You are on page 1of 425

i

K E TO G E N I C D I E T
A N D M E TA B O L I C T H E R A P I E S
ii
iii

K E TO G E N I C D I E T A N D
M E TA B O L I C T H E R A P I E S

Expanded Roles in Health and Disease

EDITED BY

S U S A N A . M A S I N O,   P H D
Vernon Roosa Professor of Applied Science
Professor of Psychology and Neuroscience
Trinity College
Hartford, CT

SECTION EDITORS

D E T L E V B O I S O N,   P H D
D O M I N I C P. D ’ AG O S T I N O,   P H D
E R I C H . KO S S O F F,   M D
J O N G M . R H O,   M D

3
iv

Oxford University Press is a department of the University of Oxford. It furthers


the University’s objective of excellence in research, scholarship, and education
by publishing worldwide. Oxford is a registered trade mark of Oxford University Press
in the UK and certain other countries.

Published in the United States of America by Oxford University Press


198 Madison Avenue, New York, NY 10016, United States of America.

© Oxford University Press 2017


All rights reserved. No part of this publication may be reproduced, stored in
a retrieval system, or transmitted, in any form or by any means, without the
prior permission in writing of Oxford University Press, or as expressly permitted
by law, by license, or under terms agreed with the appropriate reproduction
rights organization. Inquiries concerning reproduction outside the scope of the
above should be sent to the Rights Department, Oxford University Press, at the
address above.

You must not circulate this work in any other form


and you must impose this same condition on any acquirer.

Library of Congress Cataloging-in-Publication Data


Names: Masino, Susan, editor.
Title: Ketogenic diet and metabolic therapies : expanded roles in health and disease /
edited by Susan A. Masino.
Description: Oxford ; New York : Oxford University Press, [2017] | Includes bibliographical
references and index.
Identifiers: LCCN 2016019577 | ISBN 9780190497996 (alk. paper)
Subjects: | MESH: Ketogenic Diet | Metabolism—physiology
Classification: LCC RM237.73 | NLM WB 427 | DDC 613.2/83—dc23
LC record available at https://lccn.loc.gov/2016019577

This material is not intended to be, and should not be considered, a substitute for medical or
other professional advice. Treatment for the conditions described in this material is highly dependent
on the individual circumstances. And, while this material is designed to offer accurate information
with respect to the subject matter covered and to be current as of the time it was written, research and
knowledge about medical and health issues is constantly evolving and dose schedules for medications are
being revised continually, with new side effects recognized and accounted for regularly. Readers must
therefore always check the product information and clinical procedures with the most up-to-date published
product information and data sheets provided by the manufacturers and the most recent codes of conduct
and safety regulation. The publisher and the authors make no representations or warranties to readers, express
or implied, as to the accuracy or completeness of this material. Without limiting the foregoing, the publisher
and the authors make no representations or warranties as to the accuracy or efficacy of the drug dosages mentioned
in the material. The authors and the publisher do not accept, and expressly disclaim, any responsibility for
any liability, loss or risk that may be claimed or incurred as a consequence of the use and/or application of any
of the contents of this material.

1 3 5 7 9 8 6 4 2
Printed by Sheridan Books, Inc., United States of America
v

CONTENTS

Preface  ix 10. Preventing Side Effects and Diet


Contributors  xi Discontinuation  66
Cherie L. Herren, MD and
Rana R. Said, md
SECTION I: Ketogenic Diet for Epilepsy
in the Clinic SECTION II: Ketogenic Diet:
Eric H. Kossoff, MD, Section Editor Emerging Clinical Applications
1. Overview: Ketogenic Diets and Future Potential
and Pediatric Epilepsy: An Update  3 Jong M. Rho, MD, Section Editor
Eric H. Kossoff, md 11. Overview: Expanded Uses
2. “Alternative” Ketogenic Diets  5 of Ketogenic Therapies  77
Elizabeth Neal, RD, MSc, PhD Jong M. Rho, md
3. Dietary Therapy in Adults: History, 12. Metabolism-Based Treatments
Demand, and Results  16 to Counter Cancer: Scientific Rationale  79
Emily L. Johnson, md and Thomas N. Seyfried, PhD and
Mackenzie C. Cervenka, md Laura M. Shelton, PhD
4. How Do You Implement the Diet?  26 13. Ketogenic Diet as Adjunctive Therapy
for Malignant Brain Cancer  88
A. G. Christina Bergqvist, md
5. Glut1 Deficiency and Eric C. Woolf, PhD and
the Ketogenic Diets  35
Adrienne C. Scheck, PhD
14. Metabolic Therapy for Autism
Joerg Klepper, MD, PhD
Spectrum Disorder
6. Ketogenic Diet in Established and Comorbidities  101
Epilepsy Indications  40
Ning Cheng, PhD, Susan A. Masino, PhD,
Ann M. Bergin, MB, ScM, MRCP(UK) and Jong M. Rho, md
7. Ketogenic Diet for Other Epilepsies  50 15. Glucose and Ketone Metabolism in
David T. Hsieh, MD and the Aging Brain: Implications for
Elizabeth A. Thiele, MD, PhD Therapeutic Strategies to Delay the
Progression of Alzheimer’s Disease  113
8. The Ketogenic Diet
and Related Therapies in “Novel” Stephen C. Cunnane, PhD,
Situations: Idiopathic Generalized Alexandre Courchesne-Loyer, msc,
Epilepsy Syndromes  56 Valerie St-Pierre, bsc,
Camille Vandenberghe, BSC,
Sudha Kilaru Kessler, MD, MSCE Etienne Croteau, PhD, and
9. Ketogenic Diet in Status Epilepticus  60 Christian-Alexandre Castellano, PhD
Rima Nabbout, MD, PhD
vi

vi Contents

16. Ketogenic Diet and Ketones for 26. Alzheimer’s Disease: Causes


the Treatment of Traumatic Brain and Treatment  241
and Spinal Cord Injury  133
Richard L. Veech, MD, PhD, DPhil and
Femke Streijger, PhD, M. Todd King
Ward T. Plunet, PhD, and 27. Mitigation of Damage
Wolfram Tetzlaff, MD, Dr. Med, PhD from Reactive Oxygen Species
17. Anti-Inflammatory Effects of and Ionizing Radiation by
a Ketogenic Diet: Implications for Ketone Body Esters  254
New Indications  147
William Curtis, Martin Kemper, PhD,
Nina Dupuis, PhD and Alexandra Miller, PhD,
Stéphane Auvin, MD, PhD Robert Pawlosky, PHD, M. Todd King, and
18. Dietary Therapy for Neurological Richard L. Veech, MD, PhD, DPhil
Disorders: Focus on Amyotrophic 28. Metabolic Seizure Resistance via BAD
Lateral Sclerosis, Parkinson’s Disease, and KATP Channels  271
Mood Disorders, and Migraine  156
Juan Ramón Martínez-François, PhD,
Carl E. Stafstrom, MD, PhD Nika N. Danial, PhD, and
Gary Yellen, PhD
29. Lactate Dehydrogenase: A Novel
SECTION III: Ketogenic Diet in
Metabolic Target  281
the Laboratory
Nagisa Sada, PhD and
Detlev Boison, PhD, Section Editor Tsuyoshi Inoue, PhD
19. Overview of Ketogenic Diet in the 30. Effects of the Ketogenic Diet on
Laboratory: Progress on Models the Blood-Brain Barrier  289
and Mechanisms  165 Manoj Banjara, PhD and
Detlev Boison, PhD Damir Janigro, PhD
20. Ketogenic Diet and PPARgamma  167
Timothy A. Simeone, PhD SECTION IV: Ketone-Based Metabolism:
21. Ketogenic Diet in a Hippocampal General Health and Metabolic Alternatives
Slice: Models and Mechanisms  186
Dominic P. D’Agostino, PhD,
Masahito Kawamura JR., MD, PhD Section Editor
22. Metabolic Therapy and Pain  196
31. Overview of Ketone-Based
David N. Ruskin, PhD Metabolism: General Health and
23. Ketogenic Diet, Adenosine, Metabolic Alternatives  307
Epigenetics, and Antiepileptogenesis  209 Dominic P. D’Agostino, PhD
Theresa A. Lusardi, PhD and 32. Ketone Supplementation for Health
Detlev Boison, PhD and Disease  310
24. Ketogenic Diet, Aging, and Angela M. Poff, PhD,
Neurodegeneration  216 Shannon L. Kesl, PhD, and
Kui Xu, MD, PhD, Dominic P. D’Agostino, PhD
Joseph C. LaManna, PhD, and 33. Identifying the Molecular Mechanism
Michelle A. Puchowicz, PhD of the Medium Chain Triglyceride
25. Endocrine and Reproductive Effects (Ketogenic) Diet  328
of Ketogenic Diets  227 Matthew C. Walker, FRCP, PhD and
Jacob P. Harney, PhD, Robin S.B. Williams, PhD
Kathryn Gudsnuk, ms, Ami Patel, md, 34. Triheptanoin in Epilepsy and Beyond  336
Anantha R. Vellipuram, md,
Karin Borges, PhD
Sathyajit Bandaru, ms, and
David Butler, PHD
vii

Contents vii

35. Amino Acids in the Treatment of 38. Keto-Adaptation in Health


Neurological Disorders  346 and Fitness  376
Adam L. Hartman, md Parker Hyde, CSCS, CISSN,
36. 2-Deoxyglucose: Metabolic Control Vincent J. Miller, MS, and
of Seizures through Inhibition Jeff S. Volek, PhD, RD
of Glycolysis  353 39. Advancing the Awareness and
Carl E. Stafstrom, MD, PhD and Application of Ketogenic Therapies
Thomas P. Sutula, MD, PhD Globally: The Charlie Foundation
and Matthew’s Friends  386
37. Ketogenic Diets as Highly Effective
Treatments for Diabetes Mellitus Beth Zupec-Kania, RD, CD,
and Obesity  362 Jim Abrahams,
Emma Williams, MBE, and
Eric C. Westman, MD, MHS, Susan A. Masino, PhD
Emily Maguire, MSc, and
William S. Yancy Jr., MD, MHS Index  397
viii
ix

P R E FA C E

Metabolism is a fundamental cellular process, and chronic health conditions and provide general
metabolic dysfunction is associated with disease. health benefits beyond treating any particular dis-
The ketogenic diet is a metabolic therapy first pub- ease. Understanding key mechanisms underlying
lished in 1921 as an effective treatment for seizures the success of metabolic therapy is of the high-
in both children and adults, and it has been pre- est biomedical significance: it is anticipated these
scribed to a subset of patients with epilepsy ever mechanisms will apply to provide breakthroughs
since. Today there are many drugs available to for multiple common, chronic, and poorly treated
control epileptic seizures, yet this metabolic ther- disorders. Similarly, a comprehensive understand-
apy can stop seizures even when all medications ing of the range and type of acute and chronic con-
fail: for some patients a ketogenic diet is superior ditions that metabolic therapies can prevent, delay,
to all known drug treatments. The ketogenic diet or reverse is of urgent clinical importance.
was developed nearly 100 years ago because it had Here we provide a fresh view on the promise
been observed—for centuries—that fasting would of using the biochemistry of metabolism to treat
stop seizures. Adhering to a medically prescribed disease and promote health by compiling the lat-
and carefully formulated high-fat ketogenic diet est research and perspectives of leading experts on
can maintain the ketone-based metabolism used ketogenic diets and metabolic therapies. This vol-
during fasting. ume is an up-to-date and comprehensive resource
Metabolic therapy targets the most fundamen- organized into four key subsections spearheaded
tal aspect of cell function: cell energy. Targeting cell by leaders in each area: the latest clinical research
function or dysfunction metabolically is concep- for treatment of epilepsy (Eric Kossoff, MD),
tually distinct from treating a disease specifically emerging clinical applications (Jong Rho, MD),
and pharmacologically. While a pharmacological laboratory research into key mechanisms (Detlev
approach has dominated drug development, and Boison, PhD), and diverse metabolic therapies
can be effective for some symptoms and condi- to treat disease and improve health (Dominic
tions, it is also more likely to produce off-target D’Agostino, PhD). The last chapter is devoted to
side effects and less likely to produce lasting two key organizations:  the Charlie Foundation,
changes. In contrast, supporting cell energy and established in 1994 in the United States, and
promoting metabolic homeostasis can improve Matthew’s Friends, established in 2004 in the
overall health and may offer long-term benefits in United Kingdom. In the last two decades growth
preventing or modifying disease. of research in the ketogenic diet field has been
Recent basic and translational research has exponential, and the Charlie Foundation played
provided new insight into mechanisms as well an enormously important role in raising aware-
as evidence that metabolic therapy with a keto- ness and spearheading its resurgence in the clinic
genic diet can treat diverse conditions beyond and the laboratory. Ongoing efforts of the Charlie
epilepsy. New research has also provided evi- Foundation have been furthered and multiplied by
dence that alternatives which can substitute for Matthew’s Friends, and together these foundations
or complement the diet—and potentially aug- are devoted to research, education, outreach, and
ment its efficacy—may be close at hand. Evidence applications of ketogenic therapies throughout
is also mounting that ketogenic diets can reverse the world.
x

x Preface

My personal path to a research program on the with a motivated and collaborative global com-
ketogenic diet was unusual:  it arose organically munity of researchers, clinicians, patients, and
from a basic science hypothesis on the regulation advocates. The ketogenic diet has been proven to
of adenosine. Adenosine is present throughout cure devastating cases of epilepsy, and we know
the body and the central nervous system and is a that unlocking its key mechanisms—whatever
powerful neuromodulator and bioenergetic regu- they may be—will be a major biomedical break-
lator of network homeostasis. Like the ketogenic through. Together we look forward to the 100th
diet, adenosine links metabolism and brain activ- anniversary of the ketogenic diet in 2021 with
ity and has been proven to have powerful anti- optimism that metabolic therapies will offer new,
seizure, neuroprotective, and disease-modifying safe, and effective options to promote health and
benefits. Years of basic research on adenosine led cure disease.
me unexpectedly to the most important and excit- Susan A. Masino, PhD
ing work of my career thus far and connected me Hartford, CT
xi

C O N T R I BU TO R S

Jim Abrahams Karin Borges, PhD


Charlie Foundation for Ketogenic Therapies Department of Pharmacology
Santa Monica, CA School of Biomedical Sciences
The University of Queensland
Stéphane Auvin, MD, PhD St. Lucia, Australia
Service de Neurologie Pédiatrique
Hôpital Universitaire Robert-Debré David Butler, PhD
Paris, France Neuroscience Graduate Program
University of Hartford
Sathyajit Bandaru, MS West Hartford, CT
Neuroscience Graduate Program
University of Hartford Christian-Alexandre Castellano, PhD
West Hartford, CT Research Center on Aging
Departments of Medicine and
Manoj Banjara, PhD
Pharmacology and Physiology
Departments of Biomedical Engineering
University of Sherbrooke
and Cerebrovascular Research
Sherbrooke, Québec, Canada
Cleveland Clinic Foundation
Cleveland, OH
Mackenzie C. Cervenka, MD
Ann M. Bergin, MB, ScM, MRCP(UK) Director, Adult Epilepsy Diet Center
Assistant Professor of Neurology Medical Director, Epilepsy Monitoring Unit
Harvard Medical School Assistant Professor of Neurology
Children’s Hospital of Boston Johns Hopkins Hospital
Boston, MA Baltimore, MD

A. G. Christina Bergqvist, MD Ning Cheng, PhD


Medical Director, Dietary Treatment Program Department of Pediatrics
Associate Professor of Neurology and Pediatrics Alberta Children’s Hospital
Children’s Hospital of Philadelphia Research Institute
Perelman School of Medicine at Cumming School of Medicine
the University of Pennsylvania University of Calgary
Philadelphia, PA Calgary, Canada

Detlev Boison, PhD Alexandre Courchesne-Loyer, MSc


Robert Stone Dow Chair and Director Research Center on Aging
of Neurobiology Departments of Medicine and
Director of Basic and Translational Research Pharmacology and Physiology
Legacy Research Institute University of Sherbrooke
Portland, OR Sherbrooke, Québec, Canada
xii

xii Contributors

Etienne Croteau, PhD Adam L. Hartman, MD


Research Center on Aging Associate Professor of Neurology and Pediatrics
Departments of Medicine and Pharmacology Johns Hopkins Medicine
and Physiology Baltimore, MD
Université de Sherbrooke
Sherbrooke, Québec, Canada Cherie L. Herren, MD
Medical Director, OU Children’s Physicians
Stephen C. Cunnane, PhD Ketogenic Diet Program
Research Center on Aging Department of Neurology
Departments of Medicine and Pharmacology University of Oklahoma Health Science Center
and Physiology Oklahoma City, OK
University of Sherbrooke
Sherbrooke, Québec, Canada David T. Hsieh, MD
Division of Child Neurology
William Curtis Department of Pediatrics
Guest Worker, Lab of Metabolic Control San Antonio Military Medical Center
National Institute of Alcohol Abuse Fort Sam Houston, TX
and Alcoholism
Rockville, MD Parker Hyde, CSCS, CISSN
Department of Human Sciences
Dominic P. D’Agostino, PhD The Ohio State University
Associate Professor Columbus, OH
Department of Molecular Pharmacology
and Physiology Tsuyoshi Inoue, PhD
Morsani College of Medicine Department of Biophysical Chemistry
University of South Florida Graduate School of Medicine
Tampa, FL Dentistry and Pharmaceutical Sciences
Okayama University
Nika N. Danial, PhD Okayama, Japan
Department of Cancer Biology
Dana-Farber Cancer Institute Damir Janigro, PhD
Department of Cell Biology Flocel, Inc.
Harvard Medical School Cleveland, OH
Boston, MA

Nina Dupuis, PhD Emily L. Johnson, MD


Université Paris Diderot Assistant Professor of Neurology
Sorbonne Paris Cité Department of Neurology
Paris, France Johns Hopkins Hospital
Baltimore, MD
Kathryn Gudsnuk, MS
Neuroscience Graduate Program Masahito Kawamura Jr., MD, PhD
University of Hartford Department of Pharmacology
West Hartford, CT Jikei University School of Medicine
Tokyo, Japan
Jacob P. Harney, PhD
School of Mathematics and Sciences Martin Kemper, PhD
Reinhardt University Lab of Metabolic Control
Waleska, GA National Institute of Alcohol Abuse
Neuroscience Graduate Program and Alcoholism
University of Hartford National Institute of Health
West Hartford, CT Rockville, MD
xiii

Contributors xiii

Shannon L. Kesl, PhD Alexandra Miller, PhD


Department of Molecular Pharmacology Armed Forces Radiobiology
and Physiology Research Institute
Morsani College of Medicine Uniformed Services University of
University of South Florida the Health Sciences
Tampa, FL Bethesda, MD

Sudha Kilaru Kessler, MD, MSCE Vincent J. Miller, MS


Assistant Professor of Neurology and Pediatrics Department of Human Sciences
Children’s Hospital of Philadelphia The Ohio State University
Perelman School of Medicine at the University Columbus, OH
of Pennsylvania
Philadelphia, PA Rima Nabbout, MD, PhD
Reference Centre for Rare Epilepsies
M. Todd King Department of Pediatric Neurology
Lab of Metabolic Control Necker Enfants Malades Hospital
National Institute of Alcohol Abuse Assistance Publique Hopitaux de Paris
and Alcoholism Paris Descartes University
National Institute of Health Paris, France
Rockville, MD
Elizabeth Neal, RD, MSc, PhD
Joerg Klepper, MD, PhD Specialist Ketogenic Dietitian
Chief Physician Matthews Friends Clinics
Children’s Hospital Aschaffenburg Lingfield, UK
Aschaffenburg, Germany Honorary Research Associate
Department of Neuroscience
Eric H. Kossoff, MD University College London-Institute
Professor, Neurology and Pediatrics of Child Health
Medical Director, Ketogenic Diet Center London, UK
Director, Pediatric Neurology Residency Program
Johns Hopkins Hospital Ami Patel, MD
Baltimore, MD Neuroscience Graduate Program
University of Hartford
Joseph C. LaManna, PhD West Hartford, CT
Departments of Physiology and Biophysics
Case Western Reserve University
Robert Pawlosky, PhD
Cleveland, OH
Lab of Metabolic Control
National Institute of Alcohol Abuse
Theresa A. Lusardi, PhD
and Alcoholism
Robert Stone Dow Neurobiology Laboratories
National Institute of Health
Legacy Research Institute
Rockville, MD
Portland, OR

Emily Maguire, MSc Ward T. Plunet, PhD


Low Carb Genesis International Collaboration
Edinburgh, Scotland, UK on Repair Discoveries (ICORD)
University of British Columbia
Juan Ramón Martínez-François, PhD Blusson Spinal Cord Centre
Department of Neurobiology Vancouver, Canada
Harvard Medical School
Boston, MA Angela M. Poff, PhD
Department of Molecular Pharmacology
Susan A. Masino, PhD and Physiology
Vernon Roosa Professor of Applied Science Morsani College of Medicine
Professor of Psychology and Neuroscience University of South Florida
Trinity College Tampa, FL
Hartford, CT
xiv

xiv Contributors

Michelle A. Puchowicz, PhD Timothy A. Simeone, PhD


Department of Nutrition Creighton University School of Medicine
Case Western Reserve University Department of Pharmacology
Cleveland, OH Omaha, NE

Jong M. Rho, MD Valerie St-Pierre, BSc


Professor of Pediatrics and Clinical Neurosciences Research Center on Aging
Dr. Robert Haslam Chair in Pediatric Neurology Departments of Medicine and Pharmacology
Cumming School of Medicine, University and Physiology
of Calgary Université de Sherbrooke
Calgary, Canada Sherbrooke, Québec, Canada

David N. Ruskin, PhD Carl E. Stafstrom, MD, PhD


Department of Psychology and Neuroscience Professor of Neurology and Pediatrics
Program Lederer Chair in Pediatric Epilepsy
Trinity College Johns Hopkins School of Medicine
Hartford, CT Director, Division of Pediatric Neurology
Johns Hopkins Hospital
Nagisa Sada, PhD Baltimore, MD
Department of Biophysical Chemistry
Graduate School of Medicine Femke Streijger, PhD
Dentistry and Pharmaceutical Sciences International Collaboration on Repair Discoveries
Okayama University (ICORD)
Okayama, Japan Blusson Spinal Cord Centre
University of British Columbia
Rana R. Said, MD Vancouver, Canada
Director, Pediatric Neurology
Residency Program Thomas P. Sutula, MD, PhD
Department of Pediatrics, Neurology, and Department of Neurology
Neurotherapeutics School of Medicine and Public Health
UT Southwestern Medical Center University of Wisconsin
University of Texas Southwestern Madison, WI
Dallas, TX
Wolfram Tetzlaff, MD, Dr. Med, PhD
Adrienne C. Scheck, PhD Department of Zoology
Neuro-Oncology Research International Collaboration on Repair Discoveries
Barrow Brain Tumor Research Center (ICORD)
Barrow Neurological Institute® dba St. Joseph’s Blusson Spinal Cord Centre
Hospital and Medical Center University of British Columbia
Phoenix, AZ Vancouver, Canada
School of Life Sciences
Arizona State University Elizabeth A. Thiele, MD, PhD
Tempe, AZ Pediatric Epilepsy Program
Department of Neurology
Thomas N. Seyfried, PhD Massachusetts General Hospital
Biology Department Boston, MA
Boston College
Boston, MA Camille Vandenberghe, BSc
Research Center on Aging
Laura M. Shelton, PhD Departments of Medicine and Pharmacology and
Scientific Project Coordinator Physiology
Human Metabolome Technologies America University of Sherbrooke
Boston, MA Sherbrooke, Québec, Canada
xv

Contributors xv

Richard L. Veech, MD, PhD, DPhil Eric C. Woolf, PhD


Lab of Metabolic Control Neuro-Oncology Research
National Institute of Alcohol Abuse and Barrow Brain Tumor Research Center
Alcoholism Barrow Neurological Institute® dba St. Joseph’s
National Institute of Health Hospital and Medical Center
Rockville, MD Phoenix, AZ
School of Life Sciences
Anantha R. Vellipuram, MD Arizona State University
Neuroscience Graduate Program Tempe, AZ
University of Hartford
West Hartford, CT Kui Xu, MD, PhD
Departments of Physiology
Jeff S. Volek, PhD, RD and Biophysics
Department of Human Sciences Case Western Reserve University
The Ohio State University Cleveland, OH
Columbus, OH
William S. Yancy Jr., MD, MHS
Matthew C. Walker, FRCP, PhD Division of General Internal Medicine
Department of Clinical and Experimental Department of Medicine
Epilepsy Duke University Medical Center
Institute of Neurology Center for Health Services Research in
University College London Primary Care
London, UK Durham Veterans Affairs Medical Center
Duke Diet and Fitness Center
Eric C. Westman, MD, MHS Duke University Health System
Division of General Internal Medicine Durham, NC
Department of Medicine
Duke University Medical Center Gary Yellen, PhD
Durham, NC Professor of Neurobiology
Harvard Medical School
Emma Williams, MBE Boston, MA
Matthew’s Friends
Lingfield, UK Beth Zupec-Kania, RD, CD
Ketogenic Therapies LLC
Robin S.B. Williams, PhD Elm Grove, WI
Centre for Biomedical Sciences
School of Biological Sciences
Royal Holloway University of London
Egham, UK
xvi
1

SECTION I
Ketogenic Diet for Epilepsy
in the Clinic

E R I C H . K O S S O F F, M D , S E C T I O N E D I T O R
2
3

1
Overview: Ketogenic Diets and Pediatric Epilepsy
An Update

E R I C H . K O S S O F F, M D

A s it approaches its 100-year anniversary, the


ketogenic diet (KD) is reaching an interest
level not previously seen. Originally published in
with refractory epilepsy. In 2009, Dr.  Freeman
and colleagues performed the first blinded study
of the KD by having all participants consume the
1921 by Dr. Russell Wilder at the Mayo Clinic, its ketogenic diet plus a daily supplement of either
creation came at a time in which there were few saccharin (treatment group) or glucose (to pre-
other options for epilepsy (Wilder, 1921). The KD vent ketosis; control group) (Freeman et al., 2009).
was widely used for the next several decades in They found a trend toward improved seizure fre-
both children and adults, with approximately 50% quency in the saccharin group, though the effect
of patients reporting at least a 50% reduction in did not reach statistical significance, possibly due
seizures in multiple studies. The advent of phe- to complex actions of the KD that were not pre-
nytoin and other modern pharmaceutical antisei- vented with ingestion of glucose once a day. Neal
zure drugs in the 1940s and afterward relegated and colleagues randomized patients to no change
the KD to “alternative” medicine and it was largely in standard medical management or addition of
ignored by epilepsy specialists. For many decades the KD; they found that patients with refractory
it was used only as a last resort in children with epilepsy who were randomized to receive the KD
intractable epilepsy; only very select institutions were more likely to have a 50% decrease in sei-
were still implementing it sporadically. zure frequency than the control group (Neal et al.,
In 1993, one such refractory case prompted 2008). Another study by Sharma et  al. in 2013,
renewed interest in dietary therapies. Hollywood using a similar study design to Dr.  Neal’s 2008
producer Jim Abrahams brought his 2-year old trial, found the modified Atkins diet to be effective
son Charlie to Johns Hopkins Hospital, where in a randomized controlled study as well (Sharma
Charlie experienced rapid seizure control within et al., 2013). In light of the accumulating evidence
days after starting the KD. Abraham created the to support the efficacy of KDs, the International
Charlie Foundation in 1994, which revitalized Ketogenic Diet Study Group, a panel of 26 neu-
research efforts, and produced First Do No Harm, a rologists and dietitians, recommended that dietary
TV movie starring Meryl Streep, which promoted therapies be strongly considered in patients of
the KD. In 1998, the first multicenter prospective any age who had failed two to three medications
study of the KD in children with refractory epi- (Kossoff et al., 2009).
lepsy demonstrated that more than half of patients Beyond the formal prospective studies which
had a greater than 50% reduction in seizure fre- have proven efficacy, perhaps an even more impor-
quency after 6 months (Vining et al., 1998). tant factor that has led to the resurgence of dietary
In the now 20+ years since the formation of therapies has been a combination of flexibility in
the Charlie Foundation, dietary therapies have implementation and recognition of true indica-
experienced a rapid resurgence in research and tions for its use (Kossoff et al., 2009). Treating the
use. The majority of countries have implemented appropriate patients (sooner rather than later) as
KDs, and more than 100 research articles are pub- well as considering alternative diets and methods
lished yearly (Kossoff & McGrogan, 2005). Several of starting this treatment have led to widespread
randomized controlled clinical trials, crossover availability, willingness of patients and neurolo-
studies, and prospective studies have confirmed gists to consider it in their treatment algorithm,
a response rate of approximately 50% in children and better (and safer) outcomes. In this section,
4

4 section I: Ketogenic Diet for Epilepsy in the Clinic

“Ketogenic Diet for Epilepsy in the Clinic,” these understanding of just how far the clinical use of
factors are discussed in more detail. dietary therapy has come in such a short time.
First, Dr. Neal highlights that now there are not
one but four types of KD now available, each with REFERENCES
excellent reported efficacy:  the classic ketogenic Bergqvist, A.G., Schall, J.I., Gallagher, P.R., Cnaan, A.,
diet (KD), the medium chain triglyceride (MCT) and Stallings, V. A., (2005). Fasting versus grad-
diet, the low glycemic index treatment (LGIT), ual initiation of the ketogenic diet: a prospective,
and the modified Atkins diet (MAD) (chapter 2). randomized clinical trial of efficacy. Epilepsia 46,
The latter two diets have certainly been responsible 1810–1819.
for the acceptance of dietary therapies by adults, Cervenka, M.C., Henry, B., Nathan, J., Wood, S., and
which is discussed by Drs. Cervenka and Johnson Volek, J.S. (2013). Worldwide dietary therapies for
in chapter 3 (see Cervenka et al., 2013). Flexibility adults with epilepsy and other disorders. J Child
during the initiation week of the classic KD has Neurol 28, 1034–1040.
Freeman, J.M., Vining, E.P., Kossoff, E.H., Pyzik, P.L.,
also revolutionized approaches to the diet by
Ye, X., and Goodman, S.N. (2009). A blinded,
many epilepsy centers as outlined by Dr. Bergqvist
crossover study of the efficacy of the ketogenic
(chapter 4; Bergqvist et al., 2005). diet. Epilepsia 50, 322–325.
Second, pediatric epilepsy experts discuss Klepper, J. (2012). GLUT1 deficiency syndrome in
the indications for dietary therapy in pediatric clinical practice. Epilepsy Res 100, 272–277.
patients. Approximately 20  years ago, there was Kossoff, E.H., and McGrogan, J.R. (2005). Worldwide
little to no ability to predict which child would be use of the ketogenic diet. Epilepsia 46, 280–289.
a KD responder. That has radically changed due to Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
research and large cohort studies. The most famous Ballaban-Gil, K.R., Christina Bergqvist, A.G.,
indication, GLUT1 (glucose-1 transporter) defi- Blackford, R., Buchhalter, J.R., Caraballo, R.H.,
ciency syndrome, uses the KD as its primary, Helen Cross, J., Dahlin, M. G., et al. (2009). Optimal
gold-standard therapy, and Dr. Klepper has been clinical management of children receiving the keto-
involved in much of the research on this condition genic diet:  recommendations of the International
Ketogenic Diet Study Group. Epilepsia 50, 304–317.
and its response to the KD (chapter  5; Klepper,
Nabbout, R., Mazzuca, M., Hubert, P., Peudennier, S.,
2012). Drs. Bergin, Hsieh, and Thiele then discuss
Allaire, C., Flurin, V., Aberastury, M., Silva, W.,
some of the other well-known epilepsy syndromes and Dulac, O. (2010). Efficacy of ketogenic diet
and genetic indications for dietary therapy such in severe refractory status epilepticus initiating
as infantile spasms, myoclonic-astatic epilepsy, fever induced refractory epileptic encephalopa-
Dravet syndrome, Rett syndrome, tuberous scle- thy in school age children (FIRES). Epilepsia 51,
rosis complex, and more (Bergin, chapter 6; Hsieh 2033–2037.
and Thiele, chapter  7). In chapters  8 and 9 Drs. Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
Kessler and Nabbout highlight the more recent, Edwards, N., Fitzsimmons, G., Whitney, A., and
“novel” indications such as absence epilepsy, juve- Cross, J.H. (2008). The ketogenic diet for the treat-
nile myoclonic epilepsy, status epilepticus, and ment of childhood epilepsy:  a randomised con-
others that have attracted investigators in the last trolled trial. Lancet Neurol 7, 500–506.
few years (Nabbout et al., 2010). Sharma, S., Sankhyan, N., Gulati, S. & Agarwala, A.
(2013). Use of the modified Atkins diet for treat-
Lastly, Drs. Herren and Said conclude this
ment of refractory childhood epilepsy: a random-
Section with a review of the latest research on
ized controlled trial. Epilepsia 54, 481–486.
how to identify and treat the adverse effects inher- Vining, E.P., Freeman, J.M., Ballaban-Gil, K., Camfield,
ent in dietary therapy as well as how to eventually C.S., Camfield, P.R., Holmes, G.L., Shinnar, S.,
discontinue treatment when clinically indicated Shuman, R., Trevathan, E., and Wheless, J.W.
(chapter  10). This important chapter shows how (1998). A multicenter study of the efficacy of the
clinical researchers are attempting to make the diet ketogenic diet. Arch Neurol 55, 1433–1437.
safer for those who require it, especially long-term. Wilder, R.M. (1921). The effects of ketonemia on the
We hope you enjoy reading this Section and gain course of epilepsy. Mayo Clin Proc 2, 307–308.
5

2
“Alternative” Ketogenic Diets
ELIZABETH NEAL, RD, MSC, PHD

INTRODUCTION Dr. Huttenlocher and colleagues that a ketogenic


As the classical ketogenic diet fast approaches diet (KD) replacing LCT with MCT would induce
a centennial anniversary, the wider ketogenic higher ketosis and allow inclusion of significantly
landscape has expanded considerably both in more carbohydrate and protein, improving palat-
application and implementation. Although still ability and acceptance. After an initial trial of a
extensively used today, this traditional dietary KD providing 60% of total dietary energy from
therapy has been the basis for development of MCT in 12 children and adolescents with epi-
alternative ketogenic protocols. One ketogenic diet lepsy (Huttenlocher et al., 1971), further results
incorporating medium chain fatty acids is used for were reported from 18 patients aged 1.5–18 years,
many children and adolescents, who benefit from of whom 16 had over 50% seizure reduction
the generous carbohydrate allowance facilitated by (Huttenlocher 1976).
the increased ketogenic potential of medium chain Interest in the MCT diet continued with further
triglycerides. More recently, two less restrictive studies reported from the United States (Trauner
dietary approaches have been developed: the low et  al., 1985), the United Kingdom (Sills et  al.,
glycemic index treatment and the modified Atkins 1986), and Taiwan (Mak et  al., 1999). A  dietary
diet. These are now being used worldwide as the modification with less MCT (30% energy) was
advantages of a more liberal ketogenic diet are rec- also suggested in response to concerns about gas-
ognized, especially in adults and older children, trointestinal side effects of MCT given in large
supported by an increasing body of scientific data. doses (Schwartz et al., 1989). In 2008, researchers
This chapter explores the background and evi- based at Great Ormond Street Hospital in London
dence for use of these alternative ketogenic diets. published a trial of classical and MCT KDs in
intractable childhood epilepsy in which children
THE MEDIUM CHAIN aged 2–16 years were randomized to receive a diet
T R I G LY C E R I D E either immediately or after a 3-month delay with
K E TO G E N I C   D I E T no additional treatment changes (control group).
The predominant fatty acids in the human diet After 3 months, seizure frequency was significantly
contain 12 or more carbon atoms and originate lower in the 54 children in the diet group com-
from animal and plant sources of long chain tri- pared with the 49 controls (Neal et al., 2008a). Of
glycerides (LCT), which can be saturated, mono- the children who were randomized, 125 received
unsaturated, or polyunsaturated. The shorter chain dietary treatment at some stage (61 classical and
length medium chain fatty acids (6 to 12 carbon 64 MCT diets). Comparing the two diet groups
atoms) originate from medium chain triglycer- using an intention to treat analysis found no sig-
ides (MCT), whose main constituents are octa- nificant difference between the two diets; 29% of
noic (C8) and decanoic (C10) fatty acids. Dietary the MCT group had over 50% seizure reduction at
sources are limited: mainly coconut and palm 3 months (Neal et al., 2009). Tolerability or with-
kernel oils. Medium chain triglycerides have dis- drawals were also not significantly different at 3
tinct physical and metabolic differences from LCT and 6 months, with no evidence that the MCT diet
with a more efficient digestion, absorption, and caused more gastrointestinal problems; indeed a
mitochondrial transport process facilitating faster history of vomiting was significantly higher in the
metabolism to acetyl CoA. Hepatic ketone body classical KD children at 12 months. In this trial, the
production is primarily determined by the rate of MCT diet was initiated at a starting dose of 40%–
acetyl CoA generation, which led to suggestions by 50% energy from the MCT supplement, aiming to
6

6 section I: Ketogenic Diet for Epilepsy in the Clinic

BOX 2.1
MEDIUM CHAIN TRIGLYCERIDE DIETARY PROTOCOL

• Starting dose of 40%–50% energy MCT given as prescribed supplement of oil or emulsion
divided between all meals and snacks (MCT % can be increased as needed and tolerated
during dietary fine-tuning)
• Protein—10% energy, increase to ≥ 12% if low energy needs to ensure meeting protein
requirements
• Carbohydrate—15%–18% energy (may be lower in older children)
• Remaining 20%–30% energy LCT (from foods)
• Food choice lists or electronic calculation of recipes, all food weighed
• Stepwise increase to starting MCT dose over 1–2 weeks during which rest of diet can be
implemented as above, although may need extra LCT to maintain total energy intake if
slower MCT introduction required
• Full vitamin and mineral supplementation
• Carbohydrate-free medications where possible

Source: (Neal, 2012)

provide the optimal balance between gastrointes- than 70% energy can be well tolerated without side
tinal tolerance and good ketosis. However many effects (Liu and Wang, 2013). Prospective follow-
children and adolescents will need a higher dose up of 48 children and adolescents aged 1–18 years
to achieve optimal seizure control. Christiana Liu on mostly (79%) the MCT diet has recently been
reports that in her extensive experience of using reported from Holland. Responder rates were
the MCT diet in Canada, MCT at 40% to greater lower in this study, only 17% achieving over 50%

Fat - 90% Fat - 60%


Protein - 6% Protein - 30%
Carbohydrate - 4% Carbohydrate - 10%

Classical KD at a 4 : 1 ratio Low glycemic index treatment

Fat - 73% (30%-60% MCT)


Fat - 65%
Protein - 10%
Protein - 30%
Carbohydrate - 17%
Carbohydrate - 5%

Medium chain triglyceride KD Modified Atkins diet

FIGURE 2.1 Ketogenic diet therapies: a comparison of dietary energy contribution from macronutrients.
7

Chapter 2: “Alternative” Ketogenic Diets 7

seizure reduction after 3  months, increasing to Hopkins went on to trial this modified Atkins diet
23% after 6 months (Lambrechts et al., 2015). (MAD) in 20 children: 13 achieved over 50% sei-
The MCT diet is implemented using commer- zure reduction after 6 months, including four who
cially available products of MCT oil or emulsion became seizure free (Kossoff et  al., 2006). In a
(Liquigen, Nutricia, 50% MCT; Betaquik, Vitaflo, further study in 30 adults, seizures were reduced
20% MCT), which are supplied in some countries by over 50% in 10 patients after 6 months on the
on medical prescription. The remaining energy is MAD (Kossoff et al., 2008a). The real advantage of
provided from carbohydrate, protein, and LCT. this diet is that it allows free protein and calories,
Calculation of this diet is not based on the keto- so can be easier to implement and comply with
genic ratio but instead looks at the percentage of than the classical KD. Although approximating
dietary energy provided by macronutrients (Box 2.1). a ketogenic ratio of 1:1 (see Figure 2.1), the only
Total energy intake is controlled as with the classi- macronutrient strictly controlled on the MAD is
cal KD, although it will theoretically depend on the carbohydrate. A  randomized crossover compari-
figure applied for energy content of MCT, which son of daily carbohydrate limits in children sug-
is lower than LCT (Ranhotra et  al., 1995); this is gested a lower intake (10 g vs. 20 g) during the
not always reflected in the conversion factors listed initial 3 months of the MAD was associated with
on the products or used for dietary calculation. significantly higher likelihood of over 50% seizure
The MCT diet is strictly prescribed and all food reduction at 3 months, after which time carbohy-
weighed, often using food choice lists to develop drate could be increased (Kossoff et al., 2007).
meal plans. It is the most generous in carbohy- The MAD has led the way in a shift of approach
drate of all ketogenic therapies (see Figure 2.1), to implementation of ketogenic therapy. The
and many children and adolescents benefit from the emphasis had been previously on absolute preci-
flexibility this offers. Medium chain triglycerides sion in calculation and accuracy in food weigh-
should be included in all meals and snacks, and ing, albeit with very successful outcomes in many
compliance is improved by encouraging creative who followed the strict KD, but with compliance
incorporation into recipes and ketogenic drinks. problems in others. As practitioners of the diet,
The dose of MCT should be slowly built up over we initially viewed what appeared to be such a
the first week or two of treatment according to tol- liberal MAD protocol with caution. Now, over
erance, ketosis, and seizure control (see Box 2.1). 10 years on, this is being increasingly adopted as
Recent data indicate there may be additional an alternative ketogenic therapy, especially suited
specific efficacy benefits of medium chain fatty to adolescents, adults, and those unable to com-
acids. Chang et al. (2013) found C10 significantly ply with the stricter classical KD. It is used with
outperformed valproate acid in both in vitro and success worldwide in children and adults (see
in vivo models of seizure control. Neuronal cell- Table 2.1, studies included if five or more subjects),
line data from Hughes et al. (2014) suggested that and has potential for use in resource-poor coun-
C10 might increase mitochondrial number, medi- tries with more limited dietetic support (Kossoff
ated via activation of the PPARγ (peroxisome et al., 2008b).
proliferator-activated) receptor and its target genes A review in 2012 combined data from published
involved in mitochondrial biogenesis. MAD studies to examine responder rates. After
3  months of treatment, 54% of 105 children (six
THE MODIFIED studies, both retrospective and prospective) and
AT K I N S   D I E T 34% of 56 adults (three studies, all prospective) had
In 2003 Dr.  Kossoff and colleagues at Johns greater than 50% seizure reduction. Prospective
Hopkins Hospital in Baltimore published a brief data was available on 82 children (four studies), of
communication to report their use of the Atkins’s whom 52% had greater than 50% seizure reduc-
diet in six patients with epilepsy aged between 7 tion after 3  months (Auvin, 2012). The following
and 52  years; three had over 90% seizure reduc- year, Dr.  Kossoff and colleagues comprehensively
tion, of whom two became seizure free (Kossoff reviewed 10 years of the MAD with similar findings.
et al., 2003). The Atkins diet, designed in the 1970s Their review included all published primary stud-
as a weight loss treatment, restricts carbohydrates ies in which the MAD was used as the first dietary
and encourages fat in a similar way to the classical treatment and also case reports of single patients.
KD but allows free protein. It was suggested that Of a combined total of 342 children, 53% had over
this could be the basis of a less restrictive keto- 50% seizure reduction, with 15% achieving seizure
genic therapy for epilepsy, the goal being seizure freedom; in a combined total of 92 adults these
control rather than weight loss. The team at Johns figures were lower at 30% and 3%, respectively
8

TABLE 2.1 WORLDWIDE USE OF THE MODIFIED ATKINS DIET

Country Study Dietary prescription—daily


carbohydrate allowance*

Children and adolescents

Argentina Vaccarezza et al., 2014 (Retrospective, n = 9) 10% energy


Denmark Weber et al., 2009 (Prospective, n = 15) 10% energy, restricted further to 10 g
Miranda et al., 2011 (Prospective, n = 33) after 1–2 weeks if poor seizure control
(Weber)
10 g for 1st 3 months (Miranda)
Egypt El-Rashidy et al., 2013 (Prospective, n = 15) 10 g for 1st month then can increase by 5-g
increments up to 10% energy
France Porta et al., 2009 (Prospective, n = 10) 10 g for 1st month then can increase by 5-g
increments up to 10% energy
India Sharma et al., 2012 (Prospective, n = 15) 10 g
Sharma et al., 2013 (Randomized controlled
trial, n = 50 in diet group)
Sharma et al., 2015 (Retrospective, n = 25)
Iran Tonekaboni et al., 2010 (Prospective, n = 51) 10 g
Ghazavi et al., 2014 (Retrospective, n = 20)
Japan Ito et al., 2011 (Retrospective, n = 6) 10 g initially (one child needed 30 g–20 g
Kumada et al., 2012 (Prospective, n = 10) step down start, Kumada et al.)
Korea Kang et al., 2007 (Prospective, n = 14) 10 g for 1st month then can increase by
Kim et al., 2012 (Retrospective, n = 20) 5-g increments up to 10% energy
USA Kossoff et al., 2003 (Retrospective, n = 6) 10 g for 1–2 months then can increase by
Kossoff et al., 2006 (Prospective, n = 20) 5-g increments up to 20 g
Kossoff et al., 2007 (Prospective, n = 20) (20 g initially for five Sturge-Weber
Kossoff et al., 2010a (Prospective, n = 5) syndrome children in 2010 paper)
Kossoff et al., 2011a (Prospective, n = 30)
Groomes et al., 2011 (Retrospective, n = 13)
Adults
Belgium Carrette et al., 2008 (Prospective, n = 8) 20 g
Canada Smith et al., 2011 (Prospective, n = 18) 20 g
USA Kossoff et al., 2008a (Prospective, n = 30) 15 g
Cervenka et al., 2012 (Prospective, n = 25) 20 g
Kossoff et al., 2013a (Retrospective, n = 8) 20 g

* all studies allowed free calories except Kim et al., 2012, who report using 75% of recommended daily intakes

BOX 2.2
MODIFIED ATKINS DIET PROTOCOL

Carbohydrate for first month: 10 g daily for children, 10–15 g daily for adolescents and 20 g
daily for adults (does not include fiber but does include sugar alcohols)
Encourage high-fat foods, eat with each meal/snack
Free protein
Free calories but need to avoid excess weight gain
Full vitamin and mineral supplementation
Carbohydrate-free medications
Ketocal formula can be used as daily supplement for first month

Source: (Kossoff et al., 2011b)


9

Chapter 2: “Alternative” Ketogenic Diets 9

(Kossoff et al., 2013b). This review included results influencing the GI of a food include fiber content,
from Dr. Sharma and colleagues in India, who pub- cooking methods, processing, ripeness, and com-
lished the first randomized trial of the MAD in 102 bination of different macronutrients within a meal.
children with intractable epilepsy aged 2–14 years. Whole grains and other high-fiber foods will lower
Using a delayed diet start control group in a design GI, as will the addition of fat or protein. Most veg-
similar to that of Neal et  al. (2008a), seizure fre- etables and many fruits are low in GI.
quency after 3  months was significantly lower in Blood glucose levels tend to be fairly stable
the 50 diet children compared with the 52 controls while on the low-carbohydrate KD, and this obser-
(Sharma et al., 2013). One study has looked at long- vation led to the suggestion that a diet based on
term outcomes of children on the MAD: over 50% only low GI carbohydrate (<50) choices could
seizure reduction was maintained in 55% of the 54 maintain this glucose stability and facilitate a more
who continued treatment for over 6 months (Chen liberal type of ketogenic regime (see Figure 2.1).
and Kossoff, 2012). This alternative low GI treatment (LGIT) (Box 2.3)
Most centers prescribe the diet as recom- was first tested by researchers from Boston: a pre-
mended by Dr.  Kossoff and his team at Johns liminary retrospective review of 20 patients found
Hopkins (Box 2.2). A  dietary variant adopted half to have greater than 90% seizure reduction
by some UK centers and described by Magrath (Pfeifer & Thiele, 2005). Results were updated in
et al. (2012) is frequently and more appropriately 2009 with a larger review of 76 patients aged 1.5–
termed the modified ketogenic diet; this is distin- 22 years, half of whom had over 50% seizure reduc-
guished from the MAD by a more generous initial tion at 3 months, increasing to 66% by 12 months
carbohydrate allowance (up to 30 g with further (Muzykewicz et al., 2009). Interestingly, carbohy-
reduction during fine-tuning depending on sei- drate intake ranged from 15 to 150 g daily (mean
zure control) and a prescribed fat intake using 53 g at 3 months); some individuals needed a more
food choice lists. There is no published data on this restricted amount for seizure control, whereas
type of modified diet. others were able to relax the carbohydrate without
adverse effect. The same group has successfully
L O W G LY C E M I C I N D E X used LGIT in 15 children with tuberous sclero-
T R E AT M E N T sis (Larson et  al., 2012)  and a small group of six
The glycemic index (GI) is a measurement based children with Angelman syndrome studied pro-
on the blood glucose response to carbohydrate- spectively (Thibert et  al., 2012). Again, reported
containing foods (Jenkins et al., 1981); this will be carbohydrate intake was very varied, between 30
influenced by their rate of digestion and absorp- and 137 g at the 4-month follow-up.
tion, slower absorbed foods having a lower GI rat- The LGIT is now used in many centers world-
ing. Glycemic index values are compared with a wide. Results in children and adolescents have
standard reference value of glucose. Other variables been reported from Italy (retrospective data on

BOX 2.3
LOW GLYCEMIC INDEX TREATMENT DIETARY
PROTOCOL

Dietary goals set for carbohydrate, protein, and fat intake based on a target energy prescription
Carbohydrate prescribed at 10% energy (approximately 40–60 g daily depending on baseline
intake); this figure includes dietary fiber. Only carbohydrates with glycemic index < 50
allowed
Approximately 30% energy protein
Approximately 60% energy LCT
Carbohydrate should be evenly distributed over the day and always eaten with some fat and/
or protein
Foods not weighed but based on household portion sizes

Source: (Pfeifer, 2012)
10

10 section I: Ketogenic Diet for Epilepsy in the Clinic

15 patients: over half had over 50% seizure reduc- in the mean percentage of baseline seizures or
tion after a mean treatment duration of 14 months; numbers, with over 50% or 90% seizure reduc-
Coppola et al., 2011) and Iran (prospective study tion between the classical and MCT diet groups
of 42 patients: over 50% seizure reduction in 74% after 3, 6, and 12 months (Neal et al., 2009). The
after 1 month; Karimzadeh et al., 2014). An inter- authors concluded classical and MCT KDs were
esting case study reports a 13-year-old Japanese comparable in efficacy and tolerability and both
girl who was able to maintain a 50-g carbohy- had their place in the treatment of childhood epi-
drate daily LGIT choosing from specially designed lepsy. In view of the similar design between this
menus including unpolished rice and Natto (fer- trial and the more recent MAD randomized trial
mented soybeans) (Kumada et al., 2013). already mentioned (Sharma et  al., 2013), results
of the two studies are provided together in Table
A R E “A LT E R N AT I V E ” 2.2. Although we cannot directly compare data,
DIETS AS EFFECTIVE seizure improvement was lower in the KD trial,
AS THE CLAS SICAL which may in part reflect differences in baseline
K E TO G E N I C   D I E T ? patient characteristics, as a similar difference was
With the range of ketogenic therapies available, the seen in the control group.
initial clinic assessment consultation will include A number of studies have tried to compare
consideration of which diet to choose for an indi- classical KD and the MAD. A retrospective review
vidual patient. A key question for those embarking of children on classical KD or MAD found sig-
on ketogenic therapy is which diet could work best nificantly more KD children had greater than 50%
to treat the seizures. Will an alternative protocol seizure reduction at 3 months, but not at 6 months
with potential compliance and tolerance advan- (Porta et  al., 2009). A  prospective evaluation of
tages still be as effective as the strict classical KD? children on the MAD compared with a previously
The scientific literature has also discussed this treated KD group found an initial trend toward
question and certainly review of the many MAD greater KD efficacy disappeared when data were
trials shows results comparable to those for the age-adjusted (Miranda et  al., 2011). Data from
KD (Kossoff et al., 2013b), including longer-term Iran on 40 children of whom half were prescribed
follow-up data (Chen and Kossoff, 2012). Direct a classical KD and the others a MAD showed no
comparison trials of different ketogenic therapies significant difference in numbers achieving 50%
are limited. In the trial of Neal et  al. previously seizure reduction after 1–3  months (Ghazavi
discussed, there were no significant differences et al., 2014).

TABLE 2.2 RESULTS OF RANDOMIZED CONTROLLED TRIALS ON


THE KETOGENIC DIET AND THE MODIFIED ATKINS DIET

Sharma et al., 2013 (MAD) Neal et al., 2008a (classical and MCT KDs)

Subjects
Number enrolled 102 (2–14 years) 145 (2–16 years)
in study (ages)
Inclusion criteria Minimum 7 weekly seizures, tried at least 3 Minimum 7 weekly seizures, tried at least 2
anticonvulsant medications, no compli- anticonvulsant medications, no previous
ance issues or medical contraindications ketogenic diet, no medical contraindications
Results
Mean % 59% diet group (44–75) 63% diet group (50–74)
baseline seizure 95% control group (82–109) 137% control group (105–169)
frequency (CI)
>90% seizure 30% diet 7% diet
reduction 8% controls 0% controls
>50% seizure 52% diet 38% diet
reduction 12% controls 6% controls
Most frequent Constipation (46%) Constipation (33%)
side effect
11

Chapter 2: “Alternative” Ketogenic Diets 11

However other data suggest there may be randomized trials examining the use of different
benefits of a stricter diet. In a retrospectively ana- prescriptions within a particular ketogenic therapy.
lyzed multicenter group of 27 children switched A lower carbohydrate MAD initiation (10 g vs. 20
from the MAD to classical KD, additional seizure g) was associated with improved efficacy outcome
reduction was reported in 10, of whom five with (Kossoff et al., 2007), as was a higher classical KD
Doose syndrome (myoclonic astatic epilepsy) ketogenic ratio (4:1 vs. 3:1) (Seo et  al., 2007); in
became seizure-free; the authors identified the KD both these studies the benefits of a stricter diet at
as a “higher dose” of ketogenic therapy than the the outset were maintained even after increasing
MAD (Kossoff et al., 2010b). In a small pilot study, carbohydrate intake later in the course of treatment.
researchers in Egypt randomly assigned 40 young Although it is now recognized that ketosis may
children (aged 1–3 years) to either classical KD fed not be directly linked to seizure control, any cor-
by a liquid formula, MAD, or no diet treatment. relations between the two seem limited to the first
After 3 months the classical KD group showed sig- 3  months of dietary treatment (Neal et  al., 2009).
nificantly reduced seizure frequency and severity Further support is provided by results of a study of
compared with the MAD group; at 6 months the 30 children at Johns Hopkins who were given the
reduction in frequency was still significant, but not 4:1 ratio classical KD supplement Ketocal daily dur-
that in severity (El-Rashidy et al., 2013). ing the first month of the MAD; this had benefit
A randomized trial comparing efficay, saftey on seizure control when responder rate outcomes
and tolerability of classical KD and MAD in 104 were compared to published results of the MAD
children aged 1–18  years (51 KD, 53 MAD) has alone (Kossoff et  al., 2011a). This practice is now
recently been published. Mean percentage of base- frequently implemented at their institution (see
line seizures was lower in the KD group at 3 months Box 2.2), where the importance of a strict initiation
(38.6% KD, 47.9% MAD) and 6  months (33.8% period during the first month has been recognized
KD, 44.6% MAD), although differences were not (Kossoff et al., 2013b). It could perhaps be argued
statistically significant. In infants aged 1–2  years that key questions to ask when embarking on keto-
the mean and median baseline seizure frequen- genic therapy are not only “which dietary protocol
cies were much lower in the classical KD group to choose?” but also “how we are going to manage
with seizure freedom after 3 months achieved in 9 the initiation period for optimal seizure outcomes?”
(53%) of 17 KD patients compared to 4 (20%) of 20
MAD patients. The MAD showed tolerability and
side effect advantages. The authors concluded that SO WHICH DIET
while the MAD may be suitable for many children, TO   C H O O S E ?
the classical KD should always be first choice in The question of which diet to use for an individ-
those under 2 years (Kim et al., 2016). ual will also take into account age, lifestyle, food
Although one review of studies on dietary preferences, and feeding method. As illustrated
treatment in adults with refractory epilepsy con- in Figure  2.1, all three of the alternative dietary
cluded that both classical KD and MAD were protocols have less fat than the classical KD as
equally effective and tolerated (Klein et al., 2014), a proportion of the overall dietary energy. The
this was challenged by a recent meta-analysis of 12 MCT diet is the most generous in carbohydrate,
studies on ketogenic therapy in adults: 7 classical but consideration must be given to the need to
KD, 5 MAD, and one MCT. A total of 270 patients incorporate the MCT supplement into all meals
were evaluated with a combined efficacy rate of and snacks and the use of a strict prescription with
52% for classical KD and 34% for MAD and an food weighing. However this can work well for
odds ratio for therapeutic success of classical KD those who find it too difficult to follow a stricter
relative to MAD of 2.04, a significant difference carbohydrate restriction and need a more struc-
between the two types of diet (Ye et al., 2015). This tured dietary prescription. Adolescents and adults
analysis examined compliance, which was also sig- usually prefer the flexibility of the MAD with no
nificantly different between the two diets, at 38% food weighing and free protein and calories, and
classical KD and 56% MAD, suggesting that while to maintain ongoing compliance this option would
the classical KD may be more effective in adults, it routinely be recommended for these age groups.
is not as well tolerated. An alternative would be LGIT if unable to adhere
There is some evidence from the above stud- to the stricter MAD carbohydrate restriction. The
ies that a stricter diet might be more efficacious. classical KD is recommended for all ages if using a
This could be particularly important at the out- feeding tube and also for infants under 2 years. In
set of treatment, a hypothesis supported by two older children aged between 2 and 12 years it may
12

12 section I: Ketogenic Diet for Epilepsy in the Clinic

be sensible to start with a stricter KD in light of the vitamin and mineral supplementation to ensure
efficacy data previously discussed; this can then be requirements of micronutrients are met while
changed to the MAD at a later date if children and on a restricted diet. Adequate training from the
parents are happy to switch (Kossoff et al., 2013b). ketogenic team must be given prior to initiation of
The classical KD will be preferable for children any ketogenic therapy to ensure patients and car-
who have poor appetites and need small meals, the ers understand the dietary prescription and how
nutritionally at risk, and families who require close to manage the practicalities of ketogenic therapy
supervision on meal planning and the control of at home, including strategies for illness and acute
their child’s energy intake. In other cases it is clear situations.
that both the child and the family would not be All ketogenic therapies must be carefully mon-
able to comply with the high-fat classical KD and itored to ensure they can be implemented safely.
the MAD or MCT diet are better options. In view Home monitoring will include regular weight
of its reduced demands on time for training and checks; this is also important on the MAD and
supervision, the MAD would be first choice in cen- LGIT, where the calorie content is not strictly pre-
ters with fewer dietitians. This potential in devel- scribed. Traditionally, ketones would be checked
oping countries has been identified (Kossoff et al., up to twice daily at home while on the KD, as good
2008b), including the possibility of e-mail-based ketosis was thought to have been key to the success
MAD management in adults (Cervenka et  al., of treatment. This premise has been challenged
2012). However both the MAD and LGIT require with alternative diets where ketones are often
patients or families to design their own meals from much lower. Regular home testing of urine or
the food choices given; in some situations such as blood ketones is recommended on the MCT diet,
a residential multicarer setting this may be more but levels are not as high as those seen on the clas-
difficult and require greater dietetic input. sical KD (Neal et al., 2009). After the first month
The stricter classical KD has been recom- of the MAD, with a relaxing of dietary restrictions,
mended in epilepsy syndromes where rapid ketones will usually be much lower than those seen
improvement is needed, such as infantile spasms on the KD, especially in older children and adults.
or status epilepticus (Auvin, 2012)  and in Glut 1 Although Kossoff et al. (2011b) recommend only
deficiency syndrome and Doose syndrome a weekly check after this point, others suggest con-
(Miranda et  al., 2012), although the MAD has sistently high ketones above 3  mmol/L (blood
been used successfully in both spasms (Sharma β-hydroxybutyrate) in children on the MAD could
et al., 2012) and Glut 1 deficiency syndrome (Ito be important for maintaining efficacy (Kang et al.,
et al., 2011). Following observed benefits in Doose 2007). Ketones on the LGIT will usually be very
syndrome children changed from the MAD to low and undetectable in some cases; no correlation
classical KD, this switch should be considered in was seen between ketosis and seizures on this diet
this group if not seizure-free after 6–12 months on (Muzykewicz et al., 2009).
the MAD (Kossoff et al., 2010b). Clinic monitoring at regular follow-up visits
will include full laboratory studies, growth assess-
PRACTICAL ment, review of seizures, tolerance, and other
C O N S I D E R AT I O N S : benefits or adverse events. Side effects have been
I N I T I AT I O N A N D reported with the MCT diet, primarily gastrointes-
F O L L O W- U P O F tinal and usually managed with dietary manipu-
“A LT E R N AT I V E ”   D I E T S lation (Neal et  al., 2009; Liu and Wang, 2013).
Medical contraindications to ketogenic therapy Growth faltering has been reported in children on
are detailed in recommendations for clinical KD MCT diets, similar to that seen in children on a
management (Kossoff et  al., 2009). These will classical KD despite the significantly higher pro-
apply to both classical and alternative KDs and tein intake on the MCT diet (Neal et al., 2008b).
should be excluded prior to initiation with any Gastrointestinal symptoms including constipation
necessary baseline biochemistry. Although some have also been reported as a side effect of the MAD
centers do hospitalize children on initiation of (Kang et  al., 2007; Auvin, 2012; Chen & Kossoff,
the MCT diet, as with the classical KD it is now 2012; Sharma et  al., 2013). Raised lipid levels on
generally accepted that this can be started at home the MAD have been reported in children (Kang
without any prior fasting period. The daily intake et  al., 2007)  and adults (Cervenka et  al., 2014);
of MCT will be stepwise, increased as tolerated these were transient and normalized within a year
(Box 2.1). The MAD and LGIT are started on an of treatment. No significant side effects have been
out-patient basis. All dietary protocols require full reported in LGIT studies, although increases in
13

Chapter 2: “Alternative” Ketogenic Diets 13

blood urea nitrogen were detected in about a third E.H. (2012). E-mail management of the modified
of patients (Muzykewicz et al., 2009; Karimzadeh Atkins diet for adults with epilepsy is feasible and
et al., 2014). effective. Epilepsia 53, 728–732.
Fine-tuning of ketogenic therapy will aim to Cervenka, M.C., Patton, K., Eloyan, A., Henry, B., and
alleviate side effects where possible, and to opti- Kossoff, E.H. (2014). The impact of the modified
Atkins diet on lipid profiles in adults with epilepsy.
mize seizure outcomes. Adjustments to prescrip-
Nutr Neurosci [Epub ahead of print].
tions and micronutrient supplementation will also
Chang, P., Terbach, N., Plant, N., Chen, P.E., Walker,
be needed as a child grows older. The MCT dose M.C., and Williams, R.S. (2013). Seizure control
may be increased or decreased on the MCT diet, by ketogenic diet-associated medium chain fatty
as can carbohydrate intake, aiming to maximize acids. Neuropharmacology 69, 105–114.
benefit. As an addition to the MAD or modified Chen, W., and Kossoff, E.H. (2012). Long-term follow-
ketogenic diet, MCT has also been used to give a up of children treated with the modified Atkins
boost to ketosis and seizure control and aid compli- diet. J Child Neurol 27, 754–758.
ance by facilitating increased carbohydrate allow- Coppola, G., D’Aniello, A., Messana, T., Di Pasquale,
ance. This practice is evidence of a more flexible F., della Corte, R., Pascotto, A., and Verrotti, A.
approach to ketogenic therapy, designing an indi- (2011). Low glycemic index diet in children and
vidualized treatment based primarily on specific young adults with refractory epilepsy: first Italian
dietary and lifestyle requirements rather than on experience. Seizure 20, 526–528.
El-Rashidy, O.F., Nassar, M.F., Abdel-Hamid, I.A.,
a rigid diet protocol that is offered by a particular
Shatla, R.H., Abdel-Hamid, M.H., Gabr, S.S.,
hospital center. This may primarily use one type
Mohamed, S.G., El-Sayed, W.S., and Shaaban, S.Y.
of diet, but alternatively may use different aspects (2013). Modified Atkins diet vs classic ketogenic
of some, or indeed all, of the ketogenic therapies. formula in intractable epilepsy. Acta Neurol Scand
Well-defined dietary parameters are needed if 128, 402–408.
conducting research studies on a specific diet, but Ghazavi, A., Tonekaboni, S.H., Karimzadeh, P.,
anecdotal reports suggest more dietitians are tend- Nikibakhsh, A.A., Khajeh, A., and Fayyazi, A.
ing toward this “patient-tailored” prescription of (2014). The ketogenic and Atkins diets effect on
ketogenic therapy in clinical practice (Miranda intractable epilepsy:  a comparison. Iran J Child
et al., 2012). Neurol 8, 12–17.
Groomes, L.B., Pyzik, P.L., Turner, Z., Dorward, J.L.,
Goode, V.H., and Kossoff, E.H. (2011). Do patients
CONCLUSION with absence epilepsy respond to ketogenic diets? J
As worldwide use of the KD continues to grow, Child Neurol 26, 160–165.
it is clear that the alternative dietary protocols Hughes, S.D., Kanabus, M., Anderson, G., Hargreaves,
described in this chapter have an important place I.P., Rutherford, T., O’Donnell, M., Cross, J.H.,
within the treatments we can offer to children and Rahman, S., Eaton, S., and Heales, S.J. (2014).
adults with intractable seizures. The MAD in par- The ketogenic diet component decanoic acid
ticular has emerged as a therapy with great poten- increases mitochondrial citrate synthase and com-
tial for treating not only children with epilepsy plex I activity in neuronal cells. J Neurochem 129,
but also adults and those in countries with more 426–433.
limited resources. Further research will enable us Huttenlocher, P.R., Wilbourne, A.J., and Sigmore, J.M.
to optimize protocols for clinical implementa- (1971). Medium chain triglycerides as a therapy
tion to ensure the best possible outcome for those for intractable childhood epilepsy. Neurology 1,
1097–1103.
embarking on dietary treatment of epilepsy.
Huttenlocher, P.R. (1976). Ketonaemia and sei-
zures: metabolic and anticonvulsant effects of two
REFERENCES ketogenic diets in childhood epilepsy. Pediatr Res
Auvin, S. (2012). Should we routinely use modified 10, 536–540.
Atkins diet instead of regular ketogenic diet to Ito, Y., Oguni, H., Ito, S., Oguni, M., and Osawa, M.
treat children with epilepsy? Seizure 21, 237–240. (2011). A modified Atkins diet is promising as a
Carrette, E., Vonck, K., De Herdt, V., DeWaele, I., treatment for glucose transporter type 1 deficiency
Raedt, R., Goossens, L., Van Zandijcke, M., syndrome. Dev Med Child Neurol 53, 658–663.
Wadman, W., Thadani, V., and Boon P. (2008). Jenkins, D.J., Wolever, T.M., Taylor, R.H., Barker,
A pilot trial with modified Atkins’ diet in adult H., Fielden, H., Baldwin, J.M., Bowling, A.C.,
patients with refractory epilepsy. Clin Neurol Newman, H.C., Jenkins, A.L., and Goff, D.V.
Neurosurg 110, 797–803. (1981). Glycemic index of foods:  a physiological
Cervenka, M.C., Terao, N.N., Bosarge, J.L., Henry, basis for carbohydrate exchange. Am J Clin Nutr
B.J., Klees, A.A., Morrison, P.F., and Kossoff, 34, 362–366.
14

14 section I: Ketogenic Diet for Epilepsy in the Clinic

Kang, H.C., Lee, H.S., You, S.J., Kang, du C., Ko, T.S., liquid supplement during the initial month. J
and Kim, H.D. (2007). Use of a modified Atkins Child Neurol 26, 147–151.
diet in intractable childhood epilepsy. Epilepsia 48, Kossoff, E.H., Freeman, J.M., Turner, Z., and Rubenstein,
182–186. J.E. (2011b). Ketogenic Diets:  Treatments for
Karimzadeh, P., Sedighi, M., Beheshti, M., Azargashb, Epilepsy and other Disorders, 5th edition.
E., Ghofrani, M., and Abdollahe-Gorgi, F. (2014). (New York: Demos Medical Publishing).
Low glycemic index treatment in pediatric refrac- Kossoff, E.H., Henry, B.J., and Cervenka, M.C. (2013a).
tory epilepsy: the first Middle East report. Seizure Efficacy of dietary therapy for juvenile myoclonic
23, 570–572. epilepsy. Epilepsy Behav 26, 162–164.
Kim, Y.M., Vaidya, V.V., Khusainov, T., Kim, H.D., Kossoff, E.H., Cervenka, M.C., Henry, B.J., Haney,
Kim, S.H., Lee, E.J., Lee, Y.M., Lee, J.S., and Kang, C.A., and Turner, Z. (2013b). A decade of the mod-
H.C. (2012). Various indications for a modified ified Atkins diet (2003–2013): results, insights, and
Atkins diet in intractable childhood epilepsy. future directions. Epilepsy Behav 29, 437–442.
Brain Dev 34, 570–575 Kumada, T., Miyajima, T., Oda, N., Shimomura, H.,
Kim, J.A.,Yoon, J.R, Lee, E.J., Lee, J.S., Kim, J.T., Kim, Saito, K., and Fujii, T. (2012). Efficacy and toler-
H.D. and Kang, H.C. (2016). Efficacy of the classic ability of modified Atkins diet in Japanese chil-
ketogenic and the modified Atkins diets in refrac- dren with medication-resistant epilepsy. Brain Dev
tory childhood epilepsy. Epilepsia 57(1), 51–58 34, 32–38.
Klein, P., Tyrlikova, I., and Mathews, G.C. (2014). Kumada, T., Hiejima, I., Nozaki, F., Hayashi, A., and
Dietary treatment in adults with refractory epi- Fujii, T. (2013). Glycemic index treatment using
lepsy: a review. Neurology 83, 1978–1985. Japanese foods in a girl with Lennox-Gastaut syn-
Kossoff, E.H., Krauss, G.L., McGrogan, J.R., and drome. Pediatr Neurol 48, 390–392.
Freeman, J.M. (2003). Efficacy of the Atkins diet Lambrechts, D.A., de Kinderen, R.J., Vles, H.S., de
as therapy for intractable epilepsy. Neurology 61, Louw, A.J., Aldenkamp, A.P., and Majoie, M.J.
1789–1791. (2015). The MCT-ketogenic diet as a treatment
Kossoff, E.H., McGrogan, J.R., Bluml, R.M., Pillas, D.J., option in refractory childhood epilepsy:  a pro-
Rubenstein, J.E., and Vining, E.P. (2006). A modi- spective study with 2-year follow-up. Epilepsy
fied Atkins diet is effective for the treatment of Behav 51, 261–266.
intractable pediatric epilepsy. Epilepsia 47, 421–424. Larson, A.M., Pfeifer, H.H., and Thiele, E.A. (2012).
Kossoff, E.H., Turner, Z., Bluml, R.M., Pyzik, P.L., Low glycemic index treatment for epilepsy in
and Vining, E.P. (2007). A randomized, crossover tuberous sclerosis complex. Epilepsy Res 99,
comparison of daily carbohydrate limits using the 180–182.
modified Atkins diet. Epilepsy Behav 10, 432–436. Liu, Y.M., and Wang, H.S. (2013). Medium-chain tri-
Kossoff, E.H., Rowley, H., Sinha, S.R., and Vining, glyceride ketogenic diet, an effective treatment for
E.P. (2008a). A prospective study of the modi- drug-resistant epilepsy and a comparison with
fied Atkins diet for intractable epilepsy in adults. other ketogenic diets. Biomed J 36, 9–15.
Epilepsia 49, 316–319. Magrath, G., Leung, M.A., and Randall, T. (2012). The
Kossoff, E.H., Dorward, J.L., Molinero, M.R., and Modified Atkins Diet. In Dietary Treatment of
Holden, K.R. (2008b). The modified Atkins diet: a Epilepsy, E.G. Neal, ed. (Oxford: Wiley-Blackwell),
potential treatment for developing countries. pp. 89–99.
Epilepsia 49, 1646–1647. Mak, S.C., Chi, C.S., and Wan, C.J. (1999). Clinical expe-
Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E., rience of ketogenic diet on children with refractory
Ballaban-Gil, K.R., Bergqvist, A.G., Blackford, epilepsy. Acta Paediatr Taiwan 40, 97–100.
R., Buchhalter, J.R., Caraballo, R.H., Cross, Miranda, M.J., Mortensen, M., Povlsen, J.H., Nielsen,
J.H., Dahlin, M.G., et  al. (2009). Optimal clini- H., and Beniczky, S. (2011). Danish study of a mod-
cal management of children receiving the keto- ified Atkins diet for medically intractable epilepsy
genic diet:  recommendations of the International in children: can we achieve the same results as with
Ketogenic Diet Study Group. Epilepsia 50, 304–317. the classical ketogenic diet? Seizure 20, 151–155.
Kossoff, E.H., Borsage, J.L., and Comi, A.M. (2010a). Miranda, M.J., Turner, Z., and Magrath, G. (2012).
A pilot study of the modified Atkins diet for Sturge- Alternative diets to the classical ketogenic
Weber syndrome. Epilepsy Res 92, 240–243. diet--can we be more liberal? Epilepsy Res 100,
Kossoff, E.H., Bosarge, J.L., Miranda, M.J., Wiemer- 278–285.
Kruel, A., Kang, H.C., and Kim, H.D. (2010b). Will Muzykewicz, D.A., Lyczkowski, D.A., Memon, N.,
seizure control improve by switching from the Conant, K.D., Pfeifer, H.H., and Thiele, E.A.
modified Atkins diet to the traditional ketogenic (2009). Efficacy, safety, and tolerability of the low
diet? Epilepsia 51, 2496–2499. glycemic index treatment in pediatric epilepsy.
Kossoff, E.H., Dorward, J.L., Turner, Z., and Pyzik, Epilepsia 50, 1118–1126.
P. (2011a). Prospective study of the modified Neal, E.G., Chaffe, H.M., Schwartz, R., Lawson, M.,
Atkins diet in combination with a ketogenic Edwards, N., Fitzsimmons, G., Whitney, A., and
15

Chapter 2: “Alternative” Ketogenic Diets 15

Cross, J.H. (2008a). The ketogenic diet in the treat- Sharma, S., Sankhyan, N., Gulati, S., and Agarwala, A.
ment of epilepsy:  a randomised controlled trial. (2013). Use of the modified Atkins diet for treat-
Lancet Neurol 7, 500–506. ment of refractory childhood epilepsy: a random-
Neal, E.G., Chaffe, H.M., Edwards, N., Lawson, M., ized controlled trial. Epilepsia 54, 481–486.
Schwartz, R., and Cross, J.H. (2008b). Growth of Sharma, S., Jain, P., Gulati, S., Sankhyan, N., and
children on classical and MCT ketogenic diets. Agarwala A. (2015). Use of the modified Atkins
Pediatrics 122, e334–340. diet in Lennox Gastaut syndrome. J Child Neurol
Neal, E.G., Chaffe, H.M., Schwartz, R., Edwards, 30, 576–579.
N., Fitzsimmons, G., Whitney, A., and Cross, Sills, M.A., Forsythe, W.I., Haidukewych, D.,
J.H. (2009). A randomized trial of classical and MacDonald, A., and Robinson, M. (1986). The
medium-chain triglyceride ketogenic diets in medium chain triglyceride diet and intractable
the treatment of childhood epilepsy. Epilepsia epilepsy. Arch Dis Child 61, 1168–1172.
49, 1–9. Smith, M., Politzer, N., MacGarvie, D., McAndrews,
Neal, E.G. (2012). The medium chain ketogenic diet. M.P., and Del Campo, M. (2011). Efficacy and tol-
In Dietary Treatment of Epilepsy, E.G. Neal, ed. erability of the modified Atkins diet in adults with
(Oxford: Wiley-Blackwell), pp. 78–88. pharmacoresistant epilepsy:  a prospective obser-
Pfeifer, H.H., and Thiele, E. (2005). Low glycemic index vational study. Epilepsia 52, 775–780.
treatment: A liberalized ketogenic diet for treatment Thibert, R.L., Pfeifer, H.H., Larson, A.M., Raby,
of intractable epilepsy. Neurology 65, 1810–1812. A.R., Reynolds, A.A., Morgan, A.K., and Thiele,
Pfeifer, H.H. (2012). The low glycaemic index treat- E.A. (2012). Low glycemic index treatment for
ment. In Dietary Treatment of Epilepsy, E.G. seizures in Angelman syndrome. Epilepsia 53,
Neal, ed. (Oxford: Wiley-Blackwell), pp. 100–108. 1498–1502.
Porta, N., Vallée, L., Boutry, E., Fontaine, M., Dessein, Tonekaboni, S.H., Mostaghimi, P., Mirmiran, P.,
A.F., Joriot, S., Cuisset, J.M., Cuvellier, J.C., and Abbaskhanian, A., Abdollah Gorji, F., Ghofrani,
Auvin, S. (2009). Comparison of seizure reduction M., and Azizi, F. (2010). Efficacy of the Atkins
and serum fatty acid levels after receiving the keto- diet as therapy for intractable epilepsy in children.
genic and modified Atkins diet. Seizure 18, 359–364. Arch Iran Med 13, 492–497.
Ranhotra, G.S., Gelroth, J.A., and Glaser, B.K. (1995). Trauner, D.A. (1985). Medium chain triglyceride diet
Levels of medium-chain triglycerides and their in intractable seizure disorders. Neurology 35,
energy value. Cereal Chem 72, 365–367. 237–238.
Schwartz, R.H., Eaton, J., Bower, B.D., and Aynsley- Vaccarezza, M.M., Toma, M.V., Ramos Guevara, J.D.,
Green A. (1989). Ketogenic diets in the treatment Diez, C.G., and Agosta, G.E. (2014). Treatment of
of epilepsy:  short term clinical effects. Dev Med refractory epilepsy with the modified Atkins diet.
Child Neurol 31, 145–151. Arch Argent Pediatr 112, 348–351.
Seo, J.H., Lee, Y.M., Lee, J.S., Kang, H.C., and Kim HD. Weber, S., Mølgaard, C., Taudorf, K., and Uldall, P.
(2007). Efficacy and tolerability of the ketogenic (2009). Modified Atkins diet to children and ado-
diet according to lipid:nonlipid ratios:  compari- lescents with medical intractable epilepsy. Seizure
son of 3:1 with 4:1 diet. Epilepsia 48, 801–805. 18, 237–240.
Sharma, S., Sankhyan, N., Gulati, S., and Agarwala, A. Ye, F., Li, X.J., Jiang, W.L., Sun, H.B., and Liu, J. (2015).
(2012). Use of the modified Atkins diet in infantile Efficacy of and patient compliance with a keto-
spasms refractory to first-line treatment. Seizure genic diet in adults with intractable epilepsy:  a
21, 45–48. meta-analysis. J Clin Neurol 11, 26–31.
16

3
Dietary Therapy in Adults
History, Demand, and Results

E M I LY L . J O H N S O N, M D A N D M AC K E N Z I E C . C E RV E N K A , M D

H I S T O RY the benefit … may be dependent on the keto-


Dietary therapy has been used for the treatment nemia which much result from such fasts,
of epilepsy since antiquity. Hippocrates wrote of and that possibly equal good results could be
fasting “purifications” as a cure for seizures, and obtained if a ketonemia were produced by
reported that some of his contemporaries believed some other means. The ketone bodies, ace-
certain foods such as eel and goat to exacerbate toacetic acid and its derivatives, are formed
or cause seizures (Hippocrates, c. 400 bc). In the from fat and protein whenever a dispropor-
Roman era, drinking gladiators’ blood was thought tion exists between the amount of fatty acid
to be a cure for epilepsy (Barborka, 1929). and the amount of sugar actually burning in
In modern times, intermittent fasting has the tissues…. it is possible to provoke ketosis
been studied for over 100  years. Drs. Marie and by feeding diets which are very rich in fat and
Guelpa described a cyclical fasting regimen of low in carbohydrate. It is proposed, therefore,
4 days of fasting and purges followed by 4 days of to try the effect of such ketogenic diets on a
a restricted vegetarian diet. Three-quarters of the series of epileptics. (Wilder, 1921)
20 patients (adults and adolescents) with epilepsy
that were studied could not adhere to the diet for The Mayo Clinic began treating adults with
more than one cycle. Of the remaining patients, epilepsy with this “ketogenic diet” in 1924. C.J.
those who followed the diet had significant benefit Barborka wrote that “epileptic patients have an
and in some cases had seizure remission; however, unusual ability to consume and utilize fat,” and
long-term compliance with the diet was limited (in hypothesized that the benefits of ketosis may be
more than one case, by friends of the patient who due to changes in nerve cells, and “decreased irri-
provided foods that were not permitted) and they tability of nerves.” General wisdom held that the
concluded that their regimen was too difficult for acid-base balance contributed to seizures or sei-
most adults to follow (Marie and Guelpa, 1911). zure protection.
Dr. Geyelin of New York Presbyterian observed Barborka believed that dietary therapy offered
a 10-year-old boy with 4  years of refractory epi- a “ray of hope,” and recognized that while the
lepsy become cured after intermittent fasting (four diet was difficult, it was far better to try it than to
fasts over 4 months) under the care of Dr. Conklin “merely employ a sedative, and to wait.”
of Battle Creek. Dr. Geyelin then treated a 9-year- Barborka published several articles on the
old boy with a 3-day fast; his multiple daily seizures Mayo Clinic experience with the ketogenic diet.
stopped after the 2nd day. Dr. Geyelin went on to He emphasized the need for patient education, and
treat patients with intermittent fasting of length- required patients to spend 2–3 weeks under strict
ening duration (Geyelin, 1921)  and expanded supervision while learning the diet (Barborka,
these treatments to adults as well as children. In 1928). The diet was designed to mimic the metab-
22/26 patients (ages 3–35 years), he observed sei- olism of a fasting person to produce mild ketosis,
zure remission by the 10th day of fasting; 18/26 using a method originally developed for diabetics.
had marked improvement 1 year following fasting, The target maintenance diet was calculated to have
and had no further seizures. sufficient calories to maintain a neutral weight in
R.M. Wilder of the Mayo Clinic, analyzing adults; carbohydrates were limited to develop and
Geyelin’s work, was the first to speculate that maintain ketosis. The original diet consisted of six
17

Chapter 3: Dietary Therapy in Adults 17

phases with varying amounts of carbohydrates ketogenic diet is used in over 40 countries world-
and fat, with a stepwise decrease in the content of wide (Kossoff and McGrogan, 2005). Altogether,
carbohydrates and increase in the amount of fat. it is estimated that there are thousands of chil-
Sample menus reveal an emphasis on heavy cream dren currently on the ketogenic diet. While many
(100 cc of 40% cream with each meal), mayon- children do not continue dietary treatments into
naise, and butter (Barborka, 1929). Patients were adulthood, there is a large and growing population
educated to test their urine for ketosis. of children who will transition to dietary therapy
In 1930, Barborka published a series of 100 as adults.
adolescent and adult patients (ages 16–51) remain- Not all children who are treated with the keto-
ing on diet from 3 months to 5 years. Twelve of the genic diet require transition to adult epilepsy care.
100 patients achieved complete seizure remission Many children become seizure-free on the keto-
on the ketogenic diet, and of those, two relaxed genic diet and successfully wean off of the diet
to a less strict diet without food weighing, and within 2 years; however, there is a risk of seizure
maintained seizure control. Seven patients had recurrence with change to a less restrictive diet.
at least a 90% reduction in seizures, and 37 addi- At the Johns Hopkins Ketogenic Diet Center,
tional patients experienced significant benefit, Martinez et  al. reviewed 557 children who
giving a 56% response rate (Barborka, 1930). Of started the ketogenic diet between 1993 and 2007
the 44 patients who had no improvement, 23 had (Martinez et al., 2007). Sixty-six children who were
not achieved ketosis (though some patients with seizure-free discontinued the diet (after median
substantial improvement lacked consistent ketosis 2.1  years, range 0.5–8  years). Of those, 20% (13
as well). children) had seizure recurrence up to 5.5  years
In addition to seizure control, Barborka after discontinuing the diet (median:  2.4  years;
reported an improvement in patients’ cognition, minimum 0 years). Seven of those patients decided
the “appearance of intelligence, more normal atti- to restart the ketogenic diet. Risk factors for recur-
tude,” and decreased irritability. rence included an abnormal MRI and EEG with
Twelve of the 56 women had complete ces- epileptiform abnormalities. Parents and patients
sation of their menses; the seven women who with a higher risk of recurrence due to MRI and
restarted a standard diet had resumption of nor- EEG findings may elect to continue dietary ther-
mal menstrual cycles within a few months. One apy into adulthood.
woman with a history of menorrhagia had nor- Children and adolescents with chronic dis-
malization of her menstrual cycle. eases require thoughtful transition from pedi-
Across the Atlantic, Dr. C. Bastible studied 29 atric to adult specialists, generally at age 18;
institutionalized women with epilepsy in Dublin. however, discussions and planning for this tran-
Their diet included low carbohydrate biscuits sition must take place much earlier. Kossoff et al.
made with local Carrigeen moss, “an inexpensive identified 10 patients who started the ketogenic
seaweed found off the shores of Ireland … which diet or the modified Atkins diet as children in
gave excellent results.” Two of the 29 women the pediatric epilepsy center, and who remained
became seizure-free. Six of the remaining patients on dietary therapy until at least age 18 (the mean
had a 50%–90% decrease in seizures, and six had age at initiation was 10.3, range 6–16). These
an increase in seizures. Bastible concluded that patients remained on the diet from 4 to 32 years
“there is a definite hope of improvement or cure” (mean:  15.5  years). All had good to complete
for adults with epilepsy (Bastible, 1931). seizure control (2 with 100% seizure control,
With the introduction of phenytoin in 1938, 8 with 50%–99% reduction) while on dietary
which was more straightforward to initiate and to therapy. Four patients had previously attempted
maintain, the ketogenic diet was used and studied to reduce the ketogenic diet ratio or increase
less for the next 7 decades (Jóźwiak et al., 2011). carbohydrates, with immediate seizure worsen-
ing. Eight patients transitioned to adult epilepsy
DEMAND clinics; the oldest patients did so at ages 26 and
43 years (after several years of self-management).
Children Transitioning
Four patients switched from ketogenic diet to
to Adult Epilepsy Providers
modified Atkins diet (MAD; 20 grams per day
The ketogenic diet for children is widely used and net carbohydrate limit) with no worsening of
growing in popularity; in 2013, there were 148 diet seizures. All remained on anticonvulsants. Six
centers for children in North America, of which patients remained on dietary therapy, five at
half were started since 2000 (Jung et al., 2015). The the Johns Hopkins Adult Epilepsy Diet Center
18

18 section I: Ketogenic Diet for Epilepsy in the Clinic

(AEDC), and maintain good seizure control. At in the world with seizures uncontrolled by medica-
the AEDC, most patients transition to adult pro- tions. Many of these patients are not surgical can-
viders by 21 years (Kossoff et al., 2013c). didates, due to generalized epilepsy (of whom up
Children and adolescents with specific genetic to 26% may be refractory), multifocal nature, or
or mitochondrial conditions represent a popula- nonresectable locations of ictal onset.
tion that requires adult dietary therapy, as they age Patients with seizures resistant to two or more
past 18. The ketogenic diet is frequently helpful AEDs have a low chance of seizure freedom with
for mitochondrial disorders. While mitochondrial additional drugs added. In a longitudinal study of
disorders with onset in infancy or early childhood 1,098 newly diagnosed epilepsy patients followed
may be fatal within a few years, those with onset 2–26  years, 49% of patients were seizure-free on
in later childhood may benefit from the ketogenic the first AED prescribed; an additional 13.2%
diet and require transitioning to an adult epilepsy became seizure-free with the second drug tried,
provider familiar with the ketogenic diet (Kossoff 3.7% with the third AED, and 1% with the fourth;
et al., 2014). with successive AEDs added or attempted, the
Glucose transporter type 1 (GLUT1) deficiency percent of patients achieving seizure freedom with
is a rare genetic condition, caused by impaired each additional AED was less than 1% (Brodie
glucose transport into the brain and associated et al., 2012).
with an abnormality in the gene SLC2A1. The In addition to medically refractory epilepsy,
optimum treatment for GLUT1 deficiency is the the desire to avoid additional AED side effects
ketogenic diet, which may be prescribed life-long. is a consideration leading patients to pursue the
It is not known whether GLUT1 deficiency can ketogenic diet. Adverse effects from medica-
successfully transition to less restrictive forms of tions (along with psychiatric comorbidities such
dietary therapy such as the MAD (Kossoff et al., as depressive symptoms) are the largest predic-
2014). While more commonly diagnosed in child- tors of health-related quality of life in patients
hood, GLUT1 is also diagnosed in adults. Ninety- with epilepsy, much more strongly predictive of
one cases of adults with GLUT1 deficiency have patients’ perceived quality of life than seizure
been described in the literature, and the ketogenic frequency. In fact, in a study of 809 adult Italian
diet remains a cornerstone of treatment (Leen et patients with pharmacoresistant epilepsy, seizure
al., 2014). frequency and the presence of generalized tonic-
Juvenile myoclonic epilepsy (JME) is highly clonic seizures did not significantly affect qual-
treatment responsive, with 90% of patients achiev- ity of life, whereas quality of life declined with
ing seizure freedom with appropriate antiepileptic increased medication side effects (Luoni et  al.,
drugs (AEDs). However, the remaining 10% have 2011). In patients without comorbid depression,
medically resistant seizures. Dietary therapy has adverse medication effects are the main drivers of
been shown to be effective for JME in a small case health-related quality of life (Luoni et al., 2011).
series. Eight adolescents and adults (ages 15–44, The ketogenic diet has the benefit of freedom
mean 24.3) were started on the MAD for treatment from many of the adverse effects that can accom-
of JME. After 1 month, 7 remained on the MAD; pany additional medications, particularly cogni-
6 patients (75%) had >50% seizure reduction; after tive side effects.
3  months, 5 had >50% reduction. Two patients
became seizure-free (25%). The mean duration Super-Refractory Status Epilepticus
on diet at the time of publication was 13.2 months Patients with status epilepticus (prolonged sei-
(range, 0.5–40  months). Three patients had zure lasting more than 5 minutes, or recurrent
increased seizures during brief periods of non- seizures without return to baseline) are generally
compliance, but returned to seizure control when treated with benzodiazepines or other medica-
they reinitiated the diet (Kossoff et al., 2013b). As tions; if seizure activity continues despite treat-
JME is a diagnosis requiring lifelong treatment, ment with intravenous antiepileptic drugs, the
patients with medically refractory JME are a large condition is termed refractory status epilepticus,
population of adolescents and adults who could and the patient may be placed in a medically
benefit from dietary therapy. induced coma. If status epilepticus continues after
at least 24 hours of general anesthetic medications,
Refractory Epilepsy it is deemed super-refractory status epilepticus
Worldwide, there are 65 million people with epi- (SRSE), which is associated with high morbidity
lepsy; 30% are medically refractory (Moshe et al., and mortality, with up to 61% mortality reported
2015), leaving approximately 19.5  million people (Brophy et al., 2012).
19

Chapter 3: Dietary Therapy in Adults 19

The ketogenic diet has been used in children However, many people choose to continue
for SRSE since 1999 (Baumeister et al., 2004), and dietary treatment beyond the initial study periods
is now used for refractory status of different etiolo- requested (Carrette et al., 2008; Klein et al., 2010;
gies in children (O’Connor et al., 2014). Cervenka et  al., 2012), and some patients have
In 2008, the first report of ketogenic diet remained on dietary therapy as long as 32  years
for SRSE in an adult was published in France (Kossoff et al., 2013c).
(Bodenant et  al., 2008). At the University of A meta-analysis in 2015 comparing six classic
Pennsylvania, two adults in super-refractory status ketogenic diet studies and five MAD studies con-
were then successfully treated with the ketogenic cluded that adherence rates are higher in the MAD
diet, after 20  days and 101  days of seizures, with (combined compliance rate 56%) than the classic
successful medication weaning at 6 and 11  days ketogenic diet (38% adherence) (Ye et al., 2015).
following diet initiation, respectively (Wusthoff Not surprisingly, when dietary treatment
et  al., 2010). Thakur et  al. published the largest is effective, patients are motivated to continue
series of adults, a series at four medical centers of treatment. When patients decide to stop dietary
10 adult patients (median age, 33  years) treated treatment, the most common reason cited is lack
with the ketogenic diet for SRSE, of whom 70% of efficacy, followed by restrictiveness of the diet
had encephalitis. The diet started after a median (Kossoff et  al., 2008; Lambrechts et  al., 2012;
of 21.5 days (range, 2–60) and a median of seven Schoeler et al., 2014). Financial reasons have been
antiepileptic drugs were tried (range, 5–13). The cited in a few patients due to higher cost of meats
SE ceased in all nine patients who achieved ketosis, compared to processed carbohydrates (Smith
in a median of 3 days (Thakur et al., 2014). et al., 2011).
Dietary therapy is now used as an adjunct
strategy for SRSE in both children and adults, Efficacy
and in proposed treatment strategies the recom- Adults with pharmacoresistant epilepsy have
mendation has been made that the ketogenic diet response rates (defined as a ≥50% decrease in
“should probably be tried in all severe cases of seizures) of 33%–54% to the newer antiepileptic
super-refractory status epilepticus” (Shorvon & drugs (Mbizvo et al., 2012; Elger et al., 2007; Ben-
Ferlisi., 2011). Menachem et al., 2007). Rates of seizure freedom
with additional agents are much lower, with each
R E S U LT S additional add-on agent after the second pro-
viding a less than 5% chance of seizure freedom
Feasibility, Tolerability, and Adherence (Brodie et  al., 2012). Dietary therapy compares
While the ketogenic diet has been widely used in favorably with these rates in most published stud-
children in modern times, concerns over adults’ ies (Payne et al., 2011), especially as the patients
ability to tolerate the diet and maintain ketosis has starting dietary treatment are typically the most
slowed adoption for use in adults (Swink et  al., refractory patients, with mean prior AEDs tried
1997; Barborka, 1930). Modern studies report a ranging from 5.4 to 10.6 (Sirven et  al., 1999;
wide range of adherence to the ketogenic diet in Kossoff et al., 2003).
adults, 22%–75% at 3 months (Mosek et al., 2009; The classic ketogenic diet reduces seizures by
Klein et al., 2010), with significant variation. The ≥50% in 22%–55% of patients, using intent-to-
MAD, first published in 2003 as a less restrictive treat analysis (Sirven et al.,1999; Mosek et al., 2009;
alternative to the classic ketogenic diet (Kossoff Klein et al., 2014; Figure 3.1 and Table 3.1). Many
et al., 2003), has had published adherence rates of patients have even higher response rates, with 8%–
56%–100% at 3 months and 22%–77.8% at 1 year 27% of patients seeing >90% decrease in seizures
(Kossoff et al., 2003; Cervenka et al., 2012; Smith (Sirven et al.,1999; Schoeler et al., 2014) and seizure
et  al., 2011). These retention rates are somewhat freedom in up to 8% (Klein et al., 2010). In a com-
lower than those seen in add-on drug trials for parison of seizure-free months, Klein et al. found
new AEDs (75%–80% retention after 12–18 weeks; an improvement from 20% of months seizure-free
Elger et al., 2007; Ben-Menachem et al., 2007). at baseline to 56.2% of months seizure-free on the
The initial decision to begin dietary treatment ketogenic diet (Klein et al., 2010).
is not undertaken lightly. In some studies, up to The MAD has wider variability in pub-
two-thirds of eligible patients screened decline lished response rates, ranging from 12% to 67%
to participate, due to concerns about restrictive- with ≥50% seizure reduction (Smith et  al., 2011;
ness or complexity of the diet (Mosek et al., 2009, Kossoff et al., 2008; Kossoff et al., 2013b; Kossoff
18/27 declined; Klein et al., 2010, 23/35 declined). et  al., 2013c) and up to 33% of patients with
20

20 section I: Ketogenic Diet for Epilepsy in the Clinic


Response rates to dietary therapy for epilepsy in adults
(number of patients)
100%

90%

80%

70%
Percent of adults

60%
11 9 12 15 29 23 3 8 30 18 3 22 6 3 Percent decrease
50%
in seizures

40% <50%

≥50%
30% ≥90%

20%

10%

0%
2

11

11
99

09

10

14

03

08

08

13

13
01

01

01
20

20
19

20

20

20

20

20

20

20

20
s2

r2

a2
ith

la
en

ek

ei

ff

tte

ff

ff

n
ele
ht

nk
lei

se
so

so

so
N

po
os
rv

re

Sm
ec

er
ve
ho
K

os

os

os
op
M
Si

ar
br

et
K

K
er
Sc

-P
m

C
La

m
am
R
Ketogenic diet Modified Atkins diet
FIGURE 3.1 Studies of the classic ketogenic diet and the modified Atkins diet report varying response rates in adults,
ranging from 12% to 67%.

>90% reduction (Cervenka et  al., 2012; Kossoff larger proportion of ≥50% response rates (Kossoff
et al., 2013b; Kossoff et al., 2013c; Figure 3.1 and et al., 2008), though this has not been detected in
Table 3.1). other studies (Mady et al., 2003, Mosek et al., 2009).
Other dietary therapies such as the medium With regard to seizure type and response
chain triglyceride (MCT) diet and the low glyce- to diet therapies, Nei et  al. detected a trend
mic index treatment (Pfeifer and Thiele, 2005) have toward greater seizure reduction in patients with
not been widely studied in adults. In a series of 11 symptomatic generalized epilepsy, with 64% of
patients on the MCT diet (and four on the classic symptomatic generalized patients having ≥50%
ketogenic diet or a combination of MCT/ketogenic reduction versus 28% of focal epilepsies (Nei et al.,
diet during the study) Lambrechts found that 5/12 2014). In Mady et  al.’s study of 45 adolescents,
patients continued the diet at 1 year, and of those, those with multiple seizure types had a greater
two had a 50%–90% reduction in seizures, while improvement than those with complex partial
the remaining three patients had a <50% reduction or generalized seizure types alone (Mady et  al.,
(Lambrechts et  al., 2012). The mean AEDs used 2003). As discussed previously, high response
decreased slightly, from 2.7 at baseline to 2.2 at the rates and seizure freedom were seen in a small
end of the diet. series of adolescents and adults with JME, with
Disproving the initial speculations that adults 4/6 adults showing >50% decrease in seizures,
could not maintain ketosis, the majority of adults 2/6  >90% decrease, and 1/6 seizure-free (Kossoff
on ketogenic diets have been successful at achiev- et al., 2013b).
ing and maintaining urinary and/or serum ketosis In patients with GLUT1 deficiency, up to 90% of
(range of published rates, 58.3%–87.5%; Sirven patients were seizure-free on the ketogenic or MAD,
et al., 1999; Klein et al., 2010; Mosek et al., 2009); including three adults. One adult had resolution of
levels of ketosis have not been predictive of sei- generalized convulsive seizures, but had persistence
zure improvement (Mosek et al., 2009; Klein et al., of likely nonepileptic events; the other two adults
2010; Nei et al., 2014). were seizure-free (Ramm-Petersen et al., 2013).
Kossoff et al. found a trend toward patients Beyond a reduction in the number of seizures,
with more frequent seizures at baseline having a the severity or duration of seizures reportedly
21
TABLE 3.1 SUMMARY OF PUBLISHED STUDIES OF THE KETOGENIC
DIET AND MODIFIED ATKINS DIET IN ADULTS

Study first Year Journal Diet Number of >=50% >=90% Retention


author patients improvement improvement

Sirven 1999 Epilepsia Ketogenic 11 55% (6/11) 27% (3/11) 7/11 (63.6%) at 8 months
Coppola 2002 Epilepsy Res Ketogenic 5 (18–23) No age-specific information No age-specific information
Mady 2003 Epilepsia Ketogenic 45 adolescents 13/28 (46%); no age-specific 8/28 (28%); no age-specific 28/45 (62%) at 6 months
(12–19) information information
Groesbeck 2006 Dev Med Child Ketogenic 28 No age-specific 24/28 No age-specific information
Neurol information
Mosek 2009 Seizure Ketogenic 9 2/9 (22%) 2/9 (22%) at 3 months
Klein 2010 Epilepsy Behav Ketogenic 12 5/12 (42%) 2/12 (17%) 9/12 (75%) at 4 months
Lambrechts 2012 Epilepsy Behav KD or MCT 15 2/15 (13%) 5/15 (33%) at 12 months
Nei 2014 Seizure Ketogenic 29 (11–51) 13 (45%) 1/29 (3%) Mean 9 months
Schoeler 2014 Epilepsy Behav Ketogenic 23 9/23 (39%) 2/23 (8%) 9/23 (29%) at 12 months
Kossoff 2003 Neurology MAD 3 (18–52) 1/3 (33%) 1/3 (33%) 3/3 (100%) at 3 months
Carrette 2008 Clin Neurol MAD 8 1 (12%) 3/8 (37.5%) at 6 months
Neurosurg
Kossoff 2008 Epilepsia MAD 30 14/30 (47%) 1/30 (3%) 20/30 (67%) at 3 months
Smith 2011 Epilepsia MAD 18 4/18 (22%) 14/18 (78%) at 6 months
Coppola 2011 Seizure LGID 3 adults 3/3 (100%) 3/3 (100%) at 2 months
Cervenka 2012 Epilepsia MAD 22 6 (27%) 4/22 (18%) 14/22 (64%) at 3 months
Kossoff 2013 Epilepsy Behav MAD 6 adults 4/6 (67%) 2/6 (33%) 5/6 at (83%) 2 months
Ramm-Pettersen 2013 Dev Med Child MAD 3 adults 2/3 (67%) 2/3 (67%)* 1 year (at least)
Neurol

* suspicion of PNES in remaining patient not seizure-free


22

22 section I: Ketogenic Diet for Epilepsy in the Clinic

decreased, or the amount of time to recover from a of patients in one study of the ketogenic diet saw
seizure shortened in a subset of patients (Schoeler an improvement in mood and cognition, though
et al., 2014, Smith et al., 2011). 2/11 also reported impaired concentration (Sirven
Some studies have shown that weight loss et  al., 1999). Increased alertness and energy are
predicts diet efficacy, with 67% of patients with common findings, seen in 33%–65% of adults and
greater than 0.9  kg/m2 decrease in BMI having adolescents on the ketogenic diet (Mady et  al.,
>50% seizure reduction, compared with 27% of 2003; Mosek et al., 2009; Lambrechts et al., 2012;
patients with <0.9  kg/m2 decrease in BMI, with Schoeler et al., 2014). One study of the MAD that
p = .03 (Kossoff et al., 2008). However, this is not a administered detailed cognitive and depression
consistent finding in all studies (Smith et al., 2011), questionnaires found reduction in depression
and patients with weight gain can also respond to scores and improved concentration in 6/7 patients
the diet (Kossoff et al., 2008). on diet for 1 month, and in all three patients com-
pleting 6  months of the study (Carrette et  al.,
Beneficial Effects 2008). Quality of life scores tend to rise rather
Cognition and Mood than decrease on both the MAD and the keto-
Dietary treatment often has positive cognitive and genic diet, though not significantly (Carrette et al.,
mood effects in studies of adults with epilepsy 2008, Klein et al., 2010; Lambrechts et al., 2012).
(Table 3.2). Many patients report an improve- Anxiety, tension, and fatigue may all be improved
ment in cognition and mood, as well as (or even as well (Lambrechts et al., 2012).
despite the lack of) improved seizure control; in
fact, some patients with no or <50% improvement Weight Loss
in seizure frequency opt to continue dietary treat- Weight loss is often a desirable effect of ketogenic
ment for the cognitive benefits alone (Sirven et al., diet therapies, and many patients are able to lose
1999; Coppola et  al., 2002). The majority (7/11) significant amounts of weight, and may success-
fully move from a clinically “obese” body-mass
index (BMI) to a “normal” or “overweight” BMI.
TABLE 3.2 REPORTED BENEFICIAL Weight loss is of particular importance in the
AND ADVERSE EFFECTS OF DIETARY adult population as it is estimated that over one-
TREATMENT (OTHER THAN SEIZURE third of adults in the United States are obese (BMI
≥ 30 kg/m2; Ogden et al., 2014). Obesity can lead
CONTROL)
to type II diabetes, obstructive sleep apnea, and
Additional reported beneficial effects metabolic syndrome, all of which can be combated
with weight loss. In a series of studies investigating
Improved cognition weight loss with the MAD, mean weight loss was
Improved mood
7 kg over 3 months (Kossoff et al., 2008) and 10 kg
Weight loss (when desired)
over 6 months (Carrette et al., 2008), including 4 of
Increased alertness
the 11 patients who were obese when they started
Increased energy
the diet who were no longer obese at the conclu-
Reduced depression
sion (Kossoff et  al., 2008). In a separate study of
Improved quality of life
the ketogenic diet, mean BMI improved 18%,
Decreased anxiety
Decreased tension
from 33.8 (obese) to 27.5 (overweight) in 12 adults
Decreased length or severity of seizures
over 4 months, and the majority of overweight or
obese subjects had at least a 10% reduction in BMI
Reported adverse effects (Klein et al., 2010).
When patients are of average weight or under-
Nausea or vomiting
weight when starting diet therapy, total calories
Bloating
can be adjusted to prevent or reverse weight loss.
Constipation
Temporary lipid elevations
Impaired concentration Adverse Effects
Menstrual irregularities Gastrointestinal
Possible growth restriction Gastrointestinal side effects are common, with half
Kidney stones to all patients reporting some degree of nausea,
Skeletal fractures constipation, bloating, or vomiting at some point
on diet therapy; these generally resolve after the
23

Chapter 3: Dietary Therapy in Adults 23

first few days or weeks of treatment with the keto- can also cause menstrual irregularity. Barborka
genic diet (Sirven et al., 1999; Coppola et al., 2002; reported that 12/56 women had cessation of
Klein et  al., 2010). Rarely, patients are unable to their menses during ketogenic diet treatment;
continue dietary treatment due to intractable nau- however, in the seven that stopped the diet, nor-
sea or vomiting. mal menstruation resumed (Barborka, 1930).
In Sirven’s 1999 study, all nine women devel-
Lipids oped menstrual irregularities (irregular cycles
Lipids may increase on ketogenic diets and should or cessation of menses), which resolved on diet
be monitored. Sirven et  al. found a significant discontinuation (Sirven et  al., 1999). Menstrual
increase in total fasting cholesterol at 3 and at irregularities were also frequent in Mady et  al.’s
6  months on diet, with an increase of the mean 2003 study of the ketogenic diet (45% of women).
cholesterol from 208 mg/dL (range, 120–304) to Menstrual irregularities seem to be much less
291 mg/dL (220–395). Triglycerides also increased common with the MAD: there were no menstrual
at 3  months from mean 190 mg/dL (41–542) to irregularities in any of the 19 women in one study
203 mg/dL (68–417), then plateaued (Sirven et al., (Kossoff et al., 2003)  and none reported in nine
1999). The extension of this study continued to women in a second study (Smith et  al., 2011),
show a significant increase in total cholesterol and and they were present in only 1 out of 17 women
in the cholesterol/HDL ratio at the time of diet dis- in a third (Cervenka et  al., 2012). Lambrechts
continuation after up to 35  months on diet (Nei found none in two women on the ketogenic diet
et al., 2014). If extreme, lipid changes may prompt and two women on the MCT diet (Lambrechts
discontinuation of dietary therapy (Mosek et  al., et al., 2012).
2009). However, elevated lipids are not present in
all patients or in all studies, and triglycerides and Other Side Effects
LDL may not change (Klein et al., 2010). Long-term effects in patients on the ketogenic diet
Lipids increase on the MAD as well (Carrette for 6  years or more (in patients ages 7–23  years)
et al., 2008), though end lipid levels in some stud- included decrease in growth rate: at diet initiation,
ies remained within average cardiovascular risk 14/28 (50%) were at or below the 10th percentile
ranges (Kossoff et  al., 2008, Smith et  al., 2011). for weight, which increased to 23/28 (82%) at last
One study of the MAD found a decrease in tri- follow-up (Groesbeck et al., 2006). Growth restric-
glycerides with dietary treatment over 12 months tion was not related to degree of ketosis and is less
(Smith et al., 2011). of a concern in patients who begin ketogenic diets
Lipids may increase during the initial phase as adults. In one study, one-quarter of patients on
of the diet, then return to baseline: one study of ketogenic diets developed kidney stones, with a
37 adults on the MAD for at least 3 months found median of 2 years after diet onset (Groesbeck et al.,
that while total cholesterol and LDL had increased 2006). Subsequent studies have shown that urine
at 3 months, there was no difference from base- alkalinization with potassium citrate reduces the
line after 1 year (p = 0.2 and p = 0.5, respectively) risk of kidney stones (Sampath et  al., 2007). Six
(Cervenka et al., 2014). patients in the long-term study (21%) had skeletal
If cholesterol elevation is present, this may fractures, occurring a median of 18 months after
be manageable without stopping dietary therapy; diet initiation (Groesbeck et al., 2006).
one patient whose LDL doubled after 3  months Kidney stones have not been reported in other
continued the MAD, and with carnitine supple- studies of adults on dietary treatment. One patient
mentation and the substitution of saturated fats had a jaw fracture related to a seizure and stopped
for polyunsaturated fats saw his cholesterol and the diet (Mosek et  al., 2009), but other skeletal
LDL return to normal (Cervenka et  al., 2012). fractures have not been reported.
Carnitine supplementation successfully decreased
elevated triglycerides in three patients as well (Nei CONCLUSIONS
et al., 2014). Dietary treatment is feasible in adults and often
highly effective, with seizure reduction rates in
Effects on the Menstrual Cycle medically refractory populations of 33%–67%,
Menstrual irregularities and cessation of men- comparable with response rates in children.
struation are common in the starvation state. A  significant proportion of patients may become
Given that the ketogenic diet is designed to seizure-free. In addition to seizure reduction,
mimic starvation, it is not surprising that it patients may also benefit from improved mood
24

24 section I: Ketogenic Diet for Epilepsy in the Clinic

and cognition, as well as intentional weight loss. Coppola, G., Veggiotti, P., Cusmai, R., Bertoli, S.,
Lipids should be monitored, but in most cases Cardinali, S., Dionisi-Vici, C., Elia, M., Lipsi,
persistent lipid elevations can be managed with M.L., Sarnelli, C., Tagliabue., A., et al. (2002). The
diet adjustments. Diet adherence remains a major ketogenic diet in children, adolescents, and young
challenge for adults. adults with refractory epilepsy:  an Italian multi-
centric experience. Epilepsy Res 48, 221–227.
Elger, C., Bialer, M., Cramer, J.A., Maia, J., Almeida, L.,
REFERENCES and Soares-da-Silva P. (2007). Eslicarbazepine ace-
Barborka, C.J. (1928). Ketogenic diet treatment of epi- tate:  a double-blind, add-on, placebo-controlled
lepsy in adults. JAMA 91(2), 73–78. exploratory trial in adult patients with partial-
Barborka, C.J. (1929). The ketogenic diet and its use. onset seizures. Epilepsia 48, 497–504.
Med Clin North Am 12(6), 1639–1653. Geyelin, H.R. (1921). Fasting as a method of treating
Barborka, C.J. (1930). Epilepsy in adults:  results of epilepsy. M Rec 99, 1037–1039.
treatment by ketogenic diet in one hundred cases. Groesbeck, D.K., Bluml, R.M., and Kossoff, EH.
Arch Neurol Psych 23, 904–914. (2006). Long-term use of the ketogenic diet in the
Bastible, C. (1931). The ketogenic treatment of epi- treatment of epilepsy. Dev Med Child Neurol 48,
lepsy. Irish J Med Sci 6, 506–520. 978–981.
Baumeister, F.A., Oberhoffer, R., Liebhaber, G.M., Guelpa, G., and Marie, A. (1911). La lutte contre
Kunkel, J., Eberhardt, J., Holthausen, H., and l’epilepsie par la desintoication et par la reducation
Peters, J. (2004). Fatal propofol infusion syndrome alimentaire. Rev de Therap Med-Chir 78, 8–13.
in association with ketogenic diet. Neuropediatrics Hippocrates. (400 bc). On the Sacred Disease. From
35, 250–252. The Internet Classics Archive, available at: http://
Ben-Menachem, E., Biton, V., Jatuzis, D., Abou-Khalil, classics.mit.edu/ Hippocrates/ sacred.html.
B., Doty, P., and Rudd, G.D. (2007). Efficacy and Accessed July 27, 2015.
safety of oral lacosamide as adjunctive therapy in Jóźwiak, S., Kossoff, E.H., and Kotulska-Jóźwiak, K.
adults with partial-onset seizures. Epilepsia 48, (2011). Dietary treatment of epilepsy:  rebirth of
1308–1317. an ancient treatment. Neurol Neurochir Pol 45(4),
Bodenant, M., Moreau, C., Sejourne, C., Auvin, S., 370–378.
Delval, A., Cuisset, J.M., Derambure, P., Destee, A., Jung, D.E., Joshi, S.M., and Berg, A.T. (2015). How
and Defebvre, L. (2008). Interest of the ketogenic do you keto? Survey of North American pediat-
diet in refractory status epilepticus in an adult. Rev ric ketogenic diet centers. J Child Neurol 30(7),
Neurol (Paris) 164, 194–199. 868–873.
Brodie, M.J., Barry, S.J., Bamagous, G.A., Norrie, Klein, P., Janousek, J., Barber, A., and Weissberger, R.
J.D., and Kwan, P. (2012). Patterns of treatment (2010). Ketogenic diet treatment in adults with
response in newly diagnosed epilepsy. Neurology refractory epilepsy. Epilepsy Behav 19, 575–579.
78, 1548–1554. Klein, P., Tyrlikova, I., and Mathews, G.C. (2014).
Brophy, G.M., Bell, R., Claassen, J., Alldredge, B., Dietary treatment in adults with refractory epi-
Bleck, T.P., Glauser, T., LaRoche, S.M., Riviello, lepsy: a review. Neurology 18, 1978–1985.
J.J., Jr., Shutter, L., Sperling, M.R., et  al. (2012). Kossoff, E.H., Krauss, G.L., McGrogan, J.R., and
Guidelines for the evaluation and management of Freeman, JM. (2003). Efficacy of the Atkins diet as
status epilepticus. Neurocrit Care 17(1), 3–23. therapy for intractable epilepsy. Neurology 61(12),
Carrette, E., Vonck, K., de Herdt, V., Dewaele, I., Raedt, 1789–1791.
R., Goossens, L., Van Zandijcke, M., Wadman, W., Kossoff, E.H., and McGrogan, J.R. (2005). Worldwide
Thadani, V., and Boon, P. (2008). A pilot trial with use of the ketogenic diet. Epilepsia 46(2), 280–289.
modified Atkins’ diet in adult patients with refrac- Kossoff, E.H., Rowley, H., Sinha, S.R., and Vining EP.
tory epilepsy. Clin Neurol Neurosurg 110, 797–803. (2008). A prospective study of the modified Atkins
Cervenka, M., Terao, N., Bosarge, J., Henry, B., Klees, diet for intractable epilepsy in adults. Epilepsia
A., Morrison, P., and Kossoff, E. (2012). Email 49(2), 316–319.
management of the MAD for adults with epilepsy Kossoff, E.H., Cervenka, M.C., Henry, B.J., Haney,
is feasible and effective. Epilepsia 53, 728–732. C.A., and Turner, Z. (2013a). A decade of the
Cervenka, M.C., Patton, K., Eloyan, A., Henry, B., and MAD (2003–2013):  Results, insights, and future
Kossoff, E.H. (2014, Nov 10). The impact of the directions. Epilepsy Behav 29(3), 437–442.
modified Atkins diet on lipid profiles in adults with Kossoff, E.H., Henry, B.J., and Cervenka, M.C. (2013b).
epilepsy. Nutr Neurosci [Epub ahead of print]. Efficacy of dietary therapy for juvenile myoclonic
Coppola, G., D’Aniello, A., Messana, T., Di Pasquale, epilepsy. Epilepsy Behav 26, 162–164.
F., della Corge, R., Pascotto, A., and Verrotti, A. Kossoff, E.H., Henry, B.J., and Cervenka, M.C. (2013c).
(2011). Low glycemic index diet in children and Transitioning pediatric patients receiving keto-
young adults with refractory epilepsy: first Italian genic diets for epilepsy into adulthood. Seizure
experience. Seizure 20(7), 526–528. 22(6), 487–489.
25

Chapter 3: Dietary Therapy in Adults 25

Kossoff, E.H., Veggiotti, P., Genton, P., and Desguerre, Payne, N., Cross, J., Sander, J., and Sisodiya, S. (2011).
I. (2014). Transition for patients with epilepsy The ketogenic and related diets in adolescents and
due to metabolic and mitochondrial disorders. adults—A review. Epilepsia 52, 1941–1948.
Epilepsia 55(suppl 3), 37–40. Pfeifer, H.H., and Thiele, E.A. (2005). Low-glycemic-
Lambrechts, D.A., Wielders, L.H., Aldenkamp, A.P., index treatment:  a liberalized ketogenic diet for
Kessels, F.G., de Kinderen, R.J., and Majoie, M.J. treatment of intractable epilepsy. Neurology 65,
(2012). The ketogenic diet as a treatment option 1810–1812.
in adults with chronic refractory epilepsy: efficacy Ramm-Pettersen, A., Nakken, K., Skogseid, I., Randby,
and tolerability in clinical practice. Epilepsy Behav H., Skei, E., Bindoff, L., and Selmer, K.K. (2013).
23, 310–314. Good outcome in patients with early dietary treat-
Leen, W.G., Taher, M., Verbeek, M.M., Kamsteed, ment of GLUT-1 deficiency syndrome:  results
E.J., van de Warrenburg, B.P., and Willemsen, from a retrospective Norwegian study. Dev Med
MA. (2014). GLUT1 deficiency syndrome into Child Neurol 55, 440–447.
adulthood:  a follow-up study. J Neurol 261(3), Sampath, A., Kossoff, E.H., Furth, S.L., Pyzik, P.L., and
589–599. Vining, E.P. (2007). Kidney stones and the keto-
Luoni, C., Bisulli, F., Canevini, M.P., De Sarro, G., genic diet:  risk factors and prevention. J Child
Fattore, C., Galimberti, C.A., Gatti, G., La Neve, Neurol 22, 375–378.
A., Muscas, G., Specchio, L.M., et  al. (2011). Schoeler, N., Wood, S., Aldridge, V., Sander, J., Cross,
Determinants of health-related quality of life J., and Sisodiya, S. (2014). Ketogenic diet therapies
in pharmacoresistant epilepsy:  results from a for adults with epilepsy: feasibility and classifica-
large multicenter study of consecutively enrolled tion of response. Epilepsy Behav 37, 33–81.
patients using validated quantitative assessments. Shorvon, S., and Ferlisi, M. (2011). The treatment
Epilepsia 52, 2181–2191. of super-refractory status epilepticus:  a critical
Mady, M.A., Kossoff, E.H., McGregor, A.L., Wheless, review of available therapies and a clinical treat-
J.W., Pyzik, P.L., and Freeman, J.M. (2003). The ment protocol. Brain 134, 2802–2818.
ketogenic diet: adolescents can do it, too. Epilepsia Sirven, J., Whedon, B., Caplan, D., Liporace, J.,
44, 847–851. Glosser, D., O’Dwyer, J., and Sperling, M.R.
Martinez, C.C., Pyzik, P.L., and Kossoff, E.H. (2007). (1999). The ketogenic diet for intractable epi-
Discontinuing the ketogenic diet in seizure-free lepsy in adults:  preliminary results. Epilepsia 40,
children:  recurrence and risk factors. Epilepsia 1721–1726.
48(1), 187–190. Smith, M., Politzer, N., MacGarvie, D., McAndrews,
Mbizvo, G.K., Dixon, P., Hutton, J.L., and Marson, M., and del Campo, M. (2011). Efficacy and tol-
A.G. (2012). Levetiracetam add-on for drug- erability of the MAD in adults with pharmacore-
resistant focal epilepsy:  an updated Cochrane sistant epilepsy: a prospective observational study.
review. Cochrane Database Syst Rev 9. Epilepsia 52, 775–780.
Mosek, A., Natour, H., Neufeld, M., Shiff, Y., and Swink, T.D., Vining, E.P., and Freeman, J.M. (1997).
Vaisam, N. (2009). Ketogenic diet treatment in The ketogenic diet:  1997. Adv Pediatr 44,
adults with refractory epilepsy: a prospective pilot 297–329.
study. Seizure 18, 30–33. Thakur, K.T., Probasco, J. C., Hocker, S.E., Roehl, K.,
Moshe, S.L., Perucca, E., Ryvlin, P., and Tomson, Henry, B., Kossoff, E.H., Kaplan, P.W., Geocadin,
T. (2015). Epilepsy:  new advances. Lancet 385, R.G., Hartman, A.L., Venkatesan, A., et al (2014).
884–898. Ketogenic Diet for adults in super-refractory sta-
Nei, M., Ngo, L., Sirven, J., and Sperling, M. (2014). tus epilepticus. Neurology 82, 665–670.
Ketogenic diet in adolescents and adults with epi- Wilder, R.M. (1921). The effect of ketonemia on the
lepsy. Seizure 23, 439–442. course of epilepsy. Mayo Clin Proc 2, 307–308.
O’Connor, S.E., Ream, M.A., Richardson, C., Mikati, Wusthoff, C.J., Kranick, S.M., Morley, J.F., and
M.A., Trescher, W.H., Byeler, D.L., Sather, J.D., Christina Berggvist, A.G. (2010). The ketogenic
Michael, E.H., Urbanik, K.B., Richards, J.L., et  al. diet in treatment of two adults with prolonged
(2014). The ketogenic diet for the treatment of pedi- nonconvulsive status epilepticus. Epilepsia 51,
atric status epilepticus. Pediatr Neurol 50, 101–103. 1083–1085.
Ogden, C.L., Carroll, M.D., Kit, B.K., and Flegal, K.M. Ye, F., Li, X.J., Jian, W.L., Sun, H.B., and Liu, J. (2015).
(2014). Prevalence of childhood and adult obe- Efficacy of and patient compliance with a keto-
sity in the United States, 2011–2012. JAMA 311, genic diet in adults with intractable epilepsy:  a
806–814. meta-analysis. J Clin Neurol 11, 26–31.
26

4
How Do You Implement the Diet?
A . G . C H R I S T I N A B E R G Q V I S T, M D

INTRODUCTION Dietary therapies for epilepsy require periodic


The ketogenic diet (KD) has survived and thrived physical examination and laboratory testing to
for almost 100  years as an effective treatment of remain safe, promote general health, and pre-
intractable epilepsy. For many years the imple- vent side effects. This is particularly true in chil-
mentation of the diet remained in large parts dren, whose nutritional needs are continuously
unchanged from its initial conception by Wilder changing during childhood and adolescence.
(Livingston, 1951; Wilder, 1921). Management Technology has changed much of the interac-
was based on our clinical practice, strongly tied tion between the care provider and the KD team,
to its history of “fasting, calorie and fluid restric- and allows for long-distance communication.
tions.” As the use of the ketogenic diet increased Communication with the team via phone, e-mail,
locally in the United States and spread across the and electronic medical records (such as EPIC)
world, and side effects became better understood, is necessary when managing the ketogenic diet.
many of the standard practices were questioned However, it is not a replacement for a follow-up
and tested and some were changed or discontin- visit with the KD team.
ued (Kossoff and McGrogan, 2005). As a result, The ICSKD recommends that patients be
alternative implementation of the classic KD and evaluated at least four times in the first year of
creation of newer diets have emerged. Most of treatment and twice a year thereafter. Younger
the data analyzed consist of retrospective chart children, particularly infants, need more frequent
reviews but there is some support from random- (even monthly) evaluations to keep up with their
ized trials. In 2008 a statement regarding the man- nutritional needs.
agement of the KD was written by 28 authors from
nine countries. This served as a basis from which COMPOSITION OF THE
future improvements to dietary therapies were K E TO G E N I C D I E T   T E A M
made (Kossoff et  al., 2009). This document, the The ketogenic diet is best supervised by a team
International Consensus Statement for Ketogenic of experienced healthcare professionals. For a
Diet, is referred to as the ICSKD in this chapter. small program, this comprises, at a minimum, an
There are multiple books detailing site-specific epileptologist and a dietitian. For truly compre-
implementation practices of the KD (Freeman hensive care and for a larger program, this team
et  al., 2007; Kossoff et  al., 2011b; Snyder, 2006; is expanded to also include a nurse and social
Stafstrom et  al., 2008). It is not within the scope worker. Each team member contributes unique
of this chapter to provide the same detail; instead knowledge to optimize the care for these chil-
I  discuss the implementation changes that have dren and will grow as the number of patients in
occurred in the 100-year history of the KD. the dietary treatment program increases. The lat-
est addition to our team at the Children’s Hospital
NEED FOR A of Philadelphia is a chef to assist with creation of
FA M I LY- C E N T E R E D , recipes in our in-patient KD kitchen (Fenton et al.,
T E A M - BA S E D A P P ROAC H 2014; Groveman et al., 2014). Any institution that
T O   D I E TA RY T H E R A P I E S supports dietary therapies must also provide phar-
The KD is a prescribed medical diet therapy macy services to find medications with the lowest
requiring supervision from a healthcare team. It carbohydrate content, a very important task, as the
should never be confused with weight loss diets content in medication is not routinely reported by
that individuals can manage safely themselves. industry and is subject to frequent change.
27

Chapter 4: How Do You Implement the Diet? 27

Access to other subspecialties such as gastro- metabolic or neuromuscular specialists before


enterology and nutrition, nephrology, urology, attempting the KD in these disorders is recom-
endocrinology, a feeding team, and bone health mended. A  metabolic screen is recommended
experts is also essential to provide truly com- for any child for whom the KD is considered. The
prehensive care. The children started on these ICSKD suggests that this include acylcarnitine
diets today rarely have just “treatment resis- esters, urine organic acids, serum amino acids,
tant epilepsy” but carry many additional diag- lactate, and pyruvate. For screening labs that are
noses:  cerebral palsy, developmental disability, followed every 3–6 months while on the KD treat-
intellectual disability, feeding difficulties some ment, see the ICSKD.
with g-tube dependencies, behavioral difficulties,
autism spectrum disorder, and genetic conditions. I N I T I AT I O N
Adding the KD to their treatment regimen makes O F   K E TO G E N I C   D I E T
their care truly complex. Parents are important Initiating the KD can be intimidating to the
partners, and essential to the success of the diet family, caretaker and KD Team alike. A well pre-
and a keto program. They can be effective coaches pared admission and a well educated parent is the
of other parents, assisting them with nonmedi- best assurance for success. In the following section
cal information, and also become trained edu- I will review the various types of initiation meth-
cators as the keto-community grows to include ods that have been used and discuss both their
school, nursing agencies, and so forth (Chee et al., benefits and drawbacks.
2014; MacCracken and Scalisi, 1999). Creating
a keto community makes it possible to provide In-Patient versus Out-Patient Initiation
comprehensive care. The classic ketogenic diet created by Wilder, and
advocated by Johns Hopkins University (Freeman
P R E D I E T E VA L UAT I O N ( S ) , et  al., 2007)  is started in the hospital so that the
E D U C AT I O N A N D patient can be closely observed, monitored, and
COUNSELING treated if needed. Most centers in the United States
For families considering the KD, education will use an in-patient setting to start the KD, as
about dietary therapies is provided in various this makes it possible to advance the KD relatively
forms:  reading materials; referrals to Internet quickly and achieve “ketosis” within a few days
sites; foundations that advocate for the KD such (Kossoff et al., 2009). The child is closely observed
as the Charlie Foundation for ketogenic therapies by nursing staff and physicians, and interventions
(www.charliefoundation.org), Matthews Friends for hypoglycemia, acidosis, dehydration, vomiting,
(www.matthewsfriends.org), and the Epilepsy weight loss, or feeding intolerances can be insti-
Foundation (www.efa.org); DVDs, and so on. tuted in a timely fashion to minimize any com-
Some dietary treatment programs offer separate plications. Although seizures more often improve
education classes to assist the families in their deci- during the admission they can worsen from the
sion to try dietary therapies (e.g., the Children’s stress of switching metabolic substrate. In this
Hospital of Philadelphia [CHOP], www.chop.edu/ situation the in-patient setting allows for imme-
treatment/ketogenicdiet) diate adjustments in medication and ICU care,
The KD requires the body to switch from using should it be needed. While the family is in the
carbohydrates as the primary energy source to hospital, many hours of direct teaching—“hands-
using lipids. There are some disorders for which on” education—is provided by the KD team. Many
the use of the KD or fasting can lead to signifi- centers admit one child at a time to start the KD.
cant morbidity, even mortality. These include The KD team is on 24/7 permanent call to initi-
inborn errors of metabolism related to carnitine ate the KD. Organization of the admissions to a
(mitochondrial transport), beta oxidation defects, monthly basis, using a “small group setting” for
pyruvate carboxylase deficiencies, and porphyria teaching, improves efficiency and frees up the
(which requires a high-carbohydrate diet). As a team’s time to assist with out-patient management.
group, mitochondrial cytopathies are in their own The Children’s Hospital of Philadelphia now also
category, as some may benefit from KD therapies, provides cooking classes in its keto-kitchen as part
such as pyruvate dehydrogenase deficiency (Di of starting the KD (Groveman et al., 2014). These
Pisa et al., 2012; Weber et al., 2001; Wijburg et al., classes provide hands-on experience with the KD
1992), while for other cytopathies the KD would food before leaving the hospital, and improve
worsen the condition (Bergqvist, 2004; Horvath chances of acceptance and success with the KD
et  al., 2008; Kang et  al., 2007). Assistance from at home. Finally, although inborn errors in fatty
28

28 section I: Ketogenic Diet for Epilepsy in the Clinic

acid oxidation are rare, screening is not infallible. blood glucose and insulin levels, utilization of the
We have picked up a handful of children with body’s glycogen stores, slowing of the flux through
beta-oxidation defects during admission in the the glycolytic pathway, and finally utilization of
past 20 years. It is more likely that these children’s our fat stores via beta oxidation (Cahill, 1970;
defects would have been missed, and resulted in Cahill and Owen, 1970). In the process of breaking
significant morbidity had the diet been started as down fat, ketone bodies are produced and trans-
an out-patient. ported into our central nervous system for direct
Out-patient initiation of the KD can be suc- use in energy production or indirectly affecting a
cessful, and in some countries it is the standard of myriad of metabolic pathways leading to the “mir-
care (Neal et al., 2008; Rizzutti et al., 2007; Vaisleib acle of seizure reduction” (Lutas and Yellen, 2013).
et  al., 2004). The out-patient advancement of the Ketone bodies are acidic, therefore, while fast-
KD is in general slower, often over several weeks ing or while on the KD, the overall “acid load” is
before a “full KD is achieved.” The centers that increased. Ketone bodies also suppress our appetite
use out-patient initiation of the KD must have a and it is often difficult to get a lethargic, acidotic,
flexible, available staff, and the ability to sched- dehydrated child to eat a 90% fat meal without
ule these patients for frequent out-patient visits vomiting. For children, particularly young ones,
to make sure the diet is proceeding as expected this could become an issue that prolongs admis-
and that the child is safe. Transition into ketosis sions and worsens morbidity, and it frankly deters
is not directly observed. Any issues after hours some families from trying the KD. Many centers
must be directed to the emergency department. have used a “kinder, gentler, gradual” advance-
The amount of education provided is limited and ment without fast initiation. Wirrell (Wirrell
it requires that the family live near the epilepsy et al., 2002), first described their success with this
center to minimize time traveling. Benefits with approach in a retrospective case series of 14 chil-
an out-patient initiation include starting the KD in dren. The Korean group (Kim et al., 2004) stopped
the comfort of the child’s own home and a cheaper fasting all of their patients. They instead used the
overall cost. However, centers that use out-patient gradual caloric advancement approach of the 4:1
initiation often have a higher dropout rate before ratio and reported similar success in seizure reduc-
the 3-month mark when effectiveness is typically tion at 3 months in 41 patients compared with 81
determined, perhaps due to the above factors historical fasting controls. Finally, a randomized
(Levy et al., 2012). prospective trial compared the classical fasting
KD with a gradual initiation approach (Bergqvist
THE CLAS SIC et al., 2005). In 48 patients using equivalence test-
K E TO G E N I C   D I E T ing, the gradual approach (1:1, 2:1, 3:1, 4:1, daily
The classic ketogenic diet begins with fasting, advancement, full calorie KD) was equally effec-
and only fluids excluding carbohydrates are tive at reducing seizures at 3  months, compared
consumed. The duration of fasting varies. In the with the classical KD protocol. Both protocols
initial protocols from the 1920 to the 1930s fast- achieved strong ketosis by the 5th day discharge,
ing was commonly extended until 10% of body the gradual protocol about 1  day later than the
weight was lost (Livingston, 1951; Wilder, 1921). fasting. Side effects were reduced by about two-
The actual time centers fast their patients has thirds, and interventions were significantly fewer
decreased, but 12–72 hours is often implemented, in the gradual protocol, with less weight loss, mild
or “until ketones are large.” The KD is then started and severe hypoglycemia, dehydration, acidosis,
at the full 90% fat composition and at a third of need for bicarbonate and intravenous fluid admin-
the calories, advanced daily, until the full calorie istration. Vomiting was not quantified but rather
meal is tolerated. reported as present or not, and occurred in both
groups. Additional days needed in the hospital
Fasting were also reduced in the gradual group compared
The KD has a strong historical tie to fasting. The with the fasting group, with a dropout rate of 4%
father of medicine, Hippocrates prescribed fast- compared with 8% in the fasting group (very low,
ing for his epilepsy patients. It is described in the overall) (Bergqvist et al., 2005). With this data in
Bible (Mark 9:29), and was used in the early 1900s hand many centers stopped the fasting process
in a cyclic fashion (for several weeks at a time) as for their routine admission and use it only when
treatments for patients with epilepsy (Wheless, speed of achieving ketosis is of the essence, as with
2008). The KD was created to mimic the metabolic a child in status epilepticus (Cobo et  al., 2015).
changes that occur when we fast: lowering of the The gradual initiation protocol has been modified,
29

Chapter 4: How Do You Implement the Diet? 29

individualized to fit the child better. The CHOP energy expenditure (REE) and an activity factor.
protocol has eliminated the 1:1 ratio, instead using Children with epilepsy are less active than healthy
the (2:1, 3:1, 4:1) approach. Many centers do not children and those who have additional motor dis-
advance to a full 4:1 ratio, depending on the sever- abilities may need even fewer calories (Wong and
ity of the child’s epilepsy, frequency of seizures, Wirrell, 2006). All these factors makes it difficult
and the response to lower ratios (Seo et al., 2007). to estimate calories for a KD plan.
The ICSKD considers fasting as an option, but no Two studies have actually measured REE (an
longer necessary (Kossoff et al., 2009). estimate of the basal metabolic rate) in children
treated with the KD. In a short-term 6-month study
the KD did not change REE in 18 children, but
Liquid/Formula versus Food
change in the respiratory quotient (RQ) correlated
Ketogenic diet formulas have been created for chil-
with seizure reduction (Tagliabue et al., 2012). In
dren who have gastrostomy tubes, and for infants.
a longer, 15-month, prospective trial of 24 chil-
In an attempt to shorten the admission time, to
dren treated with the KD compared with 75 age-
quicken the acceptance of the high-fat KD, some
matched controls, linear growth status declined
centers use a formula or liquid eggnog to initiate
while weight status and REE were unchanged; REE
the KD with similar success. Some centers have
remained reduced in children with CP (Groleau
reported higher overall seizure reduction with the
et al., 2014). Further, although the children gained
formula-fed children, perhaps due to lower chance
weight as calories were adjusted, this weight gain
of noncompliance (Kossoff et al., 2004). The prob-
came in the form of a change in body composi-
lem with not modeling a home situation during
tion and relative increase in fat mass. That is, “you
the admission is that families are discharged hav-
become what you eat.” The increased calories did
ing had no experience or assistance cooking 90%
not prevent the height deceleration seen in these
fat meals that are palatable and have to attempt this
children, a long-term side effect now well estab-
by themselves, be it with the support of websites,
lished in several prospective studies of the KD
and other keto-coaches once home.
(Bergqvist et al., 2008; Nation et al., 2014; Spulber
et al., 2009; Vining et al., 2002; Williams et al., 2002;
Caloric Restriction see chapter on side effects). In summary, calorie
Initiation restriction is not needed for initiation or mainte-
Caloric restriction was traditionally part of the KD nance of the KD. Normal growth during the treat-
initiation, with the calories gradually advanced ment period is encouraged, but the KD appears to
during the initiation after the fast (1/3, 2/3, full cal- alter body composition and caloric adjustments do
orie 90% fat diet as tolerated). Bansal et al. found not prevent reduction in height velocity (growth
better effectiveness at 3  months in 30 children failure), which maybe IGF-1 mediated.
started on full calorie 4:1 KD without fast, com-
pared with 30 historical controls, but no difference WHEN DO WE DETERMINE
in side effects or interventions, perhaps related to EFFECTIVENES S OF THE KD?
the lack of gradual adjustment to the 90% fat diet Almost all efficacy trials related to dietary thera-
in their protocol (Bansal et al., 2014). pies determine initial effectiveness at 3  months.
This is likely due to comparisons with anticonvul-
Maintenance sive drug trials, where a 3-month design is stan-
Calories are important to a growing child, whose dard practice (Sachdeo, 2007; Schmidt et al., 2014).
needs are continuously changing during infancy Do we really need to stay on the diet that long to
and childhood. Calories are controlled on the determine whether it works? Can we stop sooner,
KD until the dietitian changes the prescription. or perhaps should we stay on the diet longer? To
Clinical practice from KD centers tells us if a answer these questions, Johns Hopkins University
child is given excessive calories and gains weight and Children’s Memorial Hospital in a combined
too quickly, it is associated with less reduction in study looked at when seizures began to reduce on
seizures and lower ketosis, until ideal body weight the ketogenic diet (Kossoff et  al., 2008)  in a ret-
is achieved (Freeman et  al., 2007). This has not rospective analysis of 99 children started on a 4:1
been tested in any trial. The calories are tradi- fasting KD or 3–4:1 gradual KD protocol. They
tionally determined by a weighed 3-day dietary found that the median time to first improvement
record provided by parents before the KD is was 5 days, with a range of 1–65 days. Reduction
started. Comparison of this caloric estimate to age in seizures occurred sooner in the children who
and gender RDA, estimated or measured resting were fasted compared with a gradual protocol, but
30

30 section I: Ketogenic Diet for Epilepsy in the Clinic

long-term effectiveness was not different between R AT I O S :   D O T H E Y M AT T E R ?


the two initiation protocols, confirming prior stud- As the KD practices were questioned and clini-
ies. Five subjects did not began to have any change cal protocols changed, less restrictive diets
in their seizures until day 60 or more. Then 4/5 had emerged:  the Modified Atkins diet (MAD)
>50%–90% reduction in seizures and one became (Kossoff et al., 2003), or unlimited protein diet, as
seizure-free. They concluded that if no change is it is called in Europe, and the low glycemic index
seen in seizures frequency by 2  months, the KD diet (Pfeifer and Thiele, 2005). These diets have
could be discontinued and that there was no need become popular alternatives to the KD because
for the full 3-month trial period. However, the of their less strict nature, less need for resources,
data can also be interpreted to confirm the use of possibly milder side-effects and claims of “similar”
a 3-month minimal trial period of the KD. A sig- effectiveness to the KD. Some have even suggested
nificant improvement including seizure freedom that the modified diets should replace the formal
in a 5% treatment resistant epilepsy cohort is not KD for the majority of our treatment-resistant
unsignificant! patients (Auvin, 2012; Miranda et al., 2012). What
data do we have to support this claim? Both diets
SEIZURE FREEDOM are lower ratio KD, MAD at most a 2:1 if tightly
Seizure freedom is the goal for majority of controlled and LGIT a 1–1.5:1 ratio.
patients trying the KD. In the short-term ran- Animal data to date suggest that higher ratios
domized trials it has been reported in 15%–55% are more effective in controlling seizures, although
( Martin et al, 2016). For longer-term studies the the ratios used in rodent models (4–6.3:1) are
data presented is not always a continuous vari- often much higher than what we would use in
able, instead often obtained just prior to the humans (Bough et  al., 1999; Bough et  al., 2000;
time of interest, thereby artificially inflating the Nylen et al., 2005). Some clinical data to compare
percentage reported as seizure-free. This makes ratios have been randomized, but not blinded. Seo
it difficult to interpret the data and counsel our et al. compared a 3:1 with a 4:1 KD in 76 random-
families appropriately. Taub et al. tried to answer ized patients, and seizure reduction was measured
this question in a retrospective chart review of at 3  months. They found that the 4:1 ratio KD
275 patients started on the KD, of whom 65 chil- reduced seizures more effectively and that there
dren (24%) achieved seizure freedom (defined were significantly more seizure-free patients in
as no seizure for 28  days) (Taub et  al., 2014). this group than the 3:1 group (Seo et al., 2007). At
All children had daily seizures before starting 3  months, they crossed the two protocols for an
the KD. The median time to becoming seizure- additional 3-month extension. Children who were
free was 1.5  months, 72% became seizure-free seizure-free at the 3-month mark on the 4:1 main-
before 3 months, (considered early seizure-free), tained their seizure freedom on the lower ratio,
while 28% became seizure-free after 3  months, indicating that perhaps a lower ratio can be used
(considered late seizure-free). The longest time later in the KD treatment without sacrificing effec-
to becoming seizure-free was 18  months in one tiveness. El-Rashidy et al. studied 40 patients with
subject. Seizure recurrence (defined as any sei- symptomatic treatment-resistant epilepsy ran-
zure after having been seizure-free for 28  days) domized to MAD, 4:1 KD formula, and standard
occurred in 84%, and median time to recur- diet continued medical treatment. They found that
rence was 3 months. However, 60% of those who the 4:1 formula at 3 and 6 months resulted in better
recurred still had less than one seizure per month. seizure reduction than the MAD and regular diet
Timing of discontinuation of the AED did not (El-Rashidy et  al., 2013). Raju et  al. randomized
affect return of seizures. In general, for those who 38 children to a 4:1 versus 2.5:1 KD and measured
became seizure-free and recurred, their seizures effectiveness at 3 months. There was no significant
did not return to their prediet frequency. The difference in responders or seizure-freedom rate
chance of remaining seizure-free at 18  months at 3 months (58% vs. 63% and 26 vs. 21%) (Raju
was only 3%. Families should be counseled that et al., 2011).
seizure freedom may occur at some point during Kossoff et al. looked at MAD versus MAD with
KD treatment, most often in the first few months. liquid 4:1 supplement in the first month and found
If it happens, recurrence is more common rather that the liquid supplement (i.e., higher ratio) had
than complete remission. In general, seizures a better reduction in seizures at 3 months (Kossoff
remain greatly improved and do not return to the et  al., 2011a). In a multicenter study, 27 patients
frequency before the KD was started. from several countries were identified who had
31

Chapter 4: How Do You Implement the Diet? 31

not responded completely to the MAD and who baseline 6 weeks and 3 months into the KD. They
were switched to the KD. Ten had further improve- had a low response rate over all, only 26%, but did
ment, 11 did not. Children with myoclonic astatic notice a proportional reduction in IED of 30%
epilepsy appeared to particularly improve with the during sleep strongly predicted reduction in sei-
formal KD (Kossoff et al., 2010). In summary, cur- zure or response status (Ebus et al., 2014).
rent data animal and human-based studies, some
prospective and randomized but none blinded, D U R AT I O N O F   T R E AT M E N T
indicate that higher ratios and the formal KD are The KD was initially created for a 2-year treat-
more effective at reducing seizures than the modi- ment period and a 6- to 12-month wean. However,
fied diets, at least in the short term. It is premature there is no definitive treatment period, and it can
to routinely discard the KD in favor for the modi- be individualized based on the child’s under-
fied diets. There are situations where the modified lying condition and response. There are some
diets are more appropriate (see chapter on modi- electroclinical epilepsy syndromes, such as infan-
fied diets). When choosing the MAD, families tile spasms, where if the patient responds and
should be counseled that these less restrictive diets becomes seizure-free, the diet treatment period
might come at the expense of less reduction in sei- may be shortened to 6–12 months, particularly if
zures. A blinded randomized trial with long-term the EEG has normalized (Kossoff et al., 2002). For
follow-up to assess retention of seizure control others, the KD has resulted in such improvement
and seizure-freedom rate is needed to definitively in the child’s overall quality of life, not only reduc-
answer this question. tion in seizures but also removal of all or most
medications, that the family chooses to stay on the
P R E D I C TO R O F   R E S P O N S E KD longer (Hallbook et  al., 2007). Most centers
The KD appears to work particularly well in some have a few patients who have been on the KD for
electroclinical epilepsy syndromes. However many >15 years (Kossoff et al., 2007). Often the ratios in
patients do to fit into one of these categories, there- the KD in this group of patients have been weaned
fore a predictor of response to the dietary therapies to the lowest ratio that successfully maintains sei-
would be very helpful when trying to guide par- zure control, but attempts to come off the KD have
ents about staying on the KD or not. In the mul- resulted in return of seizures. Continued diet use
tiple observational studies published there were no of dietary therapies has to be weighed against the
demographic parameters that predicted response risk and complications and long-term side effects
(such as age, gender, seizure type). of a high-fat diet (Zupec-Kania and Zupanc, 2008;
The electroencephalogram however, appears see chapter on side effects). Patients who are on the
to be a helpful tool. Kessler et al. in a prospective KD for life, due to their underlying inborn error of
blinded study of 37 patients on the fast or gradual metabolism such as Glut1 syndrome and pyruvate
KD initiation protocol assessed the routine EEG dehydrogenase disorder require particular care
at baseline, 1 and 3  months into KD treatment. in management of their diets (see chapter about
The EEGs were analyzed for background slowing, Glut1).
interictal epileptiform discharges (IED), power
spectrum analysis, and manual spike index. They REFERENCES
found that 70% of the patients became respond- Auvin, S. (2012). Should we routinely use modified
ers, with 27% seizure-free. Over time the EEG Atkins diet instead of regular ketogenic diet to
background slowing improved and there was an treat children with epilepsy? Seizure 21, 237–240.
overall increase in beta, but this change did not Bansal, S., Cramp, L., Blalock, D., Zelleke, T.,
predict responder status. Interictal epileptiform Carpenter, J., and Kao, A. (2014). The ketogenic
discharges were reduced by 65% by the 1-month diet:  initiation at goal calories versus gradual
EEG. The 1-month reduction in IED strongly pre- caloric advancement. Pediatr Neurol 50, 26–30.
Bergqvist, A.G., Schall, J.I., Gallagher, P.R., Cnaan, A.,
dicted response status to KD with odds ratio of
and Stallings, V.A. (2005). Fasting versus gradual
4.8 (95% CI 1.1–21.3, p  =  .03). They concluded initiation of the ketogenic diet:  a prospective,
that the routine EEG is an excellent predictor of randomized clinical trial of efficacy. Epilepsia 46,
response and can assist neurologists when coun- 1810–1819.
seling and encouraging parents about staying with Bergqvist, A.G., Schall, J.I., Stallings, V.A., and Zemel,
the KD in the first 3  months of therapy (Kessler B.S. (2008). Progressive bone mineral content loss
et  al., 2011). Ebus et  al. confirmed these find- in children with intractable epilepsy treated with
ings via 24-hour video-EEG in 34 patients on at the ketogenic diet. Am J Clin Nutr 88, 1678–1684.
32

32 section I: Ketogenic Diet for Epilepsy in the Clinic

Bergqvist, A.G.C. (2004). Indications and contraindi- Groleau, V., Schall, J.I., Stallings, V.A., and Bergqvist,
cations of the ketogenic diet. In Epilepsy and the C.A. (2014). Long-term impact of the ketogenic
Ketogenic Diet, C.E. Stafstrom and J.M. Rho, eds. diet on growth and resting energy expenditure in
(Totowa, NJ: Humana Press), pp. 115–117. children with intractable epilepsy. Dev Med Child
Bough, K.J., Chen, R.S., and Eagles, D.A. (1999). Path Neurol 56, 898–904.
analysis shows that increasing ketogenic ratio, but Groveman, S., Fenton, C., Chee, C., Rice, C., and
not beta-hydroxybutyrate, elevates seizure thresh- Bergqvist, A.G.C. (2014). Use of a ketogenic teach-
old in the rat. Dev Neurosci 21, 400–406. ing kitchen in acute care setting. In 4th Global
Bough, K.J., Yao, S.G., and Eagles, D.A. (2000). Symposium for Dietary Treatments of Epilepsy
Higher ketogenic diet ratios confer protection and Other Neurological Disorders, (Matthews
from seizures without neurotoxicity. Epilepsy Res Friends, pg 47, Liverpool, England).
38, 15–25. Hallbook, T., Lundgren, J., and Rosen, I. (2007).
Cahill, G.F., Jr. (1970). Starvation in man. N Engl J Ketogenic diet improves sleep quality in chil-
Med 282, 668–675. dren with therapy-resistant epilepsy. Epilepsia
Cahill, G.F., Jr., and Owen, O.E. (1970). Body fuels and 48, 59–65.
starvation. Int Psychiatry Clin 7, 25–36. Horvath, R., Gorman, G., and Chinnery, P.F. (2008).
Chee, C., Rice, C., Fenton, C., Groveman, S., and How can we treat mitochondrial encephalomyop-
Bergqvist, A.G.C. (2014). School personnel athies? Approaches to therapy. Neurotherapeutics
understanding of ketogenic diet. In 4th Global 5, 558–568.
Symposium for Dietary Treatments of Epilepsy Kang, H.C., Lee, Y.M., Kim, H.D., Lee, J.S., and Slama,
and other Neurological Disorders (Matthews A. (2007). Safe and effective use of the ketogenic
Friends, pg 57, Liverpool, England). diet in children with epilepsy and mitochon-
Cobo, N.H., Sankar, R., Murata, K.K., Sewak, S.L., drial respiratory chain complex defects. Epilepsia
Kezele, M.A., and Matsumoto, J.H. (2015). The 48, 82–88.
ketogenic diet as broad-spectrum treatment for Kessler, S.K., Gallagher, P.R., Shellhaas, R.A., Clancy,
super-refractory pediatric status epilepticus: chal- R.R., and Bergqvist, A.G. (2011). Early EEG
lenges in implementation in the pediatric and improvement after ketogenic diet initiation.
neonatal intensive care units. J Child Neurol 30, Epilepsy Res 94, 94–101.
259–266. Kim, D.W., Kang, H.C., Park, J.C., and Kim, H.D.
Di Pisa, V., Cecconi, I., Gentile, V., Di Pietro, E., (2004). Benefits of the nonfasting ketogenic diet
Marchiani, V., Verrotti, A., and Franzoni, E. (2012). compared with the initial fasting ketogenic diet.
Case report of pyruvate dehydrogenase deficiency Pediatrics 114, 1627–1630.
with unusual increase of fats during ketogenic diet Kossoff, E.H., Pyzik, P.L., McGrogan, J.R., Vining,
treatment. J Child Neurol 27, 1593–1596. E.P., and Freeman, J.M. (2002). Efficacy of the
Ebus, S.C., Lambrechts, D.A., Herraets, I.J., Majoie, ketogenic diet for infantile spasms. Pediatrics 109,
M.J., de Louw, A.J., Boon, P.J., Aldenkamp, A.P., 780–783.
and Arends, J.B. (2014). Can an early 24-hour Kossoff, E.H., Krauss, G.L., McGrogan, J.R., and
EEG predict the response to the ketogenic diet? Freeman, J.M. (2003). Efficacy of the Atkins diet
A prospective study in 34 children and adults with as therapy for intractable epilepsy. Neurology 61,
refractory epilepsy treated with the ketogenic diet. 1789–1791.
Seizure 23, 468–474. Kossoff, E.H., McGrogan, J.R., and Freeman, J.M.
El-Rashidy, O.F., Nassar, M.F., Abdel-Hamid, I.A., (2004). Benefits of an all-liquid ketogenic diet.
Shatla, R.H., Abdel-Hamid, M.H., Gabr, S.S., Epilepsia 45, 1163.
Mohamed, S.G., El-Sayed, W.S., and Shaaban, S.Y. Kossoff, E.H., and McGrogan, J.R. (2005). Worldwide
(2013). Modified Atkins diet vs classic ketogenic use of the ketogenic diet. Epilepsia 46, 280–289.
formula in intractable epilepsy. Acta Neurol Scand Kossoff, E.H., Turner, Z., and Bergey, G.K. (2007).
128, 402–408. Home-guided use of the ketogenic diet in a
Fenton, C., Groveman, S., Chee, C., Rice, C., and patient for more than 20 years. Pediatr Neurol 36,
Bergqvist, A.G.C. (2014). Establishing a keto- 424–425.
genic teaching kitchen in the acute care setting. Kossoff, E.H., Laux, L.C., Blackford, R., Morrison, P.F.,
In 4th Global Symposium for Dietary Treatments Pyzik, P.L., Hamdy, R.M., Turner, Z., and Nordli,
of Epilepsy and Other Neurological Disorders, D.R., Jr. (2008). When do seizures usually improve
(Matthews Friends, pg 47, Liverpool, England). with the ketogenic diet? Epilepsia 49, 329–333.
Freeman, J.M., Kossoff, E., and Freeman, M.T.K. (2007). Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
The Ketogenic Diet: A Treatment for Children and Ballaban-Gil, K.R., Christina Bergqvist, A.G.,
Others with Epilepsy (New York: Demos Medical Blackford, R., Buchhalter, J.R., Caraballo, R.H.,
Publishing). Helen Cross, J., Dahlin, M.G., et  al. (2009).
33

Chapter 4: How Do You Implement the Diet? 33

Optimal clinical management of children receiv- Efficacy of 4:1 (classic) versus 2.5:1 ketogenic ratio
ing the ketogenic diet:  recommendations of diets in refractory epilepsy in young children:  a
the International Ketogenic Diet Study Group. randomized open labeled study. Epilepsy Res 96,
Epilepsia 50, 304–317. 96–100.
Kossoff, E.H., Bosarge, J.L., Miranda, M.J., Wiemer- Rizzutti, S., Ramos, A.M., Muszkat, M., and Gabbai,
Kruel, A., Kang, H.C., and Kim, H.D. (2010). Will A.A. (2007). Is hospitalization really necessary
seizure control improve by switching from the during the introduction of the ketogenic diet? J
modified Atkins diet to the traditional ketogenic Child Neurol 22, 33–37.
diet? Epilepsia 51, 2496–2499. Sachdeo, R. (2007). Monotherapy clinical trial design.
Kossoff, E.H., Dorward, J.L., Turner, Z., and Pyzik, P.L. Neurology 69, S23–27.
(2011a). Prospective study of the modified Atkins Schmidt, D., Friedman, D., and Dichter, M.A. (2014).
diet in combination with a ketogenic liquid sup- Anti-epileptogenic clinical trial designs in epi-
plement during the initial month. J Child Neurol lepsy:  issues and options. Neurotherapeutics 11,
26, 147–151. 401–411.
Kossoff, E.H., Freeman, J.M., Turner, Z., and Rubenstein, Seo, J.H., Lee, Y.M., Lee, J.S., Kang, H.C., and Kim,
J.E. (2011b). Ketogenic Diets:  Treatments for H.D. (2007). Efficacy and tolerability of the keto-
Epilepsy and Other Disorders, 5th edition genic diet according to lipid:nonlipid ratios: com-
(New York: Demos Medical Publishing). parison of 3:1 with 4:1 diet. Epilepsia 48, 801–805.
Levy, R.G., Cooper, P.N., and Giri, P. (2012). Ketogenic Snyder, D. (2006). Keto kids (New  York:  Demos
diet and other dietary treatments for epilepsy. Medical Publishing).
Cochrane Database Syst Rev 3, CD001903. Spulber, G., Spulber, S., Hagenas, L., Amark, P.,
Livingston, S. (1951). The ketogenic diet in the treat- and Dahlin, M. (2009). Growth dependence on
ment of epilepsy in children. Postgrad Med 10, insulin-like growth factor-1 during the ketogenic
333–336. diet. Epilepsia 50, 297–303.
Lutas, A., and Yellen, G. (2013). The ketogenic Stafstrom, C.E., Zupec-Kania, B., and Rho, J.M. (2008).
diet: metabolic influences on brain excitability and Ketogenic diet and treatments:  introduction/per-
epilepsy. Trends Neurosci 36, 32–40. spectives. Epilepsia 49(Suppl 8), 1–2.
MacCracken, K.A., and Scalisi, J.C. (1999). Tagliabue, A., Bertoli, S., Trentani, C., Borrelli, P., and
Development and evaluation of a ketogenic diet Veggiotti, P. (2012). Effects of the ketogenic diet
program. J Am Diet Assoc 99, 1554–1558. on nutritional status, resting energy expenditure,
Martin, K., Jackson, C.F., Levy, R.G., Cooper, P.N. and substrate oxidation in patients with medically
(2016). Ketogenic diet and other dietary treat- refractory epilepsy: a 6-month prospective obser-
ments for epilepsy. Cochrane Database Syst Rev 4, vational study. Clin Nutr 31, 246–249.
CD001903 Taub, K.S., Kessler, S.K., and Bergqvist, A.G. (2014).
Miranda, M.J., Turner, Z., and Magrath, G. (2012). Risk of seizure recurrence after achieving initial
Alternative diets to the classical ketogenic diet: can seizure freedom on the ketogenic diet. Epilepsia
we be more liberal? Epilepsy Res 100, 278–285. 55, 579–583.
Nation, J., Humphrey, M., MacKay, M., and Boneh, A. Vaisleib, I.I., Buchhalter, J.R., and Zupanc, M.L.
(2014). Linear growth of children on a ketogenic (2004). Ketogenic diet: outpatient initiation, with-
diet:  does the protein-to-energy ratio matter? J out fluid, or caloric restrictions. Pediatr Neurol
Child Neurol 29, 1496–1501. 31, 198–202.
Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S., Vining, E.P., Pyzik, P., McGrogan, J., Hladky, H.,
Edwards, N., Fitzsimmons, G., Whitney, A., and Anand, A., Kriegler, S., and Freeman, J.M. (2002).
Cross, J.H. (2008). The ketogenic diet for the treat- Growth of children on the ketogenic diet. Dev
ment of childhood epilepsy:  a randomised con- Med Child Neurol 44, 796–802.
trolled trial. Lancet Neurol 7, 500–506. Weber, T.A., Antognetti, M.R., and Stacpoole, P.W.
Nylen, K., Likhodii, S., Abdelmalik, P.A., Clarke, J., (2001). Caveats when considering ketogenic diets
and Burnham, W.M. (2005). A comparison of the for the treatment of pyruvate dehydrogenase com-
ability of a 4:1 ketogenic diet and a 6.3:1 ketogenic plex deficiency. J Pediatr 138, 390–395.
diet to elevate seizure thresholds in adult and Wheless, J.W. (2008). History of the ketogenic diet.
young rats. Epilepsia 46, 1198–1204. Epilepsia 49(Suppl 8), 3–5.
Pfeifer, H.H., and Thiele, E.A. (2005). Low-glycemic- Wijburg, F.A., Barth, P.G., Bindoff, L.A., Birch-Machin,
index treatment:  a liberalized ketogenic diet for M.A., van der Blij, J.F., Ruitenbeek, W., Turnbull,
treatment of intractable epilepsy. Neurology 65, D.M., and Schutgens, R.B. (1992). Leigh syndrome
1810–1812. associated with a deficiency of the pyruvate dehy-
Raju, K.N., Gulati, S., Kabra, M., Agarwala, A., drogenase complex:  results of treatment with a
Sharma, S., Pandey, R.M., and Kalra, V. (2011). ketogenic diet. Neuropediatrics 23, 147–152.
34

34 section I: Ketogenic Diet for Epilepsy in the Clinic

Wilder, R.M. (1921). The effect of ketonemia on necessary when initiating the ketogenic diet? J
the course of epilepsy. Mayo Clin Bulletin 2, Child Neurol 17, 179–182.
307–308. Wong, J., and Wirrell, E. (2006). Physical activity in chil-
Williams, S., Basualdo-Hammond, C., Curtis, R., and dren/teens with epilepsy compared with that in their
Schuller, R. (2002). Growth retardation in children siblings without epilepsy. Epilepsia 47, 631–639.
with epilepsy on the ketogenic diet: a retrospective Zupec-Kania, B., and Zupanc, M.L. (2008). Long-term
chart review. J Am Diet Assoc 102, 405–407. management of the ketogenic diet: seizure moni-
Wirrell, E.C., Darwish, H.Z., Williams-Dyjur, C., toring, nutrition, and supplementation. Epilepsia
Blackman, M., and Lange, V. (2002). Is a fast 49(Suppl 8), 23–26.
35

5
Glut1 Deficiency and the Ketogenic Diets
JOERG KLEPPER, MD, PHD

GLUT1 DEFICIENCY associated with missense mutations are often pres-


In the fed state the human brain relies entirely ent in extended phenotypes and indicate that a
on glucose for energy metabolism. Glucose entry SLC2A1 analysis should be performed along with
into the brain is exclusively mediated by the facili- a diagnostic lumbar puncture (Weber et al., 2008).
tated glucose transporter protein Glut1. Impaired In approximately 85%–90% of cases the condition
glucose transport into the brain resulting from can be confirmed by heterozygous mutations in
Glut1 deficiency (Glut1D, OMIM 606777)  (De the SLC2A1 gene. Of note, the absence of SLC2A1
Vivo et al., 1991) will cause a cerebral “energy cri- mutations does not exclude the diagnosis (Klepper,
sis,” particularly as the developing brain requires 2013). Inheritance is predominantly autosomal
3–4 times more energy than the adult brain. dominant and mutations are mostly de novo, but
Clinical features are global developmental delay, individual cases of autosomal recessive trans-
early-onset epilepsy, and a complex movement mission have been described (Brockmann et  al.,
order (Klepper and Leiendecker, 2007). As such, 2001; Klepper et  al., 2009; Klepper et  al., 2001).
the disease mechanism of Glut1D is intriguingly Prenatal testing is available if the disease-causing
straightforward: mutation has been identified. Neuroimaging is
generally uninformative, but PET scanning has
1. The Glut1 defect results in low CSF been reported to show a distinctive metabolic
glucose concentrations, termed footprint in brain (Pascual et  al., 2002). Further
hypoglycorrhachia. diagnostic tools such as glucose uptake assay into
2. Hypoglycorrhachia results in an epileptic erythrocytes are available on a research basis only
encephalopathy and movement disorder. (Klepper et al., 1999).
3. Glut1D is treatable by means of a ketogenic
diet providing ketones as an alternative fuel C L I N I C A L P R E S E N TAT I O N
for brain energy metabolism (Figure 5.1). Classic Glut1D presents with unspecific develop-
mental delay associated with early-onset epilepsy,
DIAGNO SIS a complex movement disorder, and paroxysmal
The diagnosis of Glut1D rests on (1) low glucose nonepileptic events. Any combinations of these
concentrations in CSF, termed hypoglycorrha- features are possible (Figure 5.2) (Pearson et al.,
chia, determined via fasting lumbar puncture, and 2013). Severe cases develop secondary micro-
(2) mutations in the SLC2A1 gene (De Giorgis and cephaly, reflecting the impairment of the devel-
Veggiotti, 2013). The lumbar puncture should be oping brain. Seizure type may vary from cyanotic
performed in a metabolic steady-state, for exam- attacks and staring spells in early infancy to
ple, following a 4- to 6-hour fast. In Glut1D, CSF childhood absence epilepsy and myoclonic/gen-
glucose concentrations usually are below 40 mg/ eralized grand-mal seizures in later childhood.
dL. A  CSF/plasma glucose ratio below 0.45 is Certain epilepsy syndromes such as early-onset
helpful to determine hypoglycorrhachia, and CSF absence epilepsy (onset < age 4 years) and myo-
lactate should always be low to normal (for age- clonic astatic epilepsy (Doose syndrome) are
dependent reference values of CSF glucose and associated with Glut1D (Mullen et al., 2010; Suls
lactate, see Leen et al., 2013b). Patients with milder et al., 2008). The complex movement disorder in
phenotypes do have CSF glucose values above 41 Glut1D features ataxic-spastic and gait abnormal-
mg/dL but never normal (De Vivo and Wang, ities, action limb dystonia, mild chorea, cerebellar
2008). Mild or borderline hypoglycorrhachia action tremor, myoclonus, and dyspraxia (Pons
36

36 section I: Ketogenic Diet for Epilepsy in the Clinic

Glucose
KDT
1 BBB

Fat

Brain β-Ox.

Acetyl-CoA Liver
2

Ketones

TCA cycle

Energy

FIGURE 5.1 The metabolic concept of Glut1 Deficiency.


➊: the defect of the Glut1 transporter at the blood-brain barrier results in cerebral energy failure
➋: Ketones derived from either body fat (fasting) or nutritional fat (KDT) serve as an alternative fuel to restore cerebral energy.

et al., 2010). In adults, exertion-induced dystonia, K E TO G E N I C D I E T


and stomatin-associated cryohydrocytosis (Flatt THERAPIES IN GLUT1D
et  al., 2011; Weber et  al., 2008), a rare form of Currently ketogenic diet therapy (KDT) is the
hemolytic anemia, have been identified as allelic only treatment for this disorder of brain energy
diseases associated with SLC2A1-mutations. metabolism. The metabolic concept is to mimic

Epilepsy Movement
Infantile seizures Spasticity disorder
Ataxia
Absence seizures Dystonia
Chorea

Cognitive impairment
Classic phenotype Delayed adaptive behavior
Developmental Variable attention
encephalopathy

Cognitive/behavioral
disturbances
FIGURE 5.2 The complex phenotype of Glut1 deficiency (with permission from Pearson et al., 2013).
37

Chapter 5: Glut1 Deficiency and the Ketogenic Diets 37

the metabolic state of fasting and thus “refuel” the in ECG, selenium deficiency, renal stones, ele-
brain using ketones from the KDT (Figure 5.1). vated serum lipids, and atherosclerosis are serious
Anticonvulsant mechanisms described for KDT concerns in Glut1D—long-term data is only just
in intractable childhood epilepsy (Masino and emerging to assess these questions. Emerging tools
Rho, 2012) also apply in Glut1D and contribute to for follow-up screening are dexa-scans for bone
seizure control. In addition, KDTs provide unspe- density (Bergqvist et al., 2008) and ultrasound of
cific positive effects such as improved physical the carotid arteries for atherosclerosis (Coppola
endurance, improved attention span, and alert- et al., 2014; Kapetanakis et al., 2014). On follow-up
ness (Lambrechts et  al., 2012; Ramm-Pettersen into puberty, recommendations to maintain KDT
et al., 2014) and have a substantial positive impact in Glut1D throughout childhood and adolescence
on the associated movement disorder (Leen et al., often are particularly difficult. Achieving compli-
2013a). ance after years on KDT often is the most impor-
However, KDTs for Glut1D differ from KDTs tant practical problem in this age group.
for intractable childhood epilepsy in the following Novel compounds such as ketone-esters are
aspects: currently discussed to supplement or even replace
KDT in childhood epilepsy and metabolic disease.
• Ketones in Glut1D provide an alternative Tripheptanoin-based metabolic therapy and the
fuel for the brain, restoring the cerebral concept of anaplerosis are discussed elsewhere in
energy deficit. The higher the ketosis, this book (see ; Borges, chapter 34). Clinical trials
the more energy is provided for the and experimental data in Glut1-deficient mice may
developing brain. Within this concept the offer novel approaches toward Glut1D therapy.
KDT providing the highest ketosis is best
for the patient. OPEN QUESTIONS
• Introduction of KDT should be as early Clinical symptoms of Glut1D appear to be age-
as possible, even in infancy, to meet the dependent. Epilepsy is the prominent clinical
energy demands of the developing brain. feature in infants and children, but it often stabi-
• KDT in Glut1D should be continued lizes whereas movement disorders and paroxys-
throughout childhood into adolescence mal events increase with age and seem to be the
to provide ketones as an alternative main clinical problem in adolescence and adult-
fuel for adequate brain development. hood (Alter et al., 2015). Here the impact of KDT
Discontinuation of KDT after is less clear, although response to classic KDT
approximately 2 years, as discussed in and MAD have been described for Glut1D. In a
intractable childhood epilepsy, is not small subgroup of patients with confirmed Glut1D
recommended in Glut1D patients. symptoms do not respond adequately to KDT. For
reasons yet unclear, epilepsy remains uncontrolled
The practical approach to KDT in Glut1D and movement disorders, in particular paroxysmal
does not differ substantially from KDT for intrac- events, increase despite KDT.
table childhood epilepsy (Klepper, 2012). Clinical The use of KDT in adult Glut1D seems reasonable,
management such as initiation, supplementa- but is by no means an established recommendation—
tion, and maintenance of KDT follow established patients find it very difficult, symptoms such as epi-
guidelines (Kossoff et  al., 2009). In theory, any lepsy seem to stabilize in adults, and there is simply
KDT providing measurable ketosis may be used not enough data on the benefit and long-term adverse
in Glut1D according to age, clinical presenta- effects of KDT in this age group (Leen et al., 2014).
tion, and compliance. In practice, long-chain and The use of KDT in Glut1D allelic diseases such as
medium-chain KDT and the MAD are established paroxysmal exertion-induced dystonia, early-onset
therapies for Glut1D, whereas the low glycemic absence epilepsy, or stomatin-deficient cryohydro-
index treatment is not. cytosis also remains unclear—originally considered
Certain aspects are special to KDT in Glut1D. an exclusively pediatric disorder, Glut1D has now
In contrast to the efficacy of KDT in the treat- moved into the focus of adult neurology.
ment of childhood epilepsy the vast majority of
patients with Glut1D show an immediate and A B B R E V I AT I O N S
continuing response. This is even considered a
diagnostic tool:  if KDT is ineffective, reconsider Glut1D Glut1 Deficiency
the diagnosis. Long-term adverse effects of KDT PED Paroxysmal exercise-induced
such as growth retardation, prolonged QT interval dystonia
38

38 section I: Ketogenic Diet for Epilepsy in the Clinic

CSF cerebrospinal fluid uptake assay for diagnosis of glucose-transporter-


KDT ketogenic diet therapy protein syndrome. J Clin Lab Anal 13, 116–121.
MAD modified Atkins diet Klepper, J., Willemsen, M., Verrips, A., Guertsen, E.,
Herrmann, R., Kutzick, C., Florcken, A., and Voit,
T. (2001). Autosomal dominant transmission of
CONFLICTS OF INTEREST GLUT1 deficiency. Hum Mol Genet 10, 63–68.
The author has received speaker honoraria and Klepper, J., and Leiendecker, B. (2007). GLUT1 defi-
ciency syndrome -2007 update. Dev Med Child
travel funds from Nutricia GmbH, SHS, Heilbronn,
Neurol 49, 707–716.
Germany, and has received project support from Klepper, J., Scheffer, H., Elsaid, M.F., Kamsteeg,
the parents support group Glut1-Förderverein, E.J., Leferink, M., and Ben-Omran, T. (2009).
Bremen, Germany. Autosomal recessive inheritance of GLUT1 defi-
ciency syndrome. Neuropediatrics 40, 207–210.
REFERENCES Klepper, J. (2012). Glut1 deficiency syndrome in clini-
Alter, A.S., Engelstad, K., Hinton, V.J., Montes, J., cal practice. Epilepsy Res 100, 272–277.
Pearson, T.S., Akman, C.I., and De Vivo, D.C. Klepper, J. (2013). Absence of SLC2A1 mutations
(2015). Long-term clinical course of Glut1 defi- does not exclude Glut1 deficiency syndrome.
ciency syndrome. J Child Neurol 30, 160–169. Neuropediatrics 44, 235–236.
Bergqvist, A.G., Schall, J.I., Stallings, V.A., and Zemel, Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
B.S. (2008). Progressive bone mineral content loss Ballaban-Gil, K.R., Christina Bergqvist, A.G.,
in children with intractable epilepsy treated with Blackford, R., Buchhalter, J.R., Caraballo, R.H.,
the ketogenic diet. Am J Clin Nutr 88, 1678–1684. Helen Cross, J., Dahlin, M.G., et al. (2009). Optimal
Brockmann, K., Wang, D., Korenke, C.G., von Moers, clinical management of children receiving the keto-
A., Ho, Y.Y., Pascual, J.M., Kuang, K., Yang, H., Ma, genic diet:  recommendations of the International
L., Kranz-Eble, P., et al. (2001). Autosomal domi- Ketogenic Diet Study Group. Epilepsia 50, 304–317.
nant Glut-1 deficiency syndrome and familial epi- Lambrechts, D.A., Bovens, M.J., de la Parra, N.M.,
lepsy. Ann Neurol 50, 476–485. Hendriksen, J.G., Aldenkamp, A.P., and Majoie,
Coppola, G., Natale, F., Torino, A., Capasso, R., M.J. (2012). Ketogenic diet effects on cognition,
D’Aniello, A., Pironti, E., Santoro, E., Calabrò, R., mood, and psychosocial adjustment in children.
and Verrotti, A. (2014). The impact of the keto- Acta Neurol Scand. 127(2), 103–108.
genic diet on arterial morphology and endothelial Leen, W.G., Mewasingh, L., Verbeek, M.M., Kamsteeg,
function in children and young adults with epi- E.J., van de Warrenburg, B.P., and Willemsen,
lepsy: a case-control study. Seizure 23, 260–265. M.A. (2013a). Movement disorders in GLUT1
De Giorgis, V., and Veggiotti, P. (2013). Glut1 defi- deficiency syndrome respond to the modified
ciency syndrome 2013:  current state of the art. Atkins diet. Mov Disord 28, 1439–1442.
Seizure 22, 803–811. Leen, W.G., Wevers, R.A., Kamsteeg, E.J., Scheffer, H.,
De Vivo, D.C., Trifiletti, R.R., Jacobson, R.I., Ronen, Verbeek, M.M., and Willemsen, M.A. (2013b).
G.M., Behmand, R.A., and Harik, S.I. (1991). Cerebrospinal fluid analysis in the workup of
Defective glucose transport across the blood-brain GLUT1 deficiency syndrome: a systematic review.
barrier as a cause of persistent hypoglycorrhachia, JAMA Neurol 70, 1440–1444.
seizures, and developmental delay. N Engl J Med Leen, W.G., Taher, M., Verbeek, M.M., Kamsteeg, E.J.,
325, 703–709. van de Warrenburg, B.P., and Willemsen, M.A.
De Vivo, D.C., and Wang, D. (2008). Glut1 defi- (2014). GLUT1 deficiency syndrome into adult-
ciency:  CSF glucose; How low is too low? Rev hood: a follow-up study. J Neurol 261, 589–599.
Neurol (Paris) 164, 877–880. Masino, S.A., and Rho, J.M. (2012). Mechanisms of
Flatt, J.F., Guizouarn, H., Burton, N.M., Borgese, Ketogenic Diet Action. In Noebels, J.L., Avoli, M.,
F., Tomlinson, R.J., Forsyth, R.J., Baldwin, S.A., Rogawski, M.A., Olsen, R.W., Delgado-Escueta, A.V.,
Levinson, B.E., Quittet, P., Aguilar-Martinez, P., editors. Jasper's Basic Mechanisms of the Epilepsies
et al. (2011). Stomatin-deficient cryohydrocytosis [Internet]. 4th edition. Bethesda (MD):  National
results from mutations in SLC2A1: a novel form of Center for Biotechnology Information (US).
Glut1 deficiency syndrome. Blood 118, 5267–5277. Mullen, S.A., Suls, A., De Jonghe, P., Berkovic, S.F.,
Kapetanakis, M., Liuba, P., Odermarsky, M., Lundgren, and Scheffer, I.E. (2010). Absence epilepsies with
J., and Hallböök, T. (2014). Effects of ketogenic widely variable onset are a key feature of familial
diet on vascular function. Eur J Paediatr Neurol Glut1 deficiency. Neurology 75, 432–440.
18, 489–494. Pascual, J.M., Van Heertum, R.L., Wang, D., Engelstad,
Klepper, J., Garcia-Alvarez, M., O’Driscoll, K.R., K., and De Vivo, D.C. (2002). Imaging the meta-
Parides, M.K., Wang, D., Ho, Y.Y., and De Vivo, bolic footprint of Glut1 deficiency on the brain.
D.C. (1999). Erythrocyte 3-O-methyl-D-glucose Ann Neurol 52, 458–464.
39

Chapter 5: Glut1 Deficiency and the Ketogenic Diets 39

Pearson, T.S., Akman, C., Hinton, V.J., Engelstad, K., Suls, A., Dedeken, P., Goffin, K., Van Esch, H.,
and De Vivo, D.C. (2013). Phenotypic spectrum Dupont, P., Cassiman, D., Kempfle, J., Wuttke,
of glucose transporter type 1 deficiency syndrome T.V., Weber, Y., Lerche, H., et  al. (2008).
(Glut1 DS). Curr Neurol Neurosci Rep 13, 342. Paroxysmal exercise-induced dyskinesia and
Pons, R., Collins, A., Rotstein, M., Engelstad, K., and epilepsy is due to mutations in SLC2A1, encod-
De Vivo, D.C. (2010). The spectrum of movement ing the glucose transporter Glut1. Brain 131,
disorders in Glut-1 deficiency. Mov Disord 25, 183–144.
275–281. Weber, Y.G., Storch, A., Wuttke, T.V., Brockmann, K.,
Ramm-Pettersen, A., Stabell, K.E., Nakken, K.O., and Kempfle, J., Maljevic, S., Margari, L., Kamm, C.,
Selmer, K.K. (2014). Does ketogenic diet improve Schneider, S.A., Huber, S.M., et al. (2008). Glut1
cognitive function in patients with GLUT1-DS? mutations are a cause of paroxysmal exertion-
A 6- to 17-month follow-up study. Epilepsy Behav induced dyskinesias and induce hemolytic ane-
39, 111–115. mia by a cation leak. J Clin Invest 118, 2157–2168.
40

6
Ketogenic Diet in Established Epilepsy Indications
A N N M . B E R G I N, M B , S C M , M R C P ( U K )

INTRODUCTION brain injuries of hypoxic, traumatic, infectious,


Arising from biblical anecdote and early 20th- hemorrhagic origin), developmental brain mal-
century explorations into the effect of fasting for formations, genetic/metabolic disorders causing
seizure management, the ketogenic diet (KD) epilepsy as their primary problem, or resulting in
developed alongside medical knowledge of the symptomatic epilepsy, primary and secondary epi-
biochemistry of fasting/starvation and ketosis leptic encephalopathies and progressive/degenera-
(Wheless, 2014). Falling into relative disuse with tive epilepsies. In this era of molecular specificity,
the arrival of phenytoin and subsequent anticon- it seems counterintuitive to dwell on such a het-
vulsant medications, it continued to be used mainly erogeneous group. And indeed, increasing surgi-
in pediatrics, and primarily under the auspices of cal prowess and advancing ability to define specific
Dr. John Freeman and colleagues at Johns Hopkins genetic etiologies and offer individualized molecu-
University. After the development of waves of new larly based therapies will continue to “chip away”
anticonvulsant medications that were associated at this broad group. However it is in precisely such
with reduction in toxicity but not with greater effi- a heterogeneous group that the ketogenic diet’s
cacy in patients with refractory epilepsy, interest in efficacy was first described. And, in epilepsy prac-
the KD reemerged. Over the years, multiple case tice, patients presenting with refractory epilepsy
series, describing cases achieving remarkable and constitute exactly this mixed group of patients.
sustained efficacy in a substantial proportion of In treatment of epilepsy, serially trialed anti-
children with the most intractable epilepsies (>400 epileptic drugs (AEDs), after initial failure, are
seizures/month) had been published from the known to have diminishing likelihood of achiev-
Johns Hopkins group (Hemingway et  al., 2001). ing seizure freedom, with only 14% achieving
With the renewed interest, a multicenter group seizure freedom with a second or third agent
showed that other centers using the same protocol and only 3% seizure-free with a combination of
could achieve similar results (Vining et al., 1998), two drugs (Kwan and Brodie, 2000). Alternative
and finally a randomized controlled study was approaches, such as surgery for patients with
conducted in London, again supporting the effi- amenable lesions, and neurostimulation and/or
cacy of this treatment in children with refractory diet manipulation for others, are available in this
epilepsies (Neal et al., 2008). This chapter reviews setting. “Drug resistant epilepsy may be defined
diet treatment in epilepsies where its use is well as failure of adequate trials of two tolerated and
established—refractory nonsurgical epilepsies and appropriately chosen and used AED schedules
epileptic encephalopathies:  Lennox Gastaut syn- (whether as monotherapies or in combination) to
drome, infantile spasms, myoclonic astatic epi- achieve sustained seizure freedom” (Kwan et  al.,
lepsy (Doose syndrome), and severe myoclonic 2010). Once epilepsy is identified as refractory,
epilepsy of infancy (Dravet syndrome). nonpharmacologic approaches are appropriately
considered. Among the options, resective epilepsy
R E F R A C T O RY surgery offers the possibility of “cure” of epilepsy,
NONSURGICAL EPILEPSY in certain cases the best chance of seizure-free,
This broad group, defined by nonsuitability for and even medication-free survival. Consideration
a potentially curative surgery, includes lesional of surgical candidacy is therefore important at this
and nonlesional, focal, multifocal, and secondary point. Reasons for ineligibility for epilepsy surgery
generalized epilepsies. Etiologies include remote can be manifold—inability to lateralize or localize
symptomatic injuries (pre-, peri- and postnatal seizure focus, presence of multiple foci, high risk
41

Chapter 6: Ketogenic Diet in Established Epilepsy Indications 41

of injury to eloquent cortex, patient/family prefer- from around the world in similar case series have
ence. In this setting, as well as in those for whom reported roughly similar efficacy, which can be
epilepsy surgery has already failed, dietary manip- broadly stated as ~50% of refractory epilepsy
ulation is an important treatment option. patients starting the diet can expect to be respond-
For many years, evidence of diet efficacy was ers (i.e. have >50% seizure reduction in seizures),
in the realm of “expert opinion,” within a nar- 25%–30% will experience >90% seizure reduc-
row community of experts familiar with the tion, of whom approximately half will be seizure-
diet. This took the form of retrospective reviews free (Henderson et  al., 2006; Nathan et  al., 2009;
(Kinsman, 1992; Prasad et al., 1996; Hemingway, Hallbook et  al., 2015; Kossoff and McGrogan,
2001) describing diet experience in 58 of the most 2005; Kossoff et al., 2012).
severely affected patients over an 18- to 24-month Diet therapy in epilepsy is rigorous, requiring
follow-up period. Studies of this type included eti- precision in preparation and careful attention in
ologies and seizures of all kinds in children of a administration by caregivers. Formula-fed infants
broad range of ages, often with very high seizure and tube-fed infants are easiest to initiate and
frequency. Reported efficacy was surprisingly maintain on the diet, and usually use the classic
good, comparable to industry standards for effi- ketogenic diet at a ratio sufficient to induce keto-
cacy of new drugs today. Retrospective nature and sis with plasma betahydroxybutyrate ≥ 4 mmol/L
lack of blinding and control groups limited the or seizure freedom, whichever comes first. These
interpretability of these studies. Following media children are not troubled by dietary restriction and
exposure in 1994 in a Dateline NBC review of the poor palatability as older, orally fed children are,
diet and in 1997 after release of the film First Do who are are accustomed to making dietary choices
No Harm, there was a tidal wave of interest in the and may refuse unpalatable and unpreferred foods
diet among parents of children with refractory and drink. Reduced GI motility may worsen
epilepsy, pressuring their providers to provide, esophageal reflux and constipation in all, result-
or refer them for a trial of, diet treatment. In this ing in discomfort. Diet therapy is not without a
setting the first prospective studies emerged—a variety of systemic adverse effects occurring at low
multicenter study (Vining et al., 1998) and a single frequency (Kang et al., 2004; Kossoff et al., 2009)
institution report (Freeman et  al., 1998), again but at least comparable in range and significance
describing diet efficacy in patients with an aver- to those associated with anticonvulsant medica-
age monthly seizure frequency of 230/month and tions, although a significant advantage is the lack
410/month respectively. The multicenter study in of sedating and/or behavioral adverse effects.
particular assured the community of pediatric epi- It would be extremely helpful to be able to
leptologists that the efficacy of the Johns Hopkins predict which patients are most likely to respond
protocols could be reproduced in other hands. At to diet therapy in advance, instead of having to
this point, in 2000, a review of 11 reports on diet anticipate ~50% failure to achieve a useful seizure
efficacy, including the two cited prospective stud- reduction. Unfortunately, though many studies
ies, cosponsored by the BlueCross and Blue Shield and reports attempt to identify predictors, small
association, stated, “it is unlikely that this degree of study size, lack of control groups, heterogeneous
benefit can result from a placebo response and/or diagnostic groups, and rarity of specific epilepsy
spontaneous remission” and therefore concluded, syndromes have all limited the ability to reliably
“the evidence is sufficient to determine that the identify these factors. Schoeler et al. recently pub-
ketogenic diet is efficacious in reducing seizure lished an extensive review of the articles provid-
frequency in children with refractory epilepsy” ing data on effectiveness and putative predictive
(Lefevre and Aronson, 2000). Finally, in 2008, factors (Schoeler et al., 2013). Twenty-one factors
a randomized, controlled but unblinded study were examined. For each factor, the predomi-
of diet in a mixed group of children with refrac- nance of evidence in favor of an effect, the relative
tory epilepsy revealed a statistically significant strength of evidence against an effect of that factor,
improvement in seizure control in a group of chil- and the number of patients in the cohorts “for” and
dren treated with ketogenic diet now, compared “against” that factor were considered. No factor had
with a matched group treated with ketogenic diet strong evidence for a positive or negative response
3 months later, improving the quality of evidence to diet. Strong evidence for absence of effect is also
in support of diet efficacy (Neal et al., 2008). lacking, except in the case of gender and intel-
These studies have the great benefit of “gener- lectual ability, which appear to have no effect on
alizability” to the real world of refractory epilepsy. diet response. There were “mixed” findings (effect
As might therefore be expected, subsequent studies on response is reported in approximately half of
42

42 section I: Ketogenic Diet for Epilepsy in the Clinic

reported cases) for epilepsy cause/syndrome, sei- Many conditions have been recently estab-
zure type, and biochemical markers other than lished for which the ketogenic diet can be particu-
ketosis and plasma glucose. “Weak evidence” for larly beneficial. Several are epilepsy syndromes
an effect on diet response (effect reported in less with refractory generalized seizures. This chap-
than half of reported cases) exists for age of sei- ter covers the most common four conditions, for
zure onset, age at diet initiation, time from seizure which there are sufficient data to recommend the
onset to diet initiation, seizure frequency, diet ketogenic diet as potentially very helpful.
type, ratio, levels of ketosis (betahydroxybutyr-
ate, acetone), EEG parameters, AEDs (individual I N FA N T I L E   S PA S M S
or number/combinations), and BMI. Presence or Infantile spasms (IS) are an age-dependent sei-
absence of an effect of blood glucose, genetics, and zure type, typically occurring between 6 and
imaging findings could not be assessed because of 18  months of age, which are clinically charac-
a limited number of studies reporting and patients terized by serial body jerks occurring in clus-
reported. Assessment of predictive factors in this ters, often at the interfaces of waking and sleep.
way is complicated by interactions between factors They represent the most common type of epi-
that cannot be resolved in small, uncontrolled, leptic spasm. In infancy, they are often, but not
retrospective studies. Hopefully the future will always, associated with a characteristic EEG pat-
include the development of consortia of expert tern known as hypsarrhythmia, a chaotic waking
centers devoted to analysis of greater numbers of pattern of asynchronous high-voltage irregular
patients managed prospectively and in a standard slowing with superimposed multifocal spikes,
manner, which will allow dissection of factors pre- which are more synchronous and discontinu-
dictive of successful treatment. Better understand- ous in sleep. When spasms are associated with
ing may also illuminate potential mechanisms for developmental regression and hypsarrhythmia
further study and manipulation. this triad constitutes West syndrome. Infantile
Patients started on KD may have a seizure spasms can be idiopathic or associated with
response within 14  days, but most will con- genetic disorders, brain malformations, or pre-
tinue a trial of diet for at least 3 months, assum- existing brain injuries. ACTH and vigabatrin are
ing no exacerbation of seizures occurs beyond first-line treatments for this epileptic encepha-
the initial initiation period (Kossoff et  al., 2009). lopathy, which is refractory to initial therapies in
Discontinuation after successful treatment is usu- 30%–40% overall (Pellock et al., 2010; Lux et al.,
ally attempted after 2 years, though there is no data 2005). Refractory IS are associated with serious
determining this to be the optimal time. Rarely, delay in development in most survivors. Early
diet initiation is associated with persistent exacer- and effective treatment of this seizure type is con-
bation of seizures beyond the initiation itself. Diet sidered the best chance for normal developmen-
withdrawal is appropriate in this setting. Early diet tal outcome. With first-line treatment, ACTH
discontinuation (<3 months) is usually due to lack in most cases, elimination of clinical spasms by
of efficacy. Growing intolerance of dietary restric- 14 days of treatment is the marker of successful
tion can be problematic in young patients able to treatment. Resolution of hypsarrhythmia is also
make diet choices. considered a marker of successful treatment of
Barriers to provision of ketogenic diets con- IS and is usually assessed first at 14  days, with
tinue to include access to clinical expertise in evaluation for maintenance of improvement in a
diet treatment, cost of higher grade protein and further 28 days.
high-fat foods in some communities, individual In comparison with other epilepsies, assess-
feeding/dietary preferences, co-morbid medical ment of efficacy of any treatment for infantile
complexity, and systemic fragility raising con- spasms should consider the seizure-free rate to
cern for ability to tolerate the stress of dietary be the first desirable seizure outcome, with EEG
conversion. resolution of underlying hypsarrhythmia of near
In summary, for patients falling into the het- equal importance. Complicating the assessment
erogeneous category of “nonsurgical epilepsy,” of efficacy of any treatment of infantile spasms is
dietary therapy with a ketogenic diet should be the known occurrence of spontaneous remission
considered once drug resistance and ineligibil- of infantile spasms in untreated cases, which
ity for surgery has been established. In general, can occur as early as 1 month after onset, and
patients can expect a better chance of efficacy cumulatively in 10%–15% at 6 months and up to
than with trial of another AED (Kwan and Brodie, 25% of patients at 1 year (Hrachovy et al., 1991).
2000). Notably, in this retrospective cohort of untreated
43

Chapter 6: Ketogenic Diet in Established Epilepsy Indications 43

infants ~90% suffered moderate to severe devel- usefully an association of spasm response and
opmental impairment at follow-up, an average of “other seizure” response in these patients (p = .02)
80 months later. There is also a significant rate (Numis et al., 2011).
of relapse of spasms during treatment with first- Factors favoring diet therapy in IS include
line agents, which therefore should also be con- easily available ketogenic formula and lack of
sidered in assessing dietary treatments for this independent choice of diet intake in infants.
condition. Nonetheless, three of 20 caregivers discontinued
The ketogenic diet is among the treatment the diet; of these one indicated difficulty maintain-
options considered after failure of the first-line ing the protocol (Kayyali et al., 2014). In an Asian
treatments, or if their use is contraindicated for population, 9 of 43 patients discontinued the diet
any reason. In a study of ketogenic diet in infants due to “unacceptable” complications, and 7 due to
by Nordli, 17 of the 32 infants with refractory epi- intolerability. An additional 9 (21%) discontinued
lepsy had infantile spasms. Of the 32 infants, 6 due to insufficient efficacy (Eun et  al., 2006). In
achieved seizure freedom all of whom had infantile contrast, 6/26 (23%) had side effects (Numis et al.,
spasms, (Nordli et al., 2001). Another 6 patients 2011), none serious enough to discontinue the
with spasms had “worthwhile improvement,” and diet. In this group, 5/26 discontinued the diet by
as a group, patients with infantile spasms were ~1 year due to either lack of efficacy or difficulty
more responsive to ketogenic diet than infants maintaining the diet.
with other seizure types in this study. Following In these young patients, concerns regarding
on this finding, 23 children with IS were studied, (Hong et  al., 2010), weight gain and appropri-
of whom 18 remained on diet at 6 months, 13 of ate growth were amenable to adjustment down-
whom had ≥50% reduction in seizures (3 seizure- ward in ratio for increased protein intake. Numis
free), and 13/13 remaining on diet at 12 months et al. described no significant differences between
had ≥50% reduction (3 seizure-free) (Kossoff et responders and nonresponders in terms of weight-
al., 2002). In 2010, 104 infants with IS at the same for-height z scores, although the best responders
institution, treated with KD after exposure to a (>90% seizure reduction) had lower weight-for-
mean of 3.6 AEDs, including steroids or vigaba- height z scores at all points.
trin in >70% were reported (Hong et al., 2010). Among the 104 patients described by Hong
Overall, 64% had a ≥50% reduction in seizures, et  al., 10/18 in whom diet was used as the first-
and 38 achieved at least 6 months spasm free after line treatment became seizure-free, had normal
a median of 2.4 months on the diet. Of these, 30 EEG within 2  months, and were maintained on
(79%) maintained spasm freedom. In a prospec- the diet for 6  months, after which the diet was
tive case study of 20 patients with epileptic spasms, withdrawn without relapse (Hong et  al., 2010).
among 70% and 72% achieving a >50% reduction The same group reported a retrospective case-
in seizures at 3 and 6 months respectively, 3 infants controlled study of diet versus ACTH as initial
achieved and maintained seizure freedom for at therapy for new-onset spasms (Kossoff et  al.,
least 6 months (Kayyali et al., 2014). Improvement 2008). Eight of 13 (62%) patients on diet therapy
in spasms was noted within 1 month of start- became seizure-free at a median of 6.5  days of
ing diet treatment. EEG improved in 12 (60% of treatment, compared with 18/20 (90%) treated
patients) when assessed at 3–6 months, and in with high dose ACTH who were spasm free at a
five of six infants with hypsarrhythmia the pat- median of 4 days (p = .06). One of the 8 relapsed
tern resolved and did not recur up to 12 months (12.5%), versus 6 (33%) of the 18 ACTH respond-
after diet initiation. Pires et al. reported 6/17 ers (p = .23). Also, EEG normalization was more
(35%) seizure-free at 1 month after failing vigaba- likely in the ACTH group (1/11 vs. 9/17, p = .02)
trin and hydrocortisone (Pires et al., 2013). Eleven at 1  month, though the EEG normalized in all
of these 17 patients (65%) were seizure-free at 3 eight responders by 5 months of diet treatment.
months, one after the addition of felbamate. The Adverse effects were significantly less likely in the
addition of felbamate to their regimen brought five diet therapy group (p = .006). There was no dif-
more into the responder (>50% reduction) group. ference in developmental outcome between the
Dressler, in a retrospective, cross-sectional study treatment groups at their last examination at a
comparing early (<1.5 yrs) with late diet initiation, median of 12 months.
included 59 infants with IS in their population of Predicting efficacy of diet has been difficult.
115 infants. Nineteen (32%) were seizure-free at Eun et al. associated underlying etiology with effi-
3 months (Dressler et al., 2015a). Numis et al. had cacy, cryptogenic cases responding at a higher rate
9/26 (32%) seizure-free at 1–3 months. They noted than symptomatic cases, but no other predictor
44

44 section I: Ketogenic Diet for Epilepsy in the Clinic

emerged from their series (Eun et  al., 2006). No multiple times per day. Episodes of SE, often pro-
significant predictive factor of diet efficacy was longed, are common. Lennox Gastaut syndrome
identified in the small diet group in the case con- (LGS) may occur de novo or in about 20% devel-
trol comparison with ACTH. Numis et al. reported ops following West syndrome, and might also
11/18 males and 1/8 females were responders follow other remote symptomatic brain injuries
(p  =  0.04) but cited low sample size and skewed (e.g., hypoxic ischemic encephalopathy). If there
distribution of etiologies as potential biases is no preexisting developmental delay, cognitive
(Numis et al., 2011). This has not been reported in regression occurs and at least moderate intellec-
other studies. tual disability is expected, with virtually all indi-
A final area of interest is the possibility of viduals being dependent in adulthood. The EEG
synergistic effects of combination of KD with is characterized by slow (1.5–2 Hz) spike and
other treatments for IS. A recent study of epilep- wave activity, often multifocal spikes, and bursts
tic encephalopathies (EE) (IS, Lennox Gastaut of 10-Hz fast activity during sleep. Slow spike and
syndrome/other generalized epilepsies, CSWS) wave activity may wane over the years. Treatment
examined the efficacy of KD added to steroid with broad-spectrum anticonvulsant drugs is
therapy when not controlled by the initial ste- the mainstay of treatment but is rarely effective
roid treatment (Ville et al., 2015). Patients were in achieving seizure control. Side effects of leth-
on a mean of 3.5 mg/kg/day of hydrocortisone argy and drowsiness are particularly damaging,
(1–10 mg/kg/day) when starting the diet. Ketosis as these states are associated with increased sei-
was achieved in all but one case with status epi- zures. Nonpharmacologic treatments including
lepticus (SE), and no diet adjustment was made surgical approaches (corpus callosotomy, vagus
to maintain ketonuria. Plasma ketones were not nerve stimulation) and diet therapies offer non-
measured. There were 23 IS cases in the total sedating adjunctive therapies that, if successful,
of 42 with EE. Twenty were steroid-resistant, may allow reduction of medication burden and
three steroid-dependent. Overall, 5/27 (18.5%) a resultant improvement in seizures and quality
steroid-resistant cases and 9/15 (60%) steroid- of life.
dependent cases were considered responders. The early reports of efficacy in the most refrac-
The data presented do not allow specific analysis tory epilepsies typically included patients with
with respect to IS. this disorder, and this subgroup of patients were
In summary, studies of modest quality indi- found to respond to diet with a >50% reduction
cate that KD is a moderately effective therapy for in seizures within 5 days of diet initiation with 36
refractory IS, and is generally safe and tolerable hours of fasting and the development of ketosis
in this young population. As a first-line treat- (Freeman and Vining, 1999). A blinded crossover
ment, it has better efficacy for seizure freedom study of 20 LGS patients, initiated on diet and
over time, but is not as quickly effective as tradi- subsequently, in a blinded manner, given either
tional first-line treatments, in particular ACTH. 60 gm glucose with saccharin to abort diet, or sac-
Patients experiencing improvement do so within charin only (allowing continued ketosis) failed to
1  month, and definitely by 3  months. There may show a significant difference in outcome, but this
be reason to examine the KD in combination with was likely due to the failure to eliminate ketosis
first-line agents rather than serially, with the goal in the control group (Freeman et  al., 2009). In
of maximizing the chance of seizure freedom. 2012, Lemmon et  al. reported that in the litera-
High-quality studies would be best, and given the ture to that date 88/189 (47%) LGS patients had
low frequency of infantile spasms would require a responded to KD (Lemmon et al 2012). They ret-
multicenter approach. rospectively reviewed 71 patients from their insti-
tution, John Hopkins Hospital, of whom 36 (51%)
L E N N O X G A S TAU T achieved a >50% reduction in seizures, 16 (23%) a
SYNDROME >90% reduction, and one was seizure-free, using
This is an epilepsy syndrome characterized by an intention to treat analysis. The results were
peak onset between 3 and 5 years of age. It is associ- similar at 12  months. Caraballo et  al. reported
ated with a very refractory pleomorphic epilepsy, similar results in a smaller prospectively studied
tonic seizures being most characteristic, and also group of 20 LGS patients started on diet and fol-
including atypical absences and atonic and myo- lowed for a minimum of 16  months with reten-
clonic seizures (Arzimanoglou et al., 2009). Focal tion of 75% of patients on diet, and 2/20 (15%)
seizures and complex partial seizures may also seizure-free and 25% with 50%–99% reduction
occur. Seizures may be very frequent, occurring in seizures (Caraballo et  al., 2014). Five children
45

Chapter 6: Ketogenic Diet in Established Epilepsy Indications 45

discontinued diet in this study, three because of stiripentol (STP), valproic acid (VPA), and cloba-
lack of efficacy within 3 months, and two because zam (CLB). There is a higher mortality in children
of persistent vomiting, one of whom was also with this syndrome (Genton et al., 2011), due to
hypoglycemic. Patients with LGS have also been a variety of factors (sudden unexpected death in
reported to respond with similar efficacy to the epilepsy, SE, accident) further motivating efforts
modified Atkins diet (Sharma et al., 2015), 12/25 to control convulsive seizures. An Scn1A mutant
were responders and 2 (8%) were seizure-free. mouse model of DS showed decreased latency
In summary, LGS patients, some of the most to seizure onset after an epileptogenic challenge
refractory pediatric epilepsy patients, respond to compared with wild-type littermates. Feeding the
KD therapies. As a result, medication reduction mutant mouse a ketogenic diet increased latency
or withdrawal provides special benefit to these to seizures to levels that were not significantly
patients, possibly reducing medications even fur- different from the wild-type littermates (Dutton
ther and improving quality of life. Diet therapy et al., 2011).
should be considered early once diagnosis is clear In 2011, Caraballo and colleagues reported
and refractoriness established. their experience in 59 DS patients, of whom 24
were treated with a 4:1 ketogenic diet and fol-
D R AV E T S Y N D R O M E lowed for at least 2 years (Caraballo 2011). Sixteen
( S E V E R E M YO C L O N I C remained on the diet at 2 years, of whom 2 (16%)
E P I L E P S Y O F   I N FA N C Y ) were seizure-free, 10 (62.5%) had 75%–99%
Dravet syndrome (severe myoclonic epilepsy of reduction in seizures and another 4 (25%) had
infancy; DS SMEI) is a clinical syndrome charac- 50%–74% reduction in seizures. All five with SE
terized by initial febrile seizure, often febrile SE, responded to diet and had no additional SE dur-
and/or hemiclonic febrile seizures in the first year ing follow-up. Medication reduction, even in
of life, in the setting of initially normal develop- those without dramatic seizure efficacy, resulted in
ment. It occurs in 1:20,000–1:40,000 individuals. improvement in quality of life. Ten of 15 patients
Febrile and afebrile seizures are quickly recurrent followed by Nabbout et al. achieved a >75% reduc-
and associated with subsequent developmental tion in seizures (Nabbout et al., 2011). All of these
stagnation or regression. The epilepsy is pleo- patients were already receiving triple combination
morphic (multiple seizure types—myoclonic, therapy (VPA, STP, CLB). However, efficacy was
focal, generalized absence, and generalized lost in ~50% by 1 year. Behavioral improvements
motor seizures) and pharmacoresistent. Fever/ were noted in all responders.
hyperthermia continues to provoke seizures. Laux et  al. followed a subgroup of 20/48 DS
Subsequent to seizure onset, ataxia, pyramidal patients on KD at their own center, all of whom
signs and interictal myoclonus are seen. Early remained on diet at least 6  months, 17 continu-
EEGs are normal but later, generalized spike ing for at least 18 months. They saw 65% achieve a
and polyspike waves and multifocal spikes are greater than 50% reduction in seizures and 30% a
seen. Historically, many children thought to >90% reduction. They noted that the KD appeared
have pertussis vaccine-related encephalopathy to reduce the frequency of all seizure types, though
and long-term cognitive sequelae, were found quantification of myoclonic and absence seizures
to have underlying DS (Berkovic et  al., 2006). was not robust (Laux et al., 2013).
Up to 80% are associated with a heterozygous In a retrospective cross-sectional study, 10
mutation in the sodium channel gene SCN1A, of 32 mutation positive patients (31 SCN1A and
although not all SCN1A mutations result in DS. 1 GABRG mutations) with DS were treated with
Other genetic and/or environmental factors play KD (Dressler et al., 2015b) for at least 3 months.
an as yet unclear role in evolution of the syn- Seven of 10 patients were responders at 3 months,
drome. Development generally stabilizes, but at 6 at 6 months and 12 months. One child became
least a moderate degree of intellectual disability seizure-free on KD. A  striking finding in this
is the rule. The temporal relationship between group was the lack of SE in the eight patients
seizure onset and developmental impairment who had previously suffered this seizure type. No
suggests but does not prove a causal relationship child on KD, responder or not, had SE while on
between seizures, SE, and developmental regres- the diet. Given that excess mortality in this syn-
sion. Nonetheless, vigorous efforts to control and drome is partly related to SE, this is a notable
prevent seizures are appropriate. Problems with finding that would bear further study. Early with-
this approach include medication side effects. drawal from the KD treatment was due to failure
The gold standard is combination treatment with of efficacy; later withdrawals were due to difficulty
46

46 section I: Ketogenic Diet for Epilepsy in the Clinic

maintaining compliance. Responder status was encephalopathy can develop. Seizure-related falls
not associated with mutation type in this group. are a major component of associated morbid-
An interesting aspect of the Dressler study is ity, and these children are at risk of injury. Many
the comparison with other treatments (Dressler need to wear helmets for protection. The epilepsy
et al., 2015b). There was no significant difference is often highly pharmacoresistant, and medication
in responder rates between the gold standard adverse effects add to the impact of the disorder
treatment, a combination of STP, VPA, and CLB on cognitive performance and behavior regula-
(89%); bromide (78%); KD (70%); VPA mono- tion. Often EEG is associated with generalized
therapy (48%); TPM (topiramate) (35%); and VNS spike and polyspike wave activity, ranging from 2
(vagus nerve stimulation )(37%). Ketogenic diet to 5 Hz, which also underlies the myoclonic sei-
was more effective than LEV (levetiracetam ). The zures. Prominent theta activity is a feature of the
conclusions are limited by small numbers but interictal EEG. This disorder, among other refrac-
appear worthy of further study. tory childhood epilepsies, is very responsive to the
With respect to positive interaction with other ketogenic diets. This has led to exploration of the
treatments, 7 of 16 diet responders were on topi- relationship of this disorder to GLUT1 deficiency,
ramate in Caraballo’s updated group, and 3 of 7 of a hypothesis that would provide an explanation for
Dressler’s responders were also. its particular responsiveness to ketosis. However,
In summary, treatment with KD in DS is associ- only 0%–5% of cases have been shown to be asso-
ated with a reduction in seizures, and possibly in SE. ciated with this genetic disorder (Larsen et al.,
In this highly pharmacoresistant epilepsy, polyphar- 2015; Mullen et al., 2011). Cognitive outcome is
macy is often the rule, with the attendant burden of normal in about 60% of patients in the long term.
medication side effects. The goal of optimizing sei- Continued active seizures are correlated with vary-
zure control while minimizing medication adverse ing degrees of intellectual disability, ranging from
effects may be facilitated by introduction of the mild to moderate severity.
ketogenic diet, often allowing reduction in medica- Using ILAE criteria for diagnosis, Oguni et
tion burden and therefore in cognitive and behav- al. reported treatment outcome in a group of 81
ioral side effects. An as yet unanswerable question is patients with MAE (Oguni et al., 2002). Seizure
whether early use of the ketogenic diet, prior to sei- freedom for >2 years was achieved in 55/81 (65%)
zure onset, with its lack of cognitive side effects and by 50 months (+/– 16 mo). Later recurrence of
prominent effect on status epilepticus (SE), might GTCs occurred in 14% up to 18 years later but
ameliorate the developmental stagnation/regression were then easily controlled. Eighteen percent con-
associated with seizure onset in this disorder. tinued to have refractory epilepsy. Among the
treatments used in this group, KD was used for 26
DOO SE SYNDROME patients, of whom 15 (58%) became free of myo-
( M YO C L O N I C A S TAT I C clonic (MS) and atonic (AS) seizures and 9 more
EPILEPSY) had >50% reduction in these seizures. This efficacy
Doose syndrome (myoclonic astatic epilepsy; was better than that of ACTH (n = 22, 36% MS/
MAE) is a genetic generalized epilepsy, initially AS free, 23% >50% decrease) and ethosuximide
described by Doose in 1970 and distinguished (n = 32, 32% MS/AS free, 32% >50% decrease),
from other causes of myoclonic epilepsy in child- the best of the conventional AEDs used, and than
hood (SMEI, LGS), although the underlying valproic acid (n = 57, 12% MS/AS free, 28% >50%
genetic abnormality is not yet known. It occurs decrease). Generalized motor seizures gradually
in previously normal children, with onset, some- decreased thereafter in these patients.
times explosively, possibly as early as the latter half Caraballo et  al. reported 30 MAE patients
of the first year of life, but more typically between of whom 11 were treated with diet (Caraballo
2 and 5 years of age. There is a high rate of pre- et al., 2006). Six of these remained on the diet at
ceding febrile seizures, and of a family history of 18  months of whom two (18.2%) were seizure-
epilepsy. It is characterized by frequent myoclonic free, and two each had 75%–99% reduction and
and myoclonic astatic/atonic seizures; absences, 50%–74% reduction. The EEG was improved in
with or without myoclonia; and generalized tonic- all six, and was normal in the seizure-free patients
clonic, clonic, and/or tonic seizures. The occur- at 18 months. Five patients discontinued the diet
rence of nocturnal tonic seizures is thought by within the first 3 months, four for lack of efficacy
some to indicate a poor outcome. With extremely and one for persistent vomiting. Those remain-
frequent seizures, often absences interspersed with ing on the diet tolerated it without significant side
myoclonias, and nonconvulsive SE an epileptic effects. The authors remarked the good response
47

Chapter 6: Ketogenic Diet in Established Epilepsy Indications 47

to the diet and considered that for this disorder, it light of the above reports. It remains to be seen if
should be considered earlier in the course. efficacy remains high in this setting.
Kilaru et al. reported the order of exposure to
various treatments and their efficacy in 23 MAE CONCLUSION
patients (Kilaru et  al., 2007). Ketogenic diet was In conclusion, there is general agreement, based
tried last in 10 patients, and was the most effec- on the literature described in this chapter, that
tive treatment in achieving seizure remission. patients with these four epilepsies (infantile
It was initiated an average of 17  months (range spasms, Lennox-Gastaut syndrome, Dravet syn-
2–58 months) after onset, and after an average of drome, and myoclonic-astatic epilepsy) may ben-
five AED trials. Five (50%) became seizure-free, efit from a trial of diet therapy once their epilepsy
three within 1  month, one by 7  months, and the has become refractory to medication. The quality
last by 19  months after diet initiation. Three of of the data for individual epilepsy syndromes is
these patients remained seizure-free for more than modest, at best. These diagnoses are rare, so that
6  months. Ethosuximide (25%) and topiramate only multicenter studies are likely to produce more
(23%) were the next most effective in achieving robust data, comparing diet therapy to other treat-
remission of seizures. In this group, steroids (pred- ments in a controlled manner, and/or establish-
nisone or ACTH) were ineffective. The authors ing the best timing of diet trials. There is also very
report the possibility of spontaneous remission in little information on the efficacy of diet treatment
3 of 14 patients who became seizure-free in this as first-line therapy. The difficulty of administra-
group. When this is a factor, late-used treatments tion of diet therapy is likely to limit its acceptance
may appear more effective than those used early in in this position unless it becomes possible to
the disorder, when spontaneous remission is less induce ketosis without diet restrictions, that is, via
likely. Only 43% of patients in this group were cog- a supplement or pill. Future, multicenter collab-
nitively normal at last follow-up, the rest having orative studies will be necessary to examine these
mostly mild disability. Cognitive outcome was not possibilities.
related to time to seizure freedom.
Perhaps because this epilepsy seems especially REFERENCES
responsive to the ketogenic diet, use of a lower ratio, Arzimanoglou, A., French, J., Blume, W.T., Cross,
as in the modified Atkins diet has been considered J.H., Ernst, J.P., Feucht, M., Genton, P., Guerrini,
as an option for treatment—with the potential of R., Kluger, G., Pellock, J.M., et al. (2009). Lennox-
seeing efficacy with a less rigorous, more palatable Gastaut syndrome:  a consensus approach on
diet. There is little information available about effi- diagnosis, assessment, management, and trial
cacy in comparison to the classic ketogenic diet. methodology. Lancet Neurol 8, 82–93.
Simard-Tremblay used the modified Atkins diet in Berkovic, S.F., Harkin, L., McMahon, J.M., Pelekanos,
J.T., Zuberi, S.M., Wirrell, E.C., Gill, D.S., Iona,
six and KD in three of nine patients after a median
X., Mulley J.C., and Scheffer, I.E. (2006). De-novo
of four AEDs (Simard-Tremblay et al., 2015). They mutations of the sodium channel gene SCN1A in
reported seven of nine patients achieved seizure alleged vaccine encephalopathy:  a retrospective
remission, with one patient on KD and one on study. Lancet Neurol 5, 488–492.
the modified Atkins diet failing to respond. One Caraballo, R.H., Cersósimo, R.O., Sakr, D., Cresta, A.,
patient advanced from the modified Atkins diet to Escoba, N., and Fejerman, N. (2006). Ketogenic
4:1 KD and achieved seizure freedom. When 27 diet in patients with myoclonic-astatic epilepsy.
patients were switched from the modified Atkins Epileptic Disord 8, 151–155.
diet to KD there was improvement (additional Caraballo, R.H. (2011). Nonpharmacologic treatments
10% decrease in seizures) in 10 (37%) (Kossoff of Dravet syndrome: focus on the ketogenic diet.
et al., 2010). There were nine MAE patients in this Epilepsia 52(Suppl. 2), 79–82.
group, of whom seven improved with the change Caraballo, R.H., Fortini, S., Fresler, S., Armeno, M.,
to KD, five of them achieving seizure freedom. Ariela, A., Cresta, A., Mestre, G., and Escobal, N.
(2014). Ketogenic diet in patients with Lennox–
In summary, MAE appears to be more respon-
Gastaut syndrome. Seizure 23, 751–755.
sive to ketogenic diets than other epilepsies, with Doose, H., Gerken, H., Leonhardt, R., Völzke, E.,
approximately 50% achieving seizure freedom, and Völz, C. (1970). Centrencephalic myoclonic-
allowing medication withdrawal in many and ulti- astatic petit mal: clinical and genetic investigation.
mately diet withdrawal also. Assessment of efficacy Neuropadiatrie 2, 59–78.
is complicated by a possible but poorly defined Dressler, A., Trimmel-Schwahofer, P., Reithofer, E.,
spontaneous remission rate. Diet therapy is being Gröppel, G., Mühlebner, A., Samueli S., Grabner,
offered as an earlier option more frequently in V., Abraham, K., Benninger, F., and Feucht, M.
48

48 section I: Ketogenic Diet for Epilepsy in the Clinic

(2015a). The ketogenic diet in infants—Advantages Ketogenic diet efficacy in the treatment of intrac-
of early use. Epilepsy Res. 116, 53–58. table epileptic spasms. Ped Neurol 50, 224–227.
Dressler, A., Trimmel-Schwahofer, P., Reithofer, Kilaru, S., and Bergqvist, A.G.C. (2007). Current treat-
E., Mühlebner A., Gröppel, G., Reiter-Fink, R., ment of myoclonic astatic epilepsy: clinical expe-
Benninger, F., Grassl, R., and Feucht, M. (2015b). rience at the Children’s Hospital of Philadelphia.
Efficacy and tolerability of the ketogenic diet in Epilepsia 48, 1703–1707.
Dravet syndrome: comparison with various stan- Kinsman, S.L., Vining, E.P., Quaskey, S.A., Mellits, D.,
dard antiepileptic drug regimen. Epilepsy Res and Freeman, J.M. (1992). Efficacy of the keto-
109, 81–89. genic diet for intractable seizure disorders: review
Dutton, S.B., Sawyer, N.T., Kalume, F., Jumbo-Lucioni, of 58 cases. Epilepsia 33, 1132–1136.
P., Borges, K., Catterall, W.A., and Escayg, A. Kossoff, E.H., Pyzik, P.L., McGrogan, J.R., Vining, E.P.,
(2011). Protective effect of the ketogenic diet in and Freeman, J.M. (2002). Efficacy of the keto-
Scn1a mutant mice. Epilepsia 52, 2050–2056. genic diet for infantile spasms. Pediatrics 109,
Eun, S.H., Kang, H.C., Kim D.W., and Kim, H.D. 780–783.
(2006). Ketogenic diet for treatment of infantile Kossoff, E.H., and McGrogan, J.R. (2005). Worldwide
spasms. Brain Dev 28, 566–571. use of the ketogenic diet. Epilepsia 46, 280–289.
Freeman, J.M., Vining, E.P., Pillas, D.J., Pyzik, P.L., Kossoff, E.H., Hedderick E.F., Turner, Z., and Freeman,
Casey, J.C., and Kelly, L.M. (1998). The efficacy J.M. (2008). A case-control evaluation of the keto-
of the ketogenic diet:  a prospective evaluation genic diet versus ACTH for new-onset infantile
of intervention in 150 children. Pediatrics 102, spasms. Epilepsia 49, 1504–1509.
1358–1363. Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
Freeman, J.M., and Vining, E.P. (1999). Seizures Ballaban-Gil, K.R., Bergqvist, C.A.G., Blackford,
decrease rapidly after fasting: preliminary studies R., Buchhalter, J.R., Caraballo, R.H., Cross,
of the ketogenic diet. Arch Pediatr Adolesc Med H.J., Dahlin, M.G., et  al. (2009). Optimal clini-
153, 946–949. cal management of children receiving the keto-
Freeman, J.M., Vining, E.P., Kossoff, E.H., Pyzik, P.L., genic diet: recommendations of the International
Ye, X., and Goodman, S.N. (2009). A blinded, Ketogenic Diet Study Group. Epilepsia 50,
crossover study of the efficacy of the ketogenic 304–317.
diet. Epilepsia 50, 322–325. Kossoff, E.H., Bosarge, J.L., Miranda, M.J., Wiemer-
Genton, P., Velizarova, R., and Dravet, C. (2011). Kruel, A., Kang, H.C., and Kim, H.D. (2010). Will
Dravet syndrome:  the long-term outcome. seizure control improve by switching from the
Epilepsia 52(Suppl 2), 44–49. modified Atkins diet to the traditional ketogenic
Hallbook, T., Sjolander, A., Armak, P., Miranda, M., diet? Epilepsia 51, 2496–2499.
Bjurulf, B., and Dahlin, M. (2015). Effectiveness Kossoff, E.H., Caraballo, R.H., du Toit, T., Kim, H.D.,
of the ketogenic diet used to treat resistant child- MacKay, M.T., Nathan, J.K., and Philip, S.G.
hood epilepsy in Scandinavia. Eur J Paed Neurol (2012). Dietary therapy:  a world wide phenom-
19, 29–36 enon. Epilepsy Res 100, 205–209.
Hemingway, C., Freeman, J.M., Ribbs, D.J., and Pyzik, Kwan, P., Brodie, M.J. (2000). Early identification of
P.L. (2001). The ketogenic diet: a 3-6 year follow up refractory epilepsy. N Engl J Med 342, 314–319.
of 150 children prospectively enrolled. Pediatrics Kwan, P., Arzimanoglou, A., Berg, A.T., Brodie, M.J.,
108, 898–905. Hauser, A.W., Mathern, G., Moshe, S.L., Perucca,
Henderson, C.B., Filloux, F.M., Alder, S.C., Lyon, J.L., E., Wiebe, S., and French, J. (2010). Definition of
and Caplin, D.A. (2006). Efficacy of the ketogenic drug-resistent epilepsy:  consensus proposed by
diet as a treatment option for epilepsy:  meta- the ad hoc task force of the ILAE Commission on
analysis. J Child Neurol 21, 193–198. Therapeutic Strategies. Epilepsia 52, 1069–1077.
Hong, A.M., Turner, Z, Hamdy, R.F., and Kossoff, E.H. Larsen, J., Johannesen, K.M., Ek, J., Tang, S., Marini,
(2010). Infantile spasms treated with the ketogenic C., Blichfeldt, S., Kibæk, M., von Spiczak, S.,
diet:  prospective single-center experience in 104 Weckhuysen, S., Frangu, M., et al. (2015). The role
consecutive infants. Epilepsia 51, 1403–1407. of SLC2A1 mutations in myoclonic astatic epilepsy
Hrachovy, R.A., Glaze, D.J., Frost, J.D., Jr. (1991). A and absence epilepsy, and the estimated frequency
retrospective study of spontaneous remission of GLUT1 deficiency syndrome. Epilepsia *, 1–6.
and long-term outcome in patients with infantile Laux, L., and Blackford, R. (2013). The ketogenic diet
spasms. Epilepsia 32, 212–214. in Dravet syndrome. J Child Neurol 28, 1041–1044.
Kang, H.C., Cheung, D.E., Kim, D.W., and Kim, H.D. Lefevre, F., and Aronson, N. (2000). Ketogenic diet for
(2004). Early and late onset complications in the the treatment of refractory epilepsy in children: a
ketogenic diet for intractable epilepsy. Epilepsia systematic review of efficacy. Pediatrics 105, E46.
45, 1116–1123. Lemmon, M.E., Terao, N.N., Ng, Y-T., Reisig, W.,
Kayyali, H.R., Gustafson, M., Myers, T., Thompson, Rubenstein, J.E., and Kossoff, E.H. (2012).
L., Williams, M., and Abdelmoity, A. (2014). Efficacy of the ketogenic diet in Lennox–Gastaut
49

Chapter 6: Ketogenic Diet in Established Epilepsy Indications 49

syndrome:  a retrospective review of one institu- myoclonic-astatic epilepsy of early childhood.


tion’s experience and summary of the literature. Neuropediatrics 33, 122–132.
Dev Med Child Neurol 54, 464–468. Pellock, J.M., Hrachovy, R., Shinnar, S., Baram, T.Z.,
Lux, A.L., Edwards, S.W., Hancock, E., Johnson, Bettis, D., Dlugos, D., Gaillard, W.D., Gibson, P.A.,
A.L., Kennedy, C.R., Newton, R.W., O’Callaghan, Holmes, G.L., Nordli, D.R., et al. (2010). Infantile
F.J.K., Verity, C.M., Osborne, J.P., et al. (2005). The spasms:  a U.S.  consensus report. Epilepsia 51,
United Kingdom Infantile Spasms Study (UKISS) 2175–2189.
comparing hormone treatment with vigabatrin Pires, M.E., Ilea, A., Bourel, E., Bellavoinea, V.,
on developmental and epilepsy outcomes to age Merdariua, D., Berquin P., and Auvin, S. (2013).
14 months: a multicentre randomised trial. Lancet Ketogenic diet for infantile spasms refractory to
Neurol 4, 712–717. first-line treatments:  AN open prospective study.
Mullen, S.A., Marini, C., Suls, A., Mei, D., Della Epilepsy Res 105, 189–194.
Giustina, E., Buti, D., Arsov, T., Damiano, J., Prasad, A.N., Stafstrom, C.F., Holmes, GL. Alternative
Lawrence, K., De Jonghe, P., et al. (2011). Glucose epilepsy therapies:  the ketogenic diet, immuno-
transporter 1 deficiency as a treatable cause of myo- globulins, and steroids. Epilepsia 19966666;37
clonic astatic epilepsy. Arch Neurol 68, 1152–1155. Suppl:S81–95.
Nabbout, R., Copioli, C., Chipaux, M., Chemaly, N., Schoeler, N.E., Cross, J.H., Sander, J.W., and Sisodiya,
Desguerre, I., Dulac, O., and Chiron, C. (2011). S.M. (2013). Can we predict a favourable response
Ketogenic diet  also benefits Dravet syndrome to ketogenic diet therapies for drug-resistant epi-
patients receiving stiripentol:  a prospective pilot lepsy? Epilepsy Res 106, 1–16.
study. Epilepsia 52, e54–e57. Sharma, S., Jain, P., Gulati, S., Sankhyan, N., and
Nathan, J.K., Purandare, A.S., Parek, Z.B., and Agarwala, A. (2015). Use of the modified Atkins
Manohar, H.V. (2009). Ketogenic diet in Indian diet in Lennox Gastaut syndrome. J Child Neurol
children with uncontrolled epilepsy. Indian 30, 576–579.
Pediatrics 46, 669–673. Simard-Tremblay, S., Berry, P., Owens, A., Cook, W.B.,
Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S., Sittner, H.R., Mazzanti, M., Huber, J., Warner,
Edwards, N., Fitzsimmons, G., Whitney, A., and M., Shurtleff, H., and Saneto, R.P. (2015). High-
Cross, J.H. (2008). The ketogenic diet for the treat- fat diets and seizure control in myoclonic-astatic
ment of childhood epilepsy:  a randomized con- epilepsy:  a single center’s experience. Seizure 25,
trolled trial. Lancet Neurol 7, 500–506. 184–186.
Nordli, D.R., Jr., Kuroda, M.M., Carroll, J., Ville, D., Chiron C., Laschet, J., and Dulac, O. (2015).
Koenigsberger, D.Y., Hirsch, L.J., Bruneri, H.J., The ketogenic diet can be used successfully in
Seidel, W.T., and De Vivo, D.C. (2001). Experience combination with corticosteroids for epileptic
with the ketogenic diet in infants. Pediatrics 108, encephalopathies. Epilepsy Behav 48, 61–65.
129–133. Vining, E.P, Freeman, J.M., Ballaban-Gil, K., Camfield,
Numis, A.L., Yellen, M.B., Chu-Shore, C.J., Pfeifer, C.S., Camfield, P.R., Holmes, G.L., Shinnar, S.,
H.H., and Thiele, E.A. (2011). The relationship of Shuman, R., Trevathan, E., and Wheless, J.W.
ketosis and growth to the efficacy of the ketogenic (1998). A multicenter study of the efficacy of the
diet in infantile spasms. Epilepsy Res 96, 172–175. ketogenic diet. Arch Neurol 55, 1433–1437.
Oguni, H., Tanaka, T., Hayashi, K., Funatsuka, M., Wheless, J.W. (2014). History and origin of the keto-
Sakauchi, M., Shirakawa, S., and Osawa, M. genic diet. In Epilepsy and the Ketogenic Diet
(2002). Treatment and long-term prognosis of (Totowa NJ: Humana Press), chapter 2.
50

7
Ketogenic Diet for Other Epilepsies
D AV I D T. H S I E H , M D A N D E L I Z A B E T H A . T H I E L E , M D , P H D

INTRODUCTION developmental regression around 12–18  months


As discussed in previous chapters, the ketogenic of age, specifically with regression of purpose-
diet (KD) is the treatment of choice for epilepsy ful hand use and spoken language, development
in certain disorders of brain metabolism, in par- of a gait abnormality, and hand stereotypies
ticular glucose transporter protein 1 (Glut1) defi- such as hand wringing (Neul et  al., 2010). Rett
ciency and pyruvate dehydrogenase deficiency syndrome is associated with mutations in the
(PDHD). An understanding of the underlying methyl-CpG-binding protein 2 (MECP2) gene at
metabolic pathophysiology of these genetic disor- Xq28, which are detected in 95%–97% of typi-
ders has been coupled with successful clinical effi- cal RTT patients (Neul et  al., 2010). Epilepsy
cacy of the KD in patients with Glut1 deficiency occurs in about 70% of patients with RTT, with
or PDHD. However, there are also several other a median onset of 3–4  years (Nissenkorn et  al.,
genetic disorders that manifest with epilepsy for 2010; Nissenkorn et al., 2015). Seizure semiology
which dietary therapies appear to be especially includes all types, with generalized tonic-clonic
effective, but for which the possible underlying occurring more frequently than complex partial
metabolic mechanisms are not as clearly delin- and secondarily generalized seizures (Cardoza
eated. The International Ketogenic Diet Study et  al., 2011). The treatment of epilepsy in RTT
Group has listed several conditions for which usually involves the use of standard anticonvul-
the KD has been reported as being particularly sant medications, but about a third of patients
beneficial (Box 7.1) and could be offered ear- remain medication-refractory (Pintaudi et  al.,
lier. This list includes Doose syndrome, tuberous 2010). Patients with RTT are usually not typical
sclerosis complex (TSC), Rett syndrome (RTT), candidates for epilepsy surgery, thus RTT patients
Dravet syndrome, and infantile spasms (Kossoff with medication-refractory epilepsy are often
et  al., 2009b). Whether efficacy in these condi- considered for nonmedication treatments, such as
tions is due in part to the broad-spectrum effi- dietary therapy.
cacy of the KD or to specific mechanisms specific Rett syndrome is listed by the International
to these conditions is still under investigation. Ketogenic Diet Study Group as a condition for
Furthermore, patient populations who are often which the KD has been reported as having probable
fed with gastrostomy tubes, as in RTT, may be benefit, defined as having at least two publications
especially good candidates for the KD in liquid in the literature. In a published case series of seven
form, due to the relative ease of administration. In girls with clinical RTT and medication-refractory
this chapter, we discuss the use of dietary thera- epilepsy, the KD was initiated in and tolerated by
pies for the treatment of epilepsy in certain genetic five of the patients (Haas et al., 1986). Of these five
disorders, including RTT and TSC, as listed by the patients, four of them had a 50% or better decrease
International Ketogenic Diet Study Group, and in seizures, including one with a 75% decrease and
additionally discuss the use of epilepsy dietary one with a 100% decrease. All five of the patients
therapies in patients with Angelman syndrome who were able to tolerate the diet also had reported
(AS) and Sturge-Weber syndrome (SWS). slight behavioral improvements. In another pub-
lished case of an 8-year-old girl with genetically
RETT SYNDROME confirmed RTT with medication-refractory sei-
Rett syndrome is an X-linked dominant neu- zures, a 70% reduction in seizures was achieved on
rogenetic disorder characterized clinically by the KD (Liebhaber et  al., 2003). This patient was
51

Chapter 7: Ketogenic Diet for Other Epilepsies 51

BOX 7.1
CONDITIONS LISTED BY THE INTERNATIONAL KETOGENIC DIET STUDY
GROUP AS PROBABLE BENEFIT

Glucose transporter protein 1 deficiency


Pyruvate dehydrogenase deficiency
Doose syndrome
Tuberous sclerosis complex
Rett syndrome
Dravet syndrome
Infantile spasms
Children receiving only formula

able to tolerate the diet for over 4 years, and also giant cell astrocytomas, and radial migration lines.
had improvement reported in her behavior on the Epilepsy occurs in up to 85% of TSC patients, and
KD. Additionally, in a 10-year-old girl with geneti- is a significant factor in neurological morbidity.
cally confirmed RTT and medication-refractory Seizures begin within the first year of life in 63%
seizures, the KD significantly decreased seizure and prior to 3 years of age in 83% (Chu-Shore et al.,
frequency, and this patient was also reported to 2010a). The majority of patients have multiple sei-
concurrently became “more communicative” with zure types, with the most common being complex
her family members (Giampietro et al., 2006). partial seizures, followed by infantile spasms, gen-
It is uncertain whether there is a specific under- eralized tonic-clonic seizures, and atypical absence
lying metabolic process in patients with RTT that (Chu-Shore et al., 2010a). The treatment of epilepsy
would provide a physiological benefit to treatment in TSC usually starts with the use of standard anti-
with the KD. Some researchers have noted that convulsant medications, but TSC is medication-
many patients with RTT have serum markers sug- refractory in 30%–60% of patients (Chu-Shore
gestive of defects in energy metabolism, including et al., 2010a; Vignoli et al., 2013). The TSC patients
serum lactate or pyruvate elevations (Haas et  al., with medication-refractory epilepsy are often con-
1995)  or cerebrospinal fluid lactate elevations sidered early for epilepsy surgery candidacy, with
(Matsuishi et al., 2011). It has been hypothesized cortical tubers and abnormal surrounding cortex
that perhaps the KD alters energy metabolism usually the targets of interest. When surgical candi-
in a favorable direction in patients with RTT dates are chosen carefully, good outcomes with sei-
(Haas et al., 1986; Mantis et al., 2009), but further zure freedom can be achieved in about 60% (Zhang
research is needed to better understand the impli- et al., 2013). For those patients who are not surgi-
cations of the KD in patients with RTT. cal candidates, or for whom the family does not
wish to pursue surgical management, alternative
TUBEROUS SCLERO SIS epilepsy treatment options such as dietary therapy
COMPLEX should be considered.
Tuberous sclerosis complex is an autosomal domi- Tuberous sclerosis complex is listed by the
nant neurocutaneous disorder characterized by International Ketogenic Diet Study Group as a
hamartomatous growths in multiple organ sys- condition for which the KD has been reported as
tems, including the skin, central nervous system, having probable benefit, defined as having at least
eyes, kidneys, heart, and lungs. Mutations in the two publications in the literature. In a published
TSC1 and TSC2 genes, encoding for hamartin and case series of 12 children with TSC between the
tuberin, are present in over 85% of cases, result- ages of 8  months and 18  years treated with the
ing in hamartomatous growths due to unregulated KD, at 6  months, 92% had at least a 50% reduc-
activation of the mammalian target of rapamycin tion in seizures, with 67% having at least a 90%
(mTOR) pathway (Napolioni et al., 2008). The most reduction (Kossoff et  al., 2005). In this series,
common lesions found in the brain include corti- five children had at least a 5-month seizure-free
cal tubers, subependymal nodules, subependymal response. The authors recommended the KD in
52

52 section I: Ketogenic Diet for Epilepsy in the Clinic

patients with TSC for whom medications fail ANGELMAN SYNDROME


and no clear epileptogenic tuber is identified. In Angelman syndrome is a neurogenetic disorder
another case series of three children with TSC characterized by cognitive and language impair-
and medication-refractory seizures who were fol- ment, epilepsy, ataxia and tremorous move-
lowed for 12 months on the KD, two of the patients ments, and sleep disturbances, with patients also
became seizure-free, and the third patient had his tending to have an apparent happy demeanor.
drop seizures decreased (Coppola et  al., 2006). Angelman syndrome is maternally inherited,
Within a larger series of 71 patients with TSC, six with four possible known genetic mechanisms
were started on the KD, and two of the six had a resulting in reduced expression of the ubiquitin-
50% decrease in seizures (Overwater et al., 2015). protein ligase E3A (UBE3A) gene, most com-
When the KD is effective, the possibility monly from a deletion in the 15q11.2-13.1 region
of weaning off the KD can be considered after (70%) (Jiang et  al., 1999). Epilepsy is present in
2 years. Unfortunately, a diagnosis of TSC is a risk over 85% of patients with AS (Laan et al., 1997;
factor for seizure recurrence in patients weaned off Thibert et al., 2009), with the onset of epilepsy in
of the KD after 2 or more years of seizure freedom. 50% by 1 year of age, and over 75% by 3 years of
In one published series, all three patients with TSC age (Valente et al., 2006). The underlying etiology
who were weaned off of the KD after becoming of epilepsy in AS is not clear, but there are known
seizure-free had a recurrence of seizures, and only differences in epilepsy genotype-phenotypes cor-
one regained full control with reinitiation of the relations:  AS patients with maternal deletions
KD (Martinez et al., 2007). The authors concluded have higher rates of epilepsy (90%) than those
that patients with TSC may need to be treated for without (55%–75%) (Thibert et  al., 2009), have
longer than 2 years, or possibly indefinitely if sei- an earlier onset, and have a more severe epilepsy
zure control is achieved. phenotype (Valente et al., 2005; Minassian et al.,
There is also evidence that the low glyce- 1998). It has been hypothesized that there might
mic index treatment (LGIT) diet can be effective be other factors related in patients with mater-
in reducing seizures, and is better tolerated, in nal deletions specifically associated within their
patients with TSC. In a case series of 15 patients 15q11-13 deletions, such as the genes coding for
with TSC started on the LGIT, 47% had a 50% or subunits of the γ-aminobutyric acid (GABA), a
greater reduction in their seizures (Larson et  al., receptor complex within this region (Minassian
2012). Many of these patients had been unable to et  al., 1998). Epilepsy in AS typically manifests
tolerate the KD. In addition there is a report of a with multiple seizure types, including myoclonic,
Japanese patient with TSC and Lennox Gastaut atypical absence, generalized tonic-clonic, and
syndrome responding to the LGIT, achieved using atonic seizures, and AS patients commonly have
Japanese ethnic foods (Kumada et al., 2013). frequent or prolonged seizures (Thibert et  al.,
It has now been well characterized that the 2009). Most patients are medication-refractory,
manifestations in TSC are largely due to dys- with up to 77% of patients requiring combination
functional regulation of the mTOR pathway. It is medication therapy (Laan et  al., 1997; Thibert
unclear, however, whether there is a similar under- et  al., 2009). Patients with AS and epilepsy are
lying mechanism to explain why patients with unlikely to be candidates for epilepsy surgery,
TSC respond well to dietary therapies. There is thus alternative therapies such as dietary thera-
some bench evidence that markers of the mTOR pies are often considered.
pathway, including pS6 and pAkt, are reduced Several examples of successful treatment of
in the hippocampus and liver of rats fed the KD epilepsy in AS with the KD have been published.
(McDaniel et  al., 2011). However, in a series of In one series of 19 patients with genetically con-
five patients with TSC who were treated with KD, firmed AS and medication-refractory epilepsy, the
three of the five patients had progression of known KD was described as “effective” for all four patients
tumors (renal angiomyolipomas or subependy- it was initiated in (Valente et al., 2006). In a case
mal giant cell astrocytomas) or had new tumors report of an infant with AS, the KD produced a
develop while being treated with the KD (Chu- rapid and significant reduction in seizures from
Shore et al., 2010b). Thus, if there is some mTOR hundreds a day to near complete control, and was
inhibition provided by the KD, it does not appear, accompanied by increased alertness and smiling
although only in a small sample, to provide the (Stein et al., 2010). In another report, a 5-year-old
same level of tumor regression effects in TSC as with AS became seizure-free within 2 months on
better-studied mTOR inhibitors such as sirolimus the KD and was able to wean off of two of her three
and everolimus. anticonvulsant medications (Evangeliou et  al.,
53

Chapter 7: Ketogenic Diet for Other Epilepsies 53

2010). This patient also had an improvement in of epilepsy in SWS usually starts with the use of
her sleep pattern and reduced hyperactivity. In a standard anticonvulsant medications. The SWS
large electronic survey sent through the Angelman patients with medication-refractory epilepsy are
Syndrome Foundation to 461 family members often considered for an epilepsy surgical evalu-
of patients with AS, 11% reported trying dietary ation to consider options of either a lesionec-
therapy, to include the KD in 8% and LGIT in 2%. tomy or hemispherectomy (Arzimanoglou et  al.,
Of those trying the KD, 36% reported that the 2000; Kossoff et al., 2002). Hemispherectomy can
KD was the best overall treatment, although only result in seizure freedom in 81% (Kossoff et  al.,
19% reported still being on the KD (Thibert et al., 2002), without significant worsening in post-
2009). Finally in a series of six children with AS operative hemiparesis or cognitive functioning
followed prospectively on the LGIT for 4 months, (Arzimanoglou et al., 2000).
five of six of the patients had at least an 80% reduc- The successful implementation of the KD
tion in seizures, with five remaining on the LGIT has been reported in a 4-year-old patient with
for over a year (Thibert et al., 2012). SWS, although a recurrence of seizures occurred
It is also uncertain whether there are underly- 4  months into treatment (Petit et  al., 2008).
ing mechanisms explaining the efficacy of dietary There is a theoretical risk in SWS with the KD
therapies in patients with AS. The anticonvulsant due to the fasting and fluid restrictions imple-
effects of the ketogenic diet are probably multiple mented during the initiation of the KD, which
(Bough et  al., 2007). It has been reported that could possibly provoke stroke-like events that
children on the KD have an increase in GABA, SWS patients are prone to (Kossoff et al., 2010).
taurine, serine, and glycine in their cerebrospinal Thus, the use of less restrictive dietary thera-
fluid (Dahlin et al., 2005), thus a possible specific pies has been proposed for patients with SWS.
action partly explaining the efficacy of dietary In 2010, Kossoff et al. published a case series of
therapies in patients could be the increase of the modified Atkins diet (MAD) in five children
GABA synthesis in the brain. Specifically for AS, with SWS:  three of the patients had at least a
as mentioned, there are genes encoding for the 50% reduction in seizures, including one hav-
GABAa receptor within the region of the mater- ing a 90% reduction, and one becoming seizure-
nal 15q deletions associated with AS (Minassian free (Kossoff et al., 2010). Only one patient had
et  al., 1998), thus it has been hypothesized that a stroke-like event on the MAD, although this
increasing GABA levels through the KD may be patient had a prior history of stroke-like events
an important component in its effectiveness in prior to initiation of the MAD.
patients with AS.
CONCLUSIONS
STURGE-WEBER SYNDROME In conclusion, dietary therapies are effective for a
Sturge-Weber syndrome is a sporadic neurocu- wide spectrum of epilepsy types, and we present
taneous disorder characterized by capillary mal- four examples of genetic epilepsy syndromes that
formations in the distribution of the trigeminal have been reported to respond well to dietary ther-
nerve, glaucoma, and cerebral venous malforma- apies. Whether efficacy in these conditions is due
tions. Sturge-Weber syndrome is associated with in part to the broad-spectrum efficacy of the KD or
a somatic mutation in the GNAQ gene, which to specific mechanisms specific to these conditions
plays a key role in cell proliferation (Shirley is still under investigation. If specific pathways
et  al., 2013). Epilepsy is an especially common affected in particular genetic syndromes are better
manifestation, occurring in over 85% of patients characterized, this may help us further understand
(Pascual-Castroviejo et  al., 2008), with onset by the underlying pathophysiologic mechanisms con-
1 year of age in up to 75% (Sujansky et al., 1995; tributing to the efficacy of dietary therapies in the
Kossoff et al., 2009a). Most seizures are focal, but treatment of epilepsy.
generalized seizures can also occur (Ewen et  al.,
2007), with seizures commonly occurring in a DISCLOSURES
pattern of clustering and intense seizures fol- The view(s) expressed herein are those of the
lowed by prolonged seizure-free periods (Kossoff author(s) and do not reflect the official policy or
et  al., 2009a). Seizure control in SWS is particu- position of Brooke Army Medical Center, the US
larly important because of radiographic evidence Army Medical Department, the US Army Office of
of hypoperfusion that may occur ictally in some the Surgeon General, the United States Air Force,
patients, which may contribute to progressive cor- the Department of the Army and Department of
tical atrophy (Aylett et  al., 1999). The treatment Defense, or the US Government.
54

54 section I: Ketogenic Diet for Epilepsy in the Clinic

REFERENCES Sturge-Weber syndrome worldwide. Neurology


Arzimanoglou, A.A., Andermann, F., Aicardi, J., 59, 1735–1738.
Sainte-Rose, C., Beaulieu, M.A., Villemure, J.G., Kossoff, E.H., Thiele, E.A., Pfeifer, H.H., McGrogan,
Olivier, A., and Rasmussen, T. (2000). Sturge- J.R., and Freeman, J.M. (2005). Tuberous sclero-
Weber syndrome:  indications and results of sur- sis complex and the ketogenic diet. Epilepsia 46,
gery in 20 patients. Neurology 55, 1472–1479. 1684–1686.
Aylett, S.E., Neville, B.G., Cross, J.H., Boyd, S., Chong, Kossoff, E.H., Ferenc, L., and Comi, A.M. (2009a). An
W.K, and Kirkham, F.J. (1999). Sturge-Weber syn- infantile-onset, severe, yet sporadic seizure pattern
drome:  cerebral haemodynamics during seizure is common in Sturge-Weber syndrome. Epilepsia
activity. Dev Med Child Neurol 41, 480–485. 50, 2154–2157.
Bough, K.J., and Rho, J.M. (2007). Anticonvulsant Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
mechanisms of the ketogenic diet. Epilepsia Ballaban-Gil, K.R., Christina Bergqvist, A.G.,
48, 43–58. Blackford, R., Buchhalter, J.R., Caraballo, R.H.,
Cardoza, B., Clarke, A., Wilcox, J., Gibbon, F., Smith, Helen Cross, J., Dahlin, M.G., et  al. (2009b).
P.E., Archer, H., Hryniewiecka-Jaworska, A., and Optimal clinical management of children receiv-
Kerr, M. (2011). Epilepsy in Rett syndrome: asso- ing the ketogenic diet:  recommendations of
ciation between phenotype and genotype, and the International Ketogenic Diet Study Group.
implications for practice. Seizure 20, 646–649. Epilepsia 50, 304–317.
Chu-Shore, C.J., Major, P., Camposano, S., Muzykewicz, Kossoff, E.H., Borsage, J.L., and Comi, A.M. (2010). A
D., and Thiele, E.A. (2010a). The natural history of pilot study of the modified Atkins diet for Sturge-
epilepsy in tuberous sclerosis complex. Epilepsia Weber syndrome. Epilepsy Res 92, 240–243.
51, 1236–1241. Kumada, T., Hiejima, I., Nozaki, F., Hayashi, A., and
Chu-Shore, C.J., and Thiele, E.A. (2010b). Tumor Fujii, T. (2013). Glycemic index treatment using
growth in patients with tuberous sclerosis complex Japanese foods in a girl with Lennox-Gastaut syn-
on the ketogenic diet. Brain Dev 32, 318–322. drome. Pediatr Neurol 48, 390–392.
Coppola, G., Klepper, J., Ammendola, E., Fiorillo, Laan, L.A., Renier, W.O., Arts, W.F., Buntinx, I.M., vd
M., della Corte, R., Capano, G., and Pascotto, A. Burgt, I.J., Stroink, H., Beuten, J., Zwinderman,
(2006). The effects of the ketogenic diet in refrac- K.H., van Dijk, J.G., Brouwer, O.F. (1997).
tory partial seizures with reference to tuberous Evolution of epilepsy and EEG findings in
sclerosis. Eur J Paediatr Neurol 10, 148–151. Angelman syndrome. Epilepsia 38, 195–199.
Dahlin, M., Elfving, A., Ungerstedt, U., and Amark, P. Larson, A.M., Pfeifer, H.H., and Thiele, E.A. (2012).
(2005). The ketogenic diet influences the levels of Low glycemic index treatment for epilepsy in tuber-
excitatory and inhibitory amino acids in the CSF ous sclerosis complex. Epilepsy Res 99, 180–182.
in children with refractory epilepsy. Epilepsy Res Liebhaber, G.M., Riemann, E., and Baumeister, F.A.
64, 115–125. (2003). Ketogenic diet in Rett syndrome. J Child
Evangeliou, A., Doulioglou, V., Haidopoulou, K., Neurol 18, 74–75.
Aptouramani, M., Spilioti, M., and Varlamis, G. Mantis, J.G., Fritz, C.L., Marsh, J., Heinrichs, S.C., and
(2010). Ketogenic diet in a patient with Angelman Seyfried, T.N. (2009). Improvement in motor and
syndrome. Pediatr Int 52, 831–834. exploratory behavior in Rett syndrome mice with
Ewen, J.B., Comi, A.M., and Kossoff, E.H. (2007). restricted ketogenic and standard diets. Epilepsy
Myoclonic-astatic epilepsy in a child with Sturge- Behav 15, 133–141.
Weber syndrome. Pediatr Neurol 36, 115–117. Martinez, C.C., Pyzik, P.L., and Kossoff, E.H. (2007).
Giampietro, P.F., Schowalter, D.B., Merchant, S., Discontinuing the ketogenic diet in seizure-free
Campbell, L.R., Swink, T., and Roa, B.B. (2006). children: recurrence and risk factors. Epilepsia 48,
Widened clinical spectrum of the Q128P MECP2 187–190.
mutation in Rett syndrome. Childs Nerv Syst 22, Matsuishi, T., Yamashita, Y., Takahashi, T., and
320–324. Nagamitsu, S. (2011). Rett syndrome: the state of
Haas, R.H., Rice, M.A., Trauner, D.A., and Merritt, clinical basic research, and future perspectives.
T.A. (1986). Therapeutic effects of a ketogenic diet Brain Dev 33, 627–631.
in Rett syndrome. Am J Med Genet 24, 225–246. McDaniel, S.S., Rensing, N.R., Thio, L.L., Yamada,
Haas, R.H., Light, M., Rice, M., and Barshop, B.A. K.A., and Wong, M. (2011). The ketogenic diet
(1995). Oxidative metabolism in Rett syn- inhibits the mammalian target of rapamycin
drome:  1.  Clinical studies. Neuropediatrics (mTOR) pathway. Epilepsia 52, e7–e11.
26, 90–94. Minassian, B.A., DeLorey, T.M., Olsen, R.W.,
Jiang, Y., Lev-Lehman, E., Bressler, J., Tsai, T.F., and Philippart, M., Bronstein, Y., Zhang, Q., Guerrini,
Beaudet, A.L. (1999). Genetics of Angelman syn- R., Van Ness, P., Livet, M.O., Delgado-Escueta,
drome. Am J Hum Genet 65, 1–6. A.V. (1998). Angelman syndrome:  correlations
Kossoff, E.H., Buck, C., and Freeman, J.M. (2002). between epilepsy phenotypes and genotypes. Ann
Outcomes of 32 hemispherectomies for Neurol 43, 485–493.
55

Chapter 7: Ketogenic Diet for Other Epilepsies 55

Napolioni, V., and Curatolo, P. (2008). Genetics and somatic mutation in GNAQ. N Engl J Med 368,
molecular biology of tuberous sclerosis complex. 1971–1979.
Curr Genomics 9, 475–487. Stein, D., Chetty, M., and Rho, J.M. (2010). A “happy”
Neul, J.L., Kaufmann, W.E., Glaze, D.G., toddler presenting with sudden, life-threatening
Christodoulou, J., Clarke, A.J., Bahi-Buisson, N., seizures. Semin Pediatr Neurol 17, 35–38.
Leonard, H., Bailey, M.E., Schanen, N.C., Zapella, Sujansky, E., and Conradi, S. (1995). Sturge-Weber
M., et al. (2010). Rett syndrome: revised diagnos- syndrome: age of onset of seizures and glaucoma
tic criteria and nomenclature. Ann Neurol 68, and the prognosis for affected children. J Child
944–950. Neurol 10, 49–58.
Nissenkorn, A., Gak, E., Vecsler, M., Reznki, H., Thibert, R.L., Conant, K.D., Braun, E.K., Bruno,
Menascu, S., and Ben Zeev, B. (2010). Epilepsy in P., Said, R.R., Nespeca, M.P, and Thiele, E.A.
Rett syndrome: the experience of a National Rett (2009). Epilepsy in Angelman syndrome:  a
Center. Epilepsia 51, 1252–1258. questionnaire-based assessment of the natural
Nissenkorn, A., Levy-Drummer, R.S., Bondi, O., history and current treatment options. Epilepsia
Renieri, A., Villard, L., Mari, F., Mencarelli, M.A., 50, 2369–2376.
Lo Rizzo, C., Meloni, I., Pineda, M., et al. (2015). Thibert, R.L., Pfeifer, H.H., Larson, A.M., Raby,
Epilepsy in Rett syndrome—lessons from the Rett A.R., Reynolds, A.A., Morgan, A.K., and Thiele,
networked database. Epilepsia 56, 569–576. E.A. (2012). Low glycemic index treatment for
Overwater, I.E., Bindels-de Heus, K., Rietman, A.B., seizures in Angelman syndrome. Epilepsia 53,
Ten Hoopen, L.W., Vergouwe, Y., Moll, H.A., and 1498–1502.
de Wit, M.C. (2015). Epilepsy in children with Valente, K.D., Fridman, C., Varela, M.C., Koiffmann,
tuberous sclerosis complex:  chance of remission C.P., Andrade, J.Q., Grossman, R.M., Kok, F.,
and response to antiepileptic drugs. Epilepsia 56, and Marques-Dias, M.J. (2005). Angelman syn-
1239–1245. drome:  uniparental paternal disomy 15 deter-
Pascual-Castroviejo, I., Pascual-Pascual, S.I., mines mild epilepsy but has no influence on EEG
Velazquez-Fragua, R., and Viano, J. (2008). Sturge- patterns. Epilepsy Res 67, 163–168.
Weber syndrome:  study of 55 patients. Can J Valente, K.D., Koiffmann, C.P., Fridman, C., Varella,
Neurol Sci 35, 301–307. M., Kok, F., Andrade, J.Q., Grossman, R.M, and
Petit, F., Auvin S., Lamblin, M.D., and Vallee, L. (2008). Marques-Dias, M.J. (2006). Epilepsy in patients
Myoclonic astatic seizures in a child with Sturge- with Angelman syndrome caused by deletion
Weber syndrome. Rev Neurol 164, 953–956. of the chromosome 15q11-13. Arch Neurol 63,
Pintaudi, M., Calevo, M.G., Vignoli, A., Parodi, E., 122–128.
Aiello, F., Baglietto, M.G., Hayek, Y., Buini, S., Vignoli, A., La Briola, F., Turner, K., et  al. (2013).
Renieri, A., Russo, S., et al. (2010). Epilepsy in Rett Epilepsy in TSC:  certain etiology does not mean
syndrome:  clinical and genetic features. Epilepsy certain prognosis. Epilepsia 54, 2134–2142.
Behav 19, 296–300. Zhang, K., Hu, W.H., Zhang, C., Meng, F.G., Chen,
Shirley, M.D., Tang, H., Gallione, C.J., Baugher, J.D., N., and Zhang, J.G. (2013). Predictors of seizure
Frelin, L.P., Cohen, B., North, P.E., Marchuk, freedom after surgical management of tuberous
D.A., Comi, A.M., and Pevsner, J. (2013). Sturge- sclerosis complex: a systematic review and meta-
Weber syndrome and port-wine stains caused by analysis. Epilepsy Res 105, 377–383.
56

8
The Ketogenic Diet and Related Therapies
in “Novel” Situations
Idiopathic Generalized Epilepsy Syndromes

SUDHA KILARU KES SLER, MD, MSCE

INTRODUCTION carries a higher risk of treatment resistance and


The ketogenic diet (KD) and related dietary thera- lower rate of eventual remission. The occurrence
pies, which are characterized by carbohydrate of generalized tonic-clonic (GTC) seizures is more
restriction, are now widely accepted as a treatment common in JAE than in CAE, but in both syn-
option for epilepsies that are resistant to medica- dromes, GTC seizures portend a poorer prognosis
tions. Though many studies have demonstrated for remission.
the efficacy of the KD in symptomatic generalized Ethosuximide is the first-line treatment for
or mixed epilepsies including infantile spasms, CAE, and valproic acid is also efficacious but has a
Dravet syndrome, Doose syndrome, and Lennox greater impact on tests of attention than does ETX
Gastaut syndrome, the KD is perhaps less often (Glauser et  al., 2010). Lamotrigine is efficacious
considered for idiopathic generalized epilepsies, in a smaller proportion of patients. Second-line
which include childhood absence epilepsy (CAE), therapies are broad-spectrum AEDs such as topira-
juvenile absence epilepsy (JAE), juvenile myo- mate, zonisamide, and clobazam, or conventional
clonic epilepsy (JME), and generalized epilepsy benzodiazepines such as clonazepam. Antiepileptic
with generalized tonic-clonic seizures (GE-GTC). drugs for focal mechanisms of action in epilepsy,
This chapter covers these “novel” epilepsy uses. including the sodium channel blockers, are avoided
because they might exacerbate seizures with gen-
CHILDHOOD ABSENCE eralized mechanisms of action. Thus, treatment
EPILEPSY choices are limited when first-line drugs fail.
Childhood absence epilepsy (CAE) is the most Unfortunately the published literature on keto-
common pediatric epilepsy syndrome, accounting genic diet related therapies for absence epilepsy
for more than 10% of all cases of epilepsy in chil- is quite limited. One study from Johns Hopkins
dren (Berg et  al., 2000). Onset occurs between 4 specifically evaluated the efficacy of the KD and
and 8 years of age in an otherwise neurologically the modified Atkins diet (MAD) in children with
normal child. Typical seizures are staring spells, treatment-resistant absence epilepsy, and also
lasting only seconds, occurring many times daily, included a review of studies in the literature that
and characterized on EEG by 3-Hz generalized included absence epilepsy patients (Groomes et al.,
spike and wave activity with a normal background. 2011). In the Johns Hopkins cohort, 8 patients
Childhood absence epilepsy has classically been with absence epilepsy were treated with the KD
considered a benign epilepsy syndrome that and 13 patients with absence epilepsy were treated
responds to medication, remits with time, and with the MAD. Of the combined 21 patients, only
does not affect neurodevelopmental outcome. 2 had a history of GTC seizures in addition to
Now it appears that treatment response and remis- absence, but 8 had early-onset absence with sei-
sion rates are not as high as once assumed, and zures beginning prior to 4  years of age. Diet ini-
long-term psychosocial difficulties are common tiation occurred 1 to 12 years after seizure onset,
(Masur et  al., 2013; Nickels; Wirrell et  al., 1997). and the median number of antiepileptic medica-
Juvenile myoclonic epilepsy presents later in child- tions used was 4 (range 2–10). After 3 months of
hood, typically between 11 and 17 years of age, and dietary therapy, 18 children (82%) had a >50%
57

Chapter 8: The Ketogenic Diet and Related Therapies in “Novel” Situations 57

seizure reduction, and 4 (19%) were seizure-free. JME is a common and highly recognizable epilepsy
Too few patients (only three) had EEG data before syndrome with onset in adolescence or young
and during dietary therapy to determine the effect adulthood (Dreifuss, 1989). The hallmarks of the
on spike wave discharges. syndrome are myoclonic seizures (brief bilateral
The same paper also included a summary of 17 jerks with persevered awareness), often cluster-
previously published KD studies dating from 1922 ing in the morning and sometimes triggered by
to 2008, which included patients with CAE or JAE. photic stimulation, and 4-Hz or faster generalized
Of the 133 children with absence epilepsy and spike and wave or polyspike and wave discharges
documented outcomes (with responses recorded on EEG. Many patients initially come to medical
at 3  days to 3  months), more than two-thirds attention not when the myoclonic jerks begin, but
reported a greater than 50% seizure reduction and when the first GTC seizure occurs. Absence sei-
one-third experienced periods of seizure freedom. zures occur in a subset of patients, and may be a
What is not known from this historical review is marker of worse long-term seizure prognosis (Senf
the proportion of patients presenting with a CAE- et al., 2013).
like syndrome due to glucose transporter type 1 Because valproic acid (VPA) is efficacious
deficiency, which is expected to respond well to in most patients with JME, it has long been con-
the ketogenic diet. sidered the treatment of choice (Dreifuss, 1989).
A recent case report describes a 7-year-old However, growing concerns about the long-term
girl with absence epilepsy who became seizure- side effects and teratogenic potential of VPA have
free with the paleolithic ketogenic diet consisting made it a less desirable choice, particularly in
entirely of meat, offal, fish, egg, and animal fats young women (Tomson et al., 2015; Wheless et al.,
without calorie restriction, which she apparently 2005). In addition, treatment resistance, even to
tolerated well (Clemens et  al., 2013). Of note, VPA, does occur in JME. Thus, dietary therapy
dietary therapy was used as first-line antiepilep- may be considered in this population as well.
tic treatment because of parental concerns about Very little published literature has addressed
the potential for medication side effects. Also of the use of KD-related therapies in JME. In 2013,
note, the child had other neurologic abnormali- Dr. Kossoff and colleagues described the use of the
ties (developmental delays, social withdrawal, and MAD in eight JME patients ages 15 to 44 years, six
a history of febrile seizures)—and thus was atypi- of whom were female (Kossoff et al., 2013). Except
cal for classic CAE. Whether the child underwent for one patient who was medication naïve, all oth-
diagnostic testing for glucose transporter type ers were treatment-resistant to four or more AEDs,
1 deficiency is not reported. After an immedi- and all but three had tried valproate. Of note, the
ate and gradual decline in seizure frequency, the mean age of onset was 10.5 years, which is atypi-
patient became seizure-free at 6 weeks of therapy. cally young for JME. Seven of the eight remained
At 3  months, carbohydrates were reintroduced, on the diet at 1 month and achieved at least mod-
in the form of low glycemic index vegetables. erate urinary ketosis. After 3  months, five (63%)
The patient had no evidence of epileptiform dis- reported a greater than 50% drop in seizure fre-
charges on long-term video-EEG monitoring, and quency, and two became seizure-free. At the time
continued to be seizure-free 20 months after diet the paper was published, patients had remained
initiation. on the diet 0.5 to 40  months; two patients were
Interestingly several large, single-center series able to reduce medications, but none had stopped
of KD effectiveness include no patients with all AEDs.
absence epilepsy (Dressler et  al., 2010; Hallböök Just as for CAE, many of the larger, single-
et  al., 2015; Sharma et  al., 2009; Wibisono et  al., center, long-term follow-up studies of the KD
2015). In a recent large, single-center series from included no patients with JME. The case series
Australia that included 61 patients initiating from Australia discussed previously also included
the KD, the 29 patients who were responders at a single patient with JME, who was a responder
3  months included two with treatment-resistant at 3  months (no 12-month follow-up reported).
CAE, both of whom continued to be responders at A  very recent study from Norway prospectively
12 months (Thammongkol et al., 2012). evaluated 13 adults with idiopathic generalized
epilepsy (IGE), ages 16 to 57  years, initiating the
J U V E N I L E M YO C L O N I C MAD (Kverneland et al., 2015). The majority, nine,
EPILEPSY had JME, while two had CAE, one had Jeavon syn-
Another genetic generalized epilepsy syndrome drome (absence epilepsy with eyelid myoclonia),
infrequently appearing in ketogenic diet series, and one had an IGE not otherwise specified. Nine
58

58 section I: Ketogenic Diet for Epilepsy in the Clinic

patients continued the MAD for at least 4 weeks, REFERENCES


and only six reached the 12-week time point. Four Berg, A.T., Shinnar, S., Levy, S.R., Testa, F.M., Smith-
patients achieved a 50% or greater reduction in Rapaport, S., and Beckerman, B. (2000). How well
seizure frequency—in one, frequent GTC seizures can epilepsy syndromes be identified at diagno-
stopped. Lack of motivation or lack of adherence sis? A reassessment 2 years after initial diagnosis.
to the diet were the reasons most frequently cited Epilepsia 41, 1269–1275.
Clemens, Z., Kelemen, A., Fogarasi, A., and Tóth, C.
for dropping out of the study.
(2013). Childhood absence epilepsy successfully
While the reason for referral for KD treatment
treated with the paleolithic ketogenic diet. Neurol
for most IGE patients may be treatment-resistant Ther 2, 71–76.
seizures, the KD has other potential advantages Dreifuss, F.E. (1989). Juvenile myoclonic epi-
over AEDs besides efficacy. Though specific cog- lepsy:  characteristics of a primary generalized
nitive and behavioral challenges have been noted epilepsy. Epilepsia 30(Suppl 4), S1–S7; discussion
(inattention in CAE, impulsivity and other frontal S24–S27.
lobe dysfunction in JME, for example), patients Dressler, A., Stöcklin, B., Reithofer, E., Benninger, F.,
with IGE syndromes typically have normal intel- Freilinger, M., Hauser, E., Reiter-Fink, E., Seidl, R.,
ligence at baseline—thus, cognitive adverse effects Trimmel-Schwahofer, P., and Feucht, M. (2010).
of AEDs may have a substantial impact on func- Long-term outcome and tolerability of the keto-
tion, particularly for those on polytherapy. Though genic diet in drug-resistant childhood epilepsy: the
rigorous controlled studies are still lacking, the Austrian experience. Seizure 19, 404–408.
Glauser, T.A., Cnaan, A., Shinnar, S., Hirtz, D.G.,
potentially beneficial effects of the KD on cogni-
Dlugos, D., Masur, D., Clark, P.O., Capparelli, E.
tion have been repeatedly noted. Therefore, for
V, and Adamson, P.C. (2010). Ethosuximide, val-
IGE patients who are experiencing cognitive side proic acid, and lamotrigine in childhood absence
effects from one or more AEDs, the KD may be a epilepsy. N Engl J Med 362, 790–799.
desirable alternative, or a strategy for reducing the Groomes, L.B., Pyzik, P.L., Turner, Z., Dorward, J.L.,
number of AEDs. Goode, V.H., and Kossoff, E.H. (2011). Do patients
A key challenge for dietary epilepsy therapy with absence epilepsy respond to ketogenic diets? J
in patients with IGE is the feasibility and toler- Child Neurol 26, 160–165.
ability of the diet at a time when food choices are Hallböök, T., Sjölander, A., Åmark, P., Miranda, M.,
typically made more autonomously than in early Bjurulf, B., and Dahlin, M. (2015). Effectiveness of
childhood. In addition, maintaining the mini- the ketogenic diet used to treat resistant childhood
mum protein requirements in an adolescent or epilepsy in Scandinavia. Eur J Paediatr Neurol
adult in the context of a protein and carbohydrate 19, 29–36.
Kossoff, E.H., Henry, B.J., and Cervenka, M.C. (2013).
restricted, but normal caloric, KD can be difficult.
Efficacy of dietary therapy for juvenile myoclonic
The MAD, which is high-fat and low carbohydrate,
epilepsy. Epilepsy Behav 26, 162–164.
but less stringent in protein restriction, might be Kverneland, M., Selmer, K.K., Nakken, K.O., Iversen,
more feasible and tolerable than a classic KD in P.O., and Taubøll, E. (2015). A prospective study of
these patients. However, as shown in the study by the modified Atkins diet for adults with idiopathic
Kverneland and colleagues, even adult patients generalized epilepsy. Epilepsy Behav 53, 197–201.
may-find the MAD difficult to carry out con- Masur, D., Shinnar, S., Cnaan, A., Shinnar, R.C., Clark,
sistently (Kverneland et  al., 2015). There are no P., Wang, J., Weiss, E.F., Hirtz, D.G., and Glauser,
reports of the use of two other KD-related thera- T.A. (2013). Pretreatment cognitive deficits
pies, the low glycemic index treatment (LGIT) and and treatment effects on attention in childhood
the MCT oil diet, in IGE syndromes specifically. absence epilepsy. Neurology 81, 1572–1580.
The published series on LGIT include patients Muzykewicz, D.A., Lyczkowski, D.A., Memon, N.,
with generalized seizure types, but specific syn- Conant, K.D., Pfeifer, H.H., and Thiele, E.A.
drome information is not reported (Muzykewicz (2009). Efficacy, safety, and tolerability of the low
glycemic index treatment in pediatric epilepsy.
et al., 2009; Pfeifer and Thiele, 2005).
Epilepsia 50, 1118–1126.
In conclusion, the KD or MAD are viable Nickels, K. Seizure and psychosocial outcomes of
treatment options for patients with medication- childhood and juvenile onset generalized epilep-
resistant IGE syndromes, and may be preferred sies: wolf in sheep’s clothing, or well-dressed wolf?
over AEDs causing cognitive adverse effects. Epilepsy Curr 15, 114–117.
However, feasibility and tolerability continue to be Pfeifer, H.H., and Thiele, E.A. (2005). Low-glycemic-
a major barrier. Further investigations of the long- index treatment:  a liberalized ketogenic diet for
term outcomes of IGE treated with dietary treat- treatment of intractable epilepsy. Neurology 65,
ments are needed. 1810–1812.
59

Chapter 8: The Ketogenic Diet and Related Therapies in “Novel” Situations 59

Senf, P., Schmitz, B., Holtkamp, M., and Janz, D. in girls and women of childbearing potential.
(2013). Prognosis of juvenile myoclonic epilepsy Epilepsia 56, 1006–1019.
45 years after onset: seizure outcome and predic- Wheless, J.W., Clarke, D.F., and Carpenter, D. (2005).
tors. Neurology 81, 2128–2133. Treatment of pediatric epilepsy:  expert opinion,
Sharma, S., Gulati, S., Kalra, V., Agarwala, A., and 2005. J Child Neurol 20(Suppl 1), S1–S56; quiz
Kabra, M. (2009). Seizure control and biochemi- S59–S60.
cal profile on the ketogenic diet in young chil- Wibisono, C., Rowe, N., Beavis, E., Kepreotes,
dren with refractory epilepsy:  Indian experience. H., Mackie, F.E., Lawson, J.A., and Cardamone,
Seizure 18, 446–449. M. (2015). Ten-year single-center experience
Thammongkol, S., Vears, D.F., Bicknell-Royle, J., of the ketogenic diet:  factors influencing effi-
Nation, J., Draffin, K., Stewart, K.G., Scheffer, I.E., cacy, tolerability, and compliance. J Pediatr 166,
and Mackay, M.T. (2012). Efficacy of the keto- 1030–1036.e1.
genic diet: which epilepsies respond? Epilepsia 53, Wirrell, E.C., Camfield, C.S., Camfield, P.R., Dooley,
e55–e59. J.M., Gordon, K.E., and Smith, B. (1997). Long-
Tomson, T., Marson, A., Boon, P., Canevini, M.P., term psychosocial outcome in typical absence epi-
Covanis, A., Gaily, E., Kälviäinen, R., and Trinka, lepsy. Sometimes a wolf in sheeps’ clothing. Arch
E. (2015). Valproate in the treatment of epilepsy Pediatr Adolesc Med 151, 152–158.
60

9
Ketogenic Diet in Status Epilepticus
R I M A N A B B O U T, M D , P H D

INTRODUCTION (encephalitis, meningitis), autoimmune epilepsies,


Status epilepticus (SE) is defined by continuous epilepsies with presumed immune etiology such
epileptic status with alteration of consciousness. as FIRES and NORSE, metabolic disorders, and
Status epilepticus might be a single prolonged stroke. SE might occur during the course of epi-
seizure or recurrent seizures repeated frequently lepsy syndromes; tonic clonic or clonic in Dravet
without recovery of consciousness between the syndrome, tonic in Lennox Gastaut syndrome.
seizures. The incidence of SE is 3–41/100,000 In myoclonic atonic epilepsy (Doose syndrome),
individuals per year (Chin et  al., 2006, Raspall- myoclonic status might occur at onset or during
Chaure, 2007, Dham, 2014) and it is the second the first months. Myoclonic nonconvulsive status
most common neurologic emergency in adults is frequent in some genetic syndromes such as
and the first in children. The duration of SE was Angelman syndrome, and in mitochondrial dis-
long debated, and an operational definition is eases (Trinka et al., 2015).
proposed for tonic clonic status defining T1 as the Identifying a possible underlying cause and
time that emergency treatment should be started treating it is an important step in the treatment of
and T2 as the time at which long-term conse- SE. An etiology-targeted therapy should be insti-
quences on the brain might be expected (Trinka tuted as soon as the etiology is identified (anti-
et  al., 2015). For instance, T1 is from 5 minutes biotics for CNS infections, antiviral agents for
and above and T2 is over 30 minutes of duration encephalitis, immune therapy for auto-immune
(Trinka et al., 2015). 12%–43% of patients with SE encephalitis).
fail to respond to first- and second-line therapy, Benzodiazepines are the first-line therapy
and they enter refractory SE (after 2 hours of the in SE, with different molecules used and differ-
onset of SE). Super-refractory SE (SRSE) is defined ent routes of administration; buccal, intra rectal
by the persistence of SE over 24 hours (Ferlisi or intra muscular. If benzodiazepines fail to stop
and Shorvon, 2012). Overall, ~15% of SE cases the SE, phenytoin is usually the second-line drug.
admitted to the hospital become super-refractory The use of other AEDS with IV formulations is
(DeLorenzo, 1995; Krumholz et  al., 1999; Ferlisi the usual next step after the failure of phenytoin
and Shorvon, 2012). (Shorvon and Ferlisi, 2011). Barbiturate anesthesia
In adults, SRSE presents a high rate of mortal- is the most common third-line drug and is used in
ity (>60%) (Ferlisi and Shorvon, 2012). Although adults more often than in children. Propofol and
the risk of death is low in the pediatric popula- ketamine can also be used (Ferlisi et al., 2015). The
tion, the risk of subsequent neurological morbid- failure rate of anesthesic barbiturate and the fre-
ity and cognitive problems is high (Scott, 2009). quent recurrence after withdrawal with associated
The therapeutic intervention aims to reduce SRSE high mortality and morbidity make it essential to
duration, mortality, and short- and long-term develop other therapies for medication-resistant
comorbidities. SE.
Status epilepticus can be tonic clonic, tonic The KD has reported efficacy in SE (Kossoff
or myoclonic. It can be associated with acute or and Nabbout, 2014). The multiple mechanisms of
chronic brain disease or can occur in patients with action of KD make it a good choice for refractory
known epilepsy. It might occur at the onset or in SE. The inherent combination of these mechanisms
the course of the disease (Trinka et al., 2015). For can mimic AED polytherapy, an approach that is
instance,inaugural convulsive SE without previ- suggested to be a good choice for RSE (Löscher,
ous known epilepsy occurs in CNS infections 2015, Gama et al., 2015, Lusardi et al., 2015).
61

Chapter 9: Ketogenic Diet in Status Epilepticus 61

K E TO G E N I C D I E T etiologies (Table 9.1). This trend is confirmed by


I N   C O N V U L S I V E S TAT U S the largest pediatric series with FIRES (Nabbout
EPILEPTICUS et  al., 2010)  and in adult series (Thakur et  al.,
The first report of efficacy of KD in a series of 2014). In this last series of 10 patients, 4 patients
patients with highly recurrent seizures was in 2009 presented with NORSE, 2 with anti-NMDA
by Villeneuve et  al. (Villeneuve et  al., 2009). The encephalitis, and 1 with LGI1 encephalitis.
authors reported KD response in a retrospective Other etiologies, possibly involving inflam-
pediatric series of patients with focal pharmacore- mation cascades, were reported as a possible
sistant epilepsies. The efficacy of KD was higher in indication for KD in SE; the over representa-
patients who presented with SE or recent worsen- tion of these inflammatory etiologies might
ing of seizure frequency (Villeneuve et al., 2009). be due to their frequency in SRSE. Patients
In this first series, efficacy was reported in different with Rasmussen syndrome, an encephalitis
etiologies such as Sturge-Weber syndrome, FIRES, involving the activation of the T cell pathway
and cryptogenic focal SE (Villeneuve et al., 2009). were reported as responders in a few pediat-
A second paper in 2010 reported an international ric (Villeneuve et al., 2009) and adult patients
series of nine patients with SRSE due to FIRES (Wusthoff et al., 2010).
with 7/9 responders to KD (Nabbout et al., 2010). SE in mitochondrial diseases as in Alpers dis-
The KD was well tolerated via nasogastric tube in ease with POLG1 mutations (Martikainen et  al.,
the pediatric ICU setting. Ketosis was achieved 2012) and with stroke-like accidents as in MELAS
mostly within 24 hours and SE stopped during the syndrome (Steriade et  al., 2014)  are good can-
first days of the diet and in all responders within didates for KD to be introduced early at onset
the first week (Nabbout et al., 2010). (Desguerres et  al., 2014). Other small series of
Other reports of SE due to FIRES or similar patients with focal SE due to structural lesions
immune entities were further reported in the liter- were also reported as responders to KD in pediatric
ature, including pediatric (Vaccarezza et al., 2012, (Villeneuve et al., 2009, Caraballo et al., 2014, Lin
Nam et al., 2011, Sort et al., 2013, O’Connor et al., et al., 2015) or adult series (Bodenant et al, 2008).
2014)  and adult patients (Cervenka et  al., 2011,
Thakhur et  al., 2014). The pathophysiology of K E TO G E N I C D I E T
FIRES, based on activation of an inflammatory cas- I N   N O N C O N V U L S I V E S TAT U S
cade (Nabbout et al., 2011), makes this syndrome EPILEPTICUS
a possible specific target for KD. The majority of The efficacy of KD is not restricted to convulsive
patients reported had RSE due to inflammatory SE. The efficacy of KD was reported in myoclonic

TABLE 9.1 REPORTS OF EFFICACY OF KETOGENIC DIET (KD) IN SE IN CHILDREN


AND ADULTS. *KD WAS INTRA VENOUS (IV)

Author Population Diet Etiology Time to response

Bodenant (2008) 1 adult KD Partial 7 days


Villeneuve (2009) 5 children KD SWS, encephalitis… 1–10 days
Kumada (2009) 2 children MAD Frontal lobe, heterotopia 5–10 days
Wusthoff (2010) 2 adults KD Rasmussen, head trauma 8–10 days
Nabbout (2010) 9 children KD FIRES 4–6 days
Cervenka (2011) 1 adult KD Idiopathic, autoimmune 7 days
Ismail (2012) 1 child KD FIRES 10 days
Nam (2012) 4 children, 1 adult KD Encephalitis 1–19 days
Vaccarezza (2012) 5 children KD FIRES, partial status ?
Sort (2013) 3 children KD FIRES, HHE, mito 1–6 days
Thakur (2014) 10 adults KD Super refractory SE 1–7 days
(70% “encephalitis”)
O’Connor (2014) 5 children KD Mito, myoc, FIRES? 2–8 days
Caraballo (2015) 10 children (7/10) KD Focal seizures 5–7 days
Lin (2015) 1 child KD* Focal with Gen 2–3 days
Caraballo (2015) 2 children KD Myoclonic status (EE in 1) 3 days
62

62 section I: Ketogenic Diet for Epilepsy in the Clinic

SE of mitochondrial diseases and POLG muta- Enteral feeding is usually well tolerated in our
tions (Martikainen et  al., 2012, Desguerres et  al., experience, especially when initiated with con-
2014) and of myoclonic astatic epilepsy, or Doose tinuous infusion slowly increasing the feeding rate
syndrome (Kelley et  al., 2010, Caraballo et  al., to achieve the total caloric intake within 48–72
2013,), and in patients with myoclonic SE from hours. The major steps for successful implementa-
unknown etiologies (Caraballo et  al., 2015). KD tion in the ICU are summarized in Figure 9.1.
efficacy was also reported in patients with electri- Another limiting factor for the use of the diet
cal SE during slow sleep (ESES) (Reyes et al., 2015; might be the time lag for efficacy. In the reported
Veggiotti et al., 2012). series, ketosis appears within 24 to 72 hours and
seizure reduction within the first week (Table 9.1).
CHALLENGES This time lag is difficult to accept in a severe condi-
O F   K E TO G E N I C D I E T U S E tion such as SE. Although many studies confirmed
I N   S TAT U S E P I L E P T I C U S the main role of underlying etiology in the cogni-
The challenges of the KD might raise some con- tive outcome of SE, the long duration of SE might
cerns that limit its widespread use for SE in ICUs. also negatively impact the long-term outcome
Implementation of the diet could be a complex (Kilbride et al., 2013). However, after the first- and
issue in ICUs, especially in centers lacking KD second-line therapies for SE, treatment alterna-
teams. The availability of the KD team and the tives are scarce and the use of an anesthetic agent is
daily communication between the neurologists, usually the main strategy left (Ferlisi and Shorvon,
child neurologists, dietitians, and ICU teams are 2012). Anesthetic agents are potent seizure sup-
strongly recommended, enabling initiation of the pressors and might help to shorten the SE but their
diet under optimal conditions. This daily commu- use is based on expert opinion and is not evidence
nication makes it possible to respect the follow-up based. In addition, some concerns have been raised
of the diet, thus avoiding any additional glucose about possible worsening of the outcome of refrac-
load from fluids and concomitant medications. tory SE after the use of anesthetic agents (Sutter
Enteral feeding should be privileged, since par- et al., 2014, 2015, Ferlisi et al., 2015). Limiting dex-
enteral feeding cannot achieve a high-ratio KD. trose containg IV fluids early in the course of the SE

• Confirm the indication of KD with the ICU team


• Check for errors of fatty acid metabolism and for severe hypercholesterolemia
• Stop glucose intake after normal glycemia control:
• Stop all fluids with glucose
• Change medication with glucose when possible to glucose-free medications
Pre-diet • Inform the nutrition team

• Monitor serum glucose every 6 hours


• Monitor ketosi
• Monitor serum electrolytes and acidosis
• Control all fluids and medicines to avoid glucose accidental intake
Initiation of
• Initiate continuous enteralfeeding with 1/4-1/3 goal caloric rate and advance as tolerated
the KD

• Continue the control of accidental glucose intake


• Continue for at least 2 weeks if no major side effect
• Taper if no efficacy
follow-up

FIGURE 9.1 Steps to implement the diet in ICU.


63

Chapter 9: Ketogenic Diet in Status Epilepticus 63

after blood glucose has been controlled, and initiat- REFERENCES


ing the diet as soon as inborn errors of fat metabo- Amer, S., Shah, P., and Kommineni, V. (2015).
lism are ruled out might help to shorten the delay Refractory status epilepticus from NMDA receptor
of ketosis and improve the efficacy of KD. Along encephalitis successfully treated with an adjunc-
these lines, some medications used in the setting tive ketogenic diet. Ann Indian Acad Neurol 18(2),
of SE and refractory SE, such as steroids, can delay 256–257.
Bodenant, M., Moreau, C., Sejourné, C., Auvin, S.,
ketosis. They should be avoided when medically
Delval, A., Cuisset, J.M, Derambure, P., Destée, A.,
unnecessary (Nabbout et al., 2010).
and Defebvre, L. (2008). [Interest of the ketogenic
The reports of KD in SE are mainly retro- diet in a refractory status epilepticus in adults].
spective, reporting small numbers of individuals Rev Neurol (Paris) 164(2), 194–199.
treated and rarely referring to patients where the Caraballo, R.H., Chamorro, N., Darra, F., Fortini, S.,
diet failed. In addition, comedication and chang- and Arroyo H. (2013). Epilepsy with myoclonic
ing doses of comedication with the possible time atonic seizures:  an electroclinical study of 69
to achieve efficacy of the diet might make causal patients. Pediatr Neurol 48(5), 355–362.
attribution debatable. Finally, reports lack the Caraballo, R.H., Flesler, S., Armeno, M., Fortini, S.,
long-term follow-up studies reporting cognitive Agustinho, A., Mestre, G., Cresta, A., Buompadre,
and neurological outcome, which is, apart from M.C., and Escobal, N. (2014). Ketogenic diet in
mortality, the major endpoint of any treatment of pediatric patients with refractory focal status epi-
refractory SE. Indeed, KD reports share the same lepticus. Epilepsy Res 108(10), 1912–1916.
Caraballo, R.H., Valenzuela, G.R., Armeno, M.,
weakness with all third-line therapies in refrac-
Fortini, S., Mestre, G., and Cresta A. (2015, Nov
tory SE where no drug or therapy has achieved a
26). The ketogenic diet in two paediatric patients
high level of evidence-base medicine (Ferlisi and with refractory myoclonic status epilepticus.
Shorvon, 2012). Epileptic Disord [Epub ahead of print]. PubMed
The KD is well tolerated with low rates of side PMID: 26610306.
effects in the short and long term in the ICU set- Chin, R.F., Neville, B.G, Peckham, C., Bedford, H.,
ting, as detailed in different reports (Table 9.1). Wade, A., and Scott, R.C; NLSTEPSS Collaborative
The increasing number of reports worldwide dem- Group. (2006). Incidence, cause, and short-term
onstrates its possible implementation in ICUs. Its outcome of convulsive status epilepticus in child-
efficacy in inflammatory SE or in SE from other hood: prospective population-based study. Lancet
etiologies, convulsive and nonconvulsive, should 368(9531), 222–229.
make it a therapeutic option in the treatment of DeLorenzo, R.J., Pellock, J.M., Towne, A.R., and Boggs,
refractory SE. Recently a few data on possible J.G. (1995). Epidemiology of status epilepticus. J
Clin Neurophysiol 12(4), 316–325.
improvement of cognitive outcome are emerging
Desguerre, I., Hully, M., Rio, M., and Nabbout, R.
in patients with FIRES (Kramer et al., 2011, Singh
(2014). Mitochondrial disorders and epilepsy. Rev
et al., 2014). Neurol (Paris) 170(5), 375–380.
A prospective, randomized controlled trial is Dham, B.S., Hunter, K., and Rincon, F. (2014). The
necessary to validate this treatment option, as for all epidemiology of status epilepticus in the United
third-line therapies for refractory SE. This is impor- States. Neurocrit Care 20(3), 476–483.
tant for physicians—at least one patient has died Ferlisi, M., and Shorvon, S. (2012). The outcome of
after the KD was stopped following seizure arrest therapies in refractory and super-refractory con-
because “this indication was not considered as good vulsive status epilepticus and recommendations
clinical practice” (Nabbout et  al, 2010). This trial for therapy. Brain 135(Pt 8), 2314–2328.
should evaluate efficacy and tolerance and would Ferlisi, M., Hocker, S., Grade, M., Trinka, E., and
be mandatory for the acceptance of KD by physi- Shorvon, S.; International Steering Committee of
cians and by health authorities and institutions. the StEp Audit. (2015). Preliminary results of the
Outcomes should be evaluated in the short term - global audit of treatment of refractory status epi-
lepticus. Epilepsy Behav 49, 318–324.
aiming for control of SE - and also in the long-term
Gama, I.R., Trindade- Filho, E.M., Oliveira, S.L.,
(at a few months or longer) to evaluate seizure Bueno, N.B., Melo, I.T., Cabral- Junior, C.R.,
control as well as cognitive outcomes. Pending the Barros, E.M., Galvão, J.A., Pereira, W.S.,
results of such a trial, KD should be available in Ferreira, R.C., et al. (2015). Effects of ketogenic
ICUs and be part of the treatment arsenal of refrac- diets on the occurrence of pilocarpine-induced
tory SE, a critical situation where evidence-based status epilepticus of rats. Metab Brain Dis
medicine is dramatically lacking to date. 30(1), 93–98.
64

64 section I: Ketogenic Diet for Epilepsy in the Clinic

Kelley, S.A., and Kossoff, E.H. (2010). Doose syndrome Raspall-Chaure, M., Chin, R.F., Neville, B.G.,
(myoclonic-astatic epilepsy): 40 years of progress. Bedford, H., and Scott, R.C. (2007). The epi-
Dev Med Child Neurol 52(11), 988–993. demiology of convulsive status epilepticus
Kilbride, R.D., Reynolds, A.S., Szaflarski, J.P., and in children:  a critical review. Epilepsia 48(9),
Hirsch, L.J. (2013). Clinical outcomes following 1652–1663.
prolonged refractory status epilepticus (PRSE). Reyes, G., Flesler, S., Armeno, M., Fortini, S.,
Neurocrit Care 18(3), 374–385. Ariela, A., Cresta, A., Mestre, G., and Caraballo,
Kossoff, E.H., and Nabbout, R. (2013). Use of dietary R.H. (2015). Ketogenic diet in patients with
therapy for status epilepticus. J Child Neurol 28(8), epileptic encephalopathy with electrical status
1049–1051. epilepticus during slow sleep. Epilepsy Res 113,
Kramer, U., Chi, C.S., Lin, K.L., Specchio, N., Sahin, 126–131.
M., Olson, H., Nabbout, R., Kluger, G., Lin, J.J., Scott, R.C. (2009). Status epilepticus in the developing
and van Baalen, A. (2011). Febrile infection- brain:  longterm effects seen in humans. Epilesia
related epilepsy syndrome (FIRES): pathogenesis, 50(Suppl. 12), 32e3.
treatment, and outcome: a multicenter study on 77 Shorvon, S., and Ferlisi, M. (2011). The treatment
children. Epilepsia 52(11), 1956–1965. of super-refractory status epilepticus:  a critical
Krumholz, A. (1999). Epidemiology and evidence for review of available therapies and a clinical treat-
morbidity of nonconvulsive status epilepticus. J ment protocol. Brain 134(Pt 10), 2802–2818.
Clin Neurophysiol 16(4), 314–322; discussion 353. Singh, R.K., Joshi, S.M., Potter, D.M., Leber, S.M.,
Review. Carlson, M.D., and Shellhaas, R.A. (2014).
Lin, J.J., Lin, K.L., Chan, O.W., Hsia, S.H., and Wang, Cognitive outcomes in febrile infection-related
H.S.; CHEESE Study Group. (2015). Intravenous epilepsy syndrome treated with the ketogenic diet.
ketogenic diet therapy for treatment of the acute Pediatrics 134(5), e1431–1435.
stage of super-refractory status epilepticus in a Sort, R., Born, A.P., Pedersen, K.N., Fonsmark, L., and
pediatric patient. Pediatr Neurol 52(4), 442–445. Uldall, P. (2013). Ketogenic diet in 3 cases of child-
Löscher W. (2015). Single versus combinatorial thera- hood refractory status epilepticus. Eur J Paediatr
pies in status epilepticus: novel data from preclini- Neurol 17(6), 531–536.
cal models. Epilepsy Behav 49:20–25. Steriade, C., Andrade, D.M., Faghfoury, H.,
Lusardi, T.A., Akula, K.K., Coffman, S.Q., Ruskin, D.N., Tarnopolsky, M.A., and Tai, P. (2014).
Masino, S.A., and Boison, D. (2015). Ketogenic Mitochondrial encephalopathy with lactic acidosis
diet prevents epileptogenesis and disease progres- and stroke-like episodes (MELAS) may respond
sion in adult mice and rats. Neuropharmacology to adjunctive ketogenic diet. Pediatr Neurol 50(5),
99:500–509. 498–502.
Martikainen, M.H., Päivärinta, M., Jääskeläinen, S., Sutter, R., Marsch, S., Fuhr, P., Kaplan, P.W., and
and Majamaa, K. Successful treatment of POLG- Rüegg, S. (2014). Anesthetic drugs in status epi-
related mitochondrial epilepsy with antiepileptic lepticus:  risk or rescue? A  6-year cohort study.
drugs and low glycaemic index diet. Neurology 82(8), 656–664.
Nabbout, R., Mazzuca, M., Hubert, P., Peudennier, S., Sutter, R., and Kaplan, P.W. (2015). Can anesthetic
Allaire, C., Flurin, V., Aberastury, M., Silva, W., treatment worsen outcome in status epilepticus?
and Dulac, O. (2010). Efficacy of ketogenic diet Epilepsy Behav 49, 294–297.
in severe refractory status epilepticus initiating Thakur, K.T., Probasco, J.C., Hocker, S.E., Roehl, K.,
fever induced refractory epileptic encephalopathy Henry, B., Kossoff, E.H., Kaplan, P.W., Geocadin,
in school age children (FIRES). Epilepsia 51(10), R.G., Hartman, A.L., Venkatesan, A., et al. (2014).
2033–2037. Ketogenic diet for adults in super-refractory status
Nabbout, R., Vezzani, A., Dulac, O., and Chiron, C. epilepticus. Neurology 82(8), 665–670.
(2011). Acute encephalopathy with inflammation- Trinka, E., Cock, H., Hesdorffer, D., Rossetti, A.O.,
mediated status epilepticus. Lancet Neurol 10(1), Scheffer, I.E., Shinnar, S., Shorvon, S., and
99–108. Lowenstein, D.H. (2015). A definition and classi-
Nam, S.H., Lee, B.L., Lee, C.G., Yu, H.J., Joo, E.Y., Lee, fication of status epilepticus:  Report of the ILAE
J., and Lee, M. (2011). The role of ketogenic diet Task Force on Classification of Status Epilepticus.
in the treatment of refractory status epilepticus. Epilepsia 56, 1515–1523.
Epilepsia 52(11), e181–184. Vaccarezza, M., Silva, W., Maxit, C., and Agosta, G.
O’Connor, S.E., Ream, M.A., Richardson, C., Mikati, (2012). [Super-refractory status epilepticus: treat-
M.A., Trescher, W.H., Byler, D.L., Sather, J.D., ment with ketogenic diet in pediatrics]. Rev
Michael, E.H., Urbanik, K.B., Richards, J.L., et al. Neurol 55(1), 20–25. Spanish.
(2014). The ketogenic diet for the treatment of Veggiotti, P., Pera, M.C., Teutonico, F., Brazzo, D.,
pediatric status epilepticus. Pediatr Neurol 50(1), Balottin, U., and Tassinari, C.A. (2012). Therapy
101–103. of encephalopathy with status epilepticus
65

Chapter 9: Ketogenic Diet in Status Epilepticus 65

during sleep (ESES/CSWS syndrome): an update. Wusthoff, C.J., Kranick, S.M., Morley, J.F., and
Epileptic Disord 14(1), 1–11. Christina Bergqvist, A.G. (2010). The ketogenic
Villeneuve, N., Pinton, F., Bahi-Buisson, N., Dulac, O., diet in treatment of two adults with prolonged
Chiron, C., and Nabbout, R. (2009). The ketogenic nonconvulsive status epilepticus. Epilepsia 51(6),
diet improves recently worsened focal epilepsy. 1083–1085.
Dev Med Child Neurol 51(4), 276–281.
66

10
Preventing Side Effects and Diet Discontinuation
C H E R I E L . H E R R E N, M D A N D R A NA R . S A I D, M D

INTRODUCTION on the KD, as the diet will require a shift from car-
In general, the ketogenic diet (KD) is well toler- bohydrate metabolism to fat metabolism for energy.
ated. On average, 60% of patients remain on the All patients should have a thorough metabolic
diet for over 6  months. Those who stop the diet assessment prior to initiation of dietary therapy as
typically do so due to lack of efficacy rather than detailed in table 10.2. This screening should include
tolerability (Keene et  al., 2006). Adverse effects electrolytes, blood counts, lipids, acylcarnitine
of KD therapy can be divided in to common and panel, serum amino acids, urine organic acids, liver
serious. They can also be categorized according and kidney function, uninalysis, and urine calcium
to timing during KD therapy (during initiation, and creatinine (Kossoff et  al., 2009). Patients with
during maintenance, or during discontinuation). disorders of fat metabolism with impaired fatty acid
Common side effects include constipation, vom- transport and oxidation should not be started on
iting, acidosis, and vitamin/mineral deficiencies. the KD. There are other specific diagnoses for which
More significant side effects are rare, but include the KD should not be used including (but not lim-
pancreatitis, hepatitis, kidney stones, and cardio- ited to) pyruvate carboxylase deficiency, porphyria
myopathy. During initiation, transient side effects and primary carnitine deficiency.
can include dehydration, hypoglycemia, acidosis,
vomiting, and refusal to eat. Children need to be SOCIAL
monitored closely during initiation so that these To avoid complications it is imperative to screen
adverse effects can be avoided and treated. Fasting patients both medically and socially. Patients and
and fluid restriction may aggravate these side families should be adequately educated about
effects and are not uniformly implemented in KD what to expect while on the diet and the follow-up
programs. Fluid restriction contributes to dehydra- that will be needed (typically every 3–6 months).
tion and metabolic acidosis, which subsequently For those unwilling or unable to comply with the
can cause vomiting and lethargy. With appropriate needed follow-up, careful consideration should be
monitoring and supplementation, these adverse taken to determine whether the diet is an appro-
effects can be minimized so the patient can remain priate treatment option. Once patients are started
on the diet as long as indicated. In addition, there on the diet, good social support can be beneficial
may be social issues, including refusal to eat and to families to maintain adherence and can increase
managing special occasions and holidays. With the likelihood that they continue on the diet. This
support and resources, most families are able to support often comes from medical staff including
overcome these obstacles. When patients come off neurologists, dietitians, and nurses who are well
dietary therapy, this must be done gradually and educated on the diet and the demands it places on
with close supervision, as there may be an increase families. Engagement and education of teachers
in seizures. Patients must be provided support and and schools is vital to the success of KD therapy in
direction on how to safely discontinue the diet. school-age children. Reassurance and encourage-
ment can also come from support groups designed
SCREENING specifically for families and KD-specific functions,
One of the most important steps in avoiding poten- such as Halloween parties. Special occasions tend
tial side effects (potentially severe or life threaten- to be especially difficult, but families may take
ing) is proper screening of patients. Patients with comfort in knowing there are numerous recipes
disorders of fat metabolism could present with available for everything from ice cream to birth-
severe and potentially fatal complications if started day cake! Some centers also use parent coaches,
67

Chapter 10: Preventing Side Effects and Diet Discontinuation 67

TABLE 10.1 TIME PATIENT REMAINS ON KETOGENIC DIET (KEENE, 2006)

Author Total sample % Sample at 3 months % Sample at 6 months % Sample at 12 months

DiMario [2002] 48 ? 50 37
Coppola [2002] 56 75 38 7
Maydell [2001] 147 80 66 48
Hassan [1999] 52 68 39 13
Kankirawatana [2001] 35 62 57 12
Kang [2005] 199 88 61 27
Vining [1998] 51 88 69 47
Freeman [1998] 150 83 71 55
Ruthenstein [2005] 13 85 77 50
Kossoff [2002] 23 91 78 56

who can help families navigate issues such as food In instances of more prolonged feeding intoler-
refusal and menu ideas in real life. ance, half strength Pedialyte to provide a source of
calories is usually well tolerated, without signifi-
VO M I T I N G cant reduction in ketosis.
Vomiting is one of the more common side effects
seen in patients on the KD and is estimated to ACIDO SIS
occur in 5% of patients (Keene et al., 2006). This is Metabolic acidosis can be found in patients on
most commonly seen during the initiation phase the KD and can present with lethargy and vom-
of the diet. Metabolic acidosis may contribute to iting. This will typically present during initia-
vomiting, and in turn, vomiting may worsen aci- tion and be minimized with the use of buffering
dosis, creating a worsening cycle of symptoms. agents such as sodium bicarbonate or Polycitra K
In many cases reversing metabolic acidosis will (Cypress Pharmaceuticals, Madison, MS, USA).
alleviate nausea and vomiting. In patients who are Practitioners should be aware of medications
experiencing vomiting, maintaining proper hydra- that could worsen acidosis, especially common
tion, either enterally or IV, is an important factor antiseizure medications, such as topiramate and
in breaking this cycle. Initiation is a common time zonisamide. In some cases, it may be necessary
for patients to experience vomiting related to the to decrease these medications in order to man-
diet itself; however, vomiting can occur for other age acidosis. During initiation, acidosis should be
reasons (infectious, etc.) at any time while on the treated with aggressive hydration and use of oral
diet. Families and providers should be watchful alkalinizing agents. Rarely would IV alkalization
for vomiting and have a low threshold to provide be indicated. Once the patient is on a stable diet
intervention. In those patients who cannot ade- ratio, with adequate doses of alkalinizing agents,
quately maintain oral hydration due to vomiting or acidosis becomes less problematic. However, dur-
in those who have increasing lethargy, intravenous ing times of illness, with decreased oral intake and/
fluids may be required. Monitoring for constipa- or vomiting, acidosis can again become problem-
tion is also important, as children with constipa- atic and typically can be managed with hydration.
tion may be predisposed to vomit.
Families often are concerned about whether to C O N S T I PAT I O N
re-dose antiseizure medications with emesis; we Given the composition of the KD, an increase
advise our parents to repeat the dose if vomiting in constipation would not be unexpected.
occurred less than 30 minutes from dose admin- Constipation is common and occurs in up to 65%
istration. Management of constipation with poly- of patients (Wibisono et al., 2015). This can be par-
ethylene glycol is well tolerated. Some children ticularly problematic in those who had preexisting
require slower delivery of feeds, either with more constipation, and for those who have impaired
frequent, smaller meals or, if gastrostomy tube fed, mobility. Increased hydration may help with
with slower rates of infusion with a feeding pump. symptoms, but many patients will require medica-
For short-term vomiting around times of illness, tions to manage their constipation. Polyethylene
use of Powerade Zero or Propel Zero can provide glycol (Miralax) is a common choice for treatment
hydration and electrolytes, without carbohydrates. as it is not significantly absorbed in the gut and is
68

68 section I: Ketogenic Diet for Epilepsy in the Clinic

safe for patients on the KD. Glycerin suppositories, commonly, have gritty urine or pain. There may
increased dietary fiber, medium chain triglyceride be a role for periodically checking serum uric acid
(MCT) oil, or mineral oil could also be considered. and urinary calcium/creatinine ratios. Renal ultra-
sound is necessary if there is evidence of hema-
H Y P O G LY C E M I A turia. Children with family histories of kidney
Like many other potential side effects, the risk stones receiving carbonic anhydrase inhibitors
for hypoglycemia tends to be highest during the (topiramate, zonisamide, or acetazolamide) are
initiation phase of the diet. If patients have severe also at higher risk for stones (Furth et al., 2000 and
hypoglycemia (<30 mg/dL) or symptomatic hypo- Kossoff, 2002). Treatment includes fluid liberali-
glycemia (<40 mg/dL with lethargy, vomiting, dia- zation and urinary alkalization with bicarbonate
phoresis, seizures, or shakiness), small amounts (Sampath, 2007). In children with a higher risk for
of juice (10–20 mL) or dextrose (D5W infusion kidney stone formation, prophylactic treatment
until blood glucose is >60 mg/dL) can be given with Polycitra K has been shown to reduce the
to correct the blood sugar. If hypoglycemia is a incidence of kidney stones in this susceptible pop-
persistent problem, a reduction in ratio should ulation (McNally et al., 2009).
be considered. Once stable on the diet, glucose
tends to be on the low end of normal (50–70 mg/ INCREASED INFECTIONS
dL), but is overall very stable. Fasting prior to ini- Several studies have reported rates of increased
tiation of KD therapy may increase the chances of infections in up to 2%–4% of children on the KD
hypoglycemia (Bergqvist et  al., 2005)  and there- (Woody et  al., 1989; Vining et  al., 1998; Summ
fore close glucose monitoring in these patients is et  al., 1996). Woody et  al. assessed neutrophil
recommended. function in nine children on the KD, demon-
strating that while ketotic, these patients had
K I D N E Y   S TO N E S significantly less bacterial phagocytosis and kill-
Renal calculi have been reported in 3%–7% of chil- ing. These effects were reversible, with resolution
dren on the KD (Kielb et al., 2000). These typically when the diet was discontinued (Woody et  al.,
form, on average, after 18 months of KD therapy. 1989). Other conditions associated with ketosis,
However, there have been reports of development such as diabetes mellitus, alcoholism, glycogen
of urolithiasis after just 1 month on the diet. The storage disease, protein-calorie malnutrition,
majority of stones are uric acid stones, however, certain carbohydrate-restricted diets, and intra-
calcium oxalate, calcium phosphate, and mixed lipid infusions, have also reported impairments
calcium/uric acid stones are seen. in neutrophil function. The exact mechanism is
Approximately 50% of stones are uric acid not well understood but is likely related to serum
stones (Kielb et  al., 2000). This is likely due to metabolites that affect early processes in phagocy-
KD lowering urinary pH, which facilitates forma- tosis (Woody et al., 1989). Interestingly, only one
tion of uric acid crystals, as the solubility of uric of Woody’s patients experienced serious bacterial
acid decreases as pH drops. The KD also has been infections.
associated with uric acidemia and increased uri-
nary uric acid, which further predisposes to the V I TA M I N D E F I C I E N C I E S
development of uric acid stones. This can be com- Due to the restrictive nature of the diet (with
pounded by fluid restriction, which produces a limited vegetables and grains), it is known to be
more acidic urine and decreased urine flow, with deficient in several vitamins and minerals, espe-
precipitation of urate crystals. cially B vitamins, calcium, and vitamin D.  The
The KD has also been associated with hyper- International Ketogenic Study Group (Kossoff
calciuria, which can result in calcium crystal for- et  al., 2009)  made it a universal recommenda-
mation. Hypocitraturia also occurs due to chronic tion that all patients on the KD should be on a
metabolic acidosis. Urinary citrate is an inhibitor multivitamin with minerals and receive calcium
of calcium crystal formation; therefore, low uri- with vitamin D supplements. In the study, some
nary levels increase the risk of calcium stone for- did suggest additional supplementation may
mation. Fluid restriction serves to increase this be needed for zinc, magnesium, selenium, and
risk (Herzberg, 1990). phosphorous, but this was not a universal rec-
Children typically present with gross or micro- ommendation, and may be considered on an as-
scopic hematuria, therefore patients require reg- needed basis depending on laboratory values. Care
ular screening urinalysis. They may also, less should be taken to select a carbohydrate-free or
69

Chapter 10: Preventing Side Effects and Diet Discontinuation 69

low-carbohydrate multivitamin such as NanoVM development of pancreatitis while on the KD


(Solace Nutrition, Rockville, MD, USA), FruitiVits would necessitate coming off the diet (Stewart
(Vitaflo, Alexandria, VA, USA), Centrum (Wyeth, et al., 2001). Parenteral KD therapy with intralip-
Madison, NJ, USA), or Bugs Bunny Sugar Free ids also places children at high risk for developing
(Bayer, Morristown, NJ, USA). Vitamin lev- pancreatitis and should be used judiciously.
els should be followed on a routine basis during
follow-up, and early supplementation to avoid H E PAT I T I S
deficiencies is optimal. While rare, hepatitis is a potentially significant
side effect of the KD. It has been suggested that
E L E C T R O LY T E I M B A L A N C E S concomitant use of VPA may increase the risk
Electrolytes such as calcium, magnesium, sodium, of hepatitis, however Kang at al. (2004) did not
potassium, and phosphorus are often depleted or find a statistically significant increase in hepatitis
deficient in children on KD therapy. These labo- for those on VPA, compared with those not tak-
ratory values must also be monitored closely and ing this antiseizure medication. Decreased car-
adequately supplemented. In children on KD for- nitine levels may also increase the risk for liver
mulas, many of these electrolytes may be included dysfunction, however the risk appears to be low
in the formula. However, children on solid diets (Berry-Kravis et  al., 2001). Nonetheless, rou-
will often need additional supplements. These tine monitoring of serum carnitine and carnitine
can be in the form of dietary supplements (such supplementation is recommended. Regular moni-
as Morton’s Lite Salt) or prescription (K Phos toring of liver enzymes is recommended, and in
Neutral). the event that elevations are noted, hepatic ultra-
sound and comanagement with gastroenterol-
HYPERLIPIDEMIA ogy should be considered, including possible diet
Studies show that between 30% and 60% of discontinuation.
patients on the KD develop hypercholesterolemia
(Wibisono et  al., 2015; Nizamuddin et  al., 2008). CARDIAC SIDE EFFECTS
Nizamuddin found this was increased from 25% Cardiac complications have been reported in
at preinitiation (baseline). Notably, patients eat- children on the KD, including cardiomyopathy
ing a solid diet had a greater risk for developing and prolonged QT interval (Neal, 2012). These
high cholesterol compared with those who con- risks may be related to carnitine and selenium
sumed a formula-based diet, likely secondary to an deficiencies associated with the KD. Bergqvist
increase in saturated fats in solid foods compared et  al. (2003) reported a child with cardiomyopa-
with the liquid formulation. The dyslipidemia seen thy in association with selenium deficiency and
improved spontaneously and without interven- other children who had lowering of selenium lev-
tion in about half of patients, suggesting patients els while on the KD. In a study of 20 children on
are better able to metabolize the fat over time the KD, 15% (three children) had prolongation of
(Nizamuddin et al., 2008). It is unclear whether the the QTc interval. Of these three children, two had
dyslipidemia seen with the diet has any long-term preinitiation electrocardiograms (EKGs), which
cardiovascular or atherosclerotic effects, but given confirmed that the QTc abnormalities developed
the temporary use of the diet it appears unlikely while they were on the diet. Two of the patients
and the benefit of the diet outweighs the potential had left atrial and left ventricular enlargement,
risk (Kwitwerovich, 2003). Currently, in children, and one had severe ventricular dilatation and dys-
there are no universal recommendations for inter- function, with associated symptoms of heart fail-
vention for elevated cholesterol or triglyceride ure. Interestingly, all of the patients in this series
levels. had normal selenium levels. It was postulated that
higher beta-hydroxybuturate levels and more sig-
PA N C R E AT I T I S nificant metabolic acidosis may be associated with
Hyperlipidemia and hypertriglyceridemia are risk the development of cardiac complications.
factors for development of pancreatitis, which There are also reports of two children develop-
are also potential complications of KD therapy. ing severe dilated cardiomyopathy while on the KD
Concomitant therapy with valproic acid (VPA) (Ballaban-Gil et al., 1999). In these cases, the car-
may increase this risk. There has been a case diomyopathy resolved following discontinuation
report of a 9-year-old who died from acute hem- of the KD. It is suggested that baseline EKGs and
orrhagic pancreatitis while on KD therapy. The echocardiograms be completed prior to initiation
70

70 section I: Ketogenic Diet for Epilepsy in the Clinic

of the KD and while on the diet, including carnitine and vitamin D through their preinitiation diet
and selenium levels. Supplementation with carni- (Bergqvist et  al., 2008). The poor bone health
tine and selenium should be strongly considered. seen may be secondary to antiseizure medication
(ASM) use, which can interfere with calcium and
G ROW T H vitamin D metabolism and directly impacts the
There are conflicting reports of how the KD affects cells of bone formation and absorption. Despite
growth—both linear and weight—in children on adequate supplementation with calcium and vita-
the diet. Williams et  al. (2002) retrospectively min D, and an overall reduction in ASMs, once
reviewed 21 children treated with the KD for patients were started on the KD there was further
9.6  months to 2  years, specifically looking at lin- decline in bone mineral content. Chronic acido-
ear growth velocity. Eighteen children (86%) had sis may contribute to the decline in bone health,
a drop from their original height percentiles while and buffering agents such as sodium bicarbonate
on the diet. This was independent of mean age, or sodium citrate/citric acid may be beneficial
duration on the diet, protein intake, or calories in reducing the risk of osteopenia and fractures.
consumed per body weight while on the diet. They Bone density scans may not be indicated for all
concluded that decreased linear growth might be patients, but could be considered in those at
secondary to poor nutritional status. While these higher risk (younger nonambulatory patients)
children received the recommended nutrient or for those who have experienced pathologic
intake for protein, calories were typically restricted fractures.
to 75% of recommended daily intake. The authors
postulated that dietary protein was needed to fill FOOD REFUSAL
the calorie gap for energy and gluconeogenesis, Tolerability of the KD is the single most important
resulting in relative protein insufficiency to sup- factor limiting individual acceptance for initiation,
port growth (Williams et al., 2002). however, 60% of children who start the KD remain
Vining et al. prospectively reviewed the growth on it at 6 months follow-up (Table 10.1). The most
of 237 children treated with the KD. They found common reason for discontinuation of the diet is
that older children grew taller “almost normally” not food refusal or concerns with palatability of
but younger children fell more than two standard the diet, but lack of efficacy.
deviations below the mean in height (Vining et al., Food refusal is typically related to a limited
2002). However, Couch et  al. (1999) retrospec- repertoire of foods. Families are encouraged to
tively evaluated the nutritional status and growth keep several menus on rotation to minimize food
of 21 children on the KD for 6 months. They found fatigue. Even changing presentation of meals can
a statistically significant increase in both height help with tolerability; for example, one family used
and weight after 6  months on the diet. Six chil- a child’s tea service, with its smaller plates to make
dren had a decrease in mean percentile standard meals more enjoyable. Close communication with
weight for height, but no one fell below their pre- the KD registered dietitian is vital as they have a
KD height for age percentile after 6 months on the wealth of suggestions for improved food intake.
diet. However, the study was limited by the short Parent KD coaches are also a tremendous resource.
duration of follow-up (Couch et al., 1999). Food refusal may also represent a food intolerance;
Children on the KD need to be monitored changing to a lactose-free formula may be helpful.
closely for adequate protein intake and growth If food refusal becomes a significant battle, reduc-
parameters. Often children with intractable epi- ing the KD ratio or changing to an MCT-based
lepsy become more physically active as seizures diet to increase amounts of carbohydrates may
improve, and energy needs must be recalculated. be necessary. At times, changing to other dietary
Protein intake must be adequate and regular labo- therapies, such as the modified Atkins diet or low
ratory studies to assess protein, albumin, and pre- glycemic index therapy, is warranted.
albumin are necessary. Lack of weight gain can be
commonly seen; however, inadequate or slowed DISCONTINUING
growth (which is more problematic in infants) and T H E   K E TO G E N I C   D I E T
inappropriate weight loss occur less frequently. There are many reasons why the KD is discontin-
ued, the most common being lack of efficacy after
OSTEOPENIA 3–6 months or if seizures worsen. After 2 years of
Baseline bone mineral content may be low in effective use of the KD, most neurologists would
patients starting the KD, with only an estimated consider discontinuation. Gradually lowering
54%–63% of patients receiving adequate calcium the ratio of fat to protein and carbohydrate, then
71

Chapter 10: Preventing Side Effects and Diet Discontinuation 71

TABLE 10.2 RECOMMENDATIONS FOR PRE- KD EVALUATION (KOSSOFF ET AL, 2008)

Counseling
  Discuss seizure reduction, medication, and cognitive expectations
  Identify potential psychosocial barriers to the use of KD
  Review anticonvulsants and other medications for carbohydrate content
  Recommend family read parent-oriented KD information
Nutritional evaluation
  Baseline weight, height, and ideal weight for stature
  Body mass index (BMI) when appropriate
  Nutrition intake history: 3-day food record, food preferences, allergies, aversions, and intolerances
  Establish diet formulation: infant, oral, enteral, or a combination
  Decision on which diet to begin (MCT, classic, modified Atkins, or low glycemic index)
  Calculation of calories, fluid, and ketogenic ratio (or percentage of MCT oil)
  Establish nutritional supplementation products based on dietary reference intake
Laboratory evaluation
  Complete blood count with platelets
  Electrolytes to include serum bicarbonate, total protein, calcium, zinc, selenium, magnesium, and phosphate
  Serum liver and kidney tests (including albumin, AST, ALT, blood urea nitrogen, creatinine)
  Fasting lipid profile
  Serum acylcarnitine profile
  Urinalysis
  Urine calcium and creatinine
  Anticonvulsant drug levels (if applicable)
  Urine organic acids
  Serum amino acids
Ancillary testing (optional)
  Renal ultrasound and nephrology consultation (if a history of kidney stones)
  EEG
  MRI
Cerebrospinal fluid (CSF) (if no clear etiology has been identified)
EKG (echocardiogram) if history of heart disease

relaxing the weighing of ingredients, and finally risk of recurrence include an epileptiform elec-
adding new carbohydrate foods while keeping troencephalogram (EEG), focal abnormalities on
calories constant is typical. This is followed by neuroimaging, and tuberous sclerosis complex.
relaxation of calorie restriction, then gradual intro- Approximately 50% of children with recurrence
duction of carbohydrate-containing foods. The are difficult to control once again, even with
ideal weaning speed of the KD is not clear, specifi- resumption of the KD (Martinez et al., 2007).
cally as it pertains to the risk of increased seizures.
Worden et al. (2011) retrospectively reviewed 183
children who discontinued the KD. There was no CONCLUSION
significant difference in the incidence of seizures While the KD is an effective therapy for refractory
worsening between immediate (<1 week), quick epilepsy and certain metabolic disorders, there are
(1–6 weeks), or slow (>6 weeks) rates. However, potential adverse effects. Providers must be aware
there was an increased risk of seizures worsening of these risks for proper monitoring of children
in those patients specifically with a 50%–99% sei- and counseling families. Like all medical therapies,
zure reduction and those who were receiving more potential risks of treatment are weighed against
antiseizure medications (Worden et al., 2011). benefits of the diet.
Approximately 80% of children will remain
seizure-free when tapered off the KD if they have REFERENCES
completely responded to diet therapy and are Ballaban-Gil, K.R. (1999). Cardiomyopathy associated
medication free. Factors associated with a higher with the ketogenic diet. Epilepsia 40(Suppl 7), 129.
72

72 section I: Ketogenic Diet for Epilepsy in the Clinic

Berry-Kravis, E., Booth, G., Sanchez, A.C., and Keene, D.L. (2006). A systematic review of the use of
Woodbury-Kolb, J. (2001). Carnitine levels and the ketogenic diet in childhood epilepsy. Pediatr
the ketogenic diet. Epilepsia 42, 1445–1451. Neurol 35, 1–5.
Bergqvist, A.G.C, Chee, C.M., Lutchka, L., Rychik, J., Kielb, S., Koo, H.P., Bloom, D.A., and Faerber, G.J.
and Stallings, V.A. (2003). Selenium deficiency (2000). Nephrolitiasis associated with the keto-
associated with cardiomyopathy:  a complication genic diet. J Urol 164, 464–466.
of the ketogenic diet. Epilepsia 44, 618–620. Kossoff, E.H., Pyzik, P.I., Furth, S.L., Hladky, K.D.,
Bergqvist, A.G.C, Schall, J.I., Gallagher, P.R., Cnaan, A., Freeman, J.M., and Vining, E.P.G. (2002). Kidney
Stallings, V.A. (2005). Fasting versus gradual ini- stones, carbonic anhydrase inhibitors, and the
tiation of the ketogenic diet:  a prospective, ran- ketogenic diet. Epilepsia 43, 1168–1171.
domized clinical trial of efficacy. Epilepsia 46, Kossoff, E., Pyzik, P., McGrogan, J., Vining, E.,
1810–1819. Freeman, J. Efficacy of the ketogenic diet for infan-
Bergqvist, A.G.C, Schall, J.I., Stallings, V.A., and tile spasms. Pediatrics 109 (2002), pp. 780–783.
Zemel, B.S. (2008). Progressive bone mineral loss Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E.,
in children with intractable epilepsy treated with Ballaban-Gil, K.R., Bergqvist, A.G., Blackford, R.,
the ketogenic diet. Am J Clin Nutr 88, 1678–1684. Buchhalter, J.R., Baraballo, R.H., Cross, J.H.,
Coppola, G., Veggiotti, P., Cusmai, F. et al. (2002). The Dahlin, M.G., et  al. (2009). Optimal clinical
ketogenic diet in children, adolescents and young management of children receiving the keto-
adults with refractory epilepsy:  An Italian multi- genic diet: recommendations of the International
centric experience. Epilepsy Res. 48, 221–227. Ketogenic Diet Study Group. Epilepsia 50,
Couch, S.C., Schwarzman, F., Carroll, J., Koeniqsberger, 304–317.
D., Nordli, D.R., Deckelbaum, R.J., and DeFelice, Martinez, C.C., Pyzik, P.I., and Kossoff, E.H. (2007).
A.R. (1999). Growth and nutritional outcomes of Discontinuing the ketogenic diet in seizure-free
children treated with the ketogenic diet. J Am Diet children: risk factors and recurrence. Epilepsia 48,
Assoc 99, 1573–1575. 187–190.
DiMario, F.J.Jr, Holland, J. (2002). The Ketogenic Maydell, B., Wyllie, E., Akhtar, H. et al. (2001). Efficacy
diet: A review of the experience at the Connecticut of the ketogenic diet in focal versus generalized
Children’s Medical Center, Pediatr Neurol. 26, seizures. Pediatr Neurol 25, 208–212.
288–292. McNally, M.A., Pyzik, P.L., Rubenstein, J.E., Hamdy,
Freeman, J., Vining, E., Pillas, D., Pyzik, P., Casey, J., R.F., and Kossoff, E.H. (2009). Empiric use of
Kelly, M. (1998). The efficacy of the ketogenic diet– potassium citrate reduces kidney stone incidence
1998: A prospective evaluation of the intervention with ketogenic diet. Pediatrics 124, e300–304.
in 150 children. Pediatrics, 102, 1358–1363. Neal, E.G., Zupec-Kania, B., and Pfeifer, H.H. (2012).
Furth S.I., Casey J.C., Pyzik P.L., Neu A.M., Docimo, Carnitine, nutritional supplementation and dis-
S.G., Vining, E.P., Freeman, J.M., and Fivush, continuation of ketogenic diet therapies. Epilepsy
B.A. (2000). Risk factors for urolithiasis in chil- Res 100, 267–271.
dren on the ketogenic diet. Pediatr Nephrol 15, Nizamuddin, J., Turner, Z., Rubenstein, J.E., Pyzik,
125–128. P.L., and Kossoff, E.H. (2008). Management and
Hassan, A., Keene, D., Whiting, S., Jacob, P., risk factors of dyslipidemia with ketogenic diet.
Champagne, J., Humphreys, P. (1999). Ketogenic J Child Neurol 23, 758–761.
diet in the treatment of refractory epilepsy in Ruthenstein, J., Kossoff, E., Pyzik, P., Vining, E.,
childhood. Pediatr Neurol, 21, 548–552. McGrogan, J., Freeman, J. (2005). Experience in
Herzberg, G.Z., Fivush, B.A., Kinsman S.L., and the use of the ketogenic diet as early therapy. J Chil
Gearhart J.P. (1990). Urolithiasis associated with Neurol, 20, 31–34
the ketogenic diet. J Pediatr 117, 743–745. Sampath, A., Kossoff, E.H., Furth, P.L., Pyzik, P.L., and
Kang, H.C., Chung, D.E., Kim, D.W., and Kim, H.D. Vining, E.P. (2007). Kidney stones and the keto-
(2004). Early and late-onset complications of the genic diet:  risk factors and prevention. J Child
ketogenic diet for intractable epilepsy. Epilepsia Neurol 22, 375–378.
45, 1116–1123. Stewart, W.A., Gordon, K., and Camfield, P. (2001).
Kang, H., Kim, Y., Kim, D., Kim, H. (2005). Efficacy and Acute pancreatitis causing death in a child on the
safety of the ketogenic diet for intractable child- ketogenic diet. J Child Neurol. 16, 682.
hood epilepsy:  Korean Multicentric Experience. Summ, M., Woch, M.A., and McNeil, T. (1996).
Epilepsia, 46, 272–279. Success and complications of the ketogenic diet for
Kankirawatana, P., Jirapinyo, P., Kankirawatana, S., intractable childhood epilepsy. Epilepsia 37 (Suppl
Wongarn, R., Thamasasiri, N. (2001). Ketogenic 5), 109.
diet:  An alternative treatment for refractory Vining, E., Freeman, J., Ballaban-Gil, K. et al. (1998).
epilepsy of childhood. J Med Assoc Thai, 84, A multicenter study of the efficacy of the ketogenic
1027–1032. diet. Arch Neurol, 55, pp. 1433–1437.
73

Chapter 10: Preventing Side Effects and Diet Discontinuation 73

Vining, E.P., Freeman, J.M., Ballaban-Gil, efficacy, tolerability, and compliance. J Pediatr 166,
K.,Camfield, C.S., Camfield, P.R., Holmes, G.L., 1030–1036.
Shinnar, S., Shuman, R., Trevathan, E., Wheless, Williams, S., Basualdo-Hammond, C., Curtis, R., and
J.W. (1998). A multicenter study of the effi- Schuller, R. (2002). Growth retardation in children
cacy of the ketogenic diet. Arch Neurology 55, with epilepsy on the ketogenic diet: a retrospective
1433–1437. chart review. J Am Diet Assoc 102, 405–407.
Vining, E.P., Pyzik, P., McGrogan, J., Hladky, H., Woody, R.C., Steele, R.W., Knapple, W.L., and
Anand, A., Kriegler, S., and Freeman, J.M. (2002). Pilkington, N.S., Jr. (1989). Impaired neutrophil
Growth of children on the ketogenic diet. Dev function in children with seizures treated with the
Med Child Neurol. 44, 796–802. ketogenic diet. J Pediatr 115, 427–430.
Wibisono, C., Rowe, N., Beavis, E., Kepreotes, H., Worden, L.T., Turner, Z., Pyzik, P.L., Rubenstein, J.E.,
Mackie, F.E., Lawson, J.A., and Cardamone, and Kossoff, E.H. (2011). Is there an ideal way to
M. (2015). Ten-year single-center experi- discontinue the ketogenic diet? Epilepsy Res 95,
ence of the ketogenic diet:  factors influencing 232–236.
74
75

SECTION II
Ketogenic Diet

Emerging Clinical Applications


and Future Potential

JONG M. RHO, MD, SECTION EDITOR


76
77

11
Overview
Expanded Uses of Ketogenic Therapies

JONG M. RHO, MD

T he ketogenic diet (KD) is a proven therapy


for drug-resistant epilepsy (Vining et al.,
1998; Neal et al., 2008), and while the mecha-
by Dr.  Ning Cheng and co-authors (chapter  14)
of metabolic treatments for autism spectrum dis-
order, whose prevalence has been rising rapidly
nisms underlying its antiseizure effectiveness throughout the world. Next, Dr. Stephen Cunnane
remain unclear (Masino and Rho, 2012), there is and collaborators (chapter  15) provide a unique
growing experimental evidence for the neuropro- translational perspective on brain metabolism—
tective properties of this metabolism-based treat- using novel positron emission tomography tech-
ment (Barañano and Hartman, 2008; Maalouf et niques to make the case that nutritional ketosis,
al., 2009; Gano et al., 2014). Although definitive achieved through several currently available strate-
clinical evidence for use outside of epilepsy is gies, can correct the underlying problem of declin-
presently lacking, there are nevertheless ongoing ing brain fuels during aging. Dr. Wolf Tetzlaff and
human trials for a few neurological conditions coauthors (Streijger et al., chapter 16) then review
(www.ClinicalTrials.gov) assessing whether a the evidence that the KD (and in particular, ketone
ketogenic therapy can ameliorate symptoms and bodies) can prevent the metabolic, oxidative, and
even the disease processes themselves. Much of inflammatory cascades that follow after spinal
the experimental evidence has been converging cord injury and traumatic brain injury. The next
on the notion that the broad efficacy of the KD chapter, by Drs. Nina Dupuis and Stéphane Auvin
is due in major part to normalization of aberrant (chapter 17), reviews the mounting evidence that
energy metabolism (Masino and Rho, 2012). The the KD possesses anti-inflammatory properties,
concept that multiple neurological disorders are relevant not only to the pathophysiology of pain
pathophysiologically linked to energy dysregula- syndromes and multiple sclerosis but also to other
tion (Pathak et al., 2013) has been strengthened neurological disorders. Finally, Dr. Carl Stafstrom
by numerous studies evaluating brain bioenerget- (chapter 18) covers the current evidence that the
ics, and provides a solid rationale for how dietary KD may also afford beneficial effects against two
treatments that ameliorate underlying dysfunction neurodegenerative disorders (amyotrophic lateral
can treat or even prevent disease symptomatology. sclerosis and Parkinson’s disease), mood disorders,
In this section, “Ketogenic Diet—Emerging and migraine. Throughout all of these apparently
Clinical Applications and Future Potential,” lead- distinct neurological conditions, it should become
ing investigators in the field of ketogenic therapies clear to the reader that metabolic derangements
provide up-to-date reviews summarizing the evi- are now considered key to the pathophysiology of
dence that the KD may prove useful in the treat- these disorders, and that a dietary treatment such
ment of a number of neurological conditions. Drs. as the KD can potentially benefit patients afflicted
Thomas Seyfried, Adrienne Scheck, and colleagues with any one of several neurological diseases. It
put forward a compelling case for cancer as being is also the hope of the aforementioned authors
in part a metabolic (and perhaps more specifically, that those curious about how altered metabolism
a mitochondrial) disorder, and that the KD should can both induce and mitigate neurodegeneration
be strongly considered as adjunctive treatment and brain dysfunction will be compelled to pur-
for malignant brain cancer (Seyfried and Shelton, sue further laboratory and/or clinical research in
chapter 12; Woolf and Scheck, chapter 13). These this growing and fascinating area of translational
chapters are followed by a comprehensive review neurosciences.
78

78 section II: E merging Clinical Applications and Future Potential

REFERENCES the Epilepsies [Internet], 4th edition, J.L. Noebels,


Barañano, K.W., and Hartman, A.L. (2008). The M. Avoli, M.A. Rogawski, R.W. Olsen, and A.V.
ketogenic diet: uses in epilepsy and other neuro- Delgado-Escueta, eds. (Bethesda, MD:  National
logic illnesses. Curr Treat Options Neurol 10(6), Center for Biotechnology Information).
410–419. Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
Clinical Trials. NCT02046187, NCT01865162, Edwards, N., Fitzsimmons, G., Whitney, A., and
NCT02477904, NCT02174016, NCT01915433. Cross, J.H. (2008). The ketogenic diet for the treat-
www.ClinicalTrials.gov ment of childhood epilepsy:  a randomised con-
Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic trolled trial. Lancet Neurol 7(6), 500–506.
diets, mitochondria, and neurological diseases. J Pathak, D., Berthet, A., and Nakamura, K. (2013).
Lipid Res 55(11), 2211–2228. Energy failure: does it contribute to neurodegen-
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The eration? Ann Neurol 74(4), 506–516.
neuroprotective properties of calorie restriction, Vining, E.P., Freeman, J.M., Ballaban-Gil, K., Camfield,
the ketogenic diet, and ketone bodies. Brain Res C.S., Camfield, P.R., Holmes, G.L., Shinnar, S.,
Rev 59(2), 293–315. Shuman, R., Trevathan, E., and Wheless, J.W.
Masino, S.A., and Rho, J.M. (2012). Mechanisms of keto- (1998). A multicenter study of the efficacy of the
genic diet action. In Jasper’s Basic Mechanisms of ketogenic diet. Arch Neurol 55(11), 1433–1437.
79

12
Metabolism-Based Treatments to Counter Cancer
Scientific Rationale

T H O M A S N . S E Y F R I E D , P H D A N D L AU R A M . S H E LT O N, P H D

INTRODUCTION IS CANCER A GENETIC


Cancer has long been recognized as a genetic DISEASE OR A
disease involving mutations in oncogenes and M I TO C H O N D R I A L
tumor suppressor genes that reside in the tumor M E TA B O L I C D I S E A S E ?
cell nucleus. The nuclear gene mutations found in As an alternative to the somatic mutation theory,
nearly all types of tumors are considered the pri- emerging evidence suggests that cancer is primar-
mary cause of the cancer’s hallmarks, including ily a mitochondrial metabolic disease (Bartesaghi
(1)  sustained proliferative signaling, (2)  evasion et al., 2015; Hu et al., 2012; Seyfried, 2015; Seyfried
of growth suppressors, (3) resistance to cell death, et al., 2014; Seyfried and Shelton, 2010; Verschoor
(4)  replicative immortality, (5)  enhanced angio- et  al., 2013). This concept originated from the
genesis, and (6) activation of invasion and metas- experiments of Otto Warburg (Burk et  al., 1967;
tasis (Hanahan and Weinberg, 2000, 2011). The Warburg, 1956a, 1956b). According to Warburg’s
somatic mutations in tumor cells arise randomly theory, respiratory insufficiency is the origin of can-
during DNA replication in normal nontumori- cer. All other phenotypes of the disease, including
genic stem cells and are thought to be the origin of the somatic mutations and aerobic fermentation,
cancer (Tomasetti and Vogelstein, 2015). arise either directly or indirectly from insufficient
The somatic mutation theory is the most respiration (Seyfried, 2012a; Seyfried et al., 2014;
widely accepted explanation for the origin of can- Warburg, 1956a). Warburg’s metabolic theory was
cer, and is the justification for developing targeted also in line with the concepts of C. D. Darlington
or personalized approaches for treating various and others showing that cancer is largely a cyto-
forms of the disease (Hou and Ma, 2014; McLeod, plasmic mitochondrial disease (Darlington, 1948;
2013; Seyfried, 2015; Vaux, 2011). Despite numer- Seeger, 1959; Seyfried, 2012a, 2015; Seyfried
ous inconsistencies associated with the somatic and Shelton, 2010; Woods and du Buy, 1945).
mutation theory (Baker and Kramer, 2007; Advocates of the somatic mutation theory, how-
Burgio and Migliore, 2015; Cao et al., 2015; Rous, ever, consider the abnormal energy metabolism of
1959; Sonnenschein and Soto, 2000; Soto and tumor cells as simply another phenotype that “is
Sonnenschein, 2004), this theory is presented as if programmed by proliferation-inducing oncogenes
it were dogma in most current college textbooks of and defective tumor suppressor genes” (Hanahan
genetics, biochemistry, and cell biology, and is the and Weinberg, 2011; Seyfried, 2015). The gene
mainstay of the National Cancer Institute’s position theory of cancer, however, is inconsistent with the
which states, “Cancer is a genetic disease—that is, data obtained from the nuclear-cytoplasm transfer
it is caused by changes to genes that control the experiments.
way our cells function, especially how they grow A recent review of data from a broad range of
and divide” (http://www.cancer.gov/cancertopics/ nuclear-cytoplasm transfer experiments provides
what-is-cancer) (Seyfried, 2015). It should be rec- the most compelling evidence to date showing
ognized, however, that there is no place in science that somatic mutations alone cannot account for
for epistemological dogmatism, as dogma tran- the origin of cancer (Seyfried, 2015). These experi-
quilizes creative thinking and blocks consideration ments showed that cytoplasm containing normal
of viable alternatives (Rous, 1959; Sonnenschein mitochondria suppressed tumorigenesis in cells
and Soto, 2000). with tumor-derived nuclei. In contrast, cytoplasm
80

80 section II: E merging Clinical Applications and Future Potential

containing abnormal mitochondria from tumor Although the evidence supporting mitochon-
cells enhanced tumorigenesis in cells with normal drial abnormalities and OxPhos insufficiency in
nuclei. Viewed in the light of Warburg’s central cancer is now overwhelming, some metabolic
theory, the nuclear-cytoplasm transfer experi- studies on cultured tumor cells can be inconsis-
ments show that cancer originates from damage tent with the findings from intact tumor tissue.
to the mitochondria in the cytoplasm rather than The unnatural conditions of the in vitro growth
from damage to the genome in the nucleus. The environment (Crabtree effect) can make some
genomic instability seen in tumor cells follows, cancer cells appear to have normal respiration
rather than precedes, the disturbances in cellular despite their continued production of lactate. The
respiration. information on tumor cell energy metabolism
Seyfried et  al. described how the common obtained from in vitro studies, however, is some-
pathophysiological mechanism underlying can- times at odds with the data from in vivo studies.
cer, that is, “the oncogenic paradox,” was better The results from many in vitro studies suggest
explained when cancer is considered as a mito- that normal cells and tumor cells have similar
chondrial metabolic disease than when considered mitochondrial energy metabolism (Cairns, 2015;
as a genetic disease (Seyfried, 2012c; Seyfried et al., Koppenol et al., 2011). If mitochondrial structure
2014; Seyfried and Shelton, 2010), and recently is abnormal in the vast majority of tumor cells seen
outlined how all roads to the origin and progres- in various cancerous tissues, how is it possible that
sion of cancer pass through the mitochondria mitochondria and OxPhos could be normal in cul-
(Seyfried et  al., 2014). Hence, novel therapeutic tured cells from the tumors? Seyfried et al. recently
strategies for management and prevention emerge addressed several confounding issues related to
when cancer becomes recognized as a mitochon- tumor cell energy metabolism that is measured in
drial metabolic disease. cultured cells (Seyfried et al., 2014).
The reduced efficiency of mitochondrial
C A N C E R C E L L M E TA B O L I S M OxPhos for ATP production in tumor cells is
It is well known that lactate fermentation is the compensated for with an increased dependency
common metabolic malady seen in most if not all on substrate level phosphorylation for energy pro-
cancer cells (Seyfried et al., 2014; Warburg, 1931, duction. Substrate level phosphorylation occurs
1956a, 1956b, 1969). Indeed, lactate fermenta- at steps 7 and 10 in the glycolytic pathway, and at
tion can be detected even in the slowest growing the succinyl-CoA synthase reaction (Step 5) of the
tumors (Burk and Schade, 1956; Burk et al., 1967). citric acid cycle. Only those cells that can transi-
The greater the lactate production, the faster is tion their energy production from OxPhos to sub-
the rate of tumor growth. In addition to express- strate level phosphorylation can become tumor
ing robust fermentation in hypoxic environments, cells. Cells that cannot make this transition will
cancer cells also ferment lactate in aerobic envi- die and can never become tumor cells (Warburg,
ronments. Aerobic fermentation is the antithesis to 1956a). As substrate level phosphorylation is less
the Pasteur effect (suppression of lactate fermenta- efficient in producing ATP than is OxPhos, cancer
tion in the presence of oxygen) and has become cells must consume greater amounts of ferment-
known as aerobic glycolysis or the Warburg effect able fuels than normal cells in order to maintain
(Racker, 1972). Lactate production is greater in an adequate supply of ATP for viability (Seyfried
cancer cells than in normal cells, because the effi- and Shelton, 2010).
ciency of oxidative phosphorylation (OxPhos) is The increased consumption of glucose is
less in cancer cells than in normal cells. Reduced clearly seen with positron emission tomogra-
OxPhos efficiency arises from any type of defect phy (PET) scanning using radiolabeled glucose
in the number, structure, or function of mitochon- for detecting cancer in the body (Vander Heiden
dria (Arismendi-Morillo and Castellano-Ramirez, et al., 2009). Elevated glucose consumption leads
2008; Elliott et  al., 2012; Feichtinger et  al., 2015; to an increase in lactate production and acidifica-
Feichtinger et  al., 2010; Kiebish et  al., 2008; tion of the tumor cell microenvironment (Gatenby
Pedersen, 1978; Seyfried et al., 2014; Singh et al., and Gillies, 2004). Microenvironment acidifica-
2015; Srinivasan et al., 2015). Mitochondrial struc- tion increases inflammation and angiogenesis,
ture is linked directly to mitochondrial function which ultimately increases tumor progression.
(Lehninger, 1964). No malignant tumor tissue has Succinate, produced through amino acid fermen-
yet been found that contains mitochondria of nor- tation (Hochachka et  al., 1975), can also con-
mal number or structure when evaluated under tribute to microenvironment acidification and
electron microscopy. inflammation (Tannahill et  al., 2013). Hence, the
81

Chapter 12: Metabolism-Based Treatments to Counter Cancer 81

majority of cancer characteristics can be linked Several byproducts of amino acid fermentation
directly to mitochondrial dysfunction coupled can also accumulate in the tumor microenviron-
with increased glucose consumption for compen- ment including acetate, glutamate, alanine, suc-
satory fermentation. cinate, and ammonia. Although acetate has been
In addition to a reliance on glucose, cancer considered a potential fuel for supporting tumor
cells also depend heavily on glutamine for growth cell growth (Comerford et  al., 2014; Hosios and
and survival (DeBerardinis and Cheng, 2010; Vander Heiden, 2014), acetate levels are generally
Yuneva, 2008). Glutamine is anaplerotic and can low in the circulation (Ballard, 1972). Jaworski
be rapidly metabolized to glutamate and then to et  al. recently provided a comprehensive discus-
α-ketoglutarate for entry into the TCA cycle. In sion on the potential role of acetate in tumor
addition to serving as a carbon/nitrogen source for metabolism (Jaworski et  al., 2015). It should be
tumor cell growth, glutamine may also play a role in recognized that with the exception of glucose and
cancer cell survival and growth through enzymatic glutamine, none of the other potential fuels for
release of ammonia into the microenvironment tumor cell metabolism would be present in suffi-
(Huang et  al., 2013). Glucose and glutamine act ciently high quantities to maintain robust tumor
synergistically for driving rapid tumor cell growth. cell growth. Hence, the restriction of glucose and
Glutamine can produce some ATP under aerobic glutamine becomes of prime importance for tar-
conditions and can serve as fuel for lipid synthesis geting tumor cell growth and survival.
under hypoxic conditions (Ta and Seyfried, 2015).
In contrast to previous suggestions (DeBerardinis
et al., 2007), we found that only minor amounts of CALORIE RESTRICTION
glutamine are metabolized to lactate under either Calorie restriction (CR) with adequate nutrition or
normoxia or hypoxia in the VM-M3 invasive glio- underfeeding has been long known to limit tumor
blastoma cells (Ta and Seyfried, 2015). We suggest growth (Hursting et  al., 2010; Kritchevsky, 2001;
that glucose and glutamine can be either oxidized Mukherjee et  al., 1999; Tannenbaum, 1942). The
or fermented depending on the physiological state mechanisms by which CR reduces tumor growth
of the tumor microenvironment. Our findings have been described. Calorie restriction targets
suggest that glutamine targeting can be effective several of cancer’s hallmarks, including angio-
in managing systemic metastatic cancer (Shelton genesis, inflammation, edema, proliferation, and
et al., 2010b). distal invasion (Jiang and Wang, 2013; Mukherjee
Besides glucose and glutamine, other meta- et al., 1999; Mulrooney et al., 2011; Shelton et al.,
bolic fuels might also play a role in tumor cell 2010a; Thompson et al., 2004; Woolf et al., 2015).
metabolism. It has been shown recently that in Calorie restriction lowers the level of circulating
the presence of electron transport chain (ETC) glucose, the prime fermentable fuel for driving
abnormalities, aspartate can rescue cell prolifera- tumor growth (Seyfried et al., 2003). In addition,
tion without increasing ATP synthesis, suggesting CR or therapeutic fasting, elevates the level of
that the redox reactions of the ETC are required circulating ketone bodies, D-β-hydroxybutyrate
for survival (Sullivan et al., 2015). In cancer cells, and acetoacetate. Ketone bodies serve as a non-
this acts as a potential target for metabolic thera- fermentable super fuel for functional mitochon-
pies because aspartate is central to the de novo dria that also reduce reactive oxygen species
synthesis of purines and pyrimidines and pro- while increasing the ΔG’ of ATP hydrolysis and
teins, and for maintaining cellular redox balance. metabolic efficiency (Seyfried et al., 2014; Seyfried
While aspartate itself is not necessarily used for et  al., 2003; Veech, 2004). Based on these obser-
ATP synthesis, a number of amino acids such as vations, Cahill and Veech characterized ketone
serine, glycine, and the branched chain amino bodies as “Good Medicine” (Cahill and Veech,
acids can be catabolized to pyruvate, succinate, 2003). In contrast to normal cells, which transi-
and fumarate, all of which participate in the meta- tion to ketone bodies for energy when glucose
bolic pathways required for ATP generation and becomes limiting, tumor cells cannot use ketone
anaplerosis. Additionally, there is evidence sug- bodies effectively for energy (Fredericks and
gesting that in the absence of oxygen, Complex III Ramsey, 1978; Magee et  al., 1979; Maurer et  al.,
of the ETC can run in reverse, effectively reducing 2011; Tisdale, 1984). Ketone bodies can even be
fumarate to succinate, which will accumulate as a toxic to some tumor cells (Poff et al., 2015; Skinner
fermentation byproduct (Hochachka et al., 1975). et al., 2009). Shimazu et al. recently suggested that
This will maintain the redox balance and result in high-dose D-β-hydroxybutyrate could act as a his-
ATP synthesis. tone deacetylase inhibitor (Shimazu et  al., 2013).
82

82 section II: E merging Clinical Applications and Future Potential

The multiple defects in mitochondrial structure our failure to detect a therapeutic benefit against
and function are considered responsible for failure either naturally occurring seizures in EL mice, or
of tumor cells to effectively use ketone bodies for against astrocytoma growth in mice that were fed
energy (Seyfried and Mukherjee, 2005; Seyfried KDs in unrestricted amounts (Mantis et al., 2004;
and Shelton, 2010). Seyfried et al., 2003; Zhou et al., 2007).
Notwithstanding this growing experimental Reduced blood glucose is considered essential
literature, the therapeutic benefit of a 40% CR for managing either epileptic seizures or cancer.
for managing cancer in rodents would not be the As high-fat diets will elevate the appetite sup-
same as for managing cancer in humans. The basal pressor hormone, cholecystokinin, it is possible
metabolic rate in humans is about seven times less that dyslipidemia might not occur in most can-
than that of mice (Terpstra, 2001). Consequently, cer patients on a KD. A  therapeutic KD should
water-only therapeutic fasting for 2–3 weeks lower glucose, cholesterol, and triglycerides,
would be predicted to produce a therapeutic ben- while elevating ketone bodies and high-density
efit similar to that seen in rodents under 40% CR lipoprotein (Meidenbauer et al., 2014). The body
(Mahoney et al., 2006). As a ketogenic diet (KD) weight loss associated with KD is considered
can sometimes replicate the physiological condi- therapeutic, in contrast to the weight loss asso-
tions of therapeutic fasting (Seyfried et al., 2009), ciated with cachexia or toxic cancer therapies,
it will be easier for most cancer patients to use which is pathological. In contrast to the thera-
a KD than to use therapeutic fasting for tumor peutic shift in blood metabolites associated with
management. the KD weight loss, cachexic weight loss is asso-
ciated with elevations of blood glucose and insu-
T H E K E TO G E N I C   D I E T lin insensitivity that can stimulate tumor growth
The KD is a high-fat and low-carbohydrate diet (Kotler, 2000). High-dose steroids (dexametha-
that is widely used to reduce refractory epileptic sone), which elevate blood glucose levels, are
seizures in children (Freeman and Kossoff, 2010; often given to cancer patients undergoing che-
Kossoff and Hartman, 2012), and is also effective motherapy to reduce vomiting and to improve
in managing symptoms of a wide range of neuro- weight gain (Seyfried et al., 2015). It is clear from
logical and neurodegenerative diseases (Seyfried, an understanding of cancer metabolism that that
2014). The KD is also gaining recognition as an steroid administration for improved weight gain
effective therapy for any cancer that expresses would not be in the best interest of the cancer
the Warburg effect, which includes the majority patient (Wong et al., 2015). Hence, fundamental
of metastatic cancers. The anticancer mechanism metabolic differences exist between therapeutic
of action for the KD is potentially very simple. weight loss associated with a KD and the patho-
The KD lowers the prime fermentable fuel (glu- logical weight loss associated with cachexia and
cose) needed for driving the Warburg effect, while toxic cancer therapies.
elevating blood ketone bodies that require mito- The protein and fat composition of the KD dif-
chondrial function for effective metabolism. As fers from that of Atkins-type diets in having less
mitochondrial defects are ultimately responsible protein and more fat than the Atkins diets. This
for the reliance of tumor cells on fermentation, is important as several amino acids found in pro-
the KD targets the Warburg effect and tumor cell teins can be de-aminated to form pyruvate, which
energy metabolism. The KD can more effectively is then metabolized to form glucose through glu-
reduce glucose and elevate blood ketone bodies coneogenesis (Burt et  al., 1982). The fats in KDs
than can CR alone, making the KD potentially also contain more saturated medium chain triglyc-
more therapeutic against tumors than CR. There erides than do Atkins diets. Consequently, blood
are no anticancer drugs presently known that can glucose levels will be lower and ketone body lev-
target tumor cells while enhancing the metabolic els will be higher with KDs than with Atkins-type
efficiency of normal cells, as can the KD. diets. We recently developed the Glucose/Ketone
It is important to mention, however, that the Index calculator (GKIC) to assess the potential
unrestricted feeding of KDs can cause weight gain, therapeutic effects of various low-carbohydrate
insulin insensitivity, and dyslipidemia in some and KDs for cancer management (Meidenbauer
strains of mice, leading to elevated blood levels et al., 2015). The GKIC is a simple tool that mea-
of insulin and glucose (Meidenbauer et al., 2014). sures the ratio of blood glucose to blood ketones
Dyslipidemia from excessive consumption of KD and can help monitor the efficacy of metabolic
can occur despite the very low amounts of carbo- therapy in preclinical animal models and in clini-
hydrate in the diet. This could account in part for cal trials for malignant brain cancer or for any
83

Chapter 12: Metabolism-Based Treatments to Counter Cancer 83

cancer that expresses aerobic fermentation. The body β-D-hydroxybutyrate maintains the coen-
GKI can therefore serve as a biomarker to validate zyme Q couple in an oxidized state thus reducing
the therapeutic efficacy of various diets in target- the production of damaging ROS (Veech, 2004).
ing cancer cell energy metabolism. Therapeutic water-only fasting or restricted KDs
reduce tissue inflammation (Mulrooney et  al.,
THE FUTURE OF KDS 2011; Youm et al., 2015). Chronic inflammation is
FOR CANCER MANAGEMENT known to produce mitochondrial stress and can-
AND PREVENTION cer (Bissell and Hines, 2011; Coussens and Werb,
It is our view that reduced glucose with therapeu- 2002; Fosslien, 2008; Kamp et al., 2011). It is our
tic ketosis, achieved through low-carbohydrate contention that glucose/ketone ratios (GKI val-
KDs, can serve as an alternative or complemen- ues) of 1.0 or below could help reduce systemic
tary approach to cancer management. Evidence inflammation. The GKI might therefore serve as
also suggests that the therapeutic efficacy of the an effective biomarker, along with C-reactive pro-
KD against tumor growth can be enhanced when tein, for assessing systemic inflammation and for
combined with certain drugs and procedures reducing mitochondrial stress that can prevent
under a “press-pulse” paradigm as we previously cancer. Further studies will be needed to validate
mentioned (Seyfried et  al., 2014). For example, this concept.
therapeutic synergy was seen in combining the It has not escaped our attention that KDs could
calorie restricted KD with the glycolysis inhibitor, have a significant impact on global budgeting con-
2-deoxyglucose, for management of the syngeneic tracts for patient care. It is now recognized that
CT-2a glioma stem cell tumor (Marsh et al., 2008), global budget contracts with quality incentives
and in combining the KD with hyperbaric oxygen encourage changes in practice patterns that can
therapy and ketone esters for the management help reduce spending and improve quality of gen-
of systemic metastasis of the syngeneic VM-M3 eral health (Song et  al., 2014). The KDs are well
tumor (Poff et al., 2013; Poff et al., 2015). Scheck positioned to serve as a low-cost, nontoxic alterna-
and colleagues also showed that the KD could act tive for both the prevention and management of
synergistically with radiation therapy for man- cancer. In addition to cancer, we recently showed
agement of the GL261 mouse model of glioma how therapeutic ketosis could be effective for the
(Abdelwahab et al., 2012). management and prevention of a broad range of
The therapeutic effects of KDs seen in these chronic inflammatory diseases including obesity,
preclinical brain tumor studies have been corrobo- type 2 diabetes, cardiovascular disease, epilepsy,
rated in preclinical studies for several other can- Alzheimer’s disease, Parkinson’ s disease, and trau-
cer models including neuroblastoma, lung cancer, matic brain injury (Seyfried, 2014). Hence, KDs
prostate cancer, and breast and ovarian cancers could have utility for improving health and reduc-
(Allen et  al., 2013; Lv et  al., 2014; Mavropoulos ing costs for many of the most challenging diseases
et  al., 2009; Morscher et  al., 2015; Zhuang et  al., seen in Western societies.
2014). These preclinical studies are also motivat-
ing case reports and pilot studies in humans with AC K N OW L E D G M E N T S
brain cancer and other cancers (Champ et  al., This work was supported in part by the NIH,
2013; Champ et  al., 2014; Maroon et  al., 2015; Grants (HD-39722, NS-108055195, and CA-
Rieger et  al., 2014; Schmidt et  al., 2011; Zuccoli 102135); and grants from the American Institute
et al., 2010). It is clear from these studies, and from of Cancer Research; the Single Cause, Single Cure
the original observation of Linda Nebeling and Foundation; the George Yu Foundation; and the
colleagues (Nebeling et  al., 1995), that treatment Boston College Research Expense Fund.
of cancer patients with KDs is generally well toler-
ated, which is consistent with decades of research
REFERENCES
obtained from the evaluation of children treated
Abdelwahab, M.G., Fenton, K.E., Preul, M.C., Rho,
with KDs for epilepsy management. J.M., Lynch, A., Stafford, P., and Scheck, A.C.
In recognizing cancer as a mitochondrial (2012). The ketogenic diet is an effective adjuvant
metabolic disease, we suggested that protect- to radiation therapy for the treatment of malignant
ing cellular mitochondria from toxic or meta- glioma. PloS One 7, e36197.
bolic stress could best prevent cancer (Seyfried, Allen, B.G., Bhatia, S.K., Buatti, J.M., Brandt, K.E.,
2012b). Calorie-restricted KDs and therapeutic Lindholm, K.E., Button, A.M., Szweda, L.I.,
fasting are excellent ways to reduce oxidative Smith, B.J., Spitz, D.R., and Fath, M.A. (2013).
stress in mitochondria. Metabolism of the ketone Ketogenic diets enhance oxidative stress and
84

84 section II: E merging Clinical Applications and Future Potential

radio-chemo-therapy responses in lung cancer Coussens, L.M., and Werb, Z. (2002). Inflammation
xenografts. Clin Cancer Res 19, 3905–3913. and cancer. Nature 420, 860–867.
Arismendi-Morillo, G.J., and Castellano-Ramirez, Darlington, C.D. (1948). The plasmagene theory of the
A.V. (2008). Ultrastructural mitochondrial pathol- origin of cancer. Br J Cancer 2, 118–126.
ogy in human astrocytic tumors: potentials impli- DeBerardinis, R.J., Mancuso, A., Daikhin, E., Nissim,
cations pro-therapeutics strategies. J Electron I., Yudkoff, M., Wehrli, S., and Thompson, C.B.
Microsc (Tokyo) 57, 33–39. (2007). Beyond aerobic glycolysis:  transformed
Baker, S.G., and Kramer, B.S. (2007). Paradoxes in car- cells can engage in glutamine metabolism that
cinogenesis: new opportunities for research direc- exceeds the requirement for protein and nucleo-
tions. BMC Cancer 7, 151. tide synthesis. Proc Nat Acad Sci USA 104,
Ballard, F.J. (1972). Supply and utilization of acetate in 19345–19350.
mammals. Am J Clin Nutr 25, 773–779. DeBerardinis, R.J., and Cheng, T. (2010). Q’s next: the
Bartesaghi, S., Graziano, V., Galavotti, S., Henriquez, diverse functions of glutamine in metabolism, cell
N.V., Betts, J., Saxena, J., Minieri, V.A.D., biology and cancer. Oncogene 29, 313–324.
Karlsson, A., Martins, L.M., Capasso, M., et  al. Elliott, R.L., Jiang, X.P., and Head, J.F. (2012).
(2015). Inhibition of oxidative metabolism leads Mitochondria organelle transplantation:  intro-
to p53 genetic inactivation and transformation duction of normal epithelial mitochondria into
in neural stem cells. Proc Nat Acad Sci USA 112, human cancer cells inhibits proliferation and
1059–1064. increases drug sensitivity. Breast Cancer Res Treat
Bissell, M.J., and Hines, W.C. (2011). Why don’t we 136, 347–354.
get more cancer? A proposed role of the microen- Feichtinger, R.G., Zimmermann, F., Mayr, J.A.,
vironment in restraining cancer progression. Nat Neureiter, D., Hauser-Kronberger, C., Schilling,
Med 17, 320–329. F.H., Jones, N., Sperl, W., and Kofler, B. (2010).
Burgio, E., and Migliore, L. (2015). Towards a systemic Low aerobic mitochondrial energy metabolism in
paradigm in carcinogenesis:  linking epigenetics poorly- or undifferentiated neuroblastoma. BMC
and genetics. Mol Biol Rep 42, 777–790. Cancer 10, 149.
Burk, D., and Schade, A.L. (1956). On respiratory Feichtinger, R.G., Weis, S., Mayr, J.A., Zimmermann,
impairment in cancer cells. Science 124, 270–272. F.A., Bogner, B., Sperl, W., and Kofler, B. (2015).
Burk, D., Woods, M., and Hunter, J. (1967). On the Alterations of oxidative phosphorylation in
significance of glucolysis for cancer growth, with meningiomas and peripheral nerve sheath tumors.
special reference to Morris rat hepatomas. J Natl Neuro Oncol. 8(2):184–94.
Cancer Inst 38, 839–863. Fosslien, E. (2008). Cancer morphogenesis:  role
Burt, M.E., Gorschboth, C.M., and Brennan, M.F. of mitochondrial failure. Ann Clin Lab Sci 38,
(1982). A controlled, prospective, randomized 307–329.
trial evaluating the metabolic effects of enteral and Fredericks, M., and Ramsey, R.B. (1978). 3-Oxo acid
parenteral nutrition in the cancer patient. Cancer coenzyme A  transferase activity in brain and
49, 1092–1105. tumors of the nervous system. J Neurochem 31,
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? 1529–1531.
Good medicine? Trans Am Clin Climatol Assoc Freeman, J.M., and Kossoff, E.H. (2010). Ketosis
114, 149–161; discussion 162–143. and the ketogenic diet, 2010:  advances in treat-
Cairns, R.A. (2015). Drivers of the Warburg pheno- ing epilepsy and other disorders. Adv Pediatr 57,
type. Cancer J 21, 56–61. 315–329.
Cao, S., Zhang, C., and Xu, Y. (2015). Somatic muta- Gatenby, R.A., and Gillies, R.J. (2004). Why do can-
tions may not be the primary drivers of cancer for- cers have high aerobic glycolysis? Nat Rev Cancer
mation. Int J Cancer 137, 2762–2765. 4, 891–899.
Champ, C.E., Mishra, M.V., Showalter, T.N., Ohri, N., Hanahan, D., and Weinberg, R.A. (2000). The hall-
Dicker, A.P., and Simone, N.L. (2013). Dietary marks of cancer. Cell 100, 57–70.
recommendations during and after cancer treat- Hanahan, D., and Weinberg, R.A. (2011). Hallmarks
ment:  consistently inconsistent? Nutr Cancer 65, of cancer: the next generation. Cell 144, 646–674.
430–439. Hochachka, P.W., Owen, T.G., Allen, J.F., and Whittow,
Champ, C.E., Palmer, J.D., Volek, J.S., Werner-Wasik, G.C. (1975). Multiple end products of anaerobio-
M., Andrews, D.W., Evans, J.J., Glass, J., Kim, L., sis in diving vertebrates. Comp Biochem Physiol
and Shi, W. (2014). Targeting metabolism with a B 50, 17–22.
ketogenic diet during the treatment of glioblas- Hosios, A.M., and Vander Heiden, M.G. (2014).
toma multiforme. J Neurooncol 117, 125–131. Acetate metabolism in cancer cells. Cancer
Comerford, S.A., Huang, Z., Du, X., Wang, Y., Cai, L., Metab 2, 27.
Witkiewicz, A.K., Walters, H., Tantawy, M.N., Fu, Hou, J.P., and Ma, J. (2014). DawnRank:  discover-
A., Manning, H.C., et  al. (2014). Acetate depen- ing personalized driver genes in cancer. Genome
dence of tumors. Cell 159, 1591–1602. Med 6, 56.
85

Chapter 12: Metabolism-Based Treatments to Counter Cancer 85

Hu, Y., Lu, W., Chen, G., Wang, P., Chen, Z., Zhou, Mantis, J.G., Centeno, N.A., Todorova, M.T.,
Y., Ogasawara, M., Trachootham, D., Feng, L., McGowan, R., and Seyfried, T.N. (2004).
Pelicano, H., et al. (2012). K-ras(G12V) transfor- Management of multifactorial idiopathic epilepsy
mation leads to mitochondrial dysfunction and a in EL mice with caloric restriction and the keto-
metabolic switch from oxidative phosphorylation genic diet: role of glucose and ketone bodies. Nutr
to glycolysis. Cell Res 22, 399–412. Metab (Lond) 1, 11.
Huang, W., Choi, W., Chen, Y., Zhang, Q., Deng, H., Maroon, J.C., Seyfried, T.N., Donohue, J.P., and Bost,
He, W., and Shi, Y. (2013). A proposed role for glu- J. (2015). The role of metabolic therapy in treating
tamine in cancer cell growth through acid resis- glioblastoma multiforme. Surg Neurol Int 6, 61.
tance. Cell Res 23, 724–727. Marsh, J., Mukherjee, P., and Seyfried, T.N. (2008).
Hursting, S.D., Smith, S.M., Lashinger, L.M., Harvey, Drug/diet synergy for managing malignant
A.E., and Perkins, S.N. (2010). Calories and car- astrocytoma in mice:  2-deoxy-D-glucose and the
cinogenesis:  lessons learned from 30  years of restricted ketogenic diet. Nutr Metab (Lond) 5, 33.
calorie restriction research. Carcinogenesis 31, Maurer, G.D., Brucker, D.P., Baehr, O., Harter, P.N.,
83–89. Hattingen, E., Walenta, S., Mueller-Klieser, W.,
Jaworski, D.M., Namboodiri, A.M., and Moffett, Steinbach, J.P., and Rieger, J. (2011). Differential
J.R. (2015). Acetate as a metabolic and epigen- utilization of ketone bodies by neurons and glioma
etic modifier of cancer therapy. J Cell Biochem. cell lines: a rationale for ketogenic diet as experi-
117(3):574–88. mental glioma therapy. BMC Cancer 11, 315.
Jiang, Y.S., and Wang, F.R. (2013). Caloric restriction Mavropoulos, J.C., Buschemeyer, W.C., 3rd, Tewari,
reduces edema and prolongs survival in a mouse A.K., Rokhfeld, D., Pollak, M., Zhao, Y., Febbo,
glioma model. J Neurooncol 114, 25–32. P.G., Cohen, P., Hwang, D., et al. (2009). The effects
Kamp, D.W., Shacter, E., and Weitzman, S.A. (2011). of varying dietary carbohydrate and fat content
Chronic inflammation and cancer:  the role of on survival in a murine LNCaP prostate cancer
the mitochondria. Oncology (Williston Park) 25, xenograft model. Cancer Prev Res (Phila Pa) 2,
400–410, 413. 557–565.
Kiebish, M.A., Han, X., Cheng, H., Chuang, J.H., and McLeod, H.L. (2013). Cancer pharmacogenom-
Seyfried, T.N. (2008). Cardiolipin and electron ics:  early promise, but concerted effort needed.
transport chain abnormalities in mouse brain Science 339, 1563–1566.
tumor mitochondria: lipidomic evidence support- Meidenbauer, J.J., Mukherjee, P., and Seyfried, T.N.
ing the Warburg theory of cancer. J Lipid Res 49, (2015). The glucose ketone index calculator: a sim-
2545–2556. ple tool to monitor therapeutic efficacy for meta-
Koppenol, W.H., Bounds, P.L., and Dang, C.V. (2011). bolic management of brain cancer. Nutr Metab
Otto Warburg’s contributions to current con- (Lond) 12, 12.
cepts of cancer metabolism. Nat Rev Cancer 11, Meidenbauer, J.J., Ta, N., and Seyfried, T.N. (2014).
325–337. Influence of a ketogenic diet, fish-oil, and calorie
Kossoff, E.H., and Hartman, A.L. (2012). Ketogenic restriction on plasma metabolites and lipids in
diets:  new advances for metabolism-based thera- C57BL/6J mice. Nutr Metab 11, 23.
pies. Curr Opin Neurol. 25(2):173–8. Morscher, R.J., Aminzadeh-Gohari, S., Feichtinger,
Kotler, D.P. (2000). Cachexia. Ann Intern Med 133, R.G., Mayr, J.A., Lang, R., Neureiter, D., Sperl, W.,
622–634. and Kofler, B. (2015). Inhibition of neuroblastoma
Kritchevsky, D. (2001). Caloric restriction and cancer. tumor growth by ketogenic diet and/or calorie
J Nutr Sci Vitaminol (Tokyo) 47, 13–19. restriction in a CD1-Nu mouse model. PloS One
Lehninger, A.L. (1964). The Mitochondrion: Molecular 10, e0129802.
Basis of Structure and Function. (New  York: Mukherjee, P., Sotnikov, A.V., Mangian, H.J., Zhou,
Benjamin). J.R., Visek, W.J., and Clinton, S.K. (1999). Energy
Lv, M., Zhu, X., Wang, H., Wang, F., and Guan, W. intake and prostate tumor growth, angiogenesis,
(2014). Roles of caloric restriction, ketogenic diet and vascular endothelial growth factor expression.
and intermittent fasting during initiation, progres- J Natl Cancer Inst 91, 512–523.
sion and metastasis of cancer in animal models: a Mulrooney, T.J., Marsh, J., Urits, I., Seyfried, T.N., and
systematic review and meta-analysis. PloS One 9, Mukherjee, P. (2011). Influence of caloric restric-
e115147. tion on constitutive expression of NF-kappaB in
Magee, B.A., Potezny, N., Rofe, A.M., and Conyers, an experimental mouse astrocytoma. PloS One 6,
R.A. (1979). The inhibition of malignant cell e18085.
growth by ketone bodies. Aust J Exp Biol Med Sci Nebeling, L.C., Miraldi, F., Shurin, S.B., and Lerner, E.
57, 529–539. (1995). Effects of a ketogenic diet on tumor metab-
Mahoney, L.B., Denny, C.A., and Seyfried, T.N. (2006). olism and nutritional status in pediatric oncology
Caloric restriction in C57BL/6J mice mimics ther- patients:  two case reports. J Am Coll Nutr 14,
apeutic fasting in humans. Lipids Health Dis 5, 13. 202–208.
86

86 section II: E merging Clinical Applications and Future Potential

Pedersen, P.L. (1978). Tumor mitochondria and the Seyfried, T.N., Flores, R.E., Poff, A.M., and D’Agostino,
bioenergetics of cancer cells. Prog Exp Tumor Res D.P. (2014). Cancer as a metabolic disease: impli-
22, 190–274. cations for novel therapeutics. Carcinogenesis 35,
Poff, A.M., Ari, C., Seyfried, T.N., and D’Agostino, D.P. 515–527.
(2013). The ketogenic diet and hyperbaric oxygen Seyfried, T.N. (2015). Cancer as a mitochondrial meta-
therapy prolong survival in mice with systemic bolic disease. Front Cell Devel Biol 3, 43.
metastatic cancer. PloS One 8, e65522. Seyfried, T.N., Flores, R., Poff, A.M., D’Agostino, D.P.,
Poff, A.M., Ward, N., Seyfried, T.N., Arnold, P., and Mukherjee, P. (2015). Metabolic therapy:  a
and D’Agostino, D.P. (2015). Non-toxic meta- new paradigm for managing malignant brain can-
bolic management of metastatic cancer in VM cer. Cancer Lett 356, 289–300.
mice: Novel combination of ketogenic diet, ketone Shelton, L.M., Huysentruyt, L.C., Mukherjee, P., and
supplementation, and hyperbaric oxygen therapy. Seyfried, T.N. (2010a). Calorie restriction as an
PloS One 10, e0127407. anti-invasive therapy for malignant brain cancer
Racker, E. (1972). Bioenergetics and the problem of in the VM mouse. ASN Neuro 2, e00038.
tumor growth. Am Sci 60, 56–63. Shelton, L.M., Huysentruyt, L.C., and Seyfried, T.N.
Rieger, J., Bahr, O., Maurer, G.D., Hattingen, E., Franz, (2010b). Glutamine targeting inhibits systemic
K., Brucker, D., Walenta, S., Kammerer, U., Coy, metastasis in the VM-M3 murine tumor model.
J.F., Weller, M., et al. (2014). ERGO: a pilot study Int J Cancer 127, 2478–2485.
of ketogenic diet in recurrent glioblastoma. Int J Shimazu, T., Hirschey, M.D., Newman, J., He, W.,
Oncol 44, 1843–1852. Shirakawa, K., Le Moan, N., Grueter, C.A.,
Rous, P. (1959). Surmise and fact on the nature of can- Lim, H., Saunders, L.R., Stevens, R.D., et  al.
cer. Nature 183, 1357–1361. (2013). Suppression of oxidative stress by beta-
Schmidt, M., Pfetzer, N., Schwab, M., Strauss, I., and hydroxybutyrate, an endogenous histone deacety-
Kammerer, U. (2011). Effects of a ketogenic diet lase inhibitor. Science 339, 211–214.
on the quality of life in 16 patients with advanced Singh, L., Nag, T.C., and Kashyap, S. (2015).
cancer: A pilot trial. Nutr Metab 8, 54. Ultrastructural changes of mitochondria in
Seeger, P.G. (1959). [Cell respiration and cancer]. Das human retinoblastoma:  correlation with tumor
Deutsche Gesundheitswesen 14, 893–898. differentiation and invasiveness. Tumour Biol.
Seyfried, T.N., Sanderson, T.M., El-Abbadi, M.M., 37(5):5797–803.
McGowan, R., and Mukherjee, P. (2003). Role of Skinner, R., Trujillo, A., Ma, X., and Beierle, E.A.
glucose and ketone bodies in the metabolic con- (2009). Ketone bodies inhibit the viability of
trol of experimental brain cancer. Br J Cancer 89, human neuroblastoma cells. J Pediatr Surg 44,
1375–1382. 212–216; discussion 216.
Seyfried, T.N., and Mukherjee, P. (2005). Targeting Song, Z., Rose, S., Safran, D.G., Landon, B.E., Day, M.P.,
energy metabolism in brain cancer:  review and and Chernew, M.E. (2014). Changes in health care
hypothesis. Nutr Metab (Lond) 2, 30. spending and quality 4 years into global payment.
Seyfried, T.N., Mantis, J.G., Todorova, M.T., and Greene, N Engl J Med 371, 1704–1714.
A.E. (2009). Dietary management of epilepsy: role Sonnenschein, C., and Soto, A.M. (2000). Somatic
of glucose and ketone bodies. In Encyclopedia of mutation theory of carcinogenesis: why it should be
Basic Epilepsy Research, Vol. 2, P.A. Schwartzkroin, dropped and replaced. Mol Carcinog 29, 205–211.
ed. (Oxford: Academic Press), pp. 687–693. Soto, A.M., and Sonnenschein, C. (2004). The somatic
Seyfried, T.N., and Shelton, L.M. (2010). Cancer as a mutation theory of cancer: growing problems with
metabolic disease. Nutr Metab (Lond) 7, 7. the paradigm? Bioessays 26, 1097–1107.
Seyfried, T.N. (2012a). Cancer as a Metabolic Srinivasan, S., Guha, M., Dong, D.W., Whelan, K.A.,
Disease:  On the Origin, Management, and Ruthel, G., Uchikado, Y., Natsugoe, S., Nakagawa,
Prevention of Cancer (Hoboken, NJ: Wiley). H., and Avadhani, N.G. (2015). Disruption of cyto-
Seyfried, T.N. (2012b). Cancer prevention. In Cancer as chrome c oxidase function induces the Warburg
a Metabolic Disease: On the Origin, Management, effect and metabolic reprogramming. Oncogene.
and Prevention of Cancer (Hoboken, NJ: Wiley), 35(12):1585–95
pp. 375–386. Sullivan, L.B., Gui, D.Y., Hosios, A.M., Bush, L.N.,
Seyfried, T.N. (2012c). Mitochondrial respiratory Freinkman, E., and Vander Heiden, M.G. (2015).
dysfunction and the extrachromosomal origin of Supporting aspartate biosynthesis is an essential
cancer. In Cancer as a Metabolic Disease: On the function of respiration in proliferating cells. Cell
Origin, Management, and Prevention of Cancer 162, 552–563.
(Hoboken, NJ: Wiley), pp. 253–259. Ta, N.L., and Seyfried, T.N. (2015). Influence of
Seyfried, T.N. (2014). Ketone strong:  emerging evi- serum and hypoxia on incorporation of [C]-
dence for a therapeutic role of ketone bodies in D-glucose or [C]-L-glutamine into lipids and
neurological and neurodegenerative diseases. J lactate in murine glioblastoma cells. Lipids.
Lipid Res. 55(9):1815–7. 50(12):1167–84
87

Chapter 12: Metabolism-Based Treatments to Counter Cancer 87

Tannahill, G.M., Curtis, A.M., Adamik, J., Palsson- Warburg, O. (1969). Revised Lindau Lectures:  The
McDermott, E.M., McGettrick, A.F., Goel, G., Frezza, prime cause of cancer and prevention—Parts  1  &
C., Bernard, N.J., Kelly, B., Foley, N.H., et al. (2013). 2. In Meeting of the Nobel-Laureates D. Burk, ed.
Succinate is an inflammatory signal that induces IL- (Lindau, Lake Constance, Germany:  K.Triltsch),
1beta through HIF-1alpha. Nature 496, 238–242. pp. 1–9. http://www.hopeforcancer.com/OxyPlus.htm
Tannenbaum, A. (1942). The genesis and growth of Wong, E.T., Lok, E., Gautam, S., and Swanson,
tumors:  II. Effects of caloric restriction per se. K.D. (2015). Dexamethasone exerts profound
Cancer Res 2, 460–467. immunologic interference on treatment efficacy
Terpstra, A.H. (2001). Differences between humans for recurrent glioblastoma. Br J Cancer 113,
and mice in efficacy of the body fat lowering effect 232–241.
of conjugated linoleic acid: role of metabolic rate. J Woods, M.W., and du Buy, H.G. (1945). Cytoplasmic
Nutr 131, 2067–2068. diseases and cancer. Science 102, 591–593.
Thompson, H.J., McGinley, J.N., Spoelstra, N.S., Jiang, Woolf, E.C., Curley, K.L., Liu, Q., Turner, G.H.,
W., Zhu, Z., and Wolfe, P. (2004). Effect of dietary Charlton, J.A., Preul, M.C., and Scheck, A.C.
energy restriction on vascular density during mam- (2015). The ketogenic diet  alters the hypoxic
mary carcinogenesis. Cancer Res 64, 5643–5650. response and affects expression of proteins associ-
Tisdale, M.J. (1984). Role of acetoacetyl-CoA syn- ated with angiogenesis, invasive potential and vas-
thetase in acetoacetate utilization by tumor cells. cular permeability in a mouse glioma model. PloS
Cancer Biochem Biophys 7, 101–107. One 10, e0130357.
Tomasetti, C., and Vogelstein, B. (2015). Cancer eti- Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg, E.L.,
ology: variation in cancer risk among tissues can Bodogai, M., Kim, D., D’Agostino, D., Planavsky,
be explained by the number of stem cell divisions. N., Lupfer, C., Kanneganti, T.D., et al. (2015). The
Science 347, 78–81. ketone metabolite beta-hydroxybutyrate blocks
Vander Heiden, M.G., Cantley, L.C., and Thompson, NLRP3 inflammasome-mediated inflammatory
C.B. (2009). Understanding the Warburg disease. Nat Med 21, 263–269.
effect: the metabolic requirements of cell prolifera- Yuneva, M. (2008). Finding an “Achilles’ heel” of can-
tion. Science 324, 1029–1033. cer: the role of glucose and glutamine metabolism
Vaux, D.L. (2011). In defense of the somatic mutation in the survival of transformed cells. Cell Cycle 7,
theory of cancer. Bioessays 33, 341–343. 2083–2089.
Veech, R.L. (2004). The therapeutic implications of Zhou, W., Mukherjee, P., Kiebish, M.A., Markis, W.T.,
ketone bodies:  the effects of ketone bodies in Mantis, J.G., and Seyfried, T.N. (2007). The calo-
pathological conditions:  ketosis, ketogenic diet, rically restricted ketogenic diet, an effective alter-
redox states, insulin resistance, and mitochondral native therapy for malignant brain cancer. Nutr
metabolism. Prostaglandins Leukot Essent Fatty Metab (Lond) 4, 5.
Acids 70, 309–319. Zhuang, Y., Chan, D.K., Haugrud, A.B., and
Verschoor, M.L., Ungard, R., Harbottle, A., Jakupciak, Miskimins, W.K. (2014). Mechanisms by which
J.P., Parr, R.L., and Singh, G. (2013). Mitochondria low glucose enhances the cytotoxicity of metfor-
and cancer: past, present, and future. Biomed Res min to cancer cells both in vitro and in vivo. PloS
Int 2013, 612369. One 9, e108444.
Warburg, O. (1931). The Metabolism of Tumours Zuccoli, G., Marcello, N., Pisanello, A., Servadei, F.,
(New York: Richard R. Smith). Vaccaro, S., Mukherjee, P., and Seyfried, T.N.
Warburg, O. (1956a). On the origin of cancer cells. (2010). Metabolic management of glioblastoma
Science 123, 309–314. multiforme using standard therapy together with a
Warburg, O. (1956b). On the respiratory impairment restricted ketogenic diet: case report. Nutr Metab
in cancer cells. Science 124, 269–270. (Lond) 7, 33.
88

13
Ketogenic Diet as Adjunctive Therapy
for Malignant Brain Cancer
E R I C C . WO O L F, P H D A N D A D R I E N N E C . S C H E C K , P H D

INTRODUCTION but it often seemed overshadowed by discoveries


Human malignant glioma is a uniformly fatal of oncogenes, tumor suppressor genes, growth
disease due in part to the limitations of currently factor pathways, molecular subtypes of cancers,
available treatments, which include surgery, che- and so forth. There is a resurgence of interest in
motherapy, and radiation therapy. The average metabolism as a central theme in cancer, and we
survival of patients with glioblastoma multiforme continue to find that metabolic pathways intersect
(GBM) is 1.5  years. It is therefore of paramount and often regulate key components of tumor ini-
importance that new therapeutic strategies for tiation, progression and therapy response (Wolf
brain cancer patients be developed, especially et al., 2010; Nijsten and van Dam, 2009). In fact,
those that can enhance the efficacy of current altered metabolism itself has been referred to as
treatment options without damaging normal brain a hallmark of cancer (Cantor and Sabatini, 2012;
tissue. Advances in our understanding of the biol- Ward and Thompson, 2012), in addition to being
ogy of these tumors have led to an increase in the involved in virtually all of the cancer hallmarks
number of targeted therapies in preclinical and described in the seminal paper by Hanahan and
clinical trials (Nicholas et  al., 2011; Niyazi et  al., Weinberg (Hanahan and Weinberg, 2011)  (see
2011; Roesler et  al., 2010). While these therapies Figure 13.1; Lewis and Abdel-Haleem, 2013).
may prove somewhat effective, the heterogeneity The term “metabolic remodeling” has been
of this tumor often precludes the targeted mole- used to describe metabolic changes that can occur
cules from being found on all cells in the tumor, in cancer cells (Obre and Rossignol, 2015), and
thus reducing the efficacy of these treatments. In oncogene-associated pathways are now known to
contrast, one trait shared by virtually all tumor intersect with and alter metabolic pathways. For
cells is altered metabolism. example, the tumor suppressor protein p53, which
plays a pivotal role in the cellular responses to
T U M O R M E TA B O L I S M hypoxia, DNA damage, and oncogene activation,
Alterations in the metabolism of cancer cells, what is now known to regulate glycolysis and assist in
we now call the “Warburg effect” or aerobic glycol- maintaining mitochondrial integrity (Puzio-Kuter,
ysis, was first described by Otto Warburg in 1927 2011). Another important connection between
(Warburg et al., 1927). Cancer cells use glycolysis to metabolism and tumor growth is through regula-
provide energy and biomolecules regardless of the tion of c-MYC. Overexpression of c-MYC occurs
availability of oxygen. This results in the produc- in a wide variety of cancers, including gliomas. C-
tion of fewer ATP molecules per molecule of glu- MYC is a multifunctional transcription factor, and
cose, and thus tumor cells require large amounts of the list of its target genes include those involved in
glucose. This shift toward increased glycolytic flux both cell proliferation and cell metabolism (Miller
in the cytosol and away from the tricarboxylic acid et al., 2012). In addition to stimulating glycolysis,
cycle and oxidative phosphorylation in the mito- c-MYC has been found to activate glutaminoly-
chondria occurs very early in tumorigenesis. This sis and lipid synthesis from citrate (Obre and
allows for rapid cell proliferation even under con- Rossignol, 2015).
ditions of hypoxia and in the presence of dysfunc- With the advent of molecular analyses, studies
tional mitochondria. Since Warburg’s discovery, of growth factor pathways seemed to overshadow
metabolism has been of interest in the cancer field, the influence of metabolism on cancer growth.
89

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 89

mTOR Anabolism Perturbed


Enhanced serine/glycine Oxidative Glycolysis
glycolysis PI3K metabolism phosphorylation
inhibition
Akt
Ras HIF-1
PGM
r a t i ve Der
e
fe met gulate
HK-VDAC Proli ling abo d SCO2
Oxidative association signa lism
phosphorylation p53
inhibition defects
g

En licat
s
op lin

rep
si
ap isab

ha ion
to

nc
D

ed
NO
synthase
Glycosylation
VEGF
Angiogensis

Inflammation
HIF-1

Ang-2 NO
synthase
Perturbed
glycosylation IDH1

b il &
t a io n
it y
Im yste ion

ns utat
2-hydroxy-
s as
m m
ev

un

glutarate

M
Kynurenines i
e

FH
SHD
l
Eva cel
ding be d
g
su rowth Perturesion & Fumarate
Succinate
p pr a dh st a si s
Lactate e s s io n m et a HIF-1
Oxygen Acidification
independence of interstitium Prolyl 4-hydroxylases
Anaerobic
glycolysis Lactate

FIGURE  13.1 An illustration of the interconnectedness of tumor metabolism with Hanahan and Weinberg’s
hallmarks of cancer (Lewis and Abdel-Haleem, 2013).

Overactivation of the stress-responsive PI3K/AKT which is typically found in high-grade gliomas


signaling pathway is typical in many cancers and and other cancers. HIF-1 is a heterodimeric
often due to activation of growth factor signal- transcription factor that induces the transcrip-
ing pathways involved in glioma growth, such as tion of a variety of genes involved in angiogen-
platelet-derived growth factor, epidermal growth esis (vascular endothelial growth factor [VEGF]
factor, and insulin growth factor. We now know and other cytokines) in an attempt to improve
that these growth factor pathways are intertwined tissue perfusion. This results in the formation
with metabolic signaling pathways (Dibble and of abnormal blood vessels that can increase
Cantley, 2015; Courtnay et al., 2015; Roberts and inflammation and edema in brain tumors, as
Miyamoto, 2015; Martini et al., 2014; Iurlaro et al., well as induction of the transcription of a vari-
2014). The PI3K/AKT signaling pathway has been ety of genes that promote invasion, migration,
closely linked to metabolism, and under low glu- and tumor growth (Fischer et al., 2002; Fujiwara
cose conditions it results in rapid tumor cell death et  al., 2007; Hayashi et  al., 2007; Horing et  al.,
(Marie and Shinjo, 2011; Robey and Hay, 2009; 2012; Justus et al., 2015; Kaur et al., 2005; Masson
Yang et al., 2009). and Ratcliffe, 2014; Mou et al., 2010; Proescholdt
Another important “hub” linking metabolism et  al., 2012; Yang et  al., 2012). In addition to
and cancer is hypoxia-inducible factor 1 (HIF-1). specific actions that relate to the tumor cell’s
The expression of HIF-1 is activated by hypoxia, response to oxygen availability, HIF-1 interacts
90

90 section II: E merging Clinical Applications and Future Potential

with the PI3K/AKT signaling path to act as a reg- 2012; Robey et al., 2015; Gaude and Frezza, 2014;
ulator of cancer metabolism, proliferation, and Masson and Ratcliffe, 2014). The fact that meta-
glycolysis (Courtnay et  al., 2015; Justus et  al., bolic dysregulation is seen in virtually all tumor
2015; Wei et  al., 2013; Pore et  al., 2006). It also cells has led to suggestions that a promising thera-
affects the activation of nuclear factor–kappa B peutic strategy may be to exploit this feature. One
(NF-κB), a transcriptional activator that is cen- potential way to achieve this goal is through the
tral to the regulation of various signal transduc- use of the therapeutic ketogenic diet (KD) or phys-
tion pathways and to transcriptional activation iologically similar methods, such as caloric restric-
events that mediate inflammation, cell prolif- tion or intermittent fasting.
eration, cell migration, and angiogenesis. HIF-1
may, at least in part, provide the molecular basis T H E K E TO G E N I C   D I E T
for the Warburg effect by “reprograming” cel- The KD is more correctly referred to as “metabolic
lular metabolism in response to oxygen avail- therapy” rather than a “diet.” This high-fat, low-
ability (Corbet and Feron, 2015; Courtnay et al., carbohydrate diet is used to treat medically refrac-
2015). HIF-1 also is a central figure in alterations tory epilepsy (Kim and Rho, 2008; Cross, 2013) in
to the tumor microenvironment, which affects children and, more recently, in some adults. The
not only tumor cell growth but also response to diet is not without side effects; however, these are
therapy (Azzi et  al., 2013; Bayley and Devilee, typically readily managed when the patient has
2010; Bruzzese et  al., 2014; Casey et  al., 2015; appropriate supervision by a multidisciplinary
Danhier et  al., 2013; Dewhirst, 2009; Hattingen team (i.e., dietitian, nurse, and physician) skilled
et al., 2011; Horing et al., 2012; Hu et al., 2012; in its use. The KD has been shown to have neu-
Joon et  al., 2004; Justus et  al., 2015; Marie roprotective effects, and there are now studies to
and Shinjo, 2011; Masson and Ratcliffe, 2014; determine its efficacy for a number of neurological
Amberger-Murphy, 2009; Metallo et  al., 2011; disorders, including Alzheimer’s disease, traumatic
Semenza, 2013; Shweiki et  al., 1995; Stegeman brain injury, and amyotrophic lateral sclerosis
et al., 2014; Wei et al., 2011; Yamada et al., 1999; (Maalouf et al., 2009; Stafstrom and Rho, 2012).
Yang et al., 2012). The KD increases blood ketones and decreases
The molecular background of a tumor cell can blood glucose by simulating the physiological
also affect the regulation of the pathways already response to fasting, thus leading to high rates of
described. Loss of function of phosphatase and fatty acid oxidation and an increase in the produc-
tensin homolog (PTEN) or mutation of p53 also tion of acetyl coenzyme A  (acetyl-CoA). When
increase HIF-1, as does the accumulation of reac- the amount of acetyl-CoA exceeds the capacity of
tive oxygen species (ROS). ROS are multifaceted the tricarboxylic acid cycle to utilize it, there is an
effector molecules involved in numerous cellular increase in the production of the ketone bodies β-
pathways, including those regulating autophagic/ hydroxybutyrate (βHB) and acetoacetate (ACA),
apoptotic responses to genotoxic stress, hypoxia, which can be used as an energy source in the nor-
and nutrient deprivation. Cancer cells often have mal brain (Cahill and Veech, 2003; Morris, 2005;
increased levels of ROS (Fruehauf and Meyskens, Vanitallie and Nufert, 2003; Veech et  al., 2001;
2007), and they have been implicated in angio- Gasior et al., 2006). Since normal cells readily use
genesis induction and tumor growth through the ketones as an alternate energy source, they are
regulation of VEGF and HIF-1 (Weinberg and unlikely to be adversely affected by reduced glu-
Chandel, 2009). cose. In contrast, the metabolic alterations found
It is clear that cancer cell metabolism is far in cancer cells are generally thought to reduce
more complex than originally thought. A number their ability to be “flexible” regarding their pri-
of cancer-associated mutations affect metabolism, mary energy source, and thus they require glucose
and defects in mitochondria are seen in cancer (Seyfried et  al., 2011; Maurer et  al., 2011; Zhou
that also link metabolism with cancer initiation et al., 2007; Seyfried and Mukherjee, 2005; Tisdale
and progression. Although some of these interac- and Brennan, 1983; Fredericks and Ramsey, 1978;
tions are mentioned above, in-depth discussions Seyfried, 2012). The reduction in glucose that
of all of the interactions that occur between can- results from the KD essentially “starves” the tumor
cer and metabolism are beyond the scope of this cells and thus inhibits their growth (Baranano and
chapter and the reader is referred to a number of Hartman, 2008; Bozzetti and Zupec-Kania, 2015;
reviews on these subjects (Cantor and Sabatini, Freedland et  al., 2008; Klement, 2013; Maroon
2012; Gatenby and Gillies, 2004; Semenza, 2013; et  al., 2015; Seyfried et  al., 2009; Seyfried et  al.,
Vander Heiden et al., 2009; Ward and Thompson, 2015; Simone et  al., 2013; Wallace et  al., 2010).
91

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 91

Thus, when used as a therapy, the KD can take oxygen and ketone supplementation to demon-
advantage of the Warburg effect. strate reduced tumor cell growth and metastatic
In addition to the effects mediated by glucose spread in the VM metastatic tumor model (Poff
reduction, work in the epilepsy field and more et  al., 2015; Poff et  al., 2014). Scheck et  al. used
recent work in cancer research has shown that the syngeneic intracranial GL261-luc/albino C57/
the KD can exhibit antitumor effects even in the Bl6 model to demonstrate that caloric restriction
absence of glucose reduction. We demonstrated was not necessary for the antitumor effect of the
the effect of adding ketones to media containing KD (Stafford et al., 2010), particularly when a 4:1
glucose in vitro using the AO2V4 cell line (Scheck fat:carbohydrate plus protein formulation is used
et  al., 2012). This cell line was derived from a (Scheck et al., 2012).
recurrent human glioblastoma and is grown in The KD and similar diets used as a mono-
Waymouth’s MAB 87/3 media containing 28 mM therapy have a pluripotent effect on the growth
glucose and supplemented with 20% fetal calf of tumors both in vitro and in vivo. This may
serum. When 5 mM βHB plus 5 mM ACA was depend, at least in part, on the model system, the
added to complete media, cell growth was signifi- specific metabolic intervention, and the molecu-
cantly inhibited. When 1,3-bis(2-chloroethyl)-1 lar underpinnings of the tumor itself (Caso et al.,
nitrosourea (BCNU, carmustine), one of the che- 2013; Freedland et al., 2008; Hao et al., 2015; Kim
motherapeutic agents given to this patient prior et  al., 2012c; Lv et  al., 2014; Mavropoulos et  al.,
to tumor recurrence, was used in addition to 2009; Otto et al., 2008; Poff et al., 2015; Poff et al.,
ketones there was additional growth inhibition 2013; Simone et  al., 2013; Stafford et  al., 2010;
compared to either ketones or BCNU alone. More Woolf et  al., 2015). The striking feature of the
recent work has shown that the ketones themselves work done to date is that alterations in metabo-
exert antitumor effects separate from the effects of lism have a far-reaching effect on tumor cells,
reduced blood glucose (Magee et al., 1979; Scheck tumors, and the tumor microenvironment. Studies
et al., 2012; Skinner et al., 2009). The ketone βHB have shown reductions in growth rate as one
has recently been shown to inhibit histone deac- might expect; however, there are also changes in
tylases (HDACs), which can result in epigen- the formation of ROS and oxidative stress (Allen
etic suppression of the expression of a variety of et al., 2013; Stafford et al., 2010; Milder and Patel,
genes. Thus, ketones may provide a link between 2012), angiogenesis (Puchowicz et al., 2008; Woolf
metabolism and tumorigenesis, although the pre- et al., 2015; Seyfried et al., 2015; Zhou et al., 2007),
cise nature of these changes is as yet unknown hypoxia (Maurer et  al., 2011; Poff et  al., 2015;
(Newman and Verdin, 2014a, 2014b; Sassone- Woolf et al., 2015), inflammation and peritumoral
Corsi, 2013; Shimazu et al., 2013). The remainder edema (Kim et  al., 2012a; Mavropoulos et  al.,
of this chapter addresses the utility of increasing 2009; Woolf et al., 2015), metastasis and invasion
blood ketones and reducing blood glucose for the (Amann and Hellerbrand, 2009; Gluschnaider
treatment of brain tumors. et al., 2014; Hao et al., 2015; Lv et al., 2014; Otto
et al., 2008; Poff et al., 2015), and the expression of
C U R R E N T S TAT U S various transcriptional modulators such as NF-κB
O F   K N OW L E D G E (Woolf et al., 2015) and microRNAs (Wang et al.,
While the idea that metabolism may be a thera- 2015a; Wang et al., 2015c; Irani and Hussain, 2015;
peutic target for the treatment of cancer originated Bishop and Ferguson, 2015; Wang et  al., 2015b;
with Otto Warburg, there have been only a limited Chan et al., 2015).
number of case reports describing the use of a KD
for cancer therapy. Similarly, preclinical studies of Ketogenic Diet in Combination
efficacy and mechanisms of action were limited with Standard Therapies
until recently. Although evidence suggests that the KD provides
antitumor benefits on its own, perhaps the most
Preclinical Evidence effective use of the KD is in combination with
The use of metabolic alteration for the therapy of standard cancer therapies such as radiation and
brain tumors has been championed by Seyfried chemotherapy (Allen et al., 2014). The KD greatly
and colleagues. They used the VM (Shelton et al., enhanced survival in a mouse model of malignant
2010)  and CT-2A (Marsh et  al., 2008)  mouse glioma when combined with TMZ when compared
tumor models to show that a KD, especially when with either treatment alone (Figure 13.2) (Scheck
given in restricted amounts, extends survival. et  al., 2011). Using a bioluminescent, syngeneic
D’Agostino and colleagues have added hyperbaric intracranial model of malignant glioma, the KD
92

92 section II: E merging Clinical Applications and Future Potential

was shown to significantly potentiate the anti- with radiation and other anticancer therapeutics in
tumor effect of radiotherapy. In fact, 9 out of 11 both preclinical and clinical studies (Raffaghello
animals maintained on the KD and treated with et  al., 2008; Lee et  al., 2010; Seyfried et  al., 2012;
radiation had complete and sustained remission of Lee et al., 2012; Safdie et al., 2012; Saleh et al., 2013;
their implanted tumors, even after being switched Champ et  al., 2013; Champ et  al., 2014; Klement
back to a standard rodent diet (Figure 13.2) and Champ, 2014; Poff et  al., 2013; Raffaghello
(Abdelwahab et  al., 2012). Allen and colleagues et al., 2010).
reported similar results when the KD was com- The effectiveness of radiation therapy is due
bined with radiation and chemotherapy in a lung to a number of factors including relative damage
cancer xenograft model (Allen et  al., 2013). That done to tumor cells versus normal tissue and the
is, they found decreased tumor growth rate and ability of normal cells and tumor cells to repair the
increased survival. Calorie restriction and short- damage (Klement and Champ, 2014). Radiation
term fasting have also been found to be synergistic works, in part, by creating ROS through the radiol-
ysis of water. The ROS damage the DNA and other
macromolecules, causing sublethal damage that
(A) can become lethal if not repaired. The potentiation
SD
100 KetoCal® of radiation therapy by the KD or CR seems para-
p value (0.0049) doxical in light of our data demonstrating a reduc-
75
tion in ROS in tumors from animals maintained
50 on a KD (Stafford et al., 2010). However, radiation
effects do not only occur through ROS, and radia-
25 tion can directly damage DNA and other cellu-
lar macromolecules. Furthermore, in addition to
0 ROS, radiation causes the production of reactive
0 10 20 30 40 50 100 150
(B) nitrogen species (RNS), a potential source of mac-
Rad
100 KetoCal® + Rad romolecular damage following radiation (Saenko
p value (<0.0001) et al., 2013). Whether the KD or CR reduces the
75 formation of RNS is as yet unknown. In fact, the
main effect of the KD or CR may not be in alter-
50 ing the amount of radiation-induced damage, but
may in fact be in modulating the ability of tumor
25
and normal cells to repair radiation-induced dam-
0
age (Klement and Champ, 2014; Santivasi and Xia,
0 50 100 150 200 250 2014). Studies have shown that CR can enhance
(C) DNA repair in normal cells (Heydari et al., 2007);
TMZ
100 KetoCal® + TMZ however, this may not be the case in tumor cells,
p value (0.0025)
75 and the differential response of tumor cells and
normal cells to genotoxic stress may be mediated
50 by reduced IGF1 and glucose in the tumor cells.
We and others have shown that insulin-like growth
25 factor is reduced in animals maintained on a KD
(Freedland et al., 2008; Klement and Champ, 2014;
0 Mavropoulos et  al., 2009; Scheck et  al., 2012). In
0 10 20 30 40 50
addition, a number of studies have shown that
FIGURE  13.2 Kaplan-Meier survival plot of animals reduced activation of the PI3K/Akt pathway, acti-
implanted intracranially with GL261-luc2 malignant vation of the adenosine monophosphate-activated
glioma cells and (A) maintained on KetoCal® (KC, the 4:1 protein kinase (AMPK) signaling pathway, and
fat:carbohydrate plus protein formulation of the ketogenic reduction of receptor tyrosine kinase growth fac-
diet) versus standard diet (SD); (B) treated with 2 × 4 Gy tor pathways can all reduce radioresistance in
radiation versus KC plus radiation, and (C) treated with tumor cells (Zhang et al., 2014; Choi et al., 2014;
50 mg/kg temozolomide (TMZ) versus KC plus TMZ. Medova et  al., 2013; Gil Del Alcazar et  al., 2014;
Animals on KC survived significantly longer when treated Munshi and Ramesh, 2013; Li et  al., 2014; Sanli
with KC alone, when KC was combined with radiation, et al., 2014; Danhier et al., 2013; Wang et al., 2013).
and when KC was combined with TMZ (Abdelwahab Finally, ketones and the KD have been shown
et al., 2012; Scheck et al., 2011). to affect the immune system (Rahman et al.,
93

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 93

2014; Husain et al., 2013; Kim et al., 2012b; Youm was put on a 4:1 (ratio of fats:carbohydrate plus
et al., 2015), and we have shown that the KD also protein) calorie-restricted (600 kcal/day) KD
reverses tumor-mediated immune suppression during radiation and chemotherapy. During this
in a mouse model of malignant glioma (Lussier, time, her body weight dropped by 20%, she had
Woolf et al. 2016). As radiation-induced tumor reduced blood glucose, increased urinary ketones,
killing is known to expose the immune system to and, most importantly, no observable brain tumor
a greater diversity of tumor antigens, it is possible detectable by either fluoro-deoxyglucose positron
that the KD as an adjuvant works to augment the emission tomography (FDG-PET) or magnetic
effect of radiation in part by enhancing immunity resonance imaging (MRI). The tumor recurred 10
against GBM. weeks after the patient resumed her normal eat-
The variety of effects seen when glucose is low- ing habits, and she succumbed to her disease less
ered and/or ketones are increased suggests that than 2 years after diagnosis. While this patient did
this may also potentiate other therapies, including not experience long-term tumor control after ces-
newer immune- and targeted therapies. Concerns sation of the diet, this report demonstrated that
that potentiation of the antitumor effect of a par- the diet could be tolerated, even when used in a
ticular therapy may also increase its effect on nor- calorie-restricted setting.
mal brain are valid. However, we and others have Results of a phase 1 clinical trial were reported
shown that the gene expression changes seen in in 2011 by a German group (Schmidt et al., 2011).
tumor are different from those seen in normal Tolerability of a restricted calorie KD was tested in
brain (Stafford et  al., 2010; Chang et  al., 2013; 16 patients with a variety of advanced (end-stage)
Maurer et  al., 2011). Further, the KD is known cancers. There were no severe side effects, and 5 of
to have neuroprotective effects (Lund et al., 2009; the 16 patients were able to complete the 3-month
Maalouf et  al., 2009; Hartman, 2012; Puchowicz treatment. These five patients had stable disease
et al., 2008), and thus it has been postulated that while on the diet. Two of the 11 remaining patients
this may actually help to protect the normal brain died early following the beginning of the trial, one
from the deleterious effects of radio- and chemo- was unable to tolerate the diet and dropped out
therapy. Taken together, the preclinical data pro- immediately, two patients dropped out for per-
vides strong support for the clinical use of the KD sonal reasons, one was unable to continue the diet
or CR as an adjuvant therapy for the treatment of for more than a month, three had disease progres-
gliomas and other cancers. sion within less than 2 months of starting the diet,
and one dropped out to resume chemotherapy.
K E TO G E N I C D I E T While this trial demonstrated tolerability and a
IN HUMANS favorable side-effect profile, the antitumor efficacy
Studies of glucose utilization in cancer go back could not be assessed due to the variety and sever-
prior to the 1980s, including studies of metabo- ity of disease in the patients. Recently, Schwartz
lism and cancer cachexia (Fearon et  al., 1988; et  al. reported on two patients with recurrent
Tisdale et al., 1987). These and other studies sug- GBM treated with a calorie-restricted KD as
gested that a KD consisting of a high percentage monotherapy, and although the diet was tolerated,
of medium chain triglycerides (MCT) along with both patients showed tumor progression—the first
various supplements resulted in weight gain and within 4 weeks and the second within 12 weeks
improved nitrogen balance in both animals and of beginning the protocol (Schwartz et al., 2015).
humans. In 1995, Nebeling and colleagues pub- More recently, a number of prospective clinical
lished a case report in which they used a similar trials have been initiated which have been sum-
KD based on MCT oil to treat two female pediat- marized in Table 13.1. These trials include studies
ric patients with advanced stage malignant brain of up-front treatment using the KD in addition to
tumors (Nebeling and Lerner, 1995; Nebeling et al., standard radiation and chemotherapy in patients
1995). They demonstrated that dietary-induced diagnosed with GBM.
ketosis decreased the availability of glucose to The case reports described above, along with
the tumor without causing a decrease in patient numerous anecdotal reports, suggest that the KD
weight or overall nutritional status. Furthermore, may be a promising anticancer therapy. However,
both children had long-term tumor management more work is needed to determine how to best
(Nebeling et  al., 1995). The second case report utilize the KD and other metabolic therapies for
was published in 2010 by Zuccoli and coworkers the treatment of tumors. Most of the informa-
(Zuccoli et al., 2010). This patient was a 65-year- tion regarding the most effective way to use the
old female with a multicentric glioblastoma. She KD comes from the epilepsy literature. Further
94

94 section II: E merging Clinical Applications and Future Potential

research is needed to determine optimum blood present in these tumors has suggested a host of
ketone and glucose levels for anticancer effects. targets for the development of new treatments,
In addition, a variety of KDs are used for seizure targeted therapies tried to date have met with
control, and it is not clear whether one or more limited success. This is at least in part due to the
of the different formulations will provide the best molecular heterogeneity of these tumors that
results for cancer patients. Finally, while the KD prevents any one target from being present in all
has a long record of safety in the epilepsy com- cells. In contrast, metabolic dysregulation is pres-
munity, side effects that occur when used in com- ent in virtually all tumor cells and there is rapidly
bination with cancer therapies may differ in type increasing interest in using metabolic therapies
or severity. These data will come from carefully such as the KD for the treatment of various can-
controlled clinical trials that include input from cers, especially brain tumors. Preclinical data
healthcare professionals well versed in the use have demonstrated that the antitumor effects
of the KD. Furthermore, patient enrollment into of the KD and CR are multifaceted, and altera-
clinical trials requires “buy-in” from the medical tions in energy metabolism can inhibit cancer
community. Physicians must be educated on the cell growth and increase the tumor’s response
therapeutic benefits of metabolic alteration as an to therapy. This provides a strong impetus to
adjuvant therapy. As with any decision regarding continue work designed to elucidate the mecha-
therapy, the patient’s overall condition, including nisms through which the KD exerts its antican-
nutritional status, must be taken into account. As cer effects, as well as suggesting the need for the
suggested by Klement and Champ (2014), can- design of controlled clinical trials that will shed
cer patients should be comprehensively assessed light on the most effective ways to implement
for nutritional needs and tolerability of such metabolic therapies in combination with stan-
interventions. dard therapies for the treatment of malignant dis-
Concern about patients’ quality of life is ease. This is a novel therapeutic paradigm, and we
sometimes given as a reason not to employ the have only begun to scratch the surface in terms of
KD. Compliance can be made more difficult by its potential.
the use of steroids (prescribed for peritumoral
edema) that often increase hunger and raise REFERENCES
blood glucose levels. To address this, at least one Abdelwahab, M.G., Fenton, K.E., Preul, M.C., Rho,
clinical trial (NCT02046187) includes an analy- J.M., Lynch, A., Stafford, P., and Scheck, A.C.
sis of both patient and caregiver quality of life. (2012). The ketogenic diet is an effective adjuvant
Quality of life measurements are being added to radiation therapy for the treatment of malignant
to more clinical trials, as the importance of this glioma. PLoS One 7, e36197.
has become recognized at the national level Allen, B.G., Bhatia, S.K., Buatti, J.M., Brandt, K.E.,
(Dirven et  al., 2014; Boele et  al., 2013; van den Lindholm, K.E., Button, A.M., Szweda, L.I., Smith,
B., Spitz, D.R., and Fath, M.A. (2013). Ketogenic
Bent et al., 2011). While some clinicians are con-
diets enhance oxidative stress and radio-chemo-
cerned compliance will reduce quality of life, the therapy responses in lung cancer xenografts. Clin
patients that do remain on the KD often com- Cancer Res 19, 3905–3913.
ment that this allows them to participate in their Allen, B.G., Bhatia, S.K., Anderson, C.M.,
own therapy. Despite these caveats, the existing Eichenberger-Gilmore, J.M., Sibenaller, Z.A.,
preclinical data suggesting antitumor efficacy Mapuskar, K.A., Schoenfeld, J.D., Buatti, J.M.,
and a synergistic effect with standard therapies Spitz, D.R., and Fath, M.A. (2014). Ketogenic diets
provides a strong impetus to conduct controlled as an adjuvant cancer therapy:  HISTORY and
clinical trials, particularly those that will shed potential mechanism. Redox Biology 2C, 963–970.
light on the interactions between the KD and Amann, T., and Hellerbrand, C. (2009). GLUT1 as a
other therapies. therapeutic target in hepatocellular carcinoma.
Expert Opin Ther Targets 13, 1411–1427.
CONCLUSION Amberger-Murphy, V. (2009). Hypoxia helps glioma
to fight therapy. Curr Cancer Drug Targets 9,
Improvements in the survival and quality of life
381–390.
for patients with malignant brain tumors require
Azzi, S., Hebda, J.K., and Gavard, J. (2013). Vascular
the implementation of new therapeutic modali- permeability and drug delivery in cancers. Front
ties, especially those that increase the efficacy of Oncol 3, 211.
current therapies without increasing toxic side Baranano, K.W., and Hartman, A.L. (2008). The keto-
effects. While the rapid accumulation of data genic diet:  uses in epilepsy and other neurologic
defining the molecular and genetic aberrations illnesses. Curr Treat Options Neurol 10, 410–419.
95

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 95

Bayley, J.P., and Devilee, P. (2010). Warburg tumours signaling, HSP90 and histone deacetylases. BMC
and the mechanisms of mitochondrial tumour Cancer 14, 17.
suppressor genes: barking up the right tree? Curr Corbet, C., and Feron, O. (2015). Metabolic and mind
Opin Genet Dev 20, 324–329. shifts:  from glucose to glutamine and acetate
Bishop, K.S., and Ferguson, L.R. (2015). The interac- addictions in cancer. Curr Opin Clin Nutr Metab
tion between epigenetics, nutrition and the devel- Care 18, 346–353.
opment of cancer. Nutrients 7, 922–947. Courtnay, R., Ngo, D.C., Malik, N., Ververis, K.,
Boele, F.W., Heimans, J.J., Aaronson, N.K., Taphoorn, Tortorella, S.M., and Karagiannis, T.C. (2015).
M.J., Postma, T.J., Reijneveld, J.C., and Klein, M. Cancer metabolism and the Warburg effect:  the
(2013). Health-related quality of life of significant role of HIF-1 and PI3K. Mol Biol Rep 42, 841–851.
others of patients with malignant CNS versus non- Cross, J.H. (2013). New research with diets and epi-
CNS tumors:  a comparative study. J Neurooncol lepsy. J Child Neurol 28, 970–974.
115, 87–94. Danhier, P., De Saedeleer, C.J., Karroum, O., De,
Bozzetti, F., and Zupec-Kania, B. (2015). Toward a P.G., Porporato, P.E., Jordan, B.F., Gallez, B.,
cancer-specific diet. Clin Nutr 10. and Sonveaux, P. (2013). Optimization of tumor
Bruzzese, L., Fromonot, J., By, Y., Durand-Gorde, J.M., radiotherapy with modulators of cell metabo-
Condo, J., Kipson, N., Guieu, R., Fenouillet, E., lism:  toward clinical applications. Semin Radiat
and Ruf, J. (2014). NF-kappaB enhances hypoxia- Oncol 23, 262–272.
driven T-cell immunosuppression via upregula- Dewhirst, M.W. (2009). Relationships between cycling
tion of adenosine A(2A) receptors. Cell Signal 26, hypoxia, HIF-1, angiogenesis and oxidative stress.
1060–1067. Radiat Res 172, 653–665.
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? Dibble, C.C., and Cantley, L.C. (2015). Regulation of
Good medicine? Trans Am Clin Climatol Assoc mTORC1 by PI3K signaling. Trends Cell Biol 25,
114, 149–161. 545–555.
Cantor, J.R., and Sabatini, D.M. (2012). Cancer cell Dirven, L., Taphoorn, M.J., Reijneveld, J.C., Blazeby,
metabolism:  one hallmark, many faces. Cancer J., Jacobs, M., Pusic, A., La, S.E., Stupp, R., Fayers,
Discov 2, 881–898. P., and Efficace, F. (2014). The level of patient-
Casey, S.C., Amedei, A., Aquilano, K., Azmi, A.S., reported outcome reporting in randomised con-
Benencia, F., Bhakta, D., Bilsland, A.E., Boosani, trolled trials of brain tumour patients: a systematic
C.S., Chen, S., Ciriolo, M.R., et al. (2015). Cancer review. Eur J Cancer 50, 2432–2448.
prevention and therapy through the modulation Fearon, K.C., Borland, W., Preston, T., Tisdale, M.J.,
of the tumor microenvironment. Semin Cancer Shenkin, A., and Calman, K.C. (1988). Cancer
Biol 35, S199–223. cachexia: influence of systemic ketosis on substrate
Caso, J., Masko, E.M., Ii, J.A., Poulton, S.H., Dewhirst, levels and nitrogen metabolism. Am J Clin Nutr
M., Pizzo, S.V., and Freedland, S.J. (2013). The 47, 42–48.
effect of carbohydrate restriction on prostate can- Fischer, S., Wobben, M., Marti, H.H., Renz, D., and
cer tumor growth in a castrate mouse xenograft Schaper, W. (2002). Hypoxia-induced hyperper-
model. Prostate 73, 449–454. meability in brain microvessel endothelial cells
Champ, C.E., Baserga, R., Mishra, M.V., Jin, L., Sotgia, involves VEGF-mediated changes in the expression
F., Lisanti, M.P., Pestell, R.G., Dicker, A.P., and of zonula occludens-1. Microvasc Res 63, 70–80.
Simone, N.L. (2013). Nutrient restriction and Fredericks, M., and Ramsey, R.B. (1978). 3-Oxo acid
radiation therapy for cancer treatment: when less coenzyme A  transferase activity in brain and
is more. Oncologist 18, 97–103. tumors of the nervous system. J. Neurochem. 31,
Champ, C.E., Palmer, J.D., Volek, J.S., Werner-Wasik, 1529–1531.
M., Andrews, D.W., Evans, J.J., Glass, J., Kim, L., Freedland, S.J., Mavropoulos, J., Wang, A., Darshan,
and Shi, W. (2014). Targeting metabolism with a M., Demark-Wahnefried, W., Aronson, W.J.,
ketogenic diet during the treatment of glioblas- Cohen, P., Hwang, D., Peterson, B., Fields, T., et al.
toma multiforme. J Neurooncol 117, 125–131. (2008). Carbohydrate restriction, prostate cancer
Chan, B., Manley, J., Lee, J., and Singh, S.R. (2015). The growth, and the insulin-like growth factor axis.
emerging roles of microRNAs in cancer metabo- Prostate 68, 11–19.
lism. Cancer Lett 356, 301–308. Fruehauf, J.P., and Meyskens, F.L., Jr. (2007). Reactive
Chang, H.T., Olson, L.K., and Schwartz, K.A. (2013). oxygen species:  a breath of life or death? Clin
Ketolytic and glycolytic enzymatic expression pro- Cancer Res 13, 789–794.
files in malignant gliomas:  implication for keto- Fujiwara, S., Nakagawa, K., Harada, H., Nagato, S.,
genic diet therapy. Nutr Metab (Lond) 10, 47. Furukawa, K., Teraoka, M., Seno, T., Oka, K., Iwata,
Choi, E.J., Cho, B.J., Lee, D.J., Hwang, Y.H., Chun, S., and Ohnishi, T. (2007). Silencing hypoxia-
S.H., Kim, H.H., and Kim, I.A. (2014). Enhanced inducible factor-1alpha inhibits cell migration and
cytotoxic effect of radiation and temozolomide in invasion under hypoxic environment in malignant
malignant glioma cells: targeting PI3K-AKT-mTOR gliomas. Int J Oncol 30, 793–802.
96

96 section II: E merging Clinical Applications and Future Potential

Gasior, M., Rogawski, M.A., and Hartman, A.L. Husain, Z., Huang, Y., Seth, P., and Sukhatme, V.P.
(2006). Neuroprotective and disease-modifying (2013). Tumor-derived lactate modifies antitu-
effects of the ketogenic diet. Behav Pharmacol 17, mor immune response: effect on myeloid-derived
431–439. suppressor cells and NK cells. J Immunol 191,
Gatenby, R.A., and Gillies, R.J. (2004). Why do can- 1486–1495.
cers have high aerobic glycolysis? Nat Rev Cancer Irani, S., and Hussain, M.M. (2015). Role of
4, 891–899. microRNA-30c in lipid metabolism, adipogenesis,
Gaude, E., and Frezza, C. (2014). Defects in mito- cardiac remodeling and cancer. Curr Opin Lipidol
chondrial metabolism and cancer. Cancer Metab 26, 139–146.
2, 10–12. Iurlaro, R., Leon-Annicchiarico, C.L., and Munoz-
Gil Del Alcazar, C.R., Hardebeck, M.C., Mukherjee, B., Pinedo, C. (2014). Regulation of cancer metab-
Tomimatsu, N., Gao, X., Yan, J., Xie, X.J., Bachoo, olism by oncogenes and tumor suppressors.
R., Li, L., Habib, A.A., et al. (2014). Inhibition of Methods Enzymol 542, 59–80.
DNA double-strand break repair by the dual PI3K/ Joon, Y.A., Bazar, K.A., and Lee, P.Y. (2004). Tumors
mTOR inhibitor NVP-BEZ235 as a strategy for may modulate host immunity partly through
radiosensitization of glioblastoma. Clin Cancer hypoxia-induced sympathetic bias. Med
Res 20, 1235–1248. Hypotheses 63, 352–356.
Gluschnaider, U., Hertz, R., Ohayon, S., Smeir, E., Justus, C.R., Sanderlin, E.J., and Yang, L.V. (2015).
Smets, M., Pikarsky, E., and Bar-Tana, J. (2014). Molecular connections between cancer cell metab-
Long-chain fatty acid analogues suppress breast olism and the tumor microenvironment. Int J Mol
tumorigenesis and progression. Cancer Res 74, Sci 16, 11055–11086.
6991–7002. Kaur, B., Khwaja, F.W., Severson, E.A., Matheny, S.L.,
Hanahan, D., and Weinberg, R.A. (2011). Hallmarks Brat, D.J., and Van Meir, E.G. (2005). Hypoxia and
of cancer: the next generation. Cell 144, 646–674. the hypoxia-inducible-factor pathway in glioma
Hao, G.W., Chen, Y.S., He, D.M., Wang, H.Y., Wu, growth and angiogenesis. Neuro Oncol 7, 134–153.
G.H., and Zhang, B. (2015). Growth of human Kim, D.Y., and Rho, J.M. (2008). The ketogenic diet
colon cancer cells in nude mice is delayed by keto- and epilepsy. Curr Opin Clin Nutr Metab Care 11,
genic diet with or without omega-3 fatty acids and 113–120.
medium-chain triglycerides. Asian Pac J Cancer Kim, D.Y., Hao, J., Liu, R., Turner, G., Shi, F.D., and
Prev 16, 2061–2068. Rho, J.M. (2012a). Inflammation-mediated mem-
Hartman, A.L. (2012). Neuroprotection in metabolism- ory dysfunction and effects of a ketogenic diet in
based therapy. Epilepsy Res 100, 286–294. a murine model of multiple sclerosis. PLoS One 7,
Hattingen, E., Jurcoane, A., Bahr, O., Rieger, J., e35476.
Magerkurth, J., Anti, S., Steinbach, J.P., and Kim, D.Y., Hao, J., Liu, R., Turner, G., Shi, F.D., and
Pilatus, U. (2011). Bevacizumab impairs oxidative Rho, J.M. (2012b). Inflammation-mediated mem-
energy metabolism and shows antitumoral effects ory dysfunction and effects of a ketogenic diet in a
in recurrent glioblastomas: a 31P/1H MRSI and murine model of multiple sclerosis. PLoS One 7,
quantitative magnetic resonance imaging study. e35476.
Neuro Oncol 13, 1349–1363. Kim, H.S., Masko, E.M., Poulton, S.L., Kennedy, K.M.,
Hayashi, Y., Edwards, N.A., Proescholdt, M.A., Pizzo, S.V., Dewhirst, M.W., and Freedland, S.J.
Oldfield, E.H., and Merrill, M.J. (2007). Regulation (2012c). Carbohydrate restriction and lactate
and function of aquaporin-1 in glioma cells. transporter inhibition in a mouse xenograft model
Neoplasia 9, 777–787. of human prostate cancer. BJU Int 110, 1062–1069.
Heydari, A.R., Unnikrishnan, A., Lucente, L.V., Klement, R.J. (2013). Calorie or carbohydrate restric-
and Richardson, A. (2007). Caloric restriction tion? The ketogenic diet as another option for sup-
and genomic stability. Nucleic Acids Res 35, portive cancer treatment. Oncologist 18, 1056.
7485–7496. Klement, R.J., and Champ, C.E. (2014). Calories,
Horing, E., Harter, P.N., Seznec, J., Schittenhelm, J., carbohydrates, and cancer therapy with radia-
Buhring, H.J., Bhattacharyya, S., von Hattingen, tion: exploiting the five R’s through dietary manip-
E., Zachskorn, C., Mittelbronn, M., and Naumann, ulation. Cancer Metastasis Rev 33, 217–229.
U. (2012). The “go or grow” potential of gliomas is Lee, C., Safdie, F.M., Raffaghello, L., Wei, M., Madia,
linked to the neuropeptide processing enzyme car- F., Parrella, E., Hwang, D., Cohen, P., Bianchi, G.,
boxypeptidase E and mediated by metabolic stress. and Longo, V.D. (2010). Reduced levels of IGF-I
Acta Neuropathol 124, 83–97. mediate differential protection of normal and can-
Hu, Y.L., DeLay, M., Jahangiri, A., Molinaro, A.M., cer cells in response to fasting and improve che-
Rose, S.D., Carbonell, W.S., and Aghi, M.K. (2012). motherapeutic index. Cancer Res 70, 1564–1572.
Hypoxia-induced autophagy promotes tumor cell Lee, C., Raffaghello, L., Brandhorst, S., Safdie, F.M.,
survival and adaptation to antiangiogenic treat- Bianchi, G., Martin-Montalvo, A., Pistoia, V., Wei,
ment in glioblastoma. Cancer Res 72, 1773–1783. M., Hwang, S., Merlino, A., et  al. (2012). Fasting
97

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 97

cycles retard growth of tumors and sensitize a as experimental glioma therapy. BMC Cancer
range of cancer cell types to chemotherapy. Sci 11:315, 315.
Transl Med 4, 124ra27. Mavropoulos, J.C., Buschemeyer, W.C., III, Tewari,
Lewis, N.E., and Abdel-Haleem, A.M. (2013). The evo- A.K., Rokhfeld, D., Pollak, M., Zhao, Y., Febbo,
lution of genome-scale models of cancer metabo- P.G., Cohen, P., Hwang, D., Devi, G., et al. (2009).
lism. Front Physiol 4, 237. The effects of varying dietary carbohydrate and fat
Li, W., Guo, F., Wang, P., Hong, S., and Zhang, C. content on survival in a murine LNCaP prostate
(2014). miR-221/222 confers radioresistance in cancer xenograft model. Cancer Prev Res (Phila)
glioblastoma cells through activating Akt indepen- 2, 557–565.
dent of PTEN status. Curr Mol Med 14, 185–195. Medova, M., Aebersold, D.M., and Zimmer, Y.
Lussier, D. M., et al. (2016). Enhanced immunity in a (2013). The molecular crosstalk between the MET
mouse model of malignant glioma is mediated by receptor tyrosine kinase and the DNA damage
a therapeutic ketogenic diet. BMC Cancer. May 13: response-biological and clinical aspects. Cancers
310. (Basel) 6, 1–27.
Lund, T.M., Risa, O., Sonnewald, U., Schousboe, A., Metallo, C.M., Gameiro, P.A., Bell, E.L., Mattaini, K.R.,
and Waagepetersen, H.S. (2009). Availability of Yang, J., Hiller, K., Jewell, C.M., Johnson, Z.R.,
neurotransmitter glutamate is diminished when Irvine, D.J., Guarente, L., et  al. (2011). Reductive
beta-hydroxybutyrate replaces glucose in cultured glutamine metabolism by IDH1 mediates lipogen-
neurons. J Neurochem 110, 80–91. esis under hypoxia. Nature 481, 380–384.
Lv, M., Zhu, X., Wang, H., Wang, F., and Guan, W. Milder, J., and Patel, M. (2012). Modulation of oxida-
(2014). Roles of caloric restriction, ketogenic diet tive stress and mitochondrial function by the keto-
and intermittent fasting during initiation, progres- genic diet. Epilepsy Res 100, 295–303.
sion and metastasis of cancer in animal models: a Miller, D.M., Thomas, S.D., Islam, A., Muench, D., and
systematic review and meta-analysis. PLoS One 9, Sedoris, K. (2012). c-Myc and cancer metabolism.
e115147. Clin Cancer Res 18, 5546–5553.
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The Morris, A.A.M. (2005). Cerebral ketone body metabo-
neuroprotective properties of calorie restriction, lism. J Inherit Metab Dis 28, 109–121.
the ketogenic diet, and ketone bodies. Brain Res Mou, K., Chen, M., Mao, Q., Wang, P., Ni, R., Xia, X.,
Rev 59, 293–315. and Liu, Y. (2010). AQP-4 in peritumoral edema-
Magee, B.A., Potezny, N., Rofe, A.M., and Conyers, tous tissue is correlated with the degree of glioma
R.A. (1979). The inhibition of malignant cell and with expression of VEGF and HIF-alpha. J
growth by ketone bodies. Aust J Exp Biol Med Sci Neurooncol 100, 375–383.
57, 529–539. Munshi, A., and Ramesh, R. (2013). Mitogen-activated
Marie, S.K., and Shinjo, S.M. (2011). Metabolism protein kinases and their role in radiation response.
and brain cancer. Clinics (Sao Paulo). 66(Suppl Genes Cancer 4, 401–408.
1), 33–43. Nebeling, L.C., and Lerner, E. (1995). Implementing a
Maroon, J.C., Seyfried, T.N., Donohue, J.P., and Bost, ketogenic diet based on medium-chain triglycer-
J. (2015). The role of metabolic therapy in treat- ide oil in pediatric patients with cancer. J Am Diet
ing glioblastoma multiforme. Surg Neurol Int 6:61. Assoc 95, 693–697.
doi:  10.4103/2152-7806.155259. eCollection;%20 Nebeling, L.C., Miraldi, F., Shurin, S.B., and Lerner, E.
15., 61–7806 (1995). Effects of a ketogenic diet on tumor metabo-
Marsh, J., Mukherjee, P., and Seyfried, T.N. (2008). lism and nutritional status in pediatric oncology
Drug/diet synergy for managing malignant patients: two case reports. J Am Coll Nutr 14, 202–208.
astrocytoma in mice:  2-deoxy-D-glucose and Newman, J.C., and Verdin, E. (2014a). Beta-
the restricted ketogenic diet. Nutr Metab (Lond) hydroxybutyrate:  much more than a metabolite.
5:33, 33. Diabetes Res Clin Pract 106, 173–181.
Martini, M., De Santis, M.C., Braccini, L., Gulluni, F., Newman, J.C., and Verdin, E. (2014b). Ketone bodies
and Hirsch, E. (2014). PI3K/AKT signaling path- as signaling metabolites. Trends Endocrinol Metab
way and cancer: an updated review. Ann Med 46, 25, 42–52.
372–383. Nicholas, M.K., Lukas, R.V., Chmura, S., Yamini, B.,
Masson, N., and Ratcliffe, P.J. (2014). Hypoxia signal- Lesniak, M., and Pytel, P. (2011). Molecular het-
ing pathways in cancer metabolism:  the impor- erogeneity in glioblastoma: therapeutic opportuni-
tance of co-selecting interconnected physiological ties and challenges. Semin Oncol 38, 243–253.
pathways. Cancer Metab 2, 3. Nijsten, M.W., and van Dam, G.M. (2009).
Maurer, G.D., Brucker, D.P., Bahr, O., Harter, P.N., Hypothesis: using the Warburg effect against can-
Hattingen, E., Walenta, S., Mueller-Klieser, W., cer by reducing glucose and providing lactate. Med
Steinbach, J.P., and Rieger, J. (2011). Differential Hypotheses 73, 48–51.
utilization of ketone bodies by neurons and Niyazi, M., Siefert, A., Schwarz, S.B., Ganswindt,
glioma cell lines:  a rationale for ketogenic diet U., Kreth, F.W., Tonn, J.C., and Belka, C. (2011).
98

98 section II: E merging Clinical Applications and Future Potential

Therapeutic options for recurrent malignant gli- (2014). The beta-hydroxybutyrate receptor HCA2
oma. Radiother Oncol 98, 1–14. activates a neuroprotective subset of macrophages.
Obre, E., and Rossignol, R. (2015). Emerging concepts Nat Commun 5, 3944.
in bioenergetics and cancer research:  metabolic Roberts, D.J., and Miyamoto, S. (2015). Hexokinase II
flexibility, coupling, symbiosis, switch, oxida- integrates energy metabolism and cellular protec-
tive tumors, metabolic remodeling, signaling tion:  Akting on mitochondria and TORCing to
and bioenergetic therapy. Int J Biochem Cell Biol autophagy. Cell Death Differ 22, 248–257.
59C:167–181. doi: 10.1016/j.biocel.2014.12.008. Robey, R.B., and Hay, N. (2009). Is Akt the “Warburg
Otto, C., Kaemmerer, U., Illert, B., Muehling, B., kinase”?-Akt-energy metabolism interactions and
Pfetzer, N., Wittig, R., Voelker, H.U., Thiede, A., oncogenesis. Semin Cancer Biol 19, 25–31.
and Coy, J.F. (2008). Growth of human gastric Robey, R.B., Weisz, J., Kuemmerle, N.B., Salzberg,
cancer cells in nude mice is delayed by a ketogenic A.C., Berg, A., Brown, D.G., Kubik, L., Palorini,
diet supplemented with omega-3 fatty acids and R., Al-Mulla, F., Al-Temaimi, R., et  al. (2015).
medium-chain triglycerides. BMC Cancer 8, 122. Metabolic reprogramming and dysregulated
Poff, A.M., Ari, C., Seyfried, T.N., and D’Agostino, D.P. metabolism: cause, consequence and/or enabler of
(2013). The ketogenic diet and hyperbaric oxygen environmental carcinogenesis? Carcinogenesis 36,
therapy prolong survival in mice with systemic S203–S231.
metastatic cancer. PLoS One 8, e65522. Roesler, R., Brunetto, A.T., Abujamra, A.L., de Farias,
Poff, A.M., Ari, C., Arnold, P., Seyfried, T.N., and C.B., Brunetto, A.L., and Schwartsmann, G. (2010).
D’Agostino, D.P. (2014). Ketone supplementation Current and emerging molecular targets in glioma.
decreases tumor cell viability and prolongs sur- Expert Rev Anticancer Ther 10, 1735–1751.
vival of mice with metastatic cancer. Int J Cancer Saenko, Y., Cieslar-Pobuda, A., Skonieczna, M., and
135, 1711–1720. Rzeszowska-Wolny, J. (2013). Changes of reac-
Poff, A.M., Ward, N., Seyfried, T.N., Arnold, P., tive oxygen and nitrogen species and mitochon-
and D’Agostino, D.P. (2015). Non-toxic meta- drial functioning in human K562 and HL60 cells
bolic management of metastatic cancer in vm exposed to ionizing radiation. Radiat Res 180,
mice: novel combination of ketogenic diet, ketone 360–366.
supplementation, and hyperbaric oxygen therapy. Safdie, F., Brandhorst, S., Wei, M., Wang, W., Lee, C.,
PLoS One 10, e0127407. Hwang, S., Conti, P.S., Chen, T.C., and Longo, V.D.
Pore, N., Jiang, Z., Shu, H.K., Bernhard, E., Kao, G.D., (2012). Fasting enhances the response of glioma to
and Maity, A. (2006). Akt1 activation can aug- chemo- and radiotherapy. PLoS One 7, e44603.
ment hypoxia-inducible factor-1alpha expression Saleh, A.D., Simone, B.A., Palazzo, J., Savage, J.E.,
by increasing protein translation through a mam- Sano, Y., Dan, T., Jin, L., Champ, C.E., Zhao, S.,
malian target of rapamycin-independent pathway. Lim, M., et al. (2013). Caloric restriction augments
Mol Cancer Res 4, 471–479. radiation efficacy in breast cancer. Cell Cycle 12,
Proescholdt, M.A., Merrill, M.J., Stoerr, E.M., 1955–1963.
Lohmeier, A., Pohl, F., and Brawanski, A. (2012). Sanli, T., Steinberg, G.R., Singh, G., and Tsakiridis, T.
Function of carbonic anhydrase IX in glioblastoma (2014). AMP-activated protein kinase (AMPK)
multiforme. Neuro Oncol 14, 1357–1366. beyond metabolism: a novel genomic stress sensor
Puchowicz, M.A., Zechel, J.L., Valerio, J., Emancipator, participating in the DNA damage response path-
D.S., Xu, K., Pundik, S., LaManna, J.C., and Lust, way. Cancer Biol Ther 15, 156–169.
W.D. (2008). Neuroprotection in diet-induced Santivasi, W.L., and Xia, F. (2014). Ionizing radia-
ketotic rat brain after focal ischemia. J Cereb Blood tion-induced DNA damage, response, and repair.
Flow Metab 28, 1907–1916. Antioxid Redox Signal 21, 251–259.
Puzio-Kuter, A.M. (2011). The role of p53 in metabolic Sassone-Corsi, P. (2013). Physiology: when metabolism
regulation. Genes Cancer 2, 385–391. and epigenetics converge. Science 339, 148–150.
Raffaghello, L., Lee, C., Safdie, F.M., Wei, M., Madia, Scheck, A. C., Abdelwahab, M. G., Stafford, P., Kim,
F., Bianchi, G., and Longo, V.D. (2008). Starvation- D. Y., Iwai, S., Preul, M. C., and Rho, J. M. (2011).
dependent differential stress resistance protects Mechanistic studies of the ketogenic diet as an
normal but not cancer cells against high-dose adjuvant therapy for malignant gliomas. Cancer
chemotherapy. Proc Natl Acad Sci USA 105, Res 70[8 Suppl.], 638.
8215–8220. Scheck, A.C., Abdelwahab, M.G., Fenton, K., and
Raffaghello, L., Safdie, F., Bianchi, G., Dorff, T., Stafford, P. (2012). The ketogenic diet for the treat-
Fontana, L., and Longo, V.D. (2010). Fasting and ment of glioma:  insights from genetic profiling.
differential chemotherapy protection in patients. Epilepsy Res 100, 327–337.
Cell Cycle 9, 4474–4476. Schmidt, M., Pfetzer, N., Schwab, M., Strauss, I., and
Rahman, M., Muhammad, S., Khan, M.A., Chen, Kammerer, U. (2011). Effects of a ketogenic diet on
H., Ridder, D.A., Muller-Fielitz, H., Pokorna, B., the quality of life in 16 patients with advanced can-
Vollbrandt, T., Stolting, I., Nadrowitz, R., et  al. cer: a pilot trial. Nutr Metab (Lond) 8, 54.
99

Chapter 13: Ketogenic Diet as Adjunctive Therapy for Malignant Brain Cancer 99

Schwartz, K., Chang, H.T., Nikolai, M., Pernicone, ketogenic diet reverses gene expression patterns
J., Rhee, S., Olson, K., Kurniali, P.C., Hord, N.G., and reduces reactive oxygen species levels when
and Noel, M. (2015). Treatment of glioma patients used as an adjuvant therapy for glioma. Nutr
with ketogenic diets:  report of two cases treated Metab (Lond) 7, 74.
with an IRB-approved energy-restricted ketogenic Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic
diet protocol and review of the literature. Cancer diet as a treatment paradigm for diverse neuro-
Metab 3, eCollection 2015. logical disorders. Front Pharmacol 3:59.
Semenza, G.L. (2013). HIF-1 mediates metabolic Stegeman, H., Span, P.N., Kaanders, J.H., and Bussink,
responses to intratumoral hypoxia and oncogenic J. (2014). Improving chemoradiation efficacy
mutations. J Clin Invest 123, 3664–3671. by PI3-K/AKT inhibition. Cancer Treat Rev 40,
Seyfried, T.N., and Mukherjee, P. (2005). Targeting 1182–1191.
energy metabolism in brain cancer:  review and Tisdale, M.J., and Brennan, R.A. (1983). Loss of aceto-
hypothesis. Nutr Metab 2, 30–38. acetate coenzyme A transferase activity in tumours
Seyfried, B.T., Kiebish, M., Marsh, J., and Mukherjee, of peripheral tissues. Br J Cancer 47, 293–297.
P. (2009). Targeting energy metabolism in brain Tisdale, M.J., Brennan, R.A., and Fearon, K.C. (1987).
cancer through calorie restriction and the keto- Reduction of weight loss and tumour size in a
genic diet. J Cancer Res Ther 5(Suppl 1):S7–15. cachexia model by a high fat diet. Br J Cancer
Seyfried, T.N., Kiebish, M.A., Marsh, J., Shelton, L.M., 56, 39–43.
Huysentruyt, L.C., and Mukherjee, P. (2011). van den Bent, M.J., Wefel, J.S., Schiff, D., Taphoorn, M.J.,
Metabolic management of brain cancer. Biochim Jaeckle, K., Junck, L., Armstrong, T., Choucair, A.,
Biophys Acta 1807, 577–594. Waldman, A.D., Gorlia, T., et  al. (2011). Response
Seyfried, T.N. (2012). Cancer as a Metabolic Disease: assessment in neuro-oncology (a report of the
On the Origin, Management and Prevention of RANO group): assessment of outcome in trials of dif-
Cancer (Hoboken, NJ: Wiley). fuse low-grade gliomas. Lancet Oncol 12, 583–593.
Seyfried, T.N., Marsh, J., Shelton, L.M., Huysentruyt, Vander Heiden, M.G., Cantley, L.C., and Thompson,
L.C., and Mukherjee, P. (2012). Is the restricted C.B. (2009). Understanding the Warburg
ketogenic diet a viable alternative to the standard effect: the metabolic requirements of cell prolifera-
of care for managing malignant brain cancer? tion. Science 1029–1033.
Epilepsy Res 100, 310–326. Vanitallie, T.B., and Nufert, T.H. (2003).
Seyfried, T.N., Flores, R., Poff, A.M., D’Agostino, D.P., Ketones: metabolism’s ugly duckling. Nutr Rev 61,
and Mukherjee, P. (2015). Metabolic therapy:  a 327–341.
new paradigm for managing malignant brain can- Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A.,
cer. Cancer Lett 356, 289–300. and Cahill, G.F., Jr. (2001). Ketone bodies, poten-
Shelton, L.M., Huysentruyt, L.C., Mukherjee, P., and tial therapeutic uses. IUBMB Life 51, 241–247.
Seyfried, T.N. (2010). Calorie restriction as an Wallace, D.C., Fan, W., and Procaccio, V. (2010).
anti-invasive therapy for malignant brain cancer Mitochondrial energetics and therapeutics. Annu
in the VM mouse. ASN Neuro 2, e00038. Rev Pathol 5:297–348, 297–348.
Shimazu, T., Hirschey, M.D., Newman, J., He, Wang, G., Wang, J., Zhao, H., Wang, J., and Tony To
W., Shirakawa, K., Le, M.N., Grueter, C.A., SS (2015a). The role of Myc and let-7a in glioblas-
Lim, H., Saunders, L.R., Stevens, R.D., et  al. toma, glucose metabolism and response to ther-
(2013). Suppression of oxidative stress by beta- apy. Arch Biochem Biophys 580, 84–92.
hydroxybutyrate, an endogenous histone deacety- Wang, H., Xu, T., Jiang, Y., Yan, Y., Qin, R., and Chen, J.
lase inhibitor. Science 339, 211–214. (2015b). MicroRNAs in human glioblastoma: from
Shweiki, D., Neeman, M., Itin, A., and Keshet, E. bench to beside. Front Biosci (Landmark Ed) 20,
(1995). Induction of vascular endothelial growth 105–118.
factor expression by hypoxia and by glucose defi- Wang, T., Wang, G., Hao, D., Liu, X., Wang, D., Ning,
ciency in multicell spheroids:  implications for N., and Li, X. (2015c). Aberrant regulation of the
tumor angiogenesis. Proc Natl Acad Sci USA 92, LIN28A/LIN28B and let-7 loop in human malig-
768–772. nant tumors and its effects on the hallmarks of
Simone, B.A., Champ, C.E., Rosenberg, A.L., Berger, cancer. Mol Cancer 14, 125.
A.C., Monti, D.A., Dicker, A.P., and Simone, N.L. Wang, Y., Yuan, J.L., Zhang, Y.T., Ma, J.J., Xu, P., Shi,
(2013). Selectively starving cancer cells through C.H., Zhang, W., Li, Y.M., Fu, Q., Zhu, G.F., et al.
dietary manipulation: methods and clinical impli- (2013). Inhibition of both EGFR and IGF1R
cations. Future Oncol 9, 959–976. sensitized prostate cancer cells to radiation by
Skinner, R., Trujillo, A., Ma, X., and Beierle, E.A. (2009). synergistic suppression of DNA homologous
Ketone bodies inhibit the viability of human neuro- recombination repair. PLoS One 8, e68784.
blastoma cells. J Pediatr Surg 44, 212–216. Warburg, O., Wind, F., and Negelein, E. (1927). The
Stafford, P., Abdelwahab, M.G., Kim, D.Y., Preul, metabolism of tumors in the body. J Gen Physiol
M.C., Rho, J.M., and Scheck, A.C. (2010). The 8, 519–530.
100

100 section II: E merging Clinical Applications and Future Potential

Ward, P.S., and Thompson, C.B. (2012). Metabolic Glioblastoma cells require glutamate dehydroge-
reprogramming: a cancer hallmark even Warburg nase to survive impairments of glucose metabo-
did not anticipate. Cancer Cell 21, 297–308. lism or Akt signaling. Cancer Res 69, 7986–7993.
Wei, J., Wu, A., Kong, L.Y., Wang, Y., Fuller, G., Fokt, Yang, L., Lin, C., Wang, L., Guo, H., and Wang, X.
I., Melillo, G., Priebe, W., and Heimberger, A.B. (2012). Hypoxia and hypoxia-inducible fac-
(2011). Hypoxia potentiates glioma-mediated tors in glioblastoma multiforme progression
immunosuppression. PLoS One 6, e16195. and therapeutic implications. Exp Cell Res 318,
Wei, W., Shi, Q., Remacle, F., Qin, L., Shackelford, 2417–2426.
D.B., Shin, Y.S., Mischel, P.S., Levine, R.D., and Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg,
Heath, J.R. (2013). Hypoxia induces a phase tran- E.L., Bodogai, M., Kim, D., D’Agostino, D.,
sition within a kinase signaling network in cancer Planavsky, N., Lupfer, C., Kanneganti, T.D.,
cells. Proc Natl Acad Sci USA 110, E1352–E1360. et  al. (2015). The ketone metabolite beta-
Weinberg, F., and Chandel, N.S. (2009). Reactive oxy- hydroxybutyrate blocks NLRP3 inflammasome-
gen species-dependent signaling regulates cancer. mediated inflammatory disease. Nat Med 21,
Cell Mol Life Sci 66, 3663–3673. 263–269.
Wolf, A., Agnihotri, S., and Guha, A. (2010). Targeting Zhang, H., Gu, C., Yu, J., Wang, Z., Yuan, X., Yang,
metabolic remodeling in glioblastoma multiforme. L., Wang, J., Jia, Y., Liu, J., and Liu, F. (2014).
Oncotarget 1, 552–562. Radiosensitization of glioma cells by TP53-
Woolf, E.C., Curley, K.L., Liu, Q., Turner, G.H., induced glycolysis and apoptosis regulator
Charlton, J.A., Preul, M.C., and Scheck, A.C. knockdown is dependent on thioredoxin-1
(2015). The ketogenic diet  alters the hypoxic nuclear translocation. Free Radic Biol Med 69C,
response and affects expression of proteins associ- 239–248.
ated with angiogenesis, invasive potential and vas- Zhou, W., Mukherjee, P., Kiebish, M.A., Markis, W.T.,
cular permeability in a mouse glioma model. PLoS Mantis, J.G., and Seyfried, T.N. (2007). The calo-
One 10, e0130357. rically restricted ketogenic diet, an effective alter-
Yamada, M., Tomida, A., Yun, J., Cai, B., Yoshikawa, native therapy for malignant brain cancer. Nutr
H., Taketani, Y., and Tsuruo, T. (1999). Cellular Metab (Lond) 4, 5.
sensitization to cisplatin and carboplatin with Zuccoli, G., Marcello, N., Pisanello, A., Servadei, F.,
decreased removal of platinum-DNA adduct Vaccaro, S., Mukherjee, P., and Seyfried, T.N.
by glucose-regulated stress. Cancer Chemother (2010). Metabolic management of glioblastoma
Pharmacol 44, 59–64. multiforme using standard therapy together with a
Yang, C., Sudderth, J., Dang, T., Bachoo, R.G., restricted ketogenic diet: case report. Nutr Metab
McDonald, J.G., and DeBerardinis, R.J. (2009). 7, 33–53.
101

14
Metabolic Therapy for Autism Spectrum
Disorder and Comorbidities
N I N G C H E N G, P H D, S U S A N A . M A S I N O, P H D, A N D J O N G M . R H O, M D

OV E RV I E W O F   AU T I S M Disease Control and Prevention in 2014. For con-


S P E C T RU M D I S O R D E R troversial reasons, the prevalence of ASD appears
Autism spectrum disorder (hereafter referred to as to be on the rise (a 10-fold increase in 40 years),
“ASD”) is a group of complex disorders of neurode- which may be explained only partly by improved
velopment. It is characterized by persistent deficits diagnosis and awareness (Hansen et  al., 2015).
in social communication and interaction across Developmental delay in ASD can be detected as
multiple contexts, as well as restricted and repeti- early as 6  months of age, a critical time for the
tive patterns of behavior and interests (DiCicco- development of higher-order social, emotional,
Bloom et al., 2006; Lai et al., 2014; Llaneza et al., and communications functions (Courchesne et al.,
2010). Further, it is well known that ASD is a mul- 2007), and increasingly, the importance of early
tiorgan disorder that can affect the normal func- intervention is being recognized (Orinstein et al.,
tion of the gastrointestinal, immune, hepatic, and 2014). However, on average, children identified
endocrine systems (Frye et al., 2015; Goines and with ASD were not diagnosed until after 4 years of
Van de Water, 2010; Hsiao, 2013; Mayer et al., 2015; age (CDC, 2014), even though patients can be reli-
Patterson, 2011). The term “spectrum” refers to the ably diagnosed as early as 2 years of age (Kleinman
wide range of symptoms and levels of impairment et al., 2008; Lord et al., 2006).
that can occur in individuals with ASD. Severity Because ASD has broad and heterogeneous
of language impairment of the patients, for exam- manifestations and has been associated with a
ple, can range from deficits in pragmatic use of plethora of possible etiological factors that are
language to complete lack of spoken language. both genetic and environmental, ASD remains a
Another level of heterogeneity is the existence of clinical syndrome, but not a specific etiologically
many comorbidities that include both neurologic defined disorder. The prominent heterogeneity in
conditions such as epilepsy, sleep impairment, autism has made it a challenge to investigate its
delays and deficits in motor functions, and psychi- neurobiological mechanisms and find interven-
atric dysfunctions including depression, anxiety, tions for the affected individuals. Currently, only
irritability, attention deficit hyperactivity disorder, comorbid manifestations of the disorder can be
as well as physical health issues such as gastroin- alleviated, such as epileptic seizures, psychiatric
testinal disturbances. The co-occurrence rate of disturbances, hyperactivity, sleep disturbances,
one or more non-ASD developmental diagnoses and digestive issues, but not the core symptoms
is as high as 83% (Levy et al., 2010). By contrast, (DiCicco-Bloom et  al., 2006; Lai et  al., 2014;
some people with ASD show extraordinary talents Llaneza et al., 2010).
in math, music, and art. Despite much research, there remains limited
According to epidemiological studies, ASD knowledge about the pathophysiological mecha-
occurs in all racial, ethnic, and socioeconomic nisms underlying ASD. It is generally accepted
groups, and is highly prevalent. It affects tens of that genetic susceptibility factors and environmen-
millions individuals worldwide and costs a fam- tal influences both contribute to ASD (Chaste and
ily approximately $60,000 USD a year on aver- Leboyer, 2012; Kim and Leventhal, 2015; Sandin
age. In the United States, the incidence of ASD is et al., 2014; Tordjman et al., 2014), but many ques-
1 in 68 children (1 in 42 boys and 1 in 189 girls) tions still remain unanswered. Recent efforts using
based on the data released by the Centers for genome screening and sequencing have identified
102

102 section II: E merging Clinical Applications and Future Potential

rare chromosomal abnormalities and copy num- of the cellular energy, mitochondria are also criti-
ber variations, as well as hundreds of rare gene cally involved in cellular metabolism, intracellular
mutations associated with autism (Baker and Jeste, calcium signaling, generation of reactive oxygen
2015; Devlin and Scherer, 2012; Huguet et  al., species, and apoptosis (Antico Arciuch et al., 2012;
2013; Jeste and Geschwind, 2014). A small num- Murphy, 2009; Palmieri et al., 2010; Rizzuto et al.,
ber of these genetic deficits appear highly pene- 2012; Suen et al., 2008), as well as in the regulation
trant and sufficient to cause autism by themselves. of innate and adaptive immunity (Weinberg et al.,
However, the majority of the genetic changes asso- 2015). Furthermore, mitochondria are known be
ciated with ASD appear to only increase the risk affected by many of the same endogenous and
to varying degrees. In these cases, environmental exogenous risk factors of ASD, such as toxins,
factors influencing early brain development are drugs, immune activation, and metabolic distur-
thought to play a significant role in the etiology of bances (Frye and Rossignol, 2011). Thus, under-
the disorder. In other words, in the presence of a standing the role of mitochondrial dysfunction
genetic predisposition to autism, a number of pre- in ASD may help unify our understanding of this
natal, perinatal, and postnatal risk factors appear complex disorder.
to further increase a child’s risk. These include Mitochondria play a vital role especially in the
advanced parental age at time of conception (both nervous system. The brain has very high energy
mother and father), maternal illness during preg- demands, as it consumes approximately 20% of the
nancy, birth complications, and exposure to tox- basal oxygen supply in humans, and hence, calo-
ins and/or drugs during early brain development ries consumed by the body, while accounting for
(Christensen et  al., 2013; Durkin et  al., 2008; only 2% of the body weight (Raichle and Gusnard,
Gardener et al., 2011; Stromland et al., 1994). It is 2002). This is mainly because cells in the nervous
important to note that these factors do not cause system need a great amount of adenosine triphos-
ASD by themselves. Rather, they seem to modestly phate (ATP) to maintain ionic gradients across
increase the risks of developing the disorder in cellular membranes, which are essential to gener-
combination with genetic predispositions. ate membrane excitability for neurotransmission
In summary, ASD inflicts a grave burden on and plasticity (Harris et  al., 2012). In addition,
affected individuals, their families, and society as a mitochondria are involved in various aspects of
whole. Currently, ASD remains a syndrome char- neurodevelopment such as the proliferation, dif-
acterized by behaviors, but not specific etiologies. ferentiation, and maturation of neural stem cells;
Its heterogeneity in terms of both risk factors and formation of dendritic arbors and spines; develop-
clinical presentations makes it a great challenge to mental and synaptic plasticity; and the determina-
unravel the pathophysiological mechanisms and tion of cell survival and death (Kann and Kovacs,
find therapeutic treatments. 2007; Kimura and Murakami, 2014; Li et al., 2004;
Mattson et  al., 2008; Xavier et  al., 2015). Thus, it
M I TO C H O N D R I A L is not surprising that there are now multiple lines
A N D M E TA B O L I C of evidence from clinical, genetic, and biochemi-
D Y S F U N C T I O N I N   AU T I S M cal studies—in both humans and animal models—
S P E C T RU M D I S O R D E R supporting a role for mitochondrial dysfunction in
Because a combination of diverse factors such as the etiology of ASD (Dhillon et al., 2011; Frye and
genetic background and environmental exposure Rossignol, 2011; Haas, 2010; Legido et  al., 2013;
are thought to contribute to the development of Rossignol and Frye, 2012).
ASD, it has been hypothesized that these hetero- Clinical evidence points to the involvement
geneous influences elicit similar developmental of mitochondrial disease and dysfunction in
outcomes by modulating a common central nexus, ASD. The prevalence of mitochondrial disease in
a perturbation of which leads to ASD (Berg and the ASD population is estimated to be about 5%,
Geschwind, 2012; Geschwind, 2008). Identifying 500 times higher than that found in the general
such a node would provide novel insights into the population (≈0.01%). The prevalence of abnor-
development of ASD, while targeting this pathway mal values of biomarkers related to mitochon-
could form the basis of therapeutic approaches for drial function in the ASD population may be even
various forms of the disorder. A candidate for this higher, suggesting that as many as 30% of children
central node is mitochondrial function, which is with ASD may experience mitochondrial dys-
integrated into and key to many cellular pathways. function. For example, almost one-third of autis-
For example, in addition to the well-known role as tic children had elevations in plasma lactate and/
the “powerhouse of the cell,” producing the bulk or the lactate-to-pyruvate ratio, and the levels of
103

Chapter 14: Metabolic Therapy for Autism Spectrum Disorder 103

many other mitochondrial biomarkers (pyruvate, of neurological diseases including Alzheimer’s


carnitine, and ubiquinone) are significantly dif- disease, Parkinson’s disease, multiple sclerosis,
ferent between ASD and controls (Rossignol and sleep disorders, and brain cancer (Frye et al., 2013;
Frye, 2012). Conversely, the rate of ASD is higher Napoli et al., 2014; Stafstrom and Rho, 2012).
among children with mitochondrial disease There are a number of mechanisms through
(Randolph-Gips and Srinivasan, 2012). Together, which the KD may provide neuroprotective activ-
these findings suggest that mitochondrial dys- ity. Two hallmark features of biochemical changes
function likely represents a significant subclass after the KD treatment are the increase in ketone
of ASD. body production by the liver through fatty acid
Furthermore, common comorbidities of ASD oxidation and a reduction in blood glucose lev-
also suggest the involvement of mitochondrial els (Stafstrom and Rho, 2012). Specific polyun-
dysfunction. One of the most significant comor- saturated fatty acids such as arachidonic acid,
bidities associated with ASD is epilepsy. A number docosahexaenoic acid, and eicosapentaenoic acid
of studies suggest that epilepsy affects a great pro- might themselves regulate neuronal membrane
portion of individuals with ASD, with the reported excitability (Voskuyl and Vreugdenhil, 2001),
prevalence of epilepsy in the ASD population reduce inflammation (Cullingford, 2008; Jeong et
ranging from 5% to 38%, which is much higher al., 2011), or decrease the production of reactive
than the 1%–2% prevalence in the general popu- oxygen species (ROS) by mitochondria (Kim do
lation of children (Frye, 2015). In contrast, sei- and Rho, 2008). Additionally, ketone bodies have
zures have been reported to occur in about 35% to been shown to possess neuroprotective properties
60% of individuals with biochemically confirmed through improved bioenergetics. They have been
mitochondrial disease (Rahman, 2012). Thus, reported to raise ATP levels and reduce ROS pro-
shared symptoms suggest a common etiopathol- duction through enhancement of nicotinamide
ogy. Similarly, gastrointestinal dysfunction, a fre- adenine dinucleotide (NADH) oxidation and
quent comorbidity of ASD (Chaidez et al., 2014), is inhibition of mitochondrial permeability tran-
also commonly reported in mitochondrial disease sition (Kim do et al., 2007; Kim do et al., 2015).
(Frye et al., 2015). Furthermore, the KD has been shown to stimulate
Interestingly, many animal models of ASD mitochondrial biogenesis (Ahola-Erkkila et al.,
also display mitochondrial dysfunction. These 2010; Bough et al., 2006).
models include ones based on both susceptibility The second major biochemical feature of the
genes such as MECP2, UBE3A, and SLC25A12 KD is the reduction of glycolysis. In addition to
as well as environmental risk factors includ- suppressing seizures, it also improves mitochon-
ing maternal immune activation, and exposure drial function and decreases oxidative stress,
to propionic acid and valproic acid. The cur- reduces activity of pro-apoptotic factors, and
rent evidence linking mitochondrial perturba- inhibits inflammatory mediators such as inter-
tions to ASD is summarized in Table 14.1. Taken leukins and tumor necrosis factor alpha (Maalouf
together, we believe that mitochondria act as a et al., 2009).
central nexus responding to and regulating many Beyond enhancing bioenergetics and mito-
domains of cellular biology that have been impli- chondrial function, the KD has also been shown
cated in ASD. to modulate the metabolism of γ-aminobutyric
acid (GABA) and acetylcholine, two major cen-
K E TO G E N I C D I E T A S   A tral nervous system neurotransmitters (Napoli
PROMISING THERAPY et al., 2014), as well as purines (such as ATP and
F O R   AU T I S M S P E C T R U M adenosine) that have pleiotropic neural modu-
DISORDER latory roles (Masino et  al., 2010; Masino et  al.,
The ketogenic diet (KD) is a special high-fat, low- 2009). In addition, the KD has also been reported
carbohydrate diet, which is a remarkably effective to regulate energy-sensing pathways such as
nonpharmacological treatment for patients with those involving the insulin-like growth factor
medically intractable epilepsy (Neal et  al., 2008). and the mammalian target of rapamycin (Napoli
Based on the historical observation that fasting et al., 2014).
or starvation can render antiseizure effects, the As mentioned above, mitochondrial and meta-
KD was designed to reproduce the biochemical bolic dysfunction may play a key role in the patho-
changes seen in these physiological states (Masino physiology of ASD. Based on research showing
and Rho, 2012). Recently, dietary and metabolic that the KD provides numerous neuroprotective
therapies have been attempted in a wider variety effects through modulating mitochondrial and
104

TABLE 14.1 STUDIES SHOWING A LINK BET WEEN ASD


AND MITOCHONDRIAL DYSFUNCTION

Reference Cases Evidence of mitochondrial dysfunction

Studies in ASD patients*


Al-Mosalem et al., 2009 30 Increased plasma lactate levels and activity of creatine kinase
Boccuto et al., 2013 87 Decreased tryptophan metabolism in lymphoblastoid cell lines
Celestino-Soper 909 The deficiency of the trimethyllysine hydroxylase epsilon, which encodes the
et al., 2012 first enzyme in carnitine biosynthesis, was more frequent in probands from
male-male multiplex ASD families
Chen et al., 2015 78 Mitochondrial DNA copy number in peripheral blood cells was elevated
Cohen et al., 1976 25 Increased serum creatine phosphokinase levels
Correia et al., 2006 241 Increased plasma lactate levels and lactate/pyruvate ratio, but not associated
with the variation at the SLC25A12 gene
Filipek et al., 2004 100 Reduced levels of carnitine and pyruvate, increased levels of alanine and ammo-
nia in serum
Frye et al., 2013 213 Abnormal acyl-carnitine panels and glutathione metabolism in blood samples
Glessner et al., 2009 859 Copy number variations in genes involved in the ubiquitin degradation were
implicated in susceptibility for ASD
Goh et al., 2014 75 Lactate doublets detected by brain magnetic resonance spectroscopic imaging
were present at a higher rate
Kent et al., 2006 129 The 3243A>G mitochondrial DNA mutation was concluded to be a rare cause
of isolated Asperger syndrome
Kim et al., 2011 Multiple Polymorphism in a mitochondrial aspartate/glutamate carrier gene (SLC25A12)
Silverman et al., 2008 families is found to be associated with restricted repetitive behavior in autism
Kuwabara et al., 2013 25 higher plasma levels of arginine and taurine, and lower levels of 5-oxoproline
and lactic acid
Laszlo et al., 1994 30 Increased serum lactate and pyruvate levels
Maestrini et al., 2010 127 Gene encodes an inner mitochondrial membrane protease-like protein
(IMMP2L) was implicated in susceptibility for ASD
Moreno et al., 1992 60 Increased lactate and pyruvate levels
Oliveira et al., 2005 69 20% of ASD patients showed hyperlactacidemia, while 7% were classified with
definite mitochondrial respiratory chain disorder
Poling et al., 2006 159 Increase blood aspartate aminotransferase and creatine kinase levels
Rose et al., 2012 43 Primary immune cells in the blood have a more oxidized intracellular and
extracellular microenvironment and a deficit in glutathione-mediated redox/
antioxidant capacity
Rose et al., 2014 25 Mitochondrial dysfunction observed in a subset of autism lymphoblastoid cell
lines
Ramoz et al., 2004 Multiple Polymorphism in a mitochondrial aspartate/glutamate carrier gene (SLC25A12)
Segurado et al., 2005 families is found to be associated with autism
Turunen et al., 2008
Tang et al., 2013 45 Mitochondrial function and intracellular redox status were compromised in the
pyramidal neurons of the temporal cortex
Studies in animal models
Ahn et al., 2014 Mitochondrial dysfunction in a rat model of ASD using prenatal VPA exposure
Jin et al., 2015 Mecp2, whose mutations cause Rett syndrome, was observed to regulate mito-
chondrial bioenergetics through a glutamine transporter in microglia.
Kriaucionis et al., 2006 Mitochondrial abnormalities observed in Mecp2-null mouse, a model of Rett
syndrome.
Macfabe et al., 2012 Mitochondrial dysfunction observed in a rat ASD model in which pro-
pionic acid, an enteric bacterial fermentation product, is infused
intracerebroventricularly
(continued)
105

Chapter 14: Metabolic Therapy for Autism Spectrum Disorder 105

TABLE 14.1 CONTINUED

Reference Cases Evidence of mitochondrial dysfunction

Naviaux et al., 2013 Antipurinergic therapy improves the autism-like features in the maternal
Naviaux et al., 2014 immune activation mouse model and the Fragile X mouse model
Naviaux et al., 2015
Park et al., 2014 Dietary therapy with triheptanoin enhanced mitochondrial substrate use and
improved metabolism and behaviors of Mecp2-null mouse model of Rett
syndrome
Sakurai et al., 2010 Loss of a mitochondrial aspartate-glutamate carrier gene results in hypomyelin-
ation. Myelin deficits in slice cultures from KO mice are reversed by admin-
istration of pyruvate
Su et al., 2011 Mitochondrial dysfunction observed in hippocampal neurons of the UBE3A
deficient mouse model for Angelman syndrome
Zhao et al., 2010 ASD-like features observed in neuronal glucose transporter isoform 3 deficient
mice

* Only studies reporting more than 25 subjects are included in this table

metabolic pathways, it is logical to hypothesize following her initial diagnosis, the child’s CARS
that this diet could prove to be beneficial for autis- score decreased from 49 to 17, representing a
tic individuals. change from severe autism to a nonautistic state,
and her intelligence quotient increased 70 points.
THE EFFECTS OF THE Finally, Spilioti and colleagues reported the
K E TO G E N I C D I E T effects following the KD treatment in a group of
O N   AU T I S M S P E C T R U M Greek children with ASD aged between 3.5 and
D I S O R D E R PAT I E N T S 6 years (Spilioti et al., 2013). Of the 6 patients who
To date, there have been limited trials of treat- successfully implemented the diet, significant and
ing autism patients with the KD. Evangeliou and average improvement was recorded in one and two
colleagues carried out a pilot prospective study patients, respectively, while minor improvement
on autistic children aged between 4 and 10 years was reported in the remaining three patients.
to investigate the role of the KD, specifically a In summary, results from these few studies
modified medium chain triglyceride (MCT) diet show that more than 50% of the autism patients
(Evangeliou et  al., 2003). Of the 18 patients who who received the KD treatment showed moderate-
adhered to the diet, improvement was recorded in to-significant improvement, while the remaining
most individuals, based on several parameters and displayed minor improvement. These preliminary
in accordance with the Childhood Autism Rating studies suggest that the KD could be a promising
Scale (CARS). Significant (>12 units of decrease therapy for ASD; more and larger clinical studies
in CARS) and average (>8–12 units of decrease are needed.
in CARS) improvement was recorded in two and
eight patients, respectively, while minor (2–8 units T R E AT I N G A N I M A L M O D E L S
of decrease in CARS) improvement was reported O F   AU T I S M W I T H   T H E
in the remaining eight patients. K E TO G E N I C   D I E T
In a separate study, Herbert and Buckley Experiments using animal models of human dis-
reported the history of a 12-year-old child with eases have contributed much to our understand-
autism and epilepsy treated with a gluten- and ing of the mechanisms of disease, and the efficacy
casein-free KD with fats composed mostly of of potential therapeutics in preclinical settings.
medium chain triglycerides (Herbert and Buckley, Due to the complex etiology of ASD, many ani-
2013). The patient showed significant improve- mal models exist, and several of them have been
ment in seizure activity, resolution of morbid used to test the effects of the KD. For example,
obesity, and improvement in cognitive and behav- succinic semialdehyde dehydrogenase (SSADH)
ioral functioning. Over the course of several years deficiency is a rare autosomal recessive condition
106

106 section II: E merging Clinical Applications and Future Potential

that results in mild-to-moderate mental retar- Ruskin et al., 2013; Smith et al., 2014). This model
dation, disproportionate language dysfunction, was identified in an extensive effort to character-
seizures, hypotonia, hyporeflexia, hallucinations, ize 10 inbred mouse strains in terms of possess-
and autistic behaviors (Pearl et  al., 2003). In an ing autism-like behaviors (Moy et  al., 2007), and
animal model of SSADH deficiency, the SSADH was later shown to display behavioral phenotypes
knockout mouse, Nylen and colleagues found that relevant to all three core diagnostic symptoms of
KD treatment normalized electroencephalogram ASD (McFarlane et  al., 2008). Additional tests
(EEG) activity. In addition, the decrease in min- conducted in multiple independent laboratories
iature inhibitory postsynaptic currents in pyra- further confirmed that BTBR animals display
midal cells of CA1 hippocampal slices observed prominent deficits in multiple social interaction
in the mutant animals was completely restored by and communication assays, and exhibit repetitive
the KD when compared with wild-type controls. and stereotyped behaviors. Thus, the BTBR strain
In contrast, there were no significant differences has been widely regarded as a consistent and robust
between the groups in terms of miniature excit- animal model of autism, reflecting more common
atory postsynaptic currents. Moreover, GABAA forms of ASD (Gaugler et  al., 2014). During the
receptor-associated chloride channel binding relatively short time since its discovery as an ASD
of [35S]tert-butylbicyclophosphorothionate was model, the BTBR strain has been increasingly used
restored fully in hippocampus and frontoparietal to study the etiology and potential intervention of
cortex, but not in amygdala or thalamus, of KD- ASD (Llaneza et al., 2010; McFarlane et al., 2008;
fed mutant mice. Behaviorally, KD-treated mutant Moy et al., 2007; Ruskin et al., 2013). These stud-
animals experienced significantly fewer seizures ies have identified alterations in BTBR animals
compared with mutant animals fed with the con- at many levels, including genetic and epigenetic;
trol diet (Nylen et al., 2008). synaptic; neuroanatomic and functional connec-
Another study examined the effects of restricted tivity; and immunological—all of which have been
standard and ketogenic diets on a mouse model of implicated in individuals with ASD (Ellegood and
Rett syndrome. Rett syndrome is a neurodevel- Crawley, 2015; Llaneza et  al., 2010; Meyza et  al.,
opmental disorder characterized by normal early 2013; Shpyleva et  al., 2014). To test the effects of
growth and development followed by a slowing the KD on the BTBR model, Ruskin and colleagues
of development, impairment of motor functions, fed the animals with the diet for 3 weeks and then
seizure susceptibility, and intellectual disability. In performed an array of behavioral assays. They
most cases, it is caused by mutations in the methyl- reported that the BTBR mice showed increased
CpG-binding protein 2 (MECP2) gene (Amir sociability in a three-chamber test, decreased
et  al., 1999). Children with Rett syndrome often self-directed repetitive behavior, and improved
exhibit autistic-like behaviors in the early stages social communication in a food-preference assay
(Percy, 2011). By feeding animals with standard (Ruskin et  al., 2013). In addition, the authors
chow in unrestricted or restricted amounts or a showed that the behavioral improvements were
KD in restricted amounts, Mantis and colleagues probably not related to any antiseizure effect of the
found that performance in assays of motor behav- diet, because no spontaneous seizures or abnormal
ior and anxiety was significantly worse in Mecp2 EEG features were observed in the BTBR animals.
mutant mice compared with wild-type animals, These results strongly suggest that the KD may be
and restriction of either the standard diet or the able to improve the core behavioral symptoms of
KD improved motor behavior and reduced anxiety autism.
in the mutant animals (Mantis et al., 2009). As mentioned earlier, both genetic and envi-
More recently, using the BTBR T+tf/J (BTBR) ronmental factors contribute to the risks of devel-
mouse model of autism, Ruskin and colleagues oping ASD. An example of environmental risk
investigated the effects of the KD on core behav- factors is exposure to exogenous chemicals such
ioral abnormalities that define autism (Ruskin as valproic acid (VPA) during pregnancy. Valproic
et al., 2013). The BTBR inbred strain is one of the acid is a pharmacological anticonvulsant used in
most clinically relevant animal models of autism, humans primarily for the treatment of epilepsy
mainly because it displays all the core behavioral and migraine. Epidemiological studies showed
features that define the disorder—specifically, that maternal use of VPA during pregnancy was
impaired social behaviors and communication as associated with a significantly increased risk of
well as stereotyped behaviors and resistance to the offspring being autistic (Bromley et al., 2013;
change (Ellegood and Crawley, 2015; McFarlane Christensen et al., 2013). The model of VPA expo-
et  al., 2008; Meyza et  al., 2013; Moy et  al., 2007; sure is one of the most frequently studied animal
107

Chapter 14: Metabolic Therapy for Autism Spectrum Disorder 107

models of ASD (Chomiak et  al., 2013; Roullet shifts in energy metabolism and the direct actions
et  al., 2013). This model exhibits many similar of the ketone bodies on the mitochondria are two
structural and behavioral features of ASD indi- of the promising candidate mechanisms. In addi-
viduals. For example, it has been shown that VPA tion, the optimum formulation of the KD needs
exposure in both rats and mice leads to autistic- to be established, which may be different for ASD
like behavioral impairment including decreased compared with epilepsy, and distinct biomarkers
social interactions, increased repetitive behav- need to be developed that will accurately track
iors, and deficits in communication, as well as behavioral symptoms as well as disease ontogeny.
increased anxiety and increased sensitivity to both Last but not least, the potential side effects of the
painful and nonpainful stimuli. Overall, the VPA KD should be carefully investigated, especially for
rodent model possesses construct, face, and pre- ASD patients who may also suffer from intrinsic
dictive validity for ASD studies. Using this model, metabolic derangements.
Ahn and colleagues found that the KD treatment
recovered part of the play behavior of juvenile rats REFERENCES
exposed to VPA prenatally, shown by increased Ahn, Y., Narous, M., Tobias, R., Rho, J. M., and
number of play initiations/attacks. However, the Mychasiuk, R. (2014). The ketogenic diet modifies
diet did not change the disrupted pattern of play social and metabolic alterations identified in the
responses (Ahn et  al., 2014). Interestingly, the prenatal valproic acid model of autism spectrum
authors also found that prenatal exposure to VPA disorder. Dev Neurosci 36, 371–380. doi:10.1159/
altered mitochondrial respiration, and the KD was 000362645
able to partially restore it. Specifically, basal and Ahola-Erkkila, S., Carroll, C.J., Peltola-Mjosund,
K., Tulkki, V., Mattila, I., Seppanen-Laakso, T.,
ATP-linked respiration was significantly increased
Oresic, M., Tyynismaa, H., and Suomalainen, A.
in the VPA animals, but was partly normalized by (2010). Ketogenic diet slows down mitochondrial
the KD (Ahn et al., 2014). myopathy progression in mice. Hum Mol Gen 19,
1974–1984.
FUTURE RESEARCH Al-Mosalem, O. A., El-Ansary, A., Attas, O., and Al-
DIRECTIONS USING Ayadhi, L. (2009). Metabolic biomarkers related
M E TA B O L I C T H E R A P Y to energy metabolism in Saudi autistic chil-
Although strong evidence exists that mitochon- dren. Clinic Biochem 42, 949–957. doi:10.1016/
drial and metabolic dysfunction may play a piv- j.clinbiochem.2009.04.006
otal role in the complex pathophysiology of ASD, Amir, R.E., Van den Veyver, I.B., Wan, M., Tran, C.Q.,
the exact mechanisms remain undefined. For Francke, U., and Zoghbi, H.Y. (1999). Rett syn-
example, is mitochondrial dysfunction a cause, drome is caused by mutations in X-linked MECP2,
consequence, or epiphenomenon of the disrupted encoding methyl-CpG-binding protein 2. Nat
Genet 23, 185–188.
neurodevelopment observed in ASD? Considering
Antico Arciuch, V.G., Elguero, M.E., Poderoso, J.J.,
that ASD is a multisystem disorder, in which tis-
and Carreras, M.C. (2012). Mitochondrial regula-
sue, and more specifically, in which cell types is tion of cell cycle and proliferation. Antioxid Redox
mitochondrial dysfunction relevant to the dis- Signal 16, 1150–1180.
ease state? Can impaired mitochondrial function Baker, E., and Jeste, S.S. (2015). Diagnosis and man-
define a subtype of the otherwise heterogeneous agement of autism spectrum disorder in the era
patient population? Is the severity of mitochon- of genomics:  rare disorders can pave the way for
drial and metabolic disturbance correlated with targeted treatments. Pediatr Clin North Am 62,
any behavioral abnormalities or comorbidities? 607–618.
When mitochondrial function is improved, can it Berg, J.M., and Geschwind, D.H. (2012). Autism
ameliorate the general features of ASD? Answering genetics:  searching for specificity and conver-
these questions will require the collective efforts of gence. Genome Biol 13, 247.
many basic, translational, and clinical research- Boccuto, L., Chen, C. F., Pittman, A. R., Skinner,
C. D., McCartney, H. J., Jones, K., Bochner, B.
ers, as well as investigators with diverse expertise
R., Stevenson, R. E., Schwartz, C. E. (2013).
in multiorgan dysfunction, metabolism, and both
Decreased tryptophan metabolism in patients
genetic and environmental risk factors. with autism spectrum disorders. Mol Autism 4,
Similarly, although the KD has shown prom- 16. doi:10.1186/2040-2392-4-16
ising results from limited studies in both patients Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
and animal models, a mechanistic understanding Gawryluk, J.W., Greene, J.G., Shaw, R., Smith,
of its effects in ASD is lacking. Borrowing from the Y., Geiger, J.D., and Dingledine, R.J. (2006).
rich literature on the KD treatment for epilepsy, Mitochondrial biogenesis in the anticonvulsant
108

108 section II: E merging Clinical Applications and Future Potential

mechanism of the ketogenic diet. Ann Neurol 60, J. (2007). Mapping early brain development in
223–235. autism. Neuron 56, 399–413.
Bromley, R.L., Mawer, G.E., Briggs, M., Cheyne, C., Cullingford, T. (2008). Peroxisome proliferator-
Clayton-Smith, J., Garcia-Finana, M., Kneen, activated receptor alpha and the ketogenic diet.
R., Lucas, S.B., Shallcross, R., Baker, G.A., et  al. Epilepsia 49(Suppl 8), 70–72.
(2013). The prevalence of neurodevelopmental Devlin, B., and Scherer, S.W. (2012). Genetic archi-
disorders in children prenatally exposed to anti- tecture in autism spectrum disorder. Curr Opin
epileptic drugs. J Neurol Neurosurg Psychiatry 84, Genetics Dev 22, 229–237.
637–643. Dhillon, S., Hellings, J.A., and Butler, M.G. (2011).
CDC. (2014). Prevalence of autism spectrum disor- Genetics and mitochondrial abnormalities in autism
der among children aged 8  years  —autism and spectrum disorders:  a review. Curr Genomics 12,
developmental disabilities monitoring network, 11 322–332.
sites, United States, 2010. Morbidity and Mortality DiCicco-Bloom, E., Lord, C., Zwaigenbaum, L.,
Weekly Report (MMWR) Surveillance Summaries Courchesne, E., Dager, S.R., Schmitz, C., Schultz,
63, 1–21. R.T., Crawley, J., and Young, L.J. (2006). The devel-
Celestino-Soper, P. B., Violante, S., Crawford, E. L., opmental neurobiology of autism spectrum disor-
Luo, R., Lionel, A. C., Delaby, E., Cai, G., Sadikovic, der. J Neurosci 26, 6897–6906.
B., Lee, K., Lo, C., et  al. (2012). A common X- Durkin, M.S., Maenner, M.J., Newschaffer, C.J.,
linked inborn error of carnitine biosynthesis may Lee, L.C., Cunniff, C.M., Daniels, J.L., Kirby,
be a risk factor for nondysmorphic autism. Proc R.S., Leavitt, L., Miller, L., Zahorodny, W., et  al.
Natl Acad Sci U S A 109, 7974–7981. doi:10.1073/ (2008). Advanced parental age and the risk of
pnas.1120210109 autism spectrum disorder. Am J Epidemiol 168,
Chaidez, V., Hansen, R.L., and Hertz-Picciotto, I. 1268–1276.
(2014). Gastrointestinal problems in children with Ellegood, J., and Crawley, J.N. (2015). Behavioral and
autism, developmental delays or typical develop- neuroanatomical phenotypes in mouse models of
ment. J Autism Dev Disord 44, 1117–1127. autism. Neurotherapeutics 12, 521–533.
Chaste, P., and Leboyer, M. (2012). Autism risk fac- Evangeliou, A., Vlachonikolis, I., Mihailidou, H.,
tors:  genes, environment, and gene-environment Spilioti, M., Skarpalezou, A., Makaronas, N.,
interactions. Dialogues Clin Neurosci. 14, Prokopiou, A., Christodoulou, P., Liapi-Adamidou,
281–292. G., Helidonis, E., et  al. (2003). Application of a
Chen, S., Li, Z., He, Y., Zhang, F., Li, H., Liao, Y., ketogenic diet in children with autistic behav-
Wei, Z., Wan, G., Xiang, X., Hu, M., et al. (2015). ior: pilot studyJ Child Neurol 18, 113–118.
Elevated mitochondrial DNA copy number in Filipek, P.A., Juranek, J., Nguyen, M.T., Cummings,
peripheral blood cells is associated with child- C., and Gargus, J.J. (2004). Relative carnitine defi-
hood autism. BMC Psychiatry 15, 50. doi:10.1186/ ciency in autism. J Autism Dev Disord 34, 615–623
s12888-015-0432-y Frye, R.E., and Rossignol, D.A. (2011). Mitochondrial
Chomiak, T., Turner, N., and Hu, B. (2013). What dysfunction can connect the diverse medical
we have learned about autism spectrum disorder symptoms associated with autism spectrum disor-
from valproic acid. Patholog Res Int 2013, 712758. ders. Pediatr Res 69, 41R–47R.
doi: 10.1155/2013/712758. Frye, R.E., Melnyk, S., and Macfabe, D.F. (2013a).
Christensen, J., Gronborg, T.K., Sorensen, M.J., Unique acyl-carnitine profiles are potential bio-
Schendel, D., Parner, E.T., Pedersen, L.H., and markers for acquired mitochondrial disease in
Vestergaard, M. (2013). Prenatal valproate expo- autism spectrum disorder. Transl Psychiatry 3,
sure and risk of autism spectrum disorders and e220. doi:10.1038/tp.2012.143
childhood autism. JAMA 309, 1696–1703. Frye, R.E., Rossignol, D., Casanova, M.F., Brown,
Cohen, D. J., Johnson, W., Caparulo, B. K., and Young, G.L., Martin, V., Edelson, S., Coben, R., Lewine,
J. G. (1976). Creatine phosphokinase levels in J., Slattery, J.C., Lau, C., et  al. (2013b). A review
children with severe developmental disturbances. of traditional and novel treatments for seizures
Arch Gen Psychiatry 33, 683–686 in autism spectrum disorder:  findings from a
Correia, C., Coutinho, A. M., Diogo, L., Grazina, systematic review and expert panel. Front Public
M., Marques, C., Miguel, T., Ataide, A., Almeida, Health 1, 31.
J., Borges, L., Oliveira, C., et  al. (2006). Brief Frye, R.E. (2015). Metabolic and mitochondrial disor-
report: High frequency of biochemical markers for ders associated with epilepsy in children with autism
mitochondrial dysfunction in autism: no associa- spectrum disorder. Epilepsy Behav 47, 147–157.
tion with the mitochondrial aspartate/glutamate Frye, R.E., Rose, S., Slattery, J., and MacFabe, D.F.
carrier SLC25A12 gene. J Autism Dev Disord 36, (2015). Gastrointestinal dysfunction in autism
1137–1140. doi:10.1007/s10803-006-0138-6 spectrum disorder:  the role of the mitochondria
Courchesne, E., Pierce, K., Schumann, C.M., Redcay, and the enteric microbiome. Microb Ecol Health
E., Buckwalter, J.A., Kennedy, D.P., and Morgan, Dis 26, 27458.
109

Chapter 14: Metabolic Therapy for Autism Spectrum Disorder 109

Gardener, H., Spiegelman, D., and Buka, S.L. (2011). and neurotoxicity. J Neurosci 35, 2516–2529.
Perinatal and neonatal risk factors for autism:  a doi:10.1523/JNEUROSCI.2778-14.2015
comprehensive meta-analysis. Pediatrics 128, Kann, O., and Kovacs, R. (2007). Mitochondria and
344–355. neuronal activity. Am J PhysiolCell Physiol 292,
Gaugler, T., Klei, L., Sanders, S.J., Bodea, C.A., C641–657.
Goldberg, A.P., Lee, A.B., Mahajan, M., Manaa, D., Kent, L., Lambert, C., Pyle, A., Elliott, H., Wheelwright, S.,
Pawitan, Y., Reichert, J., et al. (2014). Most genetic Baron-Cohen, S., and Chinnery, P.F.The mitochon-
risk for autism resides with common variation. drial DNA A3243A>G mutation must be an infre-
Nat Genet 46, 881–885. quent cause of Asperger syndrome. J. Pediatr 149,
Geschwind, D.H. (2008). Autism:  many genes, com- 280–281. doi:10.1016/j.jpeds.2005.08.060
mon pathways? Cell 135, 391–395. Kim do, Y., Davis, L.M., Sullivan, P.G., Maalouf, M.,
Glessner, J. T., Wang, K., Cai, G., Korvatska, O., Kim, Simeone, T.A., van Brederode, J., and Rho, J.M.
C. E., Wood, S., Zhang, H., Estes, A., Brune, (2007). Ketone bodies are protective against oxi-
C. W., et  al. (2009). Autism genome-wide copy dative stress in neocortical neurons. J Neurochem
number variation reveals ubiquitin and neuro- 101, 1316–1326.
nal genes. Nature 459, 569–573. doi:10.1038/ Kim do, Y., and Rho, J.M. (2008). The ketogenic diet
nature07953 and epilepsy. Curr Opin Clin Nutr Metab Care 11,
Goh, S., Dong, Z., Zhang, Y., DiMauro, S., and Peterson, 113–120.
B.S. (2014). Mitochondrial dysfunction as a neuro- Kim do, Y., Simeone, K.A., Simeone, T.A., Pandya, J.D.,
biological subtype of autism spectrum disorder: evi- Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G.,
dence from brain imaging. JAMA Psychiatry 71, and Rho, J.M. (2015). Ketone bodies mediate anti-
665–671. doi:10.1001/jamapsychiatry.2014.179 seizure effects through mitochondrial permeabil-
Goines, P., and Van de Water, J. (2010). The immune ity transition. Ann Neurol 78, 77–87.
system’s role in the biology of autism. Curr Opin Kim, S.J., Silva, R.M., Flores, C.G., Jacob, S., Guter,
Neurol 23, 111–117. S., Valcante, G., Zaytoun, A.M., Cook, E.H.,
Haas, R.H. (2010). Autism and mitochondrial disease. and Badner, J.A. (2011). A quantitative associa-
Dev Dis Res Rev 16, 144–153. tion study of SLC25A12 and restricted repetitive
Hansen, S.N., Schendel, D.E., and Parner, E.T. (2015). behavior traits in autism spectrum disorders. Mol
Explaining the increase in the prevalence of autism Autism 2, 8. doi:10.1186/2040-2392-2-8
spectrum disorders:  the proportion attributable Kim, Y.S., and Leventhal, B.L. (2015). Genetic epide-
to changes in reporting practices. JAMA Pediatr miology and insights into interactive genetic and
169, 56–62. environmental effects in autism spectrum disor-
Harris, J.J., Jolivet, R., and Attwell, D. (2012). Synaptic ders. Biol Psychiatry 77, 66–74.
energy use and supply. Neuron 75, 762–777. Kimura, T., and Murakami, F. (2014). Evidence that
Herbert, M.R., and Buckley, J.A. (2013). Autism and dendritic mitochondria negatively regulate den-
dietary therapy: case report and review of the lit- dritic branching in pyramidal neurons in the neo-
erature. J Child Neurol 28, 975–982. cortex. J. Neurosci 34, 6938–6951.
Hsiao, E.Y. (2013). Immune dysregulation in autism Kleinman, J.M., Ventola, P.E., Pandey, J., Verbalis,
spectrum disorder. International Rev Neurobiol A.D., Barton, M., Hodgson, S., Green, J., Dumont-
113, 269–302. Mathieu, T., Robins, D.L., and Fein, D. (2008).
Huguet, G., Ey, E., and Bourgeron, T. (2013). The Diagnostic stability in very young children with
genetic landscapes of autism spectrum disorders. autism spectrum disorders. J Autism Dev Disord
Annu Rev Genomics Hum Genet 14, 191–213. 38, 606–615.
Jeong, E.A., Jeon, B.T., Shin, H.J., Kim, N., Lee, D.H., Kriaucionis, S., Paterson, A., Curtis, J., Guy, J.,
Kim, H.J., Kang, S.S., Cho, G.J., Choi, W.S., and Macleod, N., and Bird, A. (2006). Gene expression
Roh, G.S. (2011). Ketogenic diet-induced peroxi- analysis exposes mitochondrial abnormalities in a
some proliferator-activated receptor-gamma acti- mouse model of Rett syndrome. Mol Cell Biol 26,
vation decreases neuroinflammation in the mouse 5033–5042. doi:10.1128/MCB.01665-05
hippocampus after kainic acid-induced seizures. Kuwabara, H., Yamasue, H., Koike, S., Inoue, H.,
Exp Neurol 232, 195–202. Kawakubo, Y., Kuroda, M., Takano, Y., Iwashiro,
Jeste, S.S., and Geschwind, D.H. (2014). Disentangling N., Natsubori, T., Aoki, Y., et  al. (2013). Altered
the heterogeneity of autism spectrum disor- metabolites in the plasma of autism spectrum dis-
der through genetic findings. Nat Rev Neurol order:  a capillary electrophoresis time-of-flight
10, 74–81. mass spectroscopy study. PloS One 8, e73814.
Jin, L. W., Horiuchi, M., Wulff, H., Liu, X. B., doi:10.1371/journal.pone.0073814
Cortopassi, G. A., Erickson, J. D., and Maezawa, Lai, M.C., Lombardo, M.V., and Baron-Cohen, S.
I. (2015). Dysregulation of glutamine trans- (2014). Autism. Lancet 383, 896–910.
porter SNAT1 in Rett syndrome microglia:  a Laszlo, A., Horvath, E., Eck, E., and Fekete, M. (1994).
mechanism for mitochondrial dysfunction Serum serotonin, lactate and pyruvate levels in
110

110 section II: E merging Clinical Applications and Future Potential

infantile autistic children. Clinica Chimica Acta; Mattson, M.P., Gleichmann, M., and Cheng, A. (2008).
International J Clin Chem 229, 205–207 Mitochondria in neuroplasticity and neurological
Legido, A., Jethva, R., and Goldenthal, M.J. (2013). disorders. Neuron 60, 748–766.
Mitochondrial dysfunction in autism. Semin Mayer, E.A., Tillisch, K., and Gupta, A. (2015). Gut/
Pediatr Neurol 20, 163–175. brain axis and the microbiota. J Clin Inves 125,
Levy, S.E., Giarelli, E., Lee, L.C., Schieve, L.A., Kirby, R.S., 926–938.
Cunniff, C., Nicholas, J., Reaven, J., and Rice, C.E. McFarlane, H.G., Kusek, G.K., Yang, M., Phoenix, J.L.,
(2010). Autism spectrum disorder and co-occurring Bolivar, V.J., and Crawley, J.N. (2008). Autism-
developmental, psychiatric, and medical condi- like behavioral phenotypes in BTBR T+tf/J mice.
tions among children in multiple populations of the Genes, Brain, Behav 7, 152–163.
United States. J Dev Behav Pediatr 31, 267–275. Meyza, K.Z., Defensor, E.B., Jensen, A.L., Corley, M.J.,
Li, Z., Okamoto, K., Hayashi, Y., and Sheng, M. (2004). Pearson, B.L., Pobbe, R.L., Bolivar, V.J., Blanchard,
The importance of dendritic mitochondria in the D.C., and Blanchard, R.J. (2013). The BTBR T+
morphogenesis and plasticity of spines and syn- tf/J mouse model for autism spectrum disorders-
apses. Cell 119, 873–887. in search of biomarkers. Behavioural Brain Res
Llaneza, D.C., DeLuke, S.V., Batista, M., Crawley, 251, 25–34.
J.N., Christodulu, K.V., and Frye, C.A. (2010). Moreno, H., Borjas, L., Arrieta, A., Saez, L., Prassad,
Communication, interventions, and scientific A., Estevez, J., and Bonilla, E. (1992). [Clinical het-
advances in autism: a commentary. Physiol Behav erogeneity of the autistic syndrome: a study of 60
100, 268–276. families]. Invest Clin 33, 13–31.
Lord, C., Risi, S., DiLavore, P.S., Shulman, C., Thurm, Moy, S.S., Nadler, J.J., Young, N.B., Perez, A., Holloway,
A., and Pickles, A. (2006). Autism from 2 to 9 years L.P., Barbaro, R.P., Barbaro, J.R., Wilson, L.M.,
of age. Arch Gen Psychiatry 63, 694–701. Threadgill, D.W., Lauder, J.M., et al. (2007). Mouse
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The behavioral tasks relevant to autism: phenotypes of
neuroprotective properties of calorie restriction, 10 inbred strains. Behav Brain Res 176, 4–20.
the ketogenic diet, and ketone bodies. Brain Res Murphy, M.P. (2009). How mitochondria produce
Rev 59, 293–315. reactive oxygen species. Biochem J 417, 1–13.
Macfabe, D. F. (2012). Short-chain fatty acid fermenta- Napoli, E., Duenas, N., and Giulivi, C. (2014). Potential
tion products of the gut microbiome: implications therapeutic use of the ketogenic diet in autism
in autism spectrum disorders. Microb Ecol Health spectrum disorders. Front Pediatr 2, 69.
Dis 23. doi:10.3402/mehd.v23i0.19260 Naviaux, J.C., Schuchbauer, M.A., Li, K., Wang, L.,
Maestrini, E., Pagnamenta, A.T., Lamb, J.A., Bacchelli, Risbrough, V.B., Powell, S.B., and Naviaux, R.K.
E., Sykes, N.H., Sousa, I., Toma, C., Barnby, G., (2014). Reversal of autism-like behaviors and
Butler, H., Winchester, L., et  al. (2010). High- metabolism in adult mice with single-dose anti-
density SNP association study and copy number purinergic therapy. Transl Psychiatry 4, e400.
variation analysis of the AUTS1 and AUTS5 loci doi:10.1038/tp.2014.33.
implicate the IMMP2L-DOCK4 gene region in Naviaux, J.C., Wang, L., Li, K., Bright, A.T., Alaynick,
autism susceptibility. Mol Psychiatry 15, 954–968. W.A., Williams, K.R., Powell, S.B., and Naviaux,
doi:10.1038/mp.2009.34 R.K. et  al. (2015). Antipurinergic therapy cor-
Mantis, J.G., Fritz, C.L., Marsh, J., Heinrichs, S.C., and rects the autism-like features in the Fragile X
Seyfried, T.N. (2009). Improvement in motor and (Fmr1 knockout) mouse model. Mol Autism 6, 1.
exploratory behavior in Rett syndrome mice with doi:10.1186/2040-2392-6-1
restricted ketogenic and standard diets. Epilepsy Naviaux, R.K., Zolkipli, Z., Wang, L., Nakayama,
Behav 15, 133–141. T., Naviaux, J.C., Le, T.P., Schuchbauer, M.A.,
Masino, S.A., Kawamura, M., Wasser, C.D., Pomeroy, Rogac, M., Tang, Q., Dugan, L.L., et  al. (2013).
L.T., and Ruskin, D.N. (2009). Adenosine, keto- Antipurinergic therapy corrects the autism-like
genic diet and epilepsy: the emerging therapeutic features in the poly(IC) mouse model. PLoS One
relationship between metabolism and brain activ- 8, e57380. doi:10.1371/journal.pone.0057380
ity. Curr Neuropharmacol 7, 257–268. Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
Masino, S.A., Kawamura, M., Jr., Ruskin, D.N., Edwards, N., Fitzsimmons, G., Whitney, A., and
Gawryluk, J., Chen, X., and Geiger, J.D. (2010). Cross, J.H. (2008). The ketogenic diet for the treat-
Purines and the Anti-Epileptic Actions of ment of childhood epilepsy:  a randomised con-
Ketogenic Diets. Open Neurosci J 4, 58–63. trolled trial. Lancet Neurol 7, 500–506.
Masino, S.A., and Rho, J.M. (2012). Mechanisms of Nylen, K., Velazquez, J.L., Likhodii, S.S., Cortez, M.A.,
ketogenic diet action. In Jasper’s Basic Mechanisms Shen, L., Leshchenko, Y., Adeli, K., Gibson, K.M.,
of the Epilepsies, J.L. Noebels, M. Avoli, M.A. Burnham, W.M., and Snead, O.C., 3rd (2008). A
Rogawski, R.W. Olsen, and A.V. Delgado- ketogenic diet rescues the murine succinic semi-
Escueta, eds. (Bethesda, MD: National Center for aldehyde dehydrogenase deficient phenotype. Exp
Biotechnology Information), pp. 1001–1022. Neurol 210, 449–457.
111

Chapter 14: Metabolic Therapy for Autism Spectrum Disorder 111

Oliveira, G., Diogo, L., Grazina, M., Garcia, P., Ataide, generation in primary immune cells from chil-
A., Marques, C., Miguel, T., Borges, L., Vicente, dren with autism. Autism Res Treat 2012, 986519.
A.M., and Oliveira, C.R. (2005). Mitochondrial doi:10.1155/2012/986519
dysfunction in autism spectrum disorders:  a Rose, S., Frye, R.E., Slattery, J., Wynne, R., Tippett,
population-based study. Dev Med Child Neurol M., Pavliv, O., Melnyk, S., and James, S.J. (2014).
47, 185–189. Oxidative stress induces mitochondrial dysfunc-
Orinstein, A.J., Helt, M., Troyb, E., Tyson, K.E., tion in a subset of autism lymphoblastoid cell lines
Barton, M.L., Eigsti, I.M., Naigles, L., and Fein, in a well-matched case control cohort. PLoS One
D.A. (2014). Intervention for optimal outcome in 9, e85436. doi:10.1371/journal.pone.0085436
children and adolescents with a history of autism. Rossignol, D.A., and Frye, R.E. (2012). Mitochondrial
J Dev Behav Pediatr 35, 247–256. dysfunction in autism spectrum disorders:  a sys-
Palmieri, L., Papaleo, V., Porcelli, V., Scarcia, P., Gaita, tematic review and meta-analysis. Mol Psychiatry
L., Sacco, R., Hager, J., Rousseau, F., Curatolo, P., 17, 290–314.
Manzi, B., et  al. (2010). Altered calcium homeo- Roullet, F.I., Lai, J.K., and Foster, J.A. (2013). In utero
stasis in autism-spectrum disorders:  evidence exposure to valproic acid and autism—a current
from biochemical and genetic studies of the mito- review of clinical and animal studies. Neurotoxicol
chondrial aspartate/glutamate carrier AGC1. Mol Teratol 36, 47–56.
Psychiatry 15, 38–52. Ruskin, D.N., Svedova, J., Cote, J.L., Sandau, U., Rho,
Park, M.J., Aja, S., Li, Q., Degano, A.L., Penati, J., J.M., Kawamura, M., Jr., Boison, D., and Masino,
Zhuo, J., Roe, C.R., and Ronnett, G.V. (2014). S.A. (2013). Ketogenic diet improves core symp-
Anaplerotic triheptanoin diet enhances mitochon- toms of autism in BTBR mice. PLoS One 8,
drial substrate use to remodel the metabolome e65021.
and improve lifespan, motor function, and socia- Sakurai, T., Ramoz, N., Barreto, M., Gazdoiu, M.,
bility in MeCP2-null mice. PLoS One 9, e109527. Takahashi, N., Gertner, M., Dorr, N., Gama
doi:10.1371/journal.pone.0109527 Sosa, M.A., De Gasperi, R., Perez, G., et  al.
Patterson, P.H. (2011). Maternal infection and (2010). Slc25a12 disruption alters myelination
immune involvement in autism. Trends Mol Med and neurofilaments:  a model for a hypomyelin-
17, 389–394. ation syndrome and childhood neurodevelop-
Pearl, P.L., Gibson, K.M., Acosta, M.T., Vezina, L.G., mental disorders. Biol Psychiatry 67, 887–894.
Theodore, W.H., Rogawski, M.A., Novotny, E.J., doi:10.1016/j.biopsych.2009.08.042
Gropman, A., Conry, J.A., Berry, G.T., et al. (2003). Sandin, S., Lichtenstein, P., Kuja-Halkola, R.,
Clinical spectrum of succinic semialdehyde dehy- Larsson, H., Hultman, C.M., and Reichenberg,
drogenase deficiency. Neurology 60, 1413–1417. A. (2014). The familial risk of autism. JAMA 311,
Percy, A.K. (2011). Rett syndrome:  exploring the 1770–1777.
autism link. Archiv Neurol 68, 985–989. Segurado, R., Conroy, J., Meally, E., Fitzgerald, M.,
Poling, J. S., Frye, R. E., Shoffner, J., and Zimmerman, Gill, M., and Gallagher, L. (2005). Confirmation
A. W. (2006). Developmental regression and mito- of association between autism and the mitochon-
chondrial dysfunction in a child with autism. J drial aspartate/glutamate carrier SLC25A12 gene
Child Neurol 21, 170–172 on chromosome 2q31. Am J Psychiatry 162, 2182–
Rahman, S. (2012). Mitochondrial disease and epi- 2184. doi:10.1176/appi.ajp.162.11.2182
lepsy. Dev Med Child Neurol 54, 397–406. Shpyleva, S., Ivanovsky, S., de Conti, A., Melnyk,
Raichle, M.E., and Gusnard, D.A. (2002). Appraising S., Tryndyak, V., Beland, F.A., James, S.J., and
the brain’s energy budget. Proc Natl Acad Sci USA Pogribny, I.P. (2014). Cerebellar oxidative DNA
99, 10237–10239. damage and altered DNA methylation in the
Ramoz, N., Reichert, J.G., Smith, C.J., Silverman, J.M., BTBR T+tf/J mouse model of autism and similari-
Bespalova, I.N., Davis, K.L., and Buxbaum, J.D. ties with human post mortem cerebellum. PLoS
(2004). Linkage and association of the mitochon- One 9, e113712.
drial aspartate/glutamate carrier SLC25A12 gene Silverman, J.M., Buxbaum, J.D., Ramoz, N.,
with autism. Am J Psychiatry 161, 662–669. Schmeidler, J., Reichenberg, A., Hollander, E.,
Randolph-Gips, M., and Srinivasan, P. (2012). Angelo, G., Smith, C.J., and Kryzak, L.A. (2008).
Modeling autism:  a systems biology approach. J Autism-related routines and rituals associated
Clin Bioinform 2, 17. with a mitochondrial aspartate/glutamate carrier
Rizzuto, R., De Stefani, D., Raffaello, A., and SLC25A12 polymorphism. Am J Med Gen:  Part
Mammucari, C. (2012). Mitochondria as sensors B, Neuropsychiatric Genetics 147, 408–410.
and regulators of calcium signalling. Nat Rev Mol doi:10.1002/ajmg.b.30614
Cell Biol 13, 566–578. Smith, J.D., Rho, J.M., Masino, S.A., and Mychasiuk,
Rose, S., Melnyk, S., Trusty, T.A., Pavliv, O., Seidel, L., R. (2014). Inchworming: a novel motor stereotypy
Li, J., Nick, T., and James, S.J. (2012). Intracellular in the BTBR T+ Itpr3tf/J mouse model of autism. J
and extracellular redox status and free radical Vis Exp. 2014 Jul 5(89). doi: 10.3791/50791.
112

112 section II: E merging Clinical Applications and Future Potential

Spilioti, M., Evangeliou, A.E., Tramma, D., Tordjman, S., Somogyi, E., Coulon, N., Kermarrec, S.,
Theodoridou, Z., Metaxas, S., Michailidi, E., Bonti, Cohen, D., Bronsard, G., Bonnot, O., Weismann-
E., Frysira, H., Haidopoulou, A., Asprangathou, D., Arcache, C., Botbol, M., Lauth, B., et  al. (2014).
et al. (2013). Evidence for treatable inborn errors Gene x environment interactions in autism spec-
of metabolism in a cohort of 187 Greek patients trum disorders:  role of epigenetic mechanisms.
with autism spectrum disorder (ASD). Front Hum Front Psychiatry 5, 53.
Neurosci 7, 858. Turunen, J.A., Rehnstrom, K., Kilpinen, H.,
Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic Kuokkanen, M., Kempas, E., and Ylisaukko-Oja, T.
diet as a treatment paradigm for diverse neuro- (2008). Mitochondrial aspartate/glutamate carrier
logical disorders. Front Pharmacol 3, 59. SLC25A12 gene is associated with autism. Autism
Stromland, K., Nordin, V., Miller, M., Akerstrom, B., Res 1, 189–192. doi:10.1002/aur.25
and Gillberg, C. (1994). Autism in thalidomide Voskuyl, R.A., and Vreugdenhil, M. (2001). Effects
embryopathy: a population study. Dev Med Child of essential fatty acids on voltage-regulated ionic
Neurol 36, 351–356. channels and seizure thresholds in animals.
Su, H., Fan, W., Coskun, P.E., Vesa, J., Gold, J.A., In Fatty Acids:  Physiological and Behavioral
Jiang, Y.H., Potluri, P., Procaccio, V., Acab, A., Functions, Mostofsky D., Yehuda S., Salem N.Jr .
Weiss, J.H., et  al. (2011). Mitochondrial dys- eds. (Totowa, NJ: Humana Press), pp. 63–78.
function in CA1 hippocampal neurons of the Weinberg, S.E., Sena, L.A., and Chandel, N.S. (2015).
UBE3A deficient mouse model for Angelman syn- Mitochondria in the regulation of innate and
drome. Neurosci Lett 487, 129–133. doi:10.1016/ adaptive immunity. Immunity 42, 406–417.
j.neulet.2009.06.079 Xavier, J.M., Rodrigues, C.M., and Sola, S. (2015).
Suen, D.F., Norris, K.L., and Youle, R.J. (2008). Mitochondria: major regulators of neural develop-
Mitochondrial dynamics and apoptosis. Genes ment. Neuroscientist 22(4), 346–358. doi: 10.1177/
Dev 22, 1577–1590. 1073858415585472.
Tang, G., Gutierrez Rios, P., Kuo, S.H., Akman, Zhao, Y., Fung, C., Shin, D., Shin, B.C., Thamotharan,
H.O., Rosoklija, G., Tanji, K., Dwork, A., Schon, S., Sankar, R., Ehninger, D., Silva, A., and Devaskar,
E.A., Dimauro, S., Goldman, J., et  al. (2013). S.U. (2010). Neuronal glucose transporter isoform
Mitochondrial abnormalities in temporal lobe 3 deficient mice demonstrate features of autism
of autistic brain. Neurobiol Dis 54, 349–361. spectrum disorders. Mol Psychiatry 15, 286–299.
doi:10.1016/j.nbd.2013.01.006 doi:10.1038/mp.2009.51
113

15
Glucose and Ketone Metabolism in the Aging Brain
Implications for Therapeutic Strategies to Delay
the Progression of Alzheimer’s Disease

STEPHEN C. CUNNANE, PHD, ALEXANDRE COURCHESNE- LOYER, MSC,


VALERIE ST- PIERRE, BSC, CAMILLE VANDENBERGHE, BSC,
ETIENNE CROTEAU, PHD, AND CHRISTIAN- ALEXANDRE CASTELLANO, PHD

INTRODUCTION brain’s predominant fuel, but, like other organs,


The impact of deteriorating cognitive function in the brain has a back-up fuel for occasions when
older age has become a major sociomedical pre- glucose supply is insufficient, for example, dur-
occupation worldwide. Alzheimer’s disease (AD), ing fasting, starvation, strenuous exercise, or mal-
the most common form of impaired cognitive nutrition. While other organs use free fatty acids
function in older age, is debilitating and dehu- directly to replace insufficient availability of glu-
manizing, and is expensive to manage. To make cose, ketones (also known as ketone bodies) are
matters worse, there is currently no effective treat- the only significant alternative fuel to glucose for
ment. Despite considerable effort and expense in the brain. The two ketones that replace glucose
the development of antiamyloid medications, for the brain are beta-hydroxybutyrate and aceto-
there is no clear sign that this treatment strat- acetate. As the decarboxylation product of aceto-
egy will be effective in the near future (Jack et al., acetate, acetone appears to mainly be excreted on
2009; Rafii and Aisen, 2009). Some major lifestyle breath but can also be metabolized through other
risk factors for AD are well known and include pathways.
type 2 diabetes, hypertension, and sedentarity. For over 30 years now it has become increas-
Whether inadequate intake of certain nutrients ingly well established that brain glucose uptake
such as omega-3 fatty acids and certain B vitamins and metabolism are defective in AD, a problem
also contributes to AD is receiving considerable that is particularly evident in the parietal and tem-
attention but remains unclear and controversial poral cortex (reviewed in Cunnane et  al., 2015;
(Douaud et al., 2013; Jerneren et al., 2015; Quinn Cunnane et al., 2011). These results have generally
et al., 2010). As a general rule, “prevention” strate- been interpreted to mean that low brain glucose
gies based on reducing the impact of lifestyle risk uptake in AD is a consequence of neuronal fail-
factors can be implemented and have been shown ure and death. While fewer functioning neurons
in clinical trials to be beneficial in reducing the would indeed diminish the need for glucose, this
progression of not only prodromal AD but also perspective does not account for multiple exam-
other lifestyle-related chronic diseases (Brown ples of conditions in which regional brain glucose
et al., 2010; Ngandu et al., 2015; Okonkwo et al., hypometabolism is present presymptomatically in
2014). We speculate that effective strategies to individuals at elevated risk of AD, that is, before the
delay the progression of AD may have at least clinical (cognitive) onset of the disease. An emerg-
one feature in common—that of improving brain ing literature is discussed here that provides sev-
energy metabolism in older people. eral clear examples in which individuals at genetic
It is well understood that the brain requires or lifestyle risk of AD have impaired brain glucose
a disproportionately large amount of energy; presymptomatically. Given that brain function is
whereas the adult brain represents about 2% of acutely dependent on a constant supply of glucose
adult body weight, it consumes about 20%–23% and oxygen, it is crucial to know whether latent
of whole body energy requirements. Glucose is the presymptomatic deterioration in brain glucose
114

114 section II: E merging Clinical Applications and Future Potential

uptake and/or utilization could be contributing to supervised starvation. This experiment was
the development of AD, because this could have done in three obese individuals who underwent
an impact on prevention and treatment strategies. total dietary deprivation for 40  days. At that
We have developed a positron emission time, it was suspected that some other fuel was
tomography (PET) research program using the helping to meet the brain energy requirements
ketone tracer, 11C-acetoacetate (AcAc), to bet- during extreme hypoglycemia because brain
ter understand the relation between brain fuel oxygen consumption was not decreased as much
uptake and brain function in people with or at as glucose uptake, but the replacement fuel was
risk of AD. In each individual studied, we use still unknown. The report by Owen et al. (1967)
this dual tracer PET protocol to compare the was the first to demonstrate that ketones were
brain uptake of 11C-AcAc with that of the glucose the unknown replacement fuel for glucose for
tracer, 18F-fluorodeoxyglucose (FDG). We quan- the brain, that is, that ketones played a physi-
tify the magnitude of glucose and ketone uptake ologically important role in the adult brain
regionally throughout the brain. Clinical studies rather than simply being a pathological marker
are underway to test the potential therapeutic of severe insulin deficiency in decompensated
utility of ketogenic supplements based primarily type 1 diabetes. This paper also proposed that
on medium chain triglycerides (MCT), with the brain ketone uptake was proportional to blood
dual tracer PET imaging done before and after ketone concentration. Owen et  al. (1967) did
the interventions. Here, we review the rationale not describe brain ketone uptake under less
for these studies with the objective of highlighting extreme conditions, but shortly after their paper
the main points supporting a strategy that impli- was published, Gottstein et al. (1971) used AVD
cates maintenance of brain fuel supply to delay the to show that the brain also consumed ketones
onset and/or progression of AD. under the more physiological conditions of a
12- to 16-hour overnight fast. Hence, ketones
A R T E R I OV E N O U S contribute to brain energy metabolism on a
DIFFERENCE STUDIES daily basis, not just when brain glucose availa-
ACRO S S THE BRAIN: THE bility is severely limited during chronic hypo-
F O U N D AT I O N glycemia. Drenick et  al. (1972) were the first
The PET-FDG protocol was first used to image to note that the symptoms of severe glucopenia
brain glucose metabolism in the late 1970s with (as low as 0.5 mM plasma glucose) induced by
reports on AD first being published in the early controlled insulin infusion could be avoided
1980s (Benson et al., 1983). Since then, PET-FDG by up to 60  days of medically supervised star-
has been a cornerstone of human and animal vation. Their study showed that ketones could
studies on brain energy metabolism in aging and account for ≥85% of brain fuel requirements
AD (reviewed in Cunnane et  al., 2011). Indeed, in obese adults. Drenick et al. (1972) also used
without PET-FDG, it is doubtful that brain glu- AVD to show that skeletal muscle of the forearm
cose hypometabolism in AD would have become was not using ketones at all during prolonged
widely studied, because the only option besides starvation. These starvation studies were clearly
PET, the arteriovenous difference (AVD) method, extreme, but they unequivocally demonstrated
is highly invasive and used increasingly less in the physiological role of ketones particularly if
research. Nevertheless, it was the AVD method not uniquely for the brain.
that first showed that brain oxygen and glucose The first AVD report on brain energy metabo-
uptake were unaffected by healthy aging (Dastur, lism in AD was by Lying-Tunell et al. (1981), who
1985). This method produced several ground- showed that brain glucose uptake was impaired
breaking reports comparing brain ketone and by 26% but that brain ketone uptake was still nor-
glucose uptake that laid the foundation for our mal (Table 15.1). This report confirmed earlier
current understanding that ketones are an essen- reports using AVD that had demonstrated lower
tial physiological fuel working daily in tandem brain glucose uptake in AD and provided the
with glucose to assure brain energy requirements first suggestion that the disease did not necessar-
are being met. ily adversely affect the brain ketone utilization to
The first of these pioneering AVD reports the same extent as glucose, even when the cogni-
was by Owen et  al. (1967), who showed that tive deficit became severe. Hoyer et al. (1988) also
ketones could supply about two-thirds of the used the AVD method and demonstrated 45%
brain’s fuel requirement when glucose avail- lower brain glucose utilization in suspected (i.e.,
ability was severely limited during medically early stage) AD. Unlike in more advanced AD,
115

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 115

TABLE 15.1 LOW BRAIN GLUCOSE UPTAKE BUT NORMAL BRAIN BETA-


HYDROXYBUT YRATE UPTAKE IN MILD ALZHEIMER’S DISEASE COMPARED
WITH AGE- MATCHED HEALTHY CONTROLS. MEASUREMENTS WERE MADE BY ARTE-
RIOVENOUS DIFFERENCE

Fuel Controls Mild Alzheimer’s p value (versus Control) Reference

Glucose 24.8 18.7 <0.01 Lying-Tunell et al., 1981


(µmol/100 g/min) 24.9±7.2 11.6±4.0 <0.01 Ogawa et al., 1996
Ketones 0.6 0.45 ns Lying-Tunell et al., 1981
(µmol/100 g/min) 0.14±0.08 0.11±0.06 ns Ogawa et al., 1996

ns—not significant between Controls and mild Alzheimer’s

they showed that neither blood flow to the brain CMR of glucose (CMRg) is measured less and
nor oxygen uptake was adversely affected early in less because, ideally, it requires blood sampling
AD. Hoyer et al. (1988) surmised from their results from a catheter inserted into the brachial artery
that the problem with brain glucose utilization in as well as exhaustive data processing. As a result
AD probably involved impaired glycolysis, par- of declining quantification of CMRg, the actual
ticularly the conversion of glucose to pyruvate via deficit in brain glucose uptake in people at risk
pyruvate dehydrogenase (Sorbi et al., 1983). Hoyer of AD is now only infrequently measured; rather,
et al. (1992; 1988) were the first to conceptualize PET-FDG data are increasingly expressed using
the significance of lower brain glucose utilization a statistical parametric map. Statistical paramet-
as an early and possibly presymptomatic problem ric mapping clearly identifies the regions of the
in AD. However, they did not report brain ketone brain with FDG uptake below a certain statistical
uptake. threshold, but the actual magnitude of that dif-
The second pioneering AVD paper that ference compared with the controls is generally
addressed brain energy metabolism in AD was unknown (Baker et  al., 2011; Kalpouzos et  al.,
by Ogawa et al. (1996), who confirmed the report 2009). Hence, although the regional nature of
by Lying-Tunell et  al. (1981) showing that AD the deficit in brain FDG uptake in AD can be
adversely affected brain glucose but not brain clearly observed in some detail with PET (which
ketone uptake (Table 15.1). Unlike Lying-Tunell is impossible with AVD), there are fewer and
et  al. (1981) but similar to Hoyer et  al. (1988), fewer quantitative PET-FDG reports, so we have
Ogawa et  al. (1996) found that brain oxygen actually learned very little about the magnitude
uptake was less adversely affected in AD than of the regional deficit in brain glucose uptake in
brain glucose uptake. These AVD studies raised AD or how it changes as AD progresses since
the possibility that brain glucose hypometabo- the AVD studies and insights of Hoyer et  al.
lism was a relatively early problem in AD, and that (1988). Furthermore, with the focus on glucose
this problem was apparently not seen with brain as the representative brain fuel using PET-FDG,
ketone utilization. until recently, the possibility that brain ketone
While AVD is quite invasive, it is also a uptake could be less affected in AD and hence
robust method that permits quantification of possibly part of a treatment strategy has largely
global brain energy metabolism and is still in use been overlooked. However, knowing the magni-
(Dalsgaard et al., 2004; Ide et al., 2000). The PET- tude of the deficit in brain glucose uptake in AD
FDG data were initially expressed as the “cerebral and the extent to which it continues to worsen
metabolic rate” (CMR) of glucose. These units without treatment provides critical information
matched those used with AVD, which provided about existing treatments that may fortuitously
a reference method against which early PET- improve brain glucose uptake, namely, acetyl-
FDG results could be calibrated (Sokoloff et al., cholinesterase inhibitors (Nordberg, 2006)  or
1977). Both methods converge on the point that NMDA receptor antagonists (Wang et  al.,
the AD brain is glucose-deprived and at risk of 2013), or new treatments aiming to bypass or
starvation or exhaustion (Cunnane et  al., 2011; correct this problem early enough to delay the
Hoyer et al., 1988; Mamelak, 2012). However, the progression of AD.
116

116 section II: E merging Clinical Applications and Future Potential

P R E S Y M P T O M AT I C at elevated risk of the disease but in whom cogni-


BRAIN GLUCOSE tive scores are still normal.
H Y P O M E TA B O L I S M : Carriers of the presenilin-1 mutation are at
M U LT I P L E E X A M P L E S very high, almost certain, risk of developing AD
While AVD has largely become a technique of the (Blennow et al., 2006; Fox et al., 1997; Scholl et al.,
past, it helped seed our interest in applying PET- 2011). Carriers of the apolipoprotein E4 allele and
FDG to the question of causality—is brain glucose persons with a family history of AD, especially on
hypometabolism a consequence of AD or part of the maternal side, are also at increased risk of AD
the cause or, indeed, both? The stage of AD can (Bu, 2009; Kennedy et  al., 1995; Mosconi et  al.,
be difficult to determine precisely, which in turn 2005; Reiman et al., 2004), although their risk is
means that it can be difficult to determine whether still relatively low compared with presenilin-1
those on the threshold or at the early stages of carriers. Type 2 diabetes ranks among the most
the disease have a specific deficit in brain glu- important of the nongenetic or lifestyle risk fac-
cose uptake or a broader deficit that also includes tors for AD. In presymptomatic individuals with
impaired oxygen uptake and lower blood flow to these aforementioned conditions in whom the
the brain. Assessing the presence of a possible pre- results on cognitive batteries are still normal,
symptomatic neurometabolic deficit in a disease PET-FDG clearly shows that regional brain glu-
of uncertain and gradual onset like AD therefore cose hypometabolism is present before the onset
requires a different strategy such as studying those of cognitive deficit (Table 15.2). These examples

TABLE 15.2 PRESYMPTOMATIC BRAIN GLUCOSE HYPOMETAB OLISM IN PERSONS


AT RISK OF ALZHEIMER’S DISEASE (AD)

Group at risk of AD Mean age Brain region Magnitude of brain Reference


(y) affected glucose hypometabo-
lism (% difference from
control)

Young adult carriers of 30 Posterior cingulate –14 to –25 Schöll et al., 2011
Presenilin-1 Parietal cortex
Temporal cortex
Young adult carriers of 31 Parietal cortex –9 to –11 Reiman et al., 2004
Apolipoprotein-E4 Temporal cortex
Posterior cingulate
Prefrontal cortex
Maternal family history 43 Parietal cortex –12 to –21 Mosconi et al.,
of AD Temporal cortex 2006
Hippocampus
Entorhinal cortex
Posterior cingulate
Prediabetic older persons 74 Temporal cortex N/A Baker et al., 2011
Parietal cortex
Posterior cingulate
Precuneus
Prefrontal cortex
Insulin resistant young 25 Frontal cortex –9 to –14 Castellano et al.,
women with polycystic Middle temporal 2015
ovary syndrome cortex
Cognitively healthy older 72 Frontal cortex –10 to –18 Nugent et al., 2015
adults Temporal cortex
Anterior cingulate
Putamen
Thalamus

N/A—not available
117

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 117

are a good basis for concluding that presymptom- a test of working memory, suggesting that insulin
atic brain glucose hypometabolism can be pres- resistance can adversely affect both brain glucose
ent and could therefore be contributing to the metabolism and possibly cognitive performance
development of AD. in young adults (Castellano et al., 2015a). Whether
Aging is considered to be the most significant women with polycystic ovary syndrome are pre-
risk factor for AD, so we have extensively stud- disposed to a higher risk of cognitive decline as
ied brain energy metabolism in cognitively nor- they age remains to be seen but deserves further
mal older persons. Using quantitative PET-FDG, attention.
we have shown that in cognitively normal people These examples demonstrate that regional
aged 72 years, the CMRg in the superior frontal brain glucose hypometabolism can be present
cortex is about 35±5 instead of the 40±7 mmol/ in those in whom cognition is still normal even
100 g/min seen in young adults, that is, the glu- though they are at risk of AD due to old age, or
cose uptake deficit in this brain region is of the certain genetic or lifestyle factors associated with
order of 12%–13% (Nugent et  al., 2015; Nugent AD. The presymptomatic glucose uptake defi-
et al., 2014a; Nugent et al., 2014b). The CMRg did cit is commonly but not exclusively in the fron-
not differ significantly between young and older tal cortex, and its magnitude is of the order of
adults in either white or gray matter as a whole 12%–15% (Castellano et al., 2015b; Cunnane et al.,
nor did it differ in the temporal or parietal cor- 2011; Nugent et al., 2015). These observations run
tex where CMRg decreases in AD. Lower CMRg counter to the widespread perception that brain
that is present mostly if not exclusively in the glucose hypometabolism is uniquely a conse-
frontal cortex of cognitively normal older people quence of synaptic dysfunction and neuronal fail-
has been reported on several previous occasions ure or death. Indeed, they suggest that a vicious
(Cunnane et  al., 2011; Kalpouzos et  al., 2009). cycle can develop in which latent presymptomatic
Thus, lower CMRg in the temporal and parietal brain glucose hypometabolism develops and leads
cortex in AD is a fundamentally different situa- to chronic brain energy deficit, deteriorating neu-
tion (but perhaps related to) lower frontal CMRg ronal function, further decline in demand for glu-
during aging. Incidentally, in older people with cose, and further cognitive decline (Figure 15.1;
normal cognition, cortical thickness and regional Cunnane et  al., 2011). The presence of presymp-
volumes are lower in many more brain regions tomatic brain glucose hypometabolism does not
than CMRg (Nugent et al., 2015). Thus, although necessarily mean that it is a cause of AD, nor that
lower frontal CMRg is clearly presymptomatic in neuronal function is necessarily normal, nor even
relation to aging-associated cognitive decline, it that glucose hypometabolism is the first abnormal-
is by no means the only or even the most signifi- ity detectable in those at risk of AD. Nevertheless,
cant change occurring in the brain during aging. knowing that brain glucose hypometabolism can
We have also observed a deficit of about 14%
in CMRg in the superior and middle frontal cor-
tex in 24-year-old women with polycystic ovary Latent brain glucose
syndrome (Castellano et  al., 2015a). Polycystic hypometabolism:
ovary syndrome is a multifactorial endocrine ↓ glycolysis
↓ brain glucose uptake
disease involving infertility, hyperandrogenism,
and mild-to-moderate insulin resistance. It was
the mild insulin resistance that was of particu- Ketones

lar interest to us because of its association with Deteriorating brain


increased risk of AD (Baker et  al., 2011; Craft, function:
↓ ATP generation
2009, 2012; Matsuzaki et  al., 2010; Ronnemaa Alzheimer’s
↓ neurotransmitters
disease
et al., 2008; Schrijvers et al., 2010). These women
with polycystic ovary syndrome were of nor- Cognitive symptoms:
mal weight and body-mass index, so this condi- ↓ memory
↓ executive function
tion represents a possible model of the impact of ↓ processing speed
mild insulin resistance on brain glucose metabo-
lism that is independent of age and obesity. The FIGURE 15.1 Schematic representation of latent
mild insulin resistance in women with polycystic brain glucose hypometabolism leading to a vicious cycle
ovary syndrome was significantly inversely cor- of accelerating metabolic deterioration and neuronal
related to CMRg in several brain regions. These dysfunction that increases the risk of developing Alzh-
women also had borderline low-normal scores on eimer’s disease (from Cunnane et al., 2015).
118

118 section II: E merging Clinical Applications and Future Potential

be present in people at risk of AD before the onset In adults, ketogenesis is principally from long
of measurable clinical (cognitive) deficit has impli- chain fatty acids stored in adipose tissue, the release
cations for potential therapeutic strategies (Blass, of which is controlled by insulin. The branched
2008; Cunnane et al., 2015; Cunnane et al., 2011; chain amino acids, isoleucine and leucine, are
Gibson et al., 2000; Henderson et al., 2009; Veech also ketogenic (Mitchell et al., 1995). During fast-
et al., 2001). ing, blood glucose and insulin decrease, which
releases the inhibition on lipolysis in adipose tis-
B R A I N G L U C O S E U P TA K E sue, thereby allowing plasma free fatty acids to
O R M E TA B O L I S M O R   B O T H ? increase. This increase in plasma free fatty acids
The AVD studies show the magnitude of the helps meet the need for an alternative fuel to glu-
lower glucose “disappearance” into the brain in cose for most tissues with the notable exception of
AD. However, they do not establish whether the the brain. The increased supply of fatty acids enter-
problem is with brain glucose uptake, that is, its ing the liver leads to ketogenesis by condensation
transport into the brain, or with glycolysis, that is, of two acetyl-CoAs, which are present in excess
glucose metabolism within the brain, or both. The due to fatty acid beta-oxidation (Figure 15.2).
PET studies clearly show that there is a problem in Implicitly, if insulin controls the ebb and flow
AD at the level of glucose (FDG) uptake, that is, of plasma glucose, free fatty acids, and ketones,
with glucose transport into the brain and its con- insulin sensitivity becomes an important param-
version to glucose-6-phosphate by hexokinase. eter in the process. Indeed, conditions involving
However, PET-FDG provides no information decreased insulin sensitivity, that is, insulin resis-
about whether, in addition to defective glucose tance, also impair tissue glucose uptake and are
transport, the glycolytic steps are also altered. It associated with a vicious cycle of hyperglycemia,
may not matter to the brain whether the prob- hyperinsulinemia, and an eventual trend toward
lem with glucose hypometabolism is with brain type 2 diabetes, which is a major risk factor for
glucose uptake per se or with glucose metabo- AD (Baker et al., 2011; Craft, 2009). Postprandial
lism via glycolysis; either way, if uncorrected, the hyperinsulinemia inhibits fatty acid release from
brain’s main energy source is compromised and it adipose tissue and ketogenesis, which is appro-
is at increased risk of chronic energy deficit and priate as long as insulin sensitivity and tissue
eventual exhaustion. In fact, the energy supply glucose uptake are normal, that is, when and insu-
problem seems to be at the level of both brain lin returns to normal 3–4 hours postprandially.
glucose uptake and metabolism. In vitro studies However, chronic sedentarity commonly leads
show that several enzymes in glycolysis are mark- to chronic hyperinsulinemia and insulin resis-
edly impaired in AD, including phosphofructo- tance, which not only compromise tissue glucose
kinase (Bowen et al., 1979; Iwangoff et al., 1980), uptake but also ketogenesis and ketone metabo-
alpha-ketoglutarate dehydrogenase complex lism (Bickerton et al., 2008; Fukao et al., 2004). In
(Gibson et  al., 2000), and pyruvate dehydrogen- effect, this puts the aging brain in double jeopardy
ase (Cunnane et al., 2011; Perry et al., 1980; Sorbi because now it is not only getting insufficient glu-
et al., 1983). cose but is also getting less of the main alternative
fuel, ketones (Cunnane et al., 2015; Cunnane et al.,
K E TO N E S — T H E B R A I N ’ S 2011; Mamelak, 2012).
PHYSIOLOGICAL
A LT E R N AT I V E   F U E L K E TO N E K I N E T I C S
The now classic studies by Owen et al. (1967) and AND TRANSPORT
Drenick et  al. (1972) demonstrated that ketones I N   A D U LT   H U M A N S
are the main reserve fuel for the brain when glu- Elegant kinetic studies show that ketone utilization
cose supply is compromised by starvation. The essentially matches synthesis in normal weight
human liver can produce ketones at a rate of 100– adults fasted overnight (Avogaro et  al., 1990;
150 g/day (Flatt, 1972; Reichard et al., 1974), which Balasse and Fery, 1989; Hall et al., 1984). The rapid
is more than sufficient to account for the brain’s utilization of ketones as they are produced during
ketone utilization even during prolonged star- short-term fasting generally keeps plasma ketones
vation. The energy cost to the liver of producing ≤0.3 mM (Table 15.3). Exercise for 30 minutes has
ketones is mostly supplied by gluconeogenesis, the little or no effect on ketone synthesis, utilization or
rate of which parallels and may eventually actu- clearance. After 3–5 days fasting, plasma ketones
ally limit ketone production (Flatt, 1972; Garber rise about 10-fold due mainly to increased synthe-
et al., 1974). sis and lower clearance. In obese adults, overnight
119

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 119

O O
+ 2 Acetyl-CoA
H3C SCoA H3C SCoA

Thiolase

O O
Acetoacetyl-CoA
H3C SCoA

HMG-CoA synthase

O OH O
HMG-CoA
O SCoA
CH3

HMG-CoA lyase

O O
Acetoacetate
O CH3

Nonenzymatic D-beta-hydroxybutyrate
decarboxylation dehydrogenase

O O OH

H3C CH3 O CH3

Acetone D-beta-hydroxybutyrate

FIGURE 15.2 Ketogenesis arising from the condensation of two acetyl-CoAs to form the ketones, acetoacetate, beta-
hydroxybutyrate, and acetone. HMG-CoA: 3-hydroxy-3-methylglutaryl-coenzyme A.

or 3–5 days fasting has little to no effect on these the liver. However, during extended fasting, free
kinetic parameters, but synthesis may exceed uti- fatty acids compete with ketones and become the
lization, thereby raising plasma ketones a bit more main fuel for some tissues such as skeletal mus-
than in normal weight adults. In type 1 diabetes, cle, leaving most if not all of ketone production
12 hours of fasting increases plasma ketones about available to complement the available glucose in
10-fold more than in nondiabetic adults, but it is meeting the energy needs of the brain (Drenick
not clear why, as utilization appears to keep pace et  al., 1972; Owen and Reichard, 1971). Ketone
with synthesis, both of which are similar to values transport into tissues occurs via monocarboxylic
seen in nondiabetic adults after 12 hours of fasting acid transporters, of which there are at least six
(Table 15.3). subtypes (Simpson et  al., 2007). Several of the
As far as is known, ketones can be trans- monocarboxylic acid transporters are expressed
ported into and catabolized by all tissues except in the brain. Monocarboxylic acid transporter
120

120 section II: E merging Clinical Applications and Future Potential

TABLE 15.3 KETONE (BETA- HYDROXYBUT YRATE + ACETOACETATE) KINETICS


IN HUMANS SUMMARIZED FROM A SERIES OF PUBLISHED STUDIES

Condition Fasting Plasma Utilization Synthesis Metabolic Urinary


period ketones (µmol/kg/ (µmol/kg/ clearance excretion
(mM) min) min) (mL/kg/min) (µmol/min)

Healthy adults 12–16 ha,f,g 0.1–0.3 3–5 2–5 18 —


3 dd 2.5 — 10 — 4
Healthy adults 16 hf,j 0.2–0.4 6 6 21 —
+ 30 min exercise 3–5 df,h,j 4-5 20 22 4–6 —
Obesity 12–16 ha,c 0.4–0.7 4 3–8 12 0.4
≥ 3 db,c,e,i 3-7 9 5–25 3 80–240
Type I diabetes 12 ha 2.5 6 6 — —

h—hours
d—days
A
(Hall et al., 1984)
B
(Reichard et al., 1974)
C
(Balasse, 1979)
D
(Garber et al., 1974)
E
(Owen et al., 1967)
F
(Fery and Balasse, 1983)
G
(Avogaro et al., 1990)
H
(Balasse et al., 1978)
I
(Balasse, 1986)
J
(Fery and Balasse, 1986)

expression responds rapidly to hyperketonemia “pushed” into the brain in proportion to their
(Pan et  al., 2001), so, under normal conditions, plasma ketone concentration, which usually only
brain uptake of ketones is directly proportional increases when plasma glucose decreases. This
to their plasma concentration over about a 600- contrasts with glucose, which is “pulled” into
fold range of beta-hydroxybutyrate (0.02–12 the brain in proportion to its utilization. Hence,
mM; Courchesne-Loyer et  al., 2013; Cunnane the “push-pull” strategy assures that under con-
et  al., 2015; Cunnane et  al., 2011). However, ditions in which glucose availability decreases,
as with brain glucose uptake, the transport of ketone synthesis would normally be stimulated
ketones into the brain may not be the limiting and the brain’s energy supply would be main-
variable in their uptake. Rather, ketones are tained (Figure 15.3).

Blood Brain
Glucose:
Metabolic demand
Glucose Glucose

Ketones
Ketones Ketones:
Plasma availability

FIGURE 15.3 The contrasting “push-pull” mechanism of brain fuel supply. Glucose is pulled from the blood into the
brain as a function of the brain’s metabolic demand during neuronal activation. Under normal conditions (excluding
insulin resistance), ketones are pushed from the blood into the brain in direct proportion to their plasma concentration.
121

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 121

R E G I O NA L B R A I N K E TO N E 2011; Roy et  al., 2012)  studies. This dual tracer


U P TA K E I N   E A R LY technique allows for a quantitative comparison of
ALZHEIMER’S DISEASE brain uptake of glucose and ketones with a delay of
Positron emission tomography provides a means no more than 2 hours between the two tracer infu-
of directly quantifying and comparing brain glu- sions, which avoids the potentially greater biologi-
cose and ketone uptake. Using PET, Blomqvist cal variability between scans done on different days.
et al. (2002; 1995) reported the brain metabolism This dual tracer technique permitted us to com-
of 11C-beta-hydroxybutyrate in humans and con- pare brain uptake of FDG and 11C-AcAc in early
firmed the earlier AVD studies showing that the AD (Castellano et al., 2015b). The aim was three-
uptake was directly proportional to plasma beta- fold:  (1)  to confirm the AVD reports of normal
hydroxybutyrate even at very low plasma ketone ketone but low brain glucose uptake early in AD
concentrations (Cunnane et al., 2011; Lying-Tunell (Ogawa et al., 1996), (2) to assess brain fuel metab-
et  al., 1981; Figure 15.4). We found that synthe- olism in early AD rather than the more advanced
sizing 11C-AcAc was easier than synthesizing 11C- stages previously reported (Lying-Tunell et  al.,
beta-hydroxybutyrate (Tremblay et al., 2007), and 1981), and (3)  to quantify the regional pattern
since acetoacetate is the ketone that actually enters of brain uptake of both fuels under postprandial
the mitochondria and is catabolized to acetyl CoA, conditions. We confirmed that global brain FDG
we worked with it as our brain ketone PET tracer. uptake (CMRg) was 14% lower in early AD versus
We developed a PET protocol in which 11C-AcAc is cognitively normal controls, and that this global
injected intravenously first, followed by a wash-out deficit was primarily confined to the parietal cortex,
period, and then FDG is injected. We have applied posterior cingulate, and thalamus. However, nei-
this protocol in human (Castellano et  al., 2015b; ther 11C-AcAc uptake (CMRa) nor the acetoacetate
Nugent et  al., 2014b) and animal (Pifferi et  al., uptake constant (Ka) were significantly different

100
100 metabolism as ketones
(log µmol/100 g/min)

10 % of total brain
CMR ketone

1
10

0.1

0.01 1
0.01 0.1 1 10
Plasma ketones
(log mmol/L)
FIGURE 15.4 Direct, linear relation between plasma β-hydroxybutyrate (β-HB), brain ketone uptake (left-hand Y
axis), and percentage contribution to total brain energy requirement (right-hand Y axis) in adults. Two relationships
are shown, one for plasma β-HB versus the rate of brain β-HB uptake (solid line, R2 = 0.97; Y = 1.57X - 0.20; p < .0001),
and the other for plasma acetoacetate versus the rate of brain acetoacetate uptake (Castellano et  al., 2015b) (dotted
line, R2 = 0.83; Y = 3.46X - 0.03; p < .0001): Alzheimer’s disease (AD), ⚬ cognitively healthy age-matched controls.
Units are the same for both ketones—cerebral metabolic rate (CMR; μmol/100 g/min). β-HB data have been combined
from several sources: postprandial state (Blomqvist et al., 1995), (Blomqvist et al., 2002) β-HB infusion, AD

and healthy older controls(Lying-Tunell et al., 1981), 40-day fast (Owen et al., 1967), 60-day fast(Drenick et al.,
1972), and AD and healthy older controls (Ogawa et al., 1996; indicated by the arrow). All the β-HB data are from
arteriovenous difference studies except for one report, which used β-HB-PET (Blomqvist et al., 1995). The AcAc data
were obtained using 11C-AcAc PET (Castellano et al., 2015b). Each symbol represents a single individual except when

not available in the original publication: Drenick et al. (1972), for which represents the mean of n = 5 participants,
and Ogawa et al. (1996), for which and both represent the mean of n = 7. The relationship between plasma β-HB
and the percent of brain energy consumption supplied by β-HB in adults is as follows: at plasma β-HB values around
0.1 mM, ketone supply >5% of brain energy; at 1 mM β-HB, they supply about 10%–15%; at 5–7 mM β-HB, 50%–65%
and over 7–8 mM β-HB, >75% of brain energy consumption. For a given plasma AcAc concentration, AcAc is taken up
by the brain more rapidly than β-HB, which is why the dotted regression line lies above that of the solid line for β-HB.
122

122 section II: E merging Clinical Applications and Future Potential

Age-matched controls Mild Alzheimer


50 0.6

Brain 11C-AcAc uptake


Brain 18F-FDG uptake
*

(µmol/100 g/min)
(µmol/100 g/min)
40
0.4
30
20
0.2
10
0 0.0

0.08 0.05

Brain 11C-AcAc
0.04
Brain 18F-FDG
uptake constant

uptake constant
0.06
0.03

(min–1)
(min–1)

0.04
0.02
0.02 0.01
0.00 0.00

FIGURE  15.5 Whole-brain 18F-fluorodeoxyglucose (18F-FDG) uptake is 14% lower in mild Alzheimer’s disease
(white bars; n = 10) compared with cognitively normal age-matched controls (black bars; n = 30; * p = .003). In contrast
to FDG, whole-brain 11carbon-acetoacetate (11C-AcAc) uptake is not significantly different in mild Alzheimer’s disease
compared with cognitive normal, age-matched controls (from Castellano et al., 2015b).

in the brain as a whole or in any brain region in (1996). Unlike with FDG, the 11C-AcAc PET tracer
AD versus the age-matched controls (Figure 15.5). is chemically identical to acetoacetate produced by
The kinetics of 11C-AcAc metabolism (or 11C-beta- the body, so it is possible to interpret our results as
hydroxybutyrate metabolism; Blomqvist et  al., showing that both uptake and metabolism of 11C-
1995) suggest a one-compartment model in which AcAc were normal in early AD. Unlike for glucose,
brain utilization essentially matches brain uptake. which generates ATP by both oxidative phospho-
Plasma acetoacetate and CMRa were significantly rylation and aerobic glycolysis, production of ATP
positively correlated, the slope of which did not from ketones is exclusively via the citric acid cycle
differ between controls and early AD. This implies and oxidative phosphorylation. Since we observed
that brain ketone utilization in AD was still propor- that brain 11C-AcAc metabolism was normal in
tional to plasma concentration on the same slope early AD, our results with brain ketone PET there-
as in controls (Figure 15.6). These results confirm fore indirectly support the speculation by Hoyer
those of Lying-Tunell et al. (1981) and Ogawa et al. et  al. (1988) that oxidative phosphorylation may
still be normal early in AD. These observations
provide a rationale for the concept that if provided
1.0
Mild Alzheimer with more ketones, the aging brain would be able to
Age-matched controls use them which, in turn, might help it overcome its
Brain 11C-AcAc uptake

0.8
(µmol/100 g/min)

deficit in glucose uptake and metabolism, thereby


0.6 delaying brain energy exhaustion and decreasing
the risk of cognitive decline and AD.
0.4

0.2 K E TO G E N E S I S
FROM MEDIUM CHAIN
0.0 T R I G LY C E R I D E S :   D O S E -
0 50 100 150 200 250 R E S P O N S E R E L AT I O N S H I P
Plasma AcAc (µM) In adults on a normal mixed diet, fasting induces
FIGURE  15.6 The direct, linear relationship between ketosis because long chain fatty acids are beta-
plasma ketone (acetoacetate; AcAc) concentration and oxidized by the liver to acetyl CoA. Long chain
brain ketone uptake is not altered in mild Alzheimer’s fatty acids are of 14–22 carbons in length and are
disease (white triangles, dotted line; n  =  10) compared essentially the only fatty acids found in adipose tis-
with cognitively normal age-matched elderly (black sue of adults. However, breast-fed infants achieve
squares, solid line; n = 30) (from Castellano et al., 2015b). mild ketosis in part due to the presence in breast
123

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 123

1200

Maximal plasma BHB (µM)


1000

800

600

400

200

0
0 10 20 30 40 50 60 70 80
MCT dose (g)
FIGURE  15.7 Direct relationship dose of medium chain triglycerides (MCT) and maximal observed plasma β-
hydroxybutyrate [BHB] (r = 0.95; p < .0001) in young adults unless otherwise specified. MCT were given as a single or
multiple doses*: ( Alzheimer; 10 g MCT; n = 77, ⚬ Alzheimer; 20 g MCT*; n = 77; Henderson et al., 2009), ( mild
cognitive impairment, AD; 40 g MCT; n = 10; Reger et al., 2004), (× type 1 diabetes; 40 g MCT; n = 11; Page et al., 2009),
( 10 g MCT emulsion; n = 10; 20 g MCT emulsion, n = 10, 30 g MCT emulsion; n = 10, 10 g neat MCT; n = 10;
20 g neat MCT; n = 10; 30 g neat MCT; n = 10; Courchesne-Loyer et al., unpublished), ( young adults; 71 g MCT

drink; n = 22; older adults 71 g MCT drink; n = 14; Freemantle et al., 2009), (✳ 48 g; n = 7; MCT; Seaton et al., 1986),
and (+ 70 g MCT; n = 14; Pi-Sunyer et al., 1969).

milk of medium chain fatty acids, that is, fatty acids studies show similar results whether a single or
of 6–12 carbons (Cunnane and Crawford, 2014). multiple MCT doses are given, probably because
Medium chain fatty acids are mostly absorbed of rapid plasma ketone clearance.
through the portal vein, hence gaining direct and Healthy older people have the same level
more rapid access to the liver than long chain fatty of hyperketonemia and 13C-ketone oxidation to
acids, which are absorbed into the peripheral cir- 13
C-CO2 after a standard high-fat ketogenic break-
culation via the lymph. Medium chain fatty acids fast containing MCTs as middle aged or young
are also beta-oxidized without needing to be acti- adults (76  years old versus 50 or 23  years old,
vated by carnitine. The net result is rapid beta- respectively; Freemantle et al., 2009; Figure 15.8).
oxidation and ketogenesis. With rare exceptions, Indeed, hyperketonemia after 18 hours of fasting
there is normally no further opportunity to con- may actually be somewhat higher in the seventh
sume medium chain fatty acids from the diet once to eighth decade of life compared with younger
breast-feeding is terminated. The exceptions are adults (London et al., 1986). Hence, the capacity to
coconut oil and palm kernel oil, in which medium produce and utilize ketones does not appear to be
chain fatty acids make up about 15% and 10% of reduced and may actually be increasing somewhat
the fatty acid composition, respectively. The frac- during healthy aging. These studies did not distin-
tion of these oils that contains these medium guish between ketone utilization by the brain ver-
chain fatty acids can be concentrated, resulting in sus other organs, but our brain ketone PET results
a generic product containing mostly fatty acids of confirm that the brain has similar ketone uptake
eight (octanoic or caprylic acid) and 10 carbons and utilization in healthy older compared with
(decanoic or capric acid) called medium chain tri- younger persons (Nugent et al., 2014b).
glycerides (MCTs). The ratio of these two principal
fatty acid components and their proportion of the CLINICAL STUDIES
total can vary widely from one MCT product to O F   H Y P E R K E TO N E M I A
another. The MCTs are well known to be ketogenic IN CONDITIONS
(Bach and Babayan, 1982; Freund and Weinsier, OF COGNITIVE DEFICIT
1966). Notwithstanding the generic nature of Several products based on MCTs are now available
MCTs and different study designs to assess their on the market or by prescription in the United States
metabolism, there is a remarkably good correla- and in Europe. The arrival of these products was
tion in the literature between the maximal plasma greatly stimulated by reports that MCTs had benefi-
ketone levels achieved on oral doses of MCTs from cial effects on cognitive outcomes in mild-moderate
10 to 70 g (Figure 15.7). These dose-response AD after a single dose (Reger et  al., 2004)  or
124

124 section II: E merging Clinical Applications and Future Potential

0.4

Plasma β-HB change vs. young (mM)


0.2

0.0

–0.2

–0.4
0 1 2 3 4 5 6
Time (h)

FIGURE  15.8 Plasma β -hydroxybutyrate (β-HB) response after a high-fat meal in a middle-aged group (gray
triangles; 50 years old; n = 18) amd older group (white circles; 76 years old; n = 14) compared with young adults (dotted
line; 23 years old; n = 22) (from Freemantle et al., 2009).

with regular consumption over several months the adult brain (Drenick et al., 1972; Owen et al.,
(Henderson et  al., 2009). Improved cognitive out- 1967) but glucose is still essential in the process. In
comes after MCTs are also seen in type 1 diabetics addition to being the main engine to generate ATP,
during controlled acute hypoglycemia caused by the citric acid cycle also spins off molecules needed
insulin infusion (Page et al., 2009). The very high- for brain function including the neurotransmitters
fat ketogenic diet has been reported to have a simi- GABA and acetylcholine, a process known as cat-
lar beneficial effect on cognitive and cardiovascular aplerosis (Owen et  al., 2002). Cataplerosis would
outcomes in mild cognitive impairment, the pro- rapidly deplete the citric acid cycle of its inter-
dromal state to AD (Krikorian et al., 2012). These mediates except that, via oxaloacetate, glucose
reports complement the studies showing that auto- supplies carbon to replace those intermediates, a
nomic and neurological symptoms of acute severe process known as anaplerosis. Hence, glucose is
experimental hypoglycemia and starvation can be not only a major fuel but also controls the balance
avoided by ketone infusion (Table 15.4). between anaplerosis and cataplerosis, that is, the
Acetoacetate and beta-hydroxybutyrate can net flow of carbon out of the citric acid cycle via
also be directly administered orally or by infusion. products other than CO2. Like glucose, ketones
This requires that they be provided as salts or as generate ATP via the citric acid cycle and oxida-
esters (Clarke et al., 2012; Hasselbalch et al., 1996; tive phosphorylation, but, unlike glucose, ketones
Plecko et  al., 2002), with the esters being more do not contribute to anaplerosis (Brunengraber
practical for long-term use. The safety (Clarke and Roe, 2006). In fact, ketones are cataplerotic
et al., 2012) of a beta-hydroxybutyrate-monoester because they increase citric acid cycle activity (Roy
and its anecdotal utility in improving some aspects et al., 2015) but do not provide carbon to replace
of cognitive function in an advanced case of early- intermediates in the cycle. A metaphor would be
onset Alzheimer’s disease have recently been the complementary roles of gasoline and motor oil
reported (Newport et  al., 2015). The results of in an engine; glucose provides both the gasoline
these clinical studies are still preliminary, but they and the motor oil, so the engine will work effi-
support the hypothesis that brain glucose deficit ciently, but ketones only provide the gasoline, so
contributes to impaired cognition associated with if forced to run excessively on ketones, the engine
aging and that this deficit can at least in part be soon burns out. Therefore, as long as sufficient
reversed or bypassed by either a ketogenic supple- glucose is available, anaplerosis will be maintained
ment containing MCTs, a ketone ester or a very and citric acid cycle function will continue even
high-fat ketogenic diet. during extreme ketosis.
During short-term starvation, gluconeogen-
ANAPLERO SIS esis is directly proportional to ketogenesis (Garber
During prolonged starvation, ketones can sup- et al., 1974). Some of that glucose is needed by the
ply upward of 70% of the energy requirement of liver (which cannot catabolize ketones; (Fukao
125

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 125

TABLE 15.4 CLINICAL STUDIES IN WHICH HORMONAL AND COGNITIVE


RESPONSES SUGGEST THAT ACUTE OR CHRONIC KETOGENIC TREATMENTS
MAINTAIN BRAIN FUNCTION BY COMPENSATING FOR HYPOGLYCEMIA

Treatment Reference

Acute studies
Controlled insulin- Treatment: 2 h insulin infusion ± 60 d fast Drenick et al.,
induced hypoglycemia Outcomes: ↓ effect of acute severe hypoglycemia (0.5 mM 1972
± fasting in obesity (n = 9) in one case), including ↓ mental confusion, anxiety, sweat-
ing, tachycardia, blood pressure if fasted for 60 d before
the insulin infusion
Controlled insulin- Treatment: 4 h i.v. β-HB infusion Amiel et al., 1991
induced hypoglycemia Dose: 30 μmol/min/kg body weight
in healthy adults (n = 6) Outcomes: ↓hormonal response to hypoglycemia
Controlled insulin- Treatment: 6 h i.v. β-HB infusion Veneman et al.,
induced hypoglycemia Dose: 20 µmol/min/kg body weight 1994
in healthy adults (n = 13) Outcomes: ↓ hormonal and cognitive symptoms of acute
hypoglycemia.
Controlled insulin- Treatment: single oral MCT Page et al., 2012
induced hypoglycemia Dose: 40 g in three stages (20, 10, 10 g)
in Type 1 diabetes (n = 11) Outcomes: ↓ cognitive symptoms of acute hypoglycemia.
Age-associated cognitive decline
Mild cognitive Treatment: 6 weeks high-fat ketogenic diet Krikorian et al.,
Impairment (n = 23) Outcomes:↑ secondary memory performance 2012
Mild-moderate Treatment: Single dose oral of MCT (95% octanoate) Reger et al., 2004
Alzheimer’s disease Dose: 40 g
(n = 20) Outcomes: ↑ cognitive score in APOE4(-) patients
Mild-moderate Treatment: 90 days oral MCT (95% octanoate) Henderson et al.,
Alzheimer’s disease Dose: 20 g/d 2009
(n = 77) Outcomes: ↑ cognitive score in APOE4(-) patients
Severe Alzheimer’s Treatment: 20 months oral MCT + coconut oil (4:3) Newport et al.,
disease (n = 1) Dose: 165 mL/day 2015
Outcomes: ↑ mood, affect, self-care, and cognitive and daily
activities

β-HB: beta-hydroxybutyrate

et al., 2004), but part of the resulting glucose pro- brain glucose uptake and/or utilization continue
duction is undoubtedly used to sustain anaplero- to deteriorate thereby further compromising both
sis, especially in the brain. During conditions in ATP production and anaplerosis, especially if
which glucose supply to the brain is more severely ketogenesis is stimulated. There is most likely to
limited, for example, inherited glucose transporter be a trade-off between supplying more ketones to
(Glut1) deficiency (Brunengraber and Roe, 2006; compensate for the glucose deficit and oversupply-
Mochel et al., 2005; Roe and Mochel, 2006), there ing them, thereby further depleting glucose and
is insufficient glucose to maintain anaplerosis or risking burning out the citric acid cycle.
energy production in the brain, so providing a
ketogenic supplement may be beneficial. However, B R A I N E N E R G Y S TAT U S ,
beyond a certain point, supplying more ketones N E U R A L P ROT E C T I O N,
is futile because they further stimulate the citric A N D M I TO C H O N D R I A L
acid cycle (Roy et al., 2015) without contributing FUNCTION
to anaplerosis so, in essence, they burn the cycle Regardless of whether glucose or ketones are being
out (Brunengraber and Roe, 2006). Whether the catabolized, mitochondria are required to pro-
situation is analogous in AD remains to be seen, duce the bulk of the ATP being generated. Glucose
but it is clear that as AD becomes more severe, can produce some ATP via aerobic glycolysis, a
126

126 section II: E merging Clinical Applications and Future Potential

process occurring outside mitochondria, whereas MCTs does not adversely affect serum glucose,
ketones are metabolized in an obligatory manner insulin, triglycerides, cholesterol, free fatty acids,
via oxidative phosphorylation within mitochon- body weight, or body-mass index (Courchesne-
dria. Although controversial, the enzymes of mito- Loyer et al., 2013).
chondrial oxidative phosphorylation may actually
continue to function normally, at least early in AD, E A R LY B R A I N
but ATP production declines because of lower gly- DEVELOPMENT AND
colysis to acetyl CoA. If so, there may be increased H U M A N B R A I N E VO L U T I O N
reliance on lactate as a fuel and even on gluconeo- Although outside the scope of this review, it has
genesis within the brain, with adverse consequences become clear over the past 3–4 decades that ketones
for amino acid and neurotransmitter production are much more important to the developing brain
including acetylcholine (Hoyer et al., 1988). than to the adult brain. Unlike in adults, ketones
Mitochondrial damage and increased pro- are an essential fuel and brain lipid substrate in
duction of reactive oxygen species are widely infants because there is insufficient glucose avail-
considered to coincide in AD and to exacerbate able for it alone to meet brain energy require-
the disease (Gibson et  al., 2000). Mitochondrial ments in human neonates (Bougneres et al., 1986;
dysfunction has been proposed to underlie beta- Robinson and Williamson, 1980; Settergren et al.,
amyloid accumulation and cognitive deteriora- 1976). The fatness of human babies makes this
tion in AD (Swerdlow et al., 2014; Swerdlow and essential role of ketones in neonates all the more
Khan, 2004; Yao et  al., 2009). Neural protection plausible given that long chain fatty acids stored in
by ketones seems to be related to improved mito- adipose tissue are the main substrate for ketogen-
chondrial function including reduced mitochon- esis. It is unlikely to be a coincidence that ketones
drial production of reactive oxygen species in are essential for human brain development and
response to glutamate (Maalouf et al., 2009). The that humans have fat babies; rather, these two con-
energy status of the brain appears to be increased ditions probably coevolved during human evolu-
in rats on a ketogenic diet based on ATP levels and tion with the former undoubtedly being facilitated
brain glucose relative to blood glucose (Cahill, by the latter (Cunnane and Crawford, 2014). The
2006; DeVivo et  al., 1978; Veech et  al., 2001), as potential utility of ketogenic supplements to the
well as indirect measurement of citric acid cycle aging brain is perhaps analogous to their impor-
activity (Roy et  al., 2015). These changes could tant role in the developing brain.
potentially be accompanied by mitochondrial bio-
genesis and improved respiratory function (Bough PERSPECTIVES
et  al., 2006). Others have shown that brain glu- We propose that AD is in part exacerbated by a
cose uptake and Glut expression increase in rats form of chronic gradual fuel starvation or exhaus-
on a ketogenic diet (Pifferi et al., 2011; Puchowicz tion due to brain glucose deficit and that early in the
et al., 2007; Roy et al., 2012), but this has not been disease this feature may be amenable to treatments
confirmed in humans; in fact; both PET and AVD that moderately but safely raise plasma ketones. The
studies suggest that, in human adults, brain glu- broadly similar neurological/cognitive benefit of
cose uptake decreases as brain ketone availability prolonged fasting, a very high-fat ketogenic diet con-
increases (Hasselbalch et  al.,  1995, Courchesne- taining no MCTs, or a regular diet to which MCTs or
Loyer et al. 2016). ketone esters are added (Table 15.4) suggests that the
improvement in cognition is related to a common
SAFETY OF MEDIUM CHAIN denominator of raised plasma ketones, which bypass
T R I G LY C E R I D E S chronically impaired glucose uptake and utilization
In doses up to 1 g/kg/d, MCTs have a robust safety by the AD brain. However, this is still speculative;
record in all species studies including humans to date, no study has yet provided a mechanism
(Bach and Babayan, 1982; Traul et al., 2000). Thus, by which MCT or ketones improve cognitive out-
they are not associated with increased risk of comes. We have deliberately not discussed the
cancer, obesity, or other diseases. However, they beta-amyloid hypothesis of AD, but beta-amyloid
can have secondary side effects involving gastro- accumulation could arise as a result of (Meier-Ruge
intestinal distress, an issue that can be partially et al., 1994) or be contributing to (Meier-Ruge and
mitigated by gradual dose titration. The MCTs are Bertoni-Freddari, 1997)  impaired glycolysis in the
saturated fats and, as such, have long been painted AD brain. It has also not been established whether
with the brush of increasing cardiovascular risk. certain MCTs (e.g., octanoate vs. decanoate) are
However, treatment for 30  days with 30 g/d of more effective for ketogenesis than others. Octanoic
127

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 127

TABLE 15.5 ONGOING REGISTERED CLINICAL TRIALS INVOLVING KETOGENIC


TREATMENTS IN MILD COGNITIVE IMPAIRMENT OR ALZHEIMER’S DISEASE

Conditions Treatment Reference #

Mild cognitive impairment Treatment: 6 months oral MCT beverage NCT02551419


Design: Double-blind, randomized, placebo-controlled trial
(n = 50/group)
Dose: 30 g/d
Outcome: Improvement in cognitive performance and
change in brain glucose and ketone metabolism
Mild cognitive impairment Treatment: 6 months oral MCT oil NCT01669200
Design: Double-blind, randomized controlled trial (n = 25/
group)
Dose: 40 g/d
Outcome: Improvement in cognitive performance
Mild Alzheimer’s disease Treatment: 14 days oral MCT NCT01702480
Design: Double-blind, randomized, placebo-controlled trial
(n = 8)
Dose: 10, 20, 30, and 40 g/d
Outcome: Change in CANTAB task performances and P300
EEG measurement
Mild-moderate Alzheimer’s Treatment: ≥ 6 months of AXONA (MCT powder) NCT01538212
disease Design: retrospective trial (n = 200)
Outcome: Change in living situation, psychiatric condition,
daily living, and cognitive performance

acid can be taken up by the brain (Ebert et al., 2003; be undertaken at the same time as providing a
Kuge et  al., 1995), so it may have direct effects on ketogenic supplement remains to be assessed.
brain function including but not limited to conver-
sion to ketones by astrocytes (Auestad et al., 1991).
AC K N OW L E D G M E N T S
Several clinical trials are underway that combine
Christine Brodeur-Dubreuil and Eric Lavallée
either a ketogenic supplement or diet with cognitive
provided excellent technical assistance. Financial
evaluation (Table 15.5) and should produce results
support was provided by a Canada Research
before the end of 2017, which will shed further light
Chair and University Research Chair (SCC), CFI,
on the potential utility of ketotherapeutics in neuro-
CIHR, NSERC, FRQS, FQRNT, Sojecci II, The
degenerative diseases such as AD.
Alzheimer Association (USA), and the Université
Attempts to correct or bypass impaired brain
de Sherbrooke. Some MCTs used in our studies
glucose metabolism in AD have also been made
were provided as gifts by ABITEC and Nutricia.
using several other approaches including supple-
ments of glucose (Korol and Gold, 1998), a cocktail
of glucose, malate, and antioxidant (Blass, 2008) or REFERENCES
intranasal insulin (Benedict et  al., 2011; Craft, Auestad, N., Korsak, R.A., Morrow, J.W., and Edmond,
2012). The details of those studies are beyond the J. (1991). Fatty acid oxidation and ketogenesis by
scope of this review but they are important because astrocytes in primary culture. J Neurochem 56,
they show that for 20  years now, the metabolic 1376–1386.
defect in the AD brain has been viewed by many Avogaro, A., Nosadini, R., Bier, D.M., Cobelli,
different groups as a logical target for therapeutic C., Toffolo, G., Doria, A., Valerio, A., and
Christopherson, H. (1990). Ketone body kinet-
strategies in AD (Cunnane et  al., 2015; Cunnane
ics in vivo using simultaneous administration of
et al., 2011; Gibson et al., 2000; Henderson, 2008; acetoacetate and 3-hydroxybutyrate labelled with
Hertz et al., 2015; Kapogiannis and Mattson, 2011; stable isotopes. Acta Diabetol Lat 27, 41–51.
Yao et  al., 2011). Whether trying to correct the Bach, A.C., and Babayan, V.K. (1982). Medium-
problem of impaired glucose uptake-metabolism chain triglycerides: an update. Am J Clin Nutr 36,
and deteriorating mitochondrial function should 950–962.
128

128 section II: E merging Clinical Applications and Future Potential

Baker, L.D., Cross, D.J., Minoshima, S., Belongia, D., selective neuronal loss as an important cause of
Watson, G.S., and Craft, S. (2011). Insulin resis- dementia? Lancet 1, 11–14.
tance and Alzheimer-like reductions in regional Brown, A.D., McMorris, C.A., Longman, R.S., Leigh,
cerebral glucose metabolism for cognitively nor- R., Hill, M.D., Friedenreich, C.M., and Poulin,
mal adults with prediabetes or early type 2 diabe- M.J. (2010). Effects of cardiorespiratory fitness and
tes. Arch Neurol 68, 51–57. cerebral blood flow on cognitive outcomes in older
Balasse, E.O. (1979). Kinetics of ketone body metabo- women. Neurobiol Aging 31, 2047–2057.
lism in fasting humans. Metabolism 28, 41–50. Brunengraber, H., and Roe, C.R. (2006). Anaplerotic
Balasse, E.O. (1986). Importance of ketone bodies in molecules: current and future. J Inherit Metab Dis
endogenous fat transport. Clin Nutr 5, 73–80. 29, 327–331.
Balasse, E.O., Fery, F., and Neef, M.A. (1978). Changes Bu, G. (2009). Apolipoprotein E and its receptors in
induced by exercise in rates of turnover and oxi- Alzheimer’s disease:  pathways, pathogenesis and
dation of ketone bodies in fasting man. J Appl therapy. Nat Rev Neurosci 10, 333–344.
Physiol Respir Environ Exerc Physiol 44, 5–11. Cahill, G.F., Jr. (2006). Fuel metabolism in starvation.
Balasse, E.O., and Fery, F. (1989). Ketone body produc- Annu Rev Nutr 26, 1–22.
tion and disposal: effects of fasting, diabetes, and Castellano, C.A., Baillargeon, J.P., Nugent, S.,
exercise. Diabetes Metab Rev 5, 247–270. Tremblay, S., Fortier, M., Imbeault, H., Duval, J.,
Benedict, C., Frey, W.H., 2nd, Schioth, H.B., Schultes, and Cunnane, S.C. (2015a). Regional brain glucose
B., Born, J., and Hallschmid, M. (2011). Intranasal hypometabolism in young women with polycys-
insulin as a therapeutic option in the treatment tic ovary syndrome: possible link to mild insulin
of cognitive impairments. Exp Gerontol 46, resistance. PLOS One, 10(12)
112–115. Castellano, C.A., Nugent, S., Paquet, N., Tremblay, S.,
Benson, D.F., Kuhl, D.E., Hawkins, R.A., Phelps, M.E., Bocti, C., Lacombe, G., Imbeault, H., Turcotte, E.,
Cummings, J.L., and Tsai, S.Y. (1983). The fluoro- Fulop, T., and Cunnane, S.C. (2015b). Lower brain
deoxyglucose 18F scan in Alzheimer’s disease and 18F-fluorodeoxyglucose uptake but normal 11C-
multi-infarct dementia. Arch Neurol 40, 711–714. acetoacetate metabolism in mild Alzheimer’s dis-
Bickerton, A.S., Roberts, R., Fielding, B.A., Tornqvist, ease dementia. J Alzheimers Dis 43, 1343–1353.
H., Blaak, E.E., Wagenmakers, A.J., Gilbert, M., Clarke, K., Tchabanenko, K., Pawlosky, R., Carter,
Humphreys, S.M., Karpe, F., and Frayn, K.N. E., Todd King, M., Musa-Veloso, K., Ho, M.,
(2008). Adipose tissue fatty acid metabolism in Roberts, A., Robertson, J., Vanitallie, T.B., et  al.
insulin-resistant men. Diabetologia 51, 1466–1474. (2012). Kinetics, safety and tolerability of (R)-3-
Blass, J.P. (2008). A new approach to treating hydroxybutyl (R)-3-hydroxybutyrate in healthy
Alzheimer’s disease. Ann NY Acad Sci 1147, adult subjects. Regul Toxicol Pharmacol 63,
122–128. 401–408.
Blennow, K., de Leon, M.J., and Zetterberg, H. (2006). Courchesne-Loyer, A., Fortier, M., Tremblay-
Alzheimer’s disease. Lancet 368, 387–403. Mercier, J., Chouinard-Watkins, R., Roy, M.,
Blomqvist, G., Alvarsson, M., Grill, V., Von Heijne, G., Nugent, S., Castellano, C.A., and Cunnane, S.C.
Ingvar, M., Thorell, J.O., Stone-Elander, S., Widen, (2013). Stimulation of mild, sustained keto-
L., and Ekberg, K. (2002). Effect of acute hyperke- nemia by medium-chain triacylglycerols in
tonemia on the cerebral uptake of ketone bodies healthy humans:  estimated potential contribu-
in nondiabetic subjects and IDDM patients. Am J tion to brain energy metabolism. Nutrition 29,
Physiol Endocrinol Metab 283, E20–28. 635–640.
Blomqvist, G., Thorell, J.O., Ingvar, M., Grill, V., Courchesne-Loyer, A., Croteau, E., Castellano, C.A.,
Widen, L., and Stone-Elander, S. (1995). Use of St-Pierre, V., Hennebelle, M., and Cunnane, S.C.
R-beta-[1-11C]hydroxybutyrate in PET studies (2016). Inverse relationship between brain glucose
of regional cerebral uptake of ketone bodies in and ketone metabolism in adults during short-
humans. Am J Physiol 269, E948–959. term moderate dietary ketosis: a dual tracer quan-
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., titative PET study. J Cereb Blood Flow Metab.
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Craft, S. (2009). The role of metabolic disorders in
Y., Geiger, J.D., and Dingledine, R.J. (2006). Alzheimer disease and vascular dementia:  two
Mitochondrial biogenesis in the anticonvulsant roads converged. Arch Neurol 66, 300–305.
mechanism of the ketogenic diet. Ann Neurol 60, Craft, S. (2012). Alzheimer disease: Insulin resistance
223–235. and AD—extending the translational path. Nat
Bougneres, P.F., Lemmel, C., Ferre, P., and Bier, D.M. Rev Neurol 8, 360–362.
(1986). Ketone body transport in the human neo- Cunnane, S., Nugent, S., Roy, M., Courchesne-Loyer,
nate and infant. J Clin Invest 77, 42–48. A., Croteau, E., Tremblay, S., Castellano, A., Pifferi,
Bowen, D.M., White, P., Spillane, J.A., Goodhardt, F., Bocti, C., Paquet, N., et  al. (2011). Brain fuel
M.J., Curzon, G., Iwangoff, P., Meier-Ruge, W., metabolism, aging, and Alzheimer’s disease.
and Davison, A.N. (1979). Accelerated ageing or Nutrition 27, 3–20.
129

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 129

Cunnane, S.C., and Crawford, M.A. (2014). Energetic Freund, G., and Weinsier, R.L. (1966). Standardized
and nutritional constraints on infant brain devel- ketosis in man following medium chain triglycer-
opment:  implications for brain expansion during ide ingestion. Metabolism 15, 980–991.
human evolution. J Hum Evol 77, 88–98. Fukao, T., Lopaschuk, G.D., and Mitchell, G.A. (2004).
Cunnane, S.C., Courchesne-Loyer, A., St-Pierre, V., Pathways and control of ketone body metabolism: on
Vandenberghe, C., Pierrotti, T., Fortier, M., the fringe of lipid biochemistry. Prostaglandins
Croteau, E., and Castellano, A. (2015). Can ketones Leukot Essent Fatty Acids 70, 243–251.
compensate for deteriorating brain glucose uptake Garber, A.J., Menzel, P.H., Boden, G., and Owen, O.E.
during aging? Implications for the risk and treat- (1974). Hepatic ketogenesis and gluconeogenesis
ment of Alzheimer’s disease. Ann NY Acad Sci in humans. J Clin Invest 54, 981–989.
1367, 12–20. Gibson, R.E., Burns, H.D., Hamill, T.G., Eng, W.S.,
Dalsgaard, M.K., Quistorff, B., Danielsen, E.R., Francis, B.E., and Ryan, C. (2000). Non-invasive
Selmer, C., Vogelsang, T., and Secher, N.H. (2004). radiotracer imaging as a tool for drug develop-
A reduced cerebral metabolic ratio in exercise ment. Curr Pharm Des 6, 973–989.
reflects metabolism and not accumulation of Gottstein, U., Muller, W., Berghoff, W., Gartner, H.,
lactate within the human brain. J Physiol 554, and Held, K. (1971). [Utilization of non-esterified
571–578. fatty acids and ketone nbodies in human brain].
Dastur, D.K. (1985). Cerebral blood flow and metabo- Klin Wochenschr 49, 406–411.
lism in normal human aging, pathological aging, Hall, S.E., Wastney, M.E., Bolton, T.M., Braaten, J.T.,
and senile dementia. J Cereb Blood Flow Metab and Berman, M. (1984). Ketone body kinetics in
5, 1–9. humans: the effects of insulin-dependent diabetes,
DeVivo, D.C., Leckie, M.P., Ferrendelli, J.S., and obesity, and starvation. J Lipid Res 25, 1184–1194.
McDougal, D.B., Jr. (1978). Chronic ketosis and Hasselbalch, S.G., Knudsen, G.M., Jakobsen, J.,
cerebral metabolism. Ann Neurol 3, 331–337. Hageman, L.P., Holm, S., and Paulson, O.B. (1995).
Douaud, G., Refsum, H., de Jager, C.A., Jacoby, R., Blood-brain barrier permeability of glucose and
Nichols, T.E., Smith, S.M., and Smith, A.D. (2013). ketone bodies during short-term starvation in
Preventing Alzheimer’s disease-related gray mat- humans. Am J Physiol 268, E1161–1166.
ter atrophy by B-vitamin treatment. Proc Natl Hasselbalch, S.G., Madsen, P.L., Hageman, L.P., Olsen,
Acad Sci USA 110, 9523–9528. K.S., Justesen, N., Holm, S., and Paulson, O.B.
Drenick, E.J., Alvarez, L.C., Tamasi, G.C., and (1996). Changes in cerebral blood flow and carbo-
Brickman, A.S. (1972). Resistance to symptom- hydrate metabolism during acute hyperketonemia.
atic insulin reactions after fasting. J Clin Invest 51, Am J Physiol 270, E746–751.
2757–2762. Henderson, S.T. (2008). Ketone bodies as a therapeu-
Ebert, D., Haller, R.G., and Walton, M.E. (2003). tic for Alzheimer’s disease. Neurotherapeutics 5,
Energy contribution of octanoate to intact rat 470–480.
brain metabolism measured by 13C nuclear Henderson, S.T., Vogel, J.L., Barr, L.J., Garvin, F.,
magnetic resonance spectroscopy. J Neurosci 23, Jones, J.J., and Costantini, L.C. (2009). Study of
5928–5935. the ketogenic agent AC-1202 in mild to moderate
Fery, F., and Balasse, E.O. (1983). Ketone body turn- Alzheimer’s disease:  a randomized, double-blind,
over during and after exercise in overnight-fasted placebo-controlled, multicenter trial. Nutr Metab
and starved humans. Am J Physiol 245, E318–325. (Lond) 6, 31.
Fery, F., and Balasse, E.O. (1986). Response of ketone Hertz, L., Chen, Y., and Waagepetersen, H.S. (2015).
body metabolism to exercise during transition Effects of ketone bodies in Alzheimer’s disease in rela-
from postabsorptive to fasted state. Am J Physiol tion to neural hypometabolism, beta-amyloid toxic-
250, E495–501. ity, and astrocyte function. J Neurochem 134, 7–20.
Flatt, J.P. (1972). On the maximal possible rate of keto- Hoyer, S. (1992). Oxidative energy metabolism in
genesis. Diabetes 21, 50–53. Alzheimer brain. Studies in early-onset and
Fox, N.C., Kennedy, A.M., Harvey, R.J., Lantos, P.L., late-onset cases. Mol Chem Neuropathol 16,
Roques, P.K., Collinge, J., Hardy, J., Hutton, M., 207–224.
Stevens, J.M., Warrington, E.K., et  al. (1997). Hoyer, S., Oesterreich, K., and Wagner, O. (1988).
Clinicopathological features of familial Alzheimer’s Glucose metabolism as the site of the primary
disease associated with the M139V mutation in abnormality in early-onset dementia of Alzheimer
the presenilin 1 gene: pedigree but not mutation; type? J Neurol 235, 143–148.
specific age at onset provides evidence for a further Ide, K., Schmalbruch, I.K., Quistorff, B., Horn, A.,
genetic factor. Brain 120 (Pt 3), 491–501. and Secher, N.H. (2000). Lactate, glucose and O2
Freemantle, E., Vandal, M., Tremblay Mercier, J., uptake in human brain during recovery from max-
Plourde, M., Poirier, J., and Cunnane, S.C. (2009). imal exercise. J Physiol 522 Pt 1, 159–164.
Metabolic response to a ketogenic breakfast in the Iwangoff, P., Armbruster, R., Enz, A., and Meier-
healthy elderly. J Nutr Health Aging 13, 293–298. Ruge, W. (1980). Glycolytic enzymes from human
130

130 section II: E merging Clinical Applications and Future Potential

autoptic brain cortex: normal aged and demented Mamelak, M. (2012). Sporadic Alzheimer’s disease: the
cases. Mech Ageing Dev 14, 203–209. starving brain. J Alzheimers Dis 31, 459–474.
Jack, C.R., Jr., Lowe, V.J., Weigand, S.D., Wiste, H.J., Matsuzaki, T., Sasaki, K., Tanizaki, Y., Hata, J., Fujimi,
Senjem, M.L., Knopman, D.S., Shiung, M.M., K., Matsui, Y., Sekita, A., Suzuki, S.O., Kanba,
Gunter, J.L., Boeve, B.F., Kemp, B.J., et al. (2009). S., Kiyohara, Y., et  al. (2010). Insulin resistance
Serial PIB and MRI in normal, mild cognitive is associated with the pathology of Alzheimer
impairment and Alzheimer’s disease: implications disease:  the Hisayama study. Neurology 75,
for sequence of pathological events in Alzheimer’s 764–770.
disease. Brain 132, 1355–1365. Meier-Ruge, W., Bertoni-Freddari, C., and Iwangoff,
Jerneren, F., Elshorbagy, A.K., Oulhaj, A., Smith, S.M., P. (1994). Changes in brain glucose metabolism as
Refsum, H., and Smith, A.D. (2015). Brain atrophy a key to the pathogenesis of Alzheimer’s disease.
in cognitively impaired elderly:  the importance Gerontology 40, 246–252.
of long-chain omega-3 fatty acids and B vitamin Meier-Ruge, W.A., and Bertoni-Freddari, C. (1997).
status in a randomized controlled trial. Am J Clin Pathogenesis of decreased glucose turnover
Nutr 102, 215–221. and oxidative phosphorylation in ischemic and
Kalpouzos, G., Chetelat, G., Baron, J.C., Landeau, B., trauma-induced dementia of the Alzheimer type.
Mevel, K., Godeau, C., Barre, L., Constans, J.M., Ann NY Acad Sci 826, 229–241.
Viader, F., Eustache, F., et al. (2009). Voxel-based Mitchell, G.A., Kassovska-Bratinova, S., Boukaftane,
mapping of brain gray matter volume and glucose Y., Robert, M.F., Wang, S.P., Ashmarina, L.,
metabolism profiles in normal aging. Neurobiol Lambert, M., Lapierre, P., and Potier, E. (1995).
Aging 30, 112–124. Medical aspects of ketone body metabolism. Clin
Kapogiannis, D., and Mattson, M.P. (2011). Disrupted Invest Med 18, 193–216.
energy metabolism and neuronal circuit dysfunc- Mochel, F., DeLonlay, P., Touati, G., Brunengraber, H.,
tion in cognitive impairment and Alzheimer’s dis- Kinman, R.P., Rabier, D., Roe, C.R., and Saudubray,
ease. Lancet Neurol 10, 187–198. J.M. (2005). Pyruvate carboxylase deficiency: clin-
Kennedy, A.M., Frackowiak, R.S., Newman, S.K., ical and biochemical response to anaplerotic diet
Bloomfield, P.M., Seaward, J., Roques, P., therapy. Mol Genet Metab 84, 305–312.
Lewington, G., Cunningham, V.J., and Rossor, Mosconi, L., Tsui, W.H., De Santi, S., Li, J., Rusinek,
M.N. (1995). Deficits in cerebral glucose metabo- H., Convit, A., Li, Y., Boppana, M., and de Leon,
lism demonstrated by positron emission tomogra- M.J. (2005). Reduced hippocampal metabolism in
phy in individuals at risk of familial Alzheimer’s MCI and AD: automated FDG-PET image analy-
disease. Neurosci Lett 186, 17–20. sis. Neurology 64, 1860–1867.
Korol, D.L., and Gold, P.E. (1998). Glucose, memory, Newport, M.T., VanItallie, T.B., Kashiwaya, Y., King,
and aging. Am J Clin Nutr 67, 764S–771S. M.T., and Veech, R.L. (2015). A new way to pro-
Krikorian, R., Shidler, M.D., Dangelo, K., Couch, S.C., duce hyperketonemia: use of ketone ester in a case of
Benoit, S.C., and Clegg, D.J. (2012). Dietary keto- Alzheimer’s disease. Alzheimers Dement 11, 99–103.
sis enhances memory in mild cognitive impair- Ngandu, T., Lehtisalo, J., Solomon, A., Levalahti,
ment. Neurobiol Aging 33, 425 e419–427. E., Ahtiluoto, S., Antikainen, R., Backman, L.,
Kuge, Y., Yajima, K., Kawashima, H., Yamazaki, H., Hanninen, T., Jula, A., Laatikainen, T., et al. (2015).
Hashimoto, N., and Miyake, Y. (1995). Brain A 2  year multidomain intervention of diet, exer-
uptake and metabolism of [1-11C]octanoate in cise, cognitive training, and vascular risk monitor-
rats: pharmacokinetic basis for its application as a ing versus control to prevent cognitive decline in
radiopharmaceutical for studying brain fatty acid at-risk elderly people (FINGER):  a randomised
metabolism. Ann Nucl Med 9, 137–142. controlled trial. Lancet 385, 2255–2263.
London, E.D., Margolin, R.A., Duara, R., Holloway, Nordberg, A. (2006). Mechanisms behind the neuro-
H.W., Robertson-Tchabo, E.A., Cutler, N.R., and protective actions of cholinesterase inhibitors in
Rapoport, S.I. (1986). Effects of fasting on ketone Alzheimer disease. Alzheimer Dis Assoc Disord
body concentrations in healthy men of different 20, S12–18.
ages. J Gerontol 41, 599–604. Nugent, S., Castellano, C.A., Goffaux, P., Whittingstall,
Lying-Tunell, U., Lindblad, B.S., Malmlund, H.O., and K., Lepage, M., Paquet, N., Bocti, C., Fulop, T., and
Persson, B. (1981). Cerebral blood flow and met- Cunnane, S.C. (2014a). Glucose hypometabolism
abolic rate of oxygen, glucose, lactate, pyruvate, is highly localized but lower cortical thickness and
ketone bodies and amino acids. Acta Neurol Scand brain atrophy are widespread in cognitively nor-
63, 337–350. mal older adults. Am J Physiol Endocrinol Metab.
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The Nugent, S., Tremblay, S., Chen, K.W., Ayutyanont, N.,
neuroprotective properties of calorie restriction, Roontiva, A., Castellano, C.A., Fortier, M., Roy,
the ketogenic diet, and ketone bodies. Brain Res M., Courchesne-Loyer, A., Bocti, C., et al. (2014b).
Rev 59, 293–315. Brain glucose and acetoacetate metabolism:  a
131

Chapter 15: Glucose and Ketone Metabolism in the Aging Brain 131

comparison of young and older adults. Neurobiol beta-hydroxybutyrate supplementation in two


Aging 35, 1386–1395. patients with hyperinsulinemic hypoglyce-
Nugent, S., Castellano, C.A., Bocti, C., Dionne, I., mia: monitoring of beta-hydroxybutyrate levels in
Fulop, T., and Cunnane, S.C. (2015). Relationship blood and cerebrospinal fluid, and in the brain by
of metabolic and endocrine parameters to brain in vivo magnetic resonance spectroscopy. Pediatr
glucose metabolism in older adults: do cognitively- Res 52, 301–306.
normal older adults have a particular metabolic Puchowicz, M.A., Xu, K., Sun, X., Ivy, A., Emancipator,
phenotype? Biogerontology 17, 241–255. D., and LaManna, J.C. (2007). Diet-induced keto-
Ogawa, M., Fukuyama, H., Ouchi, Y., Yamauchi, H., sis increases capillary density without altered
and Kimura, J. (1996). Altered energy metabolism blood flow in rat brain. Am J Physiol Endocrinol
in Alzheimer’s disease. J Neurol Sci 139, 78–82. Metab 292, E1607–1615.
Okonkwo, O.C., Schultz, S.A., Oh, J.M., Larson, J., Quinn, J.F., Raman, R., Thomas, R.G., Yurko-Mauro,
Edwards, D., Cook, D., Koscik, R., Gallagher, K., Nelson, E.B., Van Dyck, C., Galvin, J.E.,
C.L., Dowling, N.M., Carlsson, C.M., et al. (2014). Emond, J., Jack, C.R., Jr., Weiner, M., et al. (2010).
Physical activity attenuates age-related biomarker Docosahexaenoic acid supplementation and cog-
alterations in preclinical AD. Neurology 83, nitive decline in Alzheimer disease: a randomized
1753–1760. trial. JAMA 304, 1903–1911.
Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan, Rafii, M.S., and Aisen, P.S. (2009). Recent develop-
J.M., Herrera, M.G., and Cahill, G.F., Jr. (1967). ments in Alzheimer’s disease therapeutics. BMC
Brain metabolism during fasting. J Clin Invest 46, Med 7, 7.
1589–1595. Reger, M.A., Henderson, S.T., Hale, C., Cholerton, B.,
Owen, O.E., and Reichard, G.A., Jr. (1971). Human Baker, L.D., Watson, G.S., Hyde, K., Chapman, D.,
forearm metabolism during progressive starva- and Craft, S. (2004). Effects of beta-hydroxybutyrate
tion. J Clin Invest 50, 1536–1545. on cognition in memory-impaired adults. Neurobiol
Owen, O.E., Kalhan, S.C., and Hanson, R.W. (2002). Aging 25, 311–314.
The key role of anaplerosis and cataplerosis Reichard, G.A., Jr., Owen, O.E., Haff, A.C., Paul, P., and
for citric acid cycle function. J Biol Chem 277, Bortz, W.M. (1974). Ketone-body production and
30409–30412. oxidation in fasting obese humans. J Clin Invest
Page, K.A., Williamson, A., Yu, N., McNay, E.C., 53, 508–515.
Dzuira, J., McCrimmon, R.J., and Sherwin, R.S. Reiman, E.M., Chen, K., Alexander, G.E., Caselli,
(2009). Medium-chain fatty acids improve cogni- R.J., Bandy, D., Osborne, D., Saunders, A.M., and
tive function in intensively treated type 1 diabetic Hardy, J. (2004). Functional brain abnormali-
patients and support in vitro synaptic transmis- ties in young adults at genetic risk for late-onset
sion during acute hypoglycemia. Diabetes 58, Alzheimer’s dementia. Proc Natl Acad Sci USA
1237–1244. 101, 284–289.
Pan, J.W., Telang, F.W., Lee, J.H., de Graaf, R.A., Robinson, A.M., and Williamson, D.H. (1980).
Rothman, D.L., Stein, D.T., and Hetherington, H.P. Physiological roles of ketone bodies as substrates
(2001). Measurement of beta-hydroxybutyrate and signals in mammalian tissues. Physiol Rev 60,
in acute hyperketonemia in human brain. J 143–187.
Neurochem 79, 539–544. Roe, C.R., and Mochel, F. (2006). Anaplerotic diet
Perry, E.K., Perry, R.H., Tomlinson, B.E., Blessed, G., therapy in inherited metabolic disease: therapeu-
and Gibson, P.H. (1980). Coenzyme A-acetylating tic potential. J Inherit Metab Dis 29, 332–340.
enzymes in Alzheimer’s disease:  possible cholin- Ronnemaa, E., Zethelius, B., Sundelof, J., Sundstrom,
ergic “compartment” of pyruvate dehydrogenase. J., Degerman-Gunnarsson, M., Berne, C., Lannfelt,
Neurosci Lett 18, 105–110. L., and Kilander, L. (2008). Impaired insulin
Pi-Sunyer, F.X., Hashim, S.A., and Van Itallie, T.B. secretion increases the risk of Alzheimer disease.
(1969). Insulin and ketone responses to ingestion Neurology 71, 1065–1071.
of medium and long-chain triglycerides in man. Roy, M., Nugent, S., Tremblay-Mercier, J., Tremblay,
Diabetes 18, 96–100. S., Courchesne-Loyer, A., Beaudoin, J.F.,
Pifferi, F., Tremblay, S., Croteau, E., Fortier, M., Tremblay, L., Descoteaux, M., Lecomte, R., and
Tremblay-Mercier, J., Lecomte, R., and Cunnane, Cunnane, S.C. (2012). The ketogenic diet increases
S.C. (2011). Mild experimental ketosis increases brain glucose and ketone uptake in aged rats:  a
brain uptake of (11)C-acetoacetate and (18)F- dual tracer PET and volumetric MRI study. Brain
fluorodeoxyglucose:  a dual-tracer PET imaging Res 1488, 14–23.
study in rats. Nutr Neurosci 14, 51–58. Roy, M., Beauvieux, M.C., Naulin, J., El Hamrani,
Plecko, B., Stoeckler-Ipsiroglu, S., Schober, E., Harrer, D., Gallis, J.L., Cunnane, S.C., and Bouzier-Sore,
G., Mlynarik, V., Gruber, S., Moser, E., Moeslinger, A.K. (2015). Rapid adaptation of rat brain and
D., Silgoner, H., and Ipsiroglu, O. (2002). Oral liver metabolism to a ketogenic diet: an integrated
132

132 section II: E merging Clinical Applications and Future Potential

study using (1)H- and (13)C-NMR spectroscopy. J Swerdlow, R.H., Burns, J.M., and Khan, S.M. (2014).
Cereb Blood Flow Metab 35, 1154–1162. The Alzheimer’s disease mitochondrial cascade
Scholl, M., Almkvist, O., Bogdanovic, N., Wall, A., hypothesis:  progress and perspectives. Biochim
Langstrom, B., Viitanen, M., and Nordberg, A. Biophys Acta 1842, 1219–1231.
(2011). Time course of glucose metabolism in rela- Swerdlow, R.H., and Khan, S.M. (2004). A “mitochon-
tion to cognitive performance and postmortem drial cascade hypothesis” for sporadic Alzheimer’s
neuropathology in Met146Val PSEN1 mutation disease. Med Hypotheses 63, 8–20.
carriers. J Alzheimers Dis 24, 495–506. Traul, K.A., Driedger, A., Ingle, D.L., and Nakhasi,
Schrijvers, E.M., Witteman, J.C., Sijbrands, E.J., D. (2000). Review of the toxicologic properties of
Hofman, A., Koudstaal, P.J., and Breteler, medium-chain triglycerides. Food Chem Toxicol
M.M. (2010). Insulin metabolism and the risk 38, 79–98.
of Alzheimer disease:  the Rotterdam Study. Tremblay, S., Ouellet, R., Rodrigue, S., Langlois, R.,
Neurology 75, 1982–1987. Benard, F., and Cunnane, S.C. (2007). Automated
Seaton, T.B., Welle, S.L., Warenko, M.K., and Campbell, synthesis of 11C-acetoacetic acid, a key alter-
R.G. (1986). Thermic effect of medium-chain and nate brain fuel to glucose. Appl Radiat Isot 65,
long-chain triglycerides in man. Am J Clin Nutr 934–940.
44, 630–634. Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A.,
Settergren, G., Lindblad, B.S., and Persson, B. and Cahill, G.F., Jr. (2001). Ketone bodies, poten-
(1976). Cerebral blood flow and exchange of tial therapeutic uses. IUBMB Life 51, 241–247.
oxygen, glucose, ketone bodies, lactate, pyruvate Wang, T., Huang, Q., Reiman, E.M., Chen, K., Li, X.,
and amino acids in infants. Acta Paediatr Scand Li, G., Lin, Z., Li, C., and Xiao, S. (2013). Effects of
65, 343–353. memantine on clinical ratings, fluorodeoxyglucose
Simpson, I.A., Carruthers, A., and Vannucci, S.J. positron emission tomography measurements, and
(2007). Supply and demand in cerebral energy cerebrospinal fluid assays in patients with moder-
metabolism:  the role of nutrient transporters. J ate to severe Alzheimer dementia: a 24-week, ran-
Cereb Blood Flow Metab 27, 1766–1791. domized, clinical trial. J Clin Psychopharmacol 33,
Sokoloff, L., Reivich, M., Kennedy, C., Des Rosiers, 636–642.
M.H., Patlak, C.S., Pettigrew, K.D., Sakurada, O., Yao, J., Irwin, R.W., Zhao, L., Nilsen, J., Hamilton, R.T.,
and Shinohara, M. (1977). The [14C]deoxyglu- and Brinton, R.D. (2009). Mitochondrial bioen-
cose method for the measurement of local cerebral ergetic deficit precedes Alzheimer’s pathology in
glucose utilization: theory, procedure, and normal female mouse model of Alzheimer’s disease. Proc
values in the conscious and anesthetized albino Natl Acad Sci USA 106, 14670–14675.
rat. J Neurochem 28, 897–916. Yao, J., Rettberg, J.R., Klosinski, L.P., Cadenas, E., and
Sorbi, S., Bird, E.D., and Blass, J.P. (1983). Decreased Brinton, R.D. (2011). Shift in brain metabolism
pyruvate dehydrogenase complex activity in in late onset Alzheimer’s disease: implications for
Huntington and Alzheimer brain. Ann Neurol biomarkers and therapeutic interventions. Mol
13, 72–78. Aspects Med 32, 247–257.
133

16
Ketogenic Diet and Ketones for the Treatment
of Traumatic Brain and Spinal Cord Injury
F E M K E S T R E I J G E R , P H D , WA R D T. P L U N E T, P H D ,
A N D WO L F R A M T E T Z L A F F, M D , D R . M E D , P H D

INTRODUCTION the wide range of secondary complications listed


Every year, over 1,700 Canadians suffer an acute above (SCIRE, 2105). Rehabilitation interventions
traumatic spinal cord injury (SCI) (Rick Hansen have led to significant improvements in the neu-
Registry, 2006)  (Rick Hansen Registry:  http:// rological outcome after chronic motor incomplete
www.rickhanseninstitute.org/work/our- projects- SCI, and more recently promising strides were
initiatives/rhscir), and in the United States the made in a few individuals with functionally com-
number is around 12,500 (National Spinal Cord plete SCI (Harkema et al., 2012a; Harkema et al.,
Injury Center). Spinal cord injury has a devastat- 2012b). However, there is still a large unmet clini-
ing impact on the quality of life that goes beyond cal need for treatments to protect the injured spi-
the obvious paralysis and includes pain, spasticity, nal cord from secondary damage in the wake of
sexual dysfunction, respiratory problems, bowel a primary injury. While little can be done for the
and bladder disorders, skin ulcers, autonomic dys- immediate necrosis of the tissue due to mechani-
regulation, cardiovascular disease and metabolic cal destruction by the primary impact, the injury
disease, leading to a greatly reduced life expec- triggers a cascade of secondary damage due to vas-
tancy (SCIRE, 2105). The economic burden of cular disruption, hemorrhage, edema, and isch-
traumatic SCI in Canada is enormous and ranges emia, leading to energy depletion and ion pump
from $1.47 million CAD for a person with incom- failures, depolarization, excitotoxicity, calcium
plete paraplegia and close to $3 million CAD for a overload, activation of proteases, free oxygen radi-
complete tetraplegic individual over the course of cal formation, cell membrane compromise, lipid
life (when injury is sustained at age 35) (Krueger peroxidation and protein nitrosylation, inflamma-
et al., 2013). tion, and cell death by various mechanisms. Such
The acute management of SCI is mainly focused secondary damage results in oligodendrocyte
on surgical stabilization of the spinal column and death with demyelination, neuronal losses, and
surgical decompression of the spinal cord to relieve permanent functional impairments (for reviews
extrinsic pressure (Fehlings et  al., 2012; Furlan see, Oyinbo, 2011).
et al., 2011), although the benefits from early sur- The quest for treatments to mitigate second-
gical intervention are challenging to demonstrate ary injury after neurotrauma has led to the pre-
(Furlan et al., 2011). There are currently no neu- clinical discovery of numerous “neuroprotective”
roprotective or neurorestorative therapies avail- drug candidates as well as metabolic and dietary
able for SCI patients, and the only drug approved treatments. The latter include a number of health
by the US Food and Drug Administration (FDA), supplements and dietary regimes that are neu-
methylprednisolone, showed only marginal neu- roprotective after acute SCI in rodents including
roprotective effects when administered within 8 acetyl-L-carnitine (Patel et  al., 2012; Patel et  al.,
hours after injury (Bracken et al., 1998). Due to its 2010; Patel et  al., 2014), creatine (Rabchevsky
limited efficacy and significant side effects, methyl- et  al., 2003), melatonin (Yang et  al., 2015), natu-
prednisolone is no longer recommended (Evaniew ral polyphenols from green tea, olive oil, resvera-
et  al., 2015). The reality is that the only effective trol, turmeric (for review, see Khalatbary, 2014),
treatments for people with SCI to date are reha- fish oils (King et  al., 2006; Michael-Titus and
bilitation programs together with medical care for Priestley, 2014), and inosine (Kim et al., 2013) as
134

134 section II: E merging Clinical Applications and Future Potential

well as multinutrient combinations of DHA (doc- fat, adequate in protein, and very low in carbo-
osahexaenoic acid), EPA (eicosapentaenoic acid), hydrates, and induce a fasting-like state. KDs
choline, phospholipids, and various vitamins mar- or ketones have been shown to be beneficial in
keted as Fortasyn (Pallier et  al., 2015). Many of models of Alzheimer’s disease (Hertz et al., 2015;
these approaches with “naturally occurring” nutri- Imamura et al., 2006; Kashiwaya et al., 2000; Paoli
ents and over-the-counter health supplements et al., 2014), Parkinson’s disease (Cheng et al.,
appear to have anti-inflammatory, anti-apoptotic, 2009; Kashiwaya et al., 2000; Tieu et al., 2003),
and antioxidant properties by mechanisms that Huntington’s disease (Lim et al., 2011), amyo-
are incompletely understood, while some also trophic lateral sclerosis (Zhao et al., 2012; Zhao et
enhance synaptic plasticity in the injured spi- al., 2006), stroke (Austin et al., 2012; Gibson et al.,
nal cord of rodents (Kim et  al., 2013). However, 2012; Puchowicz et al., 2008; Rahman et al., 2014;
none of these treatments have yet to become clini- Suzuki et al., 2002), multiple sclerosis (Kim do et
cal standard through validation in clinical trials. al., 2012), and TBI (Appelberg et al., 2009; Davis
As a matter of fact, the nutritional guidelines for et al., 2008a; Hu et al., 2009a; Prins et al., 2005).
acute SCI are merely based on expert opinions and Clinical trials of ketogenic regimens in various
few very small cohort studies (Class  III/IV evi- forms are ongoing for a wide range of neurolog-
dence) (Consortium-for-Spinal-Cord-Medicine, ical and nonneurological disorders. The former
2006; Rodriguez et  al., 1997; Thibault-Halman include Parkinson’s disease (NCT013644545),
et al., 2011). Nutrition is generally recommended Tay Sachs disease (NCT01364545), stroke (NCT
within 24 hours using a balanced enteral for- 01820663), multiple sclerosis (NCT01915433),
mula composed of carbohydrates, fat and protein mild cognitive impairments (NCT02521818),
(Consortium-for-Spinal-Cord-Medicine, 2006; childhood epilepsy (NCT02497105), refractory
Thibault-Halman et al., 2011). epileptic status (NCT01796574), adult epilepsy
Contrasting these clinical guidelines, our lab- (NCT01906398), autism (NCT02477904), gli-
oratory discovered in rats that a form of caloric oblastoma (NCT01865162) and acute TBI in
restriction known as intermittent fasting (food children (NCT02174016). Clinically, KDs are
withdrawal every other day) improved outcomes successfully used for drug-resistant epilepsy in
from acute cervical and thoracic SCI by promot- children and for a variety of genetic metabolic dis-
ing neuroprotection and neuroplasticity (Jeong et orders, notably Glut1 deficiency (Kossoff, 2011).
al., 2011; Plunet et al., 2010; Plunet et al., 2008). In this chapter, we review the neuroprotective
These results raise new and interesting questions, effects of KDs and ketones as they relate to neu-
especially given the widely appreciated notion that rotrauma, specifically SCI and TBI; place them
intermittent fasting can have a positive impact on into context with our own data using KDs in
multiple chronic diseases in animal models and rodents with acute SCI; and discuss the potential
on parameters of metabolic, cardiovascular and mechanisms. Due to the overlapping pathophysi-
inflammatory diseases in humans (Brandhorst ology of SCI and traumatic TBI we discuss some of
et al., 2015; Longo and Mattson, 2014; Maalouf the evidences for possible benefits of KDs in both
et al., 2009). Despite the growing enthusiasm conditions concurrently.
for fasting in the research field in recent years,
introducing such a regimen to acutely injured K E T O G E N I C D I E T I M P R OV E S
patients is met with little enthusiasm from clini- OUTCOME AFTER SPINAL
cians, despite the fact that the caloric basal met- C O R D   I N J U RY
abolic rate and energy expenditure requirements The previously established neuroprotective effects
are drastically reduced after SCI due to paralysis of KD/ketone administration and the fact that
(Cook et al., 2008; Magnuson et al., 2011)—unlike our rats treated with intermittent fasting after
in traumatic brain injury (TBI), where they may SCI showed ketosis every second day, presented
be increased (Cook et al. 2008). Fasting affects our rationale for a battery of tests of whether
multiple pathways, including the generation of KD after rodent SCI could promote neuropro-
ketone bodies by the liver, most prominently D- tection and recovery (Streijger et al., 2013). We
beta-hydroxybutyrate (βHB) and acetoacetate developed and applied a unilateral hemicontu-
(AcAc), which have become increasingly known sion model of the cervical spinal cord for adult
to be “neuroprotective” through their intensely rats (Lee et al., 2012; Streijger et al., 2013). At 4
studied role in the so-called ketogenic diet (KD) hours after injury, access to ad lib KD consist-
for epilepsy (for reviews, see Gano et al., 2014; ing of a fat:protein+carbohydrate ratio of 3:1 was
Gasior et al., 2006; Rho, 2015). KDs are high in given and maintained for 12 weeks. Blood βHB
135

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 135

levels reached 0.7 mmol/L at 24 hours and peaked (Pierre et al., 2002). Interestingly, while KD pre-
around 1.8 mmol/L at 7 days post injury. While served tissue following SCI, this was abolished
spinal cord injured rats showed a lower food following administration of the MCT inhibitor α-
intake on the first 2 days and lost more weight cyano-4-hydroxycinnamic acid (4-CIN) beginning
acutely after SCI, their weight returned to normal after injury. This suggest that transport of ketone
values within approximately 2 weeks, and both bodies and/or lactate plays a role in the observed
standard diet (SD) and KD rats gained weight protection rather than general metabolic effects
above the preinjury levels thereafter. We ruled out like low glucose levels or free fatty acid-mediated
that a 48-hour fast acutely after injury was respon- events (Streijger et al., 2013). The interpretation of
sible for the improved recoveries seen with KD this experiment, however, is complicated by the
(unpublished observation). The KD-fed rats used fact that 4-CIN also inhibits the mitochondrial
the forelimb on their injured side more frequently pyruvate transporter (McKenna et al., 2001).
than carbohydrate-based SD-fed controls during Taken together, manipulating the composi-
vertical exploration of their environment in a cyl- tion and timing of macronutrient intake can have
inder. Importantly, this improvement was main- profound effects on the outcome from spinal cord
tained beyond week 12, when KD was replaced by trauma in rodents, which questions our current,
SD. The KD-fed rats also displayed a greater active poorly evidenced clinical nutritional guidelines
range of forelimb movement during grooming. for SCI. However, our understanding of how KD
Given that hand function is the most coveted and ketone bodies mediate improved recovery
function in people with cervical SCI (Anderson, from injury is very incomplete.
2004), we performed two different reaching tests,
and on both tasks we observed an improvement K E TO G E N I C D I E T
in rats fed KD in their ability to reach forward I M P R OV E S O U T C O M E
and grasp a small pellet compared with SD-fed A F T E R   T R AU M AT I C
animals. In addition to scoring reaching success, B R A I N   I N J U RY
which indicates little about the strategy the ani- KD has also been shown to improve functional
mal uses to obtain the pellet, reaching can be bro- outcome following TBI in adolescent (35 days old)
ken down into discrete components of motion in but not in adult rats (75 days old) (Appelberg et
time-lapse video-analysis (Whishaw and Gorny, al., 2009). After a controlled cortical impact injury
1994). In our experiments, KD improved the (CCI) and subsequent feeding of a 7:1 KD imme-
pellet grasp and the subsequent supination that diately after injury, adolescent rats showed signifi-
directs the pellet toward the mouth. Histological cant improvements in the time to traverse a beam,
analysis revealed that KD treatment resulted in a measure of fine motor coordination and balance,
smaller gray matter damage and greater protec- on postinjury day 3, but not on postinjury days
tion of neuronal survival in the vicinity of the 4–7. Additionally, on post injury day 3 the number
lesion, less infiltration by inflammatory cells, and of foot slips was reduced in the 35-day-old KD-fed
increased expression of the glucose transporter CCI rats (Appelberg et al., 2009). In the Morris
Glut1. An RT-qPCR mRNA analysis did not reveal Water Maze, 35-day-old KD-fed animals showed
differences in BDNF, PDGFβ, VEGF, HIF1α, or significantly shorter escape latencies compared
SDF1α—although a trending threefold increase in with CCI SD-fed animals. Such motor and cogni-
SDF1α remains to be revisited. tive improvements were not observed among 75-
In agreement with others (Leino et al., 2001), day-old animals; this, however, might have been
KD treatment significantly increased monocar- attributable to increased hyperactivity, which has
boxylic acid transporter 1 (MCT1) expression at been shown by others in adult rats kept on KD
2 weeks post injury (Streijger et al., 2013), which (Murphy and Burnham, 2006; Ziegler et al., 2005).
coincided with elevated blood βHB levels indica- In a preceding study, Prins and coworkers showed
tive of increased cerebral uptake of ketones. that pretreatment with KD reduced CCI lesion
MCTs facilitate the transport of monocarboxylic volume by 58% in 35-day-old adolescent KD-fed
acids such as lactate, pyruvate, and ketone bod- rats and not in the adult KD-fed rats (Prins et al.,
ies across biological membranes (Halestrap and 2005). There were also fewer Fluoro-Jade positive
Wilson, 2012). In the brain, MCT1 is predomi- cells, indicative of better neuronal preservation.
nantly expressed by vascular endothelial cells, Oddly, very young rats 17-days of age did not show
astrocytes, and oligodendrocytes (Halestrap a reduction in lesion volume under these condi-
and Wilson, 2012; Pierre et al., 2002; Pierre and tions (Prins et al., 2005). These findings are inde-
Pellerin, 2005), while MCT2 is mainly neuronal pendently supported by Hu and coworkers, who
136

136 section II: E merging Clinical Applications and Future Potential

performed similar KD experiments on 35-day-old by decreased glycolysis (Prins and Matsumoto,


TBI rats using a weight drop model (Hu et al., 2014). Oxidative damage-driven activation of the
2009a: Hu et al., 2009b). They found a decreased DNA repair enzyme poly ADP-ribose polymerase
expression of the pro-apoptotic gene Bax (Bcl-2 (PARP) may lead to depletion of NAD+ (Besson
Associated X Protein), together with a reduction of et al., 2003; LaPlaca et al., 1999) that would cause a
several markers for apoptosis as well as less edema decrease in glyceraldehyde phosphate dehydroge-
(Hu et al., 2009a; Hu et al., 2009b). It is noteworthy nase activity. Together with an oxidative damage-
to recall that our KD studies in the injured spinal driven decrease in pyruvate dehydrogenase
cord were all performed in adult rats weighing activity, less glucose can be utilized for ATP gen-
around 350 g at time of injury (11–13 weeks old). eration in the TCA cycle and energy failure ensues
(Lee et al., 1999; Sharma et al., 2009; Singh et al.,
POS SIBLE MECHANISMS 2006). Exogenously applied βHB is readily taken
O F   K E TO G E N I C   D I E T S up by the injured brain after a CCI, and used to
restore the depleted ATP stores (Prins et al., 2004).
Ketones Provide an Alternative Source
Such uptake is not seen in the uninjured brain,
of Energy—Anaplerosis
where energy supplies are largely glucose-derived
It is well known that a physiologically low intake (Prins et  al., 2004). However, this depletion of
of carbohydrates during consumption of KDs or energy stores after contusion injury occurs more
fasting typically causes hepatic ketogenesis fueled slowly in adolescent rats than in adult animals
by the beta-oxidation of fatty acids, resulting in (Deng-Bryant et al., 2011). Administration of βHB
increased blood levels of the ketone bodies: βHB, results in a more pronounced increase in βHB lev-
acetoacetate (AcAc), and acetone (Cahill, 2006). els in the brain of younger rats and also restores
These ketone bodies cross the blood-brain/spinal their energy stores more effectively compared
cord barrier and enter neuronal and glial cells via with older rats. This could be explained by the fact
MCTs (Nijland et al., 2014; Pierre and Pellerin, that young rats express higher level of MCTs for
2005). Here, ketone bodies are converted into ace- the uptake of ketone bodies into the brain com-
toacetyl-CoA by the enzyme succinyl-CoA:3-CoA pared with older rats (Prins and Giza, 2006); the
transferase (SCOT) and are broken down into two age specific differences in neuroprotection seen
molecules of Acetyl-CoA that are subsequently with KD treatments after TBI are likely related. In
used in the Krebs cycle, (see, e.g., Fukao et al., 2014; addition, young rats show an elevation of ketone
Kim do and Rho, 2008). Interestingly, brain astro- levels within 6 hours, while this takes closer to a
cytes are an additional site for ketogenesis (Auestad day in adult rats (Prins et al., 2005; Prins and Giza,
et al., 1991; Guzman and Blazquez, 2001). Taken 2006). This time difference might be critical, since
together, KDs provide an effective source of energy injury triggers an immediate secondary cascade
without the need for glycolysis or mitochondrial and the rescue of neuronal tissue after injury is
complex-I and the associated free oxygen radical time sensitive.
byproducts. The brain can utilize ketones for up This metabolic concept of KD and ketone
to 60% of its energy demands (Owen et al., 1967). treatment as key players in protection is sup-
Energy supplies are further boosted by an increase ported by the pronounced neuroprotective
in mitochondrial biogenesis together with an effect after SCI and TBI of acetyl-L-carnitine,
increase of numerous energy metabolism genes in which supplies an acetyl group for mitochon-
rat hippocampi after several weeks of KD (Bough drial acetyl-CoA synthesis used in the TCA
and Rho, 2007; Bough et al., 2006). This can pro- cycle (Patel et al., 2012; Scafidi et al., 2010). Like
vide an important alternative source of energy ketones, in the absence of pyruvate, acetyl-L-
when the activity of pyruvate dehydrogenase is carnitine can be used for mitochondrial respira-
low, as seen after both SCI and TBI (McEwen et tion (Patel et al., 2012; Patel et al., 2010). Direct
al., 2011; Sharma et al., 2009). Pyruvate dehydro- proof for the metabolic role of ketone bodies
genase converts pyruvate to acetyl-CoA, which is could be provided with a transgenic mouse line
for mitochondrial ATP production. Low activity of allowing for a cell (tissue)-specific deletion of
this enzyme could trigger an energy crisis. Hence, the mitochondrial enzyme succinyl-CoA:3-CoA
some of the benefits of KD observed after SCI or transferase (SCOTfl/fl). This will discern the met-
TBI (Prins and Matsumoto, 2014) could be due to abolic role played by ketones as a biofuel for the
this metabolic rescue by ketones. TCA cycle (anaplerosis) from the nonmetabolic
A TBI triggers a transient increase and subse- functions of ketones as inhibitors of inflamma-
quent depression in glucose uptake accompanied tion and oxidation (see next section).
137

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 137

Non-Anaplerotic Effects of Ketogenic Diet It is therefore of significant interest to understand


In addition to the aforementioned anaplerotic how different lipid compositions in varying KD
effects of ketones as fuel for the TCA cycle, the regimens affect their efficacy after neurotrauma.
beneficial effects of KD may also be due to low- In addition, it would be interesting to see whether
normal glucose levels, with reduced glycolysis KD is beneficial for myelination in the aftermath
and/or due to increases in free fatty acids and a of injury when lipids and metabolic energy are
myriad of lipid signaling molecules (for review, see in high demand. Demyelination is regularly seen
Rho, 2015). For example, elevated blood glucose after SCI due to apoptosis of oligodendrocytes
levels are often observed in humans after SCI and (Crowe et al., 1997) and remyelination is notori-
have been shown to correlate with less favorable ously sluggish in the CNS (Franklin and Ffrench-
recovery (Kobayakawa et  al., 2014). Mice under Constant, 2008).
hyperglycemic conditions show exacerbation of In addition, it is becoming increasingly clear
inflammation and secondary damage, activation that ketone bodies have direct non-metabolic
of NFκB, and inferior functional outcome com- actions as ligands for various receptors, transport-
pared with mice in which glucose levels are kept ers, and regulators of enzymes. These likely con-
normal (Kobayakawa et  al., 2014). Similar con- tribute to the beneficial effects of KDs for a wide
siderations apply to TBI in humans, in particular range of neurological disorders that are covered in
when hyperglycemia is stress induced (Bosarge other chapters of this book. Of particular interest
et al., 2015), and there is consensus that blood glu- in the context of SCI are the anti-excitotoxic, anti-
cose should be carefully controlled (Badjatia et al., oxidant, and anti-inflammatory actions of ketone
2014). However in the case of our KD experiments bodies themselves.
in SCI rats, glucose levels were in the normal range
and not significantly different from controls, mak-
Anti-Excitotoxic
ing a low-glucose mediated effect unlikely.
The anti-excitotoxic mechanisms of KD and
Changes in circulating lipids due to KD may
ketone bodies have been extensively reviewed on
have significant effects on functional outcome fol-
multiple occasions due to their relevance to epi-
lowing SCI. Lipids are important constituents of
lepsy treatment (Gano et al., 2014; Kim do et al.,
all mammalian cells and have diverse biological
2015; Kossoff and Rho, 2009; Lutas and Yellen,
functions, including as (1) major building material
2013; Masino and Rho, 2012; Rho, 2015). Both TBI
for neuronal and glial membranes; (2) precursors
and SCI trigger excitotoxic neuronal damage, and
for various messenger molecules such as arachi-
a variety of drugs blocking neuronal excitation are
donic acid (AA), docosahexaenoic acid (DHA),
neuroprotective after injury (Chen et al., 2014b;
ceramide, 1,2-diacylglycerol (DAG), phosphatidic
Park et al., 2004). However, clinical translation of
acid, and lyso-phosphatidic acid; and (3) energy
glutamate receptor blockers has been challenging
reservoirs such as triglycerides. Free fatty acids
in contrast to the less specific excitation-damp-
increase the expression of mitochondrial uncou-
ening drugs like valproic acid, used in epilepsy
pling proteins (UCPs) via activation of peroxisome
(Chen et al. 2014b), or riluzole, a nonspecific
proliferator activating receptors (PPAR) (Debril et
sodium channel blocker used in ALS (Wilson and
al., 2001; Kiec-Wilk et al., 2005). UCPs regulate the
Fehlings, 2014). A clinical trial of riluzole for acute
mitochondrial transmembrane potential and pre-
SCI is currently under way (Fehlings et al., 2015).
vent free oxygen radical (ROS) production when
However, these anti-excitotoxic treatments for SCI
this potential gets too high. The addition to iso-
have in common that they need to be adminis-
lated mitochondria of AA, docosapentaenoic acid
tered within hours after SCI in order to be effec-
(DPA), eicosapentaenoic acid (EPA), palmitoleic
tive. In contrast, the antiepileptogenic effect of KD
acid, myristic acid, and butyric acid all reduced
requires several weeks of treatment to manifest,
the mitochondrial transmembrane potential and
suggesting that prevention of excessive excitation
thereby ROS production (Davis et al., 2008b;
is not a major player in the neuroprotection pro-
Sullivan et al., 2004). Both fasting and KD increase
vided by KD in the setting of acute SCI.
mitochondrial uncoupling proteins which are
likely responsible for the reduction of mitochon-
drial ROS production (Davis et al., 2008a). Along Antioxidant
the same vein, DHA has been shown on multiple Oxidative stress and damage occurs after SCI
occasions to be neuroprotective after SCI and TBI and TBI (for an excellent overview, see Bains and
(Michael-Titus, 2007; Michael-Titus and Priestley, Hall, 2012). ROS lead to peroxynitrite formation
2014), although its mode of action is very broad. and lipid peroxidation, leading to perturbation of
138

138 section II: E merging Clinical Applications and Future Potential

many cellular processes including Ca2+ and Na+/ with carnosic acid provides effective protection
K+ pumps. These failures lead to further Ca2+ over- when given within 15 minutes of injury, and some
load, mitochondrial damage, and secondary ROS benefits such as reduced cytoskeletal breakdown
production. In both TBI and SCI, the byprod- are observed with application at 8 hours after
ucts of oxidative damage can be measured within TBI (Miller et al., 2015). It remains to be shown
minutes of injury and both 3-nitrotyrosine and 4- whether KD or administration of ketone bodies
hydroxynonenal peak between 1 and 3 days. This will be able to elicit such a protective response via
implies some urgency when attempting to prevent this Nrf2-ARE pathway in the time-sensitive set-
secondary injury with an antioxidative treatment. ting of acute neurotrauma.
Indeed, in laboratory animals, most neuroprotec- A recently recognized antioxidant function of
tive treatments are effective if given prior to or at βHB is that of a direct endogenous inhibitor of
the time of injury but lose efficacy when admin- class histone deacetylases at concentrations that
istered after a delay of several hours (Kwon et al., occur with KD administration (EC50 between 2.5
2011). Similarly, in humans with SCI, methyl- and 5 mM) (Shimazu et al., 2013). Kidneys of mice
prednisolone, an antioxidant with glucocorticoid fasted for 24 hours showed increased acetylation
actions, was only somewhat successful when given at the Foxo3a and metallothionin 2 (Mt2) promot-
within 8 hours post-SCI (Bracken et al., 1998). ers due to HDAC1 inhibition by βHB. Both genes
This implies that any diet-induced benefit pro- protect against oxidative stress, whereby Foxo3a
vided by the initiation of KD in the acute stage increases the expression of mitochondrial antioxi-
of injury would likely miss much of the oxidative dant manganese superoxide dismutase (MnSOD)
damage during the first day of injury, since it takes and catalase (Shimazu et al., 2013) (for review,
16–20 hours for the endogenous ketone levels to see Newman and Verdin, 2014). Whether a simi-
significantly rise. lar inhibition of class I HDACs by βHB occurs in
The antioxidant effects of KDs have been the brain or spinal cord and how this relates to
extensively reviewed (Gano et al., 2014). KD the known antioxidant effects of KD has yet to be
up-regulates mitochondrial glutathione (GSH) clarified. What is becoming clear, however, is that
biosynthesis in the brain (Jarrett et al., 2008), ketones may have profound epigenetic effects that
enhances mitochondrial antioxidant status, and reach far beyond anaplerosis.
reduces ROS production (Maalouf et al., 2007),
protecting mtDNA from oxidant-induced dam- Anti-Inflammatory Effects of Ketones
age (Jarrett et al., 2008). The exact mechanism of It is increasingly appreciated that KD has anti-
this GSH increase is unknown. The GSH is syn- inflammatory effects in models of multiple scle-
thetized by glutamate cysteine ligase (GCL) form- rosis (Kim do et al., 2012), stroke (Rahman et
ing gamma-glutamylcycteine, to which glycine al., 2014), pain (Ruskin et al., 2009), and other
is added by glutathione synthase. The activity of disorders (Dupuis et al., 2015). While part of this
GCL, which is rate limiting, is increased by KD may be a consequence of reduced excitotoxic-
(Jarrett et al., 2008) and could be mediated by the ity and improved mitochondrial oxidation with
transcription factor Nrf2 that is found in nuclear less ROS formation, recent research has revealed
fractions of KD-fed rats (Milder et al., 2010). Nrf2 direct effects of βHB as inflammatory regulators.
binds to antioxidant response elements (ARE) of The βHB inhibits caspase 1 cleavage as part of the
several genes including GCL (Milder and Patel, NLRP3 inflammasome complex, which ultimately
2012). It is hypothesized that Nrf2 is inactivated dampens increases in IL1β and IL18 and contrib-
by forming a complex with Kelch-like ECH-asso- utes further to the anti-inflammatory actions of
ciated protein 1 under normal physiological con- ketones (Youm et al., 2015). The inflammasome is
ditions. This complex functions as a redox sensor a multiprotein complex involved in caspase 1 acti-
and releases Nrf2 in situations of stress and high vation and cleavage of pro-IL1β and pro-IL18. It
ROS levels. It remains to be shown whether a is composed of the apoptosis-associated speck-like
transient stress or ROS response after initiation protein containing a caspase recruitment domain
of KD (Milder et al., 2010) is eliciting an increase (CASD) termed ASC and a nucleotide oliomeriza-
in Nrf2 by KD after neurotrauma. Interestingly, tion domain (NOD)-like receptor (e.g., NLRP1
there is an increase in Nrf2-ARE regulated anti- or NLRP3). In vitro, inhibition of procaspase 1
oxidant genes after TBI, but it does not occur cleavage was shown in lipopolysaccharide-primed
until 24–48 hours after injury (Miller et al., 2014), macrophages on activation with ATP using con-
which may be too late to prevent much second- centrations of βHB in the 2- to 10-mM range as
ary damage. Acceleration of Nrf2-ARE activation they occur during fasting. Importantly, this effect
139

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 139

was not seen with acetoacetate or the related the past due to its favorable effects on blood lipid
butryrate and was specific to βHB (Youm et al., levels/composition and anti-inflammatory actions
2015). The authors tested a battery of other NLRP3 deemed responsible for cardiovascular benefits
activators, including the fatty acids palmitate, (Gille et al., 2008; Lukasova et al., 2011). However,
ceramide, and sphingosine and obtained similar βHB is the main known endogenous ligand of the
results (i.e., inhibition by βHB). Priming macro- HCA2 receptor at concentrations that are easily
phages with Toll-like agonists revealed a potent reached by the KD (EC50 0.7 mM) (Taggart et al.,
inhibition of Pro-IL-1β cleavage by βHB treatment 2005). While HCA2 is widely expressed in adi-
at similar concentrations. These βHB effects were pose tissue, where it regulates lipolysis (Taggart et
specific to the NLRP3 inflammasome, and accord- al., 2005), it is also found on immune cells such
ingly, the formation of ASC oligomers, aka specks, as neutrophils, macrophages, and microglia but
in primed and ATP-activated macrophages was not in astrocytes or neurons (Kostylina et al.,
greatly reduced with βHB addition. In vivo, βHB 2008; Rahman et al., 2014). HCA2 receptor acti-
reduced peritoneal infiltration by neutrophils and vation promotes neutrophil apoptosis (Kostylina
IL1β-secreting macrophages following i.p. injec- et al., 2008), and on macrophages/monocytes it
tion of uric acid crystals in mice. activates/attracts a neuroprotective Ly-6Lo subset
Mice carrying a Nlrp3 mutation that leads to (Rahman et al., 2014). The latter study is of partic-
NLRP3 inflammasome activation show severe ular interest, showing that the beneficial effects of
peritoneal neutrophilia, which is greatly reduced KD or niacin in a model of stroke by middle cere-
by feeding of oral ketone esters. Thus, even with bral artery occlusion (MCAO) in mice were lost
levels in the order of 1mM, βHB is an effective when these mice were lacking the HCA2 receptor
and specific inhibitor the NLRP3 inflammasome. (HCA2–/–). Transplantation of bone marrow from
Importantly, these actions are independent of the HCA+/+ wild-type into the HCA–/– mice restored
presence of the GPR109a (HCA2) receptor (see this rescue effect, demonstrating that monocyte/
below), UCP2, and the metabolic function of macrophage HCA2 expression is required. It was
βHB in the TCA cycle. SCI leads to a pronounced also necessary since the inversion of this trans-
increase of the NLRP3 inflammasome, including plantation paradigm (i.e., HCA–/– bone marrow
the partnering protein ASC (both mainly in acti- into wild-type mice) abolished the neuroprotec-
vated microglia) and the well-known activation of tive effect. This beneficial effect of niacin (and,
caspase-1 and pro-IL1β cleavage (Zendedel et al., by inference, βHB) on stroke outcome requires
2015). Increased ASC expression, caspase-1 acti- Cox1 and hematopoietic prostaglandin D2 synth-
vation, and cleavage of pro-IL1β had previously ase (Rahman et al., 2014). The HCA2 receptor is
been described in a moderate cervical spinal cord also activated by dimethylfumarate (DMF), a drug
contusion model at the C5 level in rats (de Rivero used clinically for MS. The HCA2 receptors have
Vaccari et al., 2008). However, these authors did been shown to mediate the inhibition of DMF on
not find the NLRP3 increase but detected NLRP1 neutrophil adhesion and migration in an inflam-
in co-immunoprecipitations with ASC with- matory mouse model of MS (Chen et al., 2014a).
out increases in NLRP1 expression. These dif- Interestingly, prostaglandin D2 and its PD1 recep-
ferences might be due to differences in the SCI tor have been implicated in the mitigation of dam-
models used. Importantly, antibody-mediated age after excitotoxic challenge or stroke (Dore and
ASC neutralization reduced caspase-1 activation Shafique Ahmad, 2015) and to dampen seizures
and cleavage of IL1β and IL18, reducing lesion (Kaushik et al., 2014). Taken together, these data
volume and improving functional recovery in indicate that βHB at concentrations measured
multiple forelimb tests (i.e., grip strength, sticker during fasting or KD treatment can modify the
removal, gait analysis), underscoring the impor- molecular phenotype of blood monocytes and
tance of inflammasome activation in secondary macrophages toward an anti-inflammatory (M2-
injury cascades after SCI. Whether KD or oral like) tissue protective response via direct activation
ketone esters are mediating their benefits in the of the HCA2 receptor. Since neutrophil invasion
injured spinal cord via this mechanism remains and M1 macrophage polarization play important
unclear. damaging roles after SCI (for review, see Donnelly
An additional way to directly modify inflam- and Popovich, 2008; Kigerl et al., 2009; Popovich,
mation by ketones is through activation of the 2014), it is conceivable that the protective effect of
hydroxyl carboxylic acid (HCA2) receptor also the KD is mediated at least in part by these HCA2
known as GPR109A, PUMA-G or niacin recep- receptor triggered mechanisms. This hypothesis
tor. This receptor has received much attention in remains to be tested, and in light of an arsenal of
140

140 section II: E merging Clinical Applications and Future Potential

HCA2 agonists developed to treat dyslipidemias KD is safe when initiated after SCI in the hospital
and atherosclerosis, it is becoming increasingly setting. Yet, there was some transient gastrointes-
attractive. tinal discomfort (diarrhea, nausea, poor appetite,
gastric pain, and abdominal distension) in five
Translational Consideration of Ketogenic patients and a low enthusiasm for the Chinese KD
Diet in the Clinical Setting formulation. This underscores the translatability of
KD and ketone bodies have powerful anti-excito- this dietary approach, but strongly motivates us to
toxic, antiepileptic and anti-inflammatory effects improve this metabolic treatment in order to mini-
that go far beyond their classic function as an mize unpleasant side effects and increase compli-
energy-providing metabolic boost and provide ance to be able to implement a 2- to 3-month-long
neuroprotection by multiple mechanisms. One regimen after SCI in a human clinical trial.
translational challenge lies in the timing of KD For the same reason, more “liberal” diets
administration, since secondary injury cascades have been explored in the epilepsy field with
begin within minutes of neurotrauma. As a matter the hope for similar benefits. These alterna-
of fact, most neuroprotective treatments are most tives share restriction in the amount/type of
effective when given prior to injury and lose their carbohydrates and vary in the lipid and protein
effects when administered more than 1–3 hours content. They include a medium chain triglyc-
after injury (Kwon et al., 2011). However, a dietary eride enriched KD diet (MCT-KD), a low glyce-
intervention with KD does not increase ketone body mic index treatment (LGIT) and the “modified
levels until the end of the first day—in particular Atkins diet” (MAD) (Miranda et al., 2012). The
in the context of injury stress as it occurs after SCI latter alternative has been the most extensively
(Streijger et al., 2013). In this regard, the neuropro- studied and was in its original form introduced
tective effects of KD after SCI represent one of the by Dr. Atkins in the 1970s. However ketosis with
more promising interventions currently available. the original Atkins is variable and in order to
While this may be encouraging, we hypothesize that mimic the ketosis of classical KD the amount of
in the acute setting of neurotrauma, a faster increase carbohydrates are strictly reduced in the MAD to
in ketone bodies would be greatly advantageous. A less than 20 g per day (in adults) while the intake
carefully monitored direct infusion of βHB for the in protein is not restricted (unlike in KD). While
first days after trauma in parallel with a KD could be still ketogenic, this MAD results in low-normal
one avenue. An alternative approach has been envi- blood glucose levels due to gluconeogenesis from
sioned by Richard Veech and colleagues by suggest- proteins. Compared with the 3:1 ratio (3 g fat to
ing the use of oral ketone-ester supplementation 1 g of protein+carbohydrate) in KD, the ratio in
after TBI (Veech et al., 2012). In vivo oral adminis- MAD is ~1:1 whereby the carbohydrates provide
tration of ketone esters readily increases βHB levels <6% of the total calories, protein ~25%–35% and
to the desired range of 3–4 mmol/L and has been the lipids ~60%–70% of the total calories; per
shown to improve cognition and histopathology in gram, fat has more than twice as many calories
3xTgAD mice that are a model for Alzheimer’s dis- as protein or carbohydrates. Thus the MAD is
ease (Kashiwaya et al., 2013). Whether or not such much easier to implement and better tolerated
supplementation with oral ketone esters after acute by the patients than the 3:1 KD, where <3% of
TBI and SCI is best combined with KD or will be the calories stem from carbohydrates, ~11%
effective by itself as a “diet in a bottle” remains to be from protein and ~88% from fat. MAD appears
determined. almost as effective as KD as long as the carbo-
Our clinical colleagues in Guangzhou (China) hydrate intake is strictly controlled (Sharma et
recently conducted a clinical trial of KD in patients al., 2013; for review, see Auvin, 2012; Kossoff,
with acute SCI to evaluate its safety and feasibility 2011, 2013; Miranda et al., 2012). A meta-anal-
(Guo et al., 2014). These investigators introduced ysis of close to 20 studies indicates that a 50%
KD treatment in the acute phase after injury in 10 rate of seizure reduction with MAD is nearly as
patients with SCI. Comparable to what was seen good as the 56% rate with classical KD (Kossoff
in the rodent models of SCI (Streijger et al., 2013), et al., 2013). Whether MAD is as effective as KD
all patients developed ketone levels above 2 mM in the SCI setting is worth further investigation.
(typically ~ 3 mmol/L) while maintaining normal It would certainly make a huge difference for
glucose levels (9 out of 10). Routine blood tests for individuals with neurotrauma and most likely
electrolytes and liver and kidney function showed increase their compliance, if we could trial a
no changes. This demonstrates that diet-induced MAD instead of a classical KD, even when con-
ketosis in patients with SCI is feasible and that the ducting a dietary treatment for only 2–3 months,
141

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 141

which is the presently anticipated treatment Besson, V.C., Croci, N., Boulu, R.G., Plotkine, M.,
duration after SCI. and Marchand-Verrecchia, C. (2003). Deleterious
We hope to close the “translational gap” from poly(ADP-ribose)polymerase-1 pathway activa-
bench to bedside by collecting the necessary data tion in traumatic brain injury in rat. Brain Res
to better understand the underlying mechanisms 989, 58–66.
Bosarge, P.L., Shoultz, T.H., Griffin, R.L., and Kerby,
responsible for the beneficial effects of KDs in the
J.D. (2015). Stress-induced hyperglycemia is
setting of SCI and by conducting the necessary
associated with higher mortality in severe trau-
preclinical experiments to decide in what exact matic brain injury. J Trauma Acute Care Surg 79,
form the benefits of the KD and ketone bodies 289–294.
could be used for clinical trials in humans with Bough, K.J., and Rho, J.M. (2007). Anticonvulsant
acute SCI and TBI. mechanisms of the ketogenic diet. Epilepsia
48, 43–58.
AC K N OW L E D G M E N T S Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
The authors thank Brett Hilton for critically read- Gawryluk, J.W., Greene, J.G., Shaw, R., Smith,
ing this manuscript; he is supported by a Banting Y., Geiger, J.D., and Dingledine, R.J. (2006).
and Best Scholarship. Wolfram Tetzlaff holds the Mitochondrial biogenesis in the anticonvulsant
John and Penny Ryan British Columbia Leadership mechanism of the ketogenic diet. Ann Neurol 60,
Chair in Spinal Cord Injury; the research in his 223–235.
Bracken, M.B., Shepard, M.J., Holford, T.R., Leo-
laboratory is supported by the Canadian Institute
Summers, L., Aldrich, E.F., Fazl, M., Fehlings,
for Health Research, the Multiple Sclerosis Society M.G., Herr, D.L., Hitchon, P.W., Marshall, L.F.,
of Canada, Wings for Life, the International Spinal et  al. (1998). Methylprednisolone or tirilazad
Research Trust, Craig H.  Nielsen Foundation, mesylate administration after acute spinal cord
and the Rick Hansen Institute and Rick Hansen injury:  1-year follow up; results of the third
Foundation. National Acute Spinal Cord Injury randomized
controlled trial. J Neurosurg 89, 699–706.
REFERENCES Brandhorst, S., Choi, I.Y., Wei, M., Cheng, C.W.,
Anderson, K.D. (2004). Targeting recovery:  pri- Sedrakyan, S., Navarrete, G., Dubeau, L., Yap, L.P.,
orities of the spinal cord-injured population. J Park, R., Vinciguerra, M., et al. (2015). A periodic
Neurotrauma 21, 1371–1383. diet that mimics fasting promotes multi-system
Appelberg, K.S., Hovda, D.A., and Prins, M.L. (2009). regeneration, enhanced cognitive performance,
The effects of a ketogenic diet on behavioral out- and healthspan. Cell Metabol 22, 86–99.
come after controlled cortical impact injury in Cahill, G.F., Jr. (2006). Fuel metabolism in starvation.
the juvenile and adult rat. J Neurotrauma 26, Annu Rev Nutr 26, 1–22.
497–506. Chen, H., Assmann, J.C., Krenz, A., Rahman, M.,
Auestad, N., Korsak, R.A., Morrow, J.W., and Edmond, Grimm, M., Karsten, C.M., Kohl, J., Offermanns,
J. (1991). Fatty acid oxidation and ketogenesis by S., Wettschureck, N., and Schwaninger, M.
astrocytes in primary culture. J Neurochem 56, (2014a). Hydroxycarboxylic acid receptor 2 medi-
1376–1386. ates dimethyl fumarate’s protective effect in EAE. J
Austin, J.W., Kang, C.E., Baumann, M.D., DiDiodato, Clin Invest 124, 2188–2192.
L., Satkunendrarajah, K., Wilson, J.R., Stanisz, G.J., Chen, S., Wu, H., Klebe, D., Hong, Y., and Zhang, J.
Shoichet, M.S., and Fehlings, M.G. (2012). The (2014b). Valproic acid: a new candidate of thera-
effects of intrathecal injection of a hyaluronan- peutic application for the acute central nervous
based hydrogel on inflammation, scarring and system injuries. Neurochem Res 39, 1621–1633.
neurobehavioural outcomes in a rat model of Cheng, B., Yang, X., An, L., Gao, B., Liu, X., and Liu,
severe spinal cord injury associated with arach- S. (2009). Ketogenic diet protects dopaminergic
noiditis. Biomaterials 33, 4555–4564. neurons against 6-OHDA neurotoxicity via up-
Auvin, S. (2012). Should we routinely use modified regulating glutathione in a rat model of Parkinson’s
Atkins diet instead of regular ketogenic diet to disease. Brain Res 1286, 25–31.
treat children with epilepsy? Seizure 21, 237–240. Consortium-for-Spinal-Cord-Medicine. (2006). Early
Badjatia, N., Vespa, P., and Participants of the Acute Management in Adults with Spinal Cord
International Multi-disciplinary Consensus Injury. http://wwwnxtbookcom/nxtbooks/pva/
Conference on Multimodality, M. (2014). earlyacutemanagement/indexphp
Monitoring nutrition and glucose in acute brain Cook, A.M., Peppard, A., and Magnuson, B. (2008).
injury. Neurocrit Care 21(Suppl 2), S159–167. Nutrition considerations in traumatic brain injury.
Bains, M., and Hall, E.D. (2012). Antioxidant therapies Nutr Clin Pract 23, 608–620.
in traumatic brain and spinal cord injury. Biochim Crowe, M.J., Bresnahan, J.C., Shuman, S.L., Masters,
Biophys Acta 1822, 675–684. J.N., and Beattie, M.S. (1997). Apoptosis and
142

142 section II: E merging Clinical Applications and Future Potential

delayed degeneration after spinal cord injury in Franklin, R.J., and Ffrench-Constant, C. (2008).
rats and monkeys. Nat Med 3, 73–76. Remyelination in the CNS: from biology to ther-
Davis, L.M., Pauly, J.R., Readnower, R.D., Rho, J.M., apy. Nat Rev Neurosci 9, 839–855.
and Sullivan, P.G. (2008a). Fasting is neuroprotec- Fukao, T., Mitchell, G., Sass, J.O., Hori, T., Orii, K., and
tive following traumatic brain injury. Journal of Aoyama, Y. (2014). Ketone body metabolism and
Neuroscience Research 86, 1812–1822. its defects. J. Inherit. Metab. Dis. 37, 541–551.
Davis, L.M., Rho, J.M., and Sullivan, P.G. (2008b). UCP- Furlan, J.C., Noonan, V., Cadotte, D.W., and Fehlings,
mediated free fatty acid uncoupling of isolated M.G. (2011). Timing of decompressive surgery of
cortical mitochondria from fasted animals: corre- spinal cord after traumatic spinal cord injury: an
lations to dietary modulations. Epilepsia 49(Suppl evidence-based examination of pre-clinical and
8), 117–119. clinical studies. J Neurotrauma 28, 1371–1399.
de Rivero Vaccari, J.P., Lotocki, G., Marcillo, A.E., Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic
Dietrich, W.D., and Keane, R.W. (2008). A diets, mitochondria, and neurological diseases. J
molecular platform in neurons regulates inflam- Lipid Res 55, 2211–2228.
mation after spinal cord injury. J Neurosci 28, Gasior, M., Rogawski, M.A., and Hartman, A.L. (2006).
3404–3414. Neuroprotective and disease-modifying effects of
Debril, M.B., Renaud, J.P., Fajas, L., and Auwerx, J. the ketogenic diet. Behav Pharmacol 17, 431–439.
(2001). The pleiotropic functions of peroxisome Gibson, C.L., Murphy, A.N., and Murphy, S.P. (2012).
proliferator-activated receptor gamma. J Mol Med Stroke outcome in the ketogenic state: a systematic
79, 30–47. review of the animal data. J Neurochem 123 (Suppl
Deng-Bryant, Y., Prins, M.L., Hovda, D.A., and Harris, 2), 52–57.
N.G. (2011). Ketogenic diet prevents alterations Gille, A., Bodor, E.T., Ahmed, K., and Offermanns,
in brain metabolism in young but not adult rats S. (2008). Nicotinic acid:  pharmacological effects
after traumatic brain injury. J Neurotrauma 28, and mechanisms of action. Annu Rev Pharmacol
1813–1825. Toxicol 48, 79–106.
Donnelly, D.J., and Popovich, P.G. (2008). Guo, C., Zhou, J., Wu, X., Jiang, H., Lu, K., Chen, J.,
Inflammation and its role in neuroprotection, axo- Wu, Z., Yu, R., Liu, J., and Zhu, Q. (2014). [A clini-
nal regeneration and functional recovery after spi- cal trial of ketogenic diet in patients with acute spi-
nal cord injury. Exp. Neurol. 209, 378–388. nal cord injury:  safety and feasibility]. Nan Fang
Dore, S., and Shafique Ahmad, A. (2015). Cytoprotective Yi Ke Da Xue Xue Bao = J Southern Med Univ 34,
role of prostaglandin D2 DP1 receptor against neu- 571–575.
ronal injury following acute excitotoxicity and cere- Guzman, M., and Blazquez, C. (2001). Is there an
bral ischemia. In Brain Neurotrauma:  Molecular, astrocyte-neuron ketone body shuttle? Trends
Neuropsychological, and Rehabilitation Aspects, Endocrinol Metabol: TEM 12, 169–173.
F.H. Kobeissy, ed. (CRC Press/Taylor & Francis, Halestrap, A.P., and Wilson, M.C. (2012). The mono-
Boca Raton, FL). carboxylate transporter family:  role and regula-
Dupuis, N., Curatolo, N., Benoist, J.F., and Auvin, S. tion. IUBMB Life 64, 109–119.
(2015). Ketogenic diet exhibits anti-inflammatory Harkema, S., Behrman, A., and Barbeau, H. (2012a).
properties. Epilepsia 56, e95–98. Evidence-based therapy for recovery of function
Evaniew, N., Belley-Cote, E.P., Fallah, N., Noonan, after spinal cord injury. Handb Clin Neurol 109,
V., Rivers, C.S., and Dvorak, M.F. (2015). 259–274.
Methylprednisolone for the treatment of patients Harkema, S.J., Hillyer, J., Schmidt-Read, M., Ardolino,
with acute spinal cord injuries:  A  systematic E., Sisto, S.A., and Behrman, A.L. (2012b).
review and meta-analysis. J Neurotrauma. Mar Locomotor training: as a treatment of spinal cord
1;33(5):468–81 injury and in the progression of neurologic reha-
Fehlings, M.G., Vaccaro, A., Wilson, J.R., Singh, A., D, bilitation. Arch Phys Med Rehabil 93, 1588–1597.
W.C., Harrop, J.S., Aarabi, B., Shaffrey, C., Dvorak, Hertz, L., Chen, Y., and Waagepetersen, H.S. (2015).
M., Fisher, C., et  al. (2012). Early versus delayed Effects of ketone bodies in Alzheimer’s disease
decompression for traumatic cervical spinal cord in relation to neural hypometabolism, beta-
injury:  results of the Surgical Timing in Acute amyloid toxicity, and astrocyte function. Journal
Spinal Cord Injury Study (STASCIS). PLoS One 7 of Neurochemistry 134, 7–20.
(2), e32037. Hu, Z.G., Wang, H.D., Jin, W., and Yin, H.X. (2009a).
Fehlings, M.G., Nakashima, H., Nagoshi, N., Chow, Ketogenic diet reduces cytochrome c release and
D.S., Grossman, R.G., and Kopjar, B. (2015). cellular apoptosis following traumatic brain injury
Rationale, design and critical end points for in juvenile rats. Ann Clin Lab Sci 39, 76–83.
the Riluzole in Acute Spinal Cord Injury Study Hu, Z.G., Wang, H.D., Qiao, L., Yan, W., Tan, Q.F., and
(RISCIS): a randomized, double-blinded, placebo- Yin, H.X. (2009b). The protective effect of the keto-
controlled parallel multi-center trial. Spinal Cord. genic diet on traumatic brain injury-induced cell
2016 Jan;54(1):8–15 death in juvenile rats. Brain Injury 23, 459–465.
143

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 143

Imamura, K., Takeshima, T., Kashiwaya, Y., King, V.R., Huang, W.L., Dyall, S.C., Curran, O.E.,
Nakaso, K., and Nakashima, K. (2006). D-beta- Priestley, J.V., and Michael-Titus, A.T. (2006).
hydroxybutyrate protects dopaminergic SH-SY5Y Omega-3 fatty acids improve recovery, whereas
cells in a rotenone model of Parkinson’s disease. J omega-6 fatty acids worsen outcome, after spi-
Neurosci Res 84, 1376–1384. nal cord injury in the adult rat. J Neurosci 26,
Jarrett, S.G., Milder, J.B., Liang, L.P., and Patel, M. 4672–4680.
(2008). The ketogenic diet increases mitochondrial Kobayakawa, K., Kumamaru, H., Saiwai, H., Kubota,
glutathione levels. J Neurochem 106, 1044–1051. K., Ohkawa, Y., Kishimoto, J., Yokota, K., Ideta, R.,
Jeong, M.A., Plunet, W., Streijger, F., Lee, J.H., Plemel, Shiba, K., Tozaki-Saitoh, H., et  al. (2014). Acute
J.R., Park, S., Lam, C.K., Liu, J., and Tetzlaff, W. hyperglycemia impairs functional improvement
(2011). Intermittent fasting improves functional after spinal cord injury in mice and humans. Sci
recovery after rat thoracic contusion spinal cord Transl Med 6, 256ra137.
injury. J Neurotrauma 28, 479–492. Kossoff, E. (2011). Ketogenic Diets:  Treatments
Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima, for Epilepsy and Other Disorders, 5th edition
K., Clarke, K., and Veech, R.L. (2000). D-beta- (New York: Demos Health).
hydroxybutyrate protects neurons in models of Kossoff, E.H. (2013). Nonpharmacological
Alzheimer’s and Parkinson’s disease. Proc Natl approaches:  diet and neurostimulation. Handb
Acad Sci USA 97, 5440–5444. Clin Neurol 111, 803–808.
Kashiwaya, Y., Bergman, C., Lee, J.H., Wan, R., King, Kossoff, E.H., and Rho, J.M. (2009). Ketogenic
M.T., Mughal, M.R., Okun, E., Clarke, K., Mattson, diets:  evidence for short- and long-term efficacy.
M.P., and Veech, R.L. (2013). A ketone ester diet Neurotherapeutics 6, 406–414.
exhibits anxiolytic and cognition-sparing proper- Kossoff, E.H., Cervenka, M.C., Henry, B.J., Haney,
ties, and lessens amyloid and tau pathologies in a C.A., and Turner, Z. (2013). A decade of the modi-
mouse model of Alzheimer’s disease. Neurobiol fied Atkins diet (2003–2013): Results, insights, and
Aging 34, 1530–1539. future directions. Epilepsy Behav 29, 437–442.
Kaushik, M.K., Aritake, K., Kamauchi, S., Hayaishi, Kostylina, G., Simon, D., Fey, M.F., Yousefi, S., and
O., Huang, Z.L., Lazarus, M., and Urade, Y. Simon, H.U. (2008). Neutrophil apoptosis medi-
(2014). Prostaglandin D(2) is crucial for seizure ated by nicotinic acid receptors (GPR109A). Cell
suppression and postictal sleep. Exp Neurol Death Differ 15, 134–142.
253, 82–90. Krueger, H., Noonan, V.K., Trenaman, L.M., Joshi, P.,
Khalatbary, A.R. (2014). Natural polyphenols and spi- and Rivers, C.S. (2013). The economic burden of
nal cord injury. Iran Biomed J 18, 120–129. traumatic spinal cord injury in Canada. Chronic
Kiec-Wilk, B., Dembinska-Kiec, A., Olszanecka, A., Dis Inj Can 33, 113–122.
Bodzioch, M., and Kawecka-Jaszcz, K. (2005). The Kwon, B.K., Okon, E., Hillyer, J., Mann, C., Baptiste,
selected pathophysiological aspects of PPARs acti- D., Weaver, L.C., Fehlings, M.G., and Tetzlaff, W.
vation. J Physiol Pharmacol 56, 149–162. (2011). A systematic review of non-invasive phar-
Kigerl, K.A., Gensel, J.C., Ankeny, D.P., Alexander, macologic neuroprotective treatments for acute
J.K., Donnelly, D.J., and Popovich, P.G. (2009). spinal cord injury. J Neurotrauma 28, 1545–1588.
Identification of two distinct macrophage subsets LaPlaca, M.C., Raghupathi, R., Verma, A., Pieper, A.A.,
with divergent effects causing either neurotoxicity Saatman, K.E., Snyder, S.H., and McIntosh, T.K.
or regeneration in the injured mouse spinal cord. J (1999). Temporal patterns of poly(ADP-ribose)
Neurosci 29, 13435–13444. polymerase activation in the cortex following
Kim, D., Zai, L., Liang, P., Schaffling, C., Ahlborn, D., experimental brain injury in the rat. J Neurochem
and Benowitz, L.I. (2013). Inosine enhances axon 73, 205–213.
sprouting and motor recovery after spinal cord Lee, J.H., Streijger, F., Tigchelaar, S., Maloon, M., Liu,
injury. PloS One 8, e81948. J., Tetzlaff, W., and Kwon, B.K. (2012). A contu-
Kim do, Y., and Rho, J.M. (2008). The ketogenic diet sive model of unilateral cervical spinal cord injury
and epilepsy. Curr Opin Clin Nutr Metab Care 11, using the infinite horizon impactor. J Vis Exp. Jul
113–120. 24;(65). pii: 3313
Kim do, Y., Hao, J., Liu, R., Turner, G., Shi, F.D., and Lee, S.M., Wong, M.D., Samii, A., and Hovda, D.A.
Rho, J.M. (2012). Inflammation-mediated mem- (1999). Evidence for energy failure following irre-
ory dysfunction and effects of a ketogenic diet in versible traumatic brain injury. Ann N Y Acad Sci
a murine model of multiple sclerosis. PloS One 7, 893, 337–340.
e35476. Leino, R.L., Gerhart, D.Z., Duelli, R., Enerson, B.E.,
Kim do, Y., Simeone, K.A., Simeone, T.A., Pandya, J.D., and Drewes, L.R. (2001). Diet-induced ketosis
Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G., increases monocarboxylate transporter (MCT1)
and Rho, J.M. (2015). Ketone bodies mediate anti- levels in rat brain. Neurochem Int 38, 519–527.
seizure effects through mitochondrial permeabil- Lim, S., Chesser, A.S., Grima, J.C., Rappold, P.M.,
ity transition. Ann Neurol 78, 77–87. Blum, D., Przedborski, S., and Tieu, K. (2011).
144

144 section II: E merging Clinical Applications and Future Potential

D-beta-hydroxybutyrate is protective in mouse traumatic brain injury in mice. Exp Neurol 264,
models of Huntington’s disease. PloS One 6, 103–110.
e24620. Miller, D.M., Wang, J.A., Buchanan, A.K., and Hall,
Longo, V.D., and Mattson, M.P. (2014). Fasting: molec- E.D. (2014). Temporal and spatial dynamics of
ular mechanisms and clinical applications. Cell nrf2-antioxidant response elements mediated gene
Metab 19, 181–192. targets in cortex and hippocampus after controlled
Lukasova, M., Malaval, C., Gille, A., Kero, J., and cortical impact traumatic brain injury in mice. J
Offermanns, S. (2011). Nicotinic acid inhibits Neurotrauma 31, 1194–1201.
progression of atherosclerosis in mice through its Miranda, M.J., Turner, Z., and Magrath, G. (2012).
receptor GPR109A expressed by immune cells. J Alternative diets to the classical ketogenic diet: can
Clin Invest 121, 1163–1173. we be more liberal? Epilepsy Res 100, 278–285.
Lutas, A., and Yellen, G. (2013). The ketogenic Murphy, P., and Burnham, W.M. (2006). The ketogenic
diet: metabolic influences on brain excitability and diet causes a reversible decrease in activity level in
epilepsy. Trends Neurosci 36, 32–40. Long-Evans rats. Exp. Neurol. 201, 84–89.
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The Newman, J.C., and Verdin, E. (2014). Ketone bodies
neuroprotective properties of calorie restriction, as signaling metabolites. Trends Endocrinol Metab
the ketogenic diet, and ketone bodies. Brain Res 25, 42–52.
Rev 59, 293–315. Nijland, P.G., Michailidou, I., Witte, M.E., Mizee, M.R.,
Maalouf, M., Sullivan, P.G., Davis, L., Kim, D.Y., and van der Pol, S.M., van Het Hof, B., Reijerkerk, A.,
Rho, J.M. (2007). Ketones inhibit mitochondrial Pellerin, L., van der Valk, P., de Vries, H.E., et al.
production of reactive oxygen species production (2014). Cellular distribution of glucose and mono-
following glutamate excitotoxicity by increasing carboxylate transporters in human brain white
NADH oxidation. Neuroscience 145, 256–264. matter and multiple sclerosis lesions. Glia 62,
Magnuson, B., Peppard, A., and Auer Flomenhoft, D. 1125–1141.
(2011). Hypocaloric considerations in patients Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan,
with potentially hypometabolic disease states. J.M., Herrera, M.G., and Cahill, G.F., Jr. (1967).
Nutr Clin Pract 26, 253–260. Brain metabolism during fasting. J Clin Invest 46,
Masino, S.A., and Rho, J.M. (2012). Mechanisms of 1589–1595.
ketogenic diet action. In Jasper’s Basic Mechanisms Oyinbo, C.A. (2011). Secondary injury mechanisms in
of the Epilepsies, J.L. Noebels, M. Avoli, M.A. traumatic spinal cord injury: a nugget of this mul-
Rogawski, R.W. Olsen, and A.V. Delgado- tiply cascade. Acta Neurobiol Exp 71, 281–299.
Escueta, eds. (National Center for Biotechnology Pallier, P.N., Poddighe, L., Zbarsky, V., Kostusiak,
Information (US), Bethesda, MD). M., Choudhury, R., Hart, T., Burguillos, M.A.,
McEwen, M.L., Sullivan, P.G., Rabchevsky, A.G., and Musbahi, O., Groenendijk, M., Sijben, J.W., et  al.
Springer, J.E. (2011). Targeting mitochondrial (2015). A nutrient combination designed to
function for the treatment of acute spinal cord enhance synapse formation and function improves
injury. Neurotherapeutics 8, 168–179. outcome in experimental spinal cord injury.
McKenna, M.C., Hopkins, I.B., and Carey, A. (2001). Neurobiol Dis 82, 504–515.
Alpha-cyano-4-hydroxycinnamate decreases both Paoli, A., Bianco, A., Damiani, E., and Bosco, G. (2014).
glucose and lactate metabolism in neurons and Ketogenic diet in neuromuscular and neurodegen-
astrocytes:  implications for lactate as an energy erative diseases. BioMed Res Int 2014, 474296.
substrate for neurons. J Neurosci Res 66, 747–754. Park, E., Velumian, A.A., and Fehlings, M.G. (2004).
Michael-Titus, A.T. (2007). Omega-3 fatty acids and The role of excitotoxicity in secondary mechanisms
neurological injury. Prostaglandins Leukot Essent of spinal cord injury:  a review with an emphasis
Fatty Acids 77, 295–300. on the implications for white matter degeneration.
Michael-Titus, A.T., and Priestley, J.V. (2014). Journal of Neurotrauma 21, 754–774.
Omega-3 fatty acids and traumatic neurological Patel, S.P., Sullivan, P.G., Lyttle, T.S., and Rabchevsky,
injury:  from neuroprotection to neuroplasticity? A.G. (2010). Acetyl-L-carnitine ameliorates mito-
Trends Neurosci 37, 30–38. chondrial dysfunction following contusion spinal
Milder, J., and Patel, M. (2012). Modulation of oxida- cord injury. J Neurochem 114, 291–301.
tive stress and mitochondrial function by the keto- Patel, S.P., Sullivan, P.G., Lyttle, T.S., Magnuson, D.S.,
genic diet. Epilepsy Res 100, 295–303. and Rabchevsky, A.G. (2012). Acetyl-L-carnitine
Milder, J.B., Liang, L.P., and Patel, M. (2010). Acute treatment following spinal cord injury improves
oxidative stress and systemic Nrf2 activation by mitochondrial function correlated with remark-
the ketogenic diet. Neurobiol Dis 40, 238–244. able tissue sparing and functional recovery.
Miller, D.M., Singh, I.N., Wang, J.A., and Hall, E.D. Neuroscience 210, 296–307.
(2015). Nrf2-ARE activator carnosic acid decreases Patel, S.P., Sullivan, P.G., Pandya, J.D., Goldstein, G.A.,
mitochondrial dysfunction, oxidative damage VanRooyen, J.L., Yonutas, H.M., Eldahan, K.C.,
and neuronal cytoskeletal degradation following Morehouse, J., Magnuson, D.S., and Rabchevsky,
145

Chapter 16: Treatment of Traumatic Brain and Spinal Cord Injury 145

A.G. (2014). N-acetylcysteine amide preserves Rodriguez, D.J., Benzel, E.C., and Clevenger, F.W.
mitochondrial bioenergetics and improves func- (1997). The metabolic response to spinal cord
tional recovery following spinal trauma. Exp injury. Spinal Cord 35, 599–604.
Neurol 257, 95–105. Ruskin, D.N., Kawamura, M., and Masino, S.A. (2009).
Pierre, K., Magistretti, P.J., and Pellerin, L. (2002). Reduced pain and inflammation in juvenile and
MCT2 is a major neuronal monocarboxylate adult rats fed a ketogenic diet. PloS One 4, e8349.
transporter in the adult mouse brain. J Cereb Scafidi, S., Racz, J., Hazelton, J., McKenna, M.C., and
Blood Flow Metab 22, 586–595. Fiskum, G. (2010). Neuroprotection by acetyl-L-
Pierre, K., and Pellerin, L. (2005). Monocarboxylate carnitine after traumatic injury to the immature
transporters in the central nervous system:  dis- rat brain. Dev Neurosci 32, 480–487.
tribution, regulation and function. J Neurochem SCIRE. (2105). Spinal Cord Injury Rehabilitation
94, 1–14. Evidence. wwwscireprojectcom.
Plunet, W.T., Lam, C.K., Lee, J.H., Liu, J., and Tetzlaff, Sharma, P., Benford, B., Li, Z.Z., and Ling, G.S. (2009).
W. (2010). Prophylactic dietary restriction may Role of pyruvate dehydrogenase complex in trau-
promote functional recovery and increase lifespan matic brain injury and measurement of pyruvate
after spinal cord injury. Ann N Y Acad Sci 1198 dehydrogenase enzyme by dipstick test. J Emerg
Suppl 1, E1–11. Trauma Shock 2, 67–72.
Plunet, W.T., Streijger, F., Lam, C.K., Lee, J.H., Liu, J., Sharma, S., Sankhyan, N., Gulati, S., and Agarwala, A.
and Tetzlaff, W. (2008). Dietary restriction started (2013). Use of the modified Atkins diet for treat-
after spinal cord injury improves functional recov- ment of refractory childhood epilepsy: a random-
ery. Exp Neurol 213, 28–35. ized controlled trial. Epilepsia 54, 481–486.
Popovich, P.G. (2014). Neuroimmunology of trau- Shimazu, T., Hirschey, M.D., Newman, J., He, W.,
matic spinal cord injury: a brief history and over- Shirakawa, K., Le Moan, N., Grueter, C.A.,
view. Exp Neurol 258, 1–4. Lim, H., Saunders, L.R., Stevens, R.D., et  al.
Prins, M.L., Fujima, L.S., and Hovda, D.A. (2005). (2013). Suppression of oxidative stress by beta-
Age-dependent reduction of cortical contusion hydroxybutyrate, an endogenous histone deacety-
volume by ketones after traumatic brain injury. J lase inhibitor. Science 339, 211–214.
Neurosci Res 82, 413–420. Singh, I.N., Sullivan, P.G., Deng, Y., Mbye, L.H., and
Prins, M.L., and Giza, C.C. (2006). Induction of mono- Hall, E.D. (2006). Time course of post-traumatic
carboxylate transporter 2 expression and ketone mitochondrial oxidative damage and dysfunc-
transport following traumatic brain injury in juve- tion in a mouse model of focal traumatic brain
nile and adult rats. Dev Neurosci 28, 447–456. injury: implications for neuroprotective therapy. J
Prins, M.L., Lee, S.M., Fujima, L.S., and Hovda, D.A. Cereb Blood Flow Metab 26, 1407–1418.
(2004). Increased cerebral uptake and oxidation of Streijger, F., Plunet, W.T., Lee, J.H., Liu, J., Lam, C.K.,
exogenous betaHB improves ATP following trau- Park, S., Hilton, B.J., Fransen, B.L., Matheson, K.A.,
matic brain injury in adult rats. J Neurochem 90, Assinck, P., et al. (2013). Ketogenic diet improves
666–672. forelimb motor function after spinal cord injury in
Prins, M.L., and Matsumoto, J.H. (2014). The collective rodents. PloS One 8, e78765.
therapeutic potential of cerebral ketone metabolism Sullivan, P.G., Rippy, N.A., Dorenbos, K., Concepcion,
in traumatic brain injury. J Lipid Res 55, 2450–2457. R.C., Agarwal, A.K., and Rho, J.M. (2004). The
Puchowicz, M.A., Zechel, J.L., Valerio, J., Emancipator, ketogenic diet increases mitochondrial uncou-
D.S., Xu, K., Pundik, S., LaManna, J.C., and Lust, pling protein levels and activity. Ann Neurol 55,
W.D. (2008). Neuroprotection in diet-induced 576–580.
ketotic rat brain after focal ischemia. J Cereb Blood Suzuki, M., Suzuki, M., Kitamura, Y., Mori, S., Sato,
Flow Metab 28, 1907–1916. K., Dohi, S., Sato, T., Matsuura, A., and Hiraide,
Rabchevsky, A.G., Sullivan, P.G., Fugaccia, I., and A. (2002). Beta-hydroxybutyrate, a cerebral func-
Scheff, S.W. (2003). Creatine diet supplement for tion improving agent, protects rat brain against
spinal cord injury: influences on functional recov- ischemic damage caused by permanent and tran-
ery and tissue sparing in rats. J Neurotrauma 20, sient focal cerebral ischemia. Jpn J Pharmacol
659–669. 89, 36–43.
Rahman, M., Muhammad, S., Khan, M.A., Chen, Taggart, A.K., Kero, J., Gan, X., Cai, T.Q., Cheng, K.,
H., Ridder, D.A., Muller-Fielitz, H., Pokorna, B., Ippolito, M., Ren, N., Kaplan, R., Wu, K., Wu, T.J.,
Vollbrandt, T., Stolting, I., Nadrowitz, R., et  al. et  al. (2005). (D)-beta-Hydroxybutyrate inhibits
(2014). The beta-hydroxybutyrate receptor HCA2 adipocyte lipolysis via the nicotinic acid receptor
activates a neuroprotective subset of macrophages. PUMA-G. J Biol Chem 280, 26649–26652.
Nat Commun 5, 3944. Thibault-Halman, G., Casha, S., Singer, S., and Christie,
Rho, J.M. (2015). How does the ketogenic diet S. (2011). Acute management of nutritional
induce anti-seizure effects? Neurosci Lett Jul 26. demands after spinal cord injury. J Neurotrauma
pii: S0304–3940(15)30054–9 28, 1497–1507.
146

146 section II: E merging Clinical Applications and Future Potential

Tieu, K., Perier, C., Caspersen, C., Teismann, P., Wu, blocks NLRP3 inflammasome-mediated inflam-
D.C., Yan, S.D., Naini, A., Vila, M., Jackson- matory disease. Nat Med 21, 263–269.
Lewis, V., Ramasamy, R., et  al. (2003). D-beta- Zendedel, A., Johann, S., Mehrabi, S., Joghataei, M.T.,
hydroxybutyrate rescues mitochondrial respiration Hassanzadeh, G., Kipp, M., and Beyer, C. (2015).
and mitigates features of Parkinson disease. J Clin Activation and regulation of NLRP3 inflam-
Nutr 112, 892–901. masome by intrathecal application of SDF-1a
Veech, R.L., Valeri, C.R., and VanItallie, T.B. (2012). in a spinal cord injury model. Mol Neurobiol
The mitochondrial permeability transition pore Jul;53(5):3063–75
provides a key to the diagnosis and treatment of Zhao, W., Varghese, M., Vempati, P., Dzhun, A., Cheng,
traumatic brain injury. IUBMB Life 64, 203–207. A., Wang, J., Lange, D., Bilski, A., Faravelli, I., and
Whishaw, I.Q., and Gorny, B. (1994). Arpeggio and Pasinetti, G.M. (2012). Caprylic triglyceride as a
fractionated digit movements used in prehension novel therapeutic approach to effectively improve
by rats. Behav Brain Res 60, 15–24. the performance and attenuate the symptoms due
Wilson, J.R., and Fehlings, M.G. (2014). Riluzole for to the motor neuron loss in ALS disease. PloS One
acute traumatic spinal cord injury:  a promis- 7, e49191.
ing neuroprotective treatment strategy. World Zhao, Z., Lange, D.J., Voustianiouk, A., MacGrogan,
Neurosurgery 81, 825–829. D., Ho, L., Suh, J., Humala, N., Thiyagarajan, M.,
Yang, L., Yao, M., Lan, Y., Mo, W., Sun, Y.L., Wang, Wang, J., and Pasinetti, G.M. (2006). A keto-
J., Wang, Y.J., and Cui, X.J. (2015). Melatonin genic diet as a potential novel therapeutic inter-
for spinal cord injury in animal models:  a sys- vention in amyotrophic lateral sclerosis. BMC
tematic review and network meta-analysis. J Neurosci 7, 29.
Neurotrauma. Ziegler, D.R., Gamaro, G.D., Araujo, E., Bassani, M.G.,
Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg, E.L., Perry, M.L., Dalmaz, C., and Goncalves, C.A.
Bodogai, M., Kim, D., D’Agostino, D., Planavsky, (2005). Nociception and locomotor activity are
N., Lupfer, C., Kanneganti, T.D., et  al. (2015). increased in ketogenic diet fed rats. Physiol Behav
The ketone metabolite beta-hydroxybutyrate 84, 421–427.
147

17
Anti-Inflammatory Effects of a Ketogenic Diet
Implications for New Indications

N I N A D U P U I S , P H D A N D S T É P H A N E AU V I N, M D , P H D

INTRODUCTION K E TO G E N I C D I E T E X H I B I T S
The ketogenic diet (KD) is a high-fat, low- A N T I - I N F L A M M AT O RY
carbohydrate diet that results in ketosis, modula- PROPERTIES
tion of glycemia, relative caloric restriction, and The use of the KD in a pain model first led to
elevations in the levels of certain fatty acids. In the idea that this high-fat, low-carbohydrate diet
clinical practice, KD is an established treatment might possess anti-inflammatory properties. In
for pharmacoresistant epilepsy, and is increasingly this study, the KD attenuated thermal nociception
being explored for some inflammation-induced and decreased peripheral edema (details below)
epileptic encephalopathies such as fever-induced (Ruskin et  al., 2009). We recently further estab-
refractory epileptic encephalopathy in school- lished the anti-inflammatory effects of the KD
age children (FIRES) (Nabbout et al., 2011). The using a rat model of fever (Dupuis et al., 2015). In
FIRES syndrome begins with severe seizures Wistar rats, 2 weeks of KD treatment before lipo-
evolving into status epilepticus, and is preceded polysaccharide (LPS) i.p. injection reduced the
by fever or common viral infection but without fever response as well as pro-inflammatory cyto-
an identifiable cause (Dupuis and Auvin, 2015). kine levels. In this fever model, the body tempera-
Whether fever itself or some other factor is the ture did not rise similarly in KD-treated animals
trigger remains to be determined. The outcome compared with the controls. The body tempera-
is very severe with adverse motor and cognitive ture was actually significantly lower in the KD
consequences as well as medically refractory epi- group (around 37.6°C vs 38.5°C) from 1 h 30 to
lepsy (Kramer et al., 2011). It is hypothesized that 4 h after LPS administration (Dupuis et al., 2015).
the synergy between inflammation and status epi- The fever profile correlated with plasma IL- IL-
lepticus, the latter resulting partly from inflam- 1B levels: 1 h after injection 130.8 ± 36.3 pg/mL
mation itself and contributing to the induction of in control versus 51.4 ± 9.1 pg/mL in KD (p < .05);
inflammation, generates a vicious cycle that leads 2 h after injection 203.3 ± 65.8 pg/mL in con-
to major neurological consequences (Nabbout trol versus 171.9 ± 38.8 pg/mL in KD (p < .05).
et al., 2011). Four hours after LPS injection, IL-1β and TNF-α
While the antiseizure mechanism(s) remain(s) plasma levels were significantly lower in the KD
unclear, new uses for the KD are increasingly group compared with the control group, although
suggested, given its broad efficacy in various no difference was observed before LPS injection.
experimental models, including animal models In contrast, 4 hours after LPS injection, PGE2
exhibiting prominent inflammatory changes. Our plasma levels were not different between the two
aim in this chapter is to review the data support- groups. Interestingly, 4 hours after LPS injection,
ing the anti-inflammatory profile of the KD, and the KD group also showed lower IL1β mRNA lev-
to link improvements in various animal models of els in the hippocampus compared with controls.
neurological disease with KD-induced modula- As expected, total fatty acid levels were increased
tion of inflammation. Finally, we discuss the possi- after 14 days of KD compared with a standard diet.
ble mechanisms underlying the anti-inflammatory The levels of the long-chain omega-3 (n-3) PUFAs
properties of the KD. eicosapentaenoic acid (EPA; C20:5 n-3) and
148

148 section II: E merging Clinical Applications and Future Potential

docosahexaenoic acid (DHA; C22:6 n-3), which Pain


are precursors of some anti-inflammatory agents, Chronic pain persists or progresses over a long
were decreased in the KD group. In contrast, period of time. It seems that some chronic pain
arachidonic acid (AA; C20:4 n-6), an n-6 PUFA patients suffer from long-term immune-system
that is a precursor for the synthesis of some pro- activation resulting in continuous release of pro-
inflammatory eicosanoids, was also reduced by inflammatory cytokines. In contrast, there is
the KD (Dupuis et al., 2015). evidence that levels of anti-inflammatory cyto-
The KD further modulated fever by decreas- kines are decreased in patients with widespread
ing peripheral inflammation and brain IL-1β pain (Ueceyler et al., 2006) and neuropathic pain
expression. Although we cannot exclude an (Uceyler et  al., 2007). This explains why chronic
effect of ketone bodies and/or caloric restric- pain may arise through an increase in pro-
tion in our recent study, we propose that the inflammatory cytokines and the resulting inhibi-
decrease of AA may be key factor in the anti- tion of anti-inflammatory cytokines.
inflammatory properties of the KD. This sugges- In a model of pain induced by complete
tion is underscored by our previous report of a Freund’s adjuvant through intraplantar injection,
decrease in AA levels in patients responding to KD treatment over 3 weeks resulted in a decrease
KD (Porta et al., 2009). in thermal nociception. This effect was associated
Another recent study evaluated the effect of with a reduction in the peripheral inflammatory
a KD on fibroblast growth factor 21 (FGF21) response as measured by paw swelling (volume
and inflammation (Asrih et  al., 2015). The measure) and plasma extravasation (measure-
authors first established that the KD promoted ment of the dye Evans Blue from dissected tissue)
weight loss through increased energy expendi- (Ruskin et  al., 2009). In this study, the authors
ture and reduced food intake. The mice treated compared the response of adult and juvenile
with a KD had increased plasma levels of FGF21; (beginning diet at P21) animals. The KD was effec-
Fgf21 gene expression was also increased in tive in both age groups, but both the hypoalgesic
the liver but decreased in white adipose tissue effect and the reduction in plasma extravasation
(WAT). The FGF21 is produced principally in were more robust in juveniles. However, the pres-
the liver and mainly acts on adipocytes. It has ence of edema is only one indicator of an inflam-
been shown that by targeting adipose tissue, matory response, so these findings only tell part of
FGF21 promotes adiponectin production and the story.
secretion; adiponectin subsequently mediates
the systemic effects of FGF21 (Holland et  al., Multiple Sclerosis
2013; Lin et al., 2013). The authors reported an Multiple sclerosis (MS) is an autoimmune inflam-
increase in the expression of pro-inflammatory matory disorder of the central nervous system
cytokines (Tnfα, Il-6) and macrophage accumu- affecting the white matter. This demyelinating dis-
lation (Emr1, Cd68, Itgam). In addition, a key order results in a wide range of symptoms includ-
mediator of the inflammasome (Nlrp3) was sig- ing motor, cognitive, and sometimes psychiatric
nificantly increased in the liver of mice treated problems. The most widely studied animal model
with a KD. Interestingly, these inflammatory of MS is the experimental autoimmune enceph-
markers were decreased in adipose tissue of KD- alomyelitis (EAE) murine model induced by the
fed mice (Asrih et al., 2015). subcutaneous injection of myelin oligodendrocyte
glycoprotein peptide in complete Freund’s adju-
K E TO G E N I C D I E T vant, followed by injection of pertussis toxin. This
I M P R OV E S T H E   O U T C O M E model is characterized by CNS neurodegeneration
OF NEUROLOGICAL and synaptic loss, particularly in the hippocampus,
DISEASE ANIMAL MODELS with resultant spatial learning and memory defi-
W I T H   I N VO LV E M E N T cits and motor impairment. Spatial learning and
O F   I N F L A M M AT I O N memory is usually quantified by the Morris water
The KD has been widely studied in experimen- maze test, where mice must repeatedly find a hid-
tal models of seizure and epilepsy, and has also den platform in clouded water, and motor impair-
been explored in other models of neurologi- ment is quantified by swim speed (latency) and
cal diseases, such as pain, multiple sclerosis, and distance (path length).
Parkinson’s disease. In these latter conditions, In the only such study to date, a KD was given
the KD appears to render broad neuroprotective 7 days prior to EAE induction and continued until
effects (Table 17.1). motor and cognitive disabilities could be assessed
149
TABLE 17.1 SUMMARY OF PRECLINICAL STUDIES ESTABLISHING THE ANTI- INFLAMMATORY EFFECT OF KD

Type of diet Species Model Anti-inflammatory effect of KD

Ruskin et al., 2009 6.6:1 KD Rats subcutaneous injection of complete Freund’s adju- Decrease both swelling and plasma extravasation
–3 weeks before Exp. vant into one hindpaw
Xan and Chen, 2010 Made by the lab C57BL/6J Mice MPTP model Decrease of activated microglia (Iba1 staining)
–2 weeks before Exp. Decrease of IL-1β, IL-6, TNFα (ELISA of SN)
Kim do et al., 2012 6.3:1 KD (Bio-Serv C57BL/6 mice Experimental autoimmune encephalomyelitis –2–2.5 fold reduction in CNS-derived CD4+ cells and
F3666 diet) –S.C. myelin oligodendrocyte glycoprotein CD11b+ CD45+ cells (macrophage and microglia
–7 days before Exp. (MOG)35–55 peptide + complete Freund’s –tendency toward increased CD4+
adjuvant (CFA) CD25+ Foxp3+ Treg cells
–I.V. 20 ng of pertussis toxin –Lymph node & CNS reduction in cytokines (IL-1β,
IL-6, TNF-α, IL-12, IL-17) and chemokines (IFN-γ,
MCP-1, MIP-1a, MIP-1b)
Asrih et al., 2015 6.3:1 KD (Bio-Serv C57BL/6 mice Liver and White Adipose Tissue (WAT) Liver: Increase of expression of Tnfα, Il-6, Emr1, Cd68,
F3666 diet) Itgam, Nlrp3
–4 weeks before Exp. WAT: Decrease of expression of Tnfα, Il-6, Emr1, Cd68,
Itgam, Nlrp3
Dupuis et al., 2015 3:1 KD (Ketocal) Wistar rats Fever model. Modulate rise in body temperature
–2 weeks before Exp. 50 μg/kg of LPS (Escherichia coli 055:B5) Blood: reduce IL-1β, TNF-α
Brain: reduce IL-1β mRNA
150

150 section II: E merging Clinical Applications and Future Potential

in comparison with a control group (Kim do et al., In addition to the improvement in motor func-
2012). The KD ameliorated both motor func- tion, the KD was found to inhibit degeneration of
tion and cognitive impairment; spatial learning- tyrosine hydroxylase (TH)-positive neurons and
memory function was also significantly improved, preserved dopamine levels in the SN (Yang and
as demonstrated with the Morris water maze and Cheng, 2010). This effect was associated with a
measurements of hippocampal long-term potenti- reduction in activated microglia as shown by iba-
ation. Further, using 7T-MRI imaging techniques, 1 immunostaining. Moreover, the KD decreased
there was a reduction in the size of periventricu- pro-inflammatory cytokine levels (specifically, IL-
lar white matter lesions and restoration of hip- 1β, IL6, and TNF-α) in the SN of MPTP-treated
pocampal volume loss after KD treatment in the animals (Yang and Cheng, 2010).
induced EAE group of mice. Importantly, this con- Most of the experimental data thus far has
stellation of beneficial effects was associated with shown a correlation between amelioration of dis-
modulation of systemic inflammation. Specifically, ease symptomatology by the KD and decreased
the KD reduced by over twofold the number of inflammation (mostly with respect to pro-
CD4-positive T cells and CD11b/CD45-positive inflammatory cytokine levels). However, whether
macrophages/microglia. The KD-fed EAE mice the beneficial effects of a KD in models of PD are a
also showed a tendency toward increases in CD4+ direct consequence of decreased inflammation has
CD25+ Foxp3+ Treg cells. Furthermore, levels yet to be firmly demonstrated.
of several cytokines (IL-1β, IL-6, TNF-α, IL-12,
IL-17) and chemokines (IFN-γ, MCP-1, MIP- POS SIBLE MECHANISMS
1a, MIP-1b) were substantially lower in the KD- SUPPORTING ANTI-
treated EAE group compared with control EAE I N F L A M M AT O RY
mice. This modulation of cytokine and chemo- PROPERTIES OF THE
kine levels was observed in both brain and blood K E TO G E N I C   D I E T
(Kim do et al., 2012). Finally, other than this anti- Although the clinical efficacy of the KD against
inflammatory modulation of lymphocyte prolif- forms of pharmacoresistant epilepsy is well estab-
eration and cytokine/chemokine expression, the lished, the underlying mechanisms are incompletely
KD decreased overall oxidative stress in the brain understood. There have been a number of intrigu-
(Kim do et al., 2012). ing hypotheses advanced to explain KD action
(Lutas and Yellen, 2013; Masino and Rho, 2010).
Parkinson’s Disease However, it is unlikely that a single mechanism can
Parkinson’s disease (PD) is a progressive neuro- explain all of the clinical effects of the KD. In the
degenerative disorder characterized by abnormal context of the present chapter, there are also proba-
shaking, rigidity, and slowness. The symptoms bly multiple mechanisms that are likely involved in
arise from the death of dopaminergic neurons the anti-inflammatory effect of KD (Figure 17.1).
in the substantia nigra (SN). Despite abundant
clinical information about PD, the mechanisms Ketone Bodies
of neurodegeneration in the SN remain unclear. The ketone bodies beta-hydroxybutyrate (BHB)
Nevertheless, there is some evidence for the role and acetoacetate (AcAc) are the products of
on inflammation in PD, since activated microglia fatty acid oxidation in hepatic mitochondria.
have been implicated in the pathogenesis and pro- Recently, several lines of research support a direct
gression of PD (Hirsch and Hunot, 2009; Hirsch role for BHB as an anti-inflammatory mediator
et al., 2012). (Rahman et  al., 2014; Youm et  al., 2015). Beta-
The murine MPTP (1-methyl-4-phenyl- hydroxybutyrate has been shown to directly inhibit
1,2,3,6-tetrahydropyridine) model of PD recapitu- the NLRP3 (NOD-, LRR-, and pyrin domain-
lates the motor dysfunction and neurochemical containing 3)  inflammasome assembly, which is
changes observed in humans diagnosed with this responsible for caspase-1 activation and the release
disorder. As with PD patients, MPTP-treated mice of the pro-inflammatory cytokines IL-1β and IL-
are characterized by selective progressive loss of 18. The NLRP3 functions primarily in cells of the
dopaminergic neurons in the SN and their pro- myeloid lineage. In mouse bone-marrow-derived
jections to the striatum. Ketogenic diet pretreat- macrophages (BMDMs) and human monocytes,
ment has been shown to provide neuroprotective BHB inhibited ATP-induced cleavage of caspase-1
and anti-inflammatory effects and to alleviate the and the processing of the active form of IL-1β in
motor dysfunction induced by MPTP (Yang and LPS-primed cells (Youm et  al., 2015). This effect
Cheng, 2010). was independent of fasting-regulated mechanisms
151

Chapter 17: Anti-Inflammatory Effects of a Ketogenic Diet 151

FIGURE 17.1 Current understanding of the possible mechanisms supporting the anti-inflammatory mechanisms of
the ketogenic diet.
AA: arachidonic acid; BHB: beta-hydroxyButyrate; CR: caloric restriction; HCA2: hydroxyl-carboxylic acid receptor 2; Nfkbia: NFκB
inhibitor alpha; NLRP3:  NOD-, LRR- and pyrin domain-containing 3; mTOR:  mammalian target of rapamycin; PPAR:  peroxisome
proliferator-activated receptors; PUFA: polyunsaturated fatty acids; Timp3: tissue inhibitor of metalloproteinases-3

such as reactive oxygen species (ROS) production that KD promotes weight loss through increased
and inhibition of glycolysis and autophagy, both of energy expenditure and reduced food intake
which are known to regulate the NLRP3 inflam- (Asrih et  al., 2015). Caloric restriction (CR) is
masome. The inhibitory effect of BHB appears a pragmatic strategy to handle excess energy,
specific to NLRP3 because BHB does not inhibit stored in adipose tissues, that might contribute
caspase-1 activation in response to pathogens acti- to systemic inflammation (Ye and Keller, 2010).
vating other inflammasomes—namely, NLRC4 Caloric restriction can increase anti-inflammatory
(NLR family, CARD domain containing 4)  and protein gene expression of mediators such as
AIM2 (absent in melanoma 2) (Youm et al., 2015). NFκB inhibitor alpha (Nfkbia), tissue inhibitor of
Additionally, BHB was shown to inhibit caspase- metalloproteinases-3 (Timp3), and peroxisome
1 activation and IL-1β secretion in mouse models proliferator-activated receptors (PPARs) (Sung
of Muckle-Wells syndrome, a condition associated et al., 2004; Swindell, 2009), and conversely inhibit
with a mutation in NLRP3. Finally, the KD and the pro-inflammatory genes such as TNF-α, IL-6,
associated elevation in BHB levels protected mice COX-2, iNOS, VCAM-1, and ICAM-1 (Higami
bearing the missense Nlrp3 mutation that leads to et al., 2006; Jung et al., 2009).
familial cold auto-inflammatory syndrome (Youm Despite these data, evidence for the potential
et al., 2015). anti-inflammatory mechanisms of CR in humans
Beta-hydroxybutyrate has also been shown to is more limited, and most of the studies address-
be a potent modulator of the HCA2 (hydroxyl- ing this have been developed in obese patients. In
carboxylic acid 2) receptor, and through this target this particular context, CR appears to reduce pro-
exerts neuroprotective effects in a rodent model of inflammatory molecules (Lee et  al., 2010; Salas-
stroke (Rahman et al., 2014). The HCA2 activation Salvado et  al., 2006), but these data reflect more
by BHB induces Ly-6CLo monocytes and/or mac- the correction of an abnormal state to the non-
rophages to deliver a neuroprotective signal to the pathologic state than the changes induced by CR
brain (Rahman et al., 2014). under healthy conditions.
Overall, the underlying mechanisms explain-
Caloric Restriction ing the anti-inflammatory effects of CR are not
The KD is frequently associated with a reduced well understood. However, since many nutri-
calorie intake. More recently, it has been shown ents and growth factors are able to activate
152

152 section II: E merging Clinical Applications and Future Potential

mammalian target of rapamycin (mTOR), the potential drug targets for seizure control (Sampath
anti-inflammatory effect of CR might be in part and Ntambi, 2005). Both PPARα and PPARγ are
the result of stimulating this pathway. Laboratory activated by PUFAs and their eicosanoid derivates,
studies have already demonstrated that the KD as well as by synthetic ligands such as the fibrates
inhibits mTOR signaling in the brain and liver of (Kersten et al., 2000). Activation of PPARs stimu-
normal rats (McDaniel et  al., 2011). This inhibi- lates the transcription of the genes involved in
tion is thought to be related to a decrease in Akt fatty acid oxidation via the formation of heterodi-
signaling in the brain and the liver. McDaniel and meric transcription-factor complexes with the
colleagues also showed that the KD prevented retinoid-X-receptor (RXR) (Smith, 2002). Three
mTOR hyperactivation after kainate-induced types of PPARs have been identified—alpha, beta,
status epilepticus (SE) (McDaniel et  al., 2011). and gamma:  PPARα is expressed mainly in liver,
However, this study did not correlate the modula- kidney, and heart muscle; PPARβ is expressed in
tion of the mTOR by the KD with any SE-induced a number of tissues, such as brain, adipose tissue,
pro-inflammatory changes. and skin; PPARγ is also expressed in the brain.
Both ALA and LA have a greater binding affinity
Polyunsaturated Fatty Acids for PPARα than EPA or DHA (Lin et  al., 1999).
Polyunsaturated fatty acids (PUFAs) are dietary Synthetic PPAR agonists reduce experimentally
lipids that contain more than one double bond. induced inflammation (Cuzzocrea et  al., 2003;
There are two groups of PUFAs: the omega-3 (n-3) Cuzzocrea et  al., 2004; Lo Verme et  al., 2005).
and the omega-6 (n-6) PUFAs. This nomenclature This effect is the result of the inhibition of pro-
refers to the position of the double bond relative to inflammatory pathways involving nuclear fac-
the methyl terminal of the molecule (Taha et  al., tor κ B, signal transduction and transcription-1,
2010). Diet is an important source of PUFAs. and nuclear factor of activated T-cells (Blanquart
Dietary n-3 PUFAs are found in flaxseed, in some et al., 2003).
nuts, in marine fish, and in marine mammals, such
as seals. In contrast, n-6 PUFAs are found in a vari- Adenosine Modulation
ety of animal products and in vegetable oils, such The KD has been shown to modulate adenosine
as canola and corn oil, and make up the major- levels. Astrocytic adenosine kinase (ADK) is an
ity of PUFAs in the modern Western diet (Taha enzyme responsible for adenosine phosphoryla-
et al., 2010). tion and clearance of adenosine from the extra-
With normal dietary intake, n-3 PUFAs can cellular space. Expression of ADK is reduced by
decrease the production of inflammatory eico- the KD, thereby increasing extracellular levels of
sanoids, cytokines, and ROS and the expression adenosine and the activation of inhibitory ade-
of adhesion molecules. Toward these ends, n-3 nosine A1 receptor (A1AR) (Masino et  al., 2011).
PUFAs act both directly (by replacing arachi- Importantly, the effect of the KD on adenosine
donic acid as an eicosanoid substrate and inhibit- has been linked to a decrease in electrographic
ing arachidonic acid metabolism) and indirectly seizure activity (Masino et  al., 2011). The anti-
(by altering the expression of inflammatory genes inflammatory effects of the KD may be explained
through effects on transcription factor activa- in part by adenosine, as this metabolic mediator
tion). The n-3 PUFAs, particularly eicosapentae- has long been known to induce anti-inflammatory
noic acid (EPA; 20:5n-3) and docosahexaenoic activity (Kowaluk et  al., 1998; Linden, 2001).
acid (DHA, 22:6n-3), also have anti-inflammatory Along these lines, adenosine has been reported
actions, mediated primarily by their hydroxylated to reduce central and peripheral inflammation.
metabolites, which include resolvins and doco- Using the A1AR(-/-) genetically engineered mice
sanoids (Hong et  al., 2003). For instance, EPA and a pretreatment with the specific A1AR ago-
and DHA have been reported to incorporate into nist (2’Me-2-chloro-N6-cyclopentyladenosine) in
human neutrophil cells and to decrease the pro- wild-type mice, it was shown that A1AR modulates
duction of the pro-inflammatory prostaglandin LPS-induced transmigration of polymorphonu-
E2 (PGE2) in a dose-dependent manner (Rees clear cells and reduced levels of TNF-α, IL-6, and
et al., 2006). CXCL2/3 in the lung (Ngamsri et al., 2010). There
Ketogenic diet treatment not only increases are also data suggesting that A1Rs are up-regulated
fatty acid levels in the blood but also increases in acute forms of neuroinflammation and down-
levels of specific PUFAs that can bind to and acti- regulated in chronic forms. Activation of cerebral
vate peroxisome proliferator-activated receptors A1R act as a brake for the microglial response after
(PPARs). The PPARs have been considered as traumatic brain injury (Haselkorn et  al., 2010).
153

Chapter 17: Anti-Inflammatory Effects of a Ketogenic Diet 153

Both A1ARs and A2ARs appear to be involved in CLINICAL EVIDENCE AND


the inflammatory response, and pharmacological F U T U R E I N D I C AT I O N S
interventions might mitigate this (Lukashev et al., There is currently no clinical open label study or
2005; Sitkovsky and Ohta, 2005). However, A2ARs controlled trial that demonstrates that the effects
as well as pharmacological intervention targeting of the KD are through an anti-inflammatory
A2ARs might have bidirectional effects on neu- mechanism. Notwithstanding this lack of data, it is
roinflammation. Specifically, the local glutamate intriguing to note once again that the KD is an effec-
level following brain injury is one of the crucial tive treatment for a condition associated with both
factors that determines the direction of an A2AR- inflammation and medically refractory seizures—
mediated effect on neuroinflammation (Dai and namely, FIRES (Nabbout et al., 2010; Singh et al.,
Zhou, 2011). 2014). At present, whether the KD would indeed
be useful for diverse conditions associated with
Reactive Oxygen Species production, neuroinflammation—for example, pain, multiple
Uncoupling Proteins, Mitochondrial sclerosis, and Parkinson’s disease, among others—
Membrane Potential remains unclear, but the current evidence point-
Both BHB and AcAc have been shown to induce ing to broad anti-inflammatory effects of the KD
neuroprotective effects. These effects have been is compelling, as this metabolism-based treatment
linked to reduction of ROS through enhanced could constitute a readily available therapy for
NADH oxidation and inhibition of mitochon- neurological disorders associated with inflamma-
drial permeability transition (Kim do et  al., tion. Detailed preclinical studies in relevant ani-
2007; Kim do et al., 2015; Maalouf et al., 2007). mal models of neurological disease are no doubt
Furthermore, the KD has been shown to enhance of paramount importance to uncover the underly-
mitochondrial biogenesis (Bough et  al., 2006). ing biology of KD action. Ultimately though, well-
The ROS and inflammation are tightly linked designed clinical trials must be implemented to
through a strong reciprocal relationship; ROS validate readily available therapeutic approaches
generated by inflammatory cells act as inflamma- such as the KD.
tory signaling molecules and participate in the
inflammatory response. Furthermore, ROS play REFERENCES
a key role in the control of nuclear factor kappa Asrih, M., Altirriba, J., Rohner-Jeanrenaud, F., and
B (NF-kappaB), activator protein-1 (AP-1), and Jornayvaz, F.R. (2015). Ketogenic diet impairs
other transcription factors involved in gene FGF21 signaling and promotes differential inflam-
expression of both inflammatory and immune matory responses in the liver and white adipose
mediators. When sustained, oxidative stress can tissue. PLoS One 10.
lead to a chronic inflammatory state and inflam- Blanquart, C., Barbier, O., Fruchart, J.C., Staels, B.,
and Glineur, C. (2003). Peroxisome proliferator-
mation can produce aggravate ROS production
activated receptors:  regulation of transcriptional
(Harijith et al., 2014). activities and roles in inflammation. J Steroid
The KD can reduce both ROS production Biochem Mol Biol 85, 267–273.
and inflammation. Recently, it has been shown Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
that ROS can activate the NLRP3 inflamma- Gawryluk, J.W., Greene, J.G., Shaw, R., Smith,
some protein, a molecular platform activated Y., Geiger, J.D., and Dingledine, R.J. (2006).
on signs of cellular “danger” to trigger innate Mitochondrial biogenesis in the anticonvulsant
immune defenses through the maturation of pro- mechanism of the ketogenic diet. Ann Neurol 60,
inflammatory cytokines such as IL-1β and IL-18 223–235.
(Zhou et al., 2011). Moreover, oxidative stress can Cuzzocrea, S., Mazzon, E., Dugo, L., Patel, N.S.A.,
act as a central regulator of HMGB1’s transloca- Serraino, I., Di Paola, R., Genovese, T., Britti, D.,
tion, release, and activity in inflammation and De Maio, M., Caputi, A.P., et al. (2003). Reduction
cell death. Thus, modulation of HMGB1 can limit in the evolution of murine type II collagen-
induced arthritis by treatment with rosiglitazone,
inflammation and reduce tissue damage during
a ligand of the peroxisome proliferator-activated
infection as well as sterile inflammation (Yu et al.,
receptor gamma. Arthritis Rheum 48, 3544–3556.
2015). Ketone bodies now appear to have the abil- Cuzzocrea, S., Pisano, B., Dugo, L., Ianaro, A., Maffia,
ity to target the inflammatory pathways in multi- P., Patel, N.S.A., Di Paola, R., Ialenti, A., Genovese,
ple ways: (1) direct action on NLRP3 as described T., Chatterjee, P.K., et  al. (2004). Rosiglitazone, a
previously (Youm et  al., 2015), and (2)  decrease ligand of the peroxisome proliferator-activated
in ROS production and levels (Kim do et  al., receptor-gamma, reduces acute inflammation. Eur
2007; Kim do et al., 2015; Maalouf et al., 2007). J Pharmacol 483, 79–93.
154

154 section II: E merging Clinical Applications and Future Potential

Dai, S.-S., and Zhou, Y.-G. (2011). Adenosine 2A Kim do, Y., Hao, J., Liu, R., Turner, G., Shi, F.D., and
receptor:  a crucial neuromodulator with bidi- Rho, J.M. (2012). Inflammation-mediated mem-
rectional effect in neuroinflammation and brain ory dysfunction and effects of a ketogenic diet in
injury. Rev Neurosci 22, 231–239. a murine model of multiple sclerosis. PLoS One 7,
Dupuis, N., and Auvin, S. (2015). Inflammation and e35476.
epilepsy in the developing brain:  clinical and Kim do, Y., Simeone, K.A., Simeone, T.A., Pandya, J.D.,
experimental evidence. CNS Neurosci Ther 21, Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G.,
141–151. and Rho, J.M. (2015). Ketone bodies mediate anti-
Dupuis, N., Curatolo, N., Benoist, J.-F., and Auvin, S. seizure effects through mitochondrial permeabil-
(2015). Ketogenic diet exhibits anti-inflammatory ity transition. Ann Neurol 78, 77–87.
properties. Epilepsia 56, e95–e98. Kowaluk, E.A., Bhagwat, S.S., and Jarvis, M.F. (1998).
Harijith, A., Ebenezer, D.L., and Natarajan, V. Adenosine kinase inhibitors. Curr. Pharm. Des. 4,
(2014). Reactive oxygen species at the cross- 403–416.
roads of inflammasome and inflammation. Front Kramer, U., Chi, C.S., Lin, K.L., Specchio, N., Sahin,
Physiol 5. M., Olson, H., Nabbout, R., Kluger, G., Lin, J.J.,
Haselkorn, M.L., Shellington, D.K., Jackson, E.K., and van Baalen, A. (2011). Febrile infection-
Vagni, V.A., Janesko-Feldman, K., Dubey, R.K., related epilepsy syndrome (FIRES): pathogenesis,
Gillespie, D.G., Cheng, D., Bell, M.J., Jenkins, treatment, and outcome A multicenter study on 77
L.W., et al. (2010). Adenosine A(1) Receptor acti- children. Epilepsia 52, 1956–1965.
vation as a brake on the microglial response after Lee, I.S., Shin, G., and Choue, R. (2010). A 12-week
experimental traumatic brain injury in mice. J regimen of caloric restriction improves levels of
Neurotrauma 27, 901–910. adipokines and pro-inflammatory cytokines in
Higami, Y., Barger, J.L., Page, G.P., Allison, D.B., Smith, Korean women with BMIs greater than 23  kg/
S.R., Prolla, T.A., and Weindruch, R. (2006). m(2). Inflamm Res 59, 399–405.
Energy restriction lowers the expression of genes Lin, Q.O., Ruuska, S.E., Shaw, N.S., Dong, D., and
linked to inflammation, the cytoskeleton, the Noy, N. (1999). Ligand selectivity of the per-
extracellular matrix, and angiogenesis in mouse oxisome proliferator-activated receptor alpha.
adipose tissue. J Nutr 136, 343–352. Biochemistry 38, 185–190.
Hirsch, E.C., and Hunot, S. (2009). Neuroinflammation Lin, Z., Tian, H., Lam, K.S.L., Lin, S., Hoo, R.C.L.,
in Parkinson’s disease:  a target for neuroprotec- Konishi, M., Itoh, N., Wang, Y., Bornstein, S.R.,
tion? Lancet Neurol 8, 382–397. Xu, A., et  al. (2013). Adiponectin mediates the
Hirsch, E.C., Vyas, S., and Hunot, S. (2012). metabolic effects of FGF21 on glucose homeosta-
Neuroinflammation in Parkinson’s disease. sis and insulin sensitivity in mice. Cell Metabol 17,
Parkinsonism Rel Disord 18 (Suppl 1), S210–212. 779–789.
Holland, W.L., Adams, A.C., Brozinick, J.T., Bui, Linden, J. (2001). Molecular approach to adenosine
H.H., Miyauchi, Y., Kusminski, C.M., Bauer, S.M., receptors:  receptor-mediated mechanisms of tis-
Wade, M., Singhal, E., Cheng, C.C., et al. (2013). sue protection. Annu Rev Pharmacol Toxicol 41,
An FGF21-adiponectin-ceramide axis controls 775–787.
energy expenditure and insulin action in mice. Lo Verme, J., Fu, J., Astarita, G., La Rana, G., Russo,
Cell Metabol 17, 790–797. R., Calignano, A., and Piomelli, D. (2005). The
Hong, S., Gronert, K., Devchand, P.R., Moussignac, nuclear receptor peroxisome proliferator-activated
R.L., and Serhan, C.N. (2003). Novel docosatri- receptor-alpha mediates the anti-inflammatory
enes and 17S-resolvins generated from docosa- actions of palmitoylethanolamide. Mol. Pharmacol
hexaenoic acid in murine brain, human blood, and 67, 15–19.
glial cells—autacoids in anti-inflammation. J Biol Lukashev, D.E., Ohta, A., and Sitkovsky, M.V. (2005).
Chem 278, 14677–14687. Physiological regulation of acute inflammation
Jung, K.J., Lee, E.K., Kim, J.Y., Zou, Y., Sung, B., Heo, by A(2A) adenosine receptor. Drug Dev. Res 64,
H.S., Kim, M.K., Lee, J., Kim, N.D., Yu, B.P., et al. 172–177.
(2009). Effect of short term calorie restriction on Lutas, A., and Yellen, G. (2013). The ketogenic
pro-inflammatory NF-kB and AP-1 in aged rat diet: metabolic influences on brain excitability and
kidney. Inflamm Res 58, 143–150. epilepsy. Trends Neurosci 36, 32–40.
Kersten, S., Desvergne, B., and Wahli, W. (2000). Maalouf, M., Sullivan, P.G., Davis, L., Kim, D.Y., and
Roles of PPARs in health and disease. Nature 405, Rho, J.M. (2007). Ketones inhibit mitochondrial
421–424. production of reactive oxygen species production
Kim do, Y., Davis, L.M., Sullivan, P.G., Maalouf, M., following glutamate excitotoxicity by increasing
Simeone, T.A., van Brederode, J., and Rho, J.M. NADH oxidation. Neuroscience 145, 256–264.
(2007). Ketone bodies are protective against oxi- Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin,
dative stress in neocortical neurons. J Neurochem D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and
101, 1316–1326. Boison, D. (2011). A ketogenic diet suppresses
155

Chapter 17: Anti-Inflammatory Effects of a Ketogenic Diet 155

seizures in mice through adenosine A(1) recep- Singh, R.K., Joshi, S.M., Potter, D.M., Leber, S.M.,
tors. J Clin Invest 121, 2679–2683. Carlson, M.D., and Shellhaas, R.A. (2014).
Masino, S.A., and Rho, J.M. (2010). Mechanisms of Cognitive outcomes in febrile infection-related
ketogenic diet action. Epilepsia 51, 85–85. epilepsy syndrome treated with the ketogenic diet.
McDaniel, S.S., Rensing, N.R., Thio, L.L., Yamada, Pediatrics 134, e1431–1435.
K.A., and Wong, M. (2011). The ketogenic diet Sitkovsky, M.V., and Ohta, A. (2005). The “danger”
inhibits the mammalian target of rapamycin sensors that STOP the immune response:  the
(mTOR) pathway. Epilepsia 52, e7–e11. A2 adenosine receptors? Trend Immunol 26,
Nabbout, R., Mazzuca, M., Hubert, P., Peudennier, S., 299–304.
Allaire, C., Flurin, V., Aberastury, M., Silva, W., and Smith, S.A. (2002). Peroxisome proliferator-activated
Dulac, O. (2010). Efficacy of ketogenic diet in severe receptors and the regulation of mammalian lipid
refractory status epilepticus initiating fever induced metabolism. Biochem Soc Trans 30, 1086–1090.
refractory epileptic encephalopathy in school age Sung, B., Park, S., Yu, B.P., and Chung, H.Y. (2004).
children (FIRES). Epilepsia 51, 2033–2037. Modulation of PPAR in aging, inflammation, and
Nabbout, R., Vezzani, A., Dulac, O., and Chiron, C. calorie restriction. J Gerontol A Biol Sci Med Sci
(2011). Acute encephalopathy with inflammation- 59, 997–1006.
mediated status epilepticus. Lancet Neurol 10, Swindell, W.R. (2009). Genes and gene expres-
99–108. sion modules associated with caloric restric-
Ngamsri, K.-C., Wagner, R., Vollmer, I., Stark, S., and tion and aging in the laboratory mouse. BMC
Reutershan, J. (2010). Adenosine receptor A1 Genomics 10.
regulates polymorphonuclear cell trafficking and Taha, A.Y., Burnham, W.M., and Auvin, S. (2010).
microvascular permeability in lipopolysaccharide- Polyunsaturated fatty acids and epilepsy. Epilepsia
induced lung injury. J Immunol 185, 4374–4384. 51, 1348–1358.
Porta, N., Vallee, L., Boutry, E., Fontaine, M., Dessein, Uceyler, N., Rogausch, J.P., Toyka, K.V., and Sommer,
A.F., Joriot, S., Cuisset, J.M., Cuvellier, J.C., and C. (2007). Differential expression of cytokines in
Auvin, S. (2009). Comparison of seizure reduc- painful and painless neuropathies. Neurology
tion and serum fatty acid levels after receiving the 69, 42–49.
ketogenic and modified Atkins diet. Seizure Eur J Ueceyler, N., Valenza, R., Stock, M., Schedel, R.,
Epilep 18, 359–364. Sprotte, G., and Sommer, C. (2006). Reduced lev-
Rahman, M., Muhammad, S., Khan, M.A., Chen, els of antiinflammatory cytokines in patients with
H., Ridder, D.A., Muller-Fielitz, H., Pokorna, B., chronic widespread pain. Arthritis Rheum 54,
Vollbrandt, T., Stolting, I., Nadrowitz, R., et  al. 2656–2664.
(2014). The beta-hydroxybutyrate receptor HCA2 Yang, X., and Cheng, B. (2010). Neuroprotective and
activates a neuroprotective subset of macrophages. anti-inflammatory activities of ketogenic diet on
Nat Commun 5, 3944. MPTP-induced neurotoxicity. J Mol Neurosci 42,
Rees, D., Miles, E.A., Banerjee, T., Wells, S.J., Roynette, 145–153.
C.E., Wahle, K.W.J., and Calder, P.C. (2006). Dose- Ye, J., and Keller, J.N. (2010). Regulation of energy
related effects of eicosapentaenoic acid on innate metabolism by inflammation: a feedback response
immune function in healthy humans: a comparison in obesity and calorie restriction. Aging-Us 2,
of young and older men. Am J Clin Nutr 83, 331–342. 361–368.
Ruskin, D.N., Kawamura, M., and Masino, S.A. (2009). Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg,
Reduced pain and inflammation in juvenile and E.L., Bodogai, M., Kim, D., D’Agostino, D.,
adult rats fed a ketogenic diet. PLoS One 4, e8349. Planavsky, N., Lupfer, C., Kanneganti, T.D.,
Salas-Salvado, J., Bullo, M., Garcia-Lorda, P., et  al. (2015). The ketone metabolite beta-
Figueredo, R., Del Castillo, D., Bonada, A., and hydroxybutyrate blocks NLRP3 inflammasome-
Balanza, R. (2006). Subcutaneous adipose tissue mediated inflammatory disease. Nat Med 21,
cytokine production is not responsible for the res- 263–269.
toration of systemic inflammation markers during Yu, Y., Tang, D., and Kang, R. (2015). Oxidative stress-
weight loss. Int J Obes 30, 1714–1720. mediated HMGB1 biology. Front Physiol 6.
Sampath, H., and Ntambi, J.M. (2005). Polyunsaturated Zhou, R., Yazdi, A.S., Menu, P., and Tschopp, J. (2011).
fatty acid regulation of genes of lipid metabolism. A role for mitochondria in NLRP3 inflammasome
Annu Rev Nut 25, 317–340. activation. Nature 469, 221–225.
156

18
Dietary Therapy for Neurological Disorders
Focus on Amyotrophic Lateral Sclerosis, Parkinson’s Disease,
Mood Disorders, and Migraine

C A R L E . S TA F S T R O M , M D ,   P H D

INTRODUCTION of altered energy metabolism points to altered


The ketogenic diet (KD) has established efficacy mitochondrial function as a common factor in
for epilepsy and is best known for this role (Neal pathogenesis (Kunz, 2002). Similar therapeutic
et al., 2008; Winesett et al., 2015). Since the 1920s, efficacy might be achieved through alternative
the KD has been used for treatment of intractable forms of the KD, including the medium chain tri-
epilepsy, often as a last resort. The mechanism glyceride (MCT) diet, modified Atkins diet, low
of the KD is complex and not fully elucidated, glycemic index treatment, or calorie restriction;
but likely involves metabolic adaptations in a these therapies are mentioned only in passing in
variety of cellular signaling pathways leading to this chapter, as their role in nonepileptic disorders
decreased neuronal excitability and neuroprotec- has barely been explored to date.
tion. As such, it is possible that the KD can be
used therapeutically in other neurological diseases T H E N E U RO P ROT E C T I V E
(Barañano and Hartman, 2008; Paoli et al., 2014; ROLE OF THE
Stafstrom and Rho, 2012; Gano et al., 2014), and K E TO G E N I C   D I E T
small preliminary trials have been performed in Over the past 15  years or so, investigators have
brain cancer, Alzheimer disease, autism spectrum identified numerous mechanisms by which the
disorder, multiple sclerosis, and pain, as discussed KD diet might provide neuroprotection (Hartman,
elsewhere in this volume. 2012; Rho and Stafstrom, 2012; Lutas and Yellen,
Here, I  review the use of the KD in amyo- 2013; Danial et  al., 2013). This complex patho-
trophic lateral sclerosis (ALS), Parkinson disease, physiology is covered in other chapters of this
mood disorders, and migraine. These diverse dis- volume. Nevertheless, a brief discussion of patho-
orders do not share a pathophysiological basis that physiological mechanisms is warranted here, as a
is immediately obvious. Amyotrophic lateral scle- basis for explaining why diverse neurologic disor-
rosis is a motor neuron disease involving degen- ders could be amenable to KD treatment.
eration of alpha motor neurons. Parkinson disease Two hallmark features of KD treatment are
entails excitotoxic degeneration of dopaminergic the increase in ketone body production by the
neurons of the substantia nigra leading to abnor- liver and a reduction in blood glucose levels.
mal motor function and cognition. Bipolar dis- Ketone elevation is largely a consequence of fatty
order and other mood disorders are related to acid oxidation. In the setting of high fat and low
dysfunction of neurochemical balance in the brain, carbohydrate intake, as in the KD, fatty acids are
especially monoamines. Migraine is a paroxysmal oxidized in the liver to produce ketone bodies:
headache disorder involving abnormal sensitivity β-hydroxybutyrate and acetoacetate. These ketones
of blood vessels and neurons, especially involving then occasion metabolic adaptations that produce
the trigeminal system. an antiseizure effect. Ketone bodies, as well as
Despite this phenotypic and mechanistic het- specific polyunsaturated fatty acids (PUFAs) such
erogeneity, each disorder involves abnormalities in as arachidonic acid, docosahexaenoic acid, and
cellular energy utilization, implying that a benefi- eicosapentaenoic acid might themselves regulate
cial effect might be possible by manipulating nutri- neuronal membrane excitability by (1)  directly
ents and metabolic substrates. Specifically, the role blocking voltage-gated sodium and calcium
157

Chapter 18: Dietary Therapy for Neurological Disorders 157

channels (Vreugdenhil et al., 1996); (2) altering the Other important mechanisms also contribute
synthesis, degradation, or reuptake of excitatory or to the neuroprotective consequences of calorie
inhibitory neurotransmitters (Yudkoff et al., 2001; restriction, including improved mitochondrial
Juge et  al., 2010); or (3)  reducing inflammation function and decreased oxidative stress (similar
through activation of peroxisome proliferator- to that seen with ketones and PUFAs), decreased
activated receptors (Hajjar et al., 2012). activity of pro-apoptotic factors, and inhibition
How could altered neuronal excitability and of inflammatory mediators such as interleukins
dysregulated cellular energy metabolism be and tumor necrosis factor alpha (Maalouf et  al.,
linked? One way is via ATP-sensitive potassium 2009). Restoring exhausted metabolic substrates
(KATP) channels. β-hydroxybutyrate increases the may constitute another novel treatment approach
probability that KATP channels open, which would (Kovac et al., 2013). In that regard, a biochemical
decrease cellular excitability and reduce firing rate process called anaplerosis aims to replenish tricar-
and seizure occurrence (Ma et al., 2007). Another boxylic acid cycle intermediates that are depleted
route is adenosine, which is produced from ATP during the intense neuronal firing that constitutes
and causes seizure suppression through activa- seizure activity (Willis et al., 2010).
tion of inhibitory adenosine A1 receptors. The KD In the end, numerous mechanisms are likely
increases ATP and thus adenosine, producing an to contribute to the neuroprotective properties
inhibitory effect on cellular excitability (Masino of the KD. While several of these mechanisms
et al., 2009). Ketone bodies have also been shown are thought to relate principally to the antiseizure
to facilitate neuroprotection by raising ATP lev- activity of the KD, the KD also likely contributes
els and reducing ROS production (Kim et  al., to cellular homeostasis and the prevention of
2007)  through enhanced NADH oxidation and neuronal dysfunction and injury. An important
inhibition of mitochondrial permeability tran- caveat, however, is that yet unidentified mecha-
sition (Kim et  al., 2015). Along similar lines of nisms operate in disorders outside of epilepsy, pre-
improved bioenergetics, the KD has been shown senting further opportunities for examining the
to stimulate mitochondrial biogenesis, resulting in pleiotropic effects of metabolism-based therapies
stabilized synaptic function (Bough et  al., 2006). at a mechanistic level. Furthermore, an important
Therefore, altered mitochondrial biogenesis or consideration applicable to all neurodegenerative
function represents a mechanistically appropriate diseases is the timing of intervention that is crucial
target for the KD in both epilepsy and nonepilep- for a neuroprotective effect to be enabled by KD
tic neurologic disorders (Milder and Patel, 2012; treatment. Neurological disorders in late stages of
Pani, 2015). progression may have such extreme neuronal dys-
The second major biochemical feature of the function and death that neuroprotective therapies
KD is decreased glycolytic flux. Reduction of gly- may no longer work. All of these potential mecha-
colysis is an essential feature of calorie restriction, nisms are discussed in greater detail in other chap-
which has been shown to suppress seizures as ters of this volume.
well as prolong the lifespan of numerous species,
including primates (Pani, 2015). While the link EXAMPLES OF NEUROLOGIC
between calorie restriction and KD effectiveness D I S O R D E R S P O T E N T I A L LY
remains controversial, it is clear that both treat- A M E NA B L E TO   T H E
ments result in reduction of blood glucose, likely K E TO G E N I C   D I E T
involving reduced glycolytic flux. In this regard, Several neurologic diseases might benefit from
2-deoxy-D-glucose (2-DG), an analog of glucose dietary therapy, including amyotrophic lateral
that blocks phosphoglucose isomerase and hence sclerosis, Parkinson disease, mood disorders, and
inhibits glycolysis, has been shown to block epi- migraine. These disorders have diverse underlying
leptogenesis in the rat kindling model by decreas- pathophysiologies, yet each entails some contribu-
ing expression of brain-derived neurotrophic tion of cellular energy dysfunction.
factor and its principal receptor, tyrosine kinase B
(Garriga-Canut et  al., 2006). 2-DG is also effec- Amyotrophic Lateral Sclerosis
tive in several acute seizure models (Stafstrom Amyotrophic lateral sclerosis (ALS) is a rapidly
et  al., 2009). Similarly, fructose-1,6-diphosphate progressive disease due to degeneration of motor
reduces glycolysis by diverting glucose to the pen- neurons of the cortex and anterior horn of the
tose phosphate pathway and is neuroprotective in spinal cord. As a consequence, voluntary motor
multiple seizure models (Lian et  al., 2007; Ding activity gradually deteriorates, leaving the affected
et al., 2013). individual profoundly weak despite largely retained
158

158 section II: E merging Clinical Applications and Future Potential

cognitive function. The essential pathophysi- et al., 2014). Both the KD and DP diets were asso-
ological mechanisms that underlie this relentless ciated with improved motor scores compared with
disorder are yet to be fully elucidated, and likely control-fed mutant mice, and the KD and KD+DP
involve oxidative damage, glutamate excitotoxic- diets prolonged life expectancy. These results raise
ity, inflammation, and mitochondrial membrane the possibility that the KD or similar metabolic
dysfunction (Vucic et  al., 2014). Similar to other therapies, by raising ketone levels, might benefit
neurodegenerative disorders, energy-producing patients with ALS. In a broader sense, the role of
systems likely play a role and mitochondrial dys- calories or simply increasing dietary fat in ALS
function probably contributes to disease patho- patients remains a subject of intense interest, since
genesis (Martin, 2011). In this regard, the KD dietary approaches are a rather straightforward
may be a promising adjunctive treatment for ALS way to potentially enhance the quality and dura-
(Siva, 2006). tion of life in this devastating disease (Paganoni
Amyotrophic lateral sclerosis is hereditary in and Wills, 2013).
an estimated 10% of cases. Approximately 20%
of patients with familial ALS harbor a mutation Parkinson Disease
in the gene encoding copper/zinc superoxide dis- The primary pathophysiology in Parkinson disease
mutase (SOD1). A transgenic mouse model of ALS (PD) is excitotoxic degeneration of dopaminergic
has been created in which the gene SOD 1-G93A is neurons in the substantia nigra pars compacta,
overexpressed, leading to progressive muscle weak- leading to abnormalities of movement, cognition,
ness and death resulting from respiratory failure as and other cortical functions. Seizures are not com-
in the human disease. Administration of the KD to mon in PD (Szot, 2012). Therefore, how could the
these mutant mice led to both histological (higher KD benefit patients with PD? A  primary patho-
motor neuron counts) and functional (preserved genic mechanism of cell death in PD is thought to
motor function on the rotarod test) improvements be a defect in complex I of the electron transport
compared with non-KD fed SOD 1-G93A trans- chain. The KD might bypass this defect and allow
genic animals (Zhao et al., 2006). However, the KD oxidative phosphorylation to proceed. In a small
did not extend survival time compared with non- clinical study, five of seven PD patients treated
KD fed control mice. Although providing proof- with the KD showed improved scores on a stand-
of-principle for using the KD in ALS, this study ard PD rating scale (Vanitallie et al., 2005). Sample
is limited by a number of factors including very sizes were small, however, and a placebo effect
small group sizes and initiation of motor testing could not be fully excluded. There is also evidence
before the onset of ALS-type symptoms. that calorie restriction alone helps patients with
Mitochondria isolated from brains of SOD 1- PD (Srivastava and Haigis, 2011), and this finding
G93A mutant mice had increased levels of ATP was verified in a primate model (Maswood et al.,
and increased rates of ATP synthesis, as well 2004); these observations support a critical role of
as decreased function of complex I  of the elec- energy dysregulation in this disorder.
tron transport chain. Administration of the β- An animal model of PD is produced by 1-
hydroxybutyrate restored complex I  activity in methyl- 4- phenyl- 1,2,3,6- tetrahydropyridine
neurons in which complex I function was blocked (MPTP) administration; this toxin causes death of
pharmacologically (Zhao et  al., 2006), a result substantia nigra neurons by inhibiting mitochon-
also seen in mitochondria isolated from brains of drial NADH dehydrogenase (Tieu et al., 2003). β-
a Parkinson’s disease animal model (Tieu et  al., hydroxybutyrate, elevated during KD treatment,
2003). An alternative approach involves caprylic ameliorated MPTP-induced mitochondrial respi-
triglyceride, a medium-chain triglyceride with ratory chain damage in this model by bypassing
antiseizure effects that is metabolized to ketone the defect in complex I  activity. Additional sup-
bodies (Wlaź et  al., 2012). Caprylic triglycer- port for the potential benefits of ketone bodies in
ide, when fed to SOD 1-G93A transgenic mice, PD comes from in vitro experiments. In substantia
reduced motor neuron loss and preserved motor nigra neuron precursors, ketone bodies protect
performance on the rotarod and other tests (Zhao against mitochondrial respiratory chain dysfunc-
et al., 2012). Finally, a recent study examined SOD tion induced exogenously by complex I  and II
1-G93A mutant mice fed a KD variant called the inhibitors and provide anti-inflammatory action
Deanna Protocol (DP, which provides alternative on MPTP-induced neurotoxicity (Kashiwaya et al.,
fuels in the form of tricarboxylic acid cycle inter- 2000; Yang and Cheng, 2010). As in ALS, the KD
mediates such as arginine-α-ketoglutarate), or effect in PD appears to require intact complex
both the standard KD and the DP together (Ari I function (Tieu et al., 2003). In another model of
159

Chapter 18: Dietary Therapy for Neurological Disorders 159

PD induced by 6-hydroxydopamine neurotoxic- pharmacological agents are used to treat both con-
ity in rats, the KD protected nigral dopaminergic ditions (e.g., topiramate, valproate, lamotrigine)
neurons from degeneration (Cheng et al., 2009). (Bialer, 2012). Migraine and epilepsy share con-
siderable phenotypic overlap (Rogawski, 2008),
Mood Disorders although the intrinsic mechanisms underlying
Beneficial effects of the KD have been hypoth- seizures and migraine attacks differ in some fun-
esized in mood disorders, based on the observa- damental respects. There are theoretical reasons to
tion that other antiseizure agents also stabilize consider the KD for chronic migraine. Migraine
mood (e.g., valproate, carbamazepine, lamotrig- involves a complex interplay between genes and
ine) (El-Mallakh and Paskitti, 2001; Bialer, 2012). environment, and some of the causative genes
In addition, the KD alters levels of metabolites of code for proteins involved in energy metabolism
biogenic amines such as dopamine and seroto- (e.g., ATP1A2 in one form of familial hemiplegic
nin, neurotransmitters critical to the pathophysi- migraine; Gritz and Radcliffe, 2013). Brain func-
ology of depression (Murphy et al., 2004; Dahlin tion is highly dependent on normal mitochondrial
et al., 2012; Szot, 2012). The KD increased levels of function to produce sufficient energy via oxida-
endogenous norepinephrine levels in rats, which tive phosphorylation, so disrupted mitochondrial
had an antiseizure effect (Weinshenker, 2008). function has been hypothesized as a causative fac-
With respect to mood disorders, only anecdotal tor in migraine (Roos-Araujo et al., 2014).
clinical studies have been conducted thus far. One It might seem unlikely that an individual
brief KD trial on a single patient with depression was with migraine would undertake such a compli-
reportedly ineffective, although urinary ketosis was cated dietary regimen as the KD, but in light of
not achieved (Yaroslavsky et al., 2002). Two women suboptimal alternatives, the KD is at least worth
with bipolar disorder type II were treated with a KD considering in some cases, particularly in per-
for 2–3  years; both subjects tolerated the diet and sons with headaches that are refractory to conven-
demonstrated improved mood (Phelps et al., 2013). tional agents (Strahlman, 2006; Maggioni et  al.,
Obviously, large-scale studies are needed. 2011). Interestingly, the first report using the KD
Depression in rats is studied using the forced for migraine appeared in 1928, only a few years
swim test, in which an animal is placed into a after the diet’s first use for epilepsy (Schnabel,
water-filled chamber with smooth walls, from 1928). Nine of 28 migraine patients reported
which it cannot escape. After an initial phase of “some improvement,” although the validity of
frantic swimming, a rat becomes immobile, sim- this clinical study is uncertain and some patients
ply floating to prevent drowning. Depressed ani- admitted to poor compliance. In patients with epi-
mals have a shorter latency to the immobile phase, lepsy, better compliance is often achieved using
regarded as a measure of despair. The role of the the less restrictive modified Atkins diet (MAD)
KD in depression has been studied using this (Kossoff and Dorward, 2008). A trial of the MAD
model. Compared with control-fed rats, those fed was attempted in adolescents with chronic daily
the KD spent significantly less time in the immo- headache (Kossoff et al., 2010). Of eight enrolled
bility phase of the test, reflecting an overall reduc- patients, only three patients were able to com-
tion in “behavioral despair” (Murphy et al., 2004). plete the 3-month trial. Although there was some
In recent experiments, mice were exposed to the improvement in reported quality of life in those
KD prenatally (by feeding dams the KD) and then three patients, all of them continued to experience
their behavior was tested in adulthood (Sussman chronic daily headaches. These findings suggest
et  al., 2015). All rats received the standard (non- that even the MAD is too restrictive for long-term
KD) diet after birth. Adult KD offspring exhibited use in adolescents with headache and any thera-
reduced susceptibility to anxiety and depression peutic benefit of ketosis in treating headache is
and were physically more active than control mice. quite modest.
The authors concluded that prenatal exposure to a One large prospective observational study of
KD was associated with decreased stress responses the KD in migraine patients has been reported
in adulthood. At present, it is not clear whether (DiLorenzo et al., 2015). Ninety-six obese female
there are any practical or translational implications patients with migraine self-referred to a dietitian
of these results. for weight control. Patients were randomized to a
KD or standard diet for 1 month. The KD group
Migraine experienced a decrease in mean baseline head-
Epilepsy and migraine involve paroxysmal excita- aches from 2.9 per month to 0.71 per month; the
bility changes in the brain, and many of the same total number of headache days was also fewer in
160

160 section II: E merging Clinical Applications and Future Potential

the KD group (0.91 headache days for the KD REFERENCES


group vs. 5.1 days in controls). When the KD was Ari, C., Poff, A.M., Held, H.E., Landon, C.S., Goldhagen,
stopped and the patients returned to a normal diet, C.R., Mavromates, N., and D’Agostino, D.P. (2014).
the higher migraine headache frequency resumed. Metabolic therapy with Deanna Protocol supple-
A case report of a pair of obese twin sisters with mentation delays disease progression and extends
migraines reported an improvement in headache survival in amyotrophic lateral sclerosis (ALS)
mouse model. PLoS One 9, e103526.
control on a KD, and decreased migraine attacks
Barañano, K.W., and Hartman, A.L. (2008). The keto-
correlated with the level of ketosis (DiLorenzo
genic diet:  uses in epilepsy and other neurologic
et al., 2013). illnesses. Curr Treat Options Neurol 10, 410–419.
Cortical spreading depression (CSD), a wave of Bialer, M. (2012). Why are antiepileptic drugs used
depolarization that spreads slowly along the cor- for nonepileptic conditions? Epilepsia 53 (Suppl
tical surface (~3 mm/min) and is thought to cor- 7), 26–33.
relate with migraine aura (Ferrari et al., 2015), is Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
associated with neurogenic inflammation, form- Gawryluk, J.W., Greene, J.G., Shaw, R., Smith,
ing a target for current antimigraine drugs (Cui Y., Geiger, J.D., and Dingledine, R.J. (2006).
et al., 2014). Laboratory studies have investigated Mitochondrial biogenesis in the anticonvulsant
the effect of the KD on CSD (de Almeida Rabello mechanism of the ketogenic diet. Ann Neurol 60,
Oliveira et al., 2008). It was found that short-term 223–235.
treatment (10 days) with either the MCT or long- Cheng, B., Yang, X., An, L., Gao, B., Liu, X., and Liu,
S. (2009). Ketogenic diet protects dopaminergic
chain triglyceride forms of the KD resulted in
neurons against 6-OHDA neurotoxicity via up-
significant reduction in CSD velocity in rats, but
regulating glutathione in a rat model of Parkinson’s
long-term treatment (7 weeks) with either diet had disease. Brain Res 1286, 25–31.
no effect on CSD. Although the clinical implica- Cui, Y., Kataoka, Y., and Watanabe, Y. (2014). Role of
tions of these results are uncertain, it is clear that cortical spreading depression in the pathophysiol-
the KD reduces cortical excitability in a time- ogy of migraine. Neurosci Bull 30, 812–822.
dependent fashion. It is unknown whether the KD Dahlin, M., Månsson, J.E., and Åmark, P. (2012). CSF
will play a role in headache treatment in the future, levels of dopamine and serotonin, but not norepi-
but the diet may provide some clues as to migraine nephrine, metabolites are influenced by the keto-
pathophysiology. For example, mitochondrial dys- genic diet in children with epilepsy. Epilepsy Res
function, leading to decreased ATP production, 99, 132–138.
has been linked to the occurrence of CSD (Sparaco Danial, N.N., Hartman, A.L., Stafstrom, C.E., and
et al., 2006). Thio, L.L. (2013). How does the ketogenic diet
work? Four potential mechanisms. J Child Neurol
28, 1027–1033.
CONCLUSIONS
de Almeida Rabello Oliveira, M., da Rocha Ataíde, T.,
This chapter has considered several neurologic de Oliveira, S.L., de Melo Lucena, A.L., de Lira,
disorders that might be amenable to metabolic C.E., Soares, A.A., de Almeida, C.B., and Ximenes-
treatment with therapies such as the KD and its da-Silva, A. (2008). Effects of short-term and long-
variants. Despite the relative lack of clinical data term treatment with medium- and long-chain
supporting the use of KD in these disorders, triglycerides ketogenic diet on cortical spreading
preliminary studies support the idea that KD- depression in young rats. Neurosci Lett 434, 66–70.
induced metabolic shifts may lead to neuropro- DiLorenzo, C., Currà, A., Sirianni, G., Coppola, G.,
tection. How can a simple dietary change lead Bracaglia, M., Cardillo, A., DeNardis, L., and
to improvement in a set of disorders with such Pierelli, F. (2013). Diet transiently improves
a huge span of pathophysiological mechanisms? migraine in two twin sisters: possible role of keto-
A  common theme appears to be alteration in genesis? Funct Neurol 28, 305–308.
energy metabolism. So while the mechanisms DiLorenzo, C., Coppola, G., Sirianni, G., DiLorenzo,
G., Bracaglia, M., DiLenola, D., Siracusano, A.,
through which the KD exerts such effects are
Rossi, P., and Pierelli, F. (2015). Migraine improve-
certainly diverse, there may be a finite set of ment during short lasting ketogenesis: a proof-of-
final common pathways that are shared mecha- concept study. Eur J Neurol 22, 170–177.
nistically. Ultimately, the details of how such Ding, Y., Wang, S., Jiang, Y., Yang, Y., Zhang, M., Guo,
metabolic factors reduce excitability, diminish Y., Wang, S., and Ding, M.P. (2013). Fructose-1,6-
ongoing neurodegeneration, or mitigate func- diphosphate protects against epileptogenesis by
tional disability remain unknown. Herein lie rich modifying cation-chloride co-transporters in a
opportunities for further investigation in both model of amygdaloid-kindling temporal epilepti-
the laboratory and the clinic. cus. Brain Res 1539, 87–94.
161

Chapter 18: Dietary Therapy for Neurological Disorders 161

El-Mallakh, R.S., and Paskitti, M.E. (2001). The keto- Kunz, W.S. (2002). The role of mitochondria in epilep-
genic diet may have mood-stabilizing properties. togenesis. Curr Opin Neurol 15, 179–184.
Med Hypotheses 57, 724–726. Lian, X.-Y., Khan, F.A., and Stringer, J.L. (2007).
Ferrari, M.D., Klever, R.R., Terwindt, G.M., Ayata, C., Fructose-1,6-biphosphate has anticonvulsant
and van den Maagdenberg, A.M. (2015). Migraine activity in models of acute seizures in adult rats. J
pathophysiology: lessons from mouse models and Neurosci 27, 12007–12011.
human genetics. Lancet Neurol 14, 65–80. Lutas, A., and Yellen, G. (2013). The ketogenic
Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic diet: metabolic influences on brain excitability and
diets, mitochondria, and neurological diseases. J epilepsy. Trends Neurosci 36, 32–40.
Lipid Res 55, 2211–2228. Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic diet
Garriga-Canut, M., Schoenike, B., Qazi, R., Bergendahl, metabolites reduce firing in central neurons by open-
K., Daley, T.J., Pfender, R.M., Morrison, J.F., ing K(ATP) channels. J Neurosci 27, 3618–3625.
Ockuly, J., Stafstrom, C., Sutula, T., et  al. (2006). Maalouf, M.A., Rho, J.M., and Mattson, M.P. (2009).
2-Deoxy-D-glucose reduces epilepsy progres- The neuroprotective properties of calorie restric-
sion by NRSF-CtBP-dependent metabolic regula- tion, the ketogenic diet, and ketone bodies. Brain
tion of chromatin structure. Nature Neurosci 9, Res Rev 59, 293–315.
1382–1387. Maggioni, F., Margoni, M., and Zanchin, G. (2011).
Gritz, S.M., and Radcliffe, R.A. (2013). Genetic effects Ketogenic diet in migraine treatment:  a brief but
of ATP1A2 in familial hemiplegic migraine type II ancient history. Cephalalgia 31, 1150–1151.
and animal models. Hum Genomics 7, 8. Martin, L.J. (2011). Mitochondrial pathobiology in
Hajjar, T., Meng, G.Y., Rajion, M.A., Vidyadaran, S., ALS. J Bioenerg Biomembr 43, 569–579.
Othman, F., Farjam, A.S., Li, T.A., and Ebrahimi, Masino, S.A., Kawamura, M., Jr., Wasser,
M. (2012). Omega-3 polyunsaturated fatty acid C.D., Pomeroy, L.T., and Ruskin, D.N.
improves spatial learning and hippocampal peroxi- (2009). Adenosine, ketogenic diet and epi-
some proliferator activated receptors (PPARα and lepsy:  the emerging therapeutic relationship
PPARγ) gene expression in rats. BMC Neurosci between metabolism and brain activity. Curr
13, 109. Neuropharmacol 7, 257–268.
Hartman, A.L. (2012). Neuroprotection in metabolism- Maswood, N., Young, J., Tilmont, E., Zhang, Z.,
based therapy. Epilepsy Res 100, 286–294. Gash, D.M., Gerhardt, G.A., Grondin, R., Roth,
Juge, N., Gray, J.A., Omote, H., Miyaji, T., Inoue, T., G.S., Mattison, J., Lane, M.A., et  al. (2004).
Hara, C., Uneyama, H., Edwards, R.H., Nicoll, Caloric restriction increases neurotrophic
R.A., and Moriyama, Y. (2010). Metabolic con- factor levels and attenuates neurochemical
trol of vesicular glutamate transport and release. and behavioral deficits in a primate model of
Neuron 68, 99–112. Parkinson’s disease. Proc Natl Acad Sci USA 52,
Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima, 18171–18176.
K., Clarke, K., and Veech, R.L. (2000). D-beta- Milder, J., and Patel, M. (2012). Modulation of oxida-
hydroxybutyrate protects neurons in models of tive stress and mitochondrial function by the keto-
Alzheimer’s and Parkinson’s disease. Proc Natl genic diet. Epilepsy Res 100, 295–303.
Acad Sci USA 97, 5440–5444. Murphy, P., Likhodii, S., Nylen, K., and Burnham,
Kim, D.Y., Davis, L.M., Sullivan, P.G., Maalouf, M., W.M. (2004). The antidepressant properties of the
Simeone, T.A., van Brederode, J., and Rho, J.M. ketogenic diet. Biol Psychiatry 56, 981–983.
(2007). Ketone bodies are protective against oxi- Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
dative stress in neocortical neurons. J Neurochem Edwards, N., Fitzsimmons, G., Whitney, A., and
101, 1316–1326. Cross, J.H. (2008). The ketogenic diet for the treat-
Kim, D.Y., Simeone, K.A., Simeone, T.A., Pandya, J.D., ment of childhood epilepsy:  a randomised con-
Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G., trolled trial. Lancet Neurol 7, 500–506.
and Rho, J.M. (2015). Ketone bodies mediate anti- Paganoni, S., and Wills, A.M. (2013). High-fat and
seizure effects through mitochondrial permeabil- ketogenic diets in amyotrophic lateral sclerosis. J
ity transition. Ann Neurol 78, 77–87. Child Neurol 28, 989–992.
Kossoff, E.H., and Dorward, J.L. (2008). The modified Pani, G. (2015). Neuroprotective effects of dietary
Atkins diet. Epilepsia 49 (Suppl 8), 37–41. restriction: Evidence and mechanisms. Semin Cell
Kossoff, E.H., Huffman, J., Turner, Z., and Gladstein, Dev Biol 40, 106–114.
J. (2010). Use of the modified Atkins diet for ado- Paoli, A., Bianco, A., Damiani, E., and Bosco, G.
lescents with chronic daily headache. Cephalalgia (2014). Ketogenic diet in neuromuscular and neu-
30, 1014–1016. rodegenerative diseases. Biomed Res Int 2014,
Kovac, S., Abramov, A.Y., and Walker, M.C. (2013). article number 474296.
Energy depletion in seizures: anaplerosis as a strat- Phelps, J.R., Siemers, S.V., and El-Mallakh, R.S. (2013).
egy for future therapies. Neuropharmacology 69, The ketogenic diet for type II bipolar disorder.
96–104. Neurocase 19, 423–426.
162

162 section II: E merging Clinical Applications and Future Potential

Rho, J.M., and Stafstrom, C.E. (2012). The ketogenic Vreugdenhil, M., Bruehl, C., Voskuyl, R.A., Kang,
diet: what has science taught us? Epilepsy Res 100, J.X., Leaf, A., and Wadman, W.J. (1996).
210–217. Polyunsaturated fatty acids modulate sodium and
Rogawski, M.A. (2008). Common pathophysiologic calcium currents in CA1 neurons. Proc Nat Acad
mechanisms in migraine and epilepsy. Arch Sci USA 93, 12559–12563.
Neurol 65, 709–714. Vucic, S., Rothstein, J.D., and Kiernan, M.C. (2014).
Roos-Araujo, D., Stuart, S., Lea, R.A., Haupt, L.M., and Advances in treating amyotrophic lateral scle-
Griffiths, L.R. (2014). Epigenetics and migraine; rosis:  insights from pathophysiological studies.
complex mitochondrial interactions contributing Trends Neurosci 37, 433–442.
to disease susceptibility. Gene 543, 1–7. Weinshenker, D. (2008). The contribution of norepi-
Schnabel, T.G. (1928). An experience with a ketogenic nephrine and orexigenic neuropeptides to the anti-
diet in migraine. Ann Int Med 2, 314–347. convulsant effect of the ketogenic diet. Epilepsia 49
Siva, N. (2006). Can ketogenic diet slow progression of (Suppl 8), 104–107.
ALS? Lancet Neurol 5, 476. Willis, S., Stoll, J., Sweetman, L., and Borges, K. (2010).
Sparaco, M., Feleppa, M., Lipton, R.B., Rapoport, Anticonvulsant effects of a triheptanoin diet in two
A.M., and Bigal, M.E. (2006). Mitochondrial dys- mouse chronic seizure models. Neurobio Dis 40,
function and migraine: evidence and hypotheses. 565–572.
Cephalalgia 26, 361–372. Winesett, S.P., Kordecki, S., and Kossoff, E.H.
Srivastava, S. and Haigis, M.C. (2011). Role of sirtuins (2015). The ketogenic diet in pharmacoresistant
and calorie restriction in neuroprotection:  impli- childhood epilepsy. Expert Rev Neurother 15,
cations in Alzheimer’s and Parkinson’s diseases. 621–628.
Curr Pharm Des 17, 3418–3433. Wlaź, P., Socała, K., Nieoczym, D., Łuszczki, J.J.,
Stafstrom, C.E., Ockuly, J.C., Murphree, L., Valley, Zarnowska, I., Zarnowski, T., Czuczwar, S.J.,
M.T., Roopra, A., and Sutula, T.P. (2009). and Gasior, M. (2012). Anticonvulsant profile of
Anticonvulsant and antiepileptic actions of 2- caprylic acid, a main constituent of the medium-
deoxy-D-glucose in epilepsy models. Ann Neurol chain triglyceride (MCT) ketogenic diet, in mice.
65, 435–447. Neuropharmacology 62, 1882–1889.
Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic Yang, X., and Cheng, B. (2010). Neuroprotective and
diet as a treatment paradigm for diverse neuro- anti-inflammatory activities of ketogenic diet on
logical disorders. Front Pharmacol 3, 1–8. MPTP-induced neurotoxicity. J Mol Neurosci 42,
Strahlman, R.S. (2006). Can ketosis help migraine suf- 145–153.
ferers? A case report. Headache 46, 182. Yaroslavsky, Y., Stahl, Z., and Belmaker, R.H.
Sussman, D., Germann, J., and Henkelman, M. (2015). (2002). Ketogenic diet in bipolar illness. Bipolar
Gestational ketogenic diet programs brain struc- Dis 4, 75.
ture and susceptibility to depression and anxiety in Yudkoff, M., Daikhin, Y., Nissim, I., Lazarow, A., and
the adult mouse offspring. Brain Behav 5, e00300. Nissim, I. (2001). Ketogenic diet, amino acid
Szot, P. (2012). Common factors among Alzheimer’s metabolism, and seizure control. J Neurosci Res
disease, Parkinson’s disease, and epilepsy:  pos- 66, 931–940.
sible role of the noradrenergic nervous system. Zhao, W., Varghese, M., Dzhun, P.V.A., Cheng, A.,
Epilepsia 53 (Suppl 1), 61–66. Wang, J., Lange, D., Bilski, A., Faravelli, I., and
Tieu, K., Perier, C., Caspersen, C., Teismann, P., Wu, Pasinetti, G.M. (2012). Caprylic triglyceride as a
D.C., Yan, S.D., Naini, A., Vila, M., Jackson- novel therapeutic approach to effectively improve
Lewis, V., Ramasamy, R., et  al. (2003). D-beta- the performance and attenuate the symptoms due
hydroxybutyrate rescues mitochondrial respiration to the motor neuron loss in ALS disease. PLoS One
and mitigates features of Parkinson disease. J Clin 7, e49191.
Invest 112, 892–901. Zhao, Z., Lange, D.J., Voustianiouk, A., MacGrogan,
Vanitallie, T.B., Nonas, C., DiRocco, A., Boyar, D., Ho, L., Suh, J., Humala, N., Thiyagarajan, M.,
K., Hyams, K., and Heymsfield, S.B. (2005). Wang, J., and Pasinetti, G.M. (2006). A keto-
Treatment of Parkinson disease with diet- genic diet as a potential novel therapeutic inter-
induced hyperketonemia:  a feasibility study. vention in amyotrophic lateral sclerosis. BMC
Neurology 64, 728–730. Neurosci 7, 29.
163

SECTION III
Ketogenic Diet in the Laboratory

D E T L E V B O I S O N, P H D, S E C T I O N E D I T O R
164
165

19
Overview of Ketogenic Diet in the Laboratory
Progress on Models and Mechanisms

D E T L E V B O I S O N, P H D

I ncreased clinical interest in the ketogenic diet


and similar metabolic treatments has naturally
spurred research into mechanisms underlying
“Ketogenic Diet in the Laboratory,” is to discuss
recent mechanistic studies.
Early in vivo work in rodents showed that KD
their anticonvulsant/antiepileptogenic efficacy; feeding was effective against some but not all types
significant progress and new insights are being of experimentally induced seizures (Appleton and
made after many decades of relative obscurity. DeVivo, 1974; Millichap et  al., 1964; Uhlemann
This section, “Ketogenic Diet in the Laboratory,” and Neims, 1972). Later mechanistic work in ani-
includes discussions of a number of molecular mals turned to in vitro preparations (e.g., Stafstrom
targets that are affected by ketogenic diet feeding. et al., 1999; Thio et al., 2000). Here, Dr. Kawamura’s
In parallel, several alternative experimental meth- chapter reviews in vitro methodologies for mod-
ods to either elucidate or mobilize ketogenic diet eling the KD. These involve direct application of
mechanisms are explored. Based on this labora- ketone bodies, modulation of glucose and intracel-
tory research, putative and novel uses for meta- lular ATP, and maintaining the metabolic milieu of
bolic treatments are proposed. KD-fed animals in acute brain slices.
The first published reports in the medi- Several chapters address anticonvulsant or
cal literature of the effect of a ketogenic diet antiepileptogenic molecular targets for the KD.
(KD) on epilepsy are two one-page reports by Dr. Simeone reviews the literature on peroxisome
R.M. Wilder in the daily Mayo Clinic Bulletin. proliferator-activated receptor γ (PPARγ), a fatty
The first makes a brief theoretical case for use acid receptor that is expressed in many brain neu-
of a high-fat, low-carbohydrate diet based on rons. He describes the regulation of this nutri-
prior successful work with fasting in epilepsy ent sensor by seizure activity and by KD feeding,
(Wilder, 1921a). The second (published the very and that pharmacological or genetic inactivation
next day) consists of case descriptions of three of PPARγ blocks the anticonvulsant effect of KD
patients who became seizure-free after KD treat- feeding. Drs. Sada and Inoue review the functions
ment (Wilder, 1921b). Interestingly, given that of the astrocyte/neuron lactate shuttle, and then
the KD became best known as a treatment for discuss their work in brain slices, in which it was
pediatric epilepsy, two of these three patients shown that lactate reverses the inhibition caused by
were adults (aged 23 and 31). Naturally, from the ketone bodies. Conversely, ketogenic diet feeding
beginning there were questions about the anti- lowered hippocampal lactate levels. Drs. Lusardi
convulsant mechanism of the KD. Wilder’s own and Boison describe the epigenetics of epilepsy,
speculation was that elevated ketone bodies were review the augmentation of the neuromodulator
crucial, given that this metabolic response is in adenosine by KD treatment, and outline the dual
common between fasting and eating a high-fat, functions of adenosine: anticonvulsant effects via
low-carbohydrate diet (Wilder, 1921a). This is modulating synaptic activity and antiepileptogenic
still seen as the key mechanism by many inves- effects via modulating DNA methylation.
tigators, though not without dispute. Other early Several chapters address mechanistic effects
postulated mechanisms such as dehydration and of KD treatment in pathophysiologies other than
acidosis have fallen by the wayside. In fact, these epilepsy. Drs. Curtis, Kemper, et  al. review how
latter two are seen now as unnecessary and treat- reactive oxygen and nitrogen species are involved
able side effects. One main point of this section, in aging and radiation damage. They then discuss
166

166 section III: Ketogenic Diet in the Laboratory

how ketone body-based metabolism generates KD with nondietary treatments. Drs. Martínez-
NADPH, thereby quenching these species through François, Danial, and Yellen review the functions
reactions driven by the [NADP+]/[NADPH] of BCL-2-associated agonist of cell death (BAD),
couple and also by increasing transcription of cer- a protein that regulates glucose metabolism. They
tain antioxidant enzymes. Also in the context of describe how genetic manipulation of BAD to
aging, Drs. Xu, LaManna, and Puchowicz describe reduce glucose metabolism results in mice resist-
the beneficial effects of KD treatment on neuro- ant to seizures, an effect involving ATP-sensitive
degenerative processes. They propose that these potassium channels. Drs. Sada and Inoue pro-
effects arise from ketosis-induced stabilization of pose lactate dehydrogenase blockers to reduce
hypoxia-inducible factor 1α, a metabolic sensor activity of the astrocyte/neuron lactate shuttle;
and transcription factor, and its target genes. Drs. Dr.  Simeone proposes agonists of PPARγ; Drs.
Veech and King review the literature on the patho- Lusardi and Boison propose blockers of adeno-
physiology of Alzheimer’s disease and present evi- sine kinase to boost adenosine and decrease DNA
dence that ketosis-inducing treatments could be methylation; and Drs. Veech and King and Drs.
beneficial in circumventing the inhibition of pyru- Curtis, Kemper, et  al. propose administration of
vate dehydrogenase found in that disease. ketone esters, which are rapidly metabolized to
Additional chapters address other physiologi- ketone bodies.
cal effects of the KD. Drs. Banjara and Janigro
review the biology of the blood-brain barrier
REFERENCES
including the process of ketone body uptake. The Appleton, D.B., and DeVivo, D.C. (1974). An ani-
regulation of glucose and ketone body transport- mal model for the ketogenic diet. Epilepsia 15,
ers in the barrier and the suggestion of barrier 211–227.
repair by KD treatment are discussed. Drs. Harney, Millichap, J.G., Jones, J.D., and Rudis, B.P. (1964).
Gudsnuk, et  al. describe delays in puberty and Mechanism of anticonvulsant action of ketogenic
effects on reproduction in female rats fed a strict diet. Am J Dis Child 107, 593–604.
KD. Effects of KD feeding on spatial reference Stafstrom, C.E., Wang, C., and Jensen, F.E. (1999).
learning in dams and their pups are also presented. Electrophysiological observations in hippocam-
Three chapters deal with novel uses of KDs that pal slices from rats treated with the ketogenic diet.
are mostly (or completely) still confined to the lab- Dev Neurosci 21, 393–399.
oratory. Such is the case with Drs. Veech and King Thio, L.L., Wong, M., and Yamada, K.A. (2000). Ketone
and Drs. Curtis, Kemper, et al. proposing KDs for bodies do not directly alter excitatory or inhibitory
hippocampal synaptic transmission. Neurology
treating Alzheimer’s disease and ionizing radia-
54, 325–331.
tion damage, respectively, both discussed previ- Uhlemann, E.R., and Neims, A.H. (1972).
ously. In addition, Dr. Ruskin reviews the evidence Anticonvulsant properties of the ketogenic diet in
for KD treatment being able to alleviate pain and mice. J Pharmacol Exp Ther 180, 231–238.
inflammation. Wilder, R.M. (1921a). The effects of ketonemia on the
Finally, among these chapters in this sec- course of epilepsy. Mayo Clin Bull 2, 307.
tion many also address treatment stategies and Wilder, R.M. (1921b). High fat diets in epilepsy. Mayo
methodologies to invoke the mechanisms of the Clin Bull 2, 308.
167

20
Ketogenic Diet and PPARgamma
TIMOTHY A. SIMEONE, PHD

INTRODUCTION that regulate this shift in gene expression. In hepa-


tocytes, fatty acids are endogenous ligands for
Historical Overview peroxisome proliferator activated receptor alpha
A switch from a normal Western diet of low-fat (PPARalpha), a nuclear receptor transcription fac-
and high-carbohydrate/protein consumption to tor that regulates genes involved in beta-oxidation
a high-fat and low-carbohydrate/protein keto- and ketogenesis. Logically, it has been hypoth-
genic diet (KD) necessitates an adjustment in esized that the KD engages PPARalpha in the
the metabolic machinery to handle new primary brain, providing increased ability to use fatty acids
fuel sources. As detailed elsewhere in this vol- to produce ATP and ketone bodies, which may be
ume, there are three prominent biochemical con- critically important for KD-mediated neuropro-
sequences of a KD:  decreased glucose, increased tective and antiseizure effects. Indeed, PPARalpha
free fatty acids, and increased ketone bodies (i.e., agonists exert antiseizure effects in acute seizure
beta-hydroxybutyrate, acetoacetate, and acetone). models (Porta et al., 2009; Puligheddu et al., 2013).
The lower glucose and higher free fatty acids, of However, the KD also exhibits anti-inflammatory
course, directly result from the formulation of the and antioxidant properties and promotes mito-
diet. The ketone bodies are a partial breakdown chondrial health, mechanisms with ever-growing
product from using fatty acids for fuel in a low- empirical support for their importance in con-
glucose environment, and themselves can be used trolling seizures in refractory epilepsy. An alter-
as an efficient, deliverable fuel source. Ketone bod- native nutrient-sensing transcription factor that
ies skip glycolysis and enter the tricarboxylic acid regulates genes in these pathways, as well as genes
cycle at the level of acetyl-CoA. involved in beta-oxidation and ketogenesis, is
In order for fatty acids to be used for fuel PPARgamma. Remarkably, PPARgamma, like the
and for ketogenesis to occur, a series of enzymes KD, is under intense investigation for therapeu-
and proteins must be present and have increased tic potential in Alzheimer’s disease, Parkinson’s
expression. These include, but are not limited to, disease, Huntington’s disease, amyotrophic lateral
beta-oxidation enzymes such as long chain acetyl- sclerosis, multiple sclerosis, stroke, and cancer
CoA synthetase I  (ACS1); carnitine palmitoyl- (reviewed in Heneke and Landreth, 2007; Collino
transferase (CPT), which commits palmitate to et  al., 2008; Mandrekar-Colucci and Landreth,
mitochondrial entry; and the ketogenic enzyme 2011; Dineley et al., 2014; Johri et al., 2014; Carta
mitochondrial 3-hydroxy-3-methylglutaryl-CoA and Simuni, 2015; Corona and Duchen, 2015;
synthase (HMGCS2). For animals that are on a Shen et  al., 2015). In this chapter, we review
ketogenic diet, this mainly occurs in the periphery PPARgamma function and modulation and the
in hepatocytes of the liver, but there is evidence evidence for PPARgamma involvement in the KD
that astrocytes are capable of local beta-oxidation mechanism of action.
and ketogenesis (Auestad et al., 1991; Cullingford
et  al., 2002a; Cullingford et  al., 2002b; Guzmán What Are Peroxisome Proliferator
and Blázquez, 2004;Takahashi et al., 2014). Activated Receptors?
There are two immediate implications from Peroxisome proliferator activated receptors
these findings. One is that dramatic dietary (PPARs) are ligand-inducible transcription factors
changes in fuel sources somehow coordinate the that belong to the superfamily of nuclear recep-
expression of large numbers of genes. The second tors (NR), which comprises 48 transcription fac-
implication is that there is(are) nutrient sensor(s) tors including receptors for endogenous steroid
168

168 section III: Ketogenic Diet in the Laboratory

hormones, thyroid hormone, lipophilic vitamins, isoforms, which underlies the differential biolog-
and cholesterol metabolites (Burris et  al., 2013). ical function of the isoforms. The DNA binding
Functional diversity between the 48 NRs is pro- domain (DBD) contains two highly conserved
vided by distinct preferences for both ligands and zinc fingers. There is a hinge region between the
DNA sequences recognized as response elements DBD and the C-terminal LBD, which is required
within the genome. In most cases the NRs func- for receptor dimerization. The LBD contains the
tion as dimers, either homodimers or heterodi- ligand-dependent transactivation domain AF2
mers, and bind response elements in the absence and is less homologous between the isoforms,
and presence of ligand. The NR binding in the resulting in isoform specificity and sensitivity to
absence of ligand may have passive or active (i.e., ligands (Glass, 1994; Kersten and Wahli, 2000).
constitutive basal transcription or silencing) con- In the nucleus, PPARs form obligate heterodimers
sequences on target gene regulation (Burris et al., with retinoid X receptors (RXR).
2013). As a group, NRs are a significant and impor- Of the three isoforms PPARgamma is the most
tant therapeutic target for human disease, which is extensively studied. Two PPARgamma isoforms,
illustrated by the fact that 10%–15% of US Food PPARgamma1 and PPARgamma2, result from
and Drug Administration-approved drugs target alternative splicing and differential promoter use
NRs (Overington et al., 2006). (Tontonoz et  al., 1994). The PPARgamma iso-
There are three different PPAR isoforms, forms are identical except that PPARgamma2
PPARalpha (also known as NR1C1), PPARbeta contains an additional 28 amino acids at its N-
(also known as delta and NR1C2), and terminus that convey a 5- to 10-fold more effec-
PPARgamma (also known as NR1C3), which are tive ligand-independent transactivation and
encoded by genes located on human chromosomes increased ligand binding affinity to the LBD rel-
22, 6, and 3, respectively (Collino et al., 2008). The ative to PPARgamma1 (Werman et al., 1997; Shao
PPARs are involved in several aspects of devel- et al., 1998; Castillo et al., 1999; Bugge et al., 2009).
opment, such as differentiation of adipose tissue, The expression of PPARgamma1 appears ubiqui-
brain, placenta, and skin; and the three isoforms tous. In contrast, PPARgamma2 is restricted to
display unique tissue- and temporal-dependent adipose tissue; however, high-fat diets can induce
expression patterns during development in cell the expression of PPARgamma2 (Vidal-Puig et al.,
types having ectodermal, mesodermal, and endo- 1996, 1997).
dermal embryonic origins (Desvergne and Wahli, PPARgamma activation is initiated by ligand
1999). Postnatally, the three isoforms exhibit con- binding. The PPARgamma ligand binding pocket
siderable tissue-specific expression. PPARalpha is large (1,300 angstroms3), which allows struc-
is highly expressed in tissues that catabolize fatty tural promiscuity for a wide variety of endogenous
acids (e.g., hepatocytes, cardiomyocytes, entero- or natural agonists (i.e., unsaturated fatty acids,
cytes, and kidney proximal tubule cells). PPARbeta eicosanoids, oxidized lipids, nitroalkenes), syn-
is ubiquitously expressed in all cell types with thetic agonists (e.g., thiazolidinediones or TZDs),
higher levels in proliferating and differentiating and synthetic anatagonists (currently there are
cells. PPARgamma is strongly expressed in adi- no known endogenous antagonists) (Itoh et  al.,
pose tissue and the immune system. All PPARs 2008; Fong et al., 2010; Kroker and Bruning, 2015;
have roles in lipid and glucose metabolism and Sauer, 2015). The nature of PPARgamma’s large
homeostasis, inflammation, cell proliferation and LBD makes it an effective sensor and transducer
differentiation, and vascular biology (Braissant of environmental nutritional and inflammatory
et al., 1996; Tontonoz and Spiegelman, 2008); how- states.
ever, a role in peroxisome proliferation is species- Ligand binding induces a conformational
dependent and it remains unclear whether PPARs change in the receptor leading to dissociation of
are involved in this function in humans (Schrader corepressors such as NCoR (nuclear receptor core-
et al., 2015). pressor 1) and SMRT (silencing mediator of reti-
Members of the nuclear hormone receptor noic acid and thyroid hormone receptor), which
superfamily contain four domains (Figure 20.1A). normally keep basal levels of PPAR-mediated tran-
The N-terminal transactivation domain (activation scription low. Without corepressor complexes, the
function 1, AF1) is involved in ligand-independent PPARgamma-RXR heterodimer is free to bind spe-
transactivation, interaction with transcriptional cific recognition sequences called PPAR-response
coactivators and influences ligand-binding affin- elements (PPREs) in the promoter regions of target
ity of the ligand binding domain (LBD). The AF1 genes. Once corepressors disengage, coactivators
sequence has minimal homology between PPAR are recruited and alter chromatin structure and
169

Chapter 20: Ketogenic Diet and PPARgamma 169

PPARγ1
(a) 477aa
N AF-1 AF-2 C
52kDa
PPARγ2
505aa
N AF-1 AF-2 C
Ligand DNA Hinge Ligand
56kDa
Independent Binding Binding
Activation Domain Domain
Domain

(b)

Agonist
Coactivator Complexes
CBP/p300
SRCs
Corepressor Complexes Corepressor Complexes
PGC-1α
PPARγ RXR PPARγ RXR

Coactivator Complexes

PPARγ RXR
Corepressor Complexes
NCoR
SMRT
PPRE

FIGURE 20.1 Illustrations of PPARgamma splice variants and activity. (A) PPARgamma has two splice variants that
have identical primary sequences except that the N-terminal of PPARgamma 2 has an extension of 28 amino acids.
PPARgamma1 and PPARgamma2 have an activation function 1 domain (AF-1) required for ligand-independent
activation, a DNA-binding domain required for sequence-specific binding to genomic DNA at a peroxisome proliferator
response element (PPRE), a Hinge domain required for receptor dimerization, a ligand-binding domain required for
ligand-dependent modulation and an activation function 2 domain (AF-2) within the ligand-binding domain that is
required for ligand-dependent activation, ligand-dependent dimerization, coactivator recruitment, and corepressor
release. The extra 28 amino acids in the AF-1 of PPARgamma2 confers 5- to 10-fold ligand-independent activation and
greater ligand binding affinity relative to PPARgamma1. (B) PPARgamma forms a heterodimer with the retinoic acid
receptor (RXR) and in the absence of agonist it is bound to corepressor complexes (e.g., NCoR and SMRT). Once an
agonist is bound, the corepressors are released, PPARgamma-RXR recruits coactivators (e.g., CBP/p300, SRCs and PGC-
1alpha), and the complex binds to PPREs in the promoter region of target genes leading to initiation of transcription by
RNA polymerase II and general transcription factors.

recruit transcriptional machinery to promote the homeostasis, insulin-sensitization, fluid homeo-


commencement of transcription (Figure  20.1b). stasis, anti-inflammation, antioxidant, and mito-
Coactivators of PPARgamma include CREB bind- chondrial health. Its best-known role is as a master
ing protein (CBP), SRC1/2/3 (steroid receptor regulator of adipogenesis, where it coordinates
coactivator), and PPARgamma coactivator 1alpha gene expression necessary for adipocyte formation
(PGC-1alpha) (Auwerx, 1999; Katsouri et al., 2012; (Auwerx, 1999; Fong et al., 2010; Ahmadian et al.,
Kroker and Bruning, 2015). The PPARgamma- 2013; Krocker and Bruning, 2015). The insulin-
RXR-corepressor or -coactivator complexes are sensitizing effects of TZDs such as pioglitazone
further regulated by several posttranslational and rosiglitazone are clinically useful in the treat-
modifications such as phosphorylation, acetyla- ment of metabolic disorders such as type II diabe-
tion, SUMOylation, and ubiquitination, which can tes mellitus. However, these full agonists can lead
increase or decrease PPARgamma transcriptional to weight gain, fluid retention, and bone loss due
activity (Ahmadian et al., 2013). to PPARgamma activation in a wide range of tis-
PPARgamma controls hundreds of genes, with sues (Ahmadian et al., 2013). Several strategies to
many involved in lipid and glucose metabolism and reduce side effects are under investigation, ranging
170

170 section III: Ketogenic Diet in the Laboratory

from partial agonism to tissue-specific activation species. In this section, we will discuss the location
(Sujii and Evans, 2011). and function of PPARgamma in the brain and its
Many of the PPARgamma regulated genes potential roles in epilepsy and the KD.
overlap with KD-responsive genes. However,
determining which genes PPARgamma regulates Where Is PPARgamma in the Brain?
at any one time is not straightforward. An addi- PPARgamma is regionally distinct with high
tional level of complexity is that the gene sets regu- expression in neurons, astrocytes, and microglia in
lated by PPARgamma depend on tissue-dependent the cortex and moderate expression in neurons in
differential expression of types of corepressors and the hippocampus (Cullingford et al., 1998; Lu et al.,
coactivators, the state of biochemical cascades 2011; Moreno et al., 2004; Sarruf et al., 2009; Zhao
that regulate post-translational modifications, and et al., 2009). Moreno et al. (2004) have performed
the nature of the bound agonist (full or partial), the most extensive study to date, cataloging the
which determines the degree of conformational expression patterns throughout the brain of not
change and ability of PPARgamma to interact only PPARgamma but also PPARalpha, PPARbeta
with particular corepressors and coactivators and the three RXR isoforms. PPARgamma has
(Ahmadian et  al., 2013; Krocker and Bruning, the most restrictive distribution in the CNS. For
2015; Sauer, 2015). For example, the full agonist example, PPARgamma is absent from neurons in
TZDs exert beneficial insulin-sensitizing effects the olfactory bulb, the perirhinal and entorhinal
as well as detrimental side effects such as weight cortices, the temporal and occipital neocortices,
gain, whereas several partial agonists or so-called the thalamic reticular nucleus, the ventral teg-
selective PPARgamma modulators (SPPARMs) mental area, and the cerebellar Purkinjie cells. It
have been shown to retain antidiabetic effects is highly expressed in neurons of the basal gan-
while eliminating weight gain via attenuated and glia, thalamic rhomboid, centromedial and para-
selective gene-regulatory activity in comparison fascicular nuclei, nuclei of the reticular formation,
with full agonists (Carmona et al, 2007; Tan et al., and cerebellar stellate, basket, and golgi cells.
2012). Additionally, phosphorylation of Ser273 PPARgamma has moderate to weak expression in
in the LBD of PPARgamma dysregulates a group neurons of the hypothalamic nuclei, septohippo-
of distinct genes resulting in insulin-insensitivity. campal nucleus, and the hippocampal formation.
TZDs prevent Ser273 phosphorylation, but their Oligodendrocytes do not express PPARgamma,
effectiveness in producing insulin-sensitization in but it is present in some astrocytes (Moreno et al.,
humans has been found to be inversely related to 2004). These descriptions are in broad agreement
the degree of PPARgamma phosphorylation (Choi with studies using neuron-specific PPARgamma
et  al., 2010). This implies that PPARgamma acti- knockout mice (Sarruf et al., 2009; Lu et al., 2011).
vation may differentially regulate gene sets based Sarruf et  al. (2009) confirmed moderate expres-
on the state of the tissue (e.g., normal brain vs. sion in the hypothalamus but found expression
oxidatively stressed and inflamed epileptic brain). in the ventral tegmental area in contrast to the
This is an intriguing prospect that warrants further previous study. They also reported that neuronal
investigation. loss of PPARgamma resulted in 90% reduction of
PPARgamma mRNA in brain, indicating that most
P PA R G A M M A I N PPARgamma in the brain is expressed in neurons
THE BRAIN (Sarruf et al., 2009). A subsequent study found that
The majority of knowledge concerning in normal mice, PPARgamma mRNA was greatest
PPARgamma has been gained from study of its in cortex, followed by cerebellum, hippocampus,
function in peripheral tissues and diseases as is diencephalon, and hypothalamus (Lu et al., 2011).
evident in the previous sections. In the brain, Knockout of PPARgamma from neurons reduced
much less is known. PPARgamma has differen- mRNA in all CNS regions except cerebellum.
tial regional expression patterns suggesting spe- The reduction was greatest in the hippocampus
cialized and complex roles in both cellular and and least in cortex (Lu et  al., 2011). If the resid-
network function. It seems that PPARgamma has ual mRNA is from expression in glia, than we can
roles in maintaining homeostasis and protection infer that hippocampal glia express minimal levels
against injury. In particular, PPARgamma acti- of PPARgamma.
vation in the brain consistently results in three Only one study has attempted to discern differ-
general effects: prevention of mitochondrial apop- ential expression of the two PPARgamma isoforms
tosis signaling pathways, suppression of inflamma- in the hippocampal CA1 region of mice (Gahring
tory mediators, and limitation of reactive oxygen et  al., 2005). The PPARgamma antibody, which
171

Chapter 20: Ketogenic Diet and PPARgamma 171

recognizes both isoforms, stained essentially all likely contribute to neuroprotection is briefly
cells of the CA1 pyramidal layer and some associ- presented.
ated cells including presumed astrocytes. However, PPARgamma has low expression in the brain
using an antibody that recognized an epitope and is primarily in neurons, however, in in vitro
within the extra 28 N-terminal amino acids of and in vivo models of various neurodegenera-
PPARgamma2, it was found that only a small sub- tive disorders PPARgamma expression increases
set of neurons associated with the pyramidal cell in neurons, astrocytes, and microglia (Kitamura
layer were stained, suggesting that PPARgamma1 et  al., 1999; Diab et  al., 2002; Victor et  al., 2006;
is the primary isoform in the hippocampus. The Fong et al., 2010; Wang et al., 2012). It is hypothe-
PPARgamma2 positive cells lacked astrocytic sized that the inflammatory response and increase
morphology, but did colabel with nicotinic ace- in oxidative stress results in the generation of fatty
tylcholine receptors alpha4 and beta4 subunits; acid metabolites such as eicosanoids, oxidized lip-
thus, the authors concluded that PPARgamma2 ids, and nitroalkenes, which are all potent endog-
was exclusively expressed in putative interneurons enous PPARgamma agonists (Figure  20.2a; Fong
(Gahring et al., 2005). This intriguing finding war- et  al., 2010). In a positive feed-forward loop,
rants confirmation and expansion to other brain PPARgamma increases expression of itself and
regions. coactivators such as PGC-1alpha (Figure  20.2b).
Experimental evidence has proven that this
What Is the Function of PPARgamma endogenous neuroprotective mechanism is impor-
in the Brain? tant in limiting damage, because when it is absent
The role of PPARgamma in the brain during nor- mice experience significantly more brain damage
mal conditions remains largely unknown, but and oxidative stress in response to an insult such
most likely involves minimal activity in order as middle cerebral artery occlusion (Victor et al.,
to maintain lipid and glucose homeostasis. 2006; Zhao et al., 2009).
PPARgamma may also have a role in neuronal The neuroprotective properties of
development and neurogenesis. PPARgamma is PPARgamma activation in all of the above-
highly expressed in mouse embryonic brain and mentioned neurodegenerative disorders have
neural stem cells (NSCs) compared to the rela- consistently been found to involve three general
tively low levels expressed in adult mouse brain areas:  prevention of mitochondrial apoptosis
(Wada et al., 2006), and may have differential roles signaling pathways (Figure  20.2c), suppression
in NSC differentiation during development and of inflammatory mediators (Figure  20.2d), and
adulthood. Activation of PPARgamma in embry- limitation of reactive oxygen species (ROS;
onic neural stem cells promotes NSC proliferation Figure  20.2e). Mitochondrial membrane poten-
and inhibits NSC differentiation. In contrast, the tial depends on several factors, one important
ever-interesting compound cannabidiol (CBD) one being the balanced presence of anti-apoptotic
increased adult rat hippocampal neurogenesis via Bcl-2 and Bcl-xl and pro-apoptotic Bad and Bax.
interaction with a PPARgamma pathway (Espisito Upon injury, Bad and Bax translocate to mito-
et al., 2011). PPARgamma has also been implicated chondria, bind Bcl-2 and Bcl-xl, and depolarize
in promoting neurite outgrowth and dendritic mitochondria. Significant depolarization of the
spine density (Brodbeck et  al., 2008; Quintanilla mitochondrial membrane potential can lead to
et al., 2013). the release of pro-apoptotic factors such as cyto-
PPARgamma’s primary action appears to be chrome C, caspase 9, and caspase 3 (Youle and
neuroprotective in environments that involve Strasser, 2008). PPARgamma activation increases
inflammation and oxidative stress, such as dur- neurotrophic alpha-1 transcription, which leads
ing Alzheimer’s disease, Parkinson’s disease, to activation of Akt signaling pathways, resulting
Huntington’s disease, amyotrophic lateral sclero- in increased Bcl-2 expression and phosphoryla-
sis, multiple sclerosis, and stroke. There are mul- tion of Bad (p-Bad). PPARgamma activation also
tiple reviews detailing PPARgamma effects in each up-regulates 14-3-3epsilon expression, which
of these neurodegenerative disorders, to which sequesters p-Bad and prevents Bad transloca-
the reader is referred (Heneke and Landreth, tion into mitochondria (Wu et  al., 2009a; Wu
2007; Collino et  al., 2008; Mandrekar-Colucci et al., 2009b; Fuenzalida et al., 2007; Thouennon
and Landreth, 2011; Dineley et  al., 2014; Johri et al., 2015).
et al., 2014; Carta and Simuni, 2015; Corona and PPARgamma activation suppresses pro-
Duchen, 2015; Shen et  al., 2015). Here, an over- inflammatory gene expression by inhibiting the
view of PPARgamma’s major effects that most transcription factor NFkappaB. PPARgamma
172

172 section III: Ketogenic Diet in the Laboratory

PUFAs (a)

FA metabolites: Energy Failure:


Eicosanoids ATP
PPARγ Oxidized lipids [Ca2+]i
Nitroalkenes ROS

RXR

Coactivator Complexes
(e)
PPARγ RXR
(c) (d) (c)
SOD/
catalase/ Mito
PPARγ PGC1α 14-3-3ε IκB
GSH/ Biogenesis
PPRE UCP2
(b)

14-3-3ε/P-Bad NFκB ROS ATP

Antioxidant Neuroprotection

Apoptosis Cytokine/
Inflammation

Neuroprotection Anti-Inflammatory

FIGURE 20.2 Schematic illustration depicting hypothetical homeostatic neuroprotection by endogenous PPARgamma


agonists during situations of energy failure experienced during epilepsy, stroke, and other neurodegenerative disorders.
(a)  During extended periods of high oxidative stress, metabolites of fatty acids (FA) are generated that have higher
affinities for PPARgamma than their parent lipids, resulting in an increased activation of PPARgamma. (b) In a positive
feed-forward loop, PPARgamma up-regulates expression of itself and the coactivator PGC-1alpha. Neuroprotection
is achieved through multiple pathways. (c)  PPARgamma decreases apoptosis by increasing the expression of 14-3-
3epsilon, which prevents the translocation of p-Bad into the mitochondria and the release of pro-apoptotic factors
and it increases mitochondrial biogenesis, which provides neurons increased ATP and additional calcium buffering.
(d)  Inflammation is dampened by PPARgamma-mediated inhibition of the pro-inflammatory transcription factor
NFκB and (e) oxidative stress is controlled by up-regulating the expression of antioxidants such as superoxide dismutase
(SOD), catalase, glutathione (GSH), and uncoupling protein 2 (UCP2).

accomplishes this by increasing the expression further neuroinflammation, and the initiation of
of IkappaBalpha and IkappaBbeta, which pre- apoptosis. PPARgamma activation limits ROS by
vent the nuclear translocation and DNA binding increasing the antioxidant genes SOD, catalase,
of NFkappaB (Heneka et al., 2003). PPARgamma glutathione, and uncoupling protein 2 (UCP2)
may also inhibit NFkappaB activity by direct (Chen et  al., 2006; Doonan et  al., 2009; Girnun
physical interaction, steric inhibition, or cofac- et al., 2002; Zhao et al., 2006; Diano et al., 2011).
tor competition (Sauer et  al., 2015). The end As mentioned previously, PPARgamma increases
effect is that PPARgamma activation decreases PGC-1alpha. PGC-1alpha also acts as a coactivator
inflammatory cytokine production (TNF-alpha, of the oxidative stress-induced transcription factor
interleukin-6, INFgamma), iNOS and COX2 Nrf2, which is a master regulator of antioxidant
expression, and astrogliosis and microglial activa- gene sets as well as mitochondrial biogenesis (St.
tion (Combs et al., 2000; Heneka et al., 2000; Lim Pierre et  al., 2006; Milder et  al., 2010; Clark and
et al., 2000 Breidert et al., 2002; Yan et al., 2003; Simon, 2009). Deficiency in PPARgamma is linked
Dehmer et  al., 2004; Sundararajan et  al., 2005; to reduced expression of antioxidants (superoxide
Zhao et al., 2005). dismutase 1, catalase, glutathione S-transferase,
Cellular injury and inflammation can increase uncoupling protein-1, UCP1), lipid metabolism
the production of ROS, which further causes cel- enzymes (lipoprotein lipase), and the transcription
lular damage by oxidizing proteins, lipids, and factor liver X receptor-alpha (Victor et  al., 2006;
DNA. This results in altered neuronal signaling, Zhao et al., 2009).
173

Chapter 20: Ketogenic Diet and PPARgamma 173

K E TO G E N I C D I E T et al., 1998; Wenzel et al., 2007; Fenoglio-Simeone


R E G U L AT I O N et al., 2009a; Fenoglio-Simeone et al., 2009b;
O F   P PA R G A M M A Glasscock et al., 2010; Simeone et al., 2013;
Simeone et al., 2014a; Simeone et al., 2014b;
PPARgamma in Epilepsy
Kim et al., 2015; Simeone et al., 2016). Similar to
PPARgamma was first cloned from Xenopus lae- previous reports, we found that PPARgamma1
vis and mouse liver in the early 1990s (Dreyer predominated in wild-type brain. In contrast,
et al., 1992; Zhu et al., 1993; Elbrecht et al., 1996). in epileptic Kcna1-null brain we found that
Since then, thousands of studies have addressed PPARgamma2 was the dominant form. The total
PPARgamma’s structure/function, regulation, nuclear PPARgamma did not change, but the
pharmacology, expression, and importance in dis- PPARgamma2/gamma1 ratio increased three-
ease primarily in peripheral organs. At the turn fold in Kcna1-null brains (Simeone et al., 2016).
of the 21st century, neuroscientists began inves- Further studies are needed to determine the cause
tigating PPARgamma’s therapeutic potential for of this flip in isoform nuclear content. Potential
neurodegenerative diseases, in which a role for causes that preferentially enhance nuclear trans-
neuroinflammation and oxidative stress in disease location of PPARgamma2 over PPARgamma1
processes has long been recognized (Kitamura may be generation of isoform specific ligands,
et al., 1999; Combs et al., 2000) and has been inves- altered expression of isoform-specific cofac-
tigated in hundreds of publications. tors, or post-translational modifications. Altern-
Interest in the epilepsy community has had atively, transcription, translation, or splicing of
a late and slow start. The first research study PPARgamma2 could be increased in epilepsy.
appeared in 2006, and the total count to date is Recently, it was found that at the initial stage of
a little over a dozen (Okada et al., 2006; Luna- adipogenesis another nuclear receptor, gluco-
Medina et al., 2007; Maurois et al., 2008; Yu et corticoid receptor (GR), is transiently recruited
al., 2008; Hong et al., 2008, 2011, 2013; Abdallah, along with the transcription factor C/EBPbeta to
2010; Han et al., 2011; Jeong et al., 2011; Adabi a complex consisting of PBP/MED1/TRAP220
Mohazab et al., 2012; Chuang et al., 2012; Hughes and p300 to enhancer regions of the Pparg2 iso-
et al., 2014; Boes et al., 2015; Simeone et al., 2016). form. In response to glucocorticoids, this results
These studies span acute seizure models, post– in a transient increase in H3K9 acetylation
status epilepticus (SE) models, and kindling and and enhances the induction of PPARgamma2,
genetic models of chronic epilepsy and, for the which becomes the principal driver of adipo-
most part, consistently support beneficial neuro- genesis (Steger et al., 2010). Intriguingly both
protective and antiseizure effects of PPARgamma C/EBPbeta and glucocorticoids increase in the
agonists in epilepsy. brain with seizures (Lu et al., 2013; Engel et al.,
Comparable to findings in chronic neurode- 2013; Maguire and Salpekar, 2013). PGC-1alpha
generative disorders and acute stroke models, the activation of PPARgamma also enhances inter-
expression of brain PPARgamma increases sub- actions with p300/CBP (Puigserver et al., 1999).
sequent to SE induced by lithium-pilocarpine, Whether PPARgamma2 regulates distinct gene
unilateral intrahippocampal kainic acid, intra- sets is unclear, but it has been shown to up-reg-
peritoneal injection of kainic acid, and electri- ulate catalase expression to a greater degree than
cally induced self-sustaining SE (Yu et al., 2008; PPARgamma1 and is important in providing pro-
Hong et al., 2008; Jeong et al., 2011; Chuang et tection against lipotoxicity (Medina-Gomez et
al., 2012; Boes et al., 2015). Concomitantly, the al., 2007a; Medina-Gomez et al., 2007b; Yakunin
products of the PPARgamma-regulated genes et al., 2014).
Pgc-1alpha and Ucp2 are increased after lith- Assuming that the seizure/injury-induced
ium-pilocarpine and intrahippocampal kainic changes in PPARgamma expression are part of
acid (Han et al., 2011; Chuang et al., 2012). In a an endogenous neuroprotective mechanism that
recent study, we examined the nuclear content of limits damage, similar to what has been proposed
both splice variants, PPARgamma1 and gamma2, for other neurodegenerative disorders, then loss of
in the brains of wild-type mice and epileptic lit- PPARgamma should exacerbate markers of injury
termates that lack the Kv1.1 potassium channel and possibly seizures. Chuang et al. (2012) found
(Kcna1-null). Kcna1-null mice develop severe that pretreatment with bilateral focal injections of
spontaneous recurrent seizures (SRS) and are the PPARgamma antagonist GW9662 (150 nl of a 12
a model of temporal lobe epilepsy and sudden mM solution or 1.5-2 mg/kg) reduces UCP2; exac-
unexpected death in epilepsy (SUDEP) (Smart erbates intrahippocampal kainic acid SE-induced
174

174 section III: Ketogenic Diet in the Laboratory

increases in ROS, oxidized proteins, mito- PPARgamma Agonist Pretreatment Raises


chondrial Bax, cytosolic cytochrome C, and Thresholds in Acute Seizure Models
DNA fragmentation; and decreases mitochon- Abdallah (2010) and Adabi Mohazab et al. (2012)
drial respiratory complex I (MRCI) activ- utilized penelenyltetrazole (PTZ) to induce sei-
ity. Unfortunately, the authors failed to report zures in mice with two different paradigms.
the details of SE, so it is not known whether Abdallah (2010) pretreated mice with pioglitazone
PPARgamma antagonism worsened the SE (10 mg/kg; p.o.) 30 minutes prior to an intraperi-
(Chuang et al., 2012). We administered GW9662 toneal (i.p.) injection of PTZ and measured latency
(1 mg/kg/day in the drinking water for 2 weeks) to stage 4/5 clonic-tonic seizures. All vehicle con-
to Kcna1-null mice and wild-type littermates, trols reached stage 4/5, whereas only 20% of the
however, contrary to expectations, inhibiting pioglitazone group reached stage 4/5 and those
PPARgamma did not increase SRS in Kcna1- that did took twice as long as the control group.
null mice, nor did it lower seizure threshold in Adabi Mohazab et al. (2012) injected PTZ intrave-
wild-type mice (Simeone et al., 2016). A possi- nously and measured the threshold dose required
ble explanation for the lack of effect on Kcna1- to induce a whole-body clonic seizure. They found
null seizures may be that the dose of GW9662 that a 1-h pretreatment with pioglitazone was inef-
was too low. Supporting this hypothesis, we fective, whereas a 4-h pretreatment provided a
found that GW9662 had no effect on the nuclear dose-dependent (10–80 mg/kg; p.o.) increase in
PPARgamma2/gamma1 ratio in nonepileptic the seizure threshold dose. Maurois et  al. (2008)
wild-type brain. It did reduce the PPARgamma2/ also found that the PPARgamma agonist FMOC-
gamma1 ratio in epileptic Kcna1-null brain, but L-leucine ((N-[9-fluorenylmethoxycarbonyl]-)-
only from threefold to twofold of wild-type ratio L-leucine; up to 100 mg/kg; i.p.) was not effective
(Simeone et al., 2016). A greater reduction may in the 6-Hz seizure test with 1-h pretreatment. We
be needed to observe an effect on SRS. We also have found similar effects exposing mice to fluro-
found that seizure thresholds of Pparg2-null thyl gas and measuring latency to clonus and gen-
mice and neuron specific-PPARgamma knockout eralized tonic-clonic seizures. A 1-h pretreatment
mice were no different than control littermates with pioglitazone had inconsistent effects, whereas
(Simeone et al., 2016). From these experimen- a 4-h pretreatment dose-dependently (1–80 mg/
tal results we can draw the tentative conclusions kg; i.p.) increased the latency to generalized tonic-
that PPARgamma itself is somehow involved in clonic seizures but had no effect on clonic seizures
increasing nuclear PPARgamma2 and that the (Simeone et al., unpublished observations).
seizure-induced increase affords some neuropro-
tection against injury, but may not raise the sei- PPARgamma Agonists Raise Thresholds
zure threshold per se. in Seizure Susceptible and Kindling Models
The EL mice develop susceptibility to stress-
PPARgamma Agonism in Epilepsy induced tonic-clonic seizures beginning 12 weeks
The PPARgamma agonists pioglitazone and rosi- postnatally. Providing pioglitazone (20 mg/kg/
glitazone have been clinically useful in the man- day) from 5 to 11 weeks postnatally delayed the
agement of type II diabetes mellitus since 1999, age of onset and duration of stress-induced sei-
but as of today there are no published reports, zures (Okada et  al., 2006). Maurois et  al. (2008)
case reports or otherwise, describing epilepsy used a dietary-induced magnesium deficiency-
patients with comorbid type II diabetes that have dependent audiogenic seizure model. Mice were
been prescribed pioglitazone or rosiglitazone. pretreated with either FOMC-L-leucine or rosigli-
Nevertheless, experimental evidence from ani- tazone (up to 100 mg/kg; i.p.) 1 h prior to acous-
mal studies supports that PPARgamma agonists tic stimuli. FOMC-L-leucine dose-dependently
will have antiseizure, neuroprotective, and pos- reduced the number of mice responding to acous-
sibly disease-modifying effects in epilepsy. Even tic stimuli by 50%, whereas rosiglitazone was inef-
though this field of research is small and there fective. The FOMC-L-leucine effects were blocked
are relatively few studies, there is wide diver- by coadministration of the PPARgamma antago-
sity in treatment protocol (agonist, dose, pre/ nist GW9662 (1 and 2 mg/kg, i.p.) (Maurois et al.,
post-treatment) and endpoints (acute seizures, 2008). Finally, Abdallah (2010) kindled mice with
SRS, inflammatory markers, cell death, cogni- daily i.p. injections of subconvulsive PTZ (40 mg/
tive comorbidities), and it is worth detailing the kg) until the mice exhibited stage 4/5 clonic-tonic
similarities and differences to gain a sense of the seizures on three consecutive days. Pretreatment
robustness of PPARgamma effects. with pioglitazone (10 mg/kg; p.o.) 30 minutes prior
175

Chapter 20: Ketogenic Diet and PPARgamma 175

to each PTZ injection resulted in only 30% of mice (10 mg/kg; i.p.) for 5 days decreases SRS and sei-
reaching stage 4/5 seizures by day 17 (Abdullah, zure severity in epileptic Kcna1-null mice by 75%
2010). However, because pioglitazone was admin- (Simeone et al., 2016).
istered before PTZ and most pioglitazone-treated
mice did become kindled as defined by the author, PPARgamma Agonists Are Neuroprotective
it is difficult to determine whether pioglitazone post-SE and during SRS
exerted any antiepileptogenic effect in addition to Within hours to days, intrahippocampal kainic
its anticonvulsant effect. acid-induced SE results in increases in UCP2, ROS,
oxidized proteins, mitochondrial Bax, cytosolic
PPARgamma Agonists Reduce SRS cytochrome c, DNA fragmentation, and decreases
and Improve Spatial Learning in MRCI activity in rat hippocampi (Chuang et al.,
Multiple studies have used SE models to later 2012). Pretreatment with bilateral focal injections
investigate neuropathology during the latent of rosiglitazone (150 nL of a 4 mM solution or
period and after development of SRS (discussed 0.7–0.8 mg/kg) further increased UCP2 and pre-
in the next section), however, only three studies vented all other changes. In fact, MRCI activity
have provided information concerning the effect was actually improved compared with the sham
of PPARgamma agonists on either seizures during controls (Chuang et al., 2012). In a similar study,
SE and/or the development of SRS (Luna-Medina Luna-Medina et  al. (2007) found that intrahip-
et al., 2007; Hong et al., 2013; Boes et al., 2015). pocampal injection of kainic acid results in sig-
Luna-Medina et al. (2007) pretreated rats with a nificant cerebral edema, hippocampal cell loss,
thiadiazolidine compound, NP031112 (50 mg/kg; astrogliosis, and the induction of TNFalpha in
p.o.), 1 hour before kainic acid injection (10 mg/ astrocytes, microglia, and neurons. Coinjection of
kg; i.p.) and monitored the development of behav- NP031112 (5–8 ng/kg) prevented edema, cell loss,
ioral seizures and SE. NP031112 had no effect on and TNFalpha induction and reduced astrogliosis.
SE. Similarly, Hong et al. (2013) found that rosi- Furthermore, pretreatment with NP031112 (50
glitazone had no effect on the development of mg/kg; p.o.), 1 hour before kainic acid injection
lithium-pilocarpine SE. Rats were pretreated with (10 mg/kg; i.p.) also prevented hippocampal neu-
rosiglitazone (0.1 mg/kg; i.c.v.) 1 hour before lith- ronal loss as quantified 72 hours post-SE (Luna-
ium chloride injection (127 mg/kg; i.p.). Twenty- Medina et al., 2007).
four hours later rats were again pretreated 1 hour In the rat lithium-pilocarpine SE model,
prior to pilocarpine hydrochloride (30 mg/kg; i.p.) post-SE daily administration of rosiglitazone
administration. Rats then received repeated injec- has antioxidant and anti-inflammatory effects.
tions of pilocarpine (10 mg/kg; i.p.) every 30 min Rosiglitazone increases hippocampal neuronal
until they developed convulsive seizures, which survival, decreases ROS and lipid peroxidation,
were scored behaviorally according to the Racine increases SOD and GSH, and decreases microglia
scale. Rosiglitazone had no effect on SE. The and astrocytic activation, TNF-alpha, and BDNF
authors continued daily administration of rosigli- (Yu et  al., 2008; Hong et  al., 2008, 2012, 2013).
tazone and examined development of SRS by per- Similarly in EL mice and the PTZ-kindling model,
forming video-EEG recordings beginning 2 weeks pioglitazone decreased TNF-alpha, IL-1beta, IL-6,
post-SE for 6 weeks. They found that 50% of rosi- IL-10, and caspase3 (Okada et al., 2006; Abdallah,
glitazone-treated rats developed SRS compared 2010). However, rosiglitazone did not reduce hip-
with 83% of vehicle-treated rats. Moreover, rosigli- pocampal cell death in the self-sustained SE model
tazone nearly doubled the latency to SRS develop- (Boes et al., 2015).
ment and decreased the frequency and duration
of SRS (Hong et al., 2013). An earlier study by Evidence for Ketogenic Diet Activation
this group demonstrated that the same protocol of PPARgamma
improved the SE rats’ spatial learning as assessed Although the possibility of a role for PPARgamma
in the Morris water maze (Hong et al., 2011). In in the mechanism of the KD has been briefly
contrast, rosiglitazone (3 mg/kg; i.p.) administered speculated in review articles for the past several
to rats daily immediately following the end of self- years (Kobow et al., 2012; Masino and Rho, 2012;
sustained SE had no effect on the development Gano et al., 2014), there have only been two stud-
and occurrence of SRS; however, the rosiglitazone- ies published exploring this possibility, one using
treated rats did have improved spatial learning in an acute SE model and one using a chronic epi-
the Morris water maze (Boes et al., 2015). Finally, lepsy model and acute seizure threshold model
we have found that administration of pioglitazone (Jeong et al., 2011; Simeone et al., 2016). Jeong
176

176 section III: Ketogenic Diet in the Laboratory

et al. (2011) fed wild-type mice a KD (4:1 ratio in the drinking water for the 2-week KD treatment
of fats to carbohydrate+protein) for 3 weeks and prevented the increase in nuclear PPARgamma2
found a threefold increase of PPARgamma pro- and prevented KD-mediated seizure reduction in
tein in cell lysate from hippocampal tissue com- Kcna1-null mice. To further test the importance of
pared with standard diet (SD)-fed mice. The PPARgamma in the KD mechanism, we obtained
mice were injected with kainic acid (30 mg/kg) to Pparg2-null mice and conditional neuron-specific
induce status and tissue collected at 2 and 6 hours PPARgamma knockout mice. Previous studies
post–kainic acid injection. The KD delayed the have demonstrated that KD treatment raises the
onset of generalized tonic-clonic seizures, but the threshold for flurothyl seizures (Rho et al., 1999;
overall seizure burden that the mice experienced Dutton et al., 2011). Similarly, we found that KD-
during the 2 and 6 hours post–kainic acid was not treatment effectively raised seizure thresholds of
reported. In the SD-fed mice, there was a fourfold control mice. However, KD-treatment was unable
and twofold increase of PPARgamma at the 2- and to raise the seizure threshold of Pparg2-null mice
6-hour time points after injection, respectively. In and conditional neuron-specific PPARgamma
contrast, the KD-induced elevated PPARgamma knockout mice. Furthermore, the pharmaco-
levels fell to normal diet control levels by 6 hours logic and genetic manipulations of PPARgamma
post–kainic acid. Hippocampal TNF-alpha fol- expression/function did not affect the stereo-
lowed the same pattern as PPARgamma, whereas typic KD increase of blood beta-hydroxybutyr-
the KD blunted the SE-induced increases in NF- ate or decrease of glucose (Simeone et al., 2016).
kappaB, microglial CD11b, and COX2 expres- Results from this study strongly support that cen-
sion. From the studies mentioned previously tral actions of PPARgamma, more specifically
detailing the effect of injury or degeneration PPARgamma2, play a critical role in the antisei-
and PPARgamma agonists on the expression of zure mechanism of the KD.
PPARgamma, one would expect that PPARgamma
would be further increased, not decreased, by SE
in the KD-fed mice. The authors do not address What Does the KD provide That Could Be
this discrepancy (Jeong et al., 2011). Nevertheless, Activating PPARgamma?
the authors do present evidence that the KD does The selective increase of nuclear PPARgamma2
have effects on PPARgamma and downstream over PPARgamma1 may be due to the additional
markers of inflammation known to be regulated 30 amino acids in the PPARgamma2 AF1 that
by PPARgamma. convey a 5- to 10-fold more effective ligand-
In a recent study, we aimed to determine independent transactivation and increased
whether the KD changes PPARgamma expres- ligand binding affinity to the LBD relative to
sion in the brains of epileptic and normal mice, PPARgamma1 (Werman et  al., 1997; Shao et  al.,
and, if so, whether it is necessary for the antisei- 1998; Castillo et  al., 1999; Bugge et  al., 2009).
zure effects of the KD. As described earlier, we PPARgamma2 is the only PPARgamma isoform
have found that in nuclear extracts from wild- regulated at the transcriptional level by nutrition
type mouse brain, PPARgamma1 is the domi- (Medina-Gomez et al., 2007b). The PPARgamma2
nant isoform, whereas PPARgamma2 is dominant expanded AF1 also confers differential interac-
in epileptic Kcna1-null brain (Simeone et al., tion with transcriptional cofactors and post-
2016). Treating Kcna1-null mice for 2 weeks with translational modifications that would most
a KD (6:1 ratio) reduced seizure incidence and likely result in tissue-specific differences in the
severity by ~75%, as we have reported previ- regulation of gene sets by the PPARgamma splice
ously (Fenoglio-Simeone et al., 2009b; Kim et al. variants. This is evident in the periphery, where
2015; Simeone et al., 2016). Ketogenic diet treat- PPARgamma2, but not PPARgamma1, is induced
ment increased PPARgamma2 in both genotypes, during high-fat diets and initiates adipogenesis,
resulting in PPARgamma2/gamma1 ratios that increases lipid-buffering, and reduces lipotoxicity
were twofold and sixfold for wild-type and Kcna1- (Medina-Gomez et al., 2007a). Therefore, the most
null brain, respectively, compared with SD-fed likely mechanisms for this isoform-specificity of
wild-type mice. the KD probably involve either regulation of sig-
To determine whether the effect of the KD naling cascades responsible for post-translational
on PPARgamma was necessary for KD anti- modifications that contribute to selective nuclear
seizure efficacy, we performed pharmacologic translocation or transcription of PPARgamma2
and genetic experiments. Coadministering the and/or providing a PPARgamma ligand, possibly
PPARgamma antagonist GW9662 (1 mg/kg/day) selective for PPARgamma2.
177

Chapter 20: Ketogenic Diet and PPARgamma 177

The KD provides plenty of fat, of course, and in oxidative phosphorylation (e.g., multiple sub-
unsaturated fatty acids, such as omega-3 and units of complexes I, II, IV, and V); induction
omega-6 long chain polyunsaturated fatty acids, of mitochondrial biogenesis; and up-regulation
are notably increased in blood serum of patients of UCP2, catalase, and glutathione (Masino and
(Fraser et al., 2003). Importantly, long chain pol- Rho, 2012; Mandrekar-Colucci et al., 2013; Fong
yunsaturated fatty acids and their metabolites, et  al., 2010; Bernardo et  al., 2006; Heneka and
eicosanoids, oxidized lipids, and nitroalkenes are Landreth, 2007; Chuang et al., 2012; Hong et al.,
all natural ligands for PPARgamma (Yamamoto 2008, 2012, 2013; Abdallah, 2010; Adabi Mohazab
et al., 2005; Fong et al., 2010). Also, it was recently et al., 2012; Bough et al., 2006; Miglio et al., 2009;
determined that the saturated fatty acid decanoic Sullivan et  al., 2004; Yang and Cheng, 2010; Yu
acid (a.k.a. capric acid), a primary constituent of et al., 2008). All of these have been suggested as
the medium chain triglyceride ketogenic diet, is a possible disease modifying targets for epilepsy.
ligand for PPARgamma at physiologically relevant Mitochondria are of particular interest in tempo-
concentrations (Malapaka et al., 2012). ral lobe epilepsy. In human epilepsies and animal
In vivo treatment suggests that decanoic acid chemoconvulsant seizure models, brain tissue
is a selective PPARgamma modulator (i.e., par- from seizure foci exhibit mitochondrial reac-
tial agonist) as it improves glucose sensitivity tive oxygen species (ROS) and oxidative damage,
and lipid profiles without weight gain in diabetic reduced ATP-producing complex I  activity and
mice (Malapaka et  al., 2012). In vitro experi- diminished antioxidant systems (Sullivan et  al.,
ments in hippocampal slices found that decanoic 2003; Wallace, 1999; Bruce and Baudry, 1995;
acid decreases PTZ-induced epileptiform activ- DiMauro et  al., 1999; Kunz et  al., 2000; Mueller
ity in a concentration-dependent manner (Chang et al., 2001; Sudha et al., 2001; Malthankar-Phatak
et  al., 2013). Furthermore, decanoic acid-induced et  al., 2006; Vielhaber et  al., 2008; Kudin et  al.,
increases in citrate synthase, catalase, and MRCI 2009; Ryan et al., 2012; Simeone et al., 2014a; Kim
activity in SH-SY5Y neuronal cultures were inhib- et al., 2015; Simeone et al., unpublished observa-
ited by a PPARgamma antagonist (Hughes et  al., tion). Oxidative stress and mitochondrial over-
2014). Alternatively, Jeong et al. (2012) found that load likely contribute to the neuronal cell loss
treatment of cultured HT22 cells (a hippocampal in severe epilepsy associated with sclerosis. In
neuronal cell line) with the ketone body acetoace- this circumstance, it is clear that the proposed
tate (5 mM) increases PPARgamma over a 12-hour actions of PPARgamma would be neuroprotec-
period; however, we have been unable to replicate tive and increase the number of surviving cells.
this finding in primary hippocampal neuronal cul- But this illustrates only one outcome of unhealthy
tures, and in fact this concentration of acetoacetate mitochondria and does not address the potential
results in significant neuronal death within 24 and consequences that chronic mitochondrial dys-
48 hours (Simeone et  al., unpublished observa- function has on neuronal hyperexcitability and
tions). Therefore, at the moment it seems that the seizure severity.
unsaturated and saturated fatty acids provided by a Several elegant studies have demonstrated that
ketogenic diet may be the ligands for PPARgamma, mitochondria regulate synaptic transmission via
and that PPARgamma may be involved in the anti- three properties: (1) production of ATP and (2) ROS
seizure mechanism of the various formulations and (3)  sequestration of cytosolic calcium (Lee
of the ketogenic diet regardless of the type of fat et al., 2007; Lee et al., 2012; Harris et al., 2012). Thus,
content. any perturbation of mitochondrial health will send
ripples of dysregulation across synaptic, neuronal,
FUNCTIONAL and network activity. Stabilizing synaptic mito-
CONSEQUENCES chondria could be another mechanism by which
O F   P PA R G A M M A PPARgamma and the KD reduce seizure activity.
A C T I VAT I O N R E L E VA N T To test this hypothesis, we turned once again to
TO   E P I L E P S Y the Kcna1-null mouse model of epilepsy, because
PPARgamma agonists and the KD regulate simi- we have found mitochondrial pathology similar
lar anti-inflammatory, antioxidant and promito- to reports from human epilepsies (Simeone et al.,
chondrial pathways. These include, but are not 2014a; Kim et al., 2015; Simeone et al., unpublished
limited to, up-regulation of IkappaB; inhibition observations). Mitochondria isolated from cortical
of NFkappaB; reduction of cytokines such as and hippocampal tissue from epileptic Kcna1-null
IL-1beta, IL-6, and TNF-alpha; up-regulation of mice have reduced ATP-producing MRCI-driven
genes encoding mitochondrial enzymes involved respiration, increased ROS, and decreased UCP2
178

178 section III: Ketogenic Diet in the Laboratory

(Simeone et al., 2014a). The decrease of UCP2, a rotenone exacerbated provoked seizure-like events
mitochondrial protein with downstream antiox- in vitro, supporting a possible role in worsening in
idant effects, may contribute to the rise in ROS. vivo seizures (Simeone et al., 2014a).
The MRCI inhibition appears to be due to post- We have further demonstrated that in vivo
translational inhibition by elevated ROS because treatment of Kcna1-null mice with either pio-
acute addition of antioxidants such as ascorbic glitazone or a KD or a cocktail of ascorbic acid,
acid restores Kcna1-null MRCI-driven respiration alpha-tocopherol, and pyruvate designed to tar-
to wild-type levels, whereas exogenous H2O2 mim- get mitochondrial health rescues MRCI respira-
ics the MRCI dysfunction of Kcna1-null mito- tion, restores mossy fiber neurotransmitter release
chondria (Simeone et al., 2014a). In addition, the probabilities, and dampens network hyperexcit-
mitochondrial membrane potential is depolarized ability (i.e., reduces incidence of SPWs, ripples,
and mitochondrial calcium sequestration capacity and fast ripples) (Simeone et  al., 2014a; Simeone
is reduced (Simeone et al., 2014a; Kim et al., 2015; et al., 2014b; Simeone et al., unpublished observa-
Simeone et al., unpublished observation). tions). Therefore, we speculate that in Kcna1-null
Thus, the three properties of mitochondria mice one important antiseizure mechanism result-
that play a role in synaptic transmission (ATP, ing from KD modulation of PPARgamma is res-
ROS, and calcium) are dysfunctional in Kcna1- toration of MRCI function, possibly by increasing
null mitochondria and predict widespread synap- endogenous antioxidant pathways and decreasing
tic and network activity. This is exactly what we ROS. Recently, Chuang et  al. (2012) implicated
observed using a multielectrode array to record just such a pathway involved in the neuroprotec-
extracellular potentials from in vitro hippocam- tive effects of rosiglitazone in the intrahippocam-
pal slices from Kcna1-null brains (Simeone et al., pal kainate model of status epilepticus. Specifically,
2013). We found that Kcna1-null hippocampi rosiglitazone increased UCP2, decreased ROS, and
generate spontaneous network activity originat- restored MRCI function.
ing in the CA3 region in the form of sharp waves
(SPWs) and ripples (80–200 Hz bandwidth) with CONCLUDING REMARKS
an increased incidence and duration compared The development of epilepsy necessarily originates
with slices from wild-type hippocampi. Also with a precipitating event (e.g., genetic predisposi-
present in Kcna1-null hippocampi were epilepsy- tion, traumatic brain injury, virus, etc.) that lowers
associated pathologic high-frequency oscillations the seizure threshold or moves the baseline activ-
in the fast ripple bandwidth (200–600 Hz), which ity of the brain closer to the seizure threshold so
can be viewed as biomarkers of hyperexcitable that previously nonsignificant internal or external
networks. Furthermore, Kcna1-null CA3 has stimuli are capable of synchronizing large net-
enhanced coupling of excitatory inputs and popu- works into high-frequency oscillatory activity. If
lation spike generation and CA3 principal cells the resulting seizure phenotype is severe, than a
have reduced spike-timing reliability. Removing chronic inflammatory and oxidative state develops
the influence of granule cell mossy fiber path with concomitant mitochondrial dysfunction. The
inputs by microdissecting the Kcna1-null CA3 result is not only death of vulnerable cell types but
region mostly rescued the network oscillatory also dysregulation of cellular, synaptic, and net-
behavior and improved spike timing. We found work excitability, which further lowers the seizure
that Kcna1-null mossy fibers are hyperexcit- threshold and exacerbates the seizure phenotype
able and reduced paired pulse ratios, suggesting (Figure 20.3, left).
increased neurotransmitter release at these termi- Experimental evidence indicates that the KD
nals (Simeone et al., 2013). Collectively, these data prevents cell death and hyperexcitability by promot-
indicate enhanced synaptic release in the Kcna1- ing mitochondrial health and anti-inflammatory
null CA3 region reduces spike timing precision of and antioxidant pathways. Limited, but strong,
individual neurons leading to disorganization of evidence that we have reviewed here suggests that
network oscillatory activity and promotion of the the KD regulates PPARgamma to achieve these
emergence of fast ripples. effects (Figure 20.3, right). This discovery presents
This synaptic phenotype is mimicked by acute multiple opportunities for both researchers and
application of the MRCI inhibitor rotenone to clinicians. The immediate translational poten-
wild-type hippocampal slices. Experimental inhi- tial resides in the availability of the PPARgamma
bition of MRCI results in increased SPWs, ripples, agonists pioglitazone and rosiglitazone that are
and fast ripples (Simeone et  al., 2014a; Simeone FDA-approved for therapeutic use in type II dia-
et  al., unpublished observations). Moreover, betes mellitus. The TZDs are not perfect and can
179

Chapter 20: Ketogenic Diet and PPARgamma 179

Diet
Precipitating Event
ic
Ketogen
(e.g., genetics, TBI, stroke, virus)

ATP
FFA
[Ca++]i
ROS
SFAs
PUFAs +

MITO Neuronal, Synaptic, Network

PPARγ2
FUNCTION HYPEREXCITABILITY

nction
ndrial Fu
[Ca++]i
Mitocho ROS

SEIZURES
ant
Antioxid

n
ammatio
Anti-Infl
CELL INFLAMMATION
DEATH (e.g., TNF-α)

FIGURE 20.3 Schematic illustration of the proposed mechanisms of action of PPARgamma2 activation underlying
the antiseizure efficacy of the ketogenic diet. (Right) A depiction of the interactions of oxidative stress, inflammation,
and mitochondrial dysfunction that may participate in worsening seizure severity and genesis. Seizure genesis originates
from a precipitating event such as a genetic predisposition, injury, stroke, or virus that results in neuronal, synaptic,
and network hyperexcitability. The ictal events lead to increases in reactive oxygen species (ROS) production and
intracellular calcium, which can overload cellular buffering capacities. Seizures can also initiate the release of pro-
inflammatory cytokines that can further increase ROS and calcium. The ROS and excess calcium cause dysregulation of
mitochondrial function most notably at complex I, resulting in decreased ATP generation, increased ROS production,
mitochondrial membrane depolarization, and further increase in cytosolic calcium as mitochondria lose their capacity
to sequester calcium. Extreme mitochondrial damage will lead to release of pro-apoptotic factors and cell death, which
will increase inflammatory processes. Chronic mitochondrial dysfunction in neurons will dysregulate neuronal,
synaptic, and network excitability and exacerbate the precipitating event-induced hyperexcitability. (Left) The ketogenic
diet increases free fatty acids. Whether this is an increase in polyunsaturated fatty acids such as docahexaenoic acid and
eicosapentaenoic acid or medium chain saturated fatty acids such as decanoic acid depends on the formulation of the
diet. Nevertheless, this increase in fatty acids represents increases in natural agonists for the nuclear transcription factor
PPARgamma. In particular, enrichment of nuclear PPARgamma2 is important for ketogenic diet antiseizure efficacy.
Upregulation of gene sets involved in promoting mitochondrial function, antioxidation, and anti-inflammation may
contribute to not only the KD’s neuroprotective properties but also the KD’s ability to dampen hyperexcitabilty.

have significant side effects. The good news is that The identification of PPARgamma as a poten-
the diabetes academic and industry communi- tial mediator of KD efficacy in epilepsy does not
ties have active research efforts into developing preclude the other mechanisms discussed in this
PPARgamma partial agonists (SPPARMs) with volume. Some may work synergistically with
improved side-effect profiles. These may also prove PPARgamma, such as ketone bodies, and others
useful in epilepsy. Furthermore, additional tar- may be downstream effectors of PPARgamma,
gets with therapeutic potential in epilepsy may be such as adenosine.
identified by focusing on the upstream pathways
or ligands leading to PPARgamma activation and AC K N OW L E D G M E N T
the downstream effectors that result in increasing This work was supported by an award from Citizens
the seizure threshold and/or dampening hyperex- United for Research in Epilepsy Foundation
citability and neuroprotection. and the NIH (NS085389). I  thank Dr.  Kristina
180

180 section III: Ketogenic Diet in the Laboratory

A.  Simeone for many insightful discussions and Bruce, A.J., Baudry, M. (1995). Oxygen free radicals
comments on this manuscript. in rat limbic structures after kainate-induced sei-
zures. Free Radic Biol Med 18, 993–1002.
Bugge, A., Grontved, L., Aagaard, M.M., Borup, R.,
REFERENCES and Mandrup, S. (2009). The PPARgamma2 A/
Abdallah, D.M. (2010). Anticonvulsant potential B-domain plays a gene-specific role in transactiva-
of the peroxisome proliferator-activated recep- tion and cofactor recruitment. Mol Endocrinol 23,
tor γ agonist pioglitazone in pentylenetetrazole- 794–808.
induced acute seizures and kindling in mice. Brain Burris, T.P., Solt, L.A., Wang, Y, Crumbley, C.,
Res 1351, 246–253. Banerjee, S., Griffett, K., Lundasen, T., Hughes,
Adabi Mohazab, R., Javadi-Paydar, M., Delfan, B., and T., and Kojetin, D.J. (2013). Nuclear receptors
Dehpour, A.R. (2012). Possible involvement of and their selective pharmacologic modulators.
PPAR-gamma receptor and nitric oxide pathway Pharmacol Rev 65, 710–778.
in the anticonvulsant effect of acute pioglitazone Carmona, M.C., Louche, K., Lefebvre, B., Pilon, A.,
on pentylenetetrazole-induced seizures in mice. Hennuyer, N., Audinot-Bouchez, V., Fievet, C.,
Epilepsy Res 101, 28–35. Torpier, G., Formstecher, P., Renard, P., et  al.;
Ahmadian, M., Suh, J.M., Hah, N., Liddle, C., Atkins, Consortium of the French Ministry of Research
A.R., Downes, M., and Evans, R.M. (2013). PPARγ and Technology. (2007). S 26948:  a new specific
signaling and metabolism: the good, the bad and peroxisome proliferator activated receptor gamma
the future. Nat Med 19, 557–566. modulator with potent antidiabetes and antiath-
Auestad, N., Korsak, R.A., Morrow, J.W., and Edmond, erogenic effects. Diabetes 56, 2797–2808.
J. (1991) Fatty acid oxidation and ketogenesis by Carta, A.R., and Simuni, T. (2015). Thiazolidinediones
astrocytes in primary culture. J Neurochem 56, under preclinical and early clinical development
1376–1386. for the treatment of Parkinson’s disease. Expert
Auwerx, J. (1999). PPARgamma, the ultimate thrifty Opin Investig Drugs 24, 219–227.
gene. Diabetologia 42, 1033–1049. Castillo, G., Brun, R.P., Rosenfield, J.K., Hauser, S., Park,
Bernardo, A., and Minghetti, L. (2006). PPAR- C.W., Troy, A.E., Wright, M.E., and Spiegelman,
gamma agonists as regulators of microglial acti- B.M. (1999). An adipogenic cofactor bound by the
vation and brain inflammation. Curr Pharm Des differentiation domain of PPARgamma. EMBO J
12, 93–109. 18, 3676–3687.
Boes, K., Russmann, V., Ongerth, T., Licko, T., Chang, P., Terbach, N., Plant, N., Chen, P.E., Walker,
Salvamoser, J.D., Siegl, C., Potschka, H. (2015). M.C., and Williams, R.S. (2013). Seizure control
Expression regulation and targeting of the peroxi- by ketogenic diet-associated medium chain fatty
some proliferator-activated receptor γ following acids. Neuropharmacology 69, 105–114.
electrically-induced status epilepticus. Neurosci Chen, S.D., Wu, H.Y., Yang, D.I., Lee, S.Y., Shaw, F.Z.,
Lett 604, 151–156. Lin, T.K., Liou, C.W., and Chuang, Y.C. (2006).
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., Effects of rosiglitazone on global ischemia-induced
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, hippocampal injury and expression of mitochon-
Y., Geiger, J.D., and Dingledine, R.J. (2006). drial uncoupling protein 2. Biochem Biophys Res
Mitochondrial biogenesis in the anticonvulsant Commun 351, 198–203.
mechanism of the ketogenic diet. Ann Neurol 60, Choi, J.H., Banks, A.S., Estall, J.L., Kajimura, S.,
223–235. Boström, P., Laznik, D., Ruas, J.L., Chalmers,
Braissant, O., Foufelle, F., Scotto, C., Dauça, M., M.J., Kamenecka, T.M., Blüher, M., et  al. (2010).
and Wahli, W. (1996). Differential expression Anti-diabetic drugs inhibit obesity-linked phos-
of peroxisome proliferator-activated receptors phorylation of PPARgamma by Cdk5. Nature 466,
(PPARs): tissue distribution of PPAR-alpha, -beta, 451–456.
and -gamma in the adult rat. Endocrinology 137, Chuang, Y.C., Lin, T.K., Huang, H.Y., Chang, W.N., Liou,
354–366. C.W., Chen, S.D., Chang, A.Y.W., and Chan, S.H.H.
Breidert, T., Callebert, J., Heneka, M.T., Landreth, G., (2012). Peroxisome proliferator-activated receptors
Launay, J.M., and Hirsch, E.C. (2002). Protective gamma/mitochondrial uncoupling protein 2 signal-
action of the peroxisome proliferator-activated ing protects against seizure-induced neuronal cell
receptor-gamma agonist pioglitazone in a mouse death in the hippocampus following experimental
model of Parkinson’s disease, J Neurochem 82, status epilepticus. J Neuroinflammation 9, 184.
615–624. Clark, J., and Simon, D.K. (2009). Transcribe to sur-
Brodbeck, J., Balestra, M. E., Saunders, A. M., Roses, vive: transcriptional control of antioxidant defense
A. D., Mahley, R. W., and Huang, Y. (2008). programs for neuroprotection in Parkinson’s dis-
Rosiglitazone increases dendritic spine density and ease. Antioxid Redox Signal 11, 509–528.
rescues spine loss caused by apolipoprotein E4 in Collino, M., Patel, N.S., and Thiemermann, C. (2008).
primary cortical neurons. PNAS 105, 1343–1346. PPARs as new therapeutic targets for the treatment
181

Chapter 20: Ketogenic Diet and PPARgamma 181

of cerebral ischemia/reperfusion injury. Ther Adv in retinal cells via up-regulation of sestrin-1 and
Cardiovasc Dis 2, 179–197. SOD-2. J Neurochem 109, 631–643.
Combs, C.K., Johnson, D.E., Karlo, J.C., Cannady, S.B., Dreyer, C., Krey, G., Keller, H., Givel, F., Helftenbein,
and Landreth, G.E. (2000). Inflammatory mecha- G., and Wahli, W. (1992). Control of the peroxi-
nisms in Alzheimer’s disease:  inhibition of beta somal beta-oxidation pathway by a novel family of
amyloid-stimulated proinflammatory responses nuclear hormone receptors. Cell 68, 879–887.
and neurotoxicity by PPARgamma agonists. J Dutton, S.B., Sawyer, N.T., Kalume, F., Jumbo-Lucioni,
Neurosci 20, 558–567. P., Borges, K., Catterall, W.A., and Escayg, A.
Corona, J.C., and Duchen, M.R. (2015). PPARγ and (2011). Protective effect of the ketogenic diet in
PGC-1α as therapeutic targets in Parkinson’s. Scn1a mutant mice. Epilepsia 52, 2050–2056.
Neurochem Res 40, 308–316. Elbrecht, A., Chen, Y., Cullinan, C.A., Hayes, N.,
Cullingford, T.E., Bhakoo, K., Peuchen, S., Dolphin, Leibowitz, Md, Moller, D.E., and Berger, J. (1996).
C.T., Patel, R., and Clark, J.B. (1998). Distribution Molecular cloning, expression and character-
of mRNAs encoding the peroxisome proliferator- ization of human peroxisome proliferator acti-
activated receptor alpha, beta, and gamma and vated receptors gamma 1 and gamma 2. Biochem
the retinoid X receptor alpha, beta, and gamma Biophys Res Commun 224, 431–437.
in rat central nervous system. J Neurochem 70, Engel, T., Sanz-Rodgriguez, A., Jimenez-Mateos, E.M.,
1366–1375. Concannon, C.G., Jimenez-Pacheco, A., Moran,
Cullingford, T.E., Dolphin, C.T., and Sato, H. (2002a). C., Mesuret, G., Petit, E., Delanty, N., Farrell, M.A.,
The peroxisome proliferator-activated receptor et al. (2013). CHOP regulates the p53-MDM2 axis
alpha-selective activator ciprofibrate upregu- and is required for neuronal survival after seizures.
lates expression of genes encoding fatty acid Brain 136, 577–592.
oxidation and ketogenesis enzymes in rat brain. Esposito, G., Scuderi, C., Valenza, M., Togna,
Neuropharmacology. 42, 724–730. G.I., Latina, V., De Filippis, D., Cipriano, M.,
Cullingford, T.E., Eagles, D.A., and Sato, H. (2002b). Carratù, M.R., Iuvone, T., and Steardo, L. (2011).
The ketogenic diet upregulates expression of the Cannabidiol reduces Aβ-induced neuroinflam-
gene encoding the key ketogenic enzyme mito- mation and promotes hippocampal neurogenesis
chondrial 3-hydroxy-3-methylglutaryl-CoA syn- through PPARγ involvement. PLoS One 6, e28668.
thase in rat brain. Epilepsy Res 49, 99–107. Fenoglio-Simeone, K., Mazarati, A., Sefidvash-
Dehmer, T., Heneka, M.T., Sastre, M., Dichgans, J., and Hockley, S., Shin, D., Wilke, J., Milligan, H.,
Schulz, J.B. (2004) Protection by pioglitazone in Sankar, R., Rho, J.M., and Maganti, R. (2009a).
the MPTP model of Parkinson’s disease correlates Anticonvulsant effects of the selective melato-
with I  kappa B alpha induction and block of NF nin receptor agonist ramelteon. Epilepsy Behav
kappa B and iNOS activation. J Neurochem 88, 16, 52–57.
494–501. Fenoglio-Simeone, K.A., Wilke, J.C., Milligan, H.L.,
Desvergne, B., and Wahli, W. (1999) Peroxisome Allen, C.N., Rho, J.M., and Maganti, R.K. (2009b).
proliferator-activated receptors: nuclear control of Ketogenic diet treatment abolishes seizure period-
metabolism. Endocr Rev 20, 649–688. icity and improves diurnal rhythmicity in epileptic
Diab, A., Deng, C., Smith, J.D., Hussain, R.Z., Kcna1-null mice. Epilepsia 50, 2027–2034.
Phanavanh, B., Lovett-Racke, A.E., Drew, P.D., Fong, W.H., Tsai, H.D., Chen, Y.C., Wu, J.S., and Lin,
and Racke, M.K. (2002). Peroxisome proliferator- T.N. (2010). Anti-apoptotic actions of PPAR-
activated receptor-gamma agonist 15-deoxy- gamma against ischemic stroke. Mol Neurobiol 41,
Delta(12,14)-prostaglandin J(2) ameliorates 180–186.
experimental autoimmune encephalomyelitis. J Fraser, D.D., Whiting, S., Andrew, R.D., Macdonald,
Immunol 168, 2508–2515. E.A., Musa-Veloso, K., and Cunnane, S.C. (2003).
Diano, S., Liu, Z.W., Jeong, J.K., Dietrich, M.O., Ruan, Elevated polyunsaturated fatty acids in blood
H.B., Kim, E., Suyama, S., Kelly, K., Gyengesi, serum obtained from children on the ketogenic
E., Arbiser, J.L., et  al. (2011). Peroxisome diet. Neurology 60, 1026–1029.
proliferation-associated control of reactive oxygen Fuenzalida, K., Quintanilla, R., Ramos, P., Piderit, D.,
species sets melanocortin tone and feeding in diet- Fuentealba, R.A., Martinez, G., Inestrosa, N.C., and
induced obesity. Nat Med 17, 1121–1127. Bronfman, M. (2007). Peroxisome proliferator-
DiMauro, S., Kulikova, R., Tanji, K., Bonilla, E., and activated receptor gamma up-regulates the Bcl-2
Hirano, M. (1999). Mitochondrial genes for gener- anti-apoptotic protein in neurons and induces
alized epilepsies. Adv Neurol 79, 411–419. mitochondrial stabilization and protection against
Dineley, K.T., Jahrling, J.B., and Denner, L. (2014). oxidative stress and apoptosis. J Biol Chem 282,
Insulin resistance in Alzheimer’s disease. 37006–37015.
Neurobiol Dis 72, 92–103. Gahring, L.C., Persiyanov, K., Days, E.L., and Rogers,
Doonan, F., Wallace, D.M., O’Driscoll, C., and Cotter, S.W. (2005). Age-related loss of neuronal nic-
T.G. (2009). Rosiglitazone acts as a neuroprotectant otinic receptor expression in the aging mouse
182

182 section III: Ketogenic Diet in the Laboratory

hippocampus corresponds with cyclooxygenase- Rahman, S., Eaton, S., and Heales, S.J. (2014) The
2 and PPAR gamma expression and is altered by ketogenic diet component decanoic acid increases
long-term NS398 administration. J Neurobiol 62, mitochondrial citrate synthase and complex
453–468. I  activity in neuronal cells. J Neurochem 129,
Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic 426–433.
diets, mitochondria, and neurological diseases. J Itoh, T., Fairall, L., Amin, K., Inaba, Y., Szanto, A.,
Lipid Res 55, 2211–2228. Balint, B.L., Nagy, L., Yamamoto, K., and Schwabe,
Girnun, G.D., Domann, F.E., Moore, S.A., and J.W. (2008). Structural basis for the activation of
Robbins, M.E. (2002). Identification of a func- PPARgamma by oxidized fatty acids. Nat Struct
tional peroxisome proliferator-activated receptor Mol Biol 15, 924–931.
response element in the rat catalase promoter. Mol Jarrett, S.G., Milder, J.B., Liang, L.P., and Patel, M.
Endocrinol 16, 2793–2801. (2008). The ketogenic diet increases mitochondrial
Guzmán, M., and Blázquez, C. (2004). Ketone body glutathione levels. J Neurochem 106, 1044–1051.
synthesis in the brain:  possible neuroprotective Jeong, E.A., Jeon, B.T., Shin, H.J., Kim, N., Lee, D.H.,
effects. Prostaglandins Leukot Essent Fatty Acids Kim, H.J., Kang, S.S., Cho, G.J., Choi, W.S., and
70, 287–292. Roh, G.S. (2011). Ketogenic diet-induced peroxi-
Han, Y., Xie, N., Cao, L., Zhao, X., Liu, X., Jiang, H., some proliferator-activated receptor-γ activation
and Chi, Z. (2011). Adenosine monophosphate- decreases neuroinflammation in the mouse hip-
activated protein kinase and peroxisome pocampus after kainic acid-induced seizures. Exp
proliferator-activated receptor gamma coactivator Neurol 232, 195–202.
1α signaling provides neuroprotection in status Johri, A., Chandra, A., and Beal, M.F. (2013). PGC-1α,
epilepticus in rats. Neurosci Lett 500, 133–138. mitochondrial dysfunction, and Huntington’s dis-
Harris, J.J., Jolivet, R., and Attwell, D. (2012). Synaptic ease. Free Radic Biol Med 62, 37–46.
energy use and supply. Neuron 75, 762–777. Katsouri, L., Blondrath, K., and Sastre, M. (2012).
Heneka, M.T., Gavrilyuk, V., Landreth, G.E., O’Banion, Peroxisome proliferator-activated receptor-γ
M.K., Weinberg, G., and Feinstein, D.L. (2003). cofactors in neurodegeneration. IUBMB Life 64,
Noradrenergic depletion increases inflammatory 958–964.
responses in brain: effects on IkappaB and HSP70 Kersten, S., and Wahli, W. (2000). Peroxisome pro-
expression. J Neurochem 85, 387–398. liferator activated receptor agonists. EXS 89,
Heneka, M.T., Klockgether, T., and Feinstein, D.L. 141–151
(2000). Peroxisome proliferator activated receptor- Kim, D.Y., Simeone, K.A., Simeone, T.A., Pandya, J.D.,
gamma ligands reduce neuronal inducible nitric Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G.,
oxide synthase expression and cell death in vivo, J. and Rho, J.M. (2015). Ketone bodies mediate anti-
Neurosci. 20, 6862–6867. seizure effects through mitochondrial permeabil-
Heneka, M.T., and Landreth, G.E. (2007). PPARs in ity transition. Ann Neurol 78, 77–87.
the brain. Biochim Biophys Acta 1771, 1031–1045. Kitamura, Y., Shimohama, S., Koike, H., Kakimura, Ji.,
Hong, S., Huang, Y., Yu, X., Li, Y., Yang, J., Li, R., Deng, Matsuoka, Y., Nomura, Y., Gebicke-Haerter, P.J., and
Y., and Zhao, G. (2008). Peroxisome proliferator- Taniguchi, T. (1999). Increased expression of cyclo-
activated receptor gamma agonist, rosiglitazone, oxygenases and peroxisome proliferator-activated
suppresses CD40 expression and attenuates receptor-gamma in Alzheimer’s disease brains.
inflammatory responses after lithium pilocarpine- Biochem Biophys Res Commun 254, 582–586.
induced status epilepticus in rats. Int J Dev Kobow, K., Auvin, S., Jensen, F., Löscher, W., Mody, I.,
Neurosci 26, 505–515. Potschka, H., Prince, D., Sierra, A., Simonato, M.,
Hong, S., Xin, Y., HaiQin, W., GuiLian, Z., Ru, Z., Pitkänen, A., et  al. (2012). Finding a better drug
ShuQin, Z., HuQing, W., Li, Y., and Yun, D. (2012). for epilepsy: antiepileptogenesis targets. Epilepsia
The PPARγ agonist rosiglitazone prevents cogni- 53, 1868–1876.
tive impairment by inhibiting astrocyte activation Kroker, A.J., and Bruning, J.B. (2015). Review of the
and oxidative stress following pilocarpine-induced structural and dynamic mechanisms of PPARγ par-
status epilepticus. Neurol Sci 33, 559–566. tial agonism. PPAR Res. doi: 10.1155/2015/816856.
Hong, S., Xin, Y., HaiQin, W., GuiLian, Z., Ru, Z., Kudin, A.P., Zsurka, G., Elger, C.E., and Kunz, W.S.
ShuQin, Z., HuQing, W., Li, Y., Ning, B., and (2009). Mitochondrial involvement in temporal
YongNan, L. (2013). The PPARγ agonist rosigli- lobe epilepsy. Exp Neurol 218, 326–332.
tazone prevents neuronal loss and attenuates devel- Kunz, W.S., Kudin, A.P., Vielhaber, S., Blümcke, I.,
opment of spontaneous recurrent seizures through Zuschratter, W., Schramm, J., Beck, H., and Elger,
BDNF/TrkB signaling following pilocarpine- C.E. (2000). Mitochondrial complex I  deficiency
induced status epilepticus. Neurochem Int 63, in the epileptic focus of patients with temporal
405–412. lobe epilepsy. Ann Neurol 48, 766–773.
Hughes, S.D., Kanabus, M., Anderson, G., Hargreaves, Lee, D., Lee, K.H., Ho, W.K., and Lee, S.H. (2007).
I.P., Rutherford, T., O’Donnell, M., Cross, J.H., Target cell-specific involvement of presynaptic
183

Chapter 20: Ketogenic Diet and PPARgamma 183

mitochondria in post-tetanic potentiation at hip- of the Epilepsies, 4th edition, J.L. Noebels, M.
pocampal mossy fiber synapses. J Neurosci 27, Avoli, M.A. Rogawski, R.W. Olsen, and A.V.
13603–13613. Delgado-Escueta, eds. (Bethesda, MD:  National
Lee, S.H., Kim, K.R., Ryu, S.Y., Son, S., Hong, H.S., Center for Biotechnology Information).
Mook-Jung, I., Lee, S.H., and Ho, W.K. (2012). Maurois, P., Rocchi, S., Pages, N., Bac, P., Stables,
Impaired short-term plasticity in mossy fiber syn- J.P., Gressens, P., and Vamecq, J. (2008). The
apses caused by mitochondrial dysfunction of den- PPARgamma agonist FMOC-L-leucine pro-
tate granule cells is the earliest synaptic deficit in a tects both mature and immature brain. Biomed
mouse model of Alzheimer’s disease. J Neurosci Pharmacother 62, 259–263.
32, 5953–5963. Medina-Gomez, G., Gray, S., and Vidal-Puig, A.
Lim, G. P., Yang, F., Chu, T., Chen, P., Beech, W., Teter, (2007a). Adipogenesis and lipotoxicity:  role of
B., Tran, T., Ubeda, O., Ashe, K. H., Frautschy, S. A., peroxisome proliferator-activated receptor gamma
and Cole, G. M. (2000). Ibuprofen suppresses plaque (PPARgamma) and PPARgammacoactivator-1
pathology and inflammation in a mouse model for (PGC1). Public Health Nutr 10, 1132–1137.
Alzheimer’s disease. J Neurosci 20, 5709–5714. Medina-Gomez, G., Gray, S.L, Yetukuri, L.,
Lu, J., Wu, D.M., Zheng, Y.L., Hu, B., Cheng, W., Shimomura, K., Virtue, S., Campbell, M., Curtis,
Zhang, Z.F., and Li, M.Q. (2013). Troxerutin coun- R.K., Jimenez-Linan, M., Blount, M., Yeo, G.S.,
teracts domoic acid-induced memory deficits in et al. (2007b). PPAR gamma 2 prevents lipotoxic-
mice by inhibiting CCAAT/enhancer binding ity by controlling adipose tissue expandability and
protein beta-mediated inflammatory response and peripheral lipid metabolism. PLoS Genet 3:e64.
oxidative stress. J Immunol 190, 3466–3479. Miglio, G., Rosa, A.C., Rattazzi, L., Collino,
Lu, M., Sarruf, D.A., Talukdar, S., Sharma, S., Li, P., M., Lombardi, G., and Fantozzi, R. (2009).
Bandyopadhyay, G., Nalbandian, S., Fan, W., PPARgamma stimulation promotes mitochon-
Gayen, J.R., Mahata, S.K., Webster, N.J., et  al. drial biogenesis and prevents glucose deprivation-
(2011). Brain PPAR-γ promotes obesity and is induced neuronal cell loss. Neurochem Int 55,
required for the insulin-sensitizing effect of thia- 496–504.
zolidinediones. Nat Med 17, 618–622. Milder, J.B., Liang, L.P., and Patel, M. (2010). Acute
Luna-Medina, R., Cortes-Canteli, M., Sanchez- oxidative stress and systemic Nrf2 activation by
Galiano, S., Morales-Garcia, J.A., Martinez, the ketogenic diet. Neurobiol Dis 40, 238–244.
A., Santos, A., and Perez-Castillo, A. (2007). Moreno, S., Farioli-Vecchioli, S., and Cerù, M.P. (2004).
NP031112, a thiadiazolidinone compound, pre- Immunolocalization of peroxisome proliferator-
vents inflammation and neurodegeneration under activated receptors and retinoid X receptors in the
excitotoxic conditions:  potential therapeutic role adult rat CNS. Neuroscience 123, 131–145.
in brain disorders. J Neurosci 27, 5766–5776. Mueller, S.G., Kollias, S.S., Trabesinger, A.H., Buck, A.,
Maguire, J., and Salpekar, J.A. (2013). Stress, seizures, Boesiger, P., and Wieser, H.G. (2001). Proton mag-
and hypothalamic-pituitary-adrenal axis targets netic resonance spectroscopy characteristics of a
for the treatment of epilepsy. Epilepsy Behav 26, focal cortical dysgenesis during status epilepticus
352–362. and in the interictal state. Seizure 10, 518–524.
Malapaka, R.R., Khoo, S., Zhang, J., Choi, J.H., Zhou, Okada, K., Yamashita, U., and Tsuji, S. (2006).
X.E., Xu, Y., Gong, Y., Li, J., Yong, E.L., Chalmers, Ameliorative effect of pioglitazone on seizure
M.J., et  al. (2012). Identification and mechanism responses in genetically epilepsy-susceptible EL
of 10-carbon fatty acid as modulating ligand of mice. Brain Res 1102, 175–178.
peroxisome proliferator-activated receptors. J Biol Overington, J.P., Al-Lazikani, B., and Hopkins, A.L.
Chem 287, 183–195. (2006) How many drug targets are there? Nat Rev
Malthankar-Phatak, G.H., de Lanerolle, N., Eid, T., Drug Discov 5, 993–996.
Spencer, D.D., Behar, K.L., Spencer, S.S., Kim, J.H., Porta, N., Vallee, L., Lecointe, C., Bouchaert, E., Staels,
and Lai, J.C. (2006). Differential glutamate dehy- B., Bordet, R., and Auvin, S. (2009). Fenofibrate,
drogenase (GDH) activity profile in patients with a peroxisome proliferator-activated receptor-alpha
temporal lobe epilepsy. Epilepsia 47, 1292–1299. agonist, exerts anticonvulsive properties. Epilepsia
Mandrekar-Colucci, S., and Landreth, G.E. (2011). 50, 943–948.
Nuclear receptors as therapeutic targets for Puigserver, P., Adelmant, G., Wu, Z., Fan, M., Xu, J.,
Alzheimer’s disease. Expert Opin Ther Targets 15, O’Malley, B., and Spiegelman, B. (1999). Activation
1085–1097. of PPARgamma coactivator-1 through transcrip-
Mandrekar-Colucci, S., Sauerbeck, A., Popovich, P.G., tion factor docking. Science 286, 1368–1371.
and McTigue, D.M. (2013). PPAR agonists as Puligheddu, M., Pillolla, G., Melis, M., Lecca, S.,
therapeutics for CNS trauma and neurological dis- Marrosu, F., De Montis, M.G., Scheggi, S., Carta,
eases. ASN Neuro 5:e00129. G., Murru, E., Aroni, S., et al. (2013). PPAR-alpha
Masino, S.A., and Rho, J.M. (2012). Mechanisms of agonists as novel antiepileptic drugs:  Preclinical
ketogenic diet action. In Jasper’s Basic Mechanisms findings. PLoS One 8, e64541.
184

184 section III: Ketogenic Diet in the Laboratory

Quintanilla, R. A., Godoy, J. A., Alfaro, I., Cabezas, D., hippocampal slices from epileptic Kv1.1α knock-
von Bernhardi, R., Bronfman, M., and Inestrosa, out mice. Epilepsia 55, e44–49.
N.C. (2013). Thiazolidinediones promote axonal Smart, S.L., Lopantsev, V., Zhang, C.L., Robbins,
growth through the activation of the JNK pathway. C.A., Wang, H., Chiu, S.Y., Schwartzkroin, P.A.,
PLoS ONE 8, e65140. Messing, A., and Tempel, B.L. (1998). Deletion of
Quintanilla, R.A., Utreras, E., and Cabezas-Opazo, the K(V)1.1 potassium channel causes epilepsy in
F.A. (2014). Role of PPAR γ in the Differentiation mice. Neuron 20, 809–819.
and Function of Neurons. PPAR Res. doi: 10.1155/ Steger, D.J., Grant, G.R., Schupp, M., Tomaru, T.,
2014/768594. Lefterova, M.I., Schug, J., Manduchi, E., Stoeckert,
Rho, J.M., Kim, D.W., Robbins, C.A., Anderson, G.D., C.J., Jr., Lazar, M.A. (2010). Propagation of adipo-
Schwartzkroin, P.A. (1999). Age-dependent differ- genic signals through an epigenomic transition
ences in flurothyl seizure sensitivity in mice treated state. Genes Dev 24, 1035–1044.
with a ketogenic diet. Epilepsy Res 37, 233–240. St-Pierre, J., Drori, S., Uldry, M., Silvaggi, J.M., Rhee,
Ryan, K., Backos, D.S., Reigan, P., and Patel, M. (2012). J., Jäger, S., Handschin, C., Zheng, K., Lin, J., Yang,
Post-translational oxidative modification and W., et  al. (2006). Suppression of reactive oxygen
inactivation of mitochondrial complex I in epilep- species and neurodegeneration by the PGC-1 tran-
togenesis. J Neurosci 32, 11250–11258. scriptional coactivators. Cell 127, 397–408.
Sarruf, D.A., Yu, F., Nguyen, H.T., Williams, D.L., Sudha, K., Rao, A.V., and Rao, A. (2001). Oxidative
Printz, R.L., Niswender, K.D., and Schwartz, M.W. stress and antioxidants in epilepsy. Clin Chim Acta
(2009). Expression of peroxisome proliferator- 303, 19–24.
activated receptor-gamma in key neuronal subsets Sugii, S., and Evans, R.M. (2011). Epigenetic codes
regulating glucose metabolism and energy homeo- of PPARγ in metabolic disease. FEBS Lett 585,
stasis. Endocrinology 150, 707–712. 2121–2128.
Sauer, S. (2015). Ligands for the nuclear peroxisome Sullivan, P.G., Dube, C., Dorenbos, K., Steward, O., and
proliferator-activated receptor gamma. Trends Baram, T.Z. (2003). Mitochondrial uncoupling
Pharmacol Sci 36, 688–704. protein-2 protects the immature brain from exci-
Schrader, M., Costello, J.L., Godinho, L.F., Azadi, A.S., totoxic neuronal death. Ann Neurol 53, 711–717.
and Islinger, M. (2015). Proliferation and fission of Sullivan, P.G., Rippy, N.A., Dorenbos, K., Concepcion,
peroxisomes—an update. Biochim Biophys Acta. R.C., Agarwal, A.K., and Rho, J.M. (2004). The
doi: 10.1016/j.bbamcr.2015.09.024 ketogenic diet increases mitochondrial uncoupling
Shao, D., Rangwala, S.M., Bailey, S.T., Krakow, protein levels and activity. Ann Neurol 55, 576–580.
S.L., Reginato, M.J., and Lazar, M.A. (1998). Sundararajan, S., Gamboa, J.L., Victor, N.A., Wanderi,
Interdomain communication regulating ligand E.W., Lust, W.D., and Landreth, G.E. (2005).
binding by PPARgamma. Nature 396, 377–380. Peroxisome proliferator-activated receptor-gamma
Shen, Y., Lu, Y., Yu, F., Zhu, C., Wang, H., and Wang, ligands reduce inflammation and infarction size
J. (2015). Peroxisome proliferator-activated in transient focal ischemia. Neuroscience 130,
receptor-γ and its ligands in the treatment of 685–696.
tumors in the nervous system. Curr Stem Cell Res Takahashi, S., Iizumi, T., Mashima, K., Abe, T., and
Ther. doi: 10.2174/1574888X10666150728122034 Suzuki, N. (2014). Roles and regulation of keto-
Simeone, T.A., Simeone, K.A., Samson, K.K., Kim, do genesis in cultured astroglia and neurons under
Y., and Rho, J.M. (2013). Loss of the Kv1.1 potas- hypoxia and hypoglycemia. ASN Neuro 6.
sium channel promotes pathologic sharp waves doi: 10.1177/1759091414550997
and high frequency oscillations in in vitro hippo- Tan, Y., Muise, E.S., Dai, H., Raubertas, R., Wong, K.K.,
campal slices. Neurobiol Dis 54, 68–81. Thompson, G.M., Wood, H.B., Meinke, P.T., Lum,
Simeone, K.A., Matthews, S.A., Samson, K.K., and P.Y., Thompson, J.R., and Berger, J.P. (2012). Novel
Simeone, T.A. (2014a). Targeting deficiencies in transcriptome profiling analyses demonstrate that
mitochondrial respiratory complex I  and func- selective peroxisome proliferator-activated receptor
tional uncoupling exerts anti-seizure effects in a γ (PPARγ) modulators display attenuated and selec-
genetic model of temporal lobe epilepsy and in a tive gene regulatory activity in comparison with
model of acute temporal lobe seizures. Exp Neurol PPARγ full agonists. Mol Pharmacol 82, 68–79.
251, 84–90. Thouennon, E., Cheng, Y., Falahatian, V., Cawley,
Simeone, T.A., Matthews, S.A., Samson, K.K., and Simeone, N.X., and Loh, Y.P. (2015). Rosiglitazone-activated
K.A. (2016). Regulation of brain PPARgamma2 con- PPARγ induces neurotrophic factor-α1 transcrip-
tributes to ketogenic diet anti-seizure efficacy. Exp tion contributing to neuroprotection. J Neurochem
Neurol. doi: 10.1016/j.expneurol.2016.08.006. 134, 463–470.
Simeone, T.A., Samson, K.K., Matthews, S.A., and Tontonoz, P., Hu, E., Graves, R.A., Budavari, A.I., and
Simeone, K.A. (2014b). In vivo ketogenic diet Spiegelman, B.M. (1994). mPPAR gamma 2: tissue-
treatment attenuates pathologic sharp waves specific regulator of an adipocyte enhancer. Genes
and high frequency oscillations in in vitro Dev 8, 1224–1234.
185

Chapter 20: Ketogenic Diet and PPARgamma 185

Tontonoz, P., and Spiegelman, B.M. (2008). Fat and Wu, J.S., Lin, T.N., and Wu, K.K. (2009b). Rosiglitazone
beyond:  the diverse biology of PPARgamma. and PPAR-gamma overexpression protect mito-
Annu Rev Biochem 77, 289–312. chondrial membrane potential and prevent apop-
Victor, N.A., Wanderi, E.W., Gamboa, J., Zhao, X., tosis by upregulating anti-apoptotic Bcl-2 family
Aronowski, J., Deininger, K., Lust, W.D., Landreth, proteins. J Cell Physiol 220, 58–71.
G.E., and Sundararajan, S. (2006). Altered Yakunin, E., Kisos, H., Kulik, W., Grigoletto, J.,
PPARgamma expression and activation after tran- Wanders, R.J., and Sharon, R. (2014). The regula-
sient focal ischemia in rats. Eur J Neurosci 24, tion of catalase activity by PPAR γ is affected by
1653–1663. α-synuclein. Ann Clin Transl Neurol 1, 145–159.
Vidal-Puig, A., Jimenez-Liñan, M., Lowell, B.B., Yamamoto, K., Itoh, T., Abe, D., Shimizu, M., Kanda,
Hamann, A., Hu, E., Spiegelman, B., Flier, J.S., T., Koyama, T., Nishikawa, M., Tamai, T., Ooizumi,
and Moller, D.E. (1996). Regulation of PPAR H., and Yamada, S. (2005). Identification of puta-
gamma gene expression by nutrition and obesity tive metabolites of docosahexaenoic acid as potent
in rodents. J Clin Invest 97, 2553–2561. PPARgamma agonists and antidiabetic agents.
Vidal-Puig, A.J., Considine, R.V., Jimenez-Liñan, M., Bioorg Med Chem Lett 15, 517–522.
Werman, A., Pories, W.J., Caro, J.F., and Flier, J.S. Yan, Q., Zhang, J., Liu, H., Babu-Khan, S., Vassar, R.,
(1997). Peroxisome proliferator-activated receptor Biere, A. L., Citron, M., and Landreth, G. (2003).
gene expression in human tissues. Effects of obe- Antiinflammatory drug therapy alters β-amyloid
sity, weight loss, and regulation by insulin and glu- processing and deposition in an animal model of
cocorticoids. J Clin Invest 99, 2416–2422. Alzheimer’s disease. J Neurosci 23, 7504–7509.
Vielhaber, S., Niessen, H.G., Debska-Vielhaber, G., Yang, X., and Cheng, B. (2010). Neuroprotective and
Kudin, A.P., Wellmer, J., Kaufmann, J., Schönfeld, anti-inflammatory activities of ketogenic diet on
M.A., Fendrich, R., Willker, W., Leibfritz, D., MPTP-induced neurotoxicity. J Mol Neurosci 42,
et  al. (2008). Subfield-specific loss of hippocam- 145–153.
pal N-acetyl aspartate in temporal lobe epilepsy. Youle, R.J., and Strasser, A. (2008). The Bcl-2 protein
Epilepsia 49, 40–50. family: opposing activities that mediate cell death.
Wada, K., Nakajima, A., Katayama, K., Kudo, C., Nat Rev Mol Cell Biol 9, 47–59
Shibuya, A., Kubota, N., Terauchi, Y., Tachibana, M., Yu, X., Shao, X.G., Sun, H., Li, Y.N., Yang, J., Deng,
Miyoshi, H., Kamisaki, Y., et al. (2006). Peroxisome Y.C., and Huang, Y.G. (2008). Activation of cere-
proliferator-activated receptor gamma-mediated bral peroxisome proliferator-activated receptors
regulation of neural stem cell proliferation and dif- gamma exerts neuroprotection by inhibiting oxi-
ferentiation. J Biol Chem 281, 12673–12681. dative stress following pilocarpine-induced status
Wallace, D.C. (1999). Mitochondrial diseases in man epilepticus. Brain Res 1200, 146–158.
and mouse. Science 283, 1482–1488. Zhao, X., Strong, R., Zhang, J., Sun, G., Tsien, J.Z., Cui,
Wang, H., Jiang, R., He, Q., Zhang, Y., Zhang, Y., Li, Z., Grotta, J.C., and Aronowski, J. (2009). Neuronal
Y., Zhuang, R., Luo, Y., Li, Y., Wan, J., et al. (2012). PPARgamma deficiency increases susceptibility to
Expression pattern of peroxisome proliferator- brain damage after cerebral ischemia. J Neurosci
activated receptors in rat hippocampus following 29, 6186–6195.
cerebral ischemia and reperfusion injury. PPAR Zhao, Y., Patzer, A., Gohlke, P., Herdegen, T., and
Res. doi: 10.1155/2012/596394. Culman, J. (2005). The intracerebral application
Wenzel, H.J., Vacher, H., Clark, E., Trimmer, J.S., of the PPARgamma-ligand pioglitazone confers
Lee, A.L., Sapolsky, R.M., Tempel, B.L., and neuroprotection against focal ischaemia in the rat
Schwartzkroin, P.A. (2007). Structural conse- brain. Eur J Neurosci 22, 278–282.
quences of Kcna1 gene deletion and transfer in the Zhao, Y., Patzer, A., Herdegen, T., Gohlke, P., and
mouse hippocampus. Epilepsia 48, 2023–2046. Culman, J. (2006). Activation of cerebral peroxi-
Werman, A., Hollenberg, A., Solanes, G., Bjorbaek, some proliferator-activated receptors gamma pro-
C., Vidal-Puig, A.J., and Flier, J.S. (1997). Ligand- motes neuroprotection by attenuation of neuronal
independent activation domain in the N termi- cyclooxygenase-2 overexpression after focal cere-
nus of peroxisome proliferator-activated receptor bral ischemia in rats. FASEB J 20, 1162–1175.
gamma (PPARgamma). Differential activity of Zhu, Y., Alvares, K., Huang, Q., Rao, M.S., and Reddy,
PPARgamma1 and -2 isoforms and influence of J.K. (1993). Cloning of a new member of the per-
insulin. J Biol Chem 272, 20230–20235. oxisome proliferator-activated receptor gene family
Wu, J.S., Cheung, W.M., Tsai, Y.S., Chen, Y.T., Fong, from mouse liver. J Biol Chem 268, 26817–26820.
W.H., Tsai, H.D., Chen, Y.C., Liou, J.Y., Shyue, S.K., Zolezzi, J.M., Bastías-Candia, S., Santos, M.J., and
Chen, J.J., et al. (2009a). Ligand-activated peroxi- Inestrosa, N.C. (2014). Alzheimer’s disease:  rel-
some proliferator activated receptor-gamma pro- evant molecular and physiopathological events
tects against ischemic cerebral infarction and affecting amyloid-β brain balance and the puta-
neuronal apoptosis by 14-3-3 epsilon upregula- tive role of PPARs. Front Aging Neurosci 6, 176.
tion. Circulation 119, 1124–1134. doi: 10.3389/fnagi.2014.00176
186

21
Ketogenic Diet in a Hippocampal Slice
Models and Mechanisms

M A S A H I T O K AWA M U R A J R . , M D , P H D

HIPPOCAMPUS AS A MODEL Interestingly, CA3 pyramidal neurons are con-


FOR TEMPORAL LOBE nected to each other by excitatory recurrent col-
EPILEPSY laterals. Thus, the hippocampal circuit is regulated
The hippocampus is well known as a brain area by a balance between excitation from recurrent
involved in learning and memory and also as the collaterals and inhibition from interneurons.
key region underlying the form of epilepsy known When the balance collapses, the hippocampal cir-
as mesial temporal lobe epilepsy. The temporal cuit becomes hyperexcitable and causes seizures
lobe refers to the ventrolateral middle part of easily. Therefore, the atrophy in the hippocampus
neocortex, and abnormal neuronal discharge or a is thought be one of the main focuses of mesial
lesion affecting this lobe causes seizures (Gastaut, temporal lobe epilepsy, and excision of hippocam-
1973). There are two main types of temporal lobe pal sclerosis with selective amygdalohippocam-
epilepsy classified by the epileptic focus:  mesial pectomy successfully improves ~70% of surgical
and lateral. The focus of mesial temporal lobe patients (Paglioli et al., 2006; Wiebe et al., 2001).
epilepsy is the hippocampus, amygdala, or para- From these clinical findings, the hippocampus
hippocampal gyrus, and that of lateral temporal is thought to be a good experimental target for
lobe epilepsy is in neocortex. Over 80% of patients investigating epileptogenesis and therapy of tem-
with temporal lobe epilepsy have the mesial form poral lobe epilepsy.
(Quarato et al., 2005; Schramm et al., 2001). The The ketogenic diet was designed in the 1920s to
typical symptom of mesial temporal lobe epilepsy treat epilepsy by mimicking fasting (Wilder, 1921).
is complex partial seizures, which have a high Despite almost 100 years of clinical use, the mech-
probability of an accompanying aura. The aura anisms underlying the success of ketogenic diet
is characteristic, usually presenting as epigastric therapy are not well understood. In recent decades,
discomfort sometimes described as nausea or ketogenic diet has increasingly been noted as a
psychiatric symptoms including fear. Complex useful therapy for medically refractory epilepsy
partial seizures often begin with arrest of motor (Hallbook et  al., 2007). The patients of temporal
activities or staring. Autonomic motor behaviors lobe epilepsy are well known to be frequently resis-
are usually oroalimentary automatisms or com- tant to antiepileptic drugs (Wiebe and Jette, 2012).
plex automatisms. Dystonic posturing lasting 1–2 As mentioned previously, the first choice of treat-
minutes often occurs involving the arm contralat- ment for mesial temporal lobe epilepsy associated
eral to the ictal discharge (Engel, 2001). In a major- with hippocampal sclerosis is surgery (anterior
ity of patients, mesial temporal lobe epilepsies are temporal lobectomy or selective amygdalohippo-
associated with hippocampal sclerosis (Watson, campectomy) because of good therapeutic out-
2003), which is atrophy with global gliosis and comes (Tanriverdi et al., 2008). For temporal lobe
loss of CA1 and/or CA3 pyramidal neurons in epilepsy patients who are not good candidates
the hippocampus (Thom, 2009). The structure of for surgery, however, ketogenic diet is one of the
the hippocampus is simple. It includes principal therapeutic options (Ray and Wyllie, 2005). Thus,
cells (granular cells of dentate gyrus and CA1-4 a natural question is how the ketogenic diet pro-
pyramidal neurons) and surrounding interneu- duces its beneficial effects in temporal lobe epi-
rons. The principal cells form an excitatory cir- lepsy. In this chapter, we focus on investigating
cuit that is modulated by inhibitory internurons. ketogenic diet antiseizure mechanisms with acute
187

Chapter 21: Ketogenic Diet in a Hippocampal Slice 187

slice preparations of the hippocampus, reviewing concentrations (Congar et  al., 2000; Dulla et  al.,
our own and other laboratories’ work. 2005; Kojima et  al., 1989; Stafstrom et  al., 2009).
All these approaches support the use of hippocam-
T H E I M P O R TA N C E pal slice preparations to elucidate epileptic mech-
O F   H I P P O C A M PA L anisms. On the other hand, the pitfall of in vitro
S L I C E P R E PA R AT I O N S recording is that the environment of acute brain
F O R   S T U DY I N G slice preparations is different from the in vivo
ANTIEPILEPTIC condition. We make a slice by cutting brain tissue
MECHANISMS and that causes traumatic injury:  reactive gliosis
O F   K E TO G E N I C   D I E T occurs in the acute hippocampal slice (Takano
Because epilepsy is caused by abnormal neuro- et  al., 2014). Also, artificial cerebrospinal fluid
nal discharges in the brain, electrophysiologi- does not exactly reproduce actual cerebrospinal
cal measurements are the most direct and useful fluid. Thus, results from in vitro hippocampal slice
approach for researching epilepsy and its treat- preparations should be confirmed by in vivo elec-
ments. There are two approaches for electrophysi- trophysiological recordings or behavioral tests as
ological recordings of any brain region:  in vivo much as possible. For that reason, both in vivo and
and in vitro. In vivo electrophysiological recording in vitro electrophysiological recordings are useful,
of hippocampus is usually done by extracellular and both are essential for epilepsy research.
recording of electrically evoked activity (Stewart
and Reid, 1993), or continuous recording of spon- T H R E E A P P ROAC H E S
taneous field activity (Li et  al., 2008)  or multiple FOR REPRODUCING
unit activity (Lin et al., 2006); these preparations THE CONDITIONS
can be acute or chronic. In vitro electrophysiologi- O F   K E TO G E N I C
cal recording is done using single-cell intracel- DIET FEEDING
lular sharp electrodes (Abe and Ogata, 1981)  or I N   H I P P O C A M PA L   S L I C E S
patch-clamp electrodes (Kawamura et  al., 2004), The major difficulty in using acute hippocampal
or extracellular field recording with single elec- slices for ketogenic diet research is the inability to
trodes (Masino and Dunwiddie, 1999) or electrode precisely reproduce or maintain the condition of
arrays (Knowles et al., 1987) using acute slices of diet therapy in vitro preparation. In vivo recording
hippocampus. As compared with in vivo hippo- clearly does not have this problem, because it uses
campal recordings, the advantages of hippocampal the whole body of experimental animals and diet-
slices are several-fold: (1) Ease of use: acute brain altered metabolism is maintained (Koranda et al.,
slices must be maintained by perfusion with oxy- 2011; Masino et al., 2011). Therefore, special strat-
genated artificial cerebrospinal fluid (Sakmann egies must be implemented for examining mecha-
et  al., 1989). Continuous perfusion allows us to nisms of the ketogenic diet using hippocampal
change the extracellular fluid easily. Thus, it is slice preparations; in this section, we review three
easy to apply and wash out several agonists and/ of these approaches (Figure 21.1).
or antagonists of various proteins such as chan-
nels, receptors, and transporters, and it is easy Direct Application of Ketone Bodies
to examine the detail of functional mechanisms The ketogenic diet was developed to mimic fast-
of neuronal activities; (2)  Reduction:  we usually ing, which alleviates epileptic seizures (Wilder,
make 3–6 brain slices from one rodent and get 1921). Ketogenic diet increases ketone bodies
3–6 recordings from them, allowing us to reduce (β-hydroxybutyrate, acetoacetate, acetone), syn-
the number of animals used; (3)  History:  a huge thesized from free fatty acids in the liver (Masino
number of electrophysiological experiments have and Rho, 2012)  and then used for energy in the
been done using hippocampal slice preparations brain instead of glucose (Masino et  al., 2009).
in the last half-century. Several methods for caus- Chronic ketosis is one of hallmarks of the keto-
ing seizure-like bursting in vitro have been used genic diet. Therefore, one approach to reproduc-
in the hippocampal slice preparation, including ing a ketogenic diet in a hippocampal slice is direct
kindling (Sayin et al., 1999), kainic acid treatment application of ketone bodies, revealing whether
(Congar et  al., 2000; Smith and Dudek, 2001), ketone bodies modulate neuronal activity directly.
inhibition of GABA receptors (Kohling et al., 2000; In this paradigm, hippocampal slices are taken
Stafstrom et al., 2009), inhibition of potassium ion from control diet–fed animals and dissolved ketone
channels (Stafstrom et  al., 2009), and neuronal bodies are applied in an extracellular solution such
hyperexcitability by changing extracellular ion as artificial cerebrospinal fluid (Figure  21.1A1,
188

188 section III: Ketogenic Diet in the Laboratory

(a) A1 Ketone body

Control diet (CD)

A2 Reduced glucose
Hippocampal slices

Increased ATP

(b)

Ketogenic diet Reduced glucose


CD (~2 weeks)

Hippocampal slices

FIGURE  21.1 Three approaches for reproducing the conditions of ketogenic diet feeding in hippocampal slices.
(A1) The first approach is the direct application of ketone bodies to hippocampal slices from control diet-fed animals.
(A2) The second approach mimics ketogenic diet conditions by using a whole-cell patch clamp recording of a single
cell in a hippocampal slice from control diet-fed animals. To mimic the ketogenic diet milieu, extracellular glucose is
reduced and intracellular ATP is increased. (b) The third approach is recording hippocampal slices made from ketogenic
diet-fed animals. Extracellular glucose is reduced during incubation and recording to maintain the in vivo effect of the
ketogenic diet.

Table 21.1). Thio et al. reported that direct appli- tetanic stimulation-induced long-term potentia-
cation of ketone bodies had no effect on synaptic tion (LTP) recorded from the CA1 region in acute
activity in acute hippocampal slices from Sprague- hippocampal slices from Wistar rats (Kimura et al.,
Dawley (SD) rats. They recorded evoked field 2012). Similar results were reported that mixed
excitatory postsynaptic potentials (fEPSP) and ketone bodies (1 mM acetoacetate and 2 mM β-
population spikes (PS) in the CA1 region stimu- hydroxybutyrate) had no effect on CA1 region
lated by Schaffer collateral fibers, and applied synaptic transmission or theta burst-induced LTP
mixed ketone bodies (1 mM acetoacetate and in SD rat acute hippocampal slices (Youssef, 2015).
2 mM β-hydroxybutyrate) to the slices (Thio et al., A  unique approach was used by Samoilova et  al.
2000). A  20-minute application of ketone bodies (Samoilova et  al., 2010). They made organotypic
did not change either fEPSP slope or PS amplitude. hippocampal slices, which were cultured with low
They also recorded potassium channel blocker glucose and 10 mM β-hydroxybutyrate medium
4-aminopyridine-induced epileptiform discharge for at least 3  days. This chronic in vitro ketosis,
from the dentate granule cell layer and CA3 however, did not alleviate intrinsic or induced epi-
region, and reported that application for 105 min- leptiform discharges (but was neuroprotective).
utes did not change the frequency or duration of All of these studies concluded that ketone bodies
these ictal events. Kimura et al. also reported that do not directly affect synaptic transmission in the
application of mixed ketone bodies (1 mM each rat hippocampus.
acetoacetate and β-hydroxybutyrate) for 20 min- Other studies, however, have found positive
utes did not change fEPSP slope and an 80-minute results. Juge et al. (2010) made acute hippocam-
application did not change the high-frequency pal slices from C57BL/6 mice and incubated the
189

TABLE 21.1 REFERENCE LISTS OF THREE APPROACHES FOR REPRODUCING THE CONDITIONS OF KETOGENIC DIET FEEDING

Reference Animal Manipulation Recording Effect

Direct Application of Ketone Bodies


Thio et al., 2000 SD rat 1 mM AA and 2 mM βHB for 20 min. fEPSP and PS recordings No change
Kimura et al., 2012 Wistar rat 1 mM AA and 1 mM βHB for 20 min fEPSP recording No change
Youssef, 2015 SD rat 1 mM AA and 2 mM βHB fEPSP recording No change
Samoilova et al., 2010 Wistar rat 10 mM βHB for 3 days (slice culture) PS recording No change
Juge et al., 2010 C57BL/6 mouse 10 mM AA for 2 hrs Whole-cell patch clamp Reducing frequency and amplitude of mEPSC
recording in CA1 pyramidal neurons
Tanner et al., 2011 C57BL/6 mouse 2 mM βHB for 40 min Single channel recording Increasing open probability of KATP channels in
granule cells
Kim et al., 2015 Kcna1-null mouse 5 mM βHB and 1 mM AA for 2 weeks Multielectrode recordings Reducing spontaneous seizure-like events
(slice culture)
Controlling glucose and ATP levels with whole-cell patch clamp.
Kawamura et al., 2010 SD rat and C57BL/6 Increased [ATP]i and reduced Whole-cell patch clamp Causing hyperpolarization in CA3 pyramidal
mouse [glucose]e recording neurons
Acute hippocampal slices from ketogenic diet-fed animals.
Stafstrom et al., 1999 SD rat KD (4.8:1 ratio) for 6-8 weeks fEPSP and PS recordings Reducing spontaneous seizure-like events
Simeone et al., 2015 Kcna1-null mouse KD (6:1 ratio) for 11-15 days Multielectrode recordings Reducing spontaneous seizure-like events
Bough et al., 2006 SD rat KD (6:1 ratio) for 20 days fEPSP recording Inhibiting reduced glucose-induced depressions
Kawamura et al., 2014 SD rat and C57BL/6 KD (6:1 ratio) for 13-18 days PS recording Reducing evoked seizure-like bursting
mouse

AA, acetoacetate; βHB, β-hydroxybutyrate; fEPSP, field excitatory postsynaptic potentials; PS, population spikes; mEPSC, miniature excitatory postsynaptic currents; KATP channels, ATP-sensitive potassium channels;
[ATP]i, intracellular ATP concentration; [glucose]e, extracellular glucose concentration; KD, ketogenic diet.
190

190 section III: Ketogenic Diet in the Laboratory

slices with 10 mM acetoacetate for over 2 hours, this ketone body reduced the number of seizures
after which they recorded EPSPs from CA1 pyra- (Kim et al., 2015).
midal neurons using whole-cell patch clamp. In sum, several studies have used direct appli-
Frequency and amplitude of miniature EPSPs cation of ketone bodies in hippocampal acute
(mEPSP) from acetoacetate-incubated slices slices or organotypic cultures, and both positive
were significantly reduced compared with con- and negative results have been found. Negative and
trol slices. Ketone bodies inhibited valinomycin- positive studies used rats and mice, respectively, so
evoked glutamate uptake by purified vesicular a simple explanation is that the discrepancy arises
glutamate transporter (VGLUT), suggesting that from species differences. However, this is unlikely,
ketone bodies inhibit synaptic transmission with because ketogenic diet is known to reduce behav-
reduction of glutamate release via direct ketone ioral seizures in both rats (Appleton and DeVivo,
body-induced suppression of glutamate uptake 1974; Bough et al., 2002; Bough et al., 2006; Hori
into vesicles. Importantly, they also investigated et al., 1997; Zhao et al., 2004) and mice (Hartman
the behavioral effects of ketone bodies. Seizures et  al., 2008; Kwon et  al., 2008; Noh et  al., 2003;
in Wistar rats induced by intrahippocampal Rho et  al., 1999; Uhlemann and Neims, 1972).
4-aminopyridine were moderated by intrahip- The methods for applying ketone bodies varied in
pocampal 10 mM acetoacetate, both infused by these reports, including concentration of ketone
microdialysis. These results clearly show that bodies, time for application, and application path-
direct application of ketone bodies modulates way (perfusion or preincubation), and these might
synaptic transmission in hippocampal slices and contribute to interstudy variation. Using similar
reduces seizure activity in vivo (Juge et al., 2010). protocols for direct application of ketone bodies
In addition, Tanner et al. (2011) recorded single would be useful for finding common mechanisms.
channel activity from dentate granule neurons
after incubating acute hippocampal slices from Controlling Glucose and ATP Levels
C57BL/6 mice with 2 mM β-hydroxybutyrate for with Whole-Cell Patch Clamp
over 40 minutes. Preincubation with this ketone Another approach mimics the altered metabolism
body increased steady-state and stimulus- during ketogenic diet treatment using single-cell
elevated open probability of ATP-sensitive patch clamp recording. Fasting and ketogenic
potassium (KATP) channels, which contribute diet are thought to cause anticonvulsant effect by
to the slow afterhyperpolarization after action changing brain metabolism. It is well known that
potential bursts to modulate spontaneous firing, ketogenic diet causes a stable, mild hypoglyce-
suggesting that direct ketone body-mediated mia in humans (Huttenlocher, 1976) and rodents
opening of KATP channels in dentate granule (Bough et al., 2006). Interestingly, plasma glucose
neurons may act as a seizure gate in the hippo- level correlates with the antiepileptic effect of the
campus. Similar results were reported from neu- ketogenic diet (Mantis et  al., 2004), indicating
rons of the substantia nigra in coronal midbrain that extracellular glucose is mechanistically rel-
slices of rats and mice from same laboratory (Ma evant. Intracellular conditions are also thought to
et  al., 2007). Kim et  al. recorded from organo- be changed by ketogenic diet-altered metabolism,
typic hippocampal slices that were cultured with and the final product of brain energy metabolism
5 mM β-hydroxybutyrate and 1 mM acetoacetate is ATP. Several studies have reported that keto-
medium for 2 weeks (Kim et al., 2015). They used genic diet increases brain ATP levels in humans
Kcna1-null mice (C3HeB/FeJ background) lack- (Pan et  al., 1995)  and rodents (DeVivo et  al.,
ing voltage-gated potassium (Kv 1.1) channels, 1978; Nakazawa et  al., 1983; Nylen et  al., 2009).
which is thought to be a model for several types Therefore, intracellular ATP is a promising target
of epilepsy including human temporal lobe epi- for mimicking the ketogenic diet in vitro. From
lepsy. Extracellular multielectrode array record- these reports, reducing and increasing extracel-
ings showed spontaneous seizure-like events in lular glucose and intracellular ATP, respectively,
organotypic hippocampal slice cultures from might in combination reproduce mechanistically
Kcna1-null mice. The application of ketone bod- important ketogenic diet conditions in acute hip-
ies for 2 weeks attenuated the seizure-like events pocampal slices (Figure 21.1A2). As mentioned in
generated by the mutant tissue. They also applied section 2, it is easy to change extracellular solu-
5 mM β-hydroxybutyrate in vivo using subcuta- tion with an in vitro brain slice (including moder-
neously implanted osmotic minipumps to Kcna1- ately lowering glucose), but trickier to change the
null mice and reported that administration of intracellular milieu. The whole-cell patch clamp
191

Chapter 21: Ketogenic Diet in a Hippocampal Slice 191

Ketogenic diet

Glucose Ketone bodies

NADH AcetylCoA
ADP Pi
ETC
ATP synthase
Panx
NADH
TCA cycle
ATP ATP FADH2

Nucleotidase

Adenosine A1R
KATP

K+
Hyperpolarization

Antiseizure
FIGURE 21.2 Schematic of pannexin-1 channel-adenosine A1 receptor-KATP channel autocrine regulation in CA3
pyramidal neurons and relationship to ketogenic diet. Ketogenic diet increases ketone body levels and reduces glucose
levels. Ketone bodies might increase intracellular ATP production, and increased ATP is released to the extracellular
space due to reduced glucose-induced opening of pannexin-1 channels (panx). After breakdown to adenosine by
nucleotidases, activated adenosine A1 receptors (A1R) open KATP channels (KATP), which hyperpolarizes CA3 pyramidal
neurons. This hyperpolarization reduces neuronal hyperexcitability and causes the ketogenic diet’s antiseizure effects.
TCA cycle, tricarboxylic acid cycle; ETC, electron transport chain.

technique is one of the methods for recording a was sufficient or increased, reduced extracellular
single cell (Kawamura et al., 2004). This technique glucose opened pannexin-1 channels and released
allows physical exchange between the intracellu- intracellular ATP to the extracellular space.
lar fluid and the artificial intracellular solution in Released ATP was rapidly hydrolyzed to adenos-
the recording pipette, which allows us to modify ine, which activated adenosine A1 receptors with
the intracellular fluid composition of the recorded subsequent opening of KATP channels. Opening of
neuron experimentally (Figure 21.1A2, Table 21.1) these potassium channels caused hyperpolariza-
including elevating intracellular ATP. We found tion and reduced excitability. These results indi-
that increased intracellular ATP and reduced cate that mimicking the ketogenic diet condition
extracellular glucose caused hyperpolarization in with increased ATP and reduced glucose reduces
hippocampal neurons (Kawamura et  al., 2010). excitability in hippocampal CA3 pyramidal neu-
We recorded from CA3 pyramidal neurons with rons with autocrine modulation via adenosine
the whole-cell patch clamp technique in acute A1 receptors, and this might be a one of the key
hippocampal slices from control diet–fed SD rats mechanisms of the anticonvulsant effects of the
or C57BL/6 mice. Reduced extracellular glucose ketogenic diet in vivo (Figure 21.2). This approach
and increased intracellular ATP caused outward for reproducing ketogenic diet conditions in acute
current (or hyperpolarization when recording hippocampal slice is useful to elucidate detailed
membrane potential) in CA3 pyramidal neu- mechanisms within single neurons. However, it
rons. This outward current was dose-dependent mimics only two of the aspects of ketogenic diet
for both extracellular glucose and intracellular feeding. Further examinations using behavioral
ATP, and importantly it was found in both rats tests and recordings from in vivo ketogenic diet-
and mice. Pharmacological and genetic experi- fed animals are needed to link the results from this
ments demonstrated that when intracellular ATP approach to the effects of ketogenic diet feeding.
192

192 section III: Ketogenic Diet in the Laboratory

Acute Hippocampal Slices from Ketogenic diet (6:1 ratio) for 13–18 days. Excitability and
Diet–Fed Animals bicuculline-induced bursting were significantly
A third approach is possibly the most direct and inhibited by reduced extracellular glucose concen-
useful way for investigating mechanisms of keto- tration in slices from ketogenic diet–fed rats and
genic diet because it uses acute hippocampal slices mice but were not changed by reduced extracellu-
from ketogenic diet-fed animals (Figure 21.1b, lar glucose in slices from control diet–fed rodents.
Table 21.1). The question about this approach is Ketogenic diet–induced suppression of bicucul-
whether or not the intra- and extracellular milieu line-induced bursting was inhibited by adenos-
produced by ketogenic diet feeding can be main- ine A1 receptor antagonist and did not occur in
tained after making and incubating brain slices. slices from adenosine A1 receptor knockout mice.
However, four reports show that it can work suc- Antagonism of KATP channels or pannexin-1 chan-
cessfully. Stafstrom et al. reported that ketogenic nels inhibited the ketogenic diet-induced suppres-
diet (fat:[protein+carbohydrate] ratio of 4.8:1) sion of bicuculline-induced bursting. These results
induced antiseizure effects in acute hippocampal suggest that ketogenic diet causes antiseizure
slices from kainic acid–treated rats (Stafstrom et effects through a pannexin-1 channel-adenos-
al., 1999). They recorded fEPSP and PS from area ine A1 receptor-KATP channel autocrine pathway
CA1 from SD rats fed a ketogenic diet for 6–8 (Figure 21.2), the same pathway revealed by the
weeks. Synaptic transmission was not significantly whole-cell patch clamp technique for mimicking
different between slices from control diet–fed and ketogenic diet conditions as described previously
ketogenic diet–fed rats. The frequency of kainic (Kawamura et al., 2010).
acid–induced spontaneous seizures was signifi- These studies used successfully acute hippo-
cantly lower in slices from ketogenic diet–fed rats campal slices from ketogenic diet–fed rodents
than from control diet–fed rats. Similar results to elucidate the changing of neuronal activities
were reported by Simeone et al. using extracellular underlying this treatment. Interestingly, reducing
multielectrode array recordings in acute hippo- extracellular glucose concentration is thought be
campal slices from ketogenic diet–fed Knca1-null one of the most important points for reproduc-
mice (Simeone et al., 2014). The pathologic sei- ing the effects of ketogenic diet in this approach.
zure-like events generated in Knca1-null mice Synaptic transmission in hippocampal slices from
slice were diminished by ketogenic diet (6:1 ratio) ketogenic diet–fed rodents was not different from
treatment for 11–15 days. However, mossy fiber- that in slices from control diet–fed rodents when
CA3 dendritic field potential slopes and fiber vol- extracellular glucose concentration in artificial
ley amplitudes of mossy fiber are not significantly cerebrospinal fluid is standard in all three reports
different between slices from control diet–fed and (however, evidence is mounting that this stan-
ketogenic diet–fed Knca1-null mice (Simeone dard glucose concentration for acute brain slices
et al., 2014). Bough et al. recorded medial per- is higher than physiological brain glucose levels;
forant pathway-evoked fEPSPs from the dentate Kealy et al., 2013; Lowry and Fillenz, 2001; Shram
molecular layer in acute hippocampal slices from et  al., 1997). Reduced glucose reveals the differ-
SD rats fed a ketogenic diet (6:1 ratio) for over 20 ence between ketogenic diet– and control diet–
days (Bough et al., 2006). Reducing extracellu- fed animals in two of these studies (Bough et al.,
lar glucose concentration from 10 mM to 2 mM 2006; Kawamura et al., 2014), which parallels the
depressed the slope of fEPSP reversibly in slices finding that the anticonvulsant effect of the keto-
from control diet–fed rats, and this depression was genic diet is correlated with plasma glucose levels
inhibited in slices from ketogenic diet–fed rats. (Mantis et  al., 2004). Therefore it would be use-
The effects were lost after slices were incubated in ful to make extracellular glucose concentrations
10 mM glucose for over 3.5 hours. They concluded lower than standard to reproduce or maintain
that synaptic transmission in hippocampal slices effects of the ketogenic diet in acute hippocampal
from ketogenic diet–fed rats were more resistant slices.
to reduced glucose than slices from control diet–
fed rats (Bough et al., 2006). We also reported that CONCLUSIONS
ketogenic diet caused antiseizure effects in acute Here we illustrate three approaches for research-
hippocampal slices of rats and mice (Kawamura ing anticonvulsant mechanisms of ketogenic diets
et al., 2014). We recorded PS and GABA receptor by using electrophysiological recording from
blocker bicuculline-induced seizure-like bursting hippocampal slices. The usefulness of hippocam-
from the CA3 region in acute hippocampal slices pal slices is that it is easy to elucidate the details
from SD rats or C57BL/6 mice fed a ketogenic of neuronal modulation by ketogenic diet, as
193

Chapter 21: Ketogenic Diet in a Hippocampal Slice 193

shown in Figure 21.2. All three approaches have activity in children with therapy resistant epilepsy.
contributed to finding detailed potential mecha- Epilepsy Res 77, 134–140.
nisms underlying ketogenic diet effects including Hartman, A.L., Lyle, M., Rogawski, M.A., and Gasior,
VGLUT modulation, KATP opening, activation of M. (2008). Efficacy of the ketogenic diet in the
adenosine receptors, and ATP release from pan- 6-Hz seizure test. Epilepsia 49, 334–339.
Hori, A., Tandon, P., Holmes, G.L., and Stafstrom,
nexin channels. It is clear that electrophysiologi-
C.E. (1997). Ketogenic diet: effects on expression
cal recording from hippocampal slices is a good
of kindled seizures and behavior in adult rats.
tool for ketogenic diet research. However, all three Epilepsia 38, 750–758.
approaches need some steps for reproducing keto- Huttenlocher, P.R. (1976). Ketonemia and seizures:
genic diet effects in vitro such as ketone applica- metabolic and anticonvulsant effects of two keto-
tion, reduced extracellular glucose, and increased genic diets in childhood epilepsy. Pediatr Res 10,
intracellular ATP. These methods are not direct 536–540.
measurements of the in vivo anticonvulsant effects Juge, N., Gray, J.A., Omote, H., Miyaji, T., Inoue, T.,
of ketogenic diet. A combination of both in vivo Hara, C., Uneyama, H., Edwards, R.H., Nicoll,
and in vitro recordings should provide further R.A., and Moriyama, Y. (2010). Metabolic con-
explanations concerning the key anticonvulsant trol of vesicular glutamate transport and release.
mechanisms underlying ketogenic diet treatment. Neuron 68, 99–112.
Kawamura, M., Gachet, C., Inoue, K., and Kato, F.
(2004). Direct excitation of inhibitory interneu-
AC K N OW L E D G M E N T
rons by extracellular ATP mediated by P2Y1
We thank Dr. David N. Ruskin for comments on receptors in the hippocampal slice. J Neurosci 24,
this manuscript. We acknowledge the support of 10835–10845.
the Jikei University Research Fund to MK. Kawamura, M., Jr., Ruskin, D.N., and Masino, S.A.
(2010). Metabolic autocrine regulation of neurons
REFERENCES involves cooperation among pannexin hemichan-
Abe, H., and Ogata, N. (1981). Effects of penicillin on nels, adenosine receptors, and KATP channels. J
electrical activities of neurons in guinea-pig hip- Neurosci 30, 3886–3895.
pocampal slies. Jpn J Pharmacol 31, 661–675. Kawamura, M., Jr., Ruskin, D.N., Geiger, J.D., Boison,
Appleton, D.B., and DeVivo, D.C. (1974). An ani- D., and Masino, S.A. (2014). Ketogenic diet sensi-
mal model for the ketogenic diet. Epilepsia 15, tizes glucose control of hippocampal excitability. J
211–227. Lipid Res 55, 2254–2260.
Bough, K.J., Gudi, K., Han, F.T., Rathod, A.H., and Kealy, J., Bennett, R., and Lowry, J.P. (2013).
Eagles, D.A. (2002). An anticonvulsant profile Simultaneous recording of hippocampal oxygen
of the ketogenic diet in the rat. Epilepsy Res 50, and glucose in real time using constant potential
313–325. amperometry in the freely-moving rat. J Neurosci
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., Methods 215, 110–120.
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Kim, D.Y., Simeone, K.A., Simeone, T.A., Pandya, J.D.,
Y., Geiger, J.D., and Dingledine, R.J. (2006). Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G.,
Mitochondrial biogenesis in the anticonvulsant and Rho, J.M. (2015). Ketone bodies mediate anti-
mechanism of the ketogenic diet. Ann Neurol 60, seizure effects through mitochondrial permeabil-
223–235. ity transition. Ann Neurol 78, 77–87.
Congar, P., Gaiarsa, J.L., Popovici, T., Ben-Ari, Y., and Kimura, R., Ma, L.Y., Wu, C., Turner, D., Shen, J.X.,
Crepel, V. (2000). Permanent reduction of seizure Ellsworth, K., Wakui, M., Maalouf, M., and Wu,
threshold in post-ischemic CA3 pyramidal neu- J. (2012). Acute exposure to the mitochondrial
rons. J Neurophysiol 83, 2040–2046. complex I  toxin rotenone impairs synaptic long-
DeVivo, D.C., Leckie, M.P., Ferrendelli, J.S., and term potentiation in rat hippocampal slices. CNS
McDougal, D.B., Jr. (1978). Chronic ketosis and Neurosci Ther 18, 641–646.
cerebral metabolism. Ann Neurol 3, 331–337. Knowles, W.D., Traub, R.D., and Strowbridge, B.W.
Dulla, C.G., Dobelis, P., Pearson, T., Frenguelli, B.G., (1987). The initiation and spread of epilepti-
Staley, K.J., and Masino, S.A. (2005). Adenosine form bursts in the in vitro hippocampal slice.
and ATP link PCO2 to cortical excitability via pH. Neuroscience 21, 441–455.
Neuron 48, 1011–1023. Kohling, R., Vreugdenhil, M., Bracci, E., and Jefferys,
Engel, J., Jr. (2001). Mesial temporal lobe epilepsy: what J.G. (2000). Ictal epileptiform activity is facilitated
have we learned? Neuroscientist 7, 340–352. by hippocampal GABAA receptor-mediated oscil-
Gastaut, H.  (1973). Dictionary of Epilepsy:  Part  1. lations. J Neurosci 20, 6820–6829.
Definitions. (Geneva: World Health Organization). Kojima, H., Kowada, M., and Katsuta, Y. (1989).
Hallbook, T., Kohler, S., Rosen, I., and Lundgren, J. Cellular substrates for epileptiform activ-
(2007). Effects of ketogenic diet on epileptiform ity induced by repeated tetanic stimulation in
194

194 section III: Ketogenic Diet in the Laboratory

hippocampal slices. Neurol Med Chir (Tokyo) acid-induced hippocampal cell death in the male
29, 1–5. ICR mice. Epilepsy Res 53, 119–128.
Koranda, J.L., Ruskin, D.N., Masino, S.A., and Blaise, Nylen, K., Velazquez, J.L., Sayed, V., Gibson, K.M.,
J.H. (2011). A ketogenic diet reduces long-term Burnham, W.M., and Snead, O.C., 3rd (2009). The
potentiation in the dentate gyrus of freely behav- effects of a ketogenic diet on ATP concentrations
ing rats. J Neurophysiol 106, 662–666. and the number of hippocampal mitochondria in
Kwon, Y.S., Jeong, S.W., Kim, D.W., Choi, E.S., and Aldh5a1(−/−) mice. Biochim Biophys Acta 1790,
Son, B.K. (2008). Effects of the ketogenic diet on 208–212.
neurogenesis after kainic acid-induced seizures in Paglioli, E., Palmini, A., Portuguez, M., Azambuja,
mice. Epilepsy Res 78, 186–194. N., da Costa, J.C., da Silva Filho, H.F., Martinez,
Li, T., Ren, G., Lusardi, T., Wilz, A., Lan, J.Q., Iwasato, J.V., and Hoeffel, J.R. (2006). Seizure and memory
T., Itohara, S., Simon, R.P., and Boison, D. (2008). outcome following temporal lobe surgery: selec-
Adenosine kinase is a target for the prediction and tive compared with nonselective approaches for
prevention of epileptogenesis in mice. J Clin Invest hippocampal sclerosis. J Neurosurg 104, 70–78.
118, 571–582. Pan, W.J., Osmanovic, S.S., and Shefner, S.A. (1995).
Lin, L., Chen, G., Xie, K., Zaia, K.A., Zhang, S., and Characterization of the adenosine A1 receptor-
Tsien, J.Z. (2006). Large-scale neural ensemble activated potassium current in rat locus ceruleus
recording in the brains of freely behaving mice. J neurons. J Pharmacol Exp Ther 273, 537–544.
Neurosci Methods 155, 28–38. Quarato, P.P., Di Gennaro, G., Mascia, A., Grammaldo,
Lowry, J.P., and Fillenz, M. (2001). Real-time moni- L.G., Meldolesi, G.N., Picardi, A., Giampa, T.,
toring of brain energy metabolism in vivo using Falco, C., Sebastiano, F., Onorati, P., et al. (2005).
microelectrochemical sensors: the effects of anes- Temporal lobe epilepsy surgery: different surgical
thesia. Bioelectrochemistry 54, 39–47. strategies after a non-invasive diagnostic protocol.
Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic J Neurol Neurosurg Psychiatry 76, 815–824.
diet metabolites reduce firing in central neurons Ray, A., and Wyllie, E. (2005). Treatment options and
by opening K(ATP) channels. J Neurosci 27, paradigms in childhood temporal lobe epilepsy.
3618–3625. Expert Rev Neurother 5, 785-801.
Mantis, J.G., Centeno, N.A., Todorova, M.T., McGowan, Rho, J.M., Kim, D.W., Robbins, C.A., Anderson, G.D.,
R., and Seyfried, T.N. (2004). Management of multi- and Schwartzkroin, P.A. (1999). Age-dependent
factorial idiopathic epilepsy in EL mice with caloric differences in flurothyl seizure sensitivity in mice
restriction and the ketogenic diet:  role of glucose treated with a ketogenic diet. Epilepsy Res 37,
and ketone bodies. Nutr Metab (Lond) 1, 11. 233–240.
Masino, S.A., and Dunwiddie, T.V. (1999). Sakmann, B., Edwards, F., Konnerth, A., and Takahashi,
Temperature-dependent modulation of excitatory T. (1989). Patch clamp techniques used for study-
transmission in hippocampal slices is mediated by ing synaptic transmission in slices of mammalian
extracellular adenosine. J Neurosci 19, 1932–1939. brain. Q J Exp Physiol 74, 1107–1118.
Masino, S.A., Kawamura, M., Wasser, C.A., Pomeroy, Samoilova, M., Weisspapir, M., Abdelmalik, P.,
L.T., and Ruskin, D.N. (2009). Adenosine, keto- Velumian, A.A., and Carlen, P.L. (2010). Chronic
genic diet and epilepsy: the emerging therapeutic in vitro ketosis is neuroprotective but not anti-
relationship between metabolism and brain activ- convulsant. J Neurochem 113, 826–835.
ity. Curr Neuropharmacol 7, 257–268. Sayin, Rutecki, P., and Sutula, T. (1999). NMDA-
Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin, dependent currents in granule cells of the dentate
D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and gyrus contribute to induction but not permanence
Boison, D. (2011). A ketogenic diet suppresses sei- of kindling. J Neurophysiol 81, 564–574.
zures in mice through adenosine A(1) receptors. J Schramm, J., Kral, T., Grunwald, T., and Blumcke, I.
Clin Invest 121, 2679–2683. (2001). Surgical treatment for neocortical tempo-
Masino, S.A., and Rho, J.M. (2012). Mechanisms of ral lobe epilepsy: clinical and surgical aspects and
ketogenic diet action. In Jasper’s Basic Mechanisms seizure outcome. J Neurosurg 94, 33–42.
of the Epilepsies, J.L. Noebels, M. Avoli, M.A. Shram, N.F., Netchiporouk, L.I., Martelet, C., Jaffrezic-
Rogawski, R.W. Olsen, and A.V. Delgado-Escueta, Renault, N., and Cespuglio, R. (1997). Brain glu-
eds. (Bethesda: National Center for Biotechnology cose:  voltammetric determination in normal and
Information), pp. 1001–1022. hyperglycaemic rats using a glucose microsensor.
Nakazawa, M., Kodama, S., and Matsuo, T. (1983). Neuroreport 8, 1109–1112.
Effects of ketogenic diet on electroconvulsive Simeone, T.A., Samson, K.K., Matthews, S.A., and
threshold and brain contents of adenosine nucleo- Simeone, K.A. (2014). In vivo ketogenic diet treat-
tides. Brain Dev 5, 375–380. ment attenuates pathologic sharp waves and high
Noh, H.S., Kim, Y.S., Lee, H.P., Chung, K.M., Kim, frequency oscillations in in vitro hippocampal
D.W., Kang, S.S., Cho, G.J., and Choi, W.S. (2003). slices from epileptic Kv 1.1alpha knockout mice.
The protective effect of a ketogenic diet on kainic Epilepsia 55, e44–49.
195

Chapter 21: Ketogenic Diet in a Hippocampal Slice 195

Smith, B.N., and Dudek, F.E. (2001). Short- and selective amygdalohippocampectomy. J Neurosurg
long-term changes in CA1 network excitability 108, 517–524.
after kainate treatment in rats. J Neurophysiol Thio, L.L., Wong, M., and Yamada, K.A. (2000). Ketone
85, 1–9. bodies do not directly alter excitatory or inhibitory
Stafstrom, C.E., Ockuly, J.C., Murphree, L., Valley, hippocampal synaptic transmission. Neurology
M.T., Roopra, A., and Sutula, T.P. (2009). 54, 325–331.
Anticonvulsant and antiepileptic actions of 2- Thom, M. (2009). Hippocampal sclerosis:  progress
deoxy-D-glucose in epilepsy models. Ann Neurol since Sommer. Brain Pathol 19, 565–572.
65, 435–447. Uhlemann, E.R., and Neims, A.H. (1972).
Stafstrom, C.E., Wang, C., and Jensen, F.E. (1999). Anticonvulsant properties of the ketogenic diet in
Electrophysiological observations in hippocam- mice. J Pharmacol Exp Ther 180, 231–238.
pal slices from rats treated with the ketogenic diet. Watson, C. (2003). Hippocampal sclerosis and the syn-
Dev Neurosci 21, 393–399. drome of medial temporal lobe epilepsy. Expert
Stewart, C., and Reid, I. (1993). Electroconvulsive Rev Neurother 3, 821–828.
stimulation and synaptic plasticity in the rat. Brain Wiebe, S., Blume, W.T., Girvin, J.P., and Eliasziw, M.
Res 620, 139–141. (2001). A randomized, controlled trial of surgery for
Takano, T., He, W., Han, X., Wang, F., Xu, Q., Wang, temporal-lobe epilepsy. N Engl J Med 345, 311–318.
X., Oberheim Bush, N.A., Cruz, N., Dienel, G.A., Wiebe, S., and Jette, N. (2012). Pharmacoresistance
and Nedergaard, M. (2014). Rapid manifestation and the role of surgery in difficult to treat epilepsy.
of reactive astrogliosis in acute hippocampal brain Nat Rev Neurol 8, 669–677.
slices. Glia 62, 78–95. Wilder, R.M. (1921). The effects of ketonemia on the
Tanner, G.R., Lutas, A., Martinez-Francois, J.R., course of epilepsy. Mayo Clinic Proc 2, 307–308.
and Yellen, G. (2011). Single K ATP channel Youssef, F.F. (2015). Ketone bodies attenuate excito-
opening in response to action potential firing in toxic cell injury in the rat hippocampal slice under
mouse dentate granule neurons. J Neurosci 31, conditions of reduced glucose availability. Neurol
8689–8696. Res 37, 211–216.
Tanriverdi, T., Olivier, A., Poulin, N., Andermann, Zhao, Q., Stafstrom, C.E., Fu, D.D., Hu, Y., and Holmes,
F., and Dubeau, F. (2008). Long-term seizure G.L. (2004). Detrimental effects of the ketogenic
outcome after mesial temporal lobe epilepsy sur- diet on cognitive function in rats. Pediatr Res 55,
gery:  corticalamygdalohippocampectomy versus 498–506.
196

22
Metabolic Therapy and Pain
D AV I D N. R U S K I N , P H D

INTRODUCTION to prescribe or even fill prescriptions for opioids


Across all cultures, patients with chronic pain have (Glajchen, 2001; Kim et  al., 2000). The NSAIDs,
among the lowest reported quality‐of‐life scores of conversely, have little to no addiction liability
any medical condition and have comorbid mood but are poorly effective against neuropathic pain
or anxiety disorders more frequently than the nor- (Moore et al., 2015) and can have serious cardiovas-
mal population (Becker et al., 1997; Gureje et al., cular and gastrointestinal side effects (Brune and
1998). In addition, pain has a notable negative Patrignani, 2015). Thus, effective and nonaddictive
impact on economies, with high direct costs (e.g., treatments with few side effects are needed. With
physician visits, physiotherapy, medication) and, the understanding that chronic pain syndromes
more strikingly, lowered productivity (e.g., lost likely involve derangement of homeostasis, there
work days, reduced performance at work) charac- is an increased interest in applying treatments
terized in numerous countries around the world that aim to normalize metabolism, namely, meta-
(Phillips, 2006). bolic therapies. Many metabolic therapies involve
Much chronic pain falls into two distinct cat- dietary changes. Whereas some dietary metabolic
egories: inflammatory pain and neuropathic pain. therapies narrowly address specific genetic disor-
Chronic inflammation is typically accompanied ders that include pain as a symptom (Roe et  al.,
by pain due to the release of prostaglandins and 2008), this survey of the literature focuses on
their consequent sensitization of sensory neurons more wholesale dietary changes, some long estab-
(Mense, 1983). Common forms of inflammatory lished, and their effects on common types of acute,
pain are rheumatoid arthritis, chronic inflamma- inflammatory, and neuropathic pain.
tory bowel disease, pancreatitis, back pain, and
some cancer pain. In contrast, neuropathic pain FA S T I N G
is caused by lesion or dysfunction of nervous tis- One type of dietary metabolic therapy with a his-
sue; common forms are associated with diabetes, tory of use in pain is fasting, or a total or near
herpes zoster infection (shingles), stroke, mul- total (<500 kcal/d) reduction in calorie intake
tiple sclerosis, cancer chemotherapy, and both (Michalsen, 2010). Fasting has been practiced for
HIV infection and antiretroviral HIV treatment. thousands of years in many cultures as a treatment
Neuropathic pain can also be idiopathic (Lauria for various disorders (as well as for religious rea-
et al., 2014). sons). Biochemically, fasting results in lowered
Severe pain is typically treated with medica- levels of circulating glucose, as intake stops and
tion, most commonly cyclooxygenase inhibitors glycogen stores are metabolized, and in greatly
such as nonsteroidal anti‐inflammatory drugs enhanced circulating levels of molecules known
(NSAIDs) and opioids. Unfortunately, these drugs as ketone bodies, which are produced by the liver
can present serious dilemmas for both patients and from fatty acids and can be used as fuel for the tri-
healthcare providers. Many patients view opioids carboxylic acid cycle in the relative absence of glu-
as undesirable because of their fear of addiction cose. Subjects report a counterintuitive reduction
or their experiences with cognitive or other side in hunger after a few days, as use of ketone bod-
effects (Welshman, 2005); prescription of opioids ies becomes maximal (Michalsen, 2010). Fasting
is contraindicated specifically in patients with pre- can be used as a treatment for obesity and meta-
existing substance abuse issues. In addition, given bolic syndrome, and is anticonvulsant in epileptic
present levels of prescription drug abuse (Allread patients (and so spawned the ketogenic diet, see
and Paul, 2014; Manchikanti, 2006), there is an below). In the context of pain, some of the earliest
appropriate hesistancy of healthcare providers work concerned rheumatoid arthritis (Eisenberg,
197

Chapter 22: Metabolic Therapy and Pain 197

1956). Seven days of fasting significantly alleviated EOD fasting generally causes weight loss clinically
joint pain and stiffness in rheumatoid arthritis suf- (Varady et al., 2015), there is minimal body weight
ferers (Kroker et al., 1984; Michalsen et al., 2005; change in many rodent studies (Tajes et al., 2010;
Sköldstam et  al., 1979), apparently through anti‐ Tikoo et al., 2007). Therefore, studies of EOD fast-
inflammatory actions including modified neutro- ing in animals could dissociate weight loss from
phil function (Hafström et  al., 1988; Udén et  al., antinociceptive effects, essentially impossible in
1983). Eight fasting days significantly improved clinical studies of prolonged standard fasting.
pain scores in osteoarthritis, an effect that lasted Animal studies suggest that subjects who do not
weeks after the end of fasting (Schmidt et  al., need or wish to lose weight may limit this effect
2010). One to 2 weeks of fasting significantly of EOD fasting by exercising (Sakamoto and
alleviated pain in the pain disorder fibromyalgia Grunewald, 1987).
(Michalsen et  al., 2005; Michalsen et  al., 2013).
Given the commonness of mood and anxiety dis- CALORIE RESTRICTION
orders in chronic pain (Gureje et al., 2008), fasting Those who wish for pain alleviation but who can-
has the additional benefit of enhancing mood after not tolerate long periods of time without food
several days, an effect not limited to pain sufferers might consider instead calorie restriction (CR).
(Michalsen, 2010). Typically, this procedure limits total daily calorie
These clinical antinociceptive effects are sup- intake to 60% of normal (not less than ~900 kcal/d).
ported by animal experiments. In a rat model of There is abundant experimental evidence for anti-
postischemic pain, allodynia (i.e., the perception nociceptive effects of CR on acute pain (Wideman
of normally nonpainful stimuli as painful) to cold et  al., 1996), inflammatory pain (Hargraves and
and tactile stimulation in the ischemic paw was Hentall, 2005), and heat hyperalgesia in arthri-
significantly reduced if the animal had been fasted tis models (Jurcovicova et  al., 2001). Clinically,
for 18 hours prior to ischemia. (Given the higher CR (applied for 12 weeks to 1 year, and typically
metabolic rate of rats, this time is comparable to a combined with exercise) in overweight patients
longer fast in a human.) In addition, the effect of reduced back pain (thought to involve inflamma-
the fast was totally reversed by glucose injection tion; Roffey et al., 2011), knee pain (White et al.,
during the ischemic event, highlighting the impor- 2015), overall body pain (Landaeta‐Díaz et  al.,
tance of lowered glucose (Ross‐Huot et al., 2011). 2013), osteoarthritic pain (de Luis et al., 2012), and
The anti‐inflammatory effects of fasting seem to prediabetic neuropathic pain (Smith et al., 2006).
be mediated by ketosis (a state of elevated ketone All these benefits could be secondary to weight
bodies) which limits oxidative stress and produc- loss, which would lessen physical wear on joints
tion of free radicals and reactive oxygen species and backs (as suggested in most of these papers),
(Veech, 2014). though this explanation does not appear to apply
Unfortunately, the clinical benefits of prolonged for the positive outcome in diabetic neuropathy, in
fasting for rheumatoid arthritis are not long lasting which pain reduction related to actual reinnerva-
(unlike for osteoarthritis; Sköldstam et al., 1979), tion of the skin (Smith et al., 2006). Testing of CR
and fasting is necessarily a time‐limited under- treatment in fibromyalgia (see below), which is not
taking, especially for subjects who do not have limited to overweight individuals, could dissociate
extra adipose tissue to burn through over days to weight loss from CR‐related pain relief.
weeks. An alternative procedure is intermittent Apart from possible weight loss effects, it is
fasting, usually taking the form of every‐other‐ unclear whether ketosis contributes to this anti-
day (EOD) fasting, which can be implemented for nociception, as none of these clinical studies
extended periods. Notably, in humans a 24-h fast measured blood or urinary ketones. However,
is sufficient to limit activation of the NLR fam- improved glycemic control might be involved. As
ily, pyrin domain containing 3 (NLRP3) inflam- already noted, low circulating glucose is neces-
masome activation (Traba et  al., 2015), which is sary for fasting amelioration of postischemic pain
associated with hyperalgesia (Bullón et al., 2015); (Ross‐Huot et al., 2011). A meta‐analysis of stud-
therefore EOD fasting might be beneficial in rheu- ies of diabetic neuropathy (including almost 8,000
matoid arthritis. This effect on the NLRP3 inflam- patients) concluded that more aggressive glucose
masome seems to be mediated by ketone bodies control was associated with a lower incidence of
(Youm et al., 2015). Studies in humans (Rothschild neuropathic pain and improved electrophysiologi-
et al., 2014) and rodents (Poon et al., 2006; Tajes cal measures (Callaghan et  al., 2012). In animal
et  al., 2010; Tikoo et  al., 2007)  have shown posi- studies, glycemic control partially alleviated dia-
tive effects of EOD fasting on various aspects of betic neuropathy (Yorek et  al., 2014). Also, par-
health, but none have addressed pain. Although tial glycolytic inhibition by 2‐deoxy‐D‐glucose
198

198 section III: Ketogenic Diet in the Laboratory

decreases sensation of acute pain (Bodnar et  al., restore balanced metabolism through an AMPK/
1979) by central mechanisms (Bodnar et al., 1981). glycolysis pathway, one that can be engaged phar-
This action does not involve endogenous opioids, macologically with AMPK activators.
but seems to involve the ubiquitous neuromodu-
lator adenosine, which has broad analgesic effects K E TO G E N I C   D I E T
(Sawynok, 2015; Yamaoka et  al., 2013)  and is Those who wish for pain alleviation but who can
released by 2‐deoxy‐D‐glucose or mildly lowered tolerate neither CR nor fasting might consider the
glucose (Kawamura et al., 2014; Zhao et al., 1997). ketogenic diet (KD). This diet was based on the
Notably, fibromyalgia has been associated with hypothesis that the beneficial effects of fasting in
disordered glycolysis (Eisinger et al., 1994). One of epilepsy were due to the elimination of carbohy-
the regulators of glycolysis is the enzyme adenosine drates (Wilder, 1921a, 1921b); thus, the KD mini-
monophosphate‐activated protein kinase (AMPK; mizes carbohydrates and replaces the lost calories
Carling, 2005), which has also been linked to sen- with fats, and is in fact strongly anticonvulsant
sitization of nociceptors in pain states (Tillu et al., (Dressler et al., 2015; Hallböök et al., 2015; Megaw
2012). Cells from fibromyalgia sufferers displayed and Wilmshurst, 2015; Walker and Said, 2015) and
underactive AMPK and a related range of meta- mounting evidence suggests that it is antiepilepto-
bolic impairments (Alcocer‐Gómez et  al., 2015; genic (Gama et al., 2015; Hu et al., 2011; Jiang et al.,
Bullón et al., 2016). These problems were reversed 2012; Lusardi et al., 2015; Muller‐Schwarze et al.,
by incubation with serum from CR‐treated mice 1999; Neal et  al., 2008; Su et  al., 2000; Todorova
(Alcocer‐Gómez et  al., 2015). Pharmacological et  al., 2000; Xu et  al., 2006). The KDs result in
blockade of AMPK produced hyperalgesia in mice ketosis and moderately lowered glucose like fast-
that was reversed by CR (Bullón et  al., 2015). In ing or sufficiently strong CR. Our animal stud-
fibromyalgia sufferers, pharmacological activation ies showed that KDs have a clear antinociceptive
of AMPK reversed metabolic impairments and sig- effect in acute thermal pain. This effect was pres-
nificantly improved a range of symptoms includ- ent in both male and female rats (Figure 22.1) and
ing pain (Bullón et  al., 2015). Thus CR seems to was long‐lasting (Figure 22.2). Notably, the effect

Plate temperature (°C) Plate temperature (°C)


46 47 48 49 50 51 46 47 48 49 50 51

60 60
Latency to nocifensive behavior (s)

Males Females
50 50

40 40

30 30

20 20
Control diet
10 Ketogenic diet 10

0 0
14 15 16 17 18 19 14 15 16 17 18 19
Days on diet treatment Days on diet treatment

FIGURE 22.1 Sprague‐Dawley rats on a control diet or a 78% fat KD were challenged with a temperature‐response
curve for nocifensive behavior. One temperature was tested per day. Top and bottom X‐axes illustrate hotplate
temperature and days on diet, respectively. Dietary treatments began at 3–3½ weeks of age. Y‐axis indicates latency
to avoidance behavior, e.g. paw lifting, licking, or fluttering. All tests had a 60-s ceiling. At left, KD treatment in young
male rats causes heat hypoalgesia at moderate 49°C, 50°C, and 51°C temperatures. Diet F = 5.7, p = .19; Temperature
F = 158.4, p < .001; Diet‐x‐Temperature interaction F = 4.0, p < .001; n = 18–20. Neuman‐Keuls comparisons at each
temperature: *p < .05, ** p < .01, ***p < .001. Adapted from Ruskin et al. (2013). At right, KD treatment in young female
rats also causes heat hypoalgesia at the same temperatures. Diet F = 28.2, p < .001; Temperature F = 134.2, p < .001; Diet ×
Temperature interaction F = 9.6, p < .001; n = 12–16. All points are mean ± standard error. Neuman‐Keuls comparisons
at each temperature: ***p < .001. Unpublished data.
199

Chapter 22: Metabolic Therapy and Pain 199

40 is poorly effective against neuropathic pain gen-


Control diet
erally, or rather has type‐specific positive effects.
Latency to nocifensive response (s)
Ketogenic diet
Regardless, abundant evidence shows that KDs are
30 clinically beneficial in diabetes (e.g. Adams, 1931;
Mobbs et al., 2013).
Antinociceptive effects of the KD might
20
be strongest in inflammatory pain, as this
diet is anti‐inflammatory (Pérez‐Guisado and
10
Muñoz‐Serrano, 2011; Ruskin et al., 2009; Sullivan
et  al., 2004; Tendler et  al., 2007), and this effect
might be due largely to ketosis, as reviewed earlier
0 for fasting. Such findings might appear contradic-
0 5 10 15 20 25 30 tory to much published literature showing that
Days on diet treatment high‐fat diets promote inflammation (Johnson
and Makowski, 2015); however, this literature
FIGURE  22.2 The development of KD effects on
refers to diets such as the so‐called Western diet,
pain over time. Male Sprague‐Dawley rats were placed
high in fat but not low in carbohydrates. The
on a control diet or a 78% fat KD at 3–3½ weeks of age.
metabolic response to dietary fats differs greatly
Temperature‐response curves as in Figure  22.1 were
depending on the presence of carbohydrates:  the
used; for clarity of the progressive effect, data from
high‐fat‐plus‐carbohydrate diet promotes fat stor-
only the highest (51°C) tested temperature are shown.
age, whereas the high fat, low‐carbohydrate diet
Each point represents a different squad of rats. Several
promotes fat metabolism.
days of treatment are necessary for the KD‐related heat
Apart from anti‐inflammatory effects, some
hypoalgesia to emerge. T‐tests at each length of treatment
antinociceptive effects of the KD might relate
were protected for multiple comparisons. (*)  p  =  .056,
directly to its high fat content. The receptor
**p < .01, ***p < .001. Adapted from Ruskin et al. (2009)
GPR40 binds and is activated by free medium‐
and Ruskin et al. (2013).
and long‐chain fatty acids, and thus is sometimes
called free fatty acid receptor 1 (Covington et al.,
2006). Although this receptor may be considered
had a slower onset than ketosis and lowered blood a nutrient sensor, it is expressed in the central
glucose (Figure 22.2), both of which were already nervous system and turns out to affect nocicep-
significant at 2 days of KD feeding (Ruskin et al., tion. In animal testing, synthetic GPR40 agonists
2013), suggesting that these biochemical changes and natural ligands injected intrathecally or intra-
are not directly involved. cerebroventricularly reduce inflammatory and
Clinically, the KD was used very early as a treat- neuropathic pain in numerous models (Harada
ment for pain (Maggioni et al., 2011). For instance, et  al., 2014; Karki et  al., 2015; Nakamoto et  al.,
in one study KD feeding improved or completely 2013; Nakamoto et  al., 2015). There are elec-
controlled migraine in 39 of 50 patients (Baborka, trophysiological correlates of these behavioral
1930). More recent results support its antimi- effects (Karki et  al., 2015). Conversely, GPR40
graine efficacy (Di Lorenzo et al., 2013, 2015). The knockout augments the arthritic phenotype in
KD treatment was effective in treating inflamma- an osteoarthritis model (Monfoulet et  al., 2015),
tory bowel syndrome, including pain symptoms although pain behavior was not studied in this
(Austin et  al., 2009). Overall body pain was sig- report. In “Western” high‐fat diets, antinocicep-
nificantly improved in one study of overweight tive effects of GPR40 activation might be unable
subjects (Guldbrand et  al., 2014)  and was very to compete against the pro-inflammatory nature
near significance in another (Yancy et  al., 2009); of these diets.
quality of life was significantly improved in both Regarding epilepsy, there is some controversy
studies. One of these studies specifically involved over whether the KD is more effective when com-
overweight diabetics (Guldbrand et al., 2014), but bined with CR (Bough et  al., 1999b; Raffo et  al.,
did not parse the types of pain involved, so it is 2008). Certainly, KD plus CR will lead to stronger
unknown whether KD feeding alleviated neuro- ketosis and a more pronounced lowering of circu-
pathic pain in these patients. We found no benefi- lating glucose. If the KD’s antinociceptive effects
cial effect of KD in models of nerve constriction are primarily mediated by fatty acids, however,
and chemotherapeutic neuropathic pain (Masino the addition of CR should add little benefit to this
and Ruskin, 2013); it remains unclear whether KD outcome.
200

200 section III: Ketogenic Diet in the Laboratory

P O LY U N S AT U R AT E D augment voltage‐activated A‐type (Kv1.4) potas-


FAT T Y   A C I D S sium channels (Tigerholm et  al., 2012; Xu et  al.,
Some have suggested that many of the beneficial 2008), which could reduce hyperexcitability of
effects of the KD in epilepsy and general health are sensitized neural tissue. Certain PUFAs also block
due to enhanced dietary levels of polyunsaturated pain due to activation of the heat‐sensing transient
fatty acids (PUFAs; (Fraser et al., 2003; Fuehrlein receptor potential vanilloid subtype I  (the “cap-
et al., 2004; Paoli et al., 2015; Taha et al., 2005; Xu saicin receptor”), an inflammation‐related recep-
et  al., 2008), based on anticonvulsant effects of tor expressed by peripheral nociceptive neurons
PUFAs in some studies (Taha et al., 2009; Voskuyl (Matta et al., 2007).
et al., 1998; Yehuda et al., 1994). These effects are
controversial (Dahlin et  al., 2007; Dupuis et  al., OT H E R M E C H A N I S M S
2015; Willis et  al., 2009). Nevertheless, whereas Ketone bodies, augmented in fasting, CR, and
saturated fatty acids bind to GPR40, PUFAs addi- KD feeding, might act on pain through mul-
tionally act on other molecular targets that could tiple mechanisms. As already noted, a ketone
modulate pain (see below), and PUFAs and their body–based metabolism limits inflammation.
epoxide metabolites indeed help in inflammatory A  ketone body–based metabolism also produces
and neuropathic pain (Wagner et al., 2014). In ani- ATP more efficiently than one based on glucose
mal studies, for instance, elimination of dietary (Veech, 2004). Thus, ketone body treatment leads
PUFAs promotes the neuropathic pain phenotype to elevated levels of ATP and phosphocreatine
after nerve injury (Martin and Avendaño, 2009). (for instance, Deng‐Bryant et al., 2011; Kim et al.,
Conversely, addition of fish oil (rich in PUFAs) 2010; Tieu et al., 2003), which might allow restora-
to the diet reverses diabetic neuropathy includ- tion of energy homeostasis in distressed and pain-
ing derangement of pain sensation (Coppey et al., ful tissue. Besides effects on energy, ketone body
2012; Coppey et  al., 2015; Redivo et  al., 2015). treatment modulates glutamate and its recep-
Clinically, PUFA efficacy is good in inflammatory tors (Chmiel‐Perzyńska et  al., 2011; Donevan
pain (Goldberg and Katz, 2007; Lee et al., 2012) but et  al., 2003; Juge et  al., 2010; Lund et  al., 2009;
not osteoarthritis (Boe and Vangsness, 2015). Żarnowsky et al., 2012), γ‐aminobutyric acid and
Some researchers believe that the various its receptors (Erecińska et  al., 1996; Yang et  al.,
health benefits of PUFA supplements are due to 2007), potassium channels (Giménez‐Cassina
the high ratio of ω‐3 to ω‐6 PUFAs, two types of et  al., 2012; Ma et  al., 2007; Tanner et  al., 2011),
PUFA that differ in the location of double bonds and the receptor PUMA‐G (Taggart et al., 2005) in
in their carbon backbones. Fish oil has a high ω‐3/ a manner often leading to reduced excitability
ω‐6 ratio. Thus, animal studies have specifically that could limit activity in sensitized nociceptive
enriched diets with ω‐3 PUFAs and found that this systems. Multiple mechanisms acting in concert
reduces acute pain and neuropathic pain after spi- might be especially effective in normalizing the
nal cord contusion (Escudero et al., 2015; Figueroa deranged metabolism related to inflammation and
et  al., 2013). An additional benefit was that ω‐3 sensitized neural tissue. Future work on ketone
PUFAs reduced the development of tolerance to bodies will surely be aided by the availability of
morphine (Escudero et  al., 2015). On the other ketone esters (Kashiwaya et  al., 2010; Viggiano
hand, there is other evidence that ω‐3 PUFAs pro- et al., 2015), which can be taken orally and are rap-
mote neuropathic pain (Pérez et al., 2005). In clini- idly metabolized to ketone bodies, thus bypassing
cal studies, ω‐3 PUFA supplementation improved the need for wholesale dietary changes.
pain and quality of life in chronic headache suffer- On the other hand, regarding the KD, some
ers (Ramsden et al., 2013; Ramsden et al., 2015). have concluded that ketone bodies are not
How might PUFAs influence pain? PUFAs are directly involved in its anticonvulsant effect
agonists of the peroxisome proliferator‐activated (Bough et  al., 1999a, 2000; Harney et  al., 2002;
receptors (PPARs), another type of nutrient sen- Likhodii et  al., 2000; Raffo et  al., 2008), similar
sor that also happens to be involved in nociception to our interpretation of KD reductions in ther-
(Freitag and Miller, 2014). For instance, activation mal pain (Ruskin et  al., 2013). In the context
of PPARγ subtype alleviates inflammatory and of pain relief, what might low blood glucose
neuropathic pain in animal models (Morgenweck do besides promote ketosis? Such a question
et  al., 2010; Morgenweck et  al., 2013); PPARα is necessarily difficult to address given that the
agonists given to patients reduce pain in a num- former produces the latter. Fatty acid oxidation,
ber of inflammatory and neuropathic conditions and hence ketone body synthesis, can be blocked
(Freitag and Miller, 2014). Additionally, PUFAs pharmacologically in vivo, but such work has
201

Chapter 22: Metabolic Therapy and Pain 201

mostly focused on studies of ingestive behavior control glycemia, selective agonists to activate
(Langhans et  al., 2011). Fortunately, the ketosis nutrient sensors, adenosine A1 receptor ago-
complication is removed in in vitro prepara- nists to limit neural tissue hyperexcitability.
tions. With single‐neuron recording in hippo- Whereas such shortcuts may be useful in cases
campal slices, we investigated a model of the KD where the dietary treatment cannot be followed,
in which intracellular ATP was at medium‐to‐ they can have their own problems; for instance,
high concentration and extracellular glucose was the striking bradycardia that occurs after suf-
moderately low. This treatment hyperpolarized ficiently strong A1 receptor activation (Jonzon
the recorded neuron, an effect mediated by ATP et al., 1986). Dietary recruitment of these mech-
released through pannexin channels, metabo- anisms will be more physiological. In addition,
lized extracellularly to adenosine, which acted this review has suggested that some of these
on adenosine A1 receptors to open potassium dietary treatments are likely to involve multiple
channels (Kawamura et al., 2010). We suggested mechanisms that might work in synergy—drug
that this autocrine inhibitory mechanism is the treatments would lack this advantage. Multiple
major contributor to the KD’s anticonvulsant mechanisms acting together could be ideal for
effect (Masino et al., 2011). If this same mecha- restoring disordered metabolism. Furthermore,
nism were recruited in nociceptive brainstem these dietary treatments are all possess a wide
and spinal cord tissue during KD (or fasting range of health benefits, making them attrac-
or CR) treatment, it would have clear analgesic tive for people to optimize their health as well as
effects, possibly most strikingly in hyperexcitable manage their pain.
sensitized tissue.
Promotion of adenosine transmission by phar- AC K N OW L E D G M E N T S
macological means (promoting synthesis, block- Supported by National Institutes of Health
ing uptake/metabolism, allosterically modulating NS065446 and AT008742.
receptors) ameliorates pain in a wide variety of
models (e.g., Hurt and Zylka, 2012; Imlach et al., REFERENCES
2015; Little et al., 2015; Maes et al., 2012; Martins Adams, S.F. (1931). The benefit of prolonged low car-
et  al., 2013; Street et  al., 2011). Even pain treat- bohydrate feeding in diabetes:  report of a case.
ment by acupuncture and exercise seems to Mayo Clin Proc 6, 534–535.
involve enhanced adenosine (Goldman et  al., Alcocer‐Gómez, E., Garrido‐Maraver, J., Bullón, P.,
2010; Martins et al., 2013). The presence of a low‐ Marín‐Aguilar, F., Cotán, D., Carrión, A.M.,
glucose‐activated adenosine‐based autoinhibitory Alvarez‐Suarez, J.M., Giampieri, F., Sánchez‐
mechanism remains to be directly shown in brain- Alcazar, J.A., Battino, M., et al. (2015). Metformin
and caloric restriction induce an AMPK‐dependent
stem and spinal cord. However, adenosine also
restoration of mitochondrial dysfunction in
has antinociceptive effects in the periphery (Lima
fibroblasts from fibromyalgia patients. Biochim
et  al., 2010; Liu et  al., 2013; Poon and Sawynok, Biophys Acta 1852, 1257–1267.
1999; Valério et  al., 2009). In addition, immune Allread, V., and Paul, S. (2014). The misuse and abuse
cells express several types of adenosine recep- of prescription medications: part 1 current trends.
tor that can influence inflammation (Kumar and MD Advis 7, 12–20.
Sharma, 2009). Austin, G.L., Dalton, C.B., Hu, Y., Morris, C.B.,
There are other nutrient sensors besides Hankins, J., Weinland, S.R., Westman, E.C., Yancy,
GPR40 and the PPARs that could be involved in W.S., Jr., and Drossman, D.A. (2009). A very low‐
nociception. GPR41 (free fatty acid receptor 3) carbohydrate diet improves symptoms and quality
is expressed in the peripheral nervous system of life in diarrhea‐predominant irritable bowel syn-
and is activated by short‐chain fatty acids and is drome. Clin Gastroenterol Hepatol 7, 706–708.e1.
yet another molecular target for ketone bodies Baborka, C.J. (1930). Migraine:  results of treatment
(Kimura et  al., 2011; Won et  al., 2013). GPR120 by ketogenic diet in fifty cases. Mayo Clin Proc 5,
190–191.
(free fatty acid receptor 4), activated by PUFAs,
Becker, N., Bondegaard‐Thomsen, A., Olsen, A.K.,
is known to be anti‐inflammatory (Cintra et  al., Sjøgren, P., Bech, P., and Eriksen, J. (1997). Pain
2012). Studies have yet to investigate any specific epidemiology and health related quality of life in
involvement in pain for either of these receptors. chronic non‐malignant pain patients referred to
Many of the postulated antinociceptive a Danish multidisciplinary pain center. Pain 73,
mechanisms of fasting, CR, KD feeding, and 393–400.
PUFA supplementation can be recruited phar- Bodnar, R.J., Kelly, D.D., and Glusman, M. (1979).
macologically:  for example, metformin to 2‐Deoxy‐D‐glucose analgesia: influences of opiate
202

202 section III: Ketogenic Diet in the Laboratory

and non‐opiate factors. Pharmacol Biochem Behav receptor 40 family (GPR40‐GPR43) and its role in
11, 297–301. nutrient sensing. Biochem Soc Trans 34, 770–773.
Bodnar, R.J., Merrigan, K.P., and Wallace, M.M. (1981). Dahlin, M., Hjelte, L., Nilsson, S., and Åmark, P.
Analgesia following intraventricular administra- (2007). Plasma phospholipid fatty acids are influ-
tion of 2‐deoxy‐D‐glucose. Pharmacol Biochem enced by a ketogenic diet enriched with n‐3 fatty
Behav 14, 579–581. acids in children with epilepsy. Epilepsy Res 73,
Boe, C., and Vangsness, C.T. (2015). Fish oil and osteo- 199–207.
arthritis:  current evidence. Am J Orthop (Belle de Luis, D.A., Izaola, O., Alonso, M.G., Aller, R.,
Mead NJ) 44, 302–305. Cabezas, G., and de la Fuente, B. (2012). Effect
Bough, K.J., Chen, R.S., and Eagles, D.A. (1999a). Path of a hypocaloric diet with a commercial for-
analysis shows that increasing ketogenic ratio, but mula in weight loss and quality of life in obese
not β‐hydroxybutyrate, elevates seizure threshold patients with chronic osteoarthritis. Nutr Hosp
in the rat. Dev Neurosci 21, 400–406. 27, 1648–1654.
Bough, K.J., Valiyil, R., Han, F.T., and Eagles, D.A. Deng‐Bryant, Y., Prins, M.L., Hovda, D.A., and Harris,
(1999b). Seizure resistance is dependent upon age N.G. (2011). Ketogenic diet prevents alterations
and calorie restriction in rats fed a ketogenic diet. in brain metabolism in young but not adult rats
Epilepsy Res 35, 21–28. after traumatic brain injury. J Neurotrauma 28,
Bough, K.J., Yao, S.G., and Eagles, D.A. (2000). 1813–1825.
Higher ketogenic diet ratios confer protection Di Lorenzo, C., Currà, A., Sirianni, G., Coppola, G.,
from seizures without neurotoxicity. Epilepsy Res Bracaglia, M., Cardillo, A., De Nardis, L., and
38, 15–25. Pierelli, F. (2013). Diet transiently improves
Brune, K., and Patrignani, P. (2015). New insights into migraine in two twin sisters: possible role of keto-
the use of currently available non‐steroidal anti‐ genesis? Funct Neurol 28, 305–308.
inflammatory drugs. J Pain Res 8, 105–118 Di Lorenzo, C., Coppola, G., Sirianni, G., Di Lorenzo,
Bullón, P., Alcocer‐Gómez, E., Carrión, A.M., G., Bracaglia, M., Di Lenola, D., Siracusano, A.,
Marín‐Aguilar, F., Garrido‐Maraver, J., Román‐ Rossi, P., and Pierelli, F. (2015). Migraine improve-
Malo, L., Ruiz‐Cabello, J., Culic, O., Ryffel, B., ment during short lasting ketogenesis: a proof‐of‐
Apetoh, L., et al. (2016). AMPK phosphorylation concept study. Eur J Neurol 22, 170–177.
modulates pain by activation of NLRP3 inflamma- Donevan, S.D., White, H.S., Anderson, G.D., and
some. Antioxid Redox Signal 24, 157–170. Rho, J.M. (2003). Voltage‐dependent block of
Callaghan, B.C., Little, A.A., Feldman, E.L., and N‐methyl‐D‐aspartate receptors by the novel
Hughes, R.A. (2012). Enhanced glucose control anticonvulsant dibenzylamine, a bioactive con-
for preventing and treating diabetic neuropathy. stituent of L‐(+)‐β‐hydroxybutyrate. Epilepsia 44,
Cochrane Database Syst Rev 6, CD007543. 1274–1279.
Carling, D. (2005). AMP‐activated protein kinase: bal- Dressler, A., Trimmel‐Schwahofer, P., Reithofer, E.,
ancing the scales. Biochimie 87, 87–91. Mühlebner, A., Gröppel, G., Reiter‐Fink, E.,
Chmiel‐Perzyńska, I., Kloc, R., Perzyński, A., Rudzki, Benninger, F., Grassl, R., and Feucht, M. (2015).
S., and Urbańska, E.M. (2011). Novel aspect of Efficacy and tolerability of the ketogenic diet in
ketone action:  β‐hydroxybutyrate increases brain Dravet syndrome: comparison with various stan-
synthesis of kynurenic acid in vitro. Neurotox Res dard antiepileptic drug regimen. Epilepsy Res
20, 40–50. 109, 81–89.
Cintra, D.E., Ropelle, E.R., Moraes, J.C., Pauli, J.R., Dupuis, N., Curatolo, N., Benoist, J.‐F., and Stephane
Morari, J., Souza, C.T., Grimaldi, R., Stahl, M., Auvin, S. (2015). Ketogenic diet exhibits anti‐
Carvalheira, J.B., Saad, M.J., et  al. (2012). inflammatory properties. Epilepsia 56, e95–e98.
Unsaturated fatty acids revert diet‐induced hypo- Eisenberg, W. (1956). Zur Behandlung des primär‐
thalamic inflammation in obesity. PLoS One 7, chronischen Gelenkrheumatismus durch Fasten.
e30571. Hippokrates 27, 688–692.
Coppey, L.J., Holmes, A., Davidson, E.P., and Yorek, Eisinger, J., Plantamura, A., and Ayavou, T. (1994).
M.A. (2012). Partial replacement with menhaden Glycolysis abnormalities in fibromyalgia. J Am
oil improves peripheral neuropathy in high‐fat‐fed Coll Nutr 13, 144–148.
low‐dose streptozotocin type 2 diabetic rat. J Nutr Erecińska, M., Nelson, D., Daikhin, Y., and Yudkoff,
Metab 2012, 950517. M. (1996). Regulation of GABA level in rat brain
Coppey, L.J., Davidson, E.P., Obrosov, A., and Yorek, synaptosomes:  fluxes through enzymes of the
M.A. (2015). Enriching the diet with menhaden GABA shunt and effects of glutamate, calcium,
oil improves peripheral neuropathy in streptozoto- and ketone bodies. J Neurochem 67, 2325–2334.
cin‐induced type 1 diabetic rats. J Neurophysiol Escudero, G.E., Romañuk, C.B., Toledo, M.E.,
113, 701–708. Olivera, M.E., Manzo, R.H., and Laino, C.H.
Covington, D.K., Briscoe, C.A., Brown, A.J., and (2015). Analgesia enhancement and preven-
Jayawickreme, C.K. (2006). The G‐protein‐coupled tion of tolerance to morphine:  beneficial effects
203

Chapter 22: Metabolic Therapy and Pain 203

of combined therapy with omega‐3 fatty acids. J Gureje, O., Von Korff, M., Kola, L., Demyttenaere, K.,
Pharm Pharmacol 67, 1251–1262. He, Y., Posada‐Villa, J., Lepine, J.P., Angermeyer,
Figueroa, J.D., Cordero, K., Serrano‐Illan, M., M.C., Levinson, D., de Girolamo, G., et al. (2008).
Almeyda, A., Baldeosingh, K., Almaguel, F.G., The relation between multiple pains and mental
and De Leon, M. (2013). Metabolomics uncovers disorders:  results from the World Mental Health
dietary omega‐3 fatty acid‐derived metabolites Surveys. Pain 135, 82–91.
implicated in anti‐nociceptive responses after Hafström, I., Ringertz, B., Gyllenhammar, H.,
experimental spinal cord injury. Neuroscience Palmblad, J., and Harms‐Ringdahl, M. (1988).
255, 1–18. Effects of fasting on disease activity, neutrophil
Fraser, D.D., Whiting, S., Andrew, R.D., Macdonald, function, fatty acid composition, and leukotriene
E.A., Musa‐Veloso, K., and Cunnane, S.C. (2003). biosynthesis in patients with rheumatoid arthritis.
Elevated polyunsaturated fatty acids in blood Arthritis Rheum 31, 585–592.
serum obtained from children on the ketogenic Hallböök, T., Sjolander, A., Amark, P., Miranda, M.,
diet. Neurology 60, 1026–1029. Bjurulf, B., and Dahlin, M. (2015). Effectiveness of
Freitag, C.M., and Miller, R.J. (2014). Peroxisome the ketogenic diet used to treat resistant childhood
proliferator‐activated receptor agonists modulate epilepsy in Scandinavia. Eur J Paediatr Neurol
neuropathic pain: a link to chemokines? Front Cell 19, 29–36.
Neurosci 8, 238. Harada, S., Haruna, Y., Aizawa, F., Matsuura, W.,
Fuehrlein, B.S., Rutenberg, M.S., Silver, J.N., Warren, Nakamoto, K., Yamashita, T., Kasuya, F., and
M.W., Theriaque, D.W., Duncan, G.E., Stacpoole, Tokuyama, S. (2014). Involvement of GPR40,
P.W., and Brantly, M.L. (2004). Differential meta- a long‐chain free fatty acid receptor, in the
bolic effects of saturated versus polyunsaturated production of central post‐stroke pain after
fats in ketogenic diets. J Clin Endocrinol Metab global cerebral ischemia. Eur J Pharmacol 744,
89, 1641–1645. 115–123.
Gama, I.R., Trindade‐Filho, E.M., Oliveira, S.L., Hargraves, W.A., and Hentall, I.D. (2005). Analgesic
Bueno, N.B., Melo, I.T., Cabral‐Junior, C.R., effects of dietary caloric restriction in adult mice.
Barros, E.M., Galvão, J.A., Pereira, W.S., Ferreira, Pain 114, 455–461.
R.C., et al. (2015). Effects of ketogenic diets on the Harney, J.P., Madara, J., Madara, J., and I’Anson, H.
occurrence of pilocarpine‐induced status epilepti- (2002). Effects of acute inhibition of fatty acid oxi-
cus of rats. Metab Brain Dis 30, 93–98. dation on latency to seizure and concentrations of
Giménez‐Cassina, A., Martínez‐François, J.R., Fisher, β hydroxybutyrate in plasma of rats maintained
J.K., Szlyk, B., Polak, K., Wiwczar, J., Tanner, G.R., on calorie restriction and/or the ketogenic diet.
Lutas, A., Yellen, G., and Danial, N.N. (2012). Epilepsy Res 49, 239–246.
BAD‐dependent regulation of fuel metabolism Hu, X.‐L., Cheng, X., Fei, J., and Xiong, Z.‐Q. (2011).
and KATP channel activity confers resistance to epi- Neuron‐restrictive silencer factor is not required
leptic seizures. Neuron 74, 719–730. for the antiepileptic effect of the ketogenic diet.
Glajchen, M. (2001). Chronic pain:  treatment barri- Epilepsia 52, 1609–1616.
ers and strategies for clinical practice. J Am Board Hurt, J.K., and Zylka, M.J. (2012). PAPupuncture has
Fam Pract 14, 211–218. localized and long‐lasting antinociceptive effects
Goldberg, R.J., and Katz, J. (2007). A meta‐analysis of in mouse models of acute and chronic pain. Mol
the analgesic effects of omega‐3 polyunsaturated Pain 8, 28.
fatty acid supplementation for inflammatory joint Imlach, W.L., Bhola, R.F., May, L.T., Christopoulos,
pain. Pain 129, 210–223. A., and Christie, M.J. (2015). A positive allosteric
Goldman, N., Chen, M., Fujita, T., Xu, Q., Peng, W., modulator of the adenosine A1 receptor selectively
Liu, W., Jensen, T.K., Pei, Y., Wang, F., Han, X., inhibits primary afferent synaptic transmission
et  al. (2010). Adenosine A1 receptors mediate in a neuropathic pain model. Mol Pharmacol 88,
local anti‐nociceptive effects of acupuncture. Nat 460–468.
Neurosci 13, 883–888. Jiang, Y., Yang, Y., Wang, S., Ding, Y., Guo, Y., Zhang,
Guldbrand, H., Lindström, T., Dizdar, B., M.‐M., Wen, S.‐Q., and Ding, M.‐P. (2012).
Bunjaku, B., Östgren, C.J., Nystrom, F.H., and Ketogenic diet protects against epileptogenesis as
Bachrach‐Lindström, M. (2014). Randomization well as neuronal loss in amygdaloid‐ kindling sei-
to a low‐carbohydrate diet advice improves health zures. Neurosci Lett 508, 22–26.
related quality of life compared with a low‐fat diet Johnson, A.R., and Makowski, L. (2015). Nutrition
at similar weight‐loss in Type 2 diabetes mellitus. and metabolic correlates of obesity and inflam-
Diabetes Res Clin Pract 106, 221–227. mation:  clinical considerations. J Nutr 145,
Gureje, O., Von Korff, M., Simon, G.E., and Gater, R. 1131S‐1136S.
(1998). Persistent pain and well‐being:  a World Jonzon, B., Bergquist, A., Li, Y.‐O., and Fredholm, B.B.
Health Organization study in primary care. J Am (1986). Effects of adenosine and two stable ade-
Med Assoc 280, 147–151. nosine analogues on blood pressure, heart rate and
204

204 section III: Ketogenic Diet in the Laboratory

colonic temperature in the rat. Acta Physiol Scand J., Pérez‐Jimenez, F., and Fuentes‐Jiménez, F.
126, 491–498. (2013). Mediterranean diet, moderate‐to‐high
Juge, N., Gray, J.A., Omote, H., Miyaji, T., Inoue, T., intensity training, and health‐related quality of
Hara, C., Uneyama, H., Edwards, R.H., Nicoll, life in adults with metabolic syndrome. Eur J Prev
R.A., and Moriyama, Y. (2010). Metabolic con- Cardiol 20, 555–564.
trol of vesicular glutamate transport and release. Langhans, W., Leitner, C., and Arnold, M. (2011).
Neuron 68, 99–112. Dietary fat sensing via fatty acid oxidation in
Jurcovicova, J., Stanciková, M., Svík, K., Ondrejicková, enterocytes:  possible role in the control of eat-
Krsova, D., Seres, J., and Rokyta, R. (2001). Stress ing. Am J Physiol Regul Integr Comp Physiol 300,
of chronic food restriction attenuates the develop- R554–565.
ment of adjuvant arthritis in male Long Evans rats. Lauria, G., Ziegler, D., Malik, R., Merkies, I.S.,
Clin Exp Rheumatol 19, 371–376. Waxman, S.G., Faber, C.G., and group, P.S. (2014).
Karki, P., Kurihara, T., Nakamachi, T., Watanabe, J., The role of sodium channels in painful diabetic
Asada, T., Oyoshi, T., Shioda, S., Yoshimura, M., and idiopathic neuropathy. Curr Diab Rep 14, 538.
Arita, K., and Miyata, A. (2015). Attenuation of Lee, Y.H., Bae, S.C., and Song, G.G. (2012). Omega‐3
inflammatory and neuropathic pain behaviors in polyunsaturated fatty acids and the treatment of
mice through activation of free fatty acid receptor rheumatoid arthritis:  a meta‐analysis. Arch Med
GPR40. Mol Pain 11, 6. Res 43, 356–362.
Kashiwaya, Y., Pawlosky, R., Markis, W., King, M.T., Likhodii, S.S., Musa, K., Mendonca, A., Dell, C.,
Bergman, C., Srivastava, S., Murray, A., Clarke, Burnham, W.M., and Cunnane, S.C. (2000).
K., and Veech, R.L. (2010). A ketone ester diet Dietary fat, ketosis, and seizure resistance in rats
increases brain malonyl‐CoA and uncoupling pro- on the ketogenic diet. Epilepsia 41, 1400–1410.
teins 4 and 5 while decreasing food intake in the Lima, F.O., Souza, G.R., Verri, W.A., Jr., Parada, C.A.,
normal Wistar rat. J Biol Chem 285, 25950–25956. Ferreira, S.H., Cunha, F.Q., and Cunha, T.M.
Kawamura, M., Jr., Ruskin, D.N., and Masino, S.A. (2010). Direct blockade of inflammatory hyper-
(2010). Metabolic autocrine regulation of neurons nociception by peripheral A1 adenosine recep-
involves cooperation among pannexin hemichan- tors:  Involvement of the NO/cGMP/PKG/KATP
nels, adenosine receptors and KATP channels. J signaling pathway. Pain 151, 506–515.
Neurosci 30, 3886–3895. Little, J.W., Ford, A., Symons‐Liguori, A.M., Chen, Z.,
Kawamura, M., Jr., Ruskin, D.N., Geiger, J.D., Boison, Janes, K., Doyle, T., Xie, J., Luongo, L., Tosh, D.K.,
D., and Masino, S.A. (2014). Ketogenic diet sensi- Maione, S., et al. (2015). Endogenous adenosine A3
tizes glucose control of hippocampal excitability. J receptor activation selectively alleviates persistent
Lipid Res 55, 2254–2260. pain states. Brain 138, 28–35.
Kim, D.Y., Vallejo, J., and Rho, J.M. (2010). Ketones Liu, J., Reid, A.R., and Sawynok, J. (2013). Spinal sero-
prevent synaptic dysfunction induced by mito- tonin 5‐HT7 and adenosine A1 receptors, as well
chondrial respiratory complex inhibitors. J as peripheral adenosine A1 receptors, are involved
Neurochem 114, 130–141. in antinociception by systemically administered
Kim, K.S., Glajchen, M., and Portenoy, R.K. (2000). amitriptyline. Eur J Pharmacol 698, 213–219.
Unwillingness to fill opioid analgesic prescriptions Lund, T.M., Risa, Ø., Sonnewald, U., Schousboe, A.,
by local pharmacies. J Pain Symptom Manage 20, and Waagepetersen, H.S. (2009). Availability of
160–162. neurotransmitter glutamate is diminished when
Kimura, I., Inoue, D., Maeda, T., Hara, T., Ichimura, β‐hydroxybutyrate replaces glucose in cultured
A., Miyauchi, S., Kobayashi, M., Hirasawa, A., and neurons. J Neurochem 110, 80–91.
Tsujimoto, G. (2011). Short‐chain fatty acids and Lusardi, T.A., Akula, K.K., Coffman, S.Q., Ruskin, D.N.,
ketones directly regulate sympathetic nervous sys- Masino, S.A., and Boison, D. (2015). Ketogenic
tem via G protein‐coupled receptor 41 (GPR41). diet prevents epileptogenesis and disease progres-
Proc Natl Acad Sci USA 108, 8030–8035. sion in adult mice and rats. Neuropharmacology
Kroker, G.F., Stroud, R.M., Marshall, R., Bullock, T., 99, 500–509.
Carroll, F.M., Greenberg, M., Randolph, T.G., Rea, Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic diet
W.J., and Smiley, R.E. (1984). Fasting and rheu- metabolites reduce firing in central neurons by
matoid arthritis: a multicenter study. Clin Ecol 2, opening KATP channels. J Neurosci 27, 3618–3625.
137–144. Maes, S.S., Pype, S., Hoffmann, V.L.H., Biermans, M.,
Kumar, V., and Sharma, A. (2009). Adenosine:  An and Meert, T.F. (2012). Antihyperalgesic activity
endogenous modulator of innate immune sys- of nucleoside transport inhibitors in models of
tem with therapeutic potential. Eur J Pharmacol inflammatory pain in guinea pigs. J Pain Res 5,
616, 7–15. 391–400.
Landaeta‐Díaz, L., Fernández, J.M., Da Maggioni, F., Margoni, M., and Zanchin, G. (2011).
Silva‐Grigoletto, M., Rosado‐Alvarez, D., Gómez‐ Ketogenic diet in migraine treatment:  a brief but
Garduño, A., Gómez‐Delgado, F., López‐Miranda, ancient history. Cephalalgia 31, 1150–1151.
205

Chapter 22: Metabolic Therapy and Pain 205

Manchikanti, L. (2006). Prescription drug abuse: what drugs for neuropathic pain. Cochrane Database
is being done to address this new drug epidemic? Syst Rev 10, CD010902.
Pain Physician 9, 287–321. Morgenweck, J., Abdel‐aleem, O.S., McNamara, K.C.,
Martin, Y.B., and Avendaño, C. (2009). Effects of Donahue, R.R., Badr, M.Z., and Taylor, B.K.
removal of dietary polyunsaturated fatty acids on (2010). Activation of peroxisome proliferator‐
plasma extravasation and mechanical allodynia activated receptor γ in brain inhibits inflamma-
in a trigeminal neuropathic pain model. Mol tory pain, dorsal horn expression of Fos, and local
Pain 5, 8. edema. Neuropharmacology 58, 337–345.
Martins, D.F., Mazzardo‐Martins, L., Soldi, F., Morgenweck, J., Griggs, R.B., Donahue, R.R., Zadina,
Stramosk, J., Piovezan, A.P., and Santos, A.R.S. J.E., and Taylor, B.K. (2013). PPARγ activation blocks
(2013). High‐intensity swimming exercise reduces development and reduces established neuropathic
neuropathic pain in an animal model of complex pain in rats. Neuropharmacology 70, 236–246.
regional pain syndrome type I: evidence for a role of Muller‐Schwarze, A.B., Tandon, P., Liu, Z., Lang,
the adenosinergic system. Neuroscience 234, 69–76. Y., Holmes, G.L., and Stafstrom, C.E. (1999).
Masino, S.A., Li, T., Theofilas, P., Sandau, U., Ruskin, Ketogenic diet reduces spontaneous seizures and
D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., mossy fiber sprouting in the kainic acid model.
and Boison, D. (2011). A ketogenic diet suppresses NeuroReport 10, 1517–1522.
seizures in mice through adenosine A1 receptors. J Nakamoto, K., Nishinaka, T., Sato, N., Mankura, M.,
Clin Invest 121, 2679–2683. Koyama, Y., Kasuya, F., and Tokuyama, S. (2013).
Masino, S.A., and Ruskin, D.N. (2013). Ketogenic diets Hypothalamic GPR40 signaling activated by free
and pain. J Child Neurol 28, 993–1001. long chain fatty acids suppresses CFA‐ induced
Matta, J.A., Miyares, R.L., and Ahern, G.P. (2007). inflammatory chronic pain. PLoS One 8, e81563.
TRPV1 is a novel target for omega‐3 polyunsatu- Nakamoto, K., Nishinaka, T., Sato, N., Aizawa, F.,
rated fatty acids. J Physiol 578, 397–411. Yamashita, T., Mankura, M., Koyama, Y., Kasuya,
Megaw, K., and Wilmshurst, J.M. (2015). The Keilah F., and Tokuyama, S. (2015). The activation of
Foundation: making the ketogenic diet viable for supraspinal GPR40/FFA1 receptor signalling
children in Africa. Epilepsia 56, 514–516. regulates the descending pain control system. Br J
Mense, S. (1983). Basic neurobiological mechanisms Pharmacol 172, 1250–1262.
of pain. Am J Med 75, 4–14. Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
Michalsen, A. (2010). Prolonged fasting as a method of Edwards, N., Fitzsimmons, G., Whitney, A., and
mood enhancement in chronic pain syndromes: a Cross, J.H. (2008). The ketogenic diet for the treat-
review of clinical evidence and mechanisms. Curr ment of childhood epilepsy:  a randomised con-
Pain Headache Rep 14, 80–87. trolled trial. Lancet Neurol 7, 500–506.
Michalsen, A., Li, C., Kaiser, K., Lüdtke, R., Meier, L., Paoli, A., Moro, T., Bosco, G., Bianco, A., Grimaldi,
Stange, R., and Kessler, C. (2013). In-patient treat- K.A., Camporesi, E., and Mangar, D. (2015).
ment of fibromyalgia: a controlled nonrandomized Effects of n‐3 polyunsaturated fatty acids (ω‐3)
comparison of conventional medicine versus inte- supplementation on some cardiovascular risk
grative medicine including fasting therapy. Evid factors with a ketogenic Mediterranean diet. Mar
Based Complement Alternat Med 2013, 908610. Drugs 13, 996–1009.
Michalsen, A., Riegert, M., Lüdtke, R., Bäcker, M., Pérez‐Guisado, J., and Muñoz‐Serrano, A. (2011). The
Langhorst, J., Schwickert, M., and Dobos, G.J. effect of the Spanish Ketogenic Mediterranean
(2005). Mediterranean diet or extended fasting’s Diet on nonalcoholic fatty liver disease:  a pilot
influence on changing the intestinal microflora, study. J Med Food 14, 677–680.
immunoglobulin A secretion and clinical outcome Pérez, J., Ware, M.A., Chevalier, S., Gougeon, R., and
in patients with rheumatoid arthritis and fibromy- Shir, Y. (2005). Dietary omega‐3 fatty acids may
algia:  an observational study. BMC Complement be associated with increased neuropathic pain in
Altern Med 5, 22. nerve‐injured rats. Anesth Analg 101, 444–448,
Mobbs, C.V., Mastaitis, J.W., Isoda, F., and Poplawski, table of contents.
M. (2013). Treatment of diabetes and diabetic Phillips, C.J. (2006). Economic burden of chronic
complications with a ketogenic diet. J Child pain. Expert Rev Pharmacoecon Outcomes Res 6,
Neurol 28, 1009–1014. 591–601.
Monfoulet, L.E., Philippe, C., Mercier, S., Coxam, V., Poon, A., and Sawynok, J. (1999). Antinociceptive
and Wittrant, Y. (2015). Deficiency of G‐ protein and anti‐inflammatory properties of an adenos-
coupled receptor 40, a lipid‐activated receptor, ine kinase inhibitor and an adenosine deaminase
heightens in vitro‐ and in vivo‐induced murine inhibitor. Eur J Pharmacol 1999, 123–138.
osteoarthritis. Exp Biol Med (Maywood) 240, Poon, H.F., Shepherd, H.M., Reed, T.T., Calabrese,
854–866. V., Stella, A.M., Pennisi, G., Cai, J., Pierce, W.M.,
Moore, R.A., Chi, C.C., Wiffen, P.J., Derry, S., and Rice, Klein, J.B., and Butterfield, D.A. (2006). Proteomics
A.S. (2015). Oral nonsteroidal anti‐inflammatory analysis provides insight into caloric restriction
206

206 section III: Ketogenic Diet in the Laboratory

mediated oxidation and expression of brain pro- Sawynok, J. (2015). Adenosine receptor targets for
teins associated with age‐related impaired cellular pain. Neuroscience. in press.
processes:  Mitochondrial dysfunction, glutamate Schmidt, S., Stange, R., Lischka, E., Kiehntopf, M.,
dysregulation and impaired protein synthesis. Deufel, T., Loth, D., and Uhlemann, C. (2010).
Neurobiol Aging 27, 1020–1034. [Uncontrolled clinical study of the efficacy of
Raffo, E., Francois, J., Ferrandon, A., Koning, E., and ambulant fasting in patients with osteoarthritis].
Nehlig, A. (2008). Calorie‐restricted ketogenic Forsch Komplementmed 17, 87–94.
diet increases thresholds to all patterns of pentyl- Sköldstam, L., Larsson, L., and Lindström, F.D.
enetetrazole‐induced seizures: importance of elec- (1979). Effect of fasting and lactovegetarian diet
troclinical assessment. Epilepsia 49, 320–328. on rheumatoid arthritis. Scand J Rheumatol 8,
Ramsden, C.E., Faurot, K.R., Zamora, D., Suchindran, 249–255.
C.M., Macintosh, B.A., Gaylord, S., Ringel, A., Smith, A.G., Russell, J., Feldman, E.L., Goldstein, J.,
Hibbeln, J.R., Feldstein, A.E., Mori, T.A., et  al. Peltier, A., Smith, S., Hamwi, J., Pollari, D., Bixby,
(2013). Targeted alteration of dietary n‐3 and B., Howard, J., et al. (2006). Lifestyle intervention
n‐6 fatty acids for the treatment of chronic head- for pre‐diabetic neuropathy. Diabetes Care 29,
aches: a randomized trial. Pain 154, 2441–2451. 1294–1299.
Ramsden, C.E., Faurot, K.R., Zamora, D., Palsson, Street, S.E., Walsh, P.L., Sowa, N.A., Taylor‐Blake,
O.S., MacIntosh, B.A., Gaylord, S., Taha, A.Y., B., Guillot, T.S., Vihko, P., Wightman, R.M., and
Rapoport, S.I., Hibbeln, J.R., Davis, J.M., et  al. Zylka, M.J. (2011). PAP and NT5E inhibit noci-
(2015). Targeted alterations in dietary n‐3 and n‐6 ceptive neurotransmission by rapidly hydrolyzing
fatty acids improve life functioning and reduce nucleotides to adenosine. Mol Pain 7, 80.
psychological distress among patients with chronic Su, S.W., Sogawa, M.R.C.Y., Silveira, D.C., Holmes,
headache:  a secondary analysis of a randomized G.L., and Stafstrom, C.E. (2000). Timing of keto-
trial. Pain 156, 587–596. genic diet initiation in an experimental epilepsy
Redivo, D.D., Schreiber, A.K., Adami, E.R., Ribeiro, model. Dev Brain Res 125, 131–138.
D.E., Joca, S.R., Zanoveli, J.M., and Cunha, J.M. Sullivan, P.G., Rippy, N.A., Dorenbos, K., Concepcion,
(2015). Effect of omega‐3 polyunsaturated fatty R.C., Agarwal, A.K., and Rho, J.M. (2004). The
acid treatment over mechanical allodynia and ketogenic diet increases mitochondrial uncou-
depressive‐like behavior associated with experi- pling protein levels and activity. Ann Neurol 55,
mental diabetes. Behav Brain Res 298, 57–64. 576–580.
Roe, C.R., Yang, B.Z., Brunengraber, H., Roe, D.S., Taggart, A.K.P., Kero, J., Gan, X., Cai, T.‐Q., Cheng,
Wallace, M., and Garritson, B.K. (2008). Carnitine K., Ippolito, M., Ren, N., Kaplan, R., Wu, K.,
palmitoyltransferase II deficiency:  successful Wu, T.‐J., et  al. (2005). (D)‐β‐hydroxybutyrate
anaplerotic diet therapy. Neurology 71, 260–264. inhibits adipocyte lipolysis via the nicotinic
Roffey, D.M., Ashdown, L.C., Dornan, H.D., Creech, acid receptor PUMA‐G. J Biol Chem 280,
M.J., Dagenais, S., Dent, R.M., and Wai, E.K. 26649–26652.
(2011). Pilot evaluation of a multidisciplinary, Taha, A.Y., Filo, E., Ma, D.W.L., and Burnham, W.M.
medically supervised, nonsurgical weight loss (2009). Dose‐dependent anticonvulsant effects
program on the severity of low back pain in obese of linoleic and α‐linolenic polyunsaturated fatty
adults. Spine J 11, 197–204. acids on pentylenetetrazol induced seizures in rats.
Ross‐Huot, M.‐C., Laferrière, A., Gi, C.M., Khorashadi, Epilepsia 50, 72–82.
M., Schrincker, T., and Coderre, T.J. (2011). Effects Taha, A.Y., Ryan, M.A.A., and Cunnane, S.C. (2005).
of glycemic regulation on chronic postischemia Despite transient ketosis, the classic high‐fat
pain. Anesthesiology 115, 614–625. ketogenic diet induces marked changes in fatty
Rothschild, J., Hoddy, K.K., Jambazian, P., and Varady, acid metabolism in rats. Metab Clin Exp 54,
K.A. (2014). Time‐restricted feeding and risk of 1127–1132.
metabolic disease: a review of human and animal Tajes, M., Gutierrez‐Cuesta, J., Folch, J.,
studies. Nutr Rev 72, 308–318. Ortuño‐Sahagun, D., Verdaguer, E., Jiménez, A.,
Ruskin, D.N., Kawamura, M., Jr., and Masino, S.A. Junyent, F., Lau, A., Camins, A., and Pallàs, M.
(2009). Reduced pain and inflammation in juve- (2010). Neuroprotective role of intermittent fast-
nile and adult rats fed a ketogenic diet. PLoS One ing in senescence‐accelerated mice P8 (SAMP8).
4, e8349. Exp Gerontol 45, 702–710.
Ruskin, D.N., Suter, T.A.C.S., Ross, J.L., and Masino, Tanner, G.R., Lutas, A., Martínez‐François, J.R., and
S.A. (2013). Ketogenic diets and thermal pain: dis- Yellen, G. (2011). Single KATP channel opening in
sociation of hypoalgesia, elevated ketones, and response to action potential firing in mouse den-
lowered glucose in rats. J Pain 14, 467–474. tate granule neurons. J Neurosci 31, 8689–8696.
Sakamoto, K., and Grunewald, K.K. (1987). Beneficial Tendler, D., Lin, S., Yancy, W.S., Jr., Mavropoulos, J.,
effects of exercise on growth of rats during inter- Sylvestre, P., Rockey, D.C., and Westman, E.C.
mittent fasting. J Nutr 117, 390–395. (2007). The effect of a low‐carbohydrate, ketogenic
207

Chapter 22: Metabolic Therapy and Pain 207

diet on nonalcoholic fatty liver disease:  a pilot in a pentylenetetrazole‐model of seizure. Brain


study. Dig Dis Sci 52, 589–593. Res 1618, 50–54.
Tieu, K., Perier, C., Caspersen, C., Teismann, P., Voskuyl, R.A., Vreugdenhil, M., Kang, J.X., and Leaf,
Wu, D.‐C., Yan, S.‐D., Niani, A., Vila, M., A. (1998). Anticonvulsant effect of polyunsatu-
Jackson‐Lewis, V., Ramasamy, R., et  al. (2003). rated fatty acid in rats, using the cortical stimula-
D‐β‐hydroxybutyrate rescues mitochondrial res- tion model. Eur J Pharmacol 341, 145–152.
piration and mitigates features of Parkinson dis- Wagner, K., Vito, S., Inceoglu, B., and Hammock,
ease. J Clin Invest 112, 892–901. B.D. (2014). The role of long chain fatty acids
Tigerholm, J., Börjesson, S.I., Lundberg, L., Elinder, and their epoxide metabolites in nociceptive
F., and Fransén, E. (2012). Dampening of hyper- signaling. Prostaglandins Other Lipid Mediat
excitability in CA1 pyramidal neurons by polyun- 113–115, 2–12.
saturated fatty acids acting on voltage‐ gated ion Walker, I., and Said, R.R. (2015). Predictors of ketogenic
channels. PLoS One 7, e44388. diet efficacy in children based on the electroenceph-
Tikoo, K., Tripathi, D.N., Kabra, D.G., Sharma, V., and alogram (EEG). J Child Neurol 30, 1270–1274.
Gaikwad, A.B. (2007). Intermittent fasting pre- Welshman, A. (2005). Palliative care. Some organi-
vents the progression of type I diabetic nephropa- zational considerations. Minerva Anestesiol 71,
thy in rats and changes the expression of Sir2 and 439–443.
p53. FEBS Lett 581, 1071–1078. White, D.K., Neogi, T., Rejeski, W.J., Walkup, M.P.,
Tillu, D.V., Melemedjian, O.K., Asiedu, M.N., Qu, N., Lewis, C.E., Nevitt, M.C., Foy, C.G., Felson, D.T.,
De Felice, M., Dussor, G., and Price, T.J. (2012). and Group, L.A.R. (2015). Can an intensive diet
Resveratrol engages AMPK to attenuate ERK and and exercise program prevent knee pain among
mTOR signaling in sensory neurons and inhib- overweight adults at high risk? Arthritis Care Res
its incision‐induced acute and chronic pain. Mol (Hoboken) 67, 965–971.
Pain 8, 5. Wideman, C.H., Murphy, H.M., and McCartney, S.B.
Todorova, M.T., Tandon, P., Madore, R.A., Stafstrom, (1996). Interactions between vasopressin and food
C.E., and Seyfried, T.N. (2000). The ketogenic diet restriction on stress‐induced analgesia. Peptides
inhibits epileptogenesis in EL mice: a genetic model 17, 63–66.
for idiopathic epilepsy. Epilepsia 41, 933–940. Wilder, R.M. (1921a). The effects of ketonemia on the
Traba, J., Kwarteng‐Siaw, M., Okoli, T.C., Li, J., course of epilepsy. Mayo Clin Bull 2, 307.
Huffstutler, R.D., Bray, A., Waclawiw, M.A., Han, Wilder, R.M. (1921b). High fat diets in epilepsy. Mayo
K., Pelletier, M., Sauve, A.A., et al. (2015). Fasting Clin Bull 2, 308.
and refeeding differentially regulate NLRP3 Willis, S., Samala, R., Rosenberger, T.A., and Borges,
inflammasome activation in human subjects. J K. (2009). Eicosapentaenoic and docosahexae-
Clin Invest 125, 4592–4600. noic acids are not anticonvulsant or neuroprotec-
Udén, A.M., Trang, L., Venizelos, N., and Palmblad, J. tive in acute mouse seizure models. Epilepsia 50,
(1983). Neutrophil functions and clinical perfor- 138–142.
mance after total fasting in patients with rheuma- Won, Y.‐J., Lu, V.B., Puhl, H.L., III, and Ikeda, S.R.
toid arthritis. Ann Rheum Dis 42, 45–51. (2013). β‐hydroxybutyrate modulates N‐ type
Valério, D.A., Ferreira, F.I., Cunha, T.M., Alves‐Filho, calcium channels in rat sympathetic neurons by
J.C., Lima, F.O., De Oliveira, J.R., Ferreira, S.H., acting as an agonist for the G‐protein‐ coupled
Cunha, F.Q., Queiroz, R.H., and Verri, W.A., Jr. receptor FFA3. J Neurosci 33, 19314–19325.
(2009). Fructose‐1,6‐bisphosphate reduces inflam- Xu, X.P., Sun, R.P., and Jin, R.F. (2006). [Effect of keto-
matory pain‐like behaviour in mice:  role of ade- genic diet on hippocampus synaptic reorganiza-
nosine acting on A1 receptors. Br J Pharmacol 158, tion and GluR5 expression in kainic acid induced
558–568. rat model of epilepsy]. Zhonghua Er Ke Za Zhi 44,
Varady, K.A., Hoddy, K.K., Kroeger, C.M., 100–104.
Trepanowski, J.F., Klempel, M.C., Barnosky, A., Xu, X.‐P., Erichsen, D., Börjesson, S.I., Dahlin, M.,
and Bhutani, S. (2015). Determinants of weight Åmark, P., and Elinder, F. (2008). Polyunsaturated
loss success with alternate day fasting. Obes Res fatty acids and cerebrospinal fluid from children
Clin Pract. on the ketogenic diet open a voltage‐gated K chan-
Veech, R.L. (2004). The therapeutic implications of nel:  a putative mechanism of antiseizure action.
ketone bodies:  the effects of ketone bodies in Epilepsy Res 80, 57–66.
pathological conditions:  ketosis, ketogenic diet, Yamaoka, G., Horiuchi, H., Morino, T., Miura, H.,
redox states, insulin resistance, and mitochondrial and Ogata, T. (2013). Different analgesic effects of
metabolism. Prostaglandins Leukot Essent Fatty adenosine between postoperative and neuropathic
Acids 70, 309–319. pain. J Orthop Sci 18, 130–136.
Viggiano, A., Pilla, R., Arnold, P., Monda, M., Yancy, W.S., Jr., Almirall, D., Maciejewski, M.L.,
D’Agostino, D., and Coppola, G. (2015). Kolotkin, R.L., McDuffie, J.R., and Westman,
Anticonvulsant properties of an oral ketone ester E.C. (2009). Effects of two weight‐loss diets on
208

208 section III: Ketogenic Diet in the Laboratory

health‐related quality of life. Qual Life Res 18, Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg, E.L.,
281–289. Bodogai, M., Kim, D., D’Agostino, D., Planavsky,
Yang, L., Zhao, J., Milutinovic, P.S., Brosnan, N., Lupfer, C., Kanneganti, T.D., et  al. (2015).
R.J., Eger, E.I., II, and Sonner, J.M. (2007). The ketone metabolite β‐ hydroxybutyrate blocks
Anesthetic properties of the ketogenic bod- NLRP3 inflammasome‐mediated inflammatory
ies β‐hydroxybutyric acid and acetone. Anesth disease. Nature Med 21, 263–269.
Analg 105, 673–679. Żarnowsky, T., Chorągiewicz, T., Tulidowicz‐Bielak,
Yehuda, S., Carasso, R.L., and Mostofsky, D.I. (1994). M., Thaler, S., Rejdak, R., Żarnowska, I., Turski,
Essential fatty acid preparation (SR‐3) raises the W.A., and Gasior, M. (2012). Ketogenic diet
seizure threshold in rats. Eur J Pharmacol 254, increases concentrations of kynurenic acid in dis-
193–198. crete brain structures of young and adult rats. J
Yorek, M.S., Obrosov, A., Shevalye, H., Lupachyk, S., Neural Transm 119, 679–684.
Harper, M.M., Kardon, R.H., and Yorek, M.A. Zhao, Y.T., Tekkök, S., and Krnjević, K. (1997).
(2014). Effect of glycemic control on corneal 2‐Deoxy‐D‐glucose‐induced changes in mem-
nerves and peripheral neuropathy in streptozoto- brane potential, input resistance, and excitatory
cin‐induced diabetic C57Bl/6J mice. J Peripher postsynaptic potentials of CA1 hippocampal neu-
Nerv Syst 19, 205–217. rons. Can J Physiol Pharmacol 75, 368–374.
209

23
Ketogenic Diet, Adenosine, Epigenetics,
and Antiepileptogenesis
T H E R E S A A . L U S A R D I , P H D A N D D E T L E V B O I S O N, P H D

INTRODUCTION considering that the individuals in controlled


Fasting has been known as a means to control trials of the diet are refractory to conventional
epilepsy for thousands of years. Formal studies of antiepileptic drugs and have been suffering from
fasting in the early years of the 20th century led epilepsy for many years at the time the diet is initi-
to the critical observation of elevated ketone bod- ated. A more comprehensive understanding of the
ies excreted during fasting, and diets tailored to underlying mechanisms could yield evidence for
elevate ketones were established (Wheless, 2008; predictive biomarkers that can identify patients
Peterman, 1925, 1924). The difficulty of compli- who are most likely to respond well to the KD.
ance in combination with the availability of anti-
epileptic drugs diminished enthusiasm for the KD A D E N O S I N E H O M E O S TA S I S
throughout much of the 20th century, but more AND SEIZURES
recent studies in patients refractory to conven- The purine ribonucleoside adenosine is a ubiqui-
tional drugs have demonstrated the KD’s clear tous homeostatic regulator of brain activity acting
efficacy for seizure suppression. While the earli- at four G-protein-coupled receptors. Receptors
est studies suggested efficacy of the KD only in are classified by their second messenger activity
children, several studies show similar therapeu- (Gi/o:  A1R and A3R; or Gs:  A2AR and A2BR) and
tic effects in adults (Barborka, 1930; Sirven et al., their affinity for adenosine (nM:  A1R, A2AR;
1999; Coppola et al., 2002). μM: A2BR, A3R). In the brain, basal adenosine lev-
The precise mechanisms by which the KD exerts els are typically in the nM range, regulated largely
antiepileptic effects remain unknown. Beneficial through a balance between ATP breakdown by
effects of the KD can manifest clinically as quickly ectonucleotideases (CD39, CD73) (Fredholm
as several days or as long as 8  months after diet et al., 2005; Cunha, 2001) and metabolic clearance
initiation. Diet reversal yields even more perplex- by astrocytic adenosine kinase (ADK) (Boison,
ing outcomes. In some individuals, seizures return 2013; Lloyd and Fredholm, 1995). In the epileptic
within hours of glucose reintroduction, suggesting brain, this balance is disrupted by overexpression
an antiseizure mechanism. Others are able to return of ADK in astrogliotic cells (Fedele et  al., 2005),
to a “normal” diet and remain seizure-free, suggest- which results in increased clearance of adenos-
ing an antiepileptogenic mechanism. A  detailed ine and loss of A1R-mediated synaptic inhibition
review of the proposed mechanisms of the KD has (Boison, 2008).
been published recently (Masino and Rho, 2012), The role of adenosine in seizure termination has
illustrating that the influences of the diet on meta- been demonstrated directly (During and Spencer,
bolic processes are broad. Indeed, this broad range 1992; Van Gompel et  al., 2014). Astrogliosis and
of metabolic influences suggests that the therapeu- ADK up-regulation is well documented in the
tic benefits of the KD are not limited to epilepsy epileptic brain (Sofroniew and Vinters, 2010;
but might extend into other neurological disorders Aronica et  al., 2011), and strategies to restore
(Stafstrom & Rho, 2012; Kossoff & Hartman, 2012). adenosine signaling have been quite successful
The dramatic reduction or elimination of for seizure suppression. Pharmacologic activation
seizures in up to 40% of persons with epilepsy of the A1R blocks seizures (Gouder et  al., 2003),
on a KD is a strong argument for continued use as does inhibition of ADK with 5-iodotubercidin
of the diet. This is particularly compelling when (ITU) (Gouder et al., 2004). Transgenic mice have
210

210 section III: Ketogenic Diet in the Laboratory

provided further evidence of the importance of contradictory roles proposed for adenosine, a
the A1R in seizure suppression and a causal role broader view of adenosine as a “retaliatory metab-
for ADK overexpression in seizure susceptibility. olite” has been proposed (Newby, 1984) to explain
A1R knockout mice are susceptible to lethal status the emerging role of adenosine in response to
epilepticus (Fedele et  al., 2006; Kochanek et  al., physiologic stress. This concept was applied to the
2006). Overexpression of ADK in the hippocam- KD, postulating a role for adenosine as the molec-
pus is associated with spontaneous seizures, which ular link between cellular metabolism and seizure
can be blocked by ITU (Fedele et  al., 2005), and suppression (Masino et  al., 2009). Indeed, subse-
ADK suppression in the hippocampus confers quent studies have shown that administration of
protection against epileptogenesis (Li et al., 2007). the KD to epileptic rats in the pilocarpine model
The mechanistic rationale and therapeutic efficacy restores adenosine levels in the hippocampus to
of adenosine homeostasis in seizure suppression is normal (nonepileptic) levels, and that returning to
well established; however, the ubiquitous nature of a normal-fat/carbohydrate diet results in a return
adenosine signaling makes direct clinical transla- to adenosine deficiency (Lusardi et  al., 2015).
tion of these therapies impractical. Chronic sys- A  causal role for adenosine in the antiseizure
temic treatment has sedative, cardiovascular, and effects of the KD has been demonstrated in A1R
renal side effects that have led to the near aban- knockout mice and ADK overexpressing mice,
donment of pharmaceutical drug development both of which have spontaneous focal seizures.
efforts. To avoid limitations of systemic adenosine Spontaneous seizures were eliminated in the ADK
augmentation, focal delivery strategies have been overexpressing mice fed with the KD, an effect
developed. It was shown that focal adenosine sup- that was blocked by the A1R antagonist dipropyl-
plementation provided similar efficacy to systemic cyclopentylxanthine. No reduction in spontane-
treatment. Cells engineered to release adenosine ous seizures was observed in A1R−/− mice on the
inhibited kindled seizures in an A1R dependent KD, though a partial reduction was measured in
manner (Huber et al., 2001) and blocked seizures A1R+/− mice, consistent with reduced A1R expres-
in a model of drug-resistant epilepsy (Li et  al., sion levels in the heterozygotic mice (Masino et al.,
2008). Similarly, a silk-based scaffold for con- 2011). These results support the hypothesis that
trolled focal adenosine release suppressed kindled the antiseizure efficacy of the KD exerts some of its
seizures and epileptogenesis in a kindling model influence through restoration of local adenosine
(Wilz et al., 2008; Szybala et al., 2009). Of particu- homeostasis in the epileptic brain.
lar interest, in the clinically relevant kainic acid
status epilepticus (KASE) model of epilepsy, silk- EPIGENETICS IN EPILEPSY
based transient adenosine release halted epilepsy Heritable factors confer a small but appre-
progression in epileptic rats with fully generalized ciable increase in risk of epilepsy development
spontaneous seizures, with lack of seizure progres- (Kullmann, 2002). Genetic loci have been iden-
sion lasting at least 3 months after the conclusion tified in subpopulations of particular epilepsy
of the adenosine supplementation, whereas seizure syndromes, but only account for a fraction of the
rate and severity continued to progress in all con- incidence, and are not well correlated with seizure
trol animals (Williams-Karnesky et al., 2013). This severity and comorbidities (Myers and Mefford,
finding for the first time suggested a potent antiep- 2015). In general, large-scale genomewide stud-
ileptogenic role of adenosine. Maladaptive changes ies have not identified broadly applicable genomic
in adenosine metabolism are tightly linked to epi- variations that would indicate a common genetic
leptogenesis, and restoration of adenosine homeo- risk in epilepsy (Kasperaviciute et al., 2010; Mefford
stasis therefore presents a potentially powerful et  al., 2010; Cavalleri et  al., 2007). However, a
means to prevent epilepsy or its progression. recent study suggests that genetic variants of the
The influence of adenosine on biologic systems Adk gene are associated with an increased risk
is complex. It may be pro- or anti-inflammatory for the development of posttraumatic epilepsy
(Hasko et  al., 2005)  and can inhibit or activate (Diamond et  al., 2015). While the role of genet-
synaptic activity (Sebastiao and Ribeiro, 2009). ics in epilepsy and epileptogenesis needs further
Adenosine concentration is under strict local con- investigation, changes in gene expression and
trol, with both intra- and extracellular enzymes regulation might play an additional role. Studies
contributing to the maintenance of adenosine of gene expression changes following seizures, in
“tone.” While our understanding of the local the latent phase of epileptogenesis before spon-
mix of adenosine receptors and their subcellu- taneous seizures develop, and in the epileptic
lar localization explains some of the apparently brain agree that many genes are (dys)regulated in
211

Chapter 23: Ketogenic Diet, Adenosine, Epigenetics, and Antiepileptogenesis 211

the epileptogenic brain (Lukasiuk and Pitkanen, other disease processes as well (Lim and Song,
2004). However, the specific genes regulated are 2012; Hardy and Tollefsbol, 2011). Food-derived
not consistent across studies, regardless of species bioactive compounds can act as substrate donors
or model specificity (Aronica and Gorter, 2007). or reaction inhibitors (Choi and Friso, 2010), and
Pathway analyses across multiple epilepsy studies dietary imbalances can promote or limit disease
have revealed that despite variations in the specific progression. Thus, epigenetic regulation of gene
genes regulated, certain functional pathways are expression not only drives the development of our
commonly identified across studies (Aronica and gross anatomy but also shapes our intellectual,
Gorter, 2007). Thus, expression changes targeting emotional, and physiologic phenotype.
intra- and extracellular signaling, transcription
and protein biosynthesis, and immune responses K E T O G E N I C D I E T,
are all well represented in all time windows evalu- A D E N O S I N E H O M E O S TA S I S ,
ated (Aronica and Gorter, 2007). This network A N D A N T I E P I L E P TO G E N E S I S
view of epilepsy risk and development presents a IN TEMPORAL LOBE
model that can account for the confounding fac- EPILEPSY
tors in genomic studies of epilepsy, including phe- The most common form of focal epilepsy,
notypic inconsistencies associated with heritable with about 80% of seizures originating in the
risk factors, and the variable susceptibility and dis- hippocampus—a critical structure for consolida-
ease progression in acquired epilepsies. tion, storage, and retrieval of memories—temporal
Epigenetic changes are biochemical altera- lobe epilepsy (TLE) is refractory to conventional
tions of the DNA or the chromatin structure, antiepilepsy drugs in up to 40% of patients, leav-
which do not affect the coding sequence of the ing removal of the hippocampus as the antiseizure
DNA, but contribute to the regulation of gene treatment of last resort for many. The growing field
transcription and entire networks. Epigenetic of epigenetic research has led to the discovery that
changes include DNA methylation and hydroxy- significant epigenetic changes occur in the epilep-
methylation, as well as histone methylation and tic hippocampus and may present a unique thera-
acetylation (Jaenisch and Bird, 2003; Kiefer, 2007). peutic opportunity.
Importantly, epigenetic changes may affect several In both clinical and experimental settings the
genes thought to represent a risk factor for epi- epileptic hippocampus was shown to be character-
lepsy simultaneously. In contrast to genetic muta- ized by altered DNA methylation when compared
tions, epigenetic changes are potentially reversible with nonepileptic hippocampus. In human TLE
and may constitute a novel target for therapeutic samples from resected hippocampus, gene targets
intervention. DNA methylation was once believed with both increased and decreased methylation
to be stable, but a critical role for the dynamic were identified, with approximately 80% of signifi-
regulation of DNA methylation has been identi- cantly regulated sites hypermethylated, an equally
fied in learning and memory (Roth and Sweatt, prominent signature in sclerotic and nonsclerotic
2009; Day and Sweatt, 2010). Environmental dis- hippocampal tissue (Miller-Delaney et  al., 2015).
ruption of physiologic epigenetic regulation has In rodent models of TLE, similar patterns of hyper-
been implicated in a broad range of diseases, and methylation in the epileptic hippocampus have
has been widely studied in the context of exposure been demonstrated in the KASE model (Williams-
to environmental toxins (Pacchierotti and Spano, Karnesky et al., 2013; Ryley Parrish et al., 2013) and
2015; Bollati and Baccarelli, 2010). In rodent stud- pilocarpine status epilepticus models (Kobow et al.,
ies environmental factors such as maternal stimu- 2013; Lusardi et al., 2015). A receptor-independent
lation and social interaction have been linked to role for adenosine in regulating DNA methyla-
epigenetic changes, which in turn influence stress tion has recently been demonstrated (Williams-
hormones, brain development, and neuropsychi- Karnesky et al., 2013). Adenosine has a mass effect
atric phenotypes (Curley et al., 2011). Clinical evi- on biochemical enzyme reactions and is an obliga-
dence supports the conclusions from those rodent tory end product of the S-adenosylmethionine
studies (McGowan et al., 2009; Zhang et al., 2013). (SAM) dependent transmethylation pathway
Beyond exposure to overt toxins, diet can influ- (Figure 23.1), necessary for the transfer of methyl
ence the epigenome. Naturally occurring com- groups onto DNA (Boison et al., 2002; Mato et al.,
pounds in our food have a known influence on 2008). As predicted by this biochemical pathway,
epigenetic regulation. Their role has been exten- exogenous application of either adenosine or its
sively studied in cancer risk and treatment, but complementary end product homocysteine inhib-
their general mechanisms are likely to influence its the reaction and reduces DNA methylation,
212

212 section III: Ketogenic Diet in the Laboratory

Methionine
SAM adenosyltransferase

X-CH3
DNMT
Met

SAH
Methionine
SAH synthase
hydrolase

ADO
HCY
ADK

AMP

FIGURE 23.1 Transmethylation pathway. The amino acid methionine (Met) is converted to S-adenosylmethionine
(SAM), a methyl group donor, by methionine adenosyl transferase. DNA methyltransferase (DNMT) enzymes catalyze
the transfer of a methyl group from SAM to DNA, with S-adenosyl homocysteine (SAH) as a rate-limiting end product.
The enzyme SAH hydrolase catalyzes SAH to adenosine (ADO) and homocysteine (HCY) in a reversible reaction.
Thus, ADO and HCY homeostasis can regulate DNMT activity and DNA methylation levels. ADO is metabolized
by adenosine kinase (ADK) into adenosine monophosphate (AMP), and HCY is recycled back to Met by methionine
synthase. Methionine synthase and methionine adenosyltransferase activity depends on the availability of bioactive
compounds such as Vitamin B12.

whereas addition of the methyl group donor SAM homocysteine (Gill and Schatz, 1985; Schatz et al.,
increases DNA methylation in the naïve rodent 1983; Sellinger et  al., 1984). In the KASE model,
brain (Williams-Karnesky et  al., 2013). Similar direct ventricular administration of adenosine for
effects can be obtained pharmacologically using 10  days significantly reduced epilepsy disease
an ADK inhibitor (5-iodotubercidin, ITU), which progression, including the progressive increase of
reduced hippocampal DNA methylation by 50%, spontaneous convulsive seizures and additional
even in the presence of caffeine, a nonspecific mossy fiber sprouting, and restored global DNA
adenosine receptor inhibitor, further demonstrat- methylation levels to control levels, lasting well
ing that the effects of adenosine on methylation are after the conclusion of the adenosine delivery
receptor independent. The ADK is up-regulated (Williams-Karnesky et  al., 2013). These findings
within epileptogenic brain areas in rodent models show that global DNA methylation levels are under
of TLE and in human TLE with hippocampal scle- the direct control of adenosine, and that disrup-
rosis (Boison, 2012), resulting in adenosine defi- tion of adenosine homeostasis (due to ADK up-
ciency, increasing the flux of methyl groups through regulation at the epileptogenic focus) affects DNA
the transmethylation pathway. Concurrently, methylation levels and alters gene expression in the
adenosine deficiency shifts the equilibrium of the epileptic brain.
S-adenosylhomocysteine (SAH) hydrolase reac- As described above, through mechanisms
tion away from the formation of SAH (Mandaviya that are not yet well understood, strict adherence
et  al., 2014), which is also known to block DNA to a KD reduces seizures through an adenosine-
methyltransferase activity by product inhibi- receptor dependent mechanism (Masino et  al.,
tion (James et  al., 2002), thereby reinforcing the 2011). Using a kindling model of epileptogenesis,
increase in DNA methylation in the epileptic the KD also delayed the acquisition of kindling in
brain. Seizures resulting from the proconvulsant mice, an effect that persisted even after a return
L-methionine-dl-sulfoximine, which increases to a standard lab diet, while the conventional
the methylation flux by increasing the SAM/ antiepileptogenic drug valproic acid attenuated
SAH ratio, can be blocked by adenosine and only the seizures without blocking the kindling
213

Chapter 23: Ketogenic Diet, Adenosine, Epigenetics, and Antiepileptogenesis 213

process. These data demonstrate persistent designed to directly restore adenosine homeosta-
effects of the KD that are not merely due to sei- sis, with the goal of developing a novel class of
zure suppression (Lusardi et al., 2015). When fed antiepileptogenic drugs.
to rats following status epilepticus, the KD not
only reduced spontaneous seizure development
but also reduced DNA methylation levels both REFERENCES
during diet administration and after a return to Aronica, E., and Gorter, J.A. (2007). Gene expression
profile in temporal lobe epilepsy. Neuroscientist
standard diet (Kobow et al., 2013; Lusardi et al.,
13, 100–108.
2015). Though a direct link between the KD and
Aronica, E., Zurolo, E., Iyer, A., et  al. (2011).
DNA methylation levels must still be demon- Upregulation of adenosine kinase in astrocytes in
strated, when evaluated together these results experimental and human temporal lobe epilepsy.
show that the lasting effects of the KD may be Epilepsia 52, 1645–1655.
conferred via adenosine regulation of the DNA Barborka, C. (1930). Epilepsy in adults. Arch Neurol
methylome, supporting a key mechanism impli- Psychiatry 23, 904–914.
cated in epilepsy and epileptogenesis. Boison, D. (2008). The adenosine kinase hypothesis of
epileptogenesis. Prog Neurobiol 84, 249–262.
CONCLUSION Boison, D. (2012). Adenosine dysfunction in epilepsy.
With up to 35% of persons with epilepsy consid- Glia 60, 1234–1243.
ered to be refractory to treatment and no therapies Boison, D. (2013). Adenosine kinase: exploitation for
available that prevent epilepsy or its progression, therapeutic gain. Pharmacol Rev 65, 906–943.
the novel epigenetic functions of KD therapy dis- Boison, D., Scheurer, L., Zumsteg, V., Rulicke, T.,
Litynski, P., Fowler, B., Brandner, S., and Mohler,
cussed here might be of therapeutic value to not
H. (2002). Neonatal hepatic steatosis by disruption
only relieve the seizure burden in patients with of the adenosine kinase gene. Proc Natl Acad Sci
epilepsy but also to modify the development of epi- USA 99, 6985–6990.
lepsy with its sequelae of pharmacoresistance and Bollati, V., and Baccarelli, A. (2010). Environmental
the development of epilepsy-associated comorbid- epigenetics. Heredity 105, 105–112.
ities. The KD presents a powerful adjunct therapy Cavalleri, G.L., Weale, M.E., Shianna, K.V., et al. (2007).
to existing pharmacologic and surgical approaches Multicentre search for genetic susceptibility loci in
to seizure relief. Renewed interest in the KD has sporadic epilepsy syndrome and seizure types:  a
led to refinements in the diet formulation and case-control study. Lancet Neurol 6, 970–980.
administration (Kossoff et  al., 2009; Wibisono Choi, S.W., and Friso, S. (2010). Epigenetics:  a new
et  al., 2015)  and improved understanding of the bridge between nutrition and health. Adv Nutr
potential positive and negative interactions with 1, 8–16.
conventional antiepileptic drugs (Morrison et al., Coppola, G., Veggiotti, P., Cusmai, R., et al. (2002). The
ketogenic diet in children, adolescents and young
2009; van der Louw et al., 2015), improving com-
adults with refractory epilepsy:  an Italian multi-
pliance and seizure suppression rates. However, centric experience. Epilepsy Res 48, 221–227.
the diet requires close monitoring by physicians Cunha, R.A. (2001). Regulation of the ecto-
and dietitians, and seemingly minor deviations nucleotidase pathway in rat hippocampal nerve
from the ketogenic regimen can negate its benefi- terminals. Neurochem Res 26, 979–991.
cial effects. While a “ketogenic diet in a pill” may Curley, J.P., Jensen, C.L., Mashoodh, R., and
be unlikely, ongoing studies to understand the bio- Champagne, F.A. (2011). Social influences on
chemical mechanisms of the KD are an essential neurobiology and behavior: epigenetic effects dur-
step in the continued refinement of antiepileptic ing development. Psychoneuroendocrinology 36,
and antiepileptogenic therapies. The neuroprotec- 352–371.
tive mechanisms of the KD are varied, and diet Day, J.J., and Sweatt, J.D. (2010). DNA methylation and
efficacy may rely on their combined influences. memory formation. Nat Neurosci 13, 1319–1323.
Among the metabolites regulated by the KD, Diamond, M.L., Ritter, A.C., Jackson, E.K., Conley,
Y.P., Kochanek, P.M., Boison, D., and Wagner, A.K.
however, adenosine has both a direct relevance to
(2015). Genetic variation in the adenosine regu-
seizure suppression by A1R activation and an indi- latory cycle is associated with posttraumatic epi-
rect influence on epilepsy and epileptogenesis via lepsy development. Epilepsia 56, 1198–1206.
regulation of DNA methylation. A clearer under- During, M.J., and Spencer, D.D. (1992). Adenosine: a
standing of how KD therapy affects adenosine potential mediator of seizure arrest and postictal
metabolism and its epigenetic sequelae may help refractoriness. Ann Neurol 32, 618–624.
us understand adenosine dysregulation in epi- Fedele, D.E., Gouder, N., Guttinger, M., Gabernet, L.,
lepsy, and may guide the development of therapies Scheurer, L., Rulicke, T., Crestani, F., and Boison, D.
214

214 section III: Ketogenic Diet in the Laboratory

(2005). Astrogliosis in epilepsy leads to overex- Kossoff, E.H., and Hartman, A.L. (2012). Ketogenic
pression of adenosine kinase, resulting in seizure diets:  new advances for metabolism-based thera-
aggravation. Brain, 128, 2383–2395. pies. Curr Opin Neurol 25, 173–178.
Fedele, D.E., Li, T., Lan, J.Q., Fredholm, B.B., and Kossoff, E.H., Zupec-Kania, B.A., Amark, P.E., et  al.
Boison, D. (2006). Adenosine A(1). receptors are (2009). Optimal clinical management of children
crucial in keeping an epileptic focus localized. Exp receiving the ketogenic diet:  recommendations
Neurol, 200, 184–190. of the International Ketogenic Diet Study Group.
Fredholm, B.B., Chen, J.F., Cunha, R.A., Svenningsson, Epilepsia 50, 304–317.
P., and Vaugeois, J.M. (2005). Adenosine and brain Kullmann, D.M. (2002). Genetics of epilepsy. J Neurol
function. Int Rev Neurobiol 63, 191–270. Neurosurg Psychiatry, 73 (Suppl 2), II32–35.
Gill, M.W., and Schatz, R.A. (1985). The effect of diaz- Li, T., Quan Lan, J., Fredholm, B.B., Simon, R.P., and
epam on brain levels of S-adenosyl-L-methionine Boison, D. (2007). Adenosine dysfunction in
and S-adenosyl-L-homocysteine: possible correla- astrogliosis: cause for seizure generation? Neuron
tion with protection from methionine sulfoximine Glia Biol 3, 353–366.
seizures. Research communications in chemical Li, T., Ren, G., Lusardi, T., Wilz, A., Lan, J.Q., Iwasato,
pathology and pharmacology 50, 349–363. T., Itohara, S., Simon, R.P., and Boison, D. (2008).
Gouder, N., Fritschy, J.M., and Boison, D. (2003). Adenosine kinase is a target for the prediction and
Seizure suppression by adenosine A1 receptor prevention of epileptogenesis in mice. J Clin Invest
activation in a mouse model of pharmacoresistant 118, 571–582.
epilepsy. Epilepsia 44, 877–885. Lim, U., and Song, M.A. (2012). Dietary and lifestyle
Gouder, N., Scheurer, L., Fritschy, J.M., and Boison, D. factors of DNA methylation. Methods Mol Biol
(2004). Overexpression of adenosine kinase in epi- 863, 359–376.
leptic hippocampus contributes to epileptogenesis. Lloyd, H.G., and Fredholm, B.B. (1995). Involvement
J Neurosci 24, 692–701. of adenosine deaminase and adenosine kinase in
Hardy, T.M., and Tollefsbol, T.O. (2011). Epigenetic regulating extracellular adenosine concentration in
diet:  impact on the epigenome and cancer. rat hippocampal slices. Neurochem Int 26, 387–395.
Epigenomics 3, 503–518. Lukasiuk, K., and Pitkanen, A. (2004). Large-scale
Hasko, G., Pacher, P., Vizi, E.S., and Illes, P. (2005). analysis of gene expression in epilepsy research: is
Adenosine receptor signaling in the brain immune synthesis already possible? Neurochem Res 29,
system. Trends Pharmacol Sci 26, 511–516. 1169–1178.
Huber, A., Padrun, V., Deglon, N., Aebischer, P., Mohler, Lusardi, T.A., Akula, K.K., Coffman, S.Q., Ruskin, D.N.,
H., and Boison, D. (2001). Grafts of adenosine- Masino, S.A., and Boison, D. (2015). Ketogenic
releasing cells suppress seizures in kindling epi- diet prevents epileptogenesis and disease progres-
lepsy. Proc Natl Acad Sci USA 98, 7611–7616. sion in adult mice and rats. Neuropharmacology
Jaenisch, R., and Bird, A. (2003). Epigenetic regulation 99, 500–509.
of gene expression:  how the genome integrates Mandaviya, P.R., Stolk, L., and Heil, S.G. (2014).
intrinsic and environmental signals. Nat Genet, 33 Homocysteine and DNA methylation: a review of
Suppl, 245–254. animal and human literature. Mol Genet Metab
James, S.J., Melnyk, S., Pogribna, M., Pogribny, 113, 243–252.
I.P., and Caudill, M.A. (2002). Elevation in S- Masino, S.A., Kawamura, M., Wasser, C.D., Pomeroy,
adenosylhomocysteine and DNA hypometh- L.T., and Ruskin, D.N. (2009). Adenosine, keto-
ylation:  potential epigenetic mechanism for genic diet and epilepsy: the emerging therapeutic
homocysteine-related pathology. J Nutr. 132, relationship between metabolism and brain activ-
2361S–2366S. ity. Curr Neuropharmacol 7, 257–268.
Kasperaviciute, D., Catarino, C.B., Heinzen, E.L., et al. Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin,
(2010). Common genetic variation and suscepti- D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and
bility to partial epilepsies: a genome-wide associa- Boison, D. (2011). A ketogenic diet suppresses sei-
tion study. Brain 133, 2136–2147. zures in mice through adenosine A(1). receptors. J
Kiefer, J.C. (2007). Epigenetics in development. Dev Clin Invest, 121, 2679–2683.
Dyn 236, 1144–1156. Masino, S.A., and Rho, J.M. (2012) Mechanisms
Kobow, K., Kaspi, A., Harikrishnan, K.N., et al. (2013). of ketogenic diet action. In Jasper’s Basic
Deep sequencing reveals increased DNA methyla- Mechanisms of the Epilepsies, J.L. Noebels, M.
tion in chronic rat epilepsy. Acta Neuropathol 126, Avoli, M.A. Rogawski, R.W. Olsen and A.V.
741–76. Delgado-Escueta eds. (Bethesda:  National
Kochanek, P.M., Vagni, V.A., Janesko, K.L., et  al. Center for Biotechnology Information), pp.
(2006). Adenosine A1 receptor knockout mice 1001–1022.
develop lethal status epilepticus after experimental Mato, J.M., Martinez-Chantar, M.L., and Lu, S.C.
traumatic brain injury. J Cereb Blood Flow Metab (2008). Methionine metabolism and liver disease.
26, 565–575. Annu Rev Nutr 28, 273–293.
215

Chapter 23: Ketogenic Diet, Adenosine, Epigenetics, and Antiepileptogenesis 215

McGowan, P.O., Sasaki, A., D’Alessio, A.C., Dymov, Sellinger, O.Z., Schatz, R.A., Porta, R., and Wilens, T.E.
S., Labonte, B., Szyf, M., Turecki, G., and Meaney, (1984). Brain methylation and epileptogenesis: the
M.J. (2009). Epigenetic regulation of the gluco- case of methionine sulfoximine. Ann Neurol, 16
corticoid receptor in human brain associates with (Suppl), S115–120.
childhood abuse. Nat Neurosci 12, 342–348. Sirven, J., Whedon, B., Caplan, D., Liporace, J.,
Mefford, H.C., Muhle, H., Ostertag, P., et  al. (2010). Glosser, D., O’Dwyer, J., and Sperling, M.R.
Genome-wide copy number variation in epi- (1999). The ketogenic diet for intractable epi-
lepsy:  novel susceptibility loci in idiopathic lepsy in adults:  preliminary results. Epilepsia 40,
generalized and focal epilepsies. PLoS Genet 6, 1721–1726.
e1000962. Sofroniew, M.V., and Vinters, H.V. (2010).
Miller-Delaney, S.F., Bryan, K., Das, S., McKiernan, Astrocytes:  biology and pathology. Acta
R.C., Bray, I.M., Reynolds, J.P., Gwinn, R., Stallings, Neuropathol 119, 7–35.
R.L., and Henshall, D.C. (2015). Differential DNA Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic
methylation profiles of coding and non-coding diet as a treatment paradigm for diverse neuro-
genes define hippocampal sclerosis in human tem- logical disorders. Front. Pharmacol. 3, 59.
poral lobe epilepsy. Brain 138, 616–631. Szybala, C., Pritchard, E.M., Lusardi, T.A., Li, T.,
Morrison, P.F., Pyzik, P.L., Hamdy, R., Hartman, A.L., Wilz, A., Kaplan, D.L., and Boison, D. (2009).
and Kossoff, E.H. (2009). The influence of concur- Antiepileptic effects of silk-polymer based ade-
rent anticonvulsants on the efficacy of the keto- nosine release in kindled rats. Exp. Neurol., 219,
genic diet. Epilepsia 50, 1999–2001. 126–135.
Myers, C.T., and Mefford, H.C. (2015). Advancing van der Louw, E.J., Desadien, R., Vehmeijer, F.O.,
epilepsy genetics in the genomic era. Genome van der Sijs, H., Catsman-Berrevoets, C.E., and
medicine 7, 91. Neuteboom, R.F. (2015). Concomitant lamotrig-
Newby, A. (1984). Adenosine and the concept of “retal- ine use is associated with decreased efficacy of the
iatory metabolites.” Trends Biochem Sci 9, 42–44. ketogenic diet in childhood refractory epilepsy.
Pacchierotti, F., and Spano, M. (2015). Environmental Seizure 32, 75–77.
impact on DNA methylation in the germline: state Van Gompel, J.J., Bower, M.R., Worrell, G.A., et  al.
of the art and gaps of knowledge. BioMed Res Int, (2014). Increased cortical extracellular adenosine
2015, 123484. correlates with seizure termination. Epilepsia 55,
Peterman, M.G. (1924). The ketogenic diet in the 233–244.
treatment of epilepsy, a preliminary report. Am J Wheless, J.W. (2008). History of the ketogenic diet.
Dis Child 28, 28–33. Epilepsia, 49 (Suppl 8), 3–5.
Peterman, M.G. (1925). The ketogenic diet in epilepsy. Wibisono, C., Rowe, N., Beavis, E., Kepreotes, H.,
JAMA 84, 1979–1983. Mackie, F.E., Lawson, J.A., and Cardamone,
Roth, T.L., and Sweatt, J.D. (2009). Regulation of chro- M. (2015). Ten-year single-center experience
matin structure in memory formation. Curr Opin of the ketogenic diet:  factors influencing effi-
Neurobiol 19, 336–342. cacy, tolerability, and compliance. J Pediatr, 166,
Ryley Parrish, R., Albertson, A.J., Buckingham, S.C., 1030–1036, e1031.
Hablitz, J.J., Mascia, K.L., Davis Haselden, W., and Williams-Karnesky, R.L., Sandau, U.S., Lusardi, T.A.,
Lubin, F.D. (2013). Status epilepticus triggers early Lytle, N.K., Farrell, J.M., Pritchard, E.M., Kaplan,
and late alterations in brain-derived neurotrophic D.L., and Boison, D. (2013). Epigenetic changes
factor and NMDA glutamate receptor Grin2b induced by adenosine augmentation therapy pre-
DNA methylation levels in the hippocampus. vent epileptogenesis. J Clin Invest 123, 3552–3563.
Neuroscience 248C, 602–619. Wilz, A., Pritchard, E.M., Li, T., Lan, J.Q., Kaplan, D.L.,
Schatz, R.A., Wilens, T.E., Tatter, S.B., Gregor, P., and and Boison, D. (2008). Silk polymer-based ade-
Sellinger, O.Z. (1983). Possible role of increased nosine release:  therapeutic potential for epilepsy.
brain methylation in methionine sulfoximine epi- Biomaterials 29, 3609–3616.
leptogenesis:  effects of administration of adenos- Zhang, T.Y., Labonte, B., Wen, X.L., Turecki, G., and
ine and homocysteine thiolactone. J Neurosci Res Meaney, M.J. (2013). Epigenetic mechanisms for
10, 437–447. the early environmental regulation of hippocam-
Sebastiao, A.M., and Ribeiro, J.A. (2009). Tuning and pal glucocorticoid receptor gene expression in
fine-tuning of synapses with adenosine. Curr rodents and humans. Neuropsychopharmacology
Neuropharmacol 7, 180–194. 38, 111–123.
216

24
Ketogenic Diet, Aging, and Neurodegeneration
K U I X U, M D , P H D , J O S E P H C . L A M A N N A , P H D ,
A N D M I C H E L L E A . P U C H OW I C Z , P H D

OV E RV I E W also maintain that ketone bodies can act as signal-


It has been established for over 50 years that brain ing molecules that target HIF1α-related cellular
can utilize ketone bodies under carbohydrate- defenses and regulatory systems. HIF1α activa-
(glucose-) sparing conditions (Cahill and Owen, tion can result in stabilization of the “redox state”
1967; Owen et al., 1967; Randle et al., 1963). The of the cell (Semenza, 2011). We have previously
use of ketogenic diet (KD) to treat intractable shown neuroprotection by ketosis (in model of
epilepsy in children continues to be an option middle cerebral artery occlusion, MCAO) in rat.
for clinicians (DeVivo et al., 1978; Freeman et al., The hypothesis that the mechanistic link between
1998; Lennox, 1928; Millichap et al., 1964; Tallian ketosis and neuroprotection is through HIF1α and
et al., 1998; Schwartzkroin, 1999), but the mecha- its downstream effects on metabolic enzymes was
nisms of action remain diverse (Bridge, 1931; established (Puchowicz et al., 2008).
Jarrett et  al., 2008; Lund et  al., 2009; Maalouf
et al., 2009; Milder and Patel, 2012; Millichap et al., K E TO N E B O D I E S A R E
1964; Nakazawa et  al., 1983; Prins and Hovda, A LT E R N AT E E N E R G Y
2009; Yudkoff et al., 2007; Masino and Rho, 2012; S U B S T R AT E S T O   G L U C O S E
Maalouf et  al., 2007; Stafstrom and Rho, 2012). Ketone bodies (or the state of ketosis) have been
The mechanistic link of ketosis and neuroprotec- shown to be neuroprotective following oxidative
tion also remains unclear and exploratory. The stress and metabolic challenges, such as those asso-
mechanisms appear to be related to the change in ciated with stroke, ischemia, injury, Alzheimer’s
the regulation of the cell’s stress responses (Milder disease, Parkinson’s disease, glucose transporter
et al., 2010) and the changes in oxidative (glucose) deficiency, and seizures (Maalouf et  al., 2009;
metabolism (Prins and Hovda, 2009; Puchowicz Prins and Hovda, 2009; Kashiwaya et  al., 2000;
et  al., 2008; Bough and Rho, 2007; Masino and Cunnane et al., 2011). Ketone bodies are alternate
Rho, 2012). Neuroprotection by ketosis is thought energy substrates to glucose and thus they have the
to be associated with improved mitochondrial potential to restore energy balance via stabilization
function, decreased reactive oxygen species (ROS) of ATP supply. They are thought to be more effi-
and apoptotic and inflammatory mediators, and cient energy substrates to glucose for utilization
increased protective pathways (Veech, 2004; Julio- by brain (Cahill and Veech, 2003; Kashiwaya et al.,
Amilpas et al., 2015). 2000; Sato et  al., 1995; Veech et  al., 2001; Veech,
This review focuses on two potential mecha- 2004). One application for the use of ketone bodies
nisms that elucidate the neuroprotective effects as an alternative substrate is for those with aging
of ketosis in the brain. We also concentrate on defects in glucose metabolism.
the applications toward the treatment of the aged Hypometabolism is a clinical presentation in
brain, and present current data from rodent stud- some Alzheimer’s disease patients and is thought
ies where KD was used to induce a chronic state of to be associated with defects in glucose metabo-
ketosis and where neuroprotection was assessed. lism (Cunnane et  al., 2011). Studies measuring
Our view is that the neuroprotection associated the ratio of 13C-amino acid /13C- lactate in brain
with diet-induced ketosis is mainly through sta- showed the fraction of amino acid carbon derived
bilization of glucose metabolism and downstream from glucose decreased with ketosis, reflecting
metabolic pathways such as the citric acid cycle and the utilization of ketones as carbon precursors
cytosolic-mitochondrial redox shuttle systems. We for amino acid synthesis (aspartate, glutamine,
217

Chapter 24: Ketogenic Diet, Aging, and Neurodegeneration 217

glutamate, GABA) (Yudkoff et al., 2007). The neu- Vannucci and Simpson, 2003)  and moderate to
roprotective role of ketone bodies may be through high blood levels of ketosis (2–6 mM) (Leino et al.,
improvement in metabolic efficiency, by sparing 2001; Prins and Hovda, 2009). More recently, we
glucose and the degradation of muscle-derived have established in our rat model of ketosis that
amino acids for substrates. metabolic adaptations also include a correla-
Ketone body utilization has properties that tion between cerebral metabolic rate for glucose
are considered metabolically favorable over glu- (CMRglc) and blood ketone levels (Zhang et  al.,
cose: (1) the direct generation of NADH, FADH2 2013). Using positron emission tomography imag-
and succinate, which makes it readily available ing technology we showed a decrease of almost
(compartmentalized) to the mitochondria for 10% in CMRglc for each 1-mM increase of total
energy production; (2)  the direct generation of plasma ketone bodies in cortical and cerebellar
acetyl-CoA, which enters the citric acid cycle via brain regions. Together with our meta-analysis,
the citrate synthase reaction, thus relieving oxi- these data revealed that the degree and duration
dative stress induced by pyruvate dehydrogenase of ketosis played a major role in determining the
inhibition, as well as making available ATP and corresponding change in CMRglc with ketosis and
lactate generated from glycolysis for other func- that preconditioning with KG diet resulted in sig-
tions, such as the shuttling of substrates between nificant glucose sparing (Zhang et al., 2013; Zhang
neurons and glia; (3)  having the potential to et al., 2015).
reduce mitochondrial ROS production associated
with reperfusion injury; and (4)  stabilization of O X I D AT I V E S T R E S S
HIF1α independent of hypoxia. I N D U C E D A LT E R E D
G L U C O S E M E TA B O L I S M
U S E O F   K E TO G E N I C Supply of energy substrate to the mitochondria
D I E T TO   I N D U C E is critical, especially during conditions of high
K E TO S I S :   A   M O D E L energy demand where glucose metabolism may be
O F   S TA B I L I Z E D C H R O N I C deficient, such as with oxidative injury. The abil-
K E TO S I S ity of the central nervous system to recover from
The metabolic modulations associated with ketone an ischemic and/or hypoxic event is its capac-
bodies require a state of stable ketosis. The use of ity to recover from metabolic stress. It is well
a KD is an approach to establish chronic stable known that the status of cellular bioenergetics of
ketosis. Diet-induced approaches avoid the con- the neurovascular unit is a major determinate of
straints (sodium or fluid overloads, pH imbal- the pathophysiologic outcome. Stoichiometric
ance, or starvation with chronic fasting) of using analysis would predict that glycolysis cannot sus-
alternate modes of inducing ketosis, such as with tain NADH demand (energy demand) for more
infusions of ketone bodies or ketone body precur- than a few hours, depending on glycogen stores.
sors (sodium salts or acids), or long-term fasting The dogma of tissue reperfusion for cell sur-
(Desrochers et  al., 1995; Puchowicz et  al., 2000). vival together with associated oxidative damage,
It is well described that ketosis can be induced immune responses, and increased risk for cell
in rodents and humans by fasting or feeding death remains problematic.
KDs (Gano et  al., 2014; Leino et  al., 200; Milder Under pathologic conditions the brain’s energy
et  al., 2010; Pan et  al., 2000; Puchowicz et  al., demand and supply are mismatched, resulting
2007; Tallian et  al., 1998; Veech, 2004; Veldhorst in energy inefficiency. For example, survival and
et al., 2010; Yudkoff et al., 2007). We have previ- recovery of neurologic function after resuscita-
ously reported that 3 weeks of diet-induced stable tion following cardiac arrest is limited by the
ketosis is necessary to induce metabolic adapta- ability of the central nervous system to recover
tions in brain of rats resulting in neuroprotection from an ischemic event due to metabolic stress
(Puchowicz et  al., 2007; Puchowicz et  al., 2008; to brain. Following cardiac arrest and resuscita-
Zhang et al., 2013; Xu et al., 2012), which is consis- tion, increased glycolytic rates are associated with
tent with another study (Milder et al., 2010). This lactic acidosis and downstream metabolic blocks
regime can be inferred to as a preconditioning in energetics. After 24 hours of reperfusion there
state where blood concentrations of ketone bod- is an “apparent” defect in glucose metabolism,
ies remain stable over time. Adaptation to chronic which may continue and occur as a second-
ketosis includes up-regulation of monocarboxyl- ary response to oxidative stress-induced injury
ate transporters (MCT) at the blood-brain bar- (Selman et  al., 1990). At this stage of oxidative
rier (Gjedde and Crone, 1975; Leino et  al., 2001; damage there is a decrease in CMRglc (Bentourkia
218

218 section III: Ketogenic Diet in the Laboratory

100

90
KG

Survival rate (%)


80

70
STD
Number of deaths Survival rate
(4d/total)
60 <1d 1–2 d 2–3 d 3–4 d
STD 2 2 3 2 55% (10/11)
KG 1 0 0 0 86% (6/7)
50
0 1 2 3 4
Recovery (days)

FIGURE 24.1 Overall survival rates 4 days following cardiac arrest and resuscitation. The rats fed with 3 weeks of
ketogenic diet (KD) had a significantly higher survival rates compared with the rats fed with standard diet (STD) (86%
vs. 55%, p < .05, Wilcoxon survival analysis) (Xu et al., 2012).

et al., 2000; DiVivo et al., 1973; Willis et al., 2002). can lead to irreversible neurological deficit.
Mitochondria are among the most susceptible Anaplerosis, a process that balances cataplero-
organelles to oxidative stress. Brain mitochon- sis, ordinarily maintains/supplies intermediates
drial function is known to decrease 2 days follow- to the citric acid cycle, which becomes vital dur-
ing ischemia reperfusion (Xu et al., 2008), which ing increased leakiness associated with ischemia
more often results in delayed neuronal death after reperfusion injury (Brunengraber and Roe, 2006;
4 days post recovery (Xu et al., 2006). The degree Kasumov et al., 2007) and other neuropathologies.
of neurological deficit is highly dependent on the The balance between cataplerosis and anaplerosis
length of time to restored blood reflow and on the is essential to maintaining energy balance and,
severity of oxidative damage. The generation of hence, mitochondrial function. In a recent study,
ROS and cytotoxic products of lipid peroxidation, we investigated the role of ketosis on the partition-
such as 4-hydroxy-2-nonenal (HNE) and glutathi- ing of glucose and BHB as substrates at the level of
one (GSH), also play a major role in the cell’s abil- the acetyl-CoA and citric acid cycle (Zhang et al.,
ity to recover. In a previous study we have shown 2015). This study revealed that glucose entry into
that KD-induced ketosis resulted in significantly the citric acid cycle was decreased with ketosis,
higher overall survival rates 4  days post cardiac whereas BHB entry was increased. The increased
arrest and resuscitation compared with the stan- contribution of BHB to central metabolism sug-
dard diet group (86% vs. 55%, p < .05, Figure 24.1). gests that carbon backbones from ketones play a
These data support the idea that diet-induced role in the balancing of the citric acid cycle. The
ketosis is neuroprotective under conditions when possibility of increased pyruvate carboxylation
glucose utilization is limited (Xu et al., 2012). via malic enzyme should also be considered as
Studies, past and present, continue to show a mechanism of ketosis in energy stabilization
evidence that altered energy metabolism is asso- (Hassel, 2000).
ciated with aging and/or oxidative damage to
key enzymes that regulate glucose metabolism D I E T- I N D U C E D K E T O S I S
(Cunnane et  al., 2011; Gibson et  al., 1998). The A S   A N E U RO P ROT E C T I V E
imbalance between nonoxidative- and oxidative- S T R AT E G Y I N   T H E
derived ATP is recognized but not clearly under- AGED: CLINICAL
stood (Hemmila and Drewes, 1993; Hempel et al., R E L E VA N C E
1977; Hoxworth et  al., 1999; Xu et  al., 2006; Xu The aging population is at risk for increased mor-
et al., 2008). One explanation may be the lack of bidity and mortality following ischemic or hypoxic
glucose carbon entry (flux) into the citric acid events, such as those related to stroke or other
cycle (i.e., via pyruvate dehydrogenase complex) neurodegenerative conditions. Stroke is a leading
resulting in a dysregulation or “leakiness” of the disease/disability in the United States, as there are
citric acid cycle (cataplerosis). These consequences few (if any) clinical treatment strategies that can
219

Chapter 24: Ketogenic Diet, Aging, and Neurodegeneration 219

STD KG
160

120
Infarct volume (mm3)

80

40

0
STD KG

FIGURE 24.2 The total brain infarct volume artery occlusion (MCAO) in 18-month-old rats fed with ketogenic or
standard diet. Rats were randomly assigned to two diet groups, fed ad libitum, ketogenic (high fat, no carbohydrate; KG)
or standard lab-chow (STD) diet for 3 weeks prior to ischemic stroke. Rats underwent 90 minutes of MCAO, the total
infarct volume was evaluated by triphenyltetrazolium chloride (TTC) staining 24 hours after reperfusion. Results: After
fed with 3 weeks of ketogenic diet, plasma ketone bodies (mM) were increased 5-fold (3.1 ± 0.8 vs. 0.6 ± 0.1, mean ±
SEM, ketogenic diet vs. Standard diet, n = 4 each). After 24 hours of reperfusion following MCAO, the infarct volumes
were 5-fold lower in the KG diet group compared with the STD diet group (22 ± 10 vs. 124 ± 26 mm3, mean ± SEM,
n = 4 each).

reverse cellular damage. All brain cells are suscep- injury, often results in energy imbalances related to
tible to infarction following ischemia-reperfusion dysregulation of glucose (oxidative) metabolism. It
(Pundik et al., 2012). Another example of a condi- could be that ketones are effective against pathol-
tion that results in moderate to severe ischemia/ ogy associated with altered glucose metabolism
reperfusion injury is cardiac arrest and resuscita- (Cahill and Veech, 2003; Prins and Hovda, 2009;
tion, as the brain is exposed to ischemic-hypoxia Sato et  al., 1995; Veech et  al., 2001; Veech, 2004;
and reperfusion. In the United States about one Veech et al., 2012). Ketones may also play a role as
million cardiac arrests occur per year; about half signaling molecules that directly or indirectly act
of the population (aged or adult) having first-time through the regulation of cell salvation pathways,
cardiac arrests will not survive the first few days. such as HIF1α. In a recent study, we showed that
Of those who do survive, about 90% will have ketosis was induced in aged rats (18 months old)
short- or long-term neurological deficits (Chugh following 3 weeks of feeding a KD and that diet-
et  al., 2004; Goldberger et  al., 2008; Safar,1993; induced ketosis was neuroprotective against focal
Zheng et al., 2001). These deficits are more often cerebral ischemia (MCAO; Figure 24.2).
related to oxidative stress induced post resuscita-
tion mortality and delayed selective neuronal cell N E U RO P ROT E C T I O N
loss (Hoxworth et  al., 1999; Xu et  al., 2006; Xu T H R O U G H   S TA B I L I Z AT I O N
et al., 2008). In the aged, failure to recover is espe- OF HIF1Α
cially pronounced, as the aged brain is susceptible The notion that ketone bodies can act as metabolic
to oxidative stress damage as result of declines in signaling molecules in addition to being alternate
repair systems and decreased antioxidant capacity. oxidative substrates to glucose has been considered
Brain pathophysiology associated with oxidative (Shimazu et al., 2013; Bough and Rho, 2007; Gano
stress and injury, such as with ischemic reperfusion et al., 2014; Maalouf et al., 2009; Milder et al., 2010;
220

220 section III: Ketogenic Diet in the Laboratory

Tannahill et al., 2013). The role of HIF1α as a key Semenza, 2001; Sharp et al., 2001; Puchowicz et al.,
regulator of oxygen homeostasis during hypoxia 2008; Helton et al., 2005).
is well described (Agani et al., 2002; Chavez et al., We have reported that HIF1α response to
2000; Chavez et  al., 2006; Semenza, 2007, 2011; hypoxia is blunted in the aged brain due to
Semenza et al., 2000; Sharp et al., 2001). However, increased PHD levels (Ndubuizu et  al., 2010).
the metabolic role of HIF1α on neuroprotection The lack of HIF1α induction following an oxida-
remains unclear. tive insult, such as with reperfusion injury and/
HIF1α is a nuclear factor associated with neu- or aging process, increases vulnerability to oxida-
roprotection via regulation of energy metabolism tive damage that can result in poor recovery or
and is a key regulator of oxygen homeostasis dur- cell death (Xu et al., 2007; Xu et al., 2008). These
ing hypoxia, but may also play a role in neuropro- findings are consistent with other reports on mice
tection following ischemia through gene regulation with neuron-specific knockdown of HIF1α and
(Shi, 2009; Guo et al., 2008). HIF1 activation can subjected to transient focal cerebral ischemia;
also promote cell survival (Bergeron et al., 2000). the results showed increased tissue damage and
The translational impact on understanding the reduced cell survival (Baranova et al., 2007).
role of ketone bodies as metabolic regulators can Neuroprotection by HIF target genes such as
lead to a more definitive approach on treatment erythropoietin (Epo) has been described (Chong
strategies that target defects in energy metabolism et al., 2003a; Ghezzi et al., 2010). As a neuroprotec-
associated with neuropathologies and degenera- tive agent Epo has many functions:  antagonizing
tive disorders. Since the aged population has an glutamate cytotoxic action, enhancing antioxidant
increased risk for ischemic events, such as those enzyme expression, reducing free radical produc-
associated with transient global or focal stroke, the tion rate, and affecting neurotransmitter release
development of treatment strategies that incorpo- (Bartesaghi et al., 2005; Leist et al., 2004). It exerts
rate ketosis may be wise. its neuroprotective effect indirectly through res-
The mechanism of HIF1α stabilization through toration of blood flow or directly by activating
ketosis has been proposed (Puchowicz et  al., transmitter molecules in neurons that also play a
2008) to be most likely through changes in cyto- role in erythrogenesis. Although the mechanism is
plasmic/mitochondrial redox state (Guo et  al., unclear, it is apparent that Epo has anti-apoptotic
2008). This is consistent with others reporting action after central and peripheral nerve injury
stabilization of HIF1α through impairment of pro- (Chong et  al., 2003b). Although apoptosis is not
teasome function, as a result of very low ratios of reversible, early intervention with neuroprotective
alpha-ketoglutarate /fumarate (Serra-Perez et  al., therapeutic procedures such as Epo administra-
2010). A  mechanism that explains diet-induced tion or preconditioning with KD may reduce the
HIF1α stabilization (via KD) is through the feed- number of neurons that undergo apoptosis and
back inhibition of the prolyl-hydroxylase (PHD) thus improve outcome.
reaction by succinate, an intermediate of energy An additional target gene of HIF is the tran-
metabolism (see scheme below). Elevated cellu- scription factor vascular endothelial growth factor
lar levels of succinate and fumarate compete with (VEGF). The VEGF is an endothelial cell-specific
alpha-ketoglutarate PHD binding, resulting in growth factor (Storkebaum et al., 2004), but recent
inhibition of PHD and thus HIF1α stabilization evidence indicates that VEGF also has direct effects
(Hou et  al., 2014; Mills and O’Neill, 2014). The on neuronal and glial cell types (Silverman et al.,
mechanisms of ketosis that are relevant to neuro- 1999). A neuroprotective mechanism of VEGF is
protection and stabilization of HIF1α (via succi- thought to be through increased perfusion of the
nate), need to be further studied. We propose that penumbra via vasodilation and angiogenesis, such
PHD inhibition occurs as a result of ketone body as with ischemic brain insults (Zhang et al., 2002).
utilization by the cell via the activation of aceto- We have previously reported that preconditioning
acetate to acetoacetyl-CoA (via 3-ketoacyl-CoA with KD results in an angiogenic response similar
transferase), which involves the generation of suc- to what we have found with mild hypoxic expo-
cinate in the mitochondria. Once activated, HIF1 sure (Puchowicz et al., 2007). However, we found
modulates energy metabolism through down- no changes in cerebral regional blood flow with
stream regulation of metabolism and related target KD-induced ketosis in adult rats. In another study
genes. HIF1α target genes include those related to we reported that VEGF expression was decreased
angiogenesis, erythropoiesis, glucose metabolism, in cerebral cortex of aged mice (Benderro and
and cell survival (Agani et  al., 2002; Baranova LaManna, 2011). Taken all together, KD might be
et al., 2007; Chavez et al., 2006; Chen et al., 2015; a therapeutic strategy that targets neuroprotective
221

Chapter 24: Ketogenic Diet, Aging, and Neurodegeneration 221

mechanisms through HIF-regulated VEGF, inde- 2008; Julio-Amilpas et al., 2015; Sato et al., 1995;
pendent of modifications in blood perfusion Veech, 2004). Second, ketosis induces the stabili-
(Chong et al., 2003b). zation of HIF1α and its downstream target genes.
One possible link between ketosis and the sta-
DISCUS SION bilization of HIF1α is through feedback product
The mechanism of HIF1 regulation under various inhibition of PHD via elevated tissue succinate
metabolic conditions, such as with stroke, inflam- (Semenza, 2007), as a result of a change in the
mation, and altered glucose metabolism, continues metabolic redox state (see Scheme figure 24.3)
to be explored. We describe two mechanisms that (Guo et al., 2008; Serra-Perez et al., 2010; Mills
explain neuroprotection through diet-induced and O’Neill, 2014). This is consistent with a study
ketosis. First, ketone bodies are alternate energy where mitochondrial redox in hippocampal tis-
substrates to glucose, thus reducing oxidative sue was improved by KD via the activation of NF
stress by relieving metabolic blocks downstream E2-related factor 2(Nrf2) transcription factor, a
of glycolysis at the level of the citric acid cycle and, primary responder to cellular stress (Milder et al.,
hence, enabling mitochondrial electron transfer 2010). One known mechanism of Nrf2 is to up-
and generation of ATP (Semenza, 2011; Prins, regulate GSH biosynthesis. On the other hand,

Cytosol Mitochondrion
Ubiquitin
HIF1α HIF1α 1
OH
VHL
OH Succinate

Proteasome O2 PHD1
PHD2
Fe2+
5 Vit C PHD3 Succinyl-CoA Succinate
Angiogenesis HIF1α α-Ketoglutarate
2
α-Ketoglutarate Fumarate
EPO Malate Malate
OAA OAA
Glut-1
VEGF
5 5 Isocitrate 3 Malate
Malate Aspartate
4 Shuttle
NADH NAD
+ +
NAD NADH
Citrate Oxaloacetate
3
Glutamate Glutamate
Aspartate Aspartate
+
NAD NADH

1 Acetyl-CoA
4
Glucose Pyruvate
Legend 6
NADH

1 Succinate Transporter
2 a-Ketoglutarate/Malate Transporter Lactate NAD
+

3 Aspartate/Glutamate Transporter Succinyl-CoA Succinate


4 Pyruvate Transporter
5 Malate Dehydrogenase 2 Ketone Bodies
BHB, AcAc
AcAc-CoA
6 Hexokinase

FIGURE 24.3 Scheme: Proposed mechanism of neuroprotection by diet-induced ketosis.


Our scheme emphasizes that cytosolic generated succinate (via PHD-coupled activity with alpha-ketoglutarate), must be
balanced by mitochondrial redox of the citric acid cycle (shown in gray text). An accumulation of succinate would result
in an imbalance of cytosolic redox; HIF1α would then be stabilized through feedback inhibition of PHD (text shown in
black, Puchowicz et al., 2008). Ketone body utilization also results in the generation of succinate. Steps 1–5 (circled):
HIF1α stabilization is shown to be through cellular redox, which is balanced by malate-aspartate shuttle. A link to the
stabilization HIF1α is through feedback product inhibition of prolyl-hydroxylase (PHD) via elevated tissue succinate
(Semenza, 2007) or reduced α-ketoglutarate (McFate et al., 2008) (steps 3,4). Thus, an accumulation of succinate (or
reduction in α-ketoglutarate) would result in an altered in redox state (malate aspartate shuttle, step 4) and stabilization
of HIF1α (via inhibition of PHD) (step 5). Other intermediates of energy metabolism acting on HIF1α regulation have
been recently described (Chen et al., 2015; Mills and O’Neill, 2014; Tannahill et al., 2013). The potential mechanism by
which ketosis stabilizes HIF1α is through a shift in “redox state” of the cell via changes in carbon flux into / out of the
malate-aspartate shuttle (Semenza, 2011); a target would include changes in succinate-to-α-ketoglutarate ratio.
222

222 section III: Ketogenic Diet in the Laboratory

oxidizing environments (high levels of ROS) may role in energy balance and the downstream innate
suppress stabilization of HIF1α (Guo et al., 2008). signaling of HIF1α. We view that the mecha-
These findings are consistent with a study show- nism of neuroprotection by KD-induced HIF1α
ing that HIF1α played a role in N-acetylcysteine- is through the regulation and activation of genes
mediated neuroprotection in rats subjected to associated with salvation pathways and stabili-
MCAO (Zhang et al., 2014). zation of energy metabolism. The neuroprotec-
Our view is that there is a feedback regulation tion by ketosis may also be through pathways
on HIF1α activation through shifts in metabolic independent of HIF1α regulation, such as AKT-
redox, such as with KD or fasting which is inde- modulation of NRF2 and nuclear factor kappa
pendent of hypoxia-induced stabilization of HIF1α B (NF-kB), and these mechanisms remain to be
(Mills and O’Neill, 2014). Thus, HIF1α acts as a elucidated.
metabolic sensor through a continued feedback
loop directly coupled to oxidative metabolism via REFERENCES
the citric acid cycle and malate aspartate shuttle Agani, F.H., Puchowicz, M., Chavez, J.C., Pichiule,
systems (Figure 24.3). It has been suggested that P., and LaManna, J. (2002). Role of nitric oxide
HIF1α may also play a role promoting apoptosis in the regulation of HIF-1alpha expression dur-
(Helton et al., 2005; Serra-Perez et al., 2010), how- ing hypoxia. Am J Physiol Cell Physiol 283,
ever there is little known about this mechanism. C178-C186.
Neuroprotection by HIF1α may be cell specific. Baranova, O., Miranda, L.F., Pichiule, P., Dragatsis, I.,
It has been reported that neuron-specific inac- Johnson, R.S., and Chavez, J.C. (2007). Neuron-
specific inactivation of the hypoxia inducible fac-
tivation of the HIF1α results in increased brain
tor 1 alpha increases brain injury in a mouse model
injury, as studied in a mouse model of transient of transient focal cerebral ischemia. J Neurosci 27,
focal cerebral ischemia (Baranova et al., 2007). The 6320–6332.
significance of these findings suggests that HIF1α Bartesaghi, S., Marinovich, M., Corsini, E., Galli, C.L.,
and target genes are regulated distinctly, especially and Viviani, B. (2005). Erythropoietin:  a novel
under pathological conditions. Although HIF1α is neuroprotective cytokine. Neurotoxicology 26,
expressed in neurons and glia, the neuroprotective 923–928.
efficacy of a KG diet (via HIF1α) may be cell spe- Benderro, G.F., and LaManna, J.C. (2011). Hypoxia-
cific and conditional (Bergeron et al., 2000; Chavez induced angiogenesis is delayed in aging mouse
and LaManna, 2002). Previously, we have reported brain. Brain Res 1389, 50–60.
that HIF1α response to hypoxia is suppressed in the Bentourkia, M., Bol, A., Ivanoiu, A., Labar, D.,
aged and proposed that this was an explanation to Sibomana, M., Coppens, A., Michel, C., Cosnard, G.,
why the aged are at greater risk for morbidity and and De Volder, A.G. (2000). Comparison of
regional cerebral blood flow and glucose metabo-
mortality following an oxidative insult (Ndubuizu
lism in the normal brain: effect of aging. J Neurol
et al., 2010). Additionally, the potential role of KD Sci 181, 19–28.
relative to HIF2α should also be explored (Chavez Bergeron, M., Gidday, J.M., Yu, A.Y., Semenza, G.L.,
et  al., 2006). Our approach to understanding the Ferriero, D.M., and Sharp, F.R. (2000). Role of
central role of ketosis in the regulation of HIF1α hypoxia-inducible factor-1 in hypoxia-induced
as a signaling molecule in brain under pathophysi- ischemic tolerance in neonatal rat brain. Ann
ological conditions will help to delineate the role of Neurol 48, 285–296.
HIF1α in neuroprotection. Bough, K.J., and Rho, J.M. (2007). Anticonvulsant
mechanisms of the ketogenic diet. Epilepsia
CONCLUSION AND 48, 43–58.
FUTURE Bridge, E.M. (1931). The mechanism of the keto-
Over the past decade our research has consistently genic diet in epilepsy. Bull Johns Hopkins Hos 48,
shown that ketosis is neuroprotective against isch- 373–389.
Brunengraber, H., and Roe, C.R. (2006). Anaplerotic
emic insults in rats (Puchowicz et al., 2008) and,
molecules: current and future. J Inherit Metab Dis
more recently, in mice (unpublished observa-
29, 327–331.
tion). Studies in ketotic rats showed significantly Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids?
decreased infarct volumes (via MCAO model) Good medicine? Trans Am Clin Climatol Assoc
and improved survival and recovery after cardiac 114, 149–161.
arrest and resuscitation (Xu et al., 2012). We pur- Cahill, G.F.J., and Owen, O.E. (1967). Starvation and
port that the mechanisms of ketosis include glu- survival. Trans Am Clin Climat Ass 79, 13–18.
cose sparing and ketone body utilization in brain Chavez, J.C., Agani, F., Pichiule, P., and LaManna,
as well as a shift in metabolic redox, which plays a J.C. (2000). Expression of hypoxia-inducible
223

Chapter 24: Ketogenic Diet, Aging, and Neurodegeneration 223

factor-1alpha in the brain of rats during chronic Cerami, A., Chopp, M., Coleman, T., et al. (2010).
hypoxia. J Appl Physiol (1985) 89, 1937–1942. Erythropoietin:  not just about erythropoiesis.
Chavez, J.C., Baranova, O., Lin, J., and Pichiule, P. Lancet 375, 2142.
(2006). The transcriptional activator hypoxia Gibson, G.E., Sheu, K.F., and Blass, J.P. (1998).
inducible factor 2 (HIF-2/EPAS-1) regulates the Abnormalities of mitochondrial enzymes in
oxygen-dependent expression of erythropoietin in Alzheimer disease. J Neural Transm 105, 855–870.
cortical astrocytes. J Neurosci 26, 9471–9481. Gjedde, A., and Crone, C. (1975). Induction processes
Chavez, J.C., and LaManna, J.C. (2002). Activation in blood-brain transfer of ketone bodies during
of hypoxia-inducible factor-1 in the rat cerebral starvation. Am J Physiol 229, 1165–1169.
cortex after transient global ischemia:  potential Goldberger, J.J., Cain, M.E., Hohnloser, S.H., Kadish,
role of insulin-like growth factor-1. J Neurosci 22, A.H., Knight, B.P., Lauer, M.S., Maron, B.J., Page,
8922–8931. R.L., Passman, R.S., Siscovick, D., et  al. (2008).
Chen, T.T., Maevsky, E.I., and Uchitel, M.L. (2015). American Heart Association/American College of
Maintenance of homeostasis in the aging hypo- Cardiology Foundation/Heart Rhythm Society sci-
thalamus: the central and peripheral roles of suc- entific statement on noninvasive risk stratification
cinate. Front Endocrinol (Lausanne) 6, 7. techniques for identifying patients at risk for sud-
Chong, Z.Z., Kang, J.Q., and Maiese, K. (2003a). Apaf- den cardiac death: a scientific statement from the
1, Bcl-xL, cytochrome c, and caspase-9 form the American Heart Association Council on Clinical
critical elements for cerebral vascular protection Cardiology Committee on Electrocardiography
by erythropoietin. J Cereb Blood Flow Metab 23, and Arrhythmias and Council on Epidemiology
320–330. and Prevention. Circulation 118, 1497–1518.
Chong, Z.Z., Kang, J.Q., and Maiese, K. (2003b). Guo, S., Bragina, O., Xu, Y., Cao, Z., Chen, H., Zhou,
Erythropoietin fosters both intrinsic and extrin- B., Morgan, M., Lin, Y., Jiang, B.H., et al. (2008).
sic neuronal protection through modulation of Glucose up-regulates HIF-1 alpha expression in
microglia, Akt1, Bad, and caspase-mediated path- primary cortical neurons in response to hypoxia
ways. Br J Pharmacol 138, 1107–1118. through maintaining cellular redox status. J
Chugh, S.S., Jui, J., Gunson, K., Stecker, E.C., John, Neurochem 105, 1849–1860.
B.T., Thompson, B., Ilias, N., Vickers, C., Dogra, Hassel, B. (2000). Carboxylation and anaplerosis in
V., Daya, M., et al. (2004). Current burden of sud- neurons and glia. Mol Neurobiol 22, 21–40.
den cardiac death:  multiple source surveillance Helton, R., Cui, J., Scheel, J.R., Ellison, J.A., Ames,
versus retrospective death certificate-based review C., Gibson, C., Blouw, B., Ouyang, L., Dragatsis,
in a large U.S. community. J Am Coll Cardiol 44, I., Zeitlin, S., et  al. (2005). Brain-specific knock-
1268–1275. out of hypoxia-inducible factor-1alpha reduces
Cunnane, S., Nugent, S., Roy, M., Courchesne-Loyer, rather than increases hypoxic-ischemic damage. J
A., Croteau, E., Tremblay, S., Castellano, A., Pifferi, Neurosci 25, 4099–4107.
F., Bocti, C., Paquet, N., et  al. (2011). Brain fuel Hemmila, J.M., and Drewes, L.R. (1993). Glucose
metabolism, aging, and Alzheimer’s disease. transporter (GLUT1) expression by canine
Nutrition 27, 3–20. brain microvessel endothelial cells in culture:  an
Desrochers, S., Dubreuil, P., Brunet, J., Jetté, M., David, immunocytochemical study. Adv Exp Med Biol
F., Landau, B.R., and Brunengraber, H. (1995). 331, 13–18.
Metabolism of (R, S)-1, 3-butanediol acetoacetate Hempel, F.G., Jobsis, F.F., LaManna, J.L., Rosenthal,
esters, potential parenteral and enteral nutrients in M.R., and Saltzman, H.A. (1977). Oxidation of
conscious pigs. Am J Physiol 268, Pt 1):E660–667. cerebral cytochrome aa3 by oxygen plus carbon
DeVivo, D.C., Leckie, M.P., Ferrendelli, J.S., and dioxide at hyperbaric pressures. J Appl Physiol 43,
McDougal, D.B., Jr. (1978). Chronic ketosis and 873–879.
cerebral metabolism. Ann Neurol 3, 331–337. Hou, P., Kuo, C.Y., Cheng, C.T., Liou, J.P., Ann, D.K.,
DiVivo, D.C., Pagliara, A.S., and Prensky, A.L. (1973). and Chen, Q. (2014). Intermediary metabolite
Ketotic hypoglycemia and the ketogenic diet ther- precursor dimethyl-2-ketoglutarate stabilizes
apy. Neurology 23, 640–649. hypoxia-inducible factor-1alpha by inhibiting
Freeman, J.M., Vining, E.P.G., Pillas, D.J., Pyzik, P.L., prolyl-4-hydroxylase PHD2. PLoS One 9, e113865.
Casey, J.C., and Kelly, M.T. (1998). The efficacy of Hoxworth, J.M., Xu, K., Zhou, Y., Lust, W.D., and
the ketogenic diet—1998: a prospective evaluation LaManna, J.C. (1999). Cerebral metabolic pro-
of intervention in 150 children. Pediatrics 102, file, selective neuron loss, and survival of acute
1358–1363. and chronic hyperglycemic rats following cardiac
Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic arrest and resuscitation. Brain Res 821, 467–479.
diets, mitochondria, and neurological diseases. J Jarrett, S.G., Milder, J.B., Liang, L.P., and Patel,
Lipid Res 55, 2211–2228. M. (2008). The ketogenic diet increases mito-
Ghezzi, P., Bernaudin, M., Bianchi, R., Blomgren, chondrial glutathione levels. J Neurochem 106,
K., Brines, M., Campana, W., Cavaletti, G., 1044–1051.
224

224 section III: Ketogenic Diet in the Laboratory

Julio-Amilpas, A., Montiel, T., Soto-Tinoco, E., Millichap, J.G., Jones, J.D., and Rudis, B.P. (1964).
Geronimo-Olvera, C., and Massieu, L. (2015). Mechanism of anti-convulsant actin of ketogenic
Protection of hypoglycemia-induced neuronal diet. Am J Dis Child 107, 593–604.
death by beta-hydroxybutyrate involves the pres- Mills, E., and O’Neill, L.A. (2014). Succinate: a meta-
ervation of energy levels and decreased produc- bolic signal in inflammation. Trends Cell Biol 24,
tion of reactive oxygen species. J Cereb Blood Flow 313–320.
Metab 35, 851–860. Nakazawa, M., Kodama, S., and Matsuo, T. (1983).
Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima, Effects of ketogenic diet on electroconvulsive
K., Clarke, K., and Veech, R.L. (2000). D-beta- threshold and brain contetents of adenosine nucle-
hydroxybutyrate protects neurons in models of otides. Brain Dev 5, 375–380.
Alzheimer’s and Parkinson’s disease. Proc Natl Ndubuizu, O.I., Tsipis, C.P., Li, A., and LaManna,
Acad Sci USA 97, 5440–5444. J.C. (2010). Hypoxia-inducible factor-1 (HIF-1)-
Kasumov, T., Cendrowski, A.V., David, F., Jobbins, independent microvascular angiogenesis in the
K.A., Anderson, V.E., and Brunengraber, H. aged rat brain. Brain Res 1366, 101–109.
(2007). Mass isotopomer study of anaplerosis Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan,
from propionate in the perfused rat heart. Arch J.M., Herrera, M.G., and Cahill, G.F., Jr. (1967).
Biochem Biophys 463, 110–117. Brain metabolism during fasting. J Clin Invest 46,
Leino, R.L., Gerhart, D.Z., Duelli, R., Enerson, B.E., 1589–1595.
and Drewes, L.R. (2001). Diet-induced ketosis Pan, J.W., Rothman, T.L., Behar, K.L., Stein, D.T., and
increases monocarboxylate transporter (MCT1) Hetherington, H.P. (2000). Human brain beta-
levels in rat brain. Neurochem Int 38, 519–527. hydroxybutyrate and lactate increase in fasting-
Leist, M., Ghezzi, P., Grasso, G., Bianchi, R., Villa, P., induced ketosis. J Cereb Blood Flow Metab 20,
Fratelli, M., Savino, C., Bianchi, M., Nielsen, J., 1502–1507.
Gerwien, J., et  al. (2004). Derivatives of erythro- Prins, M.L. (2008). Cerebral metabolic adaptation
poietin that are tissue protective but not erythro- and ketone metabolism after brain injury. J Cereb
poietic. Science 305, 239–242. Blood Flow Metab 28, 1–16.
Lennox, W.B. (1928). Ketogenic diet in the treatment Prins, M.L., and Hovda, D.A. (2009). The effects of
of epilepsy. N Engl J Med 199, 74–75. age and ketogenic diet on local cerebral metabolic
Lund, T.M., Risa, O., Sonnewald, U., Schousboe, A., rates of glucose after controlled cortical impact
and Waagepetersen, H.S. (2009). Availability of injury in rats. J Neurotrauma 26, 1083–1093.
neurotransmitter glutamate is diminished when Puchowicz, M.A., Smith, C.L., Bomont, C., Koshy, J.,
beta-hydroxybutyrate replaces glucose in cultured David, F., and Brunengraber, H. (2000). Dog model
neurons. J Neurochem 110, 80–91. of therapeutic ketosis induced by oral administra-
Maalouf, M., Sullivan, P.G., Davis, L., Kim, D.Y., and tion of R, S-1, 3-butanediol diacetoacetate. J Nutr
Rho, J.M. (2007). Ketones inhibit mitochondrial Biochem 11, 281–287.
production of reactive oxygen species production Puchowicz, M.A., Xu, K., Sun, X., Ivy, A., Emancipator,
following glutamate excitotoxicity by increasing D., and LaManna, J.C. (2007). Diet-induced keto-
NADH oxidation. Neuroscience 145, 256–264. sis increases capillary density without altered
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). The blood flow in rat brain. Am J Physiol Endocrinol
neuroprotective properties of calorie restriction, Metab 292, E1607–E1615.
the ketogenic diet, and ketone bodies. Brain Res Puchowicz, M.A., Zechel, J.L., Valerio, J., Emancipator,
Rev 59, 293–315. D.S., Xu, K., Pundik, S., LaManna, J.C., and Lust,
Masino, S.A., and Rho, J.M. (2012). Mechanisms of W.D. (2008). Neuroprotection in diet-induced
ketogenic diet action. In Jasper’s Basic Mechanisms ketotic rat brain after focal ischemia. J Cereb Blood
of the Epilepsies, J.L. Noebels, M. Avoli, M.A. Flow Metab 28, 1907–1916.
Rogawski, R.W. Olsen, and A.V. Delgado-Escueta, Pundik, S., Xu, K., and Sundararajan, S. (2012).
eds. (Bethesda: National Center for Biotechnology Reperfusion brain injury: focus on cellular bioen-
Information), pp. 1001–1022. ergetics. Neurology 79, S44-S51.
McFate, T., Mohyeldin, A., Lu, H., Thakar, J., Henriques, Randle, P.J., Garland, P.B., Hales, C.N., and Newsholme,
J., Halim, N.D., Wu, H., Schell, M.J., Tsang, T.M., E.A. (1963). The glucose-fatty acid cycle: its role in
Teahan, O., et al. (2008). Pyruvate dehydrogenase insulin sensitivity and the metabolic disturbances
complex activity controls metabolic and malig- of diabetes mellitus. Lancet 1, 785–789.
nant phenotype in cancer cells. J Biol Chem 283, Safar, P. (1993). Systematic development of cerebral
22700–22708. resuscitation after cardiac arrest. Three promising
Milder, J., and Patel, M. (2012). Modulation of oxida- treatments:  cardiopulmonary bypass, hyperten-
tive stress and mitochondrial function by the keto- sive hemodilution, and mild hypothermia. Acta
genic diet. Epilepsy Res 100, 295–303. Neurochir Suppl (Wien) 57, 110–121.
Milder, J.B., Liang, L.P., and Patel, M. (2010). Acute Sato, K., Kashiwaya, Y., Keon, C.A., Tsuchiya, N., King,
oxidative stress and systemic Nrf2 activation by M.T., Radda, G.K., Chance, B., Clarke, K., and
the ketogenic diet. Neurobiol Dis 40, 238–244. Veech, R.L. (1995). Insulin, ketone bodies, and
225

Chapter 24: Ketogenic Diet, Aging, and Neurodegeneration 225

mitochondrial energy transduction. FASEB J 9, Vannucci, S.J., and Simpson, I.A.( 2003). Developmental
651–658. switch in brain nutrient transporter expression
Schwartzkroin, P.A. (1999). Mechanisms underlying in the rat. Am J Physiol Endocrinol Metab 285,
the anti-epileptic efficacy of the ketogenic diet. E1127–1134.
Epilepsy Res 37, 171–180. Veech, R.L. (2004). The therapeutic implications of
Selman, W.R., Ricci, A.J., Crumrine, R.C., LaManna, ketone bodies:  the effects of ketone bodies in
J.C., Ratcheson, R.A., and Lust, W.D. (1990). The pathological conditions:  ketosis, ketogenic diet,
evolution of focal ischemic damage:  a metabolic redox states, insulin resistance, and mitochondrial
analysis. Metab Brain Dis 5, 33–44. metabolism. Prostaglandins Leukot Essent Fatty
Semenza, G.L. (2001). Hypoxia-inducible factor Acids 70, 309–319.
1:  oxygen homeostasis and disease pathophysiol- Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A.,
ogy. Trends Mol Med 7, 345–350. and Cahill, G.F., Jr. (2001). Ketone bodies, poten-
Semenza, G.L. (2007). Hypoxia-inducible factor 1 tial therapeutic uses. IUBMB Life 51, 241–247.
(HIF-1) pathway. Sci STKE 2007, cm8. Veech, R.L., Valeri, C.R., and Vanitallie, T.B. (2012).
Semenza, G.L. (2011). Regulation of metabolism by The mitochondrial permeability transition
hypoxia-inducible factor 1. Cold Spring Harb pore provides a key to the diagnosis and treat-
Symp Quant Biol 76, 347–353. ment of traumatic brain injury. IUBMB Life 64,
Semenza, G.L., Agani, F., Feldser, D., Iyer, N., Kotch, 203–207.
L., Laughner, E., and Yu, A. (2000). Hypoxia, HIF- Veldhorst, M.A., Westerterp, K.R., van Vught, A.J.,
1, and the pathophysiology of common human and Westerterp-Plantenga M.S. (2010). Presence
diseases. Adv Exp Med Biol 475, 123–130. or absence of carbohydrates and the proportion of
Serra-Perez, A., Planas, A.M., Nunez-O’Mara, A., fat in a high-protein diet affect appetite suppres-
Berra, E., Garcia-Villoria, J., Ribes, A., and sion but not energy expenditure in normal-weight
Santalucia, T. (2010). Extended ischemia pre- human subjects fed in energy balance. Br J Nutr
vents HIF1alpha degradation at reoxygenation by 104, 1395–1405.
impairing prolyl-hydroxylation: role of Krebs cycle Willis, M.W., Ketter, T.A., Kimbrell, T.A., George,
metabolites. J Biol Chem 285, 18217–18224. M.S., Herscovitch, P., Danielson, A.L., Benson,
Sharp, F.R., Bergeron, M., and Bernaudin, M. (2001). B.E., and Post, R.M. (2002). Age, sex and laterality
Hypoxia-inducible factor in brain. Adv Exp Med effects on cerebral glucose metabolism in healthy
Biol 502, 273–291. adults. Psychiatry Res 114, 23–37.
Shi, H. (2009). Hypoxia inducible factor 1 as a thera- Xu, K., Puchowicz, M.A., Lust, W.D., and LaManna,
peutic target in ischemic stroke. Curr Med Chem J.C. (2006). Adenosine treatment delays post-
16, 4593–4600. ischemic hippocampal CA1 loss after cardiac
Shimazu, T., Hirschey, M.D., Newman, J., He, arrest and resuscitation in rats. Brain Res 1071,
W., Shirakawa, K., Le, M.N., Grueter, C.A., 208–217.
Lim, H., Saunders, L.R., Stevens, R.D., et  al. Xu, K., Sun, X., Puchowicz, M.A., and LaManna, J.C.
(2013). Suppression of oxidative stress by beta- (2007). Increased sensitivity to transient global
hydroxybutyrate, an endogenous histone deacety- ischemia in aging rat brain. Adv Exp Med Biol 599,
lase inhibitor. Science 339, 211–214. 199–206.
Silverman, W.F., Krum, J.M., Mani, N., and Rosenstein, Xu, K., Puchowicz, M.A., Sun, X., and LaManna, J.C.
J.M. (1999). Vascular, glial and neuronal effects (2008). Mitochondrial dysfunction in aging rat
of vascular endothelial growth factor in mes- brain following transient global ischemia. Adv Exp
encephalic explant cultures. Neuroscience 90, Med Biol 614, 379–386.
1529–1541. Xu, K., LaManna, J.C., and Puchowicz, M.A. (2012).
Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic Neuroprotective properties of ketone bodies. Adv
diet as a treatment paradigm for diverse neuro- Exp Med Biol 737, 97–102.
logical disorders. Front Pharmacol 3, 59. Yudkoff, M., Daikhin, Y., Melo, T.M., Nissim, I.,
Storkebaum, E., Lambrechts, D., and Carmeliet, P. Sonnewald, U., and Nissim, I. (2007). The keto-
(2004). VEGF: once regarded as a specific angio- genic diet and brain metabolism of amino
genic factor, now implicated in neuroprotection. acids:  relationship to the anticonvulsant effect.
Bioessays 26, 943–954. Annu Rev Nutr 27, 415–430.
Tallian, K.B., Nahata, M.C., and Tsao, C.-Y. (1998). Zhang, Y., Kuang, Y., Xu, K., Harris, D., Lee, Z.,
Role of the ketogenic diet in children with intrac- LaManna, J., and Puchowicz, M.A. (2013). Ketosis
table seizures. Ann. Pharmacother. 32, 349–385. proportionately spares glucose utilization in brain.
Tannahill, G.M., Curtis, A.M., Adamik, J., Palsson- J Cereb Blood Flow Metab 33, 1307–1311.
McDermott, E.M., McGettrick, A.F., Goel, G., Zhang, Y., Zhang, S., Marin-Valencia, I., and
Frezza, C., Bernard, N.J., Kelly, B., Foley, N.H., Puchowicz, M.A. (2015). Decreased carbon shunt-
et al. (2013). Succinate is an inflammatory signal ing from glucose toward oxidative metabolism in
that induces IL-1beta through HIF-1alpha. Nature diet-induced ketotic rat brain. J Neurochem 132,
496, 238–242. 301–312.
226

226 section III: Ketogenic Diet in the Laboratory

Zhang, Z., Yan, J., Taheri, S., Liu, K.J., and Shi, H. of VEGF and angiopoietin expression with dis-
(2014). Hypoxia-inducible factor 1 contributes to ruption of blood-brain barrier and angiogenesis
N-acetylcysteine’s protection in stroke. Free Radic. after focal cerebral ischemia. J. Cereb. Blood Flow
Biol. Med. 68, 8–21. Metab 22, 379–392.
Zhang, Z.G., Zhang, L., Tsang, W., Soltanian-Zadeh, Zheng, Z.J., Croft, J.B., Giles, W.H., and Mensah,
H., Morris, D., Zhang, R., Goussev, A., Powers, G.A. (2001). Sudden cardiac death in the United
C., Yeich, T., and Chopp, M. (2002). Correlation States, 1989 to 1998. Circulation. 104, 2158–2163.
227

25
Endocrine and Reproductive Effects
of Ketogenic Diets
J A C O B P. H A R N E Y, P H D , K AT H RY N G U D S N U K , M S , A M I PAT E L , M D ,
A N A N T H A R . V E L L I P U R A M , M D , S AT H YA J I T B A N D A R U, M S ,
A N D D AV I D B U T L E R , P H D

INTRODUCTION alone into thermal pain management (Ruskin et al.,


The impacts of maternal nutrition during gesta- 2013), neural degeneration produced by a variety of
tion on reproductive success have been appreci- insults/pathologies including cerebral ischemia (Yin
ated for centuries (Wu et al., 2004). The fact that et  al., 2015), traumatic brain injury (Prins, 2008,
mothers provide nutrients, oxygen, and the means 2012; Prins and Matsumoto, 2014), amyotrophic
to eliminate metabolic waste products from the lateral sclerosis (Zhao et al., 2006), metastatic (Poff
developing offspring via the placenta links the ulti- et  al., 2015)  and brain (Maroon et  al., 2015)  can-
mate health of both individuals to the resources cers, and in Alzheimers (Brownlow et  al., 2013),
available to the mother and her ability to distribute Huntington’s (Ruskin et al., 2011), and Parkinson’s
them effectively. It is becoming increasingly clear disease (Cheng et al., 2009) mouse models. In each
that the metabolic status of the mother impacts case investigators assessed the effectiveness of the
the phenotype and metabolic disease susceptibil- diet, or ketones themselves, on limiting the states
ity of the offspring (Rando and Simmons, 2015). of injury, disorder, or dysfunction. The ketone body
The global epidemic of metabolic syndrome has acetone has anticonvulsant effects, and it could play
increased the concern over the effects of parental a role in seizure control. Multiple mechanisms have
nutrition (lifestyle and environment) on offspring been implicated in the neuroprotective effects of
growth, development, cognitive function, and a KD (Schwartzkroin, 1999; Hartman et  al., 2007;
metabolic health (Hartil et  al., 2009). With both Ruskin and Masino, 2012; Stafstrom and Rho, 2012;
extremes (starvation and obesity) prevalent across Danial et  al., 2013; Giordan et  al., 2014; Mantis
the world, a better understanding of the impacts et al., 2014; Meidenbauer and Roberts, 2014; Rho,
of diet and dietary therapies on health is critical to 2015; Rogawski et al., 2016) and while some effects
having any hope of improving quality of life world- may be temporary and/or reversible there is mount-
wide (Fontana and Partridge, 2015). ing evidence that long-term use of the diet can
impact gene expression and behavioral phenotype
H I G H - FAT / on a more permanent basis.
K E TO G E N I C   D I E T S
The complex hormonal and metabolic mecha- ENDOCRINE EFFECTS
nisms that control and are impacted by substrate O F   H I G H - FAT /
availability drive changes in behavior that can be K E TO G E N I C   D I E T S
uncovered through manipulation of diet. A high-fat Regulation of nutrient intake, metabolism, energy
or ketogenic diet (KD) has been in use as a treat- generation, storage, distribution, and utilization is
ment for intractable epilepsy in children for close mediated in an endocrine manner and is depen-
to 100 years (Vining et al., 1998; Neal et al., 2008; dent on the nature of the foodstuff (carbohydrate,
Stafstrom and Rho, 2012). The neuroprotective protein, or fat) and the present energy needs of the
effects of the diet are compelling, and in recent organism. From an evolutionary perspective there
years the potential therapeutic uses of the diet in is nothing more important than the consistent
humans (Kossoff et al., 2012; Kossoff and Hartman, provision of food/energy to allow for growth and
2013)  has been extended through animal studies successful reproduction.
228

228 section III: Ketogenic Diet in the Laboratory

Hormones involved in regulation of food and glycerol. The fatty acids can be converted to
intake and energy expenditure come from the gas- acetyl-CoA, moved into the citric acid cycle, and
trointestinal tract (GI), adipose tissue, pancreas, used as energy to produce ATP. The use of fatty
adrenal gland, and peripheral sensory receptors acids by systemic tissues reduces the need for
and converge on the central nervous system. The glucose and thus helps to maintain homeostasis.
drive to eat comes, in part, from the hypothalamus, In order for glucose levels to be sufficient for the
where the “hunger/satiety” center of the brain is brain, glycogen breakdown, glycerol conversion
influenced by nutrients, hormones, and other sig- to glucose, and gluconeogenesis (production of
naling factors. Specific signals convey informa- glucose from noncarbohydrate sources) by the
tion regarding overall body metabolism, dietary liver must be maintained during the postabsorp-
composition, and the status of carbohydrate and tive stage. In addition to gluconeogenesis, the
lipid stores, in muscle and liver (carbohydrate) liver also carries out ketogenesis (production
and adipose (lipid) tissue, respectively. During of ketone bodies from lipid and protein break-
emergencies the sympathetic division of the auto- down), resulting in the generation of acetoacetate,
nomic nervous system is activated, resulting in β-hydroxybutyrate, and acetone. Ketone bodies
the “fight or flight” response that diminishes any produced can be used by the entire body, includ-
drive for food intake until the situation is resolved. ing the brain, for energy. The efficiency of energy
Similarly, elevated stress levels result in elevated conversion is actually higher for ketones than it
glucocorticoids (corticosterone in animals, corti- is for glucose (Schwartzkroin, 1999). The produc-
sol in humans), which also decrease GI tract activ- tion of ketone bodies serves as the signal for fast-
ity and the drive for food intake. ing, or starvation, to the organism, as most diets
During nonemergencies the parasympathetic in the wild are not “ketogenic.”
division of the autonomic nervous system pre- Paoli et  al. (2012) published blood levels of
dominates and eating, digestion, absorption, and glucose, insulin, ketone bodies, and blood pH (see
transport occur without interruption. The hor- Table 25.1) in response to normal diets, KDs, and the
mones involved include insulin, amylin, glucagon, development of diabetic ketoacidosis. The elevation
and pancreatic peptide (PP) from the endocrine in blood ketones and the decrease in blood glucose,
pancreas; neurotransmitters neuropeptide Y at least in the short term, are well established. Of par-
(NPY), agouti gene-related protein (AGRP), and ticular interest is the lower range in concentrations
pro-opiomelanocortin (POMC) in the CNS; and of insulin in response to KDs versus normal diets.
ghrelin, peptide YY (PYY), somatostatin, secre- Dysfunctional insulin signaling, possibly due to
tin, and cholecystokinin (CCK) from the GI hypoinsulinemia, may be central to a link reported
tract. Normal physiology involves integration of between type II diabetes and the development of
these signals that converge on the brain and reg- Alzheimer’s disease (Gotz et al., 2009; Bandaru et al.,
ulate food intake and energy metabolism (Paoli 2010). Further investigations into insulin signaling
et al., 2015). in the brain are imperative and ongoing.
During and immediately after a meal is the There are two modes of energy regulation,
absorptive stage, when mechanical and chemical short-term, regulated by satiety signals, for
digestion are complete and amino acids, triglyc- single-meal control, and long-term, regulated by
erides, and glucose elevation stimulate insulin
release from the beta cells of the endocrine pan-
creas. Insulin stimulates uptake of glucose and TABLE 25.1 BLOOD LEVELS DURING A
glycogenesis by liver and muscle cells, uptake and
NORMAL DIET, KETOGENIC DIET, AND
immediate use of glucose for energy by all cells
DIABETIC KETOACIDOSIS
except skeletal muscle, uptake of amino acids and
protein synthesis by all tissues, and uptake of lipid Blood levels Normal Ketogenic Diabetic
by skeletal muscle for ATP and by adipose tissue diet diet ketoacidosis
for storage as triglycerides. The postabsorptive
state is the time between meals when glucocorti- Glucose (mg/dL) 80–120 65–80 >300
coids generally rise and work in concert with glu- Insulin (μU/L) 6–23 6.6–9.4 ≅0
cagon and epinephrine to tap into energy reserves Ketone bodies 0.1 7–8 >25
that were stored during the absorptive state. (mmol/L)
Glycogen and triglycerides are broken down via pH 7.4 7.4 <7.3
glycogenolysis and lipolysis, respectively, increas- Source: Paoli et  al. (2012). Published with permission from author
ing circulating levels of glucose and fatty acids (open access).
229

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 229

Ketosis

Orexigenic Anorexigenic

Circulating FFA
AMPK phosphorylation

GABA
NPY
ROS production

Circulating Adiponectin
CCK

or = post prandial glucose

Circulating Ghrelin

AMPK phosphorylation (directly by BHB)

AgRP expression

FIGURE 25.1 Schematic diagram demonstrating both the orexigenic and anorexigenic properties of ketosis. Figure
reprinted with permission from author (Paoli et al., 2015; open access).

adiposity signals, which are a reflection of energy different physiological scenarios and assessing the
availability in storage, especially body fat. While effects on ketosis and other metabolic endocrine
the body can only store enough glycogen in liver signals (Bielohuby et al., 2011).
and muscle for one day’s worth of energy demand, Ellenbroek and colleagues (2014) reported
it can store lipid capable of supporting weeks of that long-term KD treatment (22 weeks) resulted
energy demand. The arcuate nucleus in the hypo- in glucose intolerance, reduced beta cell cluster
thalamus responds with signals that either stimu- size and mass (see Figure 25.2), and the estab-
late (orexigenic) or inhibit (anorexigenic) food lishment of plasma markers associated with dys-
intake. It is here where the effects of ketones can lipidemia and inflammation in mice (see Table
be either orexigenic or anorexigenic (Paoli et  al., 25.2). Reduced beta cell mass and subsequent
2015; see Figure 25.1), the resultant behavioral inadequate insulin secretory responses result
effects being essentially opposite. in glucose intolerance and type II diabetes in
Adiponectin (released from adipose tissue), humans (Kahn et  al., 2006; Nolan et  al., 2011).
ghrelin (from the GI tract), and insulin and glu- The regulatory mechanism was also compro-
cagon (from the pancreas) are all impacted by mised with respect to glucagon as alpha cell
ketosis, resulting in mixed endocrine profiles and mass loss was even greater than beta cell mass
resultant behaviors of increased or decreased food loss (Ellenbroek et al., 2014). Glucagon is criti-
intake. The biochemical make-up of that food will cal for counter-regulatory control of blood glu-
dictate the metabolic response. Predicting the cose, and if insufficient the individual is subject
endocrine and behavioral effects of KDs is going to hypoglycemic episodes resulting in HAAF
to require step-by-step analysis of the impact of (hypoglycemic associated autonomic failure).
changing both the lipid and protein profiles under This occurs in both type I  and II diabetes and
230

230 section III: Ketogenic Diet in the Laboratory

(a) (b) (c)


2.0

Beta cell mass (mg)


1.5

1.0

0.5

0.0
(d) (e) (f) Control KD
1.5 2000 80
Is let density (Is lets per mm2)

Control
Beta cell cluster size (µm2)

**
* KD

% Beta cell clusters


1500 60
1.0

1000 40

0.5
500 20

0.0 0 0
Control KD Control KD

00

0
0

00

50

00
50

00
–5

–7

10
<2

–1
00

00

>
00
25

50

75
Beta cell cluster size (µm2)

FIGURE 25.2 Beta cell mass in control and KD-fed mice after 22 weeks. (A) representative image of beta cells (brown) in
control mice; (B) representative image of beta cells (brown) from KD-fed mice; (C) beta cell mass (n = 8–10 mice); (D) islet
density (n = 8–10 mice); (E) median beta cell cluster size (n = 8–10 mice); (F) beta cell cluster distribution (n = 8–10 mice).
* p < .05; ** p < .01 vs. control.
Ellenbroek, J.H., van Dijck, L., Töns, H.A., Rabelink, T.J., Carlotti, F., Ballieux, B.E.P.B., de Koning, E. J. P. (2014). Long-term ketogenic diet
causes glucose intolerance and reduced ß- and α-cell mass but no weight loss in mice. Am J Physiol Endocrinol Metab 306, E552–E558.
License # 3824960598813 from AJP.

TABLE 25.2 PLASMA MARKERS OF THE contributes to long-term complications asso-


METAB OLIC SYNDROME IN CONTROL ciated with low energy, fatigue, and potential
for loss of consciousness due to insufficient
MICE AND MICE FED A KETOGENIC DIET
counter-regulatory mechanisms to raise glucose
FOR 22 WEEKS
(Dagogo-Jack, 2015).
Plasma markers Control KD Collectively, results are consistent with the
notion that long-term KD treatment, while
Total cholesterol, mg/dL 92.7 ± 3.1 141.3 ± 9.5b manageable and not life-threatening from a
Triglyceride, mg/dL 42.0 ± 1.5 64.9 ± 6.8b ketoacidotic standpoint, is not ideal for long-
Leptin, ng/mL 0.63 ± 0.37 2.35 ± 0.99a term metabolic health. Enhanced and extended
MCP-1, pg/mL 6.20 ± 0.73 12.65 ± 1.40c inflammatory responses are counter to organis-
IL-1β, pg/mL 1.19 ± 0.31 3.86 ± 1.17a mal well-being and long-term survival. For the
IL-6, pg/mL 3.20 ± 2.11 19.84 ± 8.02a KD to be an effective, long-term therapy, its del-
ALT, U/L 31.3 ± 0.9 80.6 ± 15.8b eterious side effects of dyslipidemia and inflam-
AST, U/L 89.0 ± 10.7 155.2 ± 31.4a mation will have to be addressed. Whether the
Values are means ± SE; n = 8–10 mice. MCP, monocyte chemotactic beneficial effects are mediated through ketone
protein; IL, interleukin; ALT, alanine aminotransferase; AST, aspar- bodies (Kim et  al., 2015), specific fatty acids,
tate aminotransferase. a p < .05, b p < .01, c p < .001.
Source: Ellenbroek, J.H., van Dijck, L., Töns, H.A., Rabelink, T.J., altered glucose, altered insulin and glucagon, or
Carlotti, F., Ballieux, B.E.P.B., de Koning, E. J. P. (2014). Long-term a combination of the above, an understanding of
ketogenic diet causes glucose intolerance and reduced ß- and α-cell the mechanism(s) of action of the various con-
mass but no weight loss in mice. Am J Physiol Endocrinol Metab 306,
E552–E558. License # 3824960598813 from AJP. stituents is necessary.
231

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 231

B E H AV I O R A L E F F E C T S determine the levels and changes in gene expres-


O F   H I G H - FAT / sion that result in behavioral responses of the
K E TO G E N I C   D I E T S organisms.
Initial animal studies investigating the neuropro- Central to the development of type II diabe-
tective effects of KDs on seizure susceptibility in tes is the ineffectiveness of the hormone insulin
rodent models were relatively short-term feeding to lower blood glucose by stimulating uptake in
regimens (3 weeks) followed by quantitative (time peripheral tissues. The brain has been considered
to onset; time of total seizure) and qualitative to be insulin independent for glucose uptake, how-
(tonic, clonic, or grand mal) assessment of seizure ever, there are insulin receptors located in several
severity (Hori et al., 1997; Bough and Eagles, 1999; brain regions including the hippocampus, hypo-
Su et al., 2000; Harney et al., 2002; Loscher, 2011). thalamus, and cerebellum (Kahn, 1994; Ho et al.,
The success of the KD in the treatment of pediatric 2004, Plum et al., 2005; Banks and Kastin, 1998).
epilepsy (Lefevre and Aronson, 2000; Vasconcelos Over the past decade a potential link between
et al., 2004; Neal et al., 2008; Suo et al., 2013) and type II diabetes and Alzheimer’s disease has been
the relative ease with which a dietary therapy can established and a dysfunctional insulin signaling
be administered led to the natural hope that the pathway has been proposed to be involved (Gotz
KD could impact positively numerous other neu- et  al., 2009; Bandaru et  al., 2010). The cognitive
rological or metabolic disorders (Kossoff et  al., decline reported with long-term KD treatment in
2012; Stafstrom and Rho, 2012). The therapeutic this study and others (Su et al., 2000; Zhao et al.,
use of KDs has been primarily limited to children 2004; Snead 2004; Behassan and Jan, 2006) might
(Lefevre and Aronson, 2000) but studies indicate be the result of dysfunctional insulin signaling, a
that they may be effective in adult patients suf- likely outcome of long-term KD treatment and its
fering from pharmacoresistant epilepsy (Coppola metabolic side effects.
et al., 2002). Despite the potential metabolic side effects,
Behaviors reportedly impacted by KDs individuals may choose any available means to
include epilepsy, sleep, cognition (Hallbook experience relief from their primary ailment, espe-
et  al., 2012), and motor performance, but not cially epileptic seizures that can occur at any time.
cognition, in two mouse models of Alzheimer’s As such, it is imperative that the impacts of KDs
pathology (Brownlow et  al., 2013). In addition, on normal physiology, including reproduction, be
high-fat (nonketogenic) diets alone have report- investigated. The timing and duration of exposure
edly impacted hippocampal-dependent spatial to dietary treatments will likely dictate whether
learning and memory in rats (Alzoubi et  al., effects are temporal or more long-lasting (epigen-
2009)  that was exacerbated further in combi- etic modifications), as would be expected during
nation with chronic stress. Maternal high-fat embryonic, fetal, or neonatal development (Rando
diets (HFD) themselves can act as a stressor, and Simmons, 2015).
increasing maternal glucocorticoids and dis-
rupting maternal behavior and brain activa- EFFECTS OF LONG-
tion (measured as increased c-Fos) in C57BL/6J T E R M K E TO G E N I C D I E T
mice (Bellisario et al., 2015). Changes in mouse CONSUMPTION ON PUBERTY
brain gene expression in response to both keto- AND REPRODUCTION
genic and nonketogenic HFDs (Selfridge et  al., If children are going to be consuming KDs grow-
2015) suggest that the specific lipid components ing up, what is the impact going to be on normal
of the diet likely dictate the ultimate biological development? Our laboratory has begun to inves-
effects, including alterations in the brain’s aero- tigate the effects of a KD on growth, onset of nor-
bic infrastructure and mitochondrial DNA tran- mal female development including day of vaginal
scriptional efficiency. Greenwood and Winocur opening, onset of nonnormal cycles, and onset of
(1996) reported that the degree of cognitive normal 4-day cycles in rodents. Three KDs con-
impairment is highly associated with the level of taining 8%, 14%, or 18% protein, with ketogenic
saturated fatty acids fed and independent of the ratios (fat/carb + protein) of 6.2, 4.2, and 3.2,
mono- or polyunsaturated fatty acids in the HFD. respectively (Bio-Serv, NJ), or rodent chow (RC)
Davidson and coworkers (2013) reported that were administered to 48 female Long-Evans rats
cognitive impairment induced by western diets starting on Day 23. Dates of vaginal opening were
(high in saturated fats and sugar) may be dimin- recorded, and only the KD 8% group was signifi-
ished by ketones. Collectively, present results cantly delayed (see Figure 25.3), this effect likely
suggest that specific dietary components and not the result of the more shallow growth curve (see
simply the level of ketone production, ultimately Figure 25.4).
232

*
39

37

Day of vaginal opening


35

33

31

29

27

25
Control KD8% KD14% KD18%
Dietary groups

FIGURE 25.3 The average date of vaginal opening in female Long-Evans rats maintained on rodent chow (control),
KD8%, KD14%, or KD18%. The KD8% were significantly delayed compared with controls (*; p < .05). Statistical analyses
included one-way ANOVA and post hoc Fisher’s least significant difference (LSD).

Average Weight of Rats


500
450
400
350
Weight (g)

300 Control
250
8% KD
200
150 14% KD
100 18% KD
50
0
22 33 43 53 63 73 83 93 120 180
Measurements by Day

FIGURE  25.4 The average weights of female Long-Evans rats fed rodent chow (control) or KD8%, KD14%, or
KD18% from weaning (day 22) to 180 days. KD8% were significantly lower (p < .05) at days 43 and 73, but not 93.
Statistical analyses included one-way ANOVA and post hoc Fisher’s least significant difference (LSD).

TABLE 25.3 AVERAGE AGES AND WEIGHTS OF CONTROL RODENT CHOW- AND


KD- FED FEMALE LONG- EVANS RATS AT THE FIRST REGULAR 4- DAY ESTROUS CYCLE
AND ON COMPLETION OF THREE CONSECUTIVE 4- DAY ESTROUS CYCLES

Groups n Age of first regu- Weight at first n Age after three Mean Weight on
lar 4-day cycle regular cycle (g) consecutive 4-day repeatable cycling (g)
(days) estrous cycles (days)

Control 12 45.1 ± 2.4 219.6 ± 15.3 12 65.2 ± 5.4 249.5 ± 18.6


KD8% 11 51.4 ± 6.8 123 ± 18.6 9 81.7 ± 5.7 272.3 ± 22.6
KD14% 12 46.8 ± 2.9 168.4 ± 18.9 11 65.2 ± 4.5 256.3 ± 17.4
KD18% 12 45.6 ± 3.1 172.3 ± 24.1 12 64.7 ± 3.7 262.7 ± 10.8
233

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 233

140

120

Concentration of glucose (mg/dL)


100

80

60

40

20

0
Control KD8% KD14% KD18%
Dietary groups

FIGURE 25.5 Mean concentrations of blood glucose in control rodent chow- and KD-fed Long-Evans female rats
at day 70 (postpubertal), 43 days after onset of dietary treatments. No significant differences were detected. Statistical
analyses included one-way ANOVA and post hoc Fisher’s least significant difference (LSD).

The time till completion of three consecutive Blood ketones were significantly different, reflect-
4-day (normal) estrous cycles was similarly only ing the elevated ketogenic ratio of the diets as total
significantly delayed in the KD8% group (see protein plus carbohydrates drops and total fat
Table 25.3), once again likely delayed in concert increases (see Figure 25.6).
with the delayed growth. The KD8% rats were the With the establishment of multiple KD groups
lightest of the groups at first regular estrous cycle (varying in circulating ketones), investigations
despite the additional 16 days of eating. continued into effects of maternal KD administra-
Blood glucose levels were not significantly dif- tion prior to and throughout gestation.
ferent between any of the groups after 47 days on Pregnancy establishment and maintenance
the diet (see Figure 25.5). This result is consistent requires signaling between the developing con-
with long-term KD treatment in our laboratory. ceptus (embryo and extraembryonic membranes)

*
2.5
Concentration of ketones (mM)

2 * *

1.5

0.5

0
Control KD8% KD14% KD18%
Dietary groups

FIGURE 25.6 Mean concentrations of blood β-hydroxybutyrate (mM) in control rodent chow- and KD-fed Long-
Evans female rats at day 70 (postpubertal), 43 days after onset of dietary treatments. All KD groups had significantly
elevated ketones (*; p < .01) compared with rodent chow-fed controls, but were not significantly different from each
other. Statistical analyses included one-way ANOVA and post hoc Fisher’s least significant difference (LSD).
234

234 section III: Ketogenic Diet in the Laboratory

and the mother. Prior to development of a func- elucidated; however, epigenetic (external modi-
tional placenta, commonly referred to as the fications to DNA) changes appear to be involved
peri-implantation period, the conceptus relies on (Masuyama and Hiramatsu, 2012; Rando and
uterine secretions or histotroph for support (Bazer Simmons, 2015).
et al., 2013; Harney et al., 1990; Harney et al., 1993;
Harney et  al., 1994). The components of histo- EFFECTS OF LONG-TERM
troph can be impacted by the maternal diet (Wu K E TO G E N I C D I E T
et  al., 2004)  as supplementation of the diet with CONSUMPTION BEFORE AND
the amino acid arginine enhances fetal-placental D U R I N G   G E S TAT I O N
development in rodents, swine, and humans There have been limited studies focused on KD
(Bazer at al., 2013; Bazer et al., 2015). Arginine is administration throughout gestation and its effects
a nutritionally essential amino acid for embryonic on maternal and offspring-phenotypes; however
survival and fetal and neonatal growth (Wu et al., findings suggest, as we have always appreciated,
2009). The hormonal milieu (levels of estrogen that maternal diet does indeed impact pregnancy
and progesterone) combined with diet and over- outcome. Sussman and coworkers examined the
all metabolic and energy status of the mother will effects of KD administration during pregnancy
play a key role in establishing the components of on embryonic growth (Sussman et al., 2013b),
uterine histotroph and the substrates available to offspring physiological growth and brain struc-
the developing conceptus. Continued success- ture (Sussman et al., 2013a), and brain structure
ful development requires a functional placenta, and susceptibility to depression and anxiety in
delivery of necessary nutrients and oxygen, and the adult mouse offspring (Sussman et al., 2015).
removal of metabolic waste and carbon dioxide Dams fed a KD (67% fat; 15% protein) prior to
(Bazer et al., 2015). and during gestation exhibited a significant reduc-
Just as the quality of the environment impacts tion in maternal fertility and litter size, which the
health in adults, the quality of the intrauterine authors indicated could be due to protein defi-
(IU) and early postnatal environment impacts ciency. However, others have reported contrary
the development of offspring. An adverse IU can results (Derrickson, 2007), and the 15% protein
result in intrauterine growth restriction (IUGR). should have been sufficient for normal growth. In
The health of offspring later in life is impacted addition, since the dams were prone to fatal keto-
by the occurrence of IUGR, or complications in acidosis by midlactation (Sussman et al., 2013a),
the mother, including diabetes or obesity, that pups were fostered by dams on standard chow,
affect nutrient availability (Hales and Barker, allowing examination of the effects of KD expo-
1992, 2001; Ravelli et al., 1976; Ravelli et al., 1998; sure during gestation alone. The results from their
Valdez et al., 1994). Models used to examine IUGR studies in mice indicate that KD exposure during
include dietary calorie or protein restriction, glu- all of gestation and through weaning is not condu-
cocorticoid administration, and uteroplacental cive to normal anatomical, behavioral, and physi-
insufficiency induced by ligation of the uterine ological development of the offspring. They report
artery. All conditions induced a stressor into the delayed growth and significant relative decreases
maternal system during gestation and examined bilaterally in the cortex, fimbria, hippocampus,
an impact on offspring glucose metabolism into corpus callosum, and lateral ventricle. The hypo-
adulthood (Fowden and Forhead, 2004; McMillen thalamus and medulla experienced volumetric
and Robinson, 2005). Any nutritional deficiency enlargements as well. Differences were detected
will impact energy availability, metabolism, and as early as postnatal day 11.5 and were greater at
biochemical substrate availability. In studies where postnatal day 21.5 (Sussman et al., 2013a). In an
nutritional overabundance is the standard (obe- earlier report, Sussman and colleagues (2013b)
sity and type 2 diabetes), results show significant demonstrated that anatomical comparisons of
increases in establishment of the same chronic embryos at day 13.5 detected larger volumetric
diseases in offspring (Rich-Edwards et  al., 1999). size of embryos and their hearts from dams on
Collectively, these results suggest that the IU a KD but smaller brains, pharynx, cervical spi-
environment is likely impacted by any significant nal cord, hypothalamus, midbrain, and pons.
changes in maternal nutrition or behavior (espe- By day 17.5 embryos from KD dams were volu-
cially stress-induced), which, in turn, can have metrically smaller with smaller hearts and thy-
any number of phenotypic effects on the offspring. mus glands but enlarged cervical spine, thalamus,
The mechanism(s) by which maternal diet influ- midbrain, and pons (Sussman et al., 2013b). The
ences the phenotype of offspring have yet to be most recent study by Sussman’s group examines
235

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 235

Group 1 Group 2 Group 3 Group 4


Control 8% KD 14% KD 18% KD
Mothers Mothers Mothers Mothers
(n = 12) (n = 12) (n = 12) (n = 12)

29 45 65 67
offspring offspring offspring offspring

Group 1: Group 2: Group 3: Group 4:


12 female 12 female 12 female 12 female
offspring offspring offspring offspring

FIGURE  25.7 Treatment groups of Long-Evans dams maintained on control rodent chow or KD8%, KD14%, or
KD18% from weaning (day 22) through puberty, breeding, gestation, and out to 180 days postpartum and postweaning
of their pups. Twelve female offspring per group were isolated and maintained on rodent chow till day 70, when MWM
was performed.

both brain structure and susceptibility to depres- anatomical and developmental implications on
sion and anxiety in adult mouse offspring from the offspring. Likely, changes in gene expression,
KD-fed dams throughout gestation. Eight-week- which may be mediated epigenetically (Masuyama
old adult CD-1 mice exhibited reduced suscep- and Hiramatsu, 2012; Rando and Simmons, 2015),
tibility to anxiety and depression and increased ultimately impact anatomy and the resultant phys-
physical activity compared with standard chow iology and behavior into adulthood.
controls. Our studies in adult rats on KDs have Our lab has conducted experiments in Long
generated similar findings with decreased depres- Evans rats (see Figure 25.7; Gudsnuk unpub-
sion appearing as early as 14 days after onset of lished), where females were administered rodent
treatment and continuing through 6 months (A. chow (RC) or KDs with 8%, 14%, or 18% protein
Patel, unpublished results). In addition, recent from day of weaning (day 21)  through puberty,
evidence suggests that maternal high-fat diets breeding, gestation, and weaning of their pups.
might make offspring susceptible to behav- Biweekly weights and blood samples for glucose
ioral disorder responses to stressful challenges. and β-hydroxybutyrate were collected. Dams were
Collectively these results suggest that KD exposure tested for spatial memory by the Morris water
throughout gestation and weaning has significant maze (MWM) at day 180 postpartum, and all

TABLE 25.4 EFFECTS OF DIETS ON PREGNANCY, LIT TER SIZE, PUPS DELIVERED,


AND PUPS WEANED

n % Pregnant % of Pregnant # Rats died Average Total live Total pups % of Pups
Rats rats to have during labor litter size pups weaned weaned
pups

Control 6 83.3 83.3 0 10.2 51 29 56.8


KD8% 6 100 100 1 8 48 45 93.7
KD14% 6 100 100 0 12.7 76 65 83.5
KD18% 6 100 100 0 12.5 75 67 89.3
236

236 section III: Ketogenic Diet in the Laboratory

80

70

Time in correct quadrant (s)


60
*
50

40

30

20

10

0
Control pups KD8% pups KD14% pups KD18% pups
Dietary groups

FIGURE  25.8 Performance of female Long-Evans pups during diestrous (day 70–72), 47 to 49 days postweaning
from control rodent chow- or KD-fed dams. Offspring of the KD-fed dams spent less time (s) in the correct quadrant
of the water maze (where the hidden platform is located) than controls. KD8% again performed significantly worse
(p < .02) than the controls. The KD14% and KD18% also performed better than the KD8%, but not as well as the
controls. Statistical analyses included one-way ANOVA and post hoc Fisher’s least significant difference (LSD).

female pups were weaned at day 21, put on stan- the long-term use of KDs by women during ges-
dard RC, and maintained till day 70, when their tation may result in diminished cognitive func-
spatial memory was assessed by the MWM. tion and other phenotypic effects in the offspring
KD dams showed no significant difference (Hartil et  al., 2009). Bellisario and coworkers
in blood glucose at day 70 (see Figure 25.5) but (2015) reported changes in dams’ behavior includ-
had significantly elevated β-hydroxybutyrate (see ing increased aggression and decreased locomo-
Figure 25.6), prior to breeding compared with tor activity and decreased enzymatic activity
controls. (11β-dehydrogenase-2) in the placenta, which is
Two breeding trials were conducted (n = designed to protect the fetus from maternal glu-
6/group/trial) and the results from the second cocorticoids. Increased exposure to glucocorti-
trial are presented above. The KD rats all suc- coids may have detrimental consequences to the
cessfully produced litters and actually weaned developing fetus.
a greater percentage of pups than the controls
(Table 25.4). Average litter sizes were smaller for S U M M A RY
the KD8% than the other groups. This was not The increased interest in KDs is not surprising,
surprising, as the low protein delayed onset of as the accumulated data for the successful treat-
puberty and likely has other deleterious effects ment of epileptic seizures, especially, makes giv-
on the gonads. ing it at least a try a “no-brainer.” The known
The results from the MWM of the pups of KD- potential side effects of KDs, including hypogly-
or RC-fed dams tend to follow the reverse rela- cemia and its associated symptoms, especially in
tionship, where elevated ketones are coincident the short term; elevated lipids and cholesterol;
with less time spent in the correct quadrant and and an elevated risk of kidney stones, may be
thus worse spatial memory. The KD8% group per- tolerable to patients whose options are limited
formed significantly worse (p < .02) than the RC- (Schwartzkroin, 1999).
fed controls. The KD14% and KD18% tended to Dietary changes, especially radical ones,
perform better than the KD8%, but not as well as bring about metabolic changes throughout the
the RC-fed controls (see Figure 25.8). The KD-fed body as the substrates for reactions change and
dams at 180  days postpartum tended to perform the by-products of those metabolic changes
worse than the RC-fed controls (data not shown). impact homeostasis. It is certainly clear that
Although just a few studies have been con- HFDs and KDs can vary in percentage of lipid,
ducted in mice and rats, the results to date sug- source of lipid, and saturation state of lipid, thus
gest that, while effective for some neurological inducing potential orexigenic or anorexigenic
disorders, possibly in both children and adults, effects that will ultimately change the behavior
237

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 237

of the organism (Paoli et  al., 2015). The possi- fat and protein. Am J Physiol Endocrinol Metab
bility of deleterious consequences to offspring’s 300, 65–76.
metabolic regulatory system as well as cognitive Bough, K.J., and Eagles, D.A. (1999). A ketogenic diet
function means it is imperative that investiga- increases the resistance to pentylenetetrazole-
tions continue into the specific effects of HFD, induced seizures in the rat. Epilepsia 40, 138–143.
Brownlow, M.L., Benner, L., D’Agostino, D., Gordon,
both ketogenic and nonketogenic to uncover the
M.N., and Morgan, D. (2013). Ketogenic diet
components (saturated and nonsaturated fatty
improves motor performance but not cognition in
acids, protein content and source) necessary for two mouse models of Alzheimer’s pathology. PLoS
safe therapies in relation to the endocrinology, One 8(9), e75713.
development, and neuroendocrinology of ani- Cheng, B., Yang, X., An, L., Gao, B., Liu, X., and Liu, S.
mals and ultimately humans. (2009). Ketogenic diet protects dopaminergic
neurons against 6-OHDA neurotoxicity via up-
AC K N OW L E G M E N T S regulating glutathione in a rat model of Parkinson’s
The authors thank Dr.  William Neace for assis- disease. Brain Res 1286, 25-31.
tance with statistical analyses. This research was Coppola, G., Veggiotti, P., Cusmai, R., Bertoli, S.,
supported by the Neuroscience Graduate Program Cardinali, S., and Dionisi-Vici, C. (2002). The
and the College of Arts and Sciences Dean’s ketogenic diet in children, adolescents and young
Research Fund at the University of Hartford. adults with refractory epilepsy:  an Italian multi-
centric experience. Epilepsy Res 48, 221–227.
Dagogo-Jack, S. (2015). Philip E. Cryer, MD: seminal
REFERENCES contributions to the understanding of hypoglyce-
Alzoubi, K.H., Abdul-Razzak, K.K., Khabour, O.F., mia and glucose counter-regulation and the dis-
Al-Tuweiq, G.M., Alzubi, M.A., and Alkadhi, K.A. covery of HAAF (Cryer syndrome). Diabetes Care
(2009). Adverse effect of combination of 38, 2193–2199.
chronic psychosocial stress and high fat diet on Danial, N.N., Hartman, A.L., Stafstrom, C.E., and
hippocampus-dependent memory in rats. Behav Thio, L.L. (2013). How does the ketogenic diet
Brain Res 204, 117–123. work? Four potential mechanisms. J Child Neurol
Bandaru, S.S., Lin, K., Roming, S., Vellipuram, R., 28(8), 1027–1033.
and Harney, J.P. (2010). Effects of PI3K inhibition Davidson, T.E., Hargrave, S.L., Swithers, S.E., Sample,
and low docosahexaenoic acid on cognition and C.H., Fu, X., Kinzig, K.P., and Zheng, W. (2013).
behavior. Physiol Behav 100, 239–244. Inter-relationships amount diet, obesity and
Banks, W.A., and Kastin, A.J. (1998). Differential per- hippocampal-dependent cognitive function.
meability of the blood-brain barrier to two pan- Neuroscience 253,110–122.
creatic peptides: insulin and amylin. Peptides 19, Derrickson, E.M. (2007). The effects of dietary protein
883–889. levels on milk protein levels and postnatal growth
Bazer, F.W., Johnson, G.A., and Wu, G. (2015). Amino in laboratory mice (Mus Musculus). J Mammal 88,
acids and conceptus development during the peri- 1475–1481.
implantation period of pregnancy. Adv Exper Med Ellenbroek, J.H., van Dijck, L., Töns, H.A., Rabelink,
Biol 843, 23–52. T.J., Carlotti, F., Ballieux, B.E.P.B., and de Koning,
Bazer, F.W., Kim, J.Y., Song, G.W., Ka, H., Wu, G., Johnson, E. J.  P. (2014). Long-term ketogenic diet causes
G.A., and Vallet, J.L. (2013). Roles of selected nutri- glucose intolerance and reduced ß- and α-cell
ents on development of the conceptus during preg- mass but no weight loss in mice. Am J Physiol
nancy. Soc Reprod Fertil Suppl 68, 159–174. Endocrinol Metab 306, E552–E558.
Behassan, N.A., and Jan, M.M. (2006). Ketogenic Diet Fontana, L., and Partridge, L. (2015). Promoting health
Update and Application. Neurosciences (Riyadh) and longevity through diet: from model organisms
11, 235–240. to humans. Cell Press 161(1), 106–118.
Bellisario, V., Panetta, P., Balsevich, G., Baumann, V. Fowden, A.L., and Forhead, A.J. (2004). Endocrine
Noble, J., Raggi, C., Nathan, O., Berry, A., Seckl, J., mechanisms of intrauterine programming,
Schmidt, M., Holmes, M., et al. (2015). Maternal Reproduction 127, 515–526.
high-fat diet acts as a stressor increasing maternal Giordan, C., Marchio, M., Timofeeva, E., and Biagini,
glucocorticoids’ signaling to the fetus and disrupt- G. (2014). Neuroactive peptides as putative medi-
ing maternal behavior and brain activation in ators of antiepileptic ketogenic diets. Frontiers
C57BL/6J mice. Psychoneuroendo 60, 138–150. Neurol 5, 63, 1–14.
Bielohuby, M., Menhofer, D., Kirchner, H., Stoehr, B.J., Gotz, J., Ittner, L.M., and Lim, Y.A. (2009). Common
Muller, T.D., Stock, P., Hempel, M., Stemmer, K., features between diabetes mellitus and Alzheimer’s
Pfluger, P.T., Kienzle, E., et  al. (2011). Induction disease. Cell Mol Life Sci 66, 1321–1325.
of ketosis in rats fed low-carbohydrate, high-fat Greenwood, C.E., and Winocur, G. (1996). Cognitive
diets depends on the relative abundance of dietary impairment in rats fed high-fat diets:  A  specific
238

238 section III: Ketogenic Diet in the Laboratory

effect of saturated fatty-acid intake. Behavioral Kossoff, E.H., Caraballo, R.H., du Toit, T., Kim, H.D.,
Neurosci 110, 451–459. MacKay, M.T., Nathan, J.K., and Philip, S.G.
Hales, C.N., and Barker, D.J. (1992). Type 2 (non- (2012). Dietary therapies:  a worldwide phenom-
insulin-dependent) diabetes mellitus; the thrifty enon. Epilepsy Res 100, 205–209.
phenotype hypothesis. Diabetologia 35, 595–601. Kossoff, E.H., and Hartman, A.L. (2013). Ketogenic
Hales, C.N., and Barker, D.J. (2001). The thrifty pheno- diets:  new advances for metabolism-based thera-
type hypothesis. Br Med Bull 60, 5–20. pies. Curr Opin Neurol 25. 173–178.
Hallböök, T., Ji, S., Maudsley, S., Martin, B. (2012). The Lefevre, F., and Aronson, N. (2000). Ketogenic diet
effects of the ketogenic diet on behavior and cogni- for the treatment of refractory epilepsy in chil-
tion. Epilepsy Res 100:304–309. dren:  a systematic review of efficacy. Pediatrics
Harney, J.P., Ali, M., Vedeckis, W.V., and Bazer, F.W. 105, E46.
(1994). Porcine conceptus and retinoid-binding Lin, C., Shao, B., Huang, H., Zhou, Y., and Lin, Y.
proteins. Reprod Fertil Dev 6, 211–219. (2015). Maternal high fat diet programs stress-
Harney, J.P., Madara, J., Madara, J., and I’Anson, H. induced behavioral disorder in adult offspring.
(2002). Effects of acute inhibition of fatty acid oxi- Physiol Behav 152, 119–127.
dation on latency to seizure and concentrations of Loscher, W. (2011). Critical review of current animal
β hydroxybutyrate in plasma of rats maintained models of seizures and epilepsy used in the discov-
on calorie restriction and/or the ketogenic diet. ery and development of new antiepileptic drugs.
Epilepsy Res 49, 239–246. Seizure 20, 359–368.
Harney, J.P., Mirando, M.A., Smith, L.C., and Bazer, Mantis, J.G., Meidenbauer, J.J., Zimick, N.C.,
F.W. (1990). Retinol-binding protein:  a major Centeno, N.A., and Seyfried, T.N. (2014).
secretory product of the pig conceptus. Biol Glucose reduces the anticonvulsant effects of
Reprod 42, 523–532. the ketogenic diet in EL mice. Epilepsy Res 108,
Harney, J.P., Ott, T.L., Geisert, R.D., and Bazer, F.W. 1137–1144.
(1993). Retinol-binding protein gene expression Maroon, J.C., Seyfried, T.N., Donohue, J.P., and
in cyclic and pregnant endometrium of pigs, sheep Bost, J. (2015). The role of metabolic therapy in
and cattle. Biol Reprod 49, 1066–1073. treating glioblastoma multiforme. Surg Neurol
Hartil, K., Vuguin, P.M., Kruse, M., Schmuel, E., Int 6, 61.
Fiallo, A., Vargas, C., Warner, M.J., Duran, J.L., Masuyama, H., and Hiramatsu, Y. (2012). Effects of
Jelicks, L.A., and Charron, M.J. (2009). Adverse a high-fat diet exposure in utero on the meta-
effect of combination of chronic psychoso- bolic syndrome-like phenomenon in mouse
cial stress and high fat diet on hippocampus- offspring through epigenetic changes in adipo-
dependent memory in rats. Pediatr Res 66, cytokine gene expression. Endocrinology 153,
368–373. 2823–2830.
Hartman, A.L., Gasior, M., Vining, E.P., and Rogawski, McMillen, I.C., and Robinson, J.S. (2005).
M.A. (2007). The neuropharmacology of the keto- Developmental origins of the metabolic syn-
genic diet. Pediatr Neurol 36, 281–292. drome:  prediction, plasticity, and programming.
Ho, L., Qin, W., Pompl, P.N., Xiang, Z., Wang, J., Zhao, Physiol Rev 85, 571–633.
Z, Peng, Y., Cambareri, G., Rocher, A., Mobbs, Meidenbauer, J.J., and Roberts, M.F. (2014). Reduced
C.V., et  al. Diet-induced insulin resistance pro- glucose utilization underlies seizure protection
motes amyloidosis in a transgenic mouse model of with dietary therapy in epileptic EL mice. Epilepsy
Alzheimer’s disease. FASEB J. 18:902–904. Behav 39, 48–54.
Hori, A., Tandon, P., Holmes, G.L., and Stafstrom, Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson,
C.E. (1997). Ketogenic diet: effects on expression M.S., Edwards, N., Fitzsimmons, G., Whitney,
of kindled seizures and behavior in adult rats. A., and Cross, J.H. (2008). The ketogenic
Epilepsia 38, 750–758. diet for the treatment of childhood epilepsy:  a
Kahn, C. (1994). Insulin action, diabetogenesis, randomized controlled trial. Lancet Neurol 7,
and the cause of type II diabetes. Diabetes 43, 500–506.
1066–1084. Nolan, C.J., Damm, P., and Prentki, M. (2011). Type
Kahn, S.E., Hull, R.L., and Utzschneider, K.M. (2006). 2 diabetes across generations: from pathophysiol-
Mechanisms linking obesity to insulin resistance ogy to prevention and management. Lancet 387,
and type 2 diabetes. Nature 444, 840–846. 169–181.
Kim, D.Y., Abdelwahab, M.G., Lee, S.H., O’Neill, D., Paoli, A., Grimaldi, K., Toniolo, L., Canato, M.,
Thompson, R.J., Duff, H.J., Sullivan, P.G., and Rho, Bianco, A., and Fratter, A. (2012). Nutrition and
J.M. (2015). Ketones prevent oxidative impairment acne: therapeutic potential of ketogenic diets. Skin
of Hippocampal Synaptic Integrity through KATP Pharmacol Physiol 25, 111–117. doi:  10.1159/
channels. PLoS One 10, e0119316. 000336404
239

Chapter 25: Endocrine and Reproductive Effects of Ketogenic Diets 239

Paoli, A., Bosco, G., Camporesi, E.M., and Mangar, Ruskin, D.N., Suter, T.A., Ross, J.L., and Masino, S.A.
D. (2015). Ketosis, ketogenic diet and food intake (2013). Ketogenic diets and thermal pain: dissoci-
control:  a complex relationship. Frontiers in ation of hypoalgesia, elevated ketones, and lowered
Psychol 6, 27. doi: 10.3389/fpsyg.2015.00027 glucose in rats. J Pain 14(5):467–474.
Plum, L., Schubert, M., and Bruning, J.C. (2005). The Schwartzkroin, P.A. (1999). Mechanisms underlying
role of insulin receptor signaling in the brain. the antiepileptic efficacy of the ketogenic diet.
Trends Endocrinol Metab 16, 59–65. Epilepsy Res 37, 171–180.
Poff, A., M., Ward, N., Seyfried, T.N., Arnold, P., Selfridge, J.E., Wilkins, H.M., E.L., Carl, S.M., Koppel,
and D’Agostino, D.P. (2015). Non-toxic meta- S., Funk, E., Fields, T., Lu, J., Tang, E.P., Slawson, C.,
bolic management of metastatic cancer in VM et al. (2015). Effects of one month duration keto-
mice: novel combination of ketogenic diet, ketone genic and non-ketogenic high fat diets on mouse
supplementation, and hyperbaric oxygen therapy. brain bio-energetic infrastructure. J Bioenerg
PLoS One 10, 6. Biomembr 47, 1–11.
Prins, M.L. (2012). Cerebral ketone metabolism dur- Snead, C.O. (2004). The ketogenic diet:  a cautionary
ing development and injury. Epilepsy Res 100, note. Pediatric Res 55, 368–369.
218–223. Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic
Prins, M.L. (2008). Cerebral metabolic adaptation diet as a treatment paradigm for diverse neurolog-
and ketone metabolism after brain injury. J Cereb ical disorders. Frontiers in Pharmacology 3,1–8.
Blood Flow Metab 28, 1–16. Su, S.W., Cilio, M.R., Sogawa, Y., Silveira, D.C.,
Prins, M.L., and Matsumoto, J.H. (2014). The col- Holmes, G.L., and Stafstrom, C.E. (2000). Timing
lective therapeutic potential of cerebral ketone of ketogenic diet initiation in an experimental epi-
metabolism in traumatic brain injury. J Lipid Res lepsy model. Dev Brain Res 125, 131–138.
55, 2450–2457. Suo, C., Liao, J., Lu, X., Fang, K., Hu, Y., Chen, L., Cao,
Rando, O.J., and Simmons, R.A. (2015). I’m eating for D., Huang, T., Li, B., and Li, C. (2013). Efficacy and
two: parental dietary effects on offspring metabo- safety of the ketogenic diet in Chinese children.
lism. Cell 161, 93–105. Seizure 22, 174–178.
Ravelli, G.P., Stein, Z.A., and Susser, M.W. (1976). Sussman, D., Ellegood, J., and Henkelman, M. (2013a).
Obesity in young men after famine exposure A gestational ketogenic diet  alters maternal met-
in utero and early infancy. N Engl J Med 295, abolic status as well as offspring physiological
349–353. growth and brain structure in the neonatal mouse.
Ravelli, A.C., van der Meulen, J.H., Michels, R.P., BMC Pregnancy Childbirth 13,1–10.
Osmond, C., Barker, D.J., Hales, C.N., and Sussman, D., van Eede, M., Wong, M.D., Adamson,
Bleker, O.P. (1998). Glucose tolerance in adults S.L., Henkelman, M. (2013b). Effects of a ketogenic
after prenatal exposure to famine. Lancet 351, diet during pregnancy on embryonic growth in the
173–177. mouse. Pregnancy and Childbirth 13, 109–120.
Rho, J.M. (2015). How does the ketogenic diet induce Sussman, D., Germann, J., and Henkelman, M. (2015).
anti-epileptic effects?. Neurosci Lett 7, 30. Gestational ketogenic diet programs brain struc-
Rich-Edwards, J.W., Colditz, G.A., Stampfer, M.J., ture and susceptibility to depression and anxiety
Willett, W.C., Gillman, M.W., Hennekens, C.H., in the adult mouse offspring. Brain Behav 5(2),
Speizer, F.E., and Manson, J.E. (1999). Birthweight e00300.
and the risk for type 2 diabetes mellitus in adult Valdez, R., Athens, M.A., Thompson, G.H., Bradshaw,
women. Ann Intern Med 130, 278–284. B.B., and Stern, M.P. (1994). Birthweight and adult
Rogawski, M.A., Loscher, W., and Rho, J.M. (2016). health outcomes in a biethnic population in the
Mechanisms of action of anti-seizure drugs and the USA. Diabetologia 37, 624–631.
ketogenic diet. Cold Spring Harb Perspect Med, Vasconcelos, M.M., Azevedo, P.M., Esteves, L., Brito,
May 2, 6(5) 397–424. pii:  a022780. doi:  10.1101/ A.R., Olivaes, M.C., and Herdy, G.V. (2004).
cshperspect.a022780 Ketogenic diet for intractable epilepsy in children
Ruskin, D.N., and Masino, S.A. (2012). The nervous and adolescents:  report of six cases. Rev Assoc
system and metabolic dysregulation:  emerging Med Bras 5, 380–385.
evidence converges on ketogenic diet therapy. Vining, E.P., Freeman, J.M., Ballaban-Gil, K., Camfield,
Frontiers in Neurosci 6(33), 1–12. C.S., Camfield, P.R., Holmes, G.L., Shinnar, S.,
Ruskin, D.N., Ross, J.L., Kawamura Jr, M., Ruiz, T.L., Shuman, R., Trevathan, E., and Wheless, J.W.
Geiger, J.D., and Masino, S.A. (2011). A keto- (1998). A multicenter study of the efficacy of the
genic diet delays weight loss and does not impair ketogenic diet. Arch Neurol 55, 1433–1437.
working memory or motor function in the R6/2 Wu, G., Bazer, F.W., Cudd, T.A., Meininger, C.J., and
1J mouse model of Huntington’s disease. Physiol Spencer, T.A. (2004). Maternal nutrition and fetal
Behav 103, 501–507. development. J Nutr 134, 2169–2172.
240

240 section III: Ketogenic Diet in the Laboratory

Wu, G., Bazer, F.W., Davis, T.A., Kim, S.W., Li, P., Zhao, Q, Stafstrom, C.E., Fu, D.D., Hu, Y., and Holmes,
Rhoads, J.M., Satterfield, M.C., Smith, S.B., G.L. (2004). Detrimental effects of the ketogenic
Spencer, T.E., and Yin, Y. (2009). Arginine metab- diet on cognitive function in rats. Pediatr Res
olism and nutrition in growth, health and disease. 55,498–506.
Amino Acids 37, 153–168. Zhao, Z., Lange, DJ, Voustianiouk, A., MacGrogan,
Yin, J., Han, P., Tang, Z., Liu, Q., Shi, J. (2015). Sirtuin D., Ho, L., Suh, J., Humala, N., Thiyagarajan, M.,
3 mediates neuroprotection of ketones against Wang, J., and Pasinetti, G.M. (2006). A ketogenic
ischemic stroke. J Cereb Blood Flow Metab 35, diet as a potential novel therapeutic intervention
1783–1789. in amyotropic lateral sclerosis. Neurosci 7, 29.
241

26
Alzheimer’s Disease
Causes and Treatment

RICHARD L. VEECH, MD, PHD, DPHIL AND M. TODD KING

INCIDENCE Early-onset Alzheimer’s disease is associated


The prevalence of Alzheimer’s disease is strongly with three genes that increase the production or
correlated with age. In a general community, 3% deposition of β-amyloid protein. These include the
of those between 65 and 74 years of age had prob- precursor of amyloid protein, found in Alzheimer’s
able Alzheimer’s disease, compared with 18.7% of disease and in Down syndrome. Missense muta-
those between 75 and 84  years. In those over 85 tions in the presenilin 1 and 2 genes, which code
the prevalence rose to 47.2% (Evans et al., 1989). for several secretases, involved in the catabolism
In the United States it was estimated that in 2013 of amyloid, lead to the early onset of an aggressive
there were 5.2  million patients with Alzheimer’s familial form of Alzheimer’s disease. Mutations in
disease, costing $203 billion in direct medical care these genes result in the onset of Alzheimer’s dis-
and another $216 billion in care from unpaid care- ease, often in the 40s or 50s, but occasionally in the
givers (Thies & Bleiler 2013). 30s. The increased incidence of early Alzheimer’s
disease with genetic abnormalities in amyloid sug-
P R E D I S P O S I N G FA C T O R S gests that amyloid can exacerbate the inhibition
The major predisposing factor for Alzheimer’s dis- of pyruvate dehydrogenase (PDH) (Hoshi et  al.,
ease is age. Women are more likely to be affected 1996). It follows that inhibition of cerebral PDH
than men. Diabetes, obesity and lack of exercise could be the primary cause of Alzheimer’s disease
predispose to the disease. The genetic environ- and the primary factor to be overcome in its treat-
mental, nutritional and metabolic risk factors are ment and prevention (Kashiwaya et al., 2000).
discussed below.
Environmental
Genetic Radiation is known to induce production of free
Only about 10% of the cases of Alzheimer’s disease radicals, or reactive oxygen species (ROS), and
are familial; about 90% are classed as “sporadic,” induce oxidant damage (Alexander and Stacey,
with age being the major risk factor. A  primary 1959; Riley, 1994; Szilard, 1959). Low levels of
cause of the aging-associated damage is free radi- radiation, under 5 Gy, can induce both cogni-
cal damage (Harman, 1956). Factors other than tive dysfunction and the pathological changes of
aging can predispose individuals to the onset neurodegeneration seen in both Parkinson’s and
of Alzheimer’s disease, the major genetic factor Alzheimer’s disease (Kempf et  al., 2013). In cul-
being the apolipoprotein E genotype (Strittmatter tured neurons, radiation also induced tau phos-
and Roses, 1995). Apolipoprotein E (ApoE) is the phorylation (Li et al., 2014), characteristic of both
lipoprotein involved in transport of lipids and Alzheimer’s disease and frontotemporal dementia.
cholesterol in the circulatory system. There are These effects have application to cancer therapy,
three major alleles, ApoE-2, ApoE-3, and ApoE-4. prolonged space flight, and exposure to nuclear
In one large kindred group of the ApoE-4 geno- radiation (Li et al., 2014).
type, the onset of clinical symptoms was between Traumatic brain injury (TBI) prematurely
55 and 78 years of age (Martin et al., 1997), dem- causes pathology similar to both Alzheimer’s
onstrating that this genetic factor predisposes and Parkinson’s disease (Dekosky et  al., 2013;
individuals to a “late-onset” form of the disease. DeKosky et  al., 2010). In addition to the
242

242 section III: Ketogenic Diet in the Laboratory

accumulation of amyloid and phosphorylated Alterations in the alleles of the ApoE-4 lipo-
tau, patients with TBI can develop both cogni- protein (Roses, 2006) involved in cholesterol and
tive impairment and Parkinsonian symptoms. In lipid transport, are associated with predisposi-
contrast to the damaging effects of brain trauma, tion to the more common late-onset form of the
physical exercise can retard neurodegeneration disease. While the association of the ApoE-4
and decrease inflammation in brain (Cotman allele with late-onset Alzheimer’s has been con-
et al., 2007). firmed by exhaustive genomewide association
studies (GWAS), no other major genetic links to
Nutritional and Metabolic Alzheimer’s disease were found (Naj et al., 2014).
Type II diabetes is strongly associated with both The majority of cases of late-onset Alzheimer’s dis-
vascular dementia and Alzheimer’s dementia (Ott ease are not associated with any identified genetic
et  al., 1996; Xu et  al., 2009). Diabetes has been factor but are simply correlated with increased age.
shown to accompany an increase in dementia in Alzheimer’s disease is, however, strongly correlated
twin studies (Kuusisto et  al., 1997). Conversely, with insulin resistance both epidemiologically and
caloric restriction or intermittent fasting ame- pathophysiologically (Kuusisto et  al., 1997; Ott
liorates amyloid accumulation (Mouton et  al., et al., 1996; Reaven et al., 1990; Xu et al., 2009). The
2009)  and the cognitive deficits in the triple incidence of the disease can be increased by free
transgenic mouse models of Alzheimer’s disease radical damage, radiation, TBI, immune dysfunc-
(Halagappa et al., 2007). tion, and indolence (Selkoe, 2001), all of which
are also associated with insulin resistance (Zhai
PAT H O P H Y S I O L O G Y et al., 2011).
The original clinical and pathological descrip- Evidence suggests that amyloid β-1-42 frag-
tion of neurofibrillary tangles, amyloid plaques, ments? can stimulate the phosphorylation and
paranoid ideation, and memory loss was given hence the inhibition of PDH. This amyloid frag-
by Alois Alzheimer in 1907 (Alzheimer, 1907). ment can exert toxic effects on hippocampal neu-
Transgenic mouse models based on mutations rons in culture (Kashiwaya et al., 2000), which is
in amyloid precursor protein and its processing compatible with the report that amyloid can inhibit
show accumulation of amyloid and phosphory- PDH (Hoshi et  al., 1997b; Hoshi et  al., 1996).
lated tau protein in their brains (Johnson-Wood Triple transgenic mouse models of Alzheimer’s
et  al., 1997). The most prevalent hypothesis disease, in addition to showing increased amy-
to explain the pathophysiology of Alzheimer’s loid and phosphorylated tau deposits, also exhibit
disease derives from the pathological findings decreased fluorodeoxyglucose (FDG) uptake in
described by Alzheimer of increased amyloid brain (Nicholson et  al., 2010). These findings are
plaques and phosphorylated tau tangles. The also compatible with the ability of ketone body
theory that Alzheimer’s disease results from amy- metabolisms to bypass the inhibition of PDH and
loid accumulation in the brain has been called the produce large amounts of acetyl CoA required to
amyloid hypothesis (Selkoe, 1997). Alternatively, supply the Krebs cycle (Sato et  al., 1995). These
it is possible that an inhibition of brain PDH due and other reports have led to the proposal that the
to insulin resistance in the brain results in a defi- amyloid cascade hypothesis be rejected (Herrup,
ciency in substrate availability in the tricarboxylic 2015), although defenders of the amyloid hypoth-
acid (TCA) cycle. Thus brain energy deficit and esis remain and propose that amyloid accumula-
mild cognitive impairment can precede amyloid tion is a necessary accompaniment of the disease
accumulation (Biessels and Reagan, 2015; Hoyer, (Musiek and Holtzman, 2015).
1991, 1996; Kuusisto et  al., 1997; Talbot et  al., Central to Alzheimer’s disease is the decrease
2012; Willette et al., 2015). The accumulation of in cerebral glucose use. Decreases in cerebral glu-
amyloid and phosphorylated tau protein exacer- cose metabolism occurs well before the clinical
bates the preexisting inhibition of PDH (Hoshi or pathological changes of Alzheimer’s disease
et  al., 1997a; Hoshi et  al., 1996). Mutations in (Blum-Degen et  al., 1995; Cunnane et  al., 2011;
amyloid precursor protein (APP) are found in Hoyer et  al., 1991). Inhibition of brain PDH has
comparatively rare early-onset forms of the dis- long been reported in the autopsy specimens
ease. Amyloid accumulates in brains of subjects from brains of patients with Alzheimer’s disease
with Alzheimer’s disease, but its accumulation (Gibson et  al., 1998). Indeed, intracerebral injec-
does not correlate, either quantitatively or tem- tion of streptozotocin, a drug that inhibits insulin
porally, with the cognitive impairment or loss of secretion, leads to a decrease in insulin synthe-
brain volume (Josephs et al., 2008). sis in brain (Grunblatt et  al., 2007). Decreases in
243

Chapter 26: Alzheimer’s Disease 243

brain oxidative metabolism lead to altered pro- and were found to have a decrease in cerebral 18F-
cessing of amyloid precursor protein, increased fluorodeoxyglucose uptake but no decrease in cere-
amyloid accumulation (Hoyer, 1996), and to bral metabolism of 11C-acetoacetate (Castellano
neuronal death. These changes in brain nutri- et al., 2015). These results suggest that in patients
ent metabolism, which precede either the clini- with mild cognitive impairment, perfusion with a
cal or pathological changes of the disease, suggest ketone body can overcome the metabolic defect
that decreased brain insulin sensitivity (Talbot resulting from the brain’s inability to use glucose.
et  al., 2012; Willette et  al., 2015)  and decreased Indeed Alzheimer’s disease has been called type
brain energy production is an important factor 3 diabetes (Steen et al., 2005) due to the decrease
in understanding the etiology of Alzheimer’s dis- in insulin and insulin-like growth factor signaling
ease (Ding et al., 2013; Morgen and Frolich, 2015). mechanisms in autopsy specimens of patients with
Indeed, a deficit in brain energy metabolism alters Alzheimer’s disease.
the processing of brain APP (Gabuzda et al., 1994), The metabolism of ketone bodies can, there-
suggesting the possibility that the accumulation of fore, correct this insulin deficiency in brain
amyloid in the brains of patients with Alzheimer’s because the metabolism of ketone bodies mimics
disease results from a deficit in cerebral energy the metabolic effects of insulin by increasing the
metabolism. The accumulation of protein is not production of acetyl CoA for use in the Krebs cycle
unique to Alzheimer’s disease, but occurs in other in the absence of normal PDH activity (Kashiwaya
neurodegenerative conditions as reflected in the et  al., 1997; Sato et  al., 1995). This property of
accumulation of α-synuclein in Parkinson’s dis- ketone body metabolism renders the administra-
ease and huntintin in Huntington’s disease. These tion of ketone bodies a rational method for the
observations suggest that the accumulation of therapy of this disease (Figure 26.1).
amyloid in the brains of patients with Alzheimer’s In addition to insulin resistance, patients with
disease is secondary to a primary defect in cerebral Alzheimer’s disease also show multiple physiologi-
energy metabolism resulting from PDH inhibi- cal signs indicative of free radical damage (Wang
tion due to a decrease in cerebral insulin sensitiv- et al., 2013). The metabolism of ketone bodies can
ity. There is mounting evidence suggesting that reduce free radical damage by reducing the free
brain insulin resistance is the major factor in the cytosolic [NADP+]/[NADPH] ratio making it the
etiology of Alzheimer’s disease (Carro and Torres- lowest redox potential in the cell (Krebs, 1969).
Aleman, 2004; Craft et al., 2012; de la Monte, 2012; NADPH is the terminal destroyer of oxygen free
Bomfim et  al., 2012; Kleinridders et  al., 2014; radicals. Additionally, the β-hydroxybutyrate mol-
Talbot et  al., 2012). A  recent study of patients ecule itself inhibits histone deacetylase (HDAC).
with mild Alzheimer’s dementia were compared This inhibition of HDAC allows the acetyl groups
with cognitively normal age-matched controls on histones to remain in place so that the tightly

Mitochondria

Glycolysis
Glucose Pyruvate
Pyruvate
Pyruvate
dehydrogenase
Acetyl CoA

3 oxoacid Krebs
CoA transferase cycle

Acetoacetate
Alternative
Energy Source βHBDH
D-β-hydroxybutyrate D-β-hydroxybutyrate

FIGURE  26.1 D-β-hydroxybutyrate metabolism produces acetyl CoA, which enters the Krebs, tricarboxylic acid
cycle by an alternative pathway that does not require the usual path through pyruvate dehydrogenase (PDH).
244

244 section III: Ketogenic Diet in the Laboratory

packed structure can be relaxed and available to efficiency of the working heart. Moreover, ketone
the FOXO3 transcription factors which leads to the body metabolism can mimic the metabolic effects
transcription of the enzymes responsible for the of insulin (Kashiwaya et  al., 1994). This increase
destruction of ROS (Offermanns, 2006; Shimazu in the efficiency of hydraulic work of the heart
et al., 2013). The actual removal of the ROS is pow- induced by either ketone bodies or insulin indi-
ered by NADPH of the free cytosolic [NADP+]/ cated an increase in the efficiency of mitochondrial
[NADPH] ratio (Krebs and Veech, 1969). energy generation. That increase in mitochondrial
energy generation could later be translated into an
T R E AT M E N T increased physiological performance in elite ath-
The hypothesis that amyloid accumulation is the letes or in disease states where metabolic energy
central etiological factor leading to Alzheimer’s dis- generation is deficient.
ease has led multiple pharmaceutical companies to A detailed examination of the metabolites
develop monoclonal antibodies to removed amy- of the Krebs tricarboxcylic acid cycle showed
loid. Active amyloid vaccine trials were suspended that addition of insulin to the working glucose-
due to the development of meningoencephali- perfused heart increased acetyl CoA content
tis (Schenk, 2002). Passive immunotherapeutics ninefold, while addition of 4-mM ketone bod-
with two antibodies have failed to significantly ies increased acetyl CoA 15-fold. This very large
improve cognitive function or prevent progression increase in acetyl CoA in the case of insulin addi-
but are now being tested in the early stage of the tion was caused by that hormone’s increased PDH
disease (Panza et al., 2014). Newer antibodies are activity, the enzymatic gateway to the Krebs cycle
currently being tested in asymptomatic individu- (Coore et al., 1971; Taylor and Jungas, 1974; Taylor
als at risk of developing the disease, based on the et  al., 1975). The even larger increase in acetyl
amyloid β cascade hypothesis, in the hope that this CoA caused by the metabolism of ketone bodies
approach will retard the development of clinically demonstrated that metabolism of ketone bodies
observable disease. It is fair to say that, to date, the could bypass the major metabolic block in insu-
results have been very disappointing (Prins and lin sensitivity. The metabolism of ketone bodies
Scheltens, 2013). mimics the effect of insulin’s activation of PDH
An alternative treatment for Alzheimer’s dis- by producing acetyl CoA from the ketone body
ease has been developed based on the hypothesis acetoacetate via the succinyl CoA transferase reac-
that the disease results from a primary failure of tion (Sato et al., 1995). The metabolism of ketone
the brain to develop both phosphorylation and bodies mimics a major metabolic effect of insulin
redox energy. The therapy has been based on the (Kashiwaya et al., 1997) and could therefore over-
observations that during starvation ketone bod- come insulin resistance, which is a common factor
ies can replace glucose as the major metabolic in many disease (Scheuermann-Freestone et  al.,
fuel in brain (Cahill and Aoki, 1980; Owen et al., 2004; Zhai et  al., 2011)  or injury states (Li and
1967). The detailed effects of the metabolism of Messina, 2009), including the brain (Talbot et al.,
ketone bodies were not well understood. A study 2012) of Alzheimer’s disease patients.
was undertaken to explore the metabolic effects of The Krebs cycle is the major energy-producing
ketone body metabolism in the working perfused metabolic pathway, which results in the aerobic
rat heart (Kashiwaya et al., 1994; Sato et al., 1995). production of ATP by the complete oxidation of
The working perfused heart provides the complete glucose. In brain, glucose is essentially the only
data needed on the effects on redox and phosphor- energy-producing substrate under fed conditions.
ylation states, O2 consumption, and metabolic effi- The entry of glucose into the Krebs cycle occurs
ciency. Addition of ketone bodies to the glucose through the production of acetyl CoA from pyru-
perfused heart resulted in a reduction of the mito- vate in a reaction catalyzed by PDH. The speed of
chondrial NAD- couple, an increase in the ΔG’ of that essential reaction is increased by insulin and
ATP, and a decrease in glucose use, indicating that decreased by loss of sensitivity to insulin. The vari-
the metabolism of ketone bodies replaced in large ation in the speed of this reaction strongly suggests
part the heart’s need for glucose to supply its meta- the loss of sensitivity of the brain’s PDH to insulin
bolic energy (Sato et al., 1995). is the root cause of Alzheimer’s disease.
More remarkably, the addition and metabo- Other metabolic changes of importance also
lism of ketone bodies, like addition of insulin, occur with addition of ketone bodies or insu-
increased the joules of hydraulic work put out by lin. The ratio of [isocitrate]/[α-ketoglutarate] is
the heart per mole of O2 consumed (Kashiwaya increased by both, indicating a reduction of the
et  al., 1994), demonstrating that either ketone free NADP system (Krebs and Veech, 1969). The
bodies or insulin could improve the metabolic free mitochondrial [NAD+]/[NADH] ratio is also
245

Chapter 26: Alzheimer’s Disease 245

Lactate–
[NAD+]c/[NADH]c [AcCoA] G vs GI
NAD+c 3.39 nmol/mL ICW
1233
1 * *
LDH 18 G vs GK
2
3 NADHc+H+c 16 G vs GIK
[Pyr] Pyruvate– 14
0.055 µmol/mL ICW 12
2
1 10
2 * *
8
6 [Cit]
[CoA]
132 µmol/mL ICW
FAD NADHm 4
0.412 µmol/mL ICW
PDH 4
1 * * 3
2
2 CO2 FADH2 NAD+m 0
2
1
* * * * * *
CoA
Acetyl CoA [IsoCit]
H+m 0.017 µmol/mL ICW
5
4
3
Citrate3– 2
1 [αKG]
[ASP] * * *
0.194 µmol/mL ICW
4.54 µmol/mL ICW
* * *
Citrate systhase Aconitase 3
1 2
2 1
3 * *
4 Oxaloacetate2– Isocitrate3– [Glut]
[OAA]
2.33 µmol/mL ICW
0.63 µmol/mL ICW NADHm + H+m NAD(P)+m 6
1 * * *
MDH 5
2 ICDH 4
3
3 NAD+m CO2 NAD(P)Hm 2
4 1
NH4+ * * *
[Malate] Malate2– GLDH
3
0.112 µmol/mL ICW [Fum]/[Succ] α-Ketoglutarate2– Glutamate–
2 Fumarase 0.076
1 5
4
H+m
* *
2– 32
Fumarate 1
FAD NADHm [NH4+]
0.879 µmol/mL ICW
* * *
αKGDH 2
1
[Fumarate] CoQH2
FAD +
FADH2 NAD m
2 * * *
0.071 µmol/mL ICW
Succ DH CO2
2 [H+]
1
2 * FADH2 CoQ 8.16x10–3 mol/L MW
1 * * *
– 2
[Succinate] Succinate2– Succinyl CoA 3
1.00 µmol/mL ICW [SuccCoA] [NAD+]m/[NADH]m
1 * * 16.0
33.5 µmol/mL ICW
2 2 * * *
1 1
3 Succinyl CoA Ligase 2
2 *

2x [Pi]m 4
28.2 µmol/mL MW 6
* * *
1
Acetyl CoA Transferase 2 8
GTP GDP Pi 10
4
3-Oxoacid CoA Transferase 12
CoA 6
8 D-β-hydroxybutyrate–
10
12
[AcAcCoA] NAD+m
Acetoacetyl CoA 4.45 µmol/mL ICW Acetoacetate–
4 βHBDH
3
2
1 NADHm+H+m
*

FIGURE  26.2 The study of perfused rat hearts treated with glucose, glucose + insulin, glucose + ketone (D-β-
hydroxybutyrate), or glucose +insulin + ketone (D-β-hydroxybutyrate). The relevant pathways are shown along with
concentration changes relative to just glucose. Images are modified from Sato 1995 (Sato et al., 1995) originally drawn
by Y. Kashiwaya.

reduced 3- to 10-fold by the addition of either in the ratio of [Coenzyme Q]/[Coenzyme QH2]
ketone bodies or insulin. Also of major signifi- (Bergman et al., 2010). The increase in the redox
cance is the increase in the ratio of [fumarate]/ span between the free mitochondrial NAD and
[succinate], indicating an oxidation of Q as seen Q couples indicates an increase in the ΔG of ATP
246
E m/c
E m/c G GI GK*GKI*
Mitochondrial area: 32.1-36.8% –100
Water: 17 – 20% of ICW –110
–120

mV
–130
–140
Eh NAD+/NADH m Mitochondrial pH –150
Mitochondrial 7.6
* *
–260 G GI GK GKI
* inner membrane 7.4 7.6 Cytosolic pH
–270 7.2 7.4
–280 Matrix 7.0 7.2
mV

–290 6.8 7.0


–300 H+ H+ 6.6 6.8
G GI* GK* GKI* 6.6
–310 G GI GK GKI
NH4+ NH3 NH3 NH4+ ATP/ADP
Eh Q/QH2 exchanger
NAD+ DβHB–
20 ADP3– ATP4– ATP
16 4H+ 4H+
12 ∆G ATP hydrolysis
8 NADH DH 2e– Glut– ATP4–
mV

4 ADP3–
0 G 2e– NADH + H+
–4 GI* GK* GKI*
Succ DH Pi
–8
ADP
QH2 ATP
Cit3– ATP synthase Cytosolic
∆G of Q/QH2-NAD+/NADH m
CoQH2 – ∆ G of ATP hydrolysis
Fum2– 3H+ 3H+
G GI* GK* GKI* Cytochrome C G GI* GK* GKI*
–50
–50 reductase 2e– TCA cycle αKG2– –52
–52 4H+ 4H+ ADP

Kj/mol
–54 –54
Kj/mol

Succ2–
–56 –56
–58 Cyt C Red Succ CoA– Pi –58
–60 +
H -Pi –60
–62 Cytochrome Ox contransporter
oxidase 4H+ H2PO4–
4H+ 2e– AcAc CoA AcAc– H2PO4–
Red Ox 2H+ Cytosolic
Cyt aa3 EhCyt zz3 Ox/Red H+ H+ [ADP] and [Pi]
VO2 620 [Pi]c [ADP]c
10 0.16
20 Measured 600
µmol/min ICW

H2O 0.14

µmol/min ICW
19 8
Calculated
mV

580 6 0.12
18 0.10
17 560 4
½ O2 0.08
16 540
15 G GI* GK* GKI* 2 0.06
14 G GI* GK* GKI* 0 G GI GK GKI
0.04

FIGURE  26.3 The study of perfused rat hearts treated with glucose, glucose + insulin, glucose + ketone (D-β-hydroxybutyrate), or glucose +insulin +
ketones (D-β-hydroxybutyrate). The relevant pathways are shown along with the relevant measures of ∆G, Gibbs free energy calculated for the nonstandard
concentrations, E, reduction potentials based on actual concentrations, VO2 is oxygen consumption in mL/kg/min. Images are modified from Sato 1995 (Sato
et al., 1995) originally drawn by Y. Kashiwaya.
247

Chapter 26: Alzheimer’s Disease 247

hydrolysis from −53 to −60 kJ /mole in spite of the heart fails (Taegtmeyer et  al., 1980), showing
a decrease in O2 consumption from 18.5 to 16– that the excessive oxidation of the mitochondria
17 μmol/min. An increase in the energy of ATP NAD couple produced by acetoacetate impairs
hydrolysis per O2 consumed was observed with cardiac energy production. Although acetoacetate
the addition of either ketone bodies or insulin (see is attractive because of its low cost, clearly it should
Figures 26.2 and 26.3) (Sato et al., 1995). be avoided as a supplement and the more expen-
sive chiral molecule d-β-hydroxybutyrate should
W H AT F O R M S O F   K E T O N E be considered instead.
B O D I E S A R E AVA I L A B L E ? R-1,3 butanediol is converted to d-β-
Following the realization of the widespread effects hydroxybutyrate, the physiological form of the
of ketone body metabolism on the redox and ketone body, which is first oxidized to acetoac-
phosphorylation states (Sato et  al., 1995)  and etate and then activated by succinyl CoA trans-
metabolic efficiency, it was recognized that thera- ferase to acetoacetyl CoA and hence to 2 acetyl
peutic effects could result from the administration CoA s by thiolase (see Figure 26.1). In contrast, S
of ketone bodies (Cahill and Veech, 2003; Veech 1,3 butanediol is converted to l-β hydroxybutyr-
et  al., 2001). During prolonged fasting (3  days) ate, which is activated by ATP and CoA to form
blood ketone body levels reach 5–7 mM (Cahill l-β-hydroxybutyryl CoA and further metabolized
and Aoki, 1980). This blood level of ketone bodies by the β fatty acid oxidation pathway (Lehninger
results from the endogenous production of about & Greville, 1953). The metabolic fates of the d
150 g of ketone bodies per day (Reichard et  al., versus the l form of β-hydroxybutyrate are quite
1974). Therefore, to mimic the effects of the keto- different, as shown in Figure 26.4. Metabolism of
sis of starvation, about 150 g of ketone bodies, or one molecule of the physiological enantiomer d
about 1.5 moles, would be required to mimic the β-hydroxybutyrate consumes one molecule each
effects of starvation while maintaining safe levels of NAD+, succinyl-CoA, CoASH, and CoQ and
(5–7 mM) of ketones. It is important to note that forms two molecules of acetyl-CoA, one molecule
the minimum effective dose of ketone bodies in of fumarate, and one molecule each of the reduced
various disease states has not been established and forms of NAD+ and coenzyme Q (QH2). On the
may be much less than the 150 g per day produced other hand, metabolism of the nonphysiological
during prolonged starvation. enantiomer, l β-hydroxybutyrate consumes two
Administration of ketone bodies as a simple molecules of CoA-SH, one molecule of NAD+, and
acid or salt would present an unsafe load of either one molecule of ATP and forms two molecules
counter ion. Accordingly, examination of a variety of acetyl-CoA and one molecule each of AMP
of alcohols was undertaken. R-1,3 butanediol was and pyrophosphate as well as one molecule of
the alcohol chosen, because it was converted in the the reduced form of NAD+. A second molecule of
liver to ketone bodies (Mehlman and Veech, 1972). ATP is also consumed to convert the AMP formed
Thus, a monoester of d-β-hydroxybutyrate –R-1,3 to ADP.
butanediol monoester presents no other metabo- In metabolizing mid-chain triglycerides (see
lite load than the ketone body. Figure 26.5), a reduced flavoprotein is produced in
Additionally, the oxidation state of the ketone the β oxidation pathway, which results in a reduc-
body being administered needs to be considered. tion of mitochondrial coenzyme Q, narrowing
Ketone bodies are readily interconverted between the redox span between mitochondrial NAD and
the oxidized form, acetoacetate, and the reduced Q and hence decreasing the energy available for
form, d-β-hydroxybutyrate. ATP synthesis. This is in contrast to the increase
in redox span that acompanies the metabolism of
d-β-hydroxybutyrate + NAD+ ↔ acetoacetate the physiological form of the ketone body, d-β-
+ NADH + H+ hydroxybutyrate (Sato et al., 1995).
It follows from the above, that the oxidized
While both forms of the ketone body can be metab- form of ketone body, acetoacetate, should not
olized, the energetic effects are quite different. be included in esters for administration for the
Acetoacetate can readily be used in the Langendorf purpose of elevating ketone bodies because such
perfused heart, which is a simpler model than the solutions would oxidize the mitochondrial NAD
working perfused heart (Williamson and Krebs, couple and lower the energy available for the for-
1961). In the working perfused rat heart, d-β- mation of ATP. Similarly, the use of racemic R,
hydroxybutyrate can be used to power the beating S 1.3 butanediol should not be used because the
heart effectively. When acetoacetate is substituted, unphysiological S form undergoes β oxidation,
248

248 section III: Ketogenic Diet in the Laboratory

(a) Metabolic fate of L β-Hydroxybutyrate (b) Metabolic fate of D β-Hydroxybutyrate

L β-Hydroxybutyrate D β-Hydroxybutyrate

ATP + CoASH NAD+

AMP + PPi NADH, H+

L β-Hydroxybutyryl-CoA Acetoacetate

NAD+ Succ-CoA

NADH, H+ Succinate CoQ

CoQH2
Acetoacetyl-CoA Acetoacetyl-CoA
Fumarate
CoASH CoASH

2 Acetyl-CoA 2 Acetyl-CoA

(c) Sum reaction for L b-Hydroxybutyrate

L βHB + ATP + 2CoASH + NAD+ 2Ac-CoA + AMP + PPi + NADH + H+

(d) Sum reaction for L b-Hydroxybutyrate

D βHB + NAD+ + Succ-CoA + CoASH + CoQ 2Ac-CoA + NADH + H+ Fun + CoQH2

FIGURE 26.4 The metabolism and sum reactions for both D and L β-hydroxybutyrate.
Metabolism of the L form of β-hydroxybutyrate (A) uses two molecules of CoASH and one molecule each of ATP and
NAD+ and forms 2 acetyl-CoAs and one molecule each of AMP, PPi, and reduced NAD+ (NADH, H+). Metabolism
of the D form of β-hydroxybutyrate (B) on the other hand uses one molecule each of NAD+, succ-CoA, CoASH, and
CoQ and produces two molecules of acetyl-CoA and one molecule each of fumarate and the reduced forms of NAD+
(NADN, H+), and CoQ (CoQH2). Sum reactions for each isomer are given in C and D.

producing mitochondrial flavoprotein, thus cerebral energy generation, primarily due to a


decreasing the oxidation of the Q couple, which loss of insulin sensitivity in brain, and that amy-
results from the metabolism of the physiological loid accumulation results from this decrease in
R-3-hydroxybutyrate. Several studies using the cerebral metabolic energy. The primary thera-
acetoacetate and racemic 1,3 butanediol ester have peutic target would therefore become not the
appeared (D’Agostino, 2013; Desrochers et  al., removal of amyloid but the overcoming of the
1995). The effects of these ketone ester formula- block of cerebral metabolic energy production
tions would be quite different from those produced caused by cerebral insulin resistance. This could
by the d-β-hydroxybutyrate–R-1, 3 butanediol be achieved by (1) feeding a therapeutic amount
monoester. of d-β-hydroxybutyrate–R 1,3 butanediol mono-
Evidence presented here suggests that ester, producing acetyl CoA without requir-
Alzheimer’s disease results from a deficit in ing the activity of brain PDH (Kashiwaya et  al.,
249

Chapter 26: Alzheimer’s Disease 249

Fatty acyl-CoA
ETF:ubiquinone
FAD ETFred Q
oxidoreductaseox
ETF:ubiquinone
FADH2 ETFox QH2
oxidoreductasered
2
Trans-∆ –Enoyl-CoA
When the number of
H2O carbons left reaches 4,
this molecule becomes
L-β-hydroxybutyryl-CoA.
L-β–Hydroxyacyl-CoA

NAD

NADH + H+ Krebs cycle

β–Ketoacyl-CoA

CoASH

Fatty acyl-CoA + Acetyl-CoA


(2 C atoms shorter)

FIGURE 26.5 The beta-oxidation pathway of metabolizing fatty acids. The β fatty acid oxidation pathway produces
1 NADH and 1 flavoprotein compared with the two NADH’s produced by the oxidation of the physiologically normal
D-β-hydroxybutyrate. Once you cycle down to the last two carbon atoms the terminal L-β-hydroxyacyl-CoA is L-β-
hydroxybutyryl-CoA, not the D form of β-hydroxybutyrate.

2013; Newport et  al., 2015). This approach has resulted in a decrease in brain amyloid and phos-
decreased amyloid and phosphorylated tau in phorylated tau accumulation as well as improve-
the brains of triple transgenic mice and improved ment in cognitive function (Kashiwaya et al., 2013).
cognitive performance. In a single human sub- Feeding ketone ester to a patient with advanced
ject, behavioral improvement on feeding ketone Alzheimer’s disease improved his behavior and
has been noted. cognitive function (Newport et  al., 2015). These
Alternatively, increasing the brain’s concentra- results support the need for larger clinical trials of
tion of insulin by introducing insulin into brain ketone esters in the treatment of Alzheimer’s dis-
through intranasal administration of insulin has ease and other diseases where insulin resistance is
been reported. This therapy also has proven suc- a major etiological factor.
cessful in treating Alzheimer’s disease in a limited
number of cases. Cerebrospinal fluid insulin levels REFERENCES
were shown to be significantly lower in patients Alexander, P., and Stacey, K.A. (1959). Cross-linking
with Alzheimer’s than normal controls (Craft et al., of deoxyribonucleic acid in sperm heads by ion-
1998). The olfactory lobe contains the highest con- izing radiations. Nature 184, 958–960.
centration of insulin receptors of any brain area Alzheimer, A. (1907). Uber eine eigenartige
(Kleinridders et al., 2014). Intranasal administration Erkrankung der Hirnrinde. Allgemeine Zeitschrift
of large doses of insulin improve cognitive perfor- fur Psychiatrie und physchish-Gerichtliche
mance in patients with early cognitive impairment Medizin 64, 146–148.
Bergman, C., Kashiwaya, Y., and Veech, R.L. (2010).
due to Alzheimer’s disease (Reger et al., 2008).
The effect of pH and free Mg2+ on ATP linked
Ketone bodies mimic the metabolic effects of enzymes and the calculation of Gibbs free energy of
insulin. They increase the energy of ATP hydroly- ATP hydrolysis. J Phys Chem B 114, 16137–16146.
sis, decrease free radical damage, and increase the Biessels, G.J., and Reagan, L.P. (2015). Hippocampal
transcription of antioxidant enzymes by inhibiting insulin resistance and cognitive dysfunction. Nat
histone deacetylase (Shimazu et  al., 2013; Veech, Rev Neurosci 16(11), 660–671.
2014). In the genetic mouse model of early-onset Blum-Degen, D., Frolich, L., Hoyer, S., and Riederer, P.
Alzheimer’s disease, feeding mice ketone esters (1995). Altered regulation of brain glucose
250

250 section III: Ketogenic Diet in the Laboratory

metabolism as a cause of neurodegenerative de la Monte, S.M. (2012). Brain insulin resistance and
disorders? J Neural Transm Suppl 46, 139–147, deficiency as therapeutic targets in Alzheimer’s
139–147. disease. Curr Alzheimer Res 9, 35–66.
Bomfim, T.R., Forny-Germano, L., Sathler, L.B., Brito- DeKosky, S.T., Ikonomovic, M.D., and Gandy, S.
Moreira, J., Houzel, J.C., Decker, H., Silverman, (2010). Traumatic brain injury:  football, war-
M.A., Kazi, H., Melo, H.M., McClean, P.L., et  al. fare, and long-term effects. N Engl J Med 363,
(2012). An anti-diabetes agent protects the mouse 1293–1296.
brain from defective insulin signaling caused by Dekosky, S.T., Blennow, K., Ikonomovic, M.D., and
Alzheimer’s disease- associated Abeta oligomers. J Gandy, S. (2013). Acute and chronic traumatic
Clin Invest122, 1339–1353. encephalopathies:  pathogenesis and biomarkers.
Cahill, G.F., Jr., and Aoki, T.T. (1980). Alternate fuel uti- Nat Rev Neurol 9, 192–200.
lization in brain. In Cerebral metabolism and neu- Desrochers, S., Quinze, K., Dugas, H., Dubreuil, P.,
ral function, J.V. Passonneau, R.A. Hawkins, W.D. Bomont, C., David, F., Agarwal, K.C., Kumar, A.,
Lust, and F.A. Welsh, eds. (Baltimore: Williams & Soloviev, M.V., Powers, L., et al. (1995). (R,S) 1,3
Wilkins), pp. 234–242. butandiol acetoacetate esters, potential alternates
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? to lipid emulsions for total parenteral nutrition. J
Good medicine? Trans Am Clin Climatol Assoc Nutr Biochem 6, 109–116.
114, 149–161; discussion 162–163, 149–161. Ding, F., Yao, J., Rettberg, J.R., Chen, S., and Brinton,
Carro, E., and Torres-Aleman, I. (2004). The role of R.D. (2013). Early decline in glucose transport and
insulin and insulin-like growth factor I  in the metabolism precedes shift to ketogenic system in
molecular and cellular mechanisms underlying female aging and Alzheimer’s mouse brain: impli-
the pathology of Alzheimer’s disease. Eur J of cation for bioenergetic intervention. PloS One 8,
Pharmacol 490, 127–133. e79977.
Castellano, C.A., Nugent, S., Paquet, N., Tremblay, S., Evans, D.A., Funkenstein, H.H., Albert, M.S.,
Bocti, C., Lacombe, G., Imbeault, H., Turcotte, E., Scherr, P.A., Cook, N.R., Chown, M.J., Hebert,
Fulop, T., and Cunnane, S.C. (2015). Lower brain L.E., Hennekens, C.H., and Taylor, J.O. (1989).
18F-fluorodeoxyglucose uptake but normal 11C- Prevalence of Alzheimer’s disease in a commu-
acetoacetate metabolism in mild Alzheimer’s dis- nity population of older persons. Higher than
ease dementia. J Alzheimers Dis 43, 1343–1353. previously reported [see comments]. JAMA 262,
Coore, H.G., Denton, R.M., Martin, B.R., and Randle, 2551–2556.
P.J. (1971). Regulation of adipose tissue pyruvate Gabuzda, D., Busciglio, J., Chen, L.B., Matsudaira, P.,
dehydrogenase by insulin and other hormones. and Yankner, B.A. (1994). Inhibition of energy
Biochem J 125, 115–127. metabolism alters the processing of amyloid pre-
Cotman, C.W., Berchtold, N.C., and Christie, L.A. cursor protein and induces a potentially amyloido-
(2007). Exercise builds brain health:  key roles of genic derivative. J Biol Chem 269, 13623–13628.
growth factor cascades and inflammation. Trends Gibson, G.E., Sheu, K.F., and Blass, J.P. (1998).
Neurosci 30, 464–472. Abnormalities of mitochondrial enzymes in
Craft, S., Cholerton, B., and Baker, L.D. (2012). Insulin Alzheimer disease. J Neural Transm 105, 855–870.
and Alzheimer’s disease:  untangling the web. Grunblatt, E., Salkovic-Petrisic, M., Osmanovic, J.,
Journal of Alzheimer’s Disease:  JAD 33 Suppl 1, Riederer, P., and Hoyer, S. (2007). Brain insulin
S263–275. system dysfunction in streptozotocin intracere-
Craft, S., Peskind, E., Schwartz, M.W., Schellenberg, broventricularly treated rats generates hyperphos-
G.D., Raskind, M., and Porte, D., Jr. (1998). phorylated tau protein. J Neurochem 101, 757–770.
Cerebrospinal fluid and plasma insulin levels Halagappa, V.K., Guo, Z., Pearson, M., Matsuoka,
in Alzheimer’s disease:  relationship to sever- Y., Cutler, R.G., Laferla, F.M., and Mattson, M.P.
ity of dementia and apolipoprotein E genotype. (2007). Intermittent fasting and caloric restriction
Neurology 50, 164–168. ameliorate age-related behavioral deficits in the
Cunnane, S., Nugent, S., Roy, M., Courchesne-Loyer, triple-transgenic mouse model of Alzheimer’s dis-
A., Croteau, E., Tremblay, S., Castellano, A., Pifferi, ease. Neurobiol Dis 26, 212–220.
F., Bocti, C., Paquet, N., et  al. (2011). Brain fuel Harman, D. (1956). Aging:  a theory based on free
metabolism, aging, and Alzheimer’s disease. radical and radiation chemistry. J Gerontol 11,
Nutrition 27, 3–20. 298–300.1
D’Agostino, D. P., Pilla, R., Held, H. E., Landon, C. S., Herrup, K. (2015). The case for rejecting the amyloid
Puchowicz, M., Brunengraber, H., … Dean, J. B. cascade hypothesis. Nat Neurosci 8, 794–799.
(2013). Therapeutic ketosis with ketone ester delays Hoshi, M., Takashima, A., Noguchi, K., Murayama,
central nervous system oxygen toxicity seizures in M., Sato, M., Kondo, S., Saitoh, Y., Ishiguro, K.,
rats. American Journal of Physiology – Regulatory, Hoshino, T., and Imahori, K. (1996). Regulation
Integrative and Comparative Physiology, 304(10), of mitochondrial pyruvate dehydrogenase activ-
R829–R836. ity by tau protein kinase I/glycogen synthase
251

Chapter 26: Alzheimer’s Disease 251

kinase 3beta in brain. Proc Natl Acad Sci USA 93, on the brain:  cause for concern? Radiat Environ
2719–2723. Biophys 52, 5–16.
Hoshi, M., Takashima, A., Murayama, M., Yasutake, Kleinridders, A., Ferris, H.A., Cai, W., and Kahn, C.R.
K., Yoshida, N., Ishiguro, K., Hoshino, T., and (2014). Insulin action in brain regulates systemic
Imahori, K. (1997a). Nontoxic amyloid á peptide metabolism and brain function. Diabetes 63,
1-42 suppresses acetylcholine synthesis. J Biol 2232–2243.
Chem 272, 2038–2041. Krebs, H.A., and Veech, R.L. (1969). Pyridine nucle-
Hoshi, M., Takashima, A., Murayama, M., Yasutake, otide interrelations. In The Energy Level and
K., Yoshida, N., Ishiguro, K., Hoshino, T., and Metabolic Control in Mitochondria, S. Papa,
Imahori, K. (1997b). Nontoxic amyloid beta Tager, J.M., Quagliariello, E., & Slater, E.C., eds.
peptide 1-42 suppresses acetylcholine synthe- (Bari, Italy: Adriatica Editrice), pp. 329–383.
sis. Possible role in cholinergic dysfunction in Kuusisto, J., Koivisto, K., Mykkanen, L., Helkala,
Alzheimer’s disease. J Biol Chem 272, 2038–2041. F.L., Vanhunen, M., Kervinen, K., Kesaniemi,
Hoyer, S. (1991). Abnormalities of glucose metabo- Y.A., Riekkinen, P.J., and Laakso, M. (1997).
lism in Alzheimer’s disease. Ann N Y Acad Sci Association between features of the insulin resis-
640, 53–58. tance syndrome and Alzheimer’s disease inde-
Hoyer, S. (1996). Oxidative metabolism deficiencies in pendently of apolipoprotein E4 phenotype: cross
brains of patients with Alzheimer’s disease. Acta sectional population based study. Brit Med J 315,
Neurol Scand Suppl 165, 18–24, 18–24. 1045–1049.
Hoyer, S., Nitsch, R., and Oesterreich, K. (1991). Lehninger, A.L., and Greville, G.D. (1953). The enzy-
Predominant abnormality in cerebral glucose uti- matic oxidation of d and l b hydroxybutyrate.
lization in late-onset dementia of the Alzheimer Biochem Biophys Acta 12, 188–202.
type:  a cross-sectional comparison against Li, L., and Messina, J.L. (2009). Acute insulin resis-
advanced late-onset and incipient early-onset tance following injury. Trends Endocrinol Metab
cases. J Neural Transm Park Dis Dement Sect 20, 429–435.
3, 1–14. Li, L., Wang, W., Welford, S., Zhang, T., Wang, X.,
Johnson-Wood, K., Lee, M., Motter, R., Hu, K., and Zhu, X. (2014). Ionizing radiation causes
Gordon, G., Barbour, R., Khan, K., Gordon, M., increased tau phosphorylation in primary neu-
Tan, H., Games, D., et al. (1997). Amyloid precur- rons. J Neurochem 131, 86–93.
sor protein processing and A beta42 deposition in Martin, E.S., Martin, S.E., Edelsohn, L., and
a transgenic mouse model of Alzheimer disease. Borgaonkar, D.S. (1997). Studies in a large fam-
Proc Natl Acad Sci USA 94, 1550–1555. ily with late-onset Alzheimer disease (LOAD).
Josephs, K.A., Whitwell, J.L., Ahmed, Z., Shiung, M.M., Alzheimer Dis Assoc Disord 11, 163–170.
Weigand, S.D., Knopman, D.S., Boeve, B.F., Parisi, Mehlman, M.A., and Veech, R.L. (1972). Redox and
J.E., Petersen, R.C., Dickson, D.W., et  al. (2008). phosphorylation states and metabolite concentra-
Beta-amyloid burden is not associated with rates tions in frozen clamped livers of rats fed diets con-
of brain atrophy. Ann Neurol 63, 204–212. taining 1,3- butanediol and DL-carnitine. J Nutr
Kashiwaya, Y., Sato, K., Tsuchiya, N., Thomas, S., Fell, 102, 45–51.
D.A., Veech, R.L., and Passonneau, J.V. (1994). Morgen, K., and Frolich, L. (2015). The metabolism
Control of glucose utilization in working perfused hypothesis of Alzheimer’s disease:  from the con-
rat heart. J Biol Chem 269, 25502–25514. cept of central insulin resistance and associated
Kashiwaya, Y., King, M.T., and Veech, R.L. (1997). consequences to insulin therapy. J Neural Transm
Substrate signaling by insulin:  a ketone bodies 122, 499–504.
ratio mimics insulin action in heart. Am J Cardiol Mouton, P.R., Chachich, M.E., Quigley, C., Spangler,
80, 50A–64A. E., and Ingram, D.K. (2009). Caloric restriction
Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima, attenuates amyloid deposition in middle-aged dtg
K., Clarke, K., and Veech, R.L. (2000). D-beta - APP/PS1 mice. Neurosci Lett 464(3), 184–187.
Hydroxybutyrate protects neurons in models of Musiek, E.S., and Holtzman, D.M. (2015). Three
Alzheimer’s and Parkinson’s disease. Proc Natl dimensions of the amyloid hypothesis: time, space
Acad Sci USA 97, 5440–5444. and ‘wingmen’. Nat Neurosci 18, 800–806.
Kashiwaya, Y., Bergman, C., Lee, J.H., Wan, R., Todd Naj, A.C., Jun, G., Reitz, C., Kunkle, B.W., Perry, W.,
King, M., Mughal, M.R., Okun, E., Clarke, K., Park, Y.S., Beecham, G.W., Rajbhandary, R.A.,
Mattson, M.P., and Veech, R.L. (2013). A ketone Hamilton-Nelson, K.L., Wang, L.S., et al. (2014).
ester diet exhibits anxiolytic and cognition- Effects of multiple genetic loci on age at onset in
sparing properties, and lessens amyloid and tau late-onset Alzheimer disease: a genome-wide asso-
pathologies in a mouse model of Alzheimer’s dis- ciation study. JAMA Neurol 71, 1394–1404.
ease. Neurobiol Aging 34, 1530–1539. Newport, M.T., VanItallie, T.B., Kashiwaya, Y., King,
Kempf, S.J., Azimzadeh, O., Atkinson, M.J., and Tapio, M.T., and Veech, R.L. (2015). A new way to pro-
S. (2013). Long-term effects of ionising radiation duce hyperketonemia: Use of ketone ester in a case
252

252 section III: Ketogenic Diet in the Laboratory

of Alzheimer’s disease. Alzheimers Dement 11, in heart failure is related to insulin resistance.
99–103. Cardiovasc J S Afr 15, S12.
Nicholson, R.M., Kusne, Y., Nowak, L.A., LaFerla, Selkoe, D.J. (1997). Alzheimer’s disease:  genotypes,
F.M., Reiman, E.M., and Valla, J. (2010). Regional phenotypes, and treatments. Science 275, 630–631.
cerebral glucose uptake in the 3xTG model of Selkoe, D.J. (2001). Alzheimer’s disease:  genes, pro-
Alzheimer’s disease highlights common regional teins, and therapy. Physiol Rev 81, 741–766.
vulnerability across AD mouse models. Brain Res Shimazu, T., Hirschey, M.D., Newman, J., He, W.,
1347, 179–185. Shirakawa, K., Le Moan, N., Grueter, C.A.,
Offermanns, S. (2006). The nicotinic acid receptor Lim, H., Saunders, L.R., Stevens, R.D., et  al.
GPR109A (HM74A or PUMA-G) as a new thera- (2013). Suppression of oxidative stress by beta-
peutic target. Trends Pharmacol Sci 27, 384–390. hydroxybutyrate, an endogenous histone deacety-
Ott, A., Stolk, R.P., Hofman, A., van Harskamp, lase inhibitor. Science 339, 211–214.
F., Grobbee, D.E., and Breteler, M.M. (1996). Steen, E., Terry, B.M., Rivera, E.J., Cannon, J.L., Neely,
Association of diabetes mellitus and dementia: the T.R., Tavares, R., Xu, X.J., Wands, J.R., and de la
Rotterdam Study. Diabetologia 39, 1392–1397. Monte, S.M. (2005). Impaired insulin and insulin-
Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan, like growth factor expression and signaling
J.M., Herrera, M.G., and Cahill, G.F. (1967). mechanisms in Alzheimer’s disease: is this type 3
Brain metabolism during fasting. J Clin Invest 46, diabetes? J Alzheimers Dis 7, 63–80.
1589–1595. Strittmatter, W.J., and Roses, A.D. (1995).
Panza, F., Logroscino, G., Imbimbo, B.P., and Solfrizzi, Apolipoprotein E and Alzheimer disease. Proc
V. (2014). Is there still any hope for amyloid-based Natl Acad Sci USA 92, 4725–4727.
immunotherapy for Alzheimer’s disease? Curr Szilard, L. (1959). On the nature of the aging process.
Opin Psychiatry 27, 128–137. Proc Natl Acad Sci USA 45, 30–45.
Prins, N.D., and Scheltens, P. (2013). Treating Taegtmeyer, H., Hems, R., and Krebs, H.A. (1980).
Alzheimer’s disease with monoclonal antibod- Utilization of energy-providing substrates in the
ies:  current status and outlook for the future. isolated working rat heart. Biochem J 186, 701–711.
Alzheimers Res Ther 5, 56. Talbot, K., Wang, H.Y., Kazi, H., Han, L.Y., Bakshi,
Reaven, G.M., Thompson, L.W., Nahum, D., and K.P., Stucky, A., Fuino, R.L., Kawaguchi, K.R.,
Haskins, E. (1990). Relationship between hyper- Samoyedny, A.J., Wilson, R.S., et  al. (2012).
glycemia and cognitive function in older NIDDM Demonstrated brain insulin resistance in
patients. Diabetes Care 13, 16–21. Alzheimer’s disease patients is associated with
Reger, M.A., Watson, G.S., Green, P.S., Wilkinson, IGF-1 resistance, IRS-1 dysregulation, and cogni-
C.W., Baker, L.D., Cholerton, B., Fishel, M.A., tive decline. J Clin Invest 122, 1316–1338.
Plymate, S.R., Breitner, J.C., DeGroodt, W., et  al. Taylor, S.I., and Jungas, R.L. (1974). Regulation of lipo-
(2008). Intranasal insulin improves cognition and genesis in adipose tissue:  the significance of the
modulates beta-amyloid in early AD. Neurology activation of pyruvate dehydrogenase by insulin.
70, 440–448. Arch Biochem Biophys 164, 12–19.
Reichard, G.A., Owen, O.E., Haff, A.C., Paul, P., and Taylor, S.I., Mukherjee, C., and Jungas, R.L. (1975).
Bortz, W.M. (1974). Ketone-body production and Regulation of pyruvate dehydrogenase in iso-
oxidation in fasting obese humans. J Clin Invest lated rat liver mitochondria. Effects of octanoate,
53, 508–515. oxidation-reduction state, and adenosine triphos-
Riley, P.A. (1994). Free radicals in biology:  oxidative phate to adenosine diphosphate ratio. J Biol Chem
stress and the effects of ionizing radiation. Int J 250, 2028–2035.
Radiat Biol 65, 27–33. Thies, W., and Bleiler, L. (2013). 2013 Alzheimer’s
Roses, A.D. (2006). On the discovery of the genetic disease facts and figures. Alzheimers Dement 9,
association of apolipoprotein E genotypes 208–245.
and common late-onset Alzheimer disease. J Veech, R.L. (2014). Ketone ester effects on metabolism
Alzheimers Dis 9, 361–366. and transcription. J Lipid Res 55, 2004–2008.
Sato, K., Kashiwaya, Y., Keon, C.A., Tsuchiya, N., King, Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A.,
M.T., Radda, G.K., Chance, B., Clarke, K., and and Cahill, G.F., Jr. (2001). Ketone bodies, poten-
Veech, R.L. (1995). Insulin, ketone bodies, and tial therapeutic uses. IUBMB Life 51, 241–247.
mitochondrial energy transduction. FASEB J 9, Wang, X., Wang, W., Li, L., Perry, G., Lee, H.G., and
651–658. Zhu, X. (2013). Oxidative stress and mitochon-
Schenk, D. (2002). Amyloid-beta immunotherapy for drial dysfunction in Alzheimer’s disease. Biochim
Alzheimer’s disease: the end of the beginning. Nat Biophys Acta 1842(8), 1240–1247.
Rev Neurosci 3, 824–828. Willette, A.A., Bendlin, B.B., Starks, E.J., Birdsill, A.C.,
Scheuermann-Freestone, M., Neubauer, S., and Clarke, Johnson, S.C., Christian, B.T., Okonkwo, O.C., La
K. (2004). Abnormal cardiac muscle function Rue, A., Hermann, B.P., Koscik, R.L., et al. (2015).
253

Chapter 26: Alzheimer’s Disease 253

Association of insulin resistance with cerebral Xu, W., Qiu, C., Gatz, M., Pedersen, N.L., Johansson,
glucose uptake in late middle-aged adults at risk B., and Fratiglioni, L. (2009). Mid- and late-life
for Alzheimer disease. JAMA Neurology 72(9), diabetes in relation to the risk of dementia:  a
1013–1020. population-based twin study. Diabetes 58, 71–77.
Williamson, J.R., and Krebs, H.A. (1961). Acetoacetate Zhai, L., Ballinger, S.W., and Messina, J.L. (2011). Role
as fuel of respiration in the perfused rat heart. of reactive oxygen species in injury-induced insu-
Biochem J 80, 540–547. lin resistance. Mol Endocrinol 25, 492–502.
254

27
Mitigation of Damage from Reactive Oxygen Species
and Ionizing Radiation by Ketone Body Esters
WILLIAM CURTIS*, MARTIN KEMPER, PHD,
A L E X A N D R A M I L L E R , P H D , R O B E R T PAW L O S K Y, P H D ,
M . T O D D K I N G, A N D R I C H A R D L . V E E C H , M D, P H D, D P H I L

INTRODUCTION (e.g., α or β), electromagnetic (e.g., γ or X), and


Free radicals play an important role in a number neutrons. Radiation of sufficient energy will split
of chronic degenerative diseases including auto- water into a hydroxyl radical (HO•), H+, and a free
immune disorders, aging (Harman, 1956), cata- electron, e–. These three species interact with a
racts, rheumatoid arthritis, Parkinson’s disease variety of other molecules to form the cascade of
(Kashiwaya et  al., 2000), cardiovascular disease, RNS and ROS shown in Figure 27.1.
and other neurodegenerative diseases (Pham-Huy Superoxide can also be generated without radi-
et  al., 2008). However, the pathophysiology of ation by NADPH oxidases (NOXs) in white blood
these diseases can be complex. The simplest form cells in response to pathogens (Bedard and Krause,
of ROS-induced disease is radiation sickness. 2007) or as an inducible response to inflammation
and cellular stress. Enzymes producing ROS are
ORIGINS OF REACTIVE also induced after exposure to radiation, particu-
OXYGEN AND NITROGEN larly the NOX family members DUOX 1 and 2,
SPECIES which continue producing superoxide for a num-
Reactive oxygen species (ROS) and reactive nitro- ber of days post radiation (Ameziane-El-Hassani
gen species (RNS) arise from numerous sources et  al., 2015). This prolonged ROS production
including cellular metabolism, ionizing radiation, leaves a therapeutic window for post-radiation
and various enzymes. Once created, individual ROS treatment that targets these molecules.
and RNS participate in a cascade of interconversions Numerous other enzymes produce other types
(Figure  27.1) both enzymatic and not, ultimately of ROS as part of their normal function, such
being detoxified by being converted into inert mol- as monoamine oxidase (MAO) (EC 1.4.3.4), an
ecules or by damaging various biomolecules. enzyme integral in monoaminergic neurotransmis-
Basal production of ROS occurs in the mito- sion and xenobiotic metabolism. This enzyme pro-
chondria as a byproduct of normal metabolism by duces hydrogen peroxide as part of its breakdown
the spontaneous oxidation of coenzyme Q semi- of monoamines, such as dopamine in this reac-
quinone by oxygen, creating superoxide (O2•−) tion: Dopamine + H2O +O2 → Dopaldehyde + NH3 +
(Chance et al., 1979). H2O2. This is an important issue in the treatment
of Parkinsonian patients, who are often given high
doses of l-dopa, which is known to increase ROS
Q • − + O2 → O2 • − + Q
load (Acquier et al., 2013) and can lead to further
loss of dopaminergic neurons, often seen in patients
Another common origin of ROS is radiation; on high doses of l-dopa (Yamato et al., 2010).
ionizing radiation with energy (hν) above 33eV Figure 27.1 provides a roadmap showing the
can ionize water, which makes up the vast major- cascade of ROS and RNS. Superoxide plays a central
ity of human cell volume. This process occurs role in the cascade. In the case of radiation, super-
with many radiation types including particles oxide results from the free electron combining with
oxygen. The enzyme superoxide dismutase (SOD)
* Authors contributed equally. catalyzes a reaction where two superoxide ions
255

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 255

2H2O + O2
e h
las
O2 ta H+ + e–
ca
+ 1e–

2
D

X
e–

SO
O2 – H2O2 HO H2O
O2 X2
oxygen superoxide hydrogen peroxide hydroxyl radical
+ 2H+ (ROS)
(ROS) (ROS)
+ 1e– + 1e– + 1e–
Fe2+ Fe3+ + OH–
2 GSH NADP+
GSH peroxidase GSH reductase
GSSG NADPH damage DNA/RNA
2H2O
damage Protein

NO H+ damage Lipids
Nitric oxide ONOOH
peroxynitrous
(RNS) acid
(RNS)
– ONOOCO2– –
ONOO Nitrosoperoxy- CO3 + NO2
peroxynitrite carbonate carbonate nitrogen dioxide
(RNS) CO2 (RNS) radical (RNS)

Protein modification
Tyr, Cys, Met
Zn finger
Hemes

FIGURE 27.1 The origin, reactions, and target molecules of reactive oxygen species (ROS) and reactive nitrogen species
(RNS) are shown. Free radicals include superoxide, hydroxyl and nitric oxide, carbonate radical, and nitrogen dioxide. The
oxidation steps show the sequence of four single-electron reductions (addition of one electron) from oxygen to superoxide
to peroxide to hydroxyl to water. The outlined arrow +1e− indicates reduction. The reaction of ionizing radiation with
water is represented by hν, where h is Planck’s constant and the Greek letter ν is the frequency, which gives the energy
of the electromagnetic radiation by E = hν where h = 6.626 × 10–34 Js. Abbreviations: e− —electron, SOD—superoxide
dismutase, GSH—glutathione, GSSG—glutathione disulfide, TYR—tyrosine, MET—methionine, CYS—cysteine.

react to produce oxygen and hydrogen peroxide. Oxygen-containing free radicals and other
The hydrogen peroxide can decompose to hydroxyl ROS or their nitrogenous counterparts perform
radical and hydroxide in the presence of iron and useful functions in cell signaling and physiol-
other metals via the Fenton reaction. Superoxide ogy (Pham-Huy et  al., 2008). While the causes
reacts with nitric oxide (NO) to produce peroxyni- of excessive ROS and RNS in diseases may have
trite and begins the cascade of RNS. The reaction of complex or indeterminate origins, the expo-
NO with superoxide is diffusion limited and non- sure to radiation provides a model with a single
enzymatic. This is especially problematic as NO known initiator. Generation of ROS/RNS is a
is uncharged, enabling it to pass quickly through component of many common diseases including
cellular membranes, and possesses a relatively long cataracts, rheumatoid arthritis, neurodegenera-
half-life. The kinetics of the reaction of NO with tive diseases such as amyotrophic lateral scle-
superoxide make the production of peroxynitrite rosis (ALS) and Parkinson’s (Kashiwaya et  al.,
inevitable whenever NO is produced within a few 2000)  and Alzheimer’s disease, cancer (Pham-
cell diameters of superoxide (Pacher et  al., 2007). Huy et  al., 2008), and aging (Harman, 1956).
Peroxynitrite in turn can either react directly with Table 27.1 summarizes reactions that generate or
CO2 to form nitrosoperoxycarbonate, which disso- destroy RNS and ROS.
ciates to form other radical species, or be proton-
ated to form peroxynitrous acid, which dissociates D A M A G E C AU S E D
into hydroxyl radical and nitrogen dioxide radical. BY REACTIVE OXYGEN
The formation of hydroxyl radical from peroxyni- AND NITROGEN SPECIES
trite may contribute more to the formation of free Free radicals attack nucleic acids, proteins, and
radicals than the iron catalyzed Fenton reaction lipids indiscriminately. Nucleic acids are attacked
(Pacher et al., 2007). either at the sugar (ribose or deoxyribose) or the
256

256 section III: Ketogenic Diet in the Laboratory

or within one protein to form a disulfide bridge


TABLE 27.1 THE GENERATION AND
and change the folding of the protein. This can be
DESTRUCTION OF RNS AND ROS reversed as a protein disulfide bond is reduced by
ROS/RNS formation ROS/RNS Catalyst thioredoxin reduction reaction shown in Figure 27.4.
reactions destruction However, further oxidation leads to SOH, SO2H
reactions
and SO3H, which makes the protein permanently
damaged.
H2O + hν → HO• + hν Damage to lipids is primarily to polyun-
H+ + e– saturated fatty acids (PUFAs). There are three
O2 + e– → O2•– NOX, xanthine stages, initiation, propagation, and termination.
oxidase Initiation causes the PUFA to become a free radi-
Fe2+ + H2O2 → Fe3+ + spontaneous cal. Propagation generates other free radicals, oxy-
HO• + OH– gen is often involved. Oxygen is a dual free radical,
O2•– + NO• → spontaneous and one might draw it as •O2• to show the two
ONOO– unpaired electrons. Being a dual radical makes
HO• + O2•– → spontaneous •O2• especially reactive with transition metals
OH– + O2 that are free radicals and also other free radicals.
2 O2•– + 2H+ → Superoxide Having the two unpaired electrons allows •O2• to
H2O2 + O2 dismutase propagate damage as in R• + O2 → ROO• → further
2 H2O2 → O2 + Catalase reactions. NO• being a single free radical would
2H2O result in a terminating reaction with other free
2GSH + H2O2 Glutathione
radicals as RS• + NO• → RS−NO.
→ GSSG + peroxidase
Peroxynitrite (ONOO−) is very reactive but it
2H2O
is not a free radical, therefore it tends to modify
proteins on specific residues and motifs such as
methionine, tyrosine, zinc fingers, and heme (see
base that is responsible for pairing. If hydroxyl Figure 27.1).
reacts with the sugar, it results in strand breaks,
recombination, chromosomal changes and muta- THE ROLE OF REACTIVE
tions. If hydroxyl radical reacts with the base, it OXYGEN SPECIES IN AGING
adds alcohol groups, OH, to the base where none In 1956, Denham Harmon, working in the Donner
belong and interferes with the hydrogen bonding Biophysics Lab at Berkeley, wrote a paper postulating
and fit of the matching base pair, leading to point that the process of aging was the result of free radicals
mutations. Modified guanine interferes with the produced either endogenously or by environmen-
telomerase binding proteins TRF1 and TRF2 and tal sources (Harman, 1956). In 1961 Hayflick and
causes a loss of telomere function, which requires Moorman showed that cultured cells could undergo
these binding proteins for both the protective shel- only a finite number of replications for reasons that
terin complex and for telomerase to interact with were unclear. In 1973 Olovnikov postulated that the
shelterin and extend the telomere (Sfeir, 2012). See end replication of DNA would result in the short-
for example Figure 27.2. ening of that strand at every replication (Olovnikov,
Proteins have various reactions depending on 1973). In 1978, Elizabeth Blackburn published a
the amino acid. One that is vulnerable is cysteine. paper describing a special form of DNA at the end
Oxidative stress can cause cysteine on two proteins of chromosomes, which is the telomere (Blackburn

O O O

HN N HN N OH Oxidation N
HN
+ HO• OH
H2N N H2N N N H N
N H2N N
R R R
Guanine (G) C8-OH-adduct radical of 8-hydroxyquanine
guanine (8-OH-G)

FIGURE 27.2 Guanine reacts with hydroxyl radical to produce 8-OH-G. There are three guanines in the telomere
repeat sequence, TTAGGG, making it vulnerable to oxidative damage. R is deoxyribose.
257

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 257

and Gall, 1978). In 1990 it was found that the telo- 2 to 5 Gy suppresses replication of blood and gut
mere of human fibroblasts shortened with aging cells through generation of ROS, causing death in
(Harley et al., 1990). This sequence seemed to pro- weeks. In these rapidly dividing cell types, a telo-
vide a mechanism to explain the development of meric process as described by Hayflick could play
senescence, at least in culture. a more dominant role.
Cell senescence can be triggered by a number There is, however, another large group of cells
of mechanisms including telomere uncapping, that undergo almost no replication after embry-
DNA damage, oxidative stress, oncogene activ- onic life. They include heart, skeletal muscle, and
ity, lack of nutrients, and other factors via various brain cells. The death of these slowly dividing cells
signaling pathways (Ben-Porath and Weinberg, does not have a unitary mechanism. The gradual
2005). A  number of studies reported that oxida- onset of senescence that characterizes normal
tive stress shortened telomeres (von Zglinicki, aging may in part be due to the accumulation of
2002). The formation of 8-oxo-guanine lesions in genetic damage over a lifetime (Szilard, 1959).
telomeric tandem repeats by ROS-damaged telo- Mice lacking the gene for dystrophin (MDX)
meric protein binding to telomeres (Opresko et al., demonstrate little cardiac abnormalities. However,
2005) impaired their functioning, causing replica- mice in which the mRNA component of telomer-
tion to cease. ase, mTR, was deleted show progressive shortening
In the absence of disease, ROS damage depends of telomeres with age. Crossing these two strains
on the rate of metabolism and with it, the rate of to form the mdx/mTRko mouse with deleted dys-
ROS production by mitochondria, which roughly trophin and TERC produced a mouse with heart
correlate with the life span (Speakman, 2005), a abnormalities, mitochondrial dysfunction, and
phenomenon known as scaling. From these and oxidative stress with reduced levels of the mito-
other observations it was thought that the rate of chondrial regulators peroxisome proliferator acti-
metabolism inversely correlated with life span and vated receptor gamma coactivator 1 α and 1 β
slowing it by measures such as caloric restriction (PGC1-α and PGC1-β), both reduced by 80%. The
would lengthen life span (Sohal and Weindruch, pathological changes in hearts of these animals
1996), in part by limiting mitochondrial ROS were ameliorated by administration of antioxidants
generation and hence ROS damage. Studies also (Mourkioti et al., 2013). These findings show that
appeared which reported that overexpression of both mitochondrial ROS production and telomere
the antioxidant enzyme catalase increased life shortening play a role in the death and dysfunc-
span in transgenic mice (Schriner et  al., 2005). tion of non-rapidly dividing cells such as heart cells
Other antioxidant enzymes also increased life span and that these functional abnormalities induced in
in Drosophila (Orr et al., 2013). mdx/mTRko can be reduced by administration of
The most important system providing antioxi- antioxidants.
dants and defending against ROS is the NADPH The toxicity from radiation is therefore both
defense system described later in this chapter. The acute and chronic. Acute death occurs from effects
preceding reports show that ROS can accelerate on the central nervous system (CNS) at doses
telomere shortening, damage DNA directly, induce of 20 Gy, or slow, long-term accumulated DNA
senescence, and hasten the aging process (Correia- damage leading to cell death from multiple hits
Melo et al., 2014). Suppressing ROS may slow the (Szilard, 1959). An earlier speculation by Erwin
shortening of telomeres and decrease activation Schrodinger attributed life to the maintenance
of other mechanisms implicated in the pathology of negative entropy by homeostatic processes
of aging (Harman, 1956, 1981). We postulate that (Schrodinger, 1944). In this view, aging and death
using ketone body esters to suppress ROS would would be the result of an increase in entropy. This
be effective in retarding the aging process itself. general statement without a biochemical or physi-
Telomere shortening occurs during normal ological mechanism is of limited practical use,
aging, leading to uncapping, activating senes- whereas applications of thermodynamics as exem-
cence and apoptotic programs in tissues with plified by the energetics of the various redox states
high cell turnover. Rapidly dividing cells—gut, and the process of oxidative phosphorylation have
skin, hematopoietic, and immune cells—replicate heuristic value.
constantly during life, and aging is visible to any-
body in skin as it changes from thick and smooth H OW A N T I OX I DA N T S   WO R K
in youth to thin and wrinkled in the aged. The There is a basic pattern to how antioxidant small
effects of nuclear radiation are likewise ampli- molecules work. They follow the pattern of reac-
fied in these tissues, exposure in the range of tion shown in Figure 27.3.
258

258 section III: Ketogenic Diet in the Laboratory

Antioxidant + R• RH + Antioxidant •

HO O
+ R• RH +
O O

Tocopherol Tocopheroxyl Radical


HO HO

HO HO
O O + R• RH + O
O
H H

HO OH
O O

Ascorbic Acid Ascorbyl Radical


FIGURE 27.3 The basic pattern of antioxidant reactions results in an antioxidant radical, which must be reduced
before it can participate in additional reactions. The antioxidant free radicals are sometimes obscured as they are the first
step in a reaction mechanism that may go to other forms.

The antioxidant is oxidized and the free radical The redox state of the cytoplasmic NAD-
is reduced. Why then is it not a problem that the couple in vivo was first defined in 1955 by Lynen
antioxidant has become a free radical? The answer and his coworkers (Holzer et  al., 1956)  in yeast
is that the unpaired electron in the now antioxi- by measurement of the ratio of the metabolites
dant radical is delocalized and thus less reactive. [ethanol]/[acetaldehyde] in a reaction brought to
This property makes ascorbate and tocopherol near-equilibrium by the very active cytoplasmic
distinct from other cellular reducing agents, as enzyme alcohol dehydrogenase. Soon thereafter it
they remain stable compounds after a single elec- was shown by Bucher and Klingenberg (1958) that
tron transfer. This is in contrast to glutathione, a the free [NAD+]/[NADH] in rat liver cytoplasm
powerful and important reducing agent, but one could be estimated by measurement of either
that must transfer two electrons at once to remain the [lactate]/[pyruvate] or [dihydroxyacetone-P]/
stable, as the glutathione radical is highly reactive. [3-glycerophosphate] ratio. Because of the rapid-
Tocopherol is able to transfer a single electron but ity with which these cellular redox state estimates
only a single electron, ascorbate is able to trans- change with hypoxia, accurate determinations
fer two—and, importantly, two ascorbate radicals were facilitated by the development of freeze
are able to react, forming a single molecule of clamping (Wollenberger et  al., 1960). That ratio
dihydroascorbate and one molecule of ascorbate. of [lactate]/[pyruvate] was found to be between 5
Dihydroascorbate in turn is able to accept two and 10, giving a free cytosolic [NAD+]/[NADH]
electrons from glutathione and be regenerated, calculated from the measured Keq of the reac-
bridging single electron redox couples with two tion to be about 1,000 in the fed liver and 250
electron couples. Glutathione itself is reduced by in a starved liver. Following these principles,
the free [NADP+]/[NADPH] redox couple. It is in 1967 Krebs and his coworkers (Williamson
important to understand factors controlling the 1967)  showed that the free [NAD+]/[NADH]
cellular redox states to understand how the anti- ratio in mitochondria could be estimated by
oxidants get recharged (reduced). measurement of ether the [acetoacetate]/[β-d-
hydroxybutyrate] or [α-ketoglutarate]×[NH4+]/
THE CELLULAR REDOX [glutamate] ratio and the ratio of free mitochon-
S TAT E S D I F F E R F R O M   T H E drial [NAD+ ]/[NADH] was found to be between
S TA N D A R D R E D O X 2 to 10. Following a question to Krebs by George
P OT E N T I A L S Cahill, Krebs assigned Richard Veech to deter-
A great deal of confusion in the literature on oxy- mine the free cytoplasmic [NADP+]/[NADPH]
gen free radicals results from the use of tables of ratio, which was known be used in the reduc-
standard reduction potentials, ΔEo, done at pH 7.0 tive synthesis of fats. This ratio was found to be
with all reactants at 1 M. about 0.01 to 0.02 and could be estimated by
259

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 259

TABLE 27.2 THE ACTIVIT Y OF NADP- LINKED ENZYMES IN RAT LIVER

Enzyme Activity in Keq = Metabolite couple Keq value


µmol/min/g

Isocitrate DH 22 ⎡αKG2− ⎤⎦ [CO2 ][ NADPH ] 1.17 M


EC 1.1.1.41 K eq = ⎣
⎡⎣Isocitrate3− ⎤⎦ ⎡⎣ NADP + ⎤⎦
Glutathione Reductase 7
K eq =
[GSSG][NADPH] ⎡⎣H+ ⎤⎦ 100 × 10–7 (Scott et al.,
EC 1.6.4.2 1963)
[GSH]2 ⎣⎡ NADP+ ⎤⎦
6 P Gluconate DH EC 2.8 ⎡Ribulose 5P2− ⎤⎦ [CO2 ][ NADPH ] 1.72 × 10–1 M (Villet and
1.1.1.44 K eq = ⎣ Dalziel, 1969)
⎡⎣6P Gluconate3− ⎤⎦ ⎡⎣ NADP+ ⎤⎦
Glucose 6-P DH EC 1.4 Not applicable because of lactonase
1.1.1.49
Malic Enzyme EC 1.27 ⎡ pyruvate− ⎤⎦ [CO2 ][ NADPH ] 3.44 × 10–2 M
1.1.1.39 K eq = ⎣
⎡⎣ Malate2− ⎤⎦ ⎡⎣ NADP + ⎤⎦

Source: Veech et al. (1969)

measurement of [α-ketoglutarate]×[CO2]/[iso- two major intracellular antioxidants, glutathione


citrate] or several other couples (see Table 27.2) (GSH) and ascorbic acid (Vitamin C). As these
The standard potential of the NAD- couple is antioxidants are in near equilibrium with the free
about −0.32 V, but these couples have three cytoplasmic [NADP+]/[NADPH] couple, whose
potentials in the cell depending on the intracel- large reducing potential favors the reduced, active
lular location of the enzymes involved. The three forms of GSH and Vitamin C (see Table 27.2 for
redox couples have redox potentials appropriate a list of these redox couples and Figure 27.4 for a
to the metabolic pathways they drive. diagram of the ROS-quenching reactions driven
First, in cytoplasm, the ΔE of the free NAD- by the NADP couple).
couple is −0.19 V, this smaller potential pois- These major cellular redox states are related
ing it to receive reducing power from glycolysis. to one another through common intermedi-
Second, for NAD in mitochondria, it is −0.28 V, ary metabolites (Bergman et  al., 2010; Krebs,
allowing for more energy to be available for the 1969)  and also to the cellular phosphorylation
electron transport pathway to convert ADP to potential or ATP/ADPxPi ratio keeping the energy
ATP. Third, the NADP- couple in the cytosol was of ATP hydrolysis in the range of −53 to −57 kJ/
estimated to be about −0.42 V, the lowest reduc- mole (Veech et al., 1979). It can be seen that these
tion potential in the cell (Krebs & Veech 1969). intracellular redox potentials cannot be inferred by
This strongly reducing potential allows this couple study to the standard potential of redox half reac-
not only to synthesize fats but also to favor the tions, which have led to much confusion in writ-
inactivation of oxygen free radicals through the ing about cellular redox potentials (Winterbourn,

S
NADP+ TrxR-S2 TrxR Trx-S2 Protein-S2
FAD Se

SH
NADPH
+
TrxR-(SH)2 TrxR Trx-(SH)2 Protein-SH2
+H
FAD Se–

FIGURE  27.4 Thioredoxin is a family of three proteins in mammals—TrxR1 (cytosolic), TrxR2 (mitochondrial),
TrxR3 (testis specific)—that can reduce protein disulfide bonds formed by ROS/RNS damage. Abbreviations: TrxR—
thioredoxin, FAD—flavin adenine dinucleotide, S2—disulfide, SH2—two sulfhydryl moieties.
260

260 section III: Ketogenic Diet in the Laboratory

2008). This history of the development of the mea- For [pyruvate]/[lactate] = 1:10


surement of in vivo redox states has been reviewed
(Veech, 2005, 2006). E NAD/NADH = −0.32V + 0.091V = −0.229V
Measurement of each of the metabolite cou-
ples listed in Table  27.2 yields the same value of The free cytosolic [NADP+]/[NADPH] redox
the free cytosolic [NADP+]/[NADPH] ratio with a potential calculated from isocitrate dehydroge-
redox potential of about −0.4 V, the most negative nase reaction and from the malic enzyme agree
of any intracellular potential (Krebs, 1969). With well and are the most negative potential in the
small refinements of the Keq, CO2 concentrations cell, although with improved metabolite mea-
and intracellular metabolite concentrations in the surements are slightly above the −0.42V esti-
intervening years, the potential of the NADP sys- mated in the original Krebs and Veech paper
tem is more likely to be in the range of −0.36 to (Krebs, 1969).
−0.38 V, but is still the most negative potential in The redox potential from isocitrate dehydroge-
the cell. nase reaction:
The formalism to calculate the redox potential
in the cell estimated from measured intracellular RT ⎛ NADP + ⎞
metabolites and the measured Keq of the reac- E NADP / NADPH = Eo NADP / NADPH + ln
tions involved is as follows. The standard redox nF ⎜⎝ NADPH ⎟⎠
potential is derived from the equilibrium con-
stant of the reaction. The standard potential of [Isocitrate−3 ] + [NADP + ] → [α-Ketoglutarate2 − ]
both the NAD and NADP couples, Eo, is, within + [NADPH] +[CO2 ]
experimental error, −0.32 V (Burton, 1974).
The redox potential within the cell is calculated
at follows (Veech, 2005): [NADP + ] [αKG2 − ] × [CO2 ] 1
= 3−
×
[NADPH] [Isocit ] K ICDH
RT ⎛ NAD+ ⎞
E NAD / NADH = Eo NAD / NADH + ln Eqn 1
nF ⎜⎝ NADH ⎟⎠
RT ⎛ [αKG2− ] × [CO2 ] 1 ⎞
E = Eo + ln ×
Where R = 8.1345 × 10−3 kJ/mole/oK
NADP
NADPH
NADP
NADPH
nF ⎜⎝ [Isocit 3− ] K ICDH ⎟⎠
T = 311.15 oK
n = number of electrons = 2 for lactate For [α-Ketoglutarate]:[Isocitrate] = 7:1 (Liver) and
dehydrogenase [CO2] = 1.985mM
F = 96.485 kJ/mole/V
E NADP = −0.32V − 0.044 V = −0.364 V
The free cytosolic [NAD+ ]/[NADH] calculated NADPH
from the lactate dehydrogenase reaction:
The redox potential from from the malic enzyme
[lactate − ] + [ NAD + ] → [pyruvate − ]
reaction:
+ [ NADH] + [H + ]
[NADP+ ] [Pyr − ] × [CO2 ] 1
= ×
At pH 7 [NADPH] [Mal 2 − ] K MalEnz

[Pyr − ] [NADH]
× = K LDH =1.11 × 10−4 where [pyruvate]/[malate] = 1:3 in liver
[Lac − ] [NAD+ ]
E NADP = −0.32V − 0.055V = −0.375V
Rearranged and substituted into equation 1 above NADPH

RT ⎛ [Pyr − ] 1 ⎞ Because the glutathione couple reacts with the


E NAD / NADH = Eo NAD / NADH + ln ⎜ × NADP- couple (see Table 27.3), the potential of the
nF ⎝ [Lac ] K LDH ⎟⎠

NADP- couple determines the redox potential of
the glutathione redox couple [GSH]2/[GSSG]) with
For [pyruvate]/[lactate] = 1:1 which it is in near-equilibrium at −0.37V, not the
standard potential of −0.32V at pH 7.0. The termi-
E NAD / NADH = −0.32V + 0.122V = −0.198V nal reduction of the oxygen free radicals involves
261

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 261

TABLE 27.3 ENZYMATIC SYSTEMS WHICH ASSUME THE REDOX POTENTIAL


OF THE NADP- SYSTEM

Enzyme Reaction

Glutathione Reductase EC 1.6.4.2 NADPH + GSSG + H + → NADP + + 2GSH

Glutathione Dehydrogenase 2GSH + Dehydroascorbate → GSSG+ Ascorbate


EC 1.8.5.1

Glutathione Peroxidase EC 1.11.1.9 2GSH + H2O2 → GSSG + H2O

Glutathione Transferase EC 2.5.1.8 HO–Xenobiotic + GSH → GS–Xenobiotic + H2O

Thioredoxin Reductase EC 1.6.4.5 NADPH + H + + Oxidized Thioredoxin → NADP+ + Reduced Thioredoxin

glutathione, which constitutes the most abundant Thus, the potential of the ascorbate couple and
intracellular antioxidant at 5 mM (Krebs, 1969). the glutathione couple are the same as that of the
Ascorbate, at 2–3 mM, is the second most NADP- couple. Likewise, disulfide (SS) groups
abundant reducing agent and antioxidant, and is in protein are reduced to –SH by the thioredoxin
also the cofactor for a number of metal-dependent system.
oxygenases and dioxygenases, such as collagen
prolyl and lysyl hydroxylases (Linster and Van T H E R E D O X S TAT E
Schaftingen, 2007). Ascorbate, when acting as a OF OXYGEN FREE RADICALS
reducing agent, loses an electron and a proton to A N D O T H E R R E L E VA N T
form semidehydroascorbate (SDA), which upon R E A C TA N T S
losing another electron forms dehydroascorbate A list of relevant redox potentials is listed in
(DHA). Regeneration of ascorbate by glutathione Table 27.4 below. The values for the redox poten-
occurs by the reaction: DHA + 2GSH ↔ ascorbate + tials given are taken from Bergman et  al. (2010),
GSSG, which can occur spontaneously (Linster Clark (1960), Dawes (1971), and Krebs (1969).
and Van Schaftingen, 2007)  but can also be cata- This table shows the oxidizing potential of
lyzed by the enzyme glutathione dehydrogenase ROS compounds. However, it should be pointed
(EC 1.8.5.1), an enzyme with an activity in rat liver out that these half reactions do not represent the
of about 3 µmol/min/g wet weight (Carlberg and redox potential inside of cells, which are set by the
Mannervik, 1975). These reactions (Table 27.3) NAD and NADP couples which carry the domi-
ensure that the redox potential of ascorbic acid and nant metabolic flux during metabolism.
the glutathione couple in vivo assume the redox
potential of the cytosolic NADP- couple of −0.37 H O W D - Β H B A LT E R S
V and not the standard potential—this is in sharp C E L L U L A R R E D O X   S TAT E S
contrast to ascorbate’s standard reduction potential The effects of insulin, glucose, and d-β-
of −0.06 V (Table  27.4) or the standard potential hydroxybutyrate (d-βHB) on a perfused rat heart
of the glutathione couple at −0.23 V (Clark, 1960). gave insights into the changes in relative concen-
The semidehydroascorbte (SDA) is also trations of key metabolites involved in the Krebs
reduced to ascorbate by thioredoxin reductase cycle and oxidative phosphorylation and the free
(EC 1.6.4.5) (Linster and Van Schaftingen, 2007), [NADP+]/[NADPH] ratio. Prior work on the
an NADP-linked enzyme that is the major cellular effects of hearts perfused with d-βHB showed
protein disulfide reductase (Arner and Holmgren, that the [isocitrate]/[α-ketoglutarate] (Sato et  al.,
2000). This enzyme catalyzes the reaction: NADPH 1995) couple was increased. This change indicated
+ thioredoxinox → NADP+ + thioredoxinred. This a reduction of the NADP- couple (Kashiwaya
enzyme is critical in signaling as part of thiol redox et al., 1997). Figure 27.5 shows the increased ratio
control, and regulates the activity of a number of of [NADPH]/[NADP+] found in the perfused rat
transcription factors. The potential of the thiore- heart studies, which supports a highly significant
doxin system is likewise set by the potential of the increase in the capacity for cellular ROS/RNS
free cytosolic [NADP+]/[NADPH] couple. detoxification. (We are intentionally breaking a
262

TABLE 27.4 REDOX POTENTIAL OF SOME BIOCHEMICALLY IMPORTANT


HALF- REACTIONS IN THE BIOCHEMICAL HALF REACTIONS, ALL
REACTANTS ARE AT 1 M EXCEPT H + WHICH IS AT PH 7.

Half reaction Redox Potential of half reactions

H + + e − → 1 2 H2 -0.41

Acetyl − CoA + 2H + + 2e − → Acetaldehyde + CoA –0.42

Acetoacetate− + 2H + 2e − → β − hydroxybutyrate − –0.36


+ + − +
NAD + 2H + 2e → NADH+H –0.32

NADP + + 2H + + 2e − → NADPH+H + –0.32


2− + − 3−
α ketoglutarate + CO2 + 2H + 2e → isocitrate –0.318

Pyruvate + 2H + + 2e − → Lactate –0.19

FAD + 2H + + 2e − → FADH2 –0.06

Fumarate + 2H + + 2e − → Succinate +0.031

Dehydroascorbate + 2H + 2e → Ascorbate + −
+0.06
2− 3−
α − ketoglutarate + CO2 → isocitrate +0.002
+
Ubiquinone + 2H + 2e → Dihydroubiquinone −
+0.10

1
2 O 2 + H 2 O + 2e → H 2O 2 +0.30

O2 + 2H + + 2e − → H 2 O2 +0.605
3+ − 2+
Fe + e → Fe +0.77
+ −
2 O 2 + H + 2e → H 2O +0.816
1

− +
O2 + 2H → H 2 O2 +0.89
− −
•OH + e → OH +1.89

1500
Glucose
Insulin
1000 Ketones
Insulin + Ketones
Ratio

500

0
[NADH]/[NAD+] [NADPH]/[NADP+]
(x10–5)

FIGURE  27.5 Studies of a perfused rat heart demonstrated that the ketone body D-βOHB raised the ratio of
[NADPH]/[NADP+]. Modified presentation of data published in Kashiwaya et al. 1997.
263

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 263

D-β-hydroxybutyrate NAD+
H2O2
βHBDH 2GSH
glutathione peroxidase
acetoacetate NADH
3 oxoacid 2H2O
CoA transferase
acetyl CoA
citrate synthase oxaloacetate Vit Cox
H2O
citrate HO•
aconitase NADP+ R•
glutathione
dehydrogenase Vit Ered
isocitrate RH
glutathione
Isocitrate dehydrogenase reductase
LOO•
α-ketoglutarate tetrahydrobiopterin
Vit Cred Vit Eox
Dihydropiopterin reductase
LOOH
dihydrobiopterin ONOO–
2 protein-SH
thioredoxin
H2O + NO2
protein-SS-protein

NADPH GSSG

FIGURE 27.6 The “NADP system” is driven by the [NADP+]/[NADPH] couples redox potential. [NADPH] was
elevated relative to [NADP+] in perfused rat heart studies with d-bOHB. To the left of NADPH shown in the diagram
is the relevant pathway to reducing NADP+ to NADPH. NADPH also provides the reducing potential to counteract
ROS and RNS through antioxidants that must be reduced each time they are oxidized as they quench a ROS or RNS.
Abbreviations: NAD—nicotinamide adenine dinucleotide, NADH—reduced NAD+, NADP—nicotinamide adenine
dinucleotide phosphate, NADPH—reduced NADP+, GSH—glutathione, GSSG glutathione disulfide, Vit C—vitamin
C or ascorbate, Vit E—vitamin E or a tocopherol, R•—free radical on a carbon atom, RH—an organic molecule with
C-H bond, LOO•—lipid peroxyl radical, LOOH—lipid peroxide.

long-standing rule by putting the reduced form R-1,3-butanediol referred to as ketone ester (KE),
in the numerator to simplify the discussion where was used. This compound has been shown to safely
an increase in [NADP+]/[NADPH] would other- increase levels of blood d-βHB to 5–7 mM (Clarke
wise be reported as a decrease in the inverse of the et  al., 2012a; Clarke et  al., 2012b). The ester is
ratio.) hydrolyzed in the gut and blood by esterases, and
Figure 27.6 shows some of the “NADP system” the resulting R-1,3 butane diol is converted to
that is relevant to removal of ROS and RNS as well d-βHB in the liver (Figure 27.7).
as the link back to d-βHB.
T H E R E M OVA L O F   F R E E
K E TO S I S A S   A M E A N S R A D I C A L S I N   V I VO
O F   P ROT E C T I O N Clinical trials of administration of antioxidants
AGAINST IONIZING have often been disappointing (Winterbourn,
R A D I AT I O N 2008). Most placebo trials of antioxidants have
At present there are very few approved treatments failed (Steinhubl, 2008), most notably in the
for exposure to nuclear accidents or rogue nuclear long-advocated use of high-dose ascorbic acid.
bombs (Buddemeier, 2011). Experiments have The use of vitamin antioxidants has not objec-
been conducted at the Armed Forces Radiobiology tively been proven helpful in the absence of vita-
Research Institute (AFRRI). The study investigat- min deficiency and may even be harmful (Howes,
ing d-βHB protection against acute ROS injury 2011). More recently, high doses of Vitamin E are
utilized in vitro radiation lethality on HOS reported to increase melanoma metastases in mice
(human osteoblast) cells exposed to several doses (LeGal, 2015).
and types of radiation. For the studies described While it is true that, quantitatively, glutathione
below, a novel ester of D-β-hydroxybutyrate and at about 5 mM and ascorbic acid at about 2–3 mM
264

264 section III: Ketogenic Diet in the Laboratory


OH O

OH O OH OH
Gut
esterases D-β-hydroxybutyrate Alcohol
+ dehydrogenase
O
OH OH
(R)-((R)-3-hydroxybutyl) 3 -hydroxybutanoate

R 1, 3 butanediol
FIGURE 27.7 The monoester of D-βOHB and R-1,3 butanediol is (R)-((R)3- hydroxybutyl) 3- hydroxybutyrate is
converted to two molecules of D-βOHB and provides an alternative fuel to produce ATP.

in most cells are the predominant intracellular Hydrogen peroxide can react with catalase (EC
antioxidants, one cannot change the redox poten- 1.11.1.6) in cytoplasm, but not mitochondria, to
tial of an antioxidant in vivo by simply adding form water and O2.
more of the reduced form of the couple.
2 H 2O 2 → O 2 + 2H 2O
R E M OVA L O F   R E A C T I V E
OXYGEN SPECIES Alternatively, H2O2 can react with glutathione in a
B Y   G E N E R AT I O N reaction catalyzed by glutathione peroxidase (EC
OF NADPH 1.11. 1.9) to form oxidized glutathione and water.
We have discussed earlier the failure to remove
ROS by addition of antioxidants such as ascorbic 2GSH + H 2O2 → GSSG + 2H 2O
acid (Steinhubl, 2008). It is likewise not possible
to add the reduced component of most NADPH
producing metabolite couples (see Table 27.2 THE EFFECTS OF D-ΒHB
above), since these multiply-charged com- ON TRANSCRIPTION
pounds do not readily enter the interior of cells. In addition to the ability of d-βHB metabolism
It is, however, possible to introduce the reduced to generate NADPH, the metabolite d-βHB is the
ketone body, d-βHB, which readily enters cells natural inhibitor of class I and II histone deacety-
on the monocarboxylate transporter, MCT. lases (Shimazu et al., 2013; Veech, 2014) that were
While not an NADP- coupled metabolite, d-βHB formerly identified by their ability to bind nico-
produces acetyl CoA and, via the Krebs cycle, tinic acid (Offermanns, 2006; Tunaru et al., 2003).
increased concentrations of citrate and isoci- The inhibition of histone deacetylase increases
trate, but causes little change in α-ketoglutarate transcription of the enzymes of the antioxidant
(Sato et  al., 1995). This increase in the [isoci- pathway controlled by the FOXO3A and Mt1
trate]/[α-ketoglutarate ] ratio causes a reduc- transcription factors, thus increasing the activ-
tion of the NADP- system (Kashiwaya et  al., ity of Mn superoxide dismutase and catalase (see
1997) generated by the metabolism involving the Table 27.3). d-βHB thus increases the reductive
most active NADP linked dehydrogenase, isoci- capacity of the pathways used to destroy free radi-
trate dehydrogenase. The cellular metabolic pro- cals by increasing the enzymes in the pathway used
cesses thus produce an increased reducing power for free radical destruction. Recent studies have
of the NADP system, which is regenerated by the shown that HDAC inhibitors may have a signifi-
cellular metabolism. cant benefit in radiation protections (Brown et al.,
2008; Miller et al., 2011). Therefore, in vitro studies
THE ENZYMES were initiated to test whether KE could improve
D E S T R OY I N G R E A C T I V E radiation survival and decrease radiation-induced
OXYGEN SPECIES damage.
Superoxide dismutase also can combine two super-
oxide radicals, along with two protons to form the IN VITRO STUDIES
less toxic hydrogen peroxide. Human osteoblast cells (HOS) were chosen as the
model system to study KE’s potential as a radia-
O2 − + O2 − + 2H + → H 2O2 + O2 tion countermeasure. The HOS cells have been
265

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 265

(a) Pre f-Radiation (b) Post f-Radiation

1 Control 1
KE
Surviving Fraction

Surviving Fraction
0.1 0.1

0.01 0.01

0.001 0.001

0.0001 0.0001
0 2 4 6 8 10 12 0 2 4 6 8 10 12
Radiation Dose (Gy) Radiation Dose (Gy)

FIGURE 27.8 Survival curves for human osteoblasts (HOS) in culture exposed to either gamma (60Co, 66 gCy/min)
or proton radiation (4MeV). KE-treated cell cultures received 50 µM of ketone ester (KE) in the cell media 30 minutes
prior to or for 30 minutes after radiation exposure.

previously used to evaluate potential radiation greater likelihood of exposure to proton radiation
countermeasures (Miller et al., 1998; Miller et al., than other populations.
2011). The HOS cells were incubated with KE Genomic effects of ROS begin as discrete events
(50 µM, 15 min to 1 h) either before (Figure 27.8A) such as a break in the phosphate backbone in DNA,
or immediately after (Figure  27.8B) exposure to nitrogenous base oxidation, or dimerization.
gamma radiation (60Co, 0.66 cGy/min). Following Breaks in the phosphate bonds occur as either sin-
radiation, HOS cells were plated in 100-mm petri gle, clustered, or double-stranded breaks, depend-
dishes to assess survival of colony-forming cells; at ing on the level of the ROS or radiation dose and
10 days post radiation, colonies were fixed, stained, type. As the frequency of breaks increases, macro
and counted to determine the survival fraction. effects such as abnormal chromosomes, muta-
The survival curves show that KE increased sur- genesis, and inhibited cell division become more
vival when delivered either immediately before or frequent (Lomax et  al., 2013). In general, single-
immediately after radiation. These data strongly stranded breaks are quickly repaired, have low
suggest that KE would have benefit to human cytotoxicity and occur at high levels even without
health both as a protectant delivered to popula- exogenous ROS or radiation (Cadet et  al., 2008).
tions at risk of exposure or as a mitigant after radi- Single-stranded breaks are induced at levels four-
ation. The ability of KE to improve survival even to eightfold more than double-stranded breaks
when given 24 hours after radiation is compatible with exposure to electromagnetic radiation such
with the persistent production of superoxide from as gamma or x-rays (Lomax et al., 2013). Double-
NADPH oxidase s, DUOX 1 and 2, for several days stranded or clustered single-stranded breaks occur
after radiation exposure (Ameziane-El-Hassani with particle radiation, especially α and proton, or
et al., 2015). when ROS is focused in a confined area (Lomax
Proton radiation was used to determine the et  al., 2013). A  double-stranded break is defined
ability of the KE to protect against particle radia- as two breaks within 10 bases (Lomax et al., 2013).
tion, which produces less oxidative damage than These breaks are much more difficult to repair and
gamma radiation but is more likely to induce can persist for more than 24 hours when large
double-stranded DNA breaks (Lomax et al., 2013). numbers are generated (Schmid et al., 2010). These
Cells were exposed to KE before proton radia- breaks are repaired by either homologous recombi-
tion exposure, similar to that performed above in nation or by nonhomologous end joining (Lomax
Figure 27.8A with gamma radiation. The KE was et al., 2013), with the latter being the predominant
able to confer significant protection against proton mechanism in most phases of the cell cycle. It is
radiation in this model (Figure  28.8C). Overall possible to observe these breaks by examining
cell death was higher for this type of radiation, some of the hallmarks of their repair, by observ-
which is to be expected, based on previous studies. ing different types of aberrant chromosomes, or by
Protection against proton radiation is of particular observing cell cycle arrest. Indeed, when cultured
interest to NASA, as astronauts have a substantially cells were treated with d-βHB before an exposure
266

266 section III: Ketogenic Diet in the Laboratory

Percentage Aberrant Metaphases Fragments


100 1.5
Control
80
∆G
1.0
60

40
0.5
20

0 0.0
0 1 2 3 4 5 0 1 2 3 4 5

Breaks Rings & Dicentrics


0.020 0.05

0.04
0.015
0.03
0.010
0.02
0.005
0.01

0.000 0.00
0 1 2 3 4 5 0 1 2 3 4 5
Radiation Dose (Gy) Radiation Dose (Gy)

FIGURE 27.9 The percentage of chromosomal anomalies (aberrant metaphases, fragments, breaks, and rings and
dicentrics) occurring from γ-radiation-exposed human osteoblast cells (HOS) with or without KE. The 50-µM KE was
added to the cell culture media 30 minutes prior to radiation exposure and removed immediately prior to radiation
exposure. Cells were scored using a light microscope.

to γ radiation, production of chromosomal aber- unrelated compounds that all target HDACs seem
rations and cell cycle arrest were both significantly to confer similar protection and enhanced repair
suppressed (Figure 27.9). after exposure (Brown et  al., 2008; Miller et  al.,
A novel approach for stimulating repair of 2011; Shimazu et al., 2013).
radiation or ROS-induced DNA damage utilizes The effects of persistent DNA damage are most
HDAC inhibitors. Inhibitors of HDAC activate quickly seen in tissues with high cell turnover that
important DNA repair pathways such as the Nrf2 rely on actively dividing stem cells such as skin,
and Rad51 and decrease markers of damage, even the gastrointestinal system, and blood. Damage
if the inhibitor is given after radiation exposure in these systems produces some of the rapid and
(Brown et  al., 2008; Miller et  al., 2011). Histone most visible radiation effects, such as hair loss,
deacetylase (HDAC) inhibitors have broad activity, nausea/vomiting, bone marrow suppression, and
their primary function is to increase the acetyla- skin lesions. Damage to DNA is also responsible
tion of histones by preventing their deacetylation, for much of the increased risk of cancer that results
in turn making the genome less tightly packed from radiation exposure.
and more transcriptionally active (Verdin and
Ott, 2015). The deacetylating activity of HDACs E F F E C T S O F   K E TO N E
can also extend to various other cellular proteins, ESTERS AFTER EXPOSURE
modulating activity of enzymes and protein- T O   R A D I AT I O N I N   V I VO
protein binding (Verdin and Ott, 2015). With A project is underway to evaluate the ability of KE
huge numbers of potential effects resulting from to increase murine survival after gamma radia-
inhibiting HDACs, it is difficult to determine what tion exposure. Previous studies have shown that
the final effectors of the protection are, but stud- exposure of the DBA2 inbred mouse strain to 6
ies have demonstrated that a series of chemically Gy of γ-radiation results in reproducible bone
267

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 267

(a) (b)
110 100

90 Control
KE 80

Incidence per 1000 cells


Incidence per 100 cells
70 *
50 * 60
*
30
12 40
10 *
8 *
6 * 20
4 * * *
2
0 0
MPCE MNCE
s
Fr ells

dy

S
om ice s

eB s
Po aks
nt

D ak

om tric

SD
oi
om gme

re
tC

Br

pl
an

ly
id
a
rr

at
be

as
an
A

hr

gs
%

hr
in

C
R

FIGURE 27.10 The effect of KE on the incidence rate of chromosome anomalies and micronuclei in mouse bone
marrow 48 h after 6 Gy γ-radiation. KE (750 mg/kg) or saline were delivered by gavage 24 hours after radiation exposure.
(A) Chromosomal malformation incidence per 100 cells, all cells in bone marrow were examined and malformations were
scored using a light microscope. For metaphases approximately 1,000 total were counted, 200/animal. (B) Micronuclei
were counted only in reticulocytes and erythrocytes, 10,000 total were counted per animal. Data presented as average ±
SEM, N = 5 animals, *, p < .01 by t-test with a Sidack-Bonferonni multiple comparison correction.

marrow chromosomal damage in 85-day-old doses of γ-radiation (Figure 27.11) in control ani-
mice (Grahn, 1958). In our study we have tested a mals. This decrease was significantly attenuated
single KE administration of 750 mg/kg by gavage by 750mg/kg KE 24 hours after radiation. Further
24 hours after radiation exposure. This postradia- experiments to determine KE effects on radiation
tion treatment led to a consistent 50% decrease lethality are ongoing.
in numerous types of radiation-induced chromo-
somal malformations and abnormal cell divisions
as observed by microscopy 24 hours after KE 1.2 Control
Reticulocyte: Erythrocyte Ratio

administration (Figure 27.10). The importance of KE


decreasing these indicators of DNA damage and 1.0
genomic instability is underscored by the cytotox-
icity of this type of damage and the potential for +
0.8
neoplastic transformation, ultimately resulting in
+
cancer. This dose of KE also blunted the radiation-
induced increase in incidence of micronuclei, 0.6 *
which arise from improper chromatid separation +
during anaphase, in both reticulocytes and eryth-
0.4
rocytes, which were counted using the methods of 0 2 4 6
Schmid (1975). -Radiation Dose (Gy)
In order to explore KE effects on radiation
bone marrow suppression and impact on hema- FIGURE  27.11 The effect of KE on the relative
topoiesis, an important mechanism for radiation- abundance of polychromatic and normochromatic
induced mortality at the 6 Gy level, the ratio erythrocytes in mouse bone marrow 48 h after 0, 0.5, or 6
between total reticulocytes and erythrocytes in Gy γ-radiation. KE (750 mg/kg) or saline were delivered
bone marrow was determined. Bone marrow was by gavage 24 hours after radiation exposure. 10,000
extracted as before, 24 hours after KE or saline erythrocytes were counted per animal. Data presented
gavage, a total of 48 hours after either 0.5 or 6 Gy as average ± SEM, N  =  5 animals. *, p < .01 significant
γ-radiation. This ratio, which is approximately difference between KE and control, +, p < 0.05 significant
1 in bone marrow under normal circumstances, difference versus no radiation, by t-test with a Sidack-
was significantly decreased by both 0.5 and 6 Gy Bonferonni multiple comparison correction.
268

268 section III: Ketogenic Diet in the Laboratory

CONCLUSION glutathione reductase from rat liver. J Biol Chem


Reactive oxygen species play a central role in both 250, 5475–5480.
radiation damage and aging. It is proposed that Chance, B., Sies, H., and Boveris, A. (1979).
both processes can be ameliorated by the admin- Hydroperoxide metabolism in mammalian organs.
istration of KE to increase the amount of anti- Physiol Rev 59, 527–605.
Clark, W.M. (1960). Oxidation Reduction Potentials
oxidant, enzymes, and the reducing power of the
of Organic Systems (Baltimore:  Williams and
NADP- system.
Wilkins).
Clarke, K., Tchabanenko, K., Pawlosky, R., Carter, E.,
REFERENCES Knight, N.S., Murray, A.J., Cochlin, L.E., King,
Acquier, A.B., Mori Sequeiros Garcia, M., Gorostizaga, M.T., Wong, A.W., Roberts, A., et al. (2012a). Oral
A.B., Paz, C., and Mendez, C.F. (2013). Reactive 28-day and developmental toxicity studies of (R)-
oxygen species mediate dopamine-induced signal- 3-hydroxybutyl (R)-3-hydroxybutyrate. Regul
ing in renal proximal tubule cells. FEBS Lett 587, Toxicol Pharmacol 63, 196–208.
3254–3260. Clarke, K., Tchabanenko, K., Pawlosky, R., Carter,
Ameziane-El-Hassani, R., Talbot, M., de Souza Dos E., Todd King, M., Musa-Veloso, K., Ho, M.,
Santos, M.C., Al Ghuzlan, A., Hartl, D., Bidart, Roberts, A., Robertson, J., Vanitallie, T.B., et  al.
J.M., De Deken, X., Miot, F., Diallo, I., de Vathaire, (2012b). Kinetics, safety and tolerability of (R)-
F., et  al. (2015). NADPH oxidase DUOX1 pro- 3-hydroxybutyl (R)-3-hydroxybutyrate in healthy
motes long-term persistence of oxidative stress adult subjects. Regul Toxicol Pharmacol 63,
after an exposure to irradiation. Proc Natl Acad 401–408.
Sci U S A 112, 5051–5056. Correia-Melo, C., Hewitt, G., and Passos, J.F. (2014).
Arner, E.S., and Holmgren, A. (2000). Physiological Telomeres, oxidative stress and inflammatory
functions of thioredoxin and thioredoxin reduc- factors:  partners in cellular senescence? Longev
tase. Eur J Biochem 267, 6102–6109. Healthspan 3, 1.
Bedard, K., and Krause, K.H. (2007). The NOX family Dawes, E.A. (1971). Quantitative problems in bio-
of ROS-generating NADPH oxidases:  physiology chemistry, Vol. 3, 5th edition (Edinburgh &
and pathophysiology. Physiol Rev 87, 245–313. London: E. & S. Livingstone).
Ben-Porath, I., and Weinberg, R.A. (2005). The signals Grahn, D. (1958). Acute radiation response of mice
and pathways activating cellular senescence. Int J from a cross between radiosensitive and radiore-
Biochem Cell Biol 37, 961–976. sistant strains. Radiat Res 4, 189–198.
Bergman, C., Kashiwaya, Y., and Veech, R.L. (2010). Harley, C.B., Futcher, A.B., and Greider, C.W. (1990).
The effect of pH and free Mg2+ on ATP linked Telomeres shorten during ageing of human fibro-
enzymes and the calculation of Gibbs free energy of blasts. Nature 345, 458–460.
ATP hydrolysis. J Phys Chem B 114, 16137–16146. Harman, D. (1956). Aging:  a theory based on free
Blackburn, E.H., and Gall, J.G. (1978). A tandemly radical and radiation chemistry. J Gerontol 11,
repeated sequence at the termini of the extrachro- 298–300.
mosomal ribosomal RNA genes in Tetrahymena. J Harman, D. (1981). The aging process. Proc Natl Acad
Mol Biol 120, 33–53. Sci USA 78, 7124–7128.
Brown, S.L., Kolozsvary, A., Liu, J., Ryu, S., and Kim, Holzer, H., Schultz, G., and Lynen, F. (1956).
J.H. (2008). Histone deacetylase inhibitors protect Bestimmung des quotienten DPNH/DPN in leb-
against and mitigate the lethality of total-body enden hefezellen durch analyse stationarer alkohol
irradiation in mice. Radiat Res 169, 474–478. und acetaldehyde konzentrationen. Biochem Z
Bucher, T., and Klingenberg, M. (1958). Wege des 332, 252–263.
Wasserstoffs in der lebendigen Organisation. Howes, R. (2011). Mythology of antioxidant
Angewandte Chemie 70, 552–570. enzymes. J Evidence Based Compl Altern Med
Buddemeier, B.R. (2011). Key Response Planning Factors 16, 149–159.
for the Aftermath of Nuclear Terrorism (FEMA, Kashiwaya, Y., King, M.T., and Veech, R.L. (1997).
Homeland Security, Sandia National Laboratory, Substrate signaling by insulin:  a ketone bodies
Lawrence Livermore National Laboratory). ratio mimics insulin action in heart. Am J Cardiol
Burton, K. (1974). The enthalpy change for the reduc- 80, 50A–64A.
tion of nicotinamide-adenine dinucleotide. Kashiwaya, Y., Takeshima, T., Mori, N., Nakashima,
Biochem J 143, 365–368. K., Clarke, K., and Veech, R.L. (2000). D-beta-
Cadet, J., Douki, T., and Ravanat, J.L. (2008). Hydroxybutyrate protects neurons in models of
Oxidatively generated damage to the guanine moi- Alzheimer’s and Parkinson’s disease. Proc Natl
ety of DNA: mechanistic aspects and formation in Acad Sci USA 97, 5440–5444.
cells. Acc Chem Res 41, 1075–1083. Krebs, H.A., and Veech, R.L. (1969). Pyridine nucle-
Carlberg, I., and Mannervik, B. (1975). Purification otide interrelations. In The Energy Level and
and characterization of the flavoenzyme Metabolic Control in Mitochondria, S. Papa,
269

Chapter 27: Mitigation of Damage from Reactive Oxygen Species 269

Tager, J.M., Quagliariello, E., & Slater, E.C., eds. after exposure to low and high LET radiation. Int J
(Bari, Italy: Adriatica Editrice), pp. 329–384. Radiat Biol 86, 682–691.
LeGal, K. (2015). Antioxidants can increase melanoma Schmid, W. (1975). The micronucleus test. Mutation
metastasis in mice. Sci Transl Med 7, 1–7. Research 31, 9–15.
Linster, C.L., and Van Schaftingen, E. (2007). Vitamin Schriner, S.E., Linford, N.J., Martin, G.M., Treuting, P.,
C:  biosynthesis, recycling and degradation in Ogburn, C.E., Emond, M., Coskun, P.E., Ladiges,
mammals. FEBS J 274, 1–22. W., Wolf, N., Van Remmen, H., et  al. (2005).
Lomax, M.E., Folkes, L.K., and O’Neill, P. (2013). Extension of murine life span by overexpression
Biological consequences of radiation-induced of catalase targeted to mitochondria. Science 308,
DNA damage:  relevance to radiotherapy. Clin. 1909–1911.
Oncol. 25, 578–585. Schrodinger, E. (1944). What Is Life Mind and
Miller, A.C., Blakely, W.F., Livengood, D., Whittaker, Matter (Cambridge, UK:  Cambridge University
T., Xu, J., Ejnik, J.W., Hamilton, M.M., Parlette, Press).
E., John, T.S., Gerstenberg, H.M., et  al. (1998). Scott, E.M., Duncan, I.W., and Ekstrand, V. (1963).
Transformation of human osteoblast cells to the Purification and properties of glutathione reduc-
tumorigenic phenotype by depleted uranium- tase of human erythrocytes. J Biol Chem 238,
uranyl chloride. Environ Health Perspect 106, 3928–3933.
465–471. Sfeir, A. (2012). Telomeres at a glance. J Cell Sci 125,
Miller, A.C., Cohen, S., Stewart, M., Rivas, R., and 4173–4178.
Lison, P. (2011). Radioprotection by the histone Shimazu, T., Hirschey, M.D., Newman, J., He, W.,
deacetylase inhibitor phenylbutyrate. Radiat Shirakawa, K., Le Moan, N., Grueter, C.A.,
Environ Biophys 50, 585–596. Lim, H., Saunders, L.R., Stevens, R.D., et  al.
Mourkioti, F., Kustan, J., Kraft, P., Day, J.W., Zhao, (2013). Suppression of oxidative stress by beta-
M.M., Kost-Alimova, M., Protopopov, A., hydroxybutyrate, an endogenous histone deacety-
DePinho, R.A., Bernstein, D., Meeker, A.K., et al. lase inhibitor. Science 339, 211–214.
(2013). Role of telomere dysfunction in cardiac Sohal, R.S., and Weindruch, R. (1996). Oxidative
failure in Duchenne muscular dystrophy. Nat. Cell stress, caloric restriction, and aging. Science
Bio 15, 895–904. 273, 59–63.
Offermanns, S. (2006). The nicotinic acid receptor Speakman, J.R. (2005). Body size, energy metabolism
GPR109A (HM74A or PUMA-G) as a new thera- and lifespan. J Exp Biol 208, 1717–1730.
peutic target. Trends Pharmacol Sci 27, 384–390. Steinhubl, S.R. (2008). Why have antioxidants failed in
Olovnikov, A.M. (1973). A theory of marginotomy. clinical trials? Am J Cardiol 101, 14D-19D.
The incomplete copying of template margin in Szilard, L. (1959). On the nature of the aging process.
enzymic synthesis of polynucleotides and biologi- Proc Natl Acad Sci USA 45, 30–45.
cal significance of the phenomenon. J Theor Biol Tunaru, S., Kero, J., Schaub, A., Wufka, C., Blaukat,
41, 181–190. A., Pfeffer, K., and Offermanns, S. (2003). PUMA-
Opresko, P.L., Fan, J., Danzy, S., Wilson, D.M., 3rd, and G and HM74 are receptors for nicotinic acid
Bohr, V.A. (2005). Oxidative damage in telomeric and mediate its anti-lipolytic effect. Nat Med 9,
DNA disrupts recognition by TRF1 and TRF2. 352–355.
Nucleic Acids Res 33, 1230–1239. Veech, R.L. (2005). Mitochondrial bioenergetics in
Orr, W.C., Radyuk, S.N., and Sohal, R.S. (2013). intact tissues. In Mitochondria in Health and
Involvement of redox state in the aging of Disease, C.D. Berdanier, ed. (New  York:  CRC
Drosophila melanogaster. Antioxid Redox Signal Press, Taylor & Francis), pp. 65–94.
19, 788–803. Veech, R.L. (2006). The determination of the redox
Pacher, P., Beckman, J.S., and Liaudet, L. (2007). Nitric states and phosphorylation potential in living tis-
oxide and peroxynitrite in health and disease. sues and their relationship to metabolic control
Physiol Rev 87, 315–424. of disease phenotypes. Biochem Mol Biol Edu 34,
Pham-Huy, L.A., He, H., and Pham-Huy, C. (2008). 168–179.
Free radicals, antioxidants in disease and health. Veech, R.L. (2014). Ketone ester effects on metabolism
Int J Biomed Sci 4, 89–96. and transcription. J Lipid Res 55, 2004–2008.
Sato, K., Kashiwaya, Y., Keon, C.A., Tsuchiya, N., King, Veech, R.L., Eggleston, L.V., and Krebs, H.A. (1969).
M.T., Radda, G.K., Chance, B., Clarke, K., and The redox state of free nicotinamide-adenine
Veech, R.L. (1995). Insulin, ketone bodies, and dinucleotide phosphate in the cytoplasm of rat
mitochondrial energy transduction. FASEB J 9, liver. Biochem J 115, 609–619.
651–658. Veech, R.L., Lawson, J.W.R., Cornell, N.W., and Krebs,
Schmid, T.E., Dollinger, G., Beisker, W., Hable, V., H.A. (1979). Cytosolic phosphorylation potential.
Greubel, C., Auer, S., Mittag, A., Tarnok, A., Journal of Biological Chemistry 254, 6538–6547.
Friedl, A.A., Molls, M., et al. (2010). Differences in Verdin, E., and Ott, M. (2015). 50  years of protein
the kinetics of gamma-H2AX fluorescence decay acetylation:  from gene regulation to epigenetics,
270

270 section III: Ketogenic Diet in the Laboratory

metabolism and beyond. Nat Rev Mol Cell Biol 16, Winterbourn, C.C. (2008). Reconciling the chemistry
258–264. and biology of reactive oxygen species. Nat Chem
Villet, R.H., and Dalziel, K. (1969). The nature of the Biol 4, 278–286.
carbon dioxide substrate and equilibrium constant Wollenberger, A., Ristau, O., and Schoffa, G. (1960).
of the 6-phosphogluconate dehydrogenase reac- Eine einfache Technik der extrem schnellen
tion. Biochem J 115, 633–638. Abkuhlung grssáerer Gewebestacke. Pflugers
von Zglinicki, T. (2002). Oxidative stress shortens Archiv 270, 399–412.
telomeres. Trends in Biochemical Sciences 27, Yamato, M., Kudo, W., Shiba, T., Yamada,
339–344. K.I., Watanabe, T., and Utsumi, H. (2010).
Williamson, D. H., Lund, P., & Krebs, H. A. (1967). The Determination of reactive oxygen species associ-
redox state of free nicotinamide-adenine dinucleo- ated with the degeneration of dopaminergic neu-
tide in the cytoplasm and mitochondria of rat liver. rons during dopamine metabolism. Free Radic Res
Biochem J, 103(2), 514–527. 44, 249–257.
271

28
Metabolic Seizure Resistance via BAD
and KATP Channels
J UA N R A M Ó N M A R T Í N E Z - F R A N Ç O I S , P H D , N I K A N. D A N I A L , P H D ,
A N D G A RY Y E L L E N, P H D

INTRODUCTION provides a better opportunity to dissect the


The mechanisms of the ketogenic diet are poorly mechanisms involved in metabolic seizure resis-
understood, but it has been used for almost a tance (Giménez-Cassina et al., 2012). This model
century and is arguably the most effective single involves metabolic changes in brain cells medi-
treatment for epilepsy (Hartman et al., 2007; Neal ated by the protein BAD (BCL-2-associated ago-
et al., 2009; Thiele, 2003). This diet, when adhered nist of cell death). Alteration of BAD’s function
to strictly, may reduce seizures in children by up to reduces the capacity of cells to utilize glucose and
50%, with 10%–15% of children becoming seizure- increases the capacity to use ketone bodies. The
free. Unfortunately, maintaining the ketogenic key metabolic change involved in seizure protec-
diet’s strict regimen is challenging. Thus, it would tion appears to be the switch away from glucose
be very valuable to understand the ketogenic diet’s as the preferred energy source toward utilization
mechanisms and harness them to create better epi- of other fuels, rather than elevation of circulating
lepsy therapies. ketone bodies. In addition, BAD alteration leads to
On a ketogenic diet the liver produces the increased activity of metabolically sensitive ATP-
ketone bodies β-hydroxybutyrate and acetoacetate sensitive potassium (KATP) channels in the brain.
from fatty acids, elevating ketone bodies to mil- Modulation of KATP channel activity may also play
limolar concentrations in the blood. Under these an important role in the seizure-protective actions
conditions, ketone bodies provide an alternative of the ketogenic diet.
fuel source to tissues, including the brain, which
otherwise would primarily utilize glucose (DeVivo B A D M O D U L AT E S G L U C O S E
et al., 1978; Mergenthaler et al., 2013; Owen et al., A N D K E TO N E B O DY
1967; Zielke et al., 2009). The remarkable antiepi- M E TA B O L I S M
leptic effect of increased ketone body metabolism BAD is best-known as a pro-apoptotic protein
points to a link between fuel utilization and neu- that is a member of the BCL-2 family of proteins
ronal excitability. (Czabotar et  al., 2014; Danial and Korsmeyer,
However, the molecular mechanisms of this 2004; Moldoveanu et  al., 2014). Besides its role
link are poorly understood (Danial et  al., 2013; in apoptosis, BAD modulates glucose metabo-
Hartman et  al., 2007; Lutas and Yellen, 2013). lism in multiple cell types, including hepatocytes,
Some of the mechanisms that have been proposed pancreatic β-cells, and fibroblasts (Giménez-
include increased adenosine signaling through A1 Cassina and Danial, 2015). The switch between
purinergic receptors (Masino et al., 2011), changes BAD’s apoptotic and metabolic roles is mediated
in gene expression following glycolysis reduction through phosphorylation of its serine 155, located
(Garriga-Canut et  al., 2006), suppression of glu- within an alpha helical segment known as the BH-
tamate release (Juge et  al., 2010), and changes in 3 domain (Danial et al., 2008; Datta et al., 2000).
excitability mediated by lactate dehydrogenase In hepatocytes and pancreatic β-cells, serine 155
activity (Sada et al., 2015). (equivalent to serine 118 in human BAD) phos-
A mouse model with altered metabolism simi- phorylation promotes mitochondrial metabolism
lar to the ketogenic diet produces similar anti- of glucose. In the presence of apoptotic signals,
seizure effects, and this nondietary manipulation dephosphorylated BAD promotes apoptosis.
272

272 section III: Ketogenic Diet in the Laboratory

The reduction in glucose metabolism pro- To corroborate that the effects of Bad disrup-
voked by BAD modification is reminiscent of met- tion in the metabolism of cultured neurons and
abolic changes associated with the ketogenic diet, astrocytes were due to the metabolic, and not
that is, reduced glycolysis and increased ketone the apoptotic role of BAD, mitochondrial res-
body metabolism. Thus, modifying BAD may be pirometry was also performed in BadS155A cells
a productive avenue to study the mechanisms that (Giménez-Cassina et  al., 2012). Indeed, BadS155A
underlie the seizure protective effects of changes neurons and astrocytes also preferentially utilize
in metabolism. Given the systemic, nonspecific β-hydroxybutyrate over glucose, like the BAD
effects of dietary manipulation, the study of BAD knockout. Conversely, BAD phosphorylation on
alteration enables the study of metabolic seizure serine 155 inhibits ketone body utilization. This
resistance mechanisms in a more specific manner confirms that BAD, through its metabolic role,
and without the use of dietary interventions. changes the ability of brain cells to metabolize
different fuels.
M E TA B O L I C C H A N G E S
I N   B A D - A LT E R E D A LT E R AT I O N O F   B A D
P R I M A RY N E U R O N S A N D FUNCTION PRODUCES
ASTROCYTES S E I Z U R E R E S I S TA N C E
Two key mouse models have been used to study Alteration of BAD’s metabolic role by either knock-
the effects of BAD on neurons and astrocytes:  a out of the protein or knockin of a phosphorylation-
Bad knockout and a Bad knockin. The knockin resistant mutant variant leads to altered metabolism
mouse expresses a mutant of BAD that contains without dietary manipulations. This metabolic
a nonphosphorylatable residue, alanine, at posi- switch from brain glucose to ketone body utiliza-
tion 155 (BADS155A). In knockin animals, BAD’s tion also influences excitability and produces sei-
metabolic role is altered while leaving its apop- zure resistance in vivo (Giménez-Cassina et  al.,
totic role intact, as BADS155A cannot be phosphory- 2012). The seizure resistance has been studied in
lated (Danial et al., 2008; Giménez-Cassina et al., two acute chemoconvulsant models of seizure,
2014). Importantly, these mutant mice display nei- involving intraperitoneal injection of kainate (cf.
ther gross neuroanatomical nor neurobehavioral Ben-Ari et al., 1980) or subcutaneous injection of
abnormalities. Therefore, it is unlikely that genetic pentylenetretrazole (PTZ) (cf. Ferraro et al., 1999).
modification in these mouse models substantially When injected with kainate, wild-type mice
impairs normal brain function. display a series of seizures of increasing severity
To elucidate whether BAD has similar effects that peak 1–2 hours after kainate injection and
on metabolism in the brain as it does in other tis- then slowly decay (Giménez-Cassina et  al., 2012;
sues, Giménez-Cassina et  al. (2012) measured Figure  28.1A). Most wild-type mice experience
the mitochondrial oxygen consumption rates status epilepticus with very severe tonic-clonic sei-
in primary cultures of neurons and astrocytes zures, and many die. In contrast, seizure severity
using mitochondrial respirometry. Bad-ablated in Bad−/− mice is much milder than in wild-type
cells exhibited reduced glucose metabolism and mice, and mutant mice rarely go into status epilep-
elevated capacity to metabolize ketone bodies ticus or die (Figures 28.1 and 28.3). Electrographic
compared with wild-type cells. Similar changes in seizures are also substantially milder (Figure 28.3).
brain metabolism are produced by the presence The in vivo seizure protection is due to BAD’s met-
of ketone bodies. In hippocampal brain slices, β- abolic, not apoptotic role, because BadS155A mice
hydroxybutyrate competes with glucose, lactate, were similarly resistant to kainate-induced sei-
and pyruvate for the generation of acetyl-CoA, zures. Thus, disruption of the metabolic function
inhibiting total glycolytic flux upstream of pyruvate of BAD alleviates both forebrain seizure activity
kinase (Valente-Silva et al., 2015). Additionally, in and generalized behavioral seizures during status
humans, glucose flux in the brain is inversely cor- epilepticus.
related with the degree of ketosis, allowing ketone BAD deletion is similarly protective for PTZ-
bodies to partially replace glucose in cerebral induced seizures (Figure  28.1C); PTZ produces
metabolism (Haymond et  al., 1983; Owen et  al., seizures by a different mechanism than kainate, by
1967). These observations cement the idea that inhibiting GABA action rather than by stimulat-
disrupting BAD function may mimic brain meta- ing glutamate receptors (Bough and Eagles, 1999;
bolic changes produced by the ketogenic diet and Ferraro et al., 1999). It remains to be seen whether
is a useful model to study the cellular mechanisms BAD mutations can also be protective in chronic
linking metabolism to excitability. epilepsy models.
273

Chapter 28: Metabolic Seizure Resistance via BAD and KATP Channels 273

(a) Kainate (b) Kainate (c) PTZ


Seizure Severity (Racine Scale) 4 120 *** 120 ***
WT
Bad –/– 100 100
3

Seizure severity (%)

Seizure severity (%)


80 80
2 60 60

40 40
1
20 20
0
0 1 2 3 4 0 0
Time (hours) WT Bad –/– WT Bad –/–

FIGURE 28.1 Bad−/− mice are resistant to kainate- or pentylenetetrazole-induced acute seizures. (A) Raw seizure
scores in Bad−/− 8- to 10-week-old male mice compared with wild-type (WT) mice over a 4-hour period after a single
intraperitoneal injection of kainate (30 mg/kg). (B) Kainate-induced seizure severity calculated as ∑(all scores of a given
mouse)/(time of experiment) for wild-type (WT) or Bad−/− mice. The mean of the seizure severity values from WT mice
was assigned a value of 100%. This value was then used to normalize the severity of the other tested genotypes and/
or conditions within the same scale. WT, n = 42; Bad–/–, n = 13. (C) Integrated seizure severity (calculated as in B) in
Bad−/− and WT mice (n = 16) subjected to a single subcutaneous injection of pentylenetetrazole (PTZ) at 80 mg/kg
monitored over a 70-minute period. Data in A–C are presented as mean ± SEM. *** p < .001; two-tailed Student’s t-test.
(Adapted from Giménez-Cassina et al., 2012)

BAD EFFECTS ON SEIZURE BAD has also been implicated in regulating


ARE UNLIKE EFFECTS synaptic transmission. The BAD-BAX-caspase-
O F   O T H E R B C L - 2 FA M I LY 3 cascade can induce long-term depression in
P ROT E I N S CA1 hippocampal neurons (Jiao and Li, 2011).
Due to their apoptotic roles, certain BCL-2 fam- Additionally, ABT-737, an inhibitor of BCL-2,
ily proteins such as BIM and PUMA have been BCL-XL, and BCL-w survival proteins, slows the
implicated in protection against neuronal loss and recovery of neurotransmission after intense synap-
seizure damage after prolonged periods of status tic activity and decreases hypoxia-triggered dam-
epilepticus (Engel et al., 2010, 2011; Murphy et al., age to synaptic function (Hickman et al., 2008).
2010). In a chronic seizure model (intra-amygdala This effect of ABT-737 could result from BAD
microinjection of kainate), neuronal death in the function, since this compound is likely acting on
hippocampus 24 hours after seizure initiation is downstream signaling partners of BAD. However,
decreased in Bim−/− or Puma−/− mice, demonstrat- ABT-737 also blocks binding of other BH3-only
ing a role for these pro-apototic proteins in cell proteins to BCL-2 and BCL-XL, which involves
death after chronic seizures. In contrast, loss of the same binding pocket used by BAD. Thus,
BIM or PUMA is not seizure protective to seizures disruption of these protein interactions by ABT-
immediately after kainate injection (Engel et  al., 737 is not indicative of specific actions of BAD in
2010; Murphy et  al., 2010). Similarly, knockout synaptic transmission. In any case, these synaptic
of BIM or another BH3-only protein, BID, does changes are unlikely to be part of the anticonvul-
not produce resistance to acute seizures elicited sant action of BAD deletion. For these synaptic
by intraperitoneal kainate injection (Giménez- effects, Bad−/− and BadS155A mice would have oppo-
Cassina et al., 2012). Unlike BAD, neither of these site phenotypes, as these alterations are dependent
proteins affects glucose metabolism (Giménez- on the interaction of nonphosphorylated BAD
Cassina and Danial, 2015). It appears that certain with the downstream BCL-2 proteins; but in fact,
BCL-2 family members may play a role in epilepto- both genotypes produce seizure protection. These
genesis by promoting hippocampal neuron death, observations highlight the varied and complex
but that this is distinct from the role of BAD in roles of BAD in neuronal physiology, and uniquely
altering acute seizure susceptibility, which involves distinguish the switch in fuel preference as a potent
a switch in fuel preference rather than apoptosis. mechanism to produce seizure protection.
274

274 section III: Ketogenic Diet in the Laboratory

T H E M E TA B O L I C S E I Z U R E cells, the intracellular ATP:ADP ratio determines


R E S I S TA N C E O F   B A D − / − KATP channel activity. In basal conditions, KATP
M I C E L I K E LY O C C U R S channels are spontaneously active and maintain
IN THE BRAIN the cell’s resting membrane potential near the
Upon BAD modification, the capacity to metabo- potassium equilibrium potential, that is, hyperpo-
lize glucose is decreased. This recapitulates changes larized. Upon an increase in extracellular glucose
in brain fuel utilization during fasting or on the concentration, and consequently an increase in
ketogenic diet. In contrast, the switch in fuel choice the intracellular ATP concentration, KATP chan-
produced by BAD alteration occurs without any nels are inhibited. This decrease in KATP channel
dietary alterations. As opposed to dietary manip- activity depolarizes the cell and leads to insulin
ulations that affect the whole organism, seizure release. This exemplifies how KATP channels can
protection arising from BAD modification seems link metabolism to cell excitability, but the effect
to specifically originate from metabolic changes is likely not unique to glucosensing cells like the
in the brain. Knockdown of BAD by adenovirus- β-cells.
produced shRNA in the liver, the major source of Two brain regions that are particularly
ketone body production, does not protect against enriched in KATP channels are the hippocampus
kainate-induced seizures, though it does produce and the substantia nigra pars reticulata (SNr)
the same metabolic effects observed in the liver (Dunn-Meynell et al., 1998; Karschin et al., 1997;
of Bad−/− animals (Giménez-Cassina et  al., 2012; Ma et al., 2007; Tanner et al., 2011; Yamada et al.,
Giménez-Cassina et al., 2014). 2001; Zawar et al., 1999). KATP channels present in
Interestingly, circulating levels of ketone bod- these two areas are octamers comprising two types
ies are similar in Bad−/− and wild-type mice, but of subunits:  four Kir6.2 pore-forming subunits
β-hydroxybutyrate levels are increased in whole- and four SUR1 regulatory subunits. Both of these
brain tissue derived from Bad−/−. This suggests brain regions are involved in the generation of
that in the knockout animals, ketogenesis is being seizure activity (Depaulis et al.,1994; Heinemann
increased specifically in the brain; it also argues et  al., 1992; Hsu, 2007; Iadarola and Gale, 1982;
that changes in brain metabolism upon BAD Krook-Magnuson et  al., 2015; Lothman et  al.,
alteration are unlikely to be generated by systemic 1992; McNamara et  al., 1984), and excitability of
modifications in metabolism. The precise changes neurons in these areas can be modulated by KATP
in metabolism responsible for BAD’s seizure- channels (Lutas et  al., 2014; Stanford and Lacey,
protective effect remain to be established. Given 1996; Tanner et  al., 2011; Yamada et  al., 2001;
that the ketogenic diet’s antiseizure effect can be Zawar et al., 1999).
reversed in patients promptly after carbohydrate Neuronal KATP channel activity appears gen-
consumption (Huttenlocher, 1976; Pfeifer et  al., erally to be low, implying that the intracellular
2008), it seems likely that the transition in fuel ATP concentration in neurons is often sufficiently
type utilization is the crucial factor. Metabolomic high to inhibit these channels (Giménez-Cassina
studies comparing wild-type to Bad-altered geno- et al., 2012; Haller et al., 2001; Pelletier et al., 2000;
types could shed light on the metabolic pathways Schwanstecher and Panten, 1993; Tanner et  al.,
implicated in BAD’s effect. 2011; Yamada et al., 2001). Substantial metabolic
insults to the brain are known to be capable of
K AT P C H A N N E L S activating the channels; for instance in brain isch-
L I N K M E TA B O L I S M emia, intracellular ATP and ATP/ADP markedly
W I T H   E X C I TA B I L I T Y decrease after ~2 minutes (Katsura et al., 1992). In
One likely candidate for linking altered brain the CA1 area of the hippocampus, a brain region
metabolism with altered excitability, in the keto- particularly susceptible to ischemic insult (Davolio
genic diet and in BAD-altered animals, is the and Greenamyre, 1995; Schmidt-Kastner and
ATP-sensitive potassium (KATP) channel. KATP Freund, 1991; Smith et al., 1984), hypoxia triggers
channels are inhibited by intracellular ATP, neuronal hyperpolarization by activating potas-
activated by intracellular ADP, and are widely sium conductances, including KATP channel cur-
expressed throughout the body, including the rents (Yamada and Inagaki, 2005; Yamada et  al.,
brain (Liss and Roeper, 2001; Nichols, 2006; Proks 2001). Consistent with this, KATP channel inhibi-
and Ashcroft, 2009). Their role linking metabo- tors, such as the sulfonylurea drugs glibenclamide
lism with cell excitability has been best studied or tolbutamide, can suppress the hypoxia-induced
in control of insulin release by pancreatic β-cells hyperpolarization. Furthermore, mice lacking
(Ashcroft, 2005; Ashcroft et al., 1984). In these either Kir6.2 or SUR1 exhibit a markedly reduced
275

Chapter 28: Metabolic Seizure Resistance via BAD and KATP Channels 275

threshold for generalized seizures upon a hypoxic increased in Bad−/− mouse DGNs, from ~0.5% in
insult, that is, they are hypersensitive to hypoxia- wild-type to ~20% in Bad−/− (Giménez-Cassina
induced seizures (Hernández-Sánchez et al., 2001; et  al., 2012). Confirming this increase in basal
Yamada et al., 2001). Interestingly, similar to Bad- KATP channel activity in DGNs with disrupted
altered mice, mice overexpressing SUR1 display a BAD function, whole-cell KATP current is also
decrease in kainate-induced seizure susceptibility. inhibited by dialyzing the intracellular medium
These observations indicate that KATP channels of either Bad−/− or BadS155A DGNs with high ATP
play a crucial role in mediating seizure protec- (4 mM)—a phenomenon denominated “wash-
tive effects provoked by metabolic insufficiency or down” (Figure 28.2A). On the other hand, wash-
hypoxia. down does not occur in wild-type DGNs (as KATP
Besides ischemic/hypoxic insult, other meta- channels are mostly closed in this case) or in
bolic changes in brain regions implicated in sei- DGNs lacking both BAD and Kir6.2.
zure activity can also modulate KATP channel These whole-cell results corroborate the
activity. Poisoning of GABAergic neurons in the fact that basal KATP channel open probability is
SNr or hippocampal CA1 neurons with cyanide increased in Bad−/− DGNs. Additionally, the num-
activates KATP channels (Matsumoto et  al., 2002; ber of functional KATP channels in DGNs is not
Schwanstecher and Panten, 1993). In addition, in affected by altering BAD’s metabolic role. The
dentate granule neurons (DGNs) of the hippo- maximal KATP conductance, observed in whole-
campus, a much more subtle energetic challenge cell recordings dialyzing the neuron’s intracellu-
can lead to KATP channel opening: neuronal firing lar medium with a low concentration of ATP (0.3
elicited by antidromic stimulation increases single mM), is unchanged between wild-type and Bad−/−
KATP channel activity (Tanner et al., 2011). Firing or BadS155A DGNs.
of action potentials produces metabolic changes The increased KATP channel activity observed
by activating the pumps, for example the Na/ in Bad-altered DGNs suggests that KATP channels
K ATPase, that maintain intracellular ionic con- might lead to decreased neuronal excitability. Even
centrations (Ivannikov et  al., 2010; Mercer and though further studies are needed to demonstrate
Dunham, 1981). Activation of these pumps trig- such a scenario, consistent with this idea, studies
gers ATP consumption, leading to an opening of using a hippocampal neuron cell line have shown
KATP channels (Haller et  al., 2001; Tanner et  al., that KATP channels can mediate changes in hip-
2011). It has also been shown that KATP channel pocampal neuron excitability (Huang et al., 2006,
open probability in DGNs increases by extracel- 2007). Thus, a decrease in excitability mediated by
lular application of β-hydroxybutyrate (Tanner KATP channels could be a central mechanism in the
et  al., 2011). These observations are consistent ketogenic diet’s seizure protective effects.
with the idea that KATP channels could mediate
the anticonvulsant effect of the ketogenic diet. K AT P C H A N N E L S M E D I AT E
First, KATP channels might open in conditions of S E I Z U R E P ROT E C T I O N
hyperexcitability, such as during a seizure. Second,
ELICITED BY BAD
elevated levels of circulating ketone bodies could
also increase KATP channel activity. These effects D I S RU P T I O N
are synergistic, and the resulting increase in KATP Lack of BAD leads to a remarkable increase in KATP
channel current would reduce excitability and pro- channel activity and also produces seizure protec-
duce seizure resistance. tion. It appears that these two effects are causally
related:  eliminating Kir6.2 (Kcnj11) in a Bad−/−
background nearly abolishes the seizure resistance
BA D D I S RU P T I O N produced by Bad ablation (Figure  28.2B). This
I N C R E A S E S K AT P C H A N N E L substantial attenuation of seizure resistance is not
ACTIVITY due to an increase in seizure severity produced by
As previously stated, KATP channels link metabo- lack of Kir6.2; deletion of Kir6.2 alone does not
lism to excitability and are activated by both increase mouse seizure sensitivity compared with
ketone bodies and electrical activity in the brain. wild-type animals. These results constitute genetic
This raises the question of whether these channels evidence that KATP channels are necessary in medi-
mediate the seizure resistance elicited by BAD dis- ating BAD’s effect on seizure protection.
ruption. In single-channel, cell-attached experi- KATP channels are well-known mediators of
ments in DGNs in hippocampal brain slices, the changes in metabolism with neuronal excitability.
basal KATP channel open probability is dramatically Indeed, either glucose deprivation or ketone body
276

276 section III: Ketogenic Diet in the Laboratory

(a) (b) n.s.


***
WT 120
2.0 *** **
Bad –/–
Normalized Conductance to G3min

BadS155A 100
Bad –/–; Kir6.2 –/–

Seizure Severity (%)


1.5
80

1.0 60

40
0
20

0 0
0 1 2 3 WT Bad –/– Bad –/–; Kir6.2 –/–
Time from break-in (min) Kir6.2 –/–

FIGURE 28.2 KATP channels mediate effects of BAD alteration. (A) Whole-cell conductance recorded from dentate
granule neurons (DGNs) in acute hippocampal slices showing “washdown” of an initially high KATP channel conductance
with high ATP (4 mM) in the recording pipette. Washdown is observed in Bad–/– and BadS155A DGNs but not in wild-type
(WT) or Bad–/–;Kir6.2–/– double-mutant DGNs. The time course of slope conductance during whole-cell recording was
normalized to the value 3 min after break-in. (B) Integrated seizure severity in Bad–/– or Kir6.2–/– single mutants, or Bad–/–;
Kir6.2–/– double mutant mice compared with WT mice after a single intraperitoneal injection of kainate monitored and
analyzed as in Figure 28.1. Data are presented as mean ± SEM. ** p < .01; *** p < .001; n.s., nonsignificant; two-tailed
Student’s t-test. (Adapted from Giménez-Cassina et al., 2012).

metabolism can increase KATP channel activity. possibility is that glucose metabolism is special
Furthermore, these effects can be potentiated by in its ability to respond rapidly to acute energy
neuronal firing, for example, during seizure activity. challenges, as might occur in the elevated brain
The mechanism by which a metabolic fuel switch activity leading to seizures. Thus, BAD alteration,
could increase KATP channel activity is unknown. by switching metabolism away from glucose con-
Given that ATP and ADP directly control the open sumption, may produce a decrease in the ATP con-
probability of KATP channels, changes in ATP/ADP centration at the plasma membrane, which then
local to KATP channels might be different in the pres- could trigger KATP channel opening and reduced
ence or absence of BAD’s metabolic effects. neuronal excitability.
Why would ATP/ADP change in the presence Even though modulation of KATP channel activ-
of abundant alternative fuels? One possibility is ity by ATP/ADP is a straightforward explanation,
that KATP channel activity is controlled by local KATP channel regulation is complex, and other
glycolysis. Glycolysis is bypassed and inhibited processes, such as PIP2 binding or phosphoryla-
by mitochondrial metabolism of ketone bod- tion, can also alter KATP channel activity (Nichols,
ies (DeVivo et  al.,1978; Haymond et  al., 1983; 2006; Proks and Ashcroft, 2009). In addition,
Valente-Silva et al., 2015). This metabolic change β-hydroxybutyrate levels are increased in Bad−/−
would increase ATP produced by mitochondria compared with wild-type brains, and, as men-
and decrease ATP produced by glycolysis. tioned above, this can induce KATP channel open-
Indeed, KATP channels and glycolytic enzymes ing. While it is unlikely that the effects of ketone
can be found associated in large complexes in bodies on KATP channel activity explain the total-
the plasma membrane (Dhar-Chowdhury et  al., ity of the observed KATP channel activation, these
2007; Dubinsky et  al., 1998; Hong et  al., 2011). metabolites may constitute part of multiple actions
Evidence suggests that these glycolytic enzyme needed to exert the full effect observed upon BAD
complexes may produce ATP compartmentation alteration.
in the cell (Chu et al., 2012; Hoffman et al., 2009; How does KATP channel activation in certain
Proverbio and Hoffman, 1977). Furthermore, gly- brain cells elicit seizure protection? A  simple
colysis inhibition can trigger the opening of KATP explanation is that elevated activity, which would
channels (Matsumoto et al., 2002; Schwanstecher ordinarily lead to a seizure in a susceptible indi-
and Panten, 1993; Tantama et al., 2013). Another vidual, produces increased KATP channel current,
277

Chapter 28: Metabolic Seizure Resistance via BAD and KATP Channels 277

Glucose
BAD deletion produces BAD
altered metabolism Alternative fuels

WT Bad –/–

Increased KATP channel


activity in dentate
granule neurons

2 pA
0.5 s

Bad –/– 0.5 *

High Energy Spiking State


0.4

Fraction of Time in
KA inj KA inj

0.3

Seizure protection 0.2

0.1

500 V 0
WT Bad –/–
1 min

FIGURE 28.3 Summary of effects through BAD and KATP channels that lead to seizure resistance. Ablation of Bad
leads to decreased glucose metabolism and increased consumption of alternative fuels. This metabolic switch increases
KATP channel activity, as seen in cell-attached recordings of KATP channels in dentate granule neurons in hippocampal
brain slices. Compared with wild-type (WT) mice, Bad–/– mice are resistant to acute kainate-induced seizures exhibiting
reduced cortical seizure activity recorded by electroencephalogram (EEG) as shown by representative EEG traces of
approximately1 hour for WT or Bad–/– mice (bottom left and middle panels), and reduced fraction of time spent in high
energy spiking state (bottom right panel). WT, n = 17; Bad–/–, n = 20; mean ± SEM, * p < .05 by two-tailed Student’s t-test.
(Adapted from Giménez-Cassina et al., 2012).

which then reduces subsequent neuronal activity CONCLUSION


by hyperpolarizing neurons. It will be important In summary (Figure 28.3), certain manipula-
to reveal the relevant brain regions associated with tions of BAD’s metabolic role produce a meta-
this seizure protective effect. KATP channels are bolic change in brain cells, switching away from
present in two brain regions associated with sei- glucose utilization toward consumption of alter-
zure activity: the DGNs in the hippocampus and native fuels. The metabolic changes triggered
the GABAergic neurons of the SNr. It has been by BAD modification are reminiscent of those
shown that reducing neuronal firing in either elicited by a ketogenic diet. Remarkably, BAD
of these two brain areas inhibits seizure activity disruption produces robust seizure protection
(Akman et  al., 2015; Coulter and Carlson, 2007; in the absence of dietary treatments. At the cel-
Krook-Magnuson et  al., 2013; Paz et  al., 2007; lular level, it appears that KATP channels mediate
Vercueil et  al., 1998). Studies on KATP channel BAD’s anticonvulsant effect. Disruption of BAD
activity or in vivo seizure models in conditional function increases KATP channel activity in neu-
Kir6.2 (Kcnj11) knockout lines could be used to rons in the dentate gyrus, a brain region that is
define the brain regions and brain cell types impli- likely a gate for seizure activity. The mechanisms
cated in seizure resistance. by which BAD modulates brain metabolism and
278

278 section III: Ketogenic Diet in the Laboratory

how these metabolic changes affect excitability work? Four potential mechanisms. J Child Neurol
have just started to be revealed. These seizure- 28, 1027–1033.
protective mechanisms will hopefully serve as Datta, S.R., Katsov, A., Hu, L., Petros, A., Fesik, S.W.,
the basis for the development of more effective Yaffe, M.B., and Greenberg, M.E. (2000). 14-3-3
therapies for epilepsy. proteins and survival kinases cooperate to inacti-
vate BAD by BH3 domain phosphorylation. Mol
Cell 6, 41–51.
AC K N OW L E D G M E N T S
Davolio, C., and Greenamyre, J.T. (1995). Selective
We thank members of the Yellen lab for critical vulnerability of the CA1 region of hippocampus
review and discussion of our manuscript. This to the indirect excitotoxic effects of malonic acid.
work was supported by grants from the National Neurosci Lett 192, 29–32.
Institutes of Health (R01 NS083844 to G.Y.  and Depaulis, A., Vergnes, M., and Marescaux, C. (1994).
N.N.D. and R01 NS055031 to G.Y.) Endogenous control of epilepsy: the nigral inhibi-
tory system. Prog Neurobiol 42, 33–52.
REFERENCES DeVivo, D.C., Leckie, M.P., Ferrendelli, J.S., and
Akman, O., Gulcebi, M.I., Carcak, N., Ketenci Ozatman, McDougal, D.B. (1978). Chronic ketosis and cere-
S., Eryigit, T., Moshé, S.L., Galanopoulou, A.S., bral metabolism. Ann Neurol 3, 331–337.
and Yilmaz Onat, F. (2015). The role of the sub- Dhar-Chowdhury, P., Malester, B., Rajacic, P., and
stantia nigra pars reticulata in kindling resistance Coetzee, W.A. (2007). The regulation of ion chan-
in rats with genetic absence epilepsy. Epilepsia 56, nels and transporters by glycolytically derived
1793–1802. ATP. Cell Mol Life Sci 64, 3069–3083.
Ashcroft, F.M. (2005). ATP-sensitive potassium chan- Dubinsky, W.P., Mayorga-Wark, O., and Schultz,
nelopathies:  focus on insulin secretion. J Clin S.G. (1998). Colocalization of glycolytic enzyme
Invest 115, 2047–2058. activity and KATP channels in basolateral mem-
Ashcroft, F.M., Harrison, D.E., and Ashcroft, S.J. brane of Necturus enterocytes. Am J Physiol 275,
(1984). Glucose induces closure of single potas- C1653–C1659.
sium channels in isolated rat pancreatic beta-cells. Dunn-Meynell, A.A., Rawson, N.E., and Levin, B.E.
Nature 312, 446–448. (1998). Distribution and phenotype of neurons
Ben-Ari, Y., Tremblay, E., Ottersen, O.P., and Meldrum, containing the ATP-sensitive K+ channel in rat
B.S. (1980). The role of epileptic activity in hippo- brain. Brain Res 814, 41–54.
campal and “remote” cerebral lesions induced by Engel, T., Hatazaki, S., Tanaka, K., Prehn, J.H.M., and
kainic acid. Brain Res 191, 79–97. Henshall, D.C. (2010). Deletion of Puma protects
Bough, K.J., and Eagles, D.A. (1999). A ketogenic diet hippocampal neurons in a model of severe status
increases the resistance to pentylenetetrazole- epilepticus. Neuroscience 168, 443–450.
induced seizures in the rat. Epilepsia 40, Engel, T., Plesnila, N., Prehn, J.H.M., and Henshall,
138–143. D.C. (2011). In vivo contributions of BH3-only
Chu, H., Puchulu-Campanella, E., Galan, J.A., proteins to neuronal death following seizures,
Tao, W.A., Low, P.S., and Hoffman, J.F. (2012). ischemia, and traumatic brain injury. J Cereb
Identification of cytoskeletal elements enclos- Blood Flow Metab 31, 1196–1210.
ing the ATP pools that fuel human red blood cell Ferraro, T.N., Golden, G.T., Smith, G.G., St Jean, P.,
membrane cation pumps. Proc Natl Acad Sci USA Schork, N.J., Mulholland, N., Ballas, C., Schill, J.,
109, 12794–12799. Buono, R.J., and Berrettini, W.H. (1999). Mapping
Coulter, D.A., and Carlson, G.C. (2007). Functional loci for pentylenetetrazol-induced seizure suscep-
regulation of the dentate gyrus by GABA-mediated tibility in mice. J Neurosci 19, 6733–6739.
inhibition. Prog Brain Res 163, 235–243. Garriga-Canut, M., Schoenike, B., Qazi, R.,
Czabotar, P.E., Lessene, G., Strasser, A., and Adams, Bergendahl, K., Daley, T.J., Pfender, R.M.,
J.M. (2014). Control of apoptosis by the BCL-2 Morrison, J.F., Ockuly, J., Stafstrom, C., Sutula, T.,
protein family:  implications for physiology and et al. (2006). 2-Deoxy-D-glucose reduces epilepsy
therapy. Nat Rev Mol Cell Biol 15, 49–63. progression by NRSF-CtBP-dependent metabolic
Danial, N.N., and Korsmeyer, S.J. (2004). Cell regulation of chromatin structure. Nat Neurosci 9,
death: critical control points. Cell 116, 205–219. 1382–1387.
Danial, N.N., Walensky, L.D., Zhang, C.-Y., Choi, Giménez-Cassina, A., and Danial, N.N. (2015).
C.S., Fisher, J.K., Molina, A.J.A., Datta, S.R., Regulation of mitochondrial nutrient and energy
Pitter, K.L., Bird, G.H., Wikstrom, J.D., et  al. metabolism by BCL-2 family proteins. Trends
(2008). Dual role of proapoptotic BAD in insu- Endocrinol Metab 26, 165–175.
lin secretion and beta cell survival. Nat Med 14, Giménez-Cassina, A., Martínez-François, J.R., Fisher,
144–153. J.K., Szlyk, B., Polak, K., Wiwczar, J., Tanner, G.R.,
Danial, N.N., Hartman, A.L., Stafstrom, C.E., and Lutas, A., Yellen, G., and Danial, N.N. (2012).
Thio, L.L. (2013). How does the ketogenic diet BAD- dependent regulation of fuel metabolism
279

Chapter 28: Metabolic Seizure Resistance via BAD and KATP Channels 279

and KATP channel activity confers resistance to epi- by gamma-aminobutyric acid. Science 218,
leptic seizures. Neuron 74, 719–730. 1237–1240.
Giménez-Cassina, A., Garcia-Haro, L., Choi, C.S., Ivannikov, M.V., Sugimori, M., and Llinás, R.R. (2010).
Osundiji, M.A., Lane, E.A., Huang, H., Yildirim, Calcium clearance and its energy requirements in
M.A., Szlyk, B., Fisher, J.K., Polak, K., et al. (2014). cerebellar neurons. Cell Calcium 47, 507–513.
Regulation of hepatic energy metabolism and glu- Jiao, S., and Li, Z. (2011). Nonapoptotic function of
coneogenesis by BAD. Cell Metab 19, 272–284. BAD and BAX in long-term depression of synaptic
Haller, M., Mironov, S.L., Karschin, A., and Richter, transmission. Neuron 70, 758–772.
D.W. (2001). Dynamic activation of KATP chan- Juge, N., Gray, J.A., Omote, H., Miyaji, T., Inoue, T.,
nels in rhythmically active neurons. J Physiol 537, Hara, C., Uneyama, H., Edwards, R.H., Nicoll,
69–81. R.A., and Moriyama, Y. (2010). Metabolic con-
Hartman, A.L., Gasior, M., Vining, E.P.G., and trol of vesicular glutamate transport and release.
Rogawski, M.A. (2007). The neuropharmacology Neuron 68, 99–112.
of the ketogenic diet. Pediatr Neurol 36, 281–292. Karschin, C., Ecke, C., Ashcroft, F.M., and Karschin,
Haymond, M.W., Howard, C., Ben-Galim, E., and A. (1997). Overlapping distribution of KATP chan-
DeVivo, D.C. (1983). Effects of ketosis on glucose nel-forming Kir6.2 subunit and the sulfonylurea
flux in children and adults. Am J Physiol 245, receptor SUR1 in rodent brain. FEBS Lett 401,
E373–E378. 59–64.
Heinemann, U., Beck, H., Dreier, J.P., Ficker, E., Stabel, Katsura, K., Minamisawa, H., Ekholm, A., Folbergrová,
J., and Zhang, C.L. (1992). The dentate gyrus as a J., and Siesjö, B.K. (1992). Changes of labile metab-
regulated gate for the propagation of epileptiform olites during anoxia in moderately hypo- and
activity. Epilepsy Res (Suppl 7), 273–280. hyperthermic rats: correlation to membrane fluxes
Hernández-Sánchez, C., Basile, A.S., Fedorova, I., of K+. Brain Res 590, 6–12.
Arima, H., Stannard, B., Fernandez, A.M., Ito, Y., Krook-Magnuson, E., Armstrong, C., Oijala, M., and
and LeRoith, D. (2001). Mice transgenically over- Soltesz, I. (2013). On-demand optogenetic control
expressing sulfonylurea receptor 1 in forebrain of spontaneous seizures in temporal lobe epilepsy.
resist seizure induction and excitotoxic neuron Nat Commun 4, 1376.
death. Proc Natl Acad Sci USA 98, 3549–3554. Krook-Magnuson, E., Armstrong, C., Bui, A., Lew, S.,
Hickman, J.A., Hardwick, J.M., Kaczmarek, L.K., and Oijala, M., and Soltesz, I. (2015). In vivo evalua-
Jonas, E.A. (2008). Bcl-xL inhibitor ABT-737 tion of the dentate gate theory in epilepsy. J Physiol
reveals a dual role for Bcl-xL in synaptic transmis- 593, 2379–2388.
sion. J Neurophysiol 99, 1515–1522. Liss, B., and Roeper, J. (2001). Molecular physiology
Hoffman, J.F., Dodson, A., and Proverbio, F. (2009). On of neuronal KATP channels. Mol Membr Biol 18,
the functional use of the membrane compartmen- 117–127.
talized pool of ATP by the Na+ and Ca++ pumps in Lothman, E.W., Stringer, J.L., and Bertram, E.H.
human red blood cell ghosts. J Gen Physiol 134, (1992). The dentate gyrus as a control point for
351–361. seizures in the hippocampus and beyond. Epilepsy
Hong, M., Kefaloyianni, E., Bao, L., Malester, B., Res (Suppl 7), 301–313.
Delaroche, D., Neubert, T.A., and Coetzee, W.A. Lutas, A., and Yellen, G. (2013). The ketogenic
(2011). Cardiac ATP-sensitive K+ channel associ- diet: metabolic influences on brain excitability and
ates with the glycolytic enzyme complex. FASEB J epilepsy. Trends Neurosci 36, 32–40.
25, 2456–2467. Lutas, A., Birnbaumer, L., and Yellen, G. (2014).
Hsu, D. (2007). The dentate gyrus as a filter or gate: a Metabolism regulates the spontaneous firing of
look back and a look ahead. Prog Brain Res 163, substantia nigra pars reticulata neurons via KATP
601–613. and nonselective cation channels. J Neurosci 34,
Huang, C.-W., Huang, C.-C., and Wu, S.-N. (2006). 16336–16347.
The opening effect of pregabalin on ATP-sensitive Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic
potassium channels in differentiated hippocampal diet metabolites reduce firing in central neu-
neuron–derived H19-7 Cells. Epilepsia 47, 720–72 rons by opening KATP channels. J Neurosci 27,
Huang, C.-W., Huang, C.-C., Cheng, J.-T., Tsai, J.-J., 3618–3625.
and Wu, S.-N. (2007). Glucose and hippocampal Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin,
neuronal excitability: Role of ATP-sensitive potas- D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and
sium channels. J Neurosci Res 85, 1468–1477. Boison, D. (2011). A ketogenic diet suppresses sei-
Huttenlocher, P.R. (1976). Ketonemia and sei- zures in mice through adenosine A1 receptors. J
zures: metabolic and anticonvulsant effects of two Clin Invest 121, 2679–2683.
ketogenic diets in childhood epilepsy. Pediatr Res Matsumoto, N., Komiyama, S., and Akaike, N. (2002).
10, 536–540. Pre- and postsynaptic ATP- sensitive potassium
Iadarola, M.J., and Gale, K. (1982). Substantia channels during metabolic inhibition of rat hippo-
nigra:  site of anticonvulsant activity mediated campal CA1 neurons. J Physiol 541, 511–520.
280

280 section III: Ketogenic Diet in the Laboratory

McNamara, J.O., Galloway, M.T., Rigsbee, L.C., and enzymes with a stiripentol analog to treat epilepsy.
Shin, C. (1984). Evidence implicating substantia Science 347, 1362–1367.
nigra in regulation of kindled seizure threshold. J Schmidt-Kastner, R., and Freund, T.F. (1991). Selective
Neurosci 4, 2410–2417. vulnerability of the hippocampus in brain isch-
Mercer, R.W., and Dunham, P.B. (1981). Membrane- emia. Neuroscience 40, 599–636.
bound ATP fuels the Na/K pump. Studies on Schwanstecher, C., and Panten, U. (1993). Tolbutamide-
membrane-bound glycolytic enzymes on inside- and diazoxide-sensitive K+ channel in neurons
out vesicles from human red cell membranes. J of substantia nigra pars reticulata. Naunyn.
Gen Physiol. 78, 547–568. Schmiedebergs Arch Pharmacol 348, 113–117.
Mergenthaler, P., Lindauer, U., Dienel, G.A., and Smith, M.L., Auer, R.N., and Siesjö, B.K. (1984). The
Meisel, A. (2013). Sugar for the brain: the role of density and distribution of ischemic brain injury
glucose in physiological and pathological brain in the rat following 2-10 min of forebrain isch-
function. Trends Neurosci 36, 587–597. emia. Acta Neuropathol (Berl) 64, 319–332.
Moldoveanu, T., Follis, A.V., Kriwacki, R.W., and Stanford, I.M., and Lacey, M.G. (1996).
Green, D.R. (2014). Many players in BCL-2 family Electrophysiological investigation of adenosine
affairs. Trends Biochem Sci 39, 101–111. trisphosphate-sensitive potassium channels in the
Murphy, B.M., Engel, T., Paucard, A., Hatazaki, S., rat substantia nigra pars reticulata. Neuroscience
Mouri, G., Tanaka, K., Tuffy, L.P., Jimenez-Mateos, 74, 499–509.
E.M., Woods, I., Dunleavy, M., et  al. (2010). Tanner, G.R., Lutas, A., Martínez-François, J.R.,
Contrasting patterns of Bim induction and neuro- and Yellen, G. (2011). Single KATP channel open-
protection in Bim-deficient mice between hippo- ing in response to action potential firing in
campus and neocortex after status epilepticus. Cell mouse dentate granule neurons. J Neurosci 31,
Death Differ 17, 459–468. 8689–8696.
Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S., Tantama, M., Martínez-François, J.R., Mongeon, R.,
Edwards, N., Fitzsimmons, G., Whitney, A., and and Yellen, G. (2013). Imaging energy status in live
Cross, J.H. (2009). A randomized trial of classical cells with a fluorescent biosensor of the intracellu-
and medium-chain triglyceride ketogenic diets in lar ATP-to-ADP ratio. Nat Commun 4, 2550.
the treatment of childhood epilepsy. Epilepsia 50, Thiele E.A. (2003). Assessing the efficacy of antiepi-
1109–1117. leptic treatments: the ketogenic diet. Epilepsia 44
Nichols, C.G. (2006). KATP channels as molecular sen- (Suppl 7), 26–29.
sors of cellular metabolism. Nature 440, 470–476. Valente-Silva, P., Lemos, C., Köfalvi, A., Cunha,
Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan, J.M., R.A., and Jones, J.G. (2015). Ketone bodies effec-
Herrera, M.G., and Cahill, G.F. (1967). Brain metab- tively compete with glucose for neuronal acetyl-
olism during fasting. J Clin Invest 46, 1589–1595. CoA generation in rat hippocampal slices. NMR
Paz, J.T., Chavez, M., Saillet, S., Deniau, J.M., and Biomed 28, 1111–1116.
Charpier, S. (2007). Activity of ventral medial tha- Vercueil, L., Benazzouz, A., Deransart, C., Bressand,
lamic neurons during absence seizures and modu- K., Marescaux, C., Depaulis, A., and Benabid, A.L.
lation of cortical paroxysms by the nigrothalamic (1998). High-frequency stimulation of the subtha-
pathway. J Neurosci 27, 929–941. lamic nucleus suppresses absence seizures in the
Pelletier, M.R., Pahapill, P.A., Pennefather, P.S., and rat: comparison with neurotoxic lesions. Epilepsy
Carlen, P.L. (2000). Analysis of single KATP chan- Res 31, 39–46.
nels in mammalian dentate gyrus granule cells. J Yamada, K., and Inagaki, N. (2005). Neuroprotection
Neurophysiol 84, 2291–2301. by KATP channels. J Mol Cell Cardiol 38, 945–949.
Pfeifer, H.H., Lyczkowski, D.A., and Thiele, E.A. Yamada, K., Ji, J.J., Yuan, H., Miki, T., Sato, S.,
(2008). Low glycemic index treatment: implemen- Horimoto, N., Shimizu, T., Seino, S., and Inagaki,
tation and new insights into efficacy. Epilepsia 49 N. (2001). Protective role of ATP-sensitive potas-
(Suppl 8), 42–45. sium channels in hypoxia- induced generalized
Proks, P., and Ashcroft, F.M. (2009). Modeling KATP seizure. Science 292, 1543–1546.
channel gating and its regulation. Prog Biophys Zawar, C., Plant, T.D., Schirra, C., Konnerth, A., and
Mol Biol 99, 7–19. Neumcke, B. (1999). Cell-type specific expression
Proverbio, F., and Hoffman, J.F. (1977). Membrane of ATP-sensitive potassium channels in the rat
compartmentalized ATP and its preferential use by hippocampus. J Physiol 514 ( Pt 2), 327–341.
the Na,K-ATPase of human red cell ghosts. J Gen Zielke, H.R., Zielke, C.L., and Baab, P.J. (2009). Direct
Physiol 69, 605–632. measurement of oxidative metabolism in the living
Sada, N., Lee, S., Katsu, T., Otsuki, T., and Inoue, brain by microdialysis: a review. J Neurochem 109
T. (2015). Epilepsy treatment. Targeting LDH (Suppl 1), 24–29.
281

29
Lactate Dehydrogenase
A Novel Metabolic Target

N AG I S A S A D A , P H D A N D T S U Y O S H I I N O U E , P H D

T H E K E TO G E N I C D I E T the molecular mechanisms underlying the antiepi-


F O R   D R U G - R E S I S TA N T leptic effects of the ketogenic diet.
EPILEPSY
Epilepsy is a neurological disorder that is charac- MECHANISMS RESPONSIBLE
terized by the hyperexcitation of electrical activi- FOR THE ANTIEPILEPTIC
ties in the brain. Approximately 1% of the world’s EFFECTS OF THE
population has epilepsy, and one-third of epileptic K E TO G E N I C   D I E T
patients are resistant to currently available antiepi- The ketogenic diet mainly elicits two metabolic
leptic drugs (Kwan and Brodie, 2000). Therefore, changes in the body; increases in ketone bodies
it is important to develop new antiepileptic drugs and decreases in glucose (Bough et al., 2006). The
that can treat the drug-resistant epilepsy. As a step neural inhibition and antiepileptic effects elicited
toward achieving this goal, “endogenous proteins” by these two metabolic changes have been actively
that have the ability to suppress epileptic seizures studied (Lutas and Yellen, 2013). The following
need to be identified, and will become target mol- rodent studies have demonstrated the antiepileptic
ecules for the development of antiepileptic drugs. effects of ketone bodies. Ketone bodies consist of
“Chemical compounds” that act on these target acetoacetate, β-hydroxybutyrate, and acetone. The
molecules and suppress epileptic seizures must ketogenic diet was previously shown to increase
then be identified, and will become prototypes β-hydroxybutyrate levels up to ~8 mM in the
of new antiepileptic drugs. Thus, the search for blood plasma of rodents (Bough et al., 1999). An
new target molecules in basic neuroscience will intraperitoneal injection of ketone bodies directly
ultimately lead to drug development for drug- suppresses seizures in vivo in audiogenic seizure-
resistant epilepsy. susceptible mice (Rho et al., 2002). At the molecu-
Since epileptic seizures are caused by the lar and cellular levels, ketone bodies decrease the
hyperexcitation of electrical activities in the brain, firing rate of neurons in the substantia nigra via
antiepileptic drugs also need to be designed in adenosine 5’-triphosphate (ATP)-sensitive K+
order to suppress the neuronal hyperexcitation. channels (KATP channels) (Ma et al., 2007). Single
Electrical activities in the brain are generated by channel recordings have revealed that ketone
ion channels, synaptic receptors, and neurotrans- bodies open KATP channels in the dentate granule
mitter transporters. Therefore, currently avail- cells of the hippocampus (Tanner et  al., 2011).
able antiepileptic drugs have been designed to act Acetoacetate is also known to be an inhibitor of
on the molecules generating electrical currents vesicular glutamate transporters (VGLUTs) (Juge
(Meldrum and Rogawski, 2007). However, these et al., 2010). The role of the VGLUTs is to fill gluta-
antiepileptic drugs are ineffective for one-third of mate into synaptic vesicles and regulate excitatory
epileptic patients (Kwan and Brodie, 2000); that synaptic transmission in the brain, and thus, ace-
is, other molecular targets need to be identified. toacetate reduces miniature excitatory postsyn-
The ketogenic diet is known to be effective for aptic currents (miniature EPSCs) and suppresses
some patients with drug-resistant epilepsy (Neal 4-aminopyridine-induced acute seizures in vivo
et  al., 2008), but there are no antiepileptic drugs (Juge et al., 2010).
that mimic the ketogenic diet. Based on this back- In terms of decreases in glucose, seizures are
ground, neuroscientists have recently focused on suppressed by 2-deoxy-D-glucose, a glycolytic
282

282 section III: Ketogenic Diet in the Laboratory

inhibitor (Garriga-Canut et  al., 2006). Glucose initially transported into astrocytes, and converted
metabolism is reduced in BAD (BCL-2-associated to lactate by lactate dehydrogenase (LDH). This
agonist of cell death) knockout mice, which opens lactate is then released to extracellular spaces,
metabolically sensitive KATP channels and leads to transported into neurons, and converted to pyru-
resistance to convulsants such as kainic acid and vate by LDH in glycolysis, which produces ATP in
pentylenetetrazol (Giménez-Cassina et al., 2012). the TCA cycle (Figure 29.1). This metabolic com-
The ketogenic diet has also been shown to sup- munication from astrocytes to neurons is called
press seizures in mice by increasing the activation the astrocyte-neuron lactate shuttle (Bélanger et
of adenosine A1 receptors (Masino et  al., 2011). al., 2011).
In vitro recordings from hippocampal slices have Several lines of evidence support that this
revealed that reductions in glucose hyperpolarize astrocyte-neuron lactate shuttle is used in the sup-
pyramidal neurons, and this is mediated by ade- ply of energy to neurons. First, astrocytes show
nosine A1 receptors and KATP channels (Kawamura lower rates of oxidative metabolism than neu-
et  al., 2010). Consistent with these findings, the rons, and consequently release a large amount
direct activation of adenosine A1 receptors has of lactate to the extracellular spaces (Itoh et  al.,
been shown to suppress chronic seizures in a 2003; Bouzier-Sore et  al., 2006). Second, lactate
mouse model of mesial temporal lobe epilepsy appears to be used as an energy source preferred
(Gouder et al., 2003). over glucose in the brain (Larrabee, 1995; Smith
These extensive studies have uncovered the et al., 2003). Third, although glucose uptakes into
mechanisms underlying the antiepileptic effects neurons and astrocytes are similar in the resting
of the ketogenic diet. However, all of the mol- state of the brain (Nehlig et  al., 2004; Chuquet
ecules revealed by these studies are ion channels et al., 2010), increases in neuronal activities elicit
(KATP channels), synaptic receptors (adenosine the higher glucose uptakes in astrocytes (Chuquet
A1 receptors), and neurotransmitter transporters et  al., 2010)  and also elicit sustained increases
(VGLUTs); namely, molecules regulating elec- in extracellular lactate (Hu and Wilson, 1997).
trical currents. Since the ketogenic diet changes Thus, the energy supply to neurons is achieved by
energy metabolites (glucose and ketone bodies), not only the direct glucose pathway to neurons
it is hypothesized that the brain has “metabolic but also the astrocyte-neuron lactate shuttle (see
pathways” that play key roles in the antiepilep- Figure 29.1).
tic effects of the ketogenic diet. In other words, There is also growing evidence to show that the
there are assumed to be “metabolic enzymes” astrocyte-neuron lactate shuttle regulates electri-
that regulate electrical activities in neurons and cal activities in neurons. First of all, the following
suppress seizures in vivo in the same manner studies have demonstrated that lactate regulates
with the ketogenic diet. In order to explore these electrical activities in neurons. The inhibition of
metabolic pathways and enzymes, we focused on glycolysis reduces synaptic transmission in the
the astrocyte-neuron lactate shuttle in the brain, hippocampus, and this is rescued by the presence
because this lactate shuttle was known to be a met- of lactate (Schurr et al., 1988), which demonstrates
abolic pathway involved in the regulation of elec- that the metabolism of lactate to pyruvate in neu-
trical activities in the brain (Rouach et  al., 2008, rons contributes to the maintenance of synaptic
Parsons and Hirasawa, 2010). transmission. Consistent with this finding, the
inhibition of monocarboxylate transporters that
ASTROCYTE-NEURON transport lactate into neurons also reduces synap-
L A C TAT E S H U T T L E tic transmission in the hippocampus (Izumi et al.,
Glucose is the main energy source in the brain, and 1997). Furthermore, orexin-containing neurons in
produces ATP in neurons and glial cells. The con- the hypothalamus are hyperpolarized by the inhi-
centration of glucose in vivo is approximately 2.5 bition of monocarboxylate transporters (Parsons
mM in the brain (Silver and Erecińska, 1994), and and Hirasawa, 2010). Importantly, they are also
is maintained at a lower concentration than that hyperpolarized by fluoroacetate, a glial toxin, and
in the blood. Glucose is directly transported into this hyperpolarization is recovered by the applica-
neurons and then converted to pyruvate by glycol- tion of lactate (Parsons and Hirasawa, 2010), These
ysis in neurons, which produces ATP in the TCA findings suggest that lactate released from astro-
cycle (Figure 29.1). The role of astrocytes (a type cytes regulates membrane potentials in neurons.
of glial cell) as the energy supplier to neurons has Rouach and colleagues provided more direct
recently received increasing attention (Bélanger evidence to show that electrical activities in neu-
et al., 2011). As shown in Figure 29.1, glucose is rons are regulated by the metabolic pathway from
283

Chapter 29: Lactate Dehydrogenase 283

Astrocyte Neuron
Lactate Glycolysis
LDH
LDH

Pyruvate

Glucose
TCA cycle
Ketone
body

FIGURE 29.1 The astrocyte-neuron lactate shuttle. Glucose is directly transported into neurons, and then converted
to pyruvate by glycolysis. As an alternative pathway, glucose is transported into astrocytes and converted to lactate by
LDH, which is released into extracellular spaces: lactate is then transported into neurons, and converted to pyruvate
by glycolysis. This pathway is called the astrocyte-neuron lactate shuttle. Decreases in glucose and increases in ketone
bodies are elicited by the ketogenic diet. Ketone bodies are transported into neurons, which bypass glycolysis and
directly enter the TCA cycle in neurons.

astrocytes (Rouach et al., 2008). Astrocytes in the ACSF) to ketone bodies. The switch from glucose
hippocampus are connected to one another by gap to ketone bodies hyperpolarized subthalamic neu-
junctions, and thus, small molecules injected into rons; and this hyperpolarization was recovered by
single astrocytes using patch pipettes diffuse into the addition of lactate in ACSF (Sada et al., 2015).
many neighboring astrocytes (D’Ambrosio et  al., Based on the metabolic pathways shown in Figure
1998). By using this property, a group of nearby 29.1, it is likely that the hyperpolarization is elic-
astrocytes can be controlled by selective filling of ited by the decrease in extracellular lactate derived
active small molecules into single astrocytes using from the switch of glucose to ketone bodies, and
patch pipettes (Rouach et  al., 2008; Henneberger recovered by the addition of lactate to ACSF. Thus,
et  al., 2010). Rouach and colleagues made filling the astrocyte-neuron lactate shuttle is likely to reg-
of glucose into single astrocytes in hippocampal ulate membrane potentials in neurons.
slices, and then examined the effects of the depri- In order to confirm this idea, we focused on
vation of extracellular glucose on hippocampal LDH, a metabolic enzyme located in the astrocyte-
synaptic transmission. They demonstrated that, neuron lactate shuttle (Figure 29.1). Thus, this lac-
although glucose deprivation reduced synaptic tate shuttle can be suppressed by inhibiting LDH.
transmission, these reductions were rescued by the We used oxamate, an LDH inhibitor that has been
selective filling of glucose into astrocytes (Rouach used to inhibit LDH in the brain (Lam et al., 2005),
et  al., 2008), showing that hippocampal synaptic and examined the effects of the LDH inhibitor on
transmission is regulated by metabolic communi- membrane potentials in neurons (Sada et al., 2015;
cation from astrocytes. Rouach and colleagues also see Figure 29.2). The inhibition of LDH hyperpo-
showed that epileptiform activities in hippocam- larized neurons in the subthalamic nucleus, and
pal slices were regulated by the selective filling of also hyperpolarized pyramidal cells in the hippo-
glucose into astrocytes (Rouach et al., 2008). campus (Figure  29.2A). Furthermore, the hyper-
polarization elicited by the LDH inhibition was not
L A C TAT E D E H Y D R O G E N A S E : recovered by the addition of lactate, but was recov-
A   M E TA B O L I C TA R G E T ered by the addition of pyruvate (Figure  29.2B).
FOR EPILEPSY Lactate is an upstream metabolite of LDH in neu-
The findings of these studies prompted us to inves- rons, whereas pyruvate is a downstream metabo-
tigate electrical regulation by the astrocyte-neuron lite in neurons (see Figure 29.1). Thus, neuronal
lactate shuttle, and we recently reported that this LDH in the lactate shuttle regulates membrane
lactate shuttle is involved in the antiepileptic potentials in neurons (Figure 29.2C).
effects of the ketogenic diet (Sada et al., 2015). The We also obtained direct evidence to show that
ketogenic diet is known to increase ketone bodies membrane potentials in neurons are regulated by
and decrease glucose in the body (Bough et  al., lactate released from astrocytes (Sada et al., 2015;
2006). Therefore, we made patch clamp recordings see Figure 29.3). Patch-clamp recordings were
from neurons in the subthalamic nucleus in mouse obtained from neighboring pairs of pyramidal cells
brain slices, and switched glucose in the extra- and astrocytes in hippocampal slices, and oxam-
cellular perfusate (artificial cerebrospinal fluid; ate was selectively applied to recorded astrocytes
284

284 section III: Ketogenic Diet in the Laboratory

(a)

–60 mV
3 mV
5 min

Oxamate
(b)

Oxamate Oxamate
28 min 2 mV 28 min 2 mV
2 min 2 min

Oxamate Oxamate

Lactate Pyruvate

(c)
Astrocyte Neuron
Lactate
Glycolysis
LDH
LDH

Pyruvate

Glucose

TCA cycle
Neuronal
Inhibition

FIGURE 29.2 LDH is an electrical regulator in neurons. (A) Pyramidal cells in the hippocampus are hyperpolarized
by oxamate, an LDH inhibitor. (B) The hyperpolarization elicited by the LDH inhibitor is recovered by pyruvate, but not
by lactate. Lactate and pyruvate are located upstream and downstream of neuronal LDH on the astrocyte-neuron lactate
shuttle (see Figure 29.1), respectively. Thus, the lactate-to-pyruvate conversion via LDH in neurons is critical for this
electrical regulation. (C) The summary scheme showing the neuronal inhibition elicited by inhibiting LDH in neurons.
Reproduced from Sada et al. (2015).

through the patch pipette (Figure  29.3A). The membrane potentials in neurons. We also obtained
selective inhibition of LDH in astrocytes elicited evidence to show that this lactate release is actually
hyperpolarization in neighboring pyramidal cells weakened by the ketogenic diet (Sada et al., 2015;
(Figure 29.3B), which demonstrated that LDH in see Figure 29.4). Mice were fed a ketogenic diet
astrocytes regulates membrane potentials in neu- for about 3 weeks, and the lactate concentration
rons. In order to further confirm that this hyper- in the hippocampus was measured and compared
polarization in pyramidal cells was actually due to with that in the hippocampus of age-matched
reductions in the release of lactate from astrocytes mice fed a standard diet. The measurement of lac-
(see Figure 29.3A), we performed the same experi- tate concentration revealed that the hippocampal
ments under the extracellular perfusion of lactate. lactate was lower in mice fed the ketogenic diet
The hyperpolarization induced by filling oxamate than in those fed the standard diet (Figure 29.4A).
into astrocytes (see Figure 29.3B) was not observed Thus, the release of lactate in this lactate shuttle
when lactate was present in the extracellular per- is lowered by the ingestion of the ketogenic diet
fusate (Figure 29.3C). Thus, astrocytic LDH in this (Figure 29.4B).
lactate shuttle also regulates membrane potentials Taken together, these findings show that the
in neurons (Figure 29.3D). astrocyte-neuron lactate shuttle is regulated by the
These in vitro studies revealed that the release ketogenic diet, and LDH in this lactate shuttle is a
of lactate from astrocytes to neurons regulates key enzyme that regulates membrane potentials in
285

(A) (C) Paired recording in lactate


O Glucose Glucose Oxamate Lactate Oxamate Lactate
xa
m
at Rupture
e
Patch Patch
pipette pipette “Rupture”
LDH Lactate LDH

Pyruvate

Astrocyte Pyramidal cell


Astrocyte
(B) Paired recording –77 mV
10 mV
Oxamate Oxamate
2 min
Rupture
“Rupture”

–59 mV
Pyramidal 1 mV
Astrocyte cell 2 min

–76 mV (D)
10 mV
2 min
Astrocyte Neuron
Lactate Glycolysis
LDH
–61 mV LDH

Pyruvate
Pyramidal
cell
Glucose

Neuronal TCA cycle

1 mV Inhibition
2 min

FIGURE  29.3 Electrical regulation by the astrocyte-neuron lactate shuttle. (A) The experimental design used for
simultaneous recordings from pyramidal cells and astrocytes in the hippocampus. LDH in astrocytes are selectively
inhibited by the intracellular application of oxamate via patch pipettes. (B)  Pyramidal cells are hyperpolarized by
the LDH inhibition in astrocytes. Neurons are recorded in the whole-cell mode using normal intracellular solution,
whereas astrocytes are recorded in the cell-attached mode using intracellular solution including oxamate. Oxamate
is selectively applied by rupturing the patch membranes in astrocytes. (C) Pyramidal cells are not hyperpolarized by
the LDH inhibition in astrocytes when lactate is present extracellularly. Thus, lactate is a mediator for this astrocyte-
induced electrical regulation in neurons. (D) The summary scheme showing the neuronal inhibition by inhibiting LDH
in astrocytes. Reproduced from Sada et al. (2015).

(A) (B)
0.05
Astrocyte Neuron
Lactate in the Hippocampus

0.04 **
Lactate Glycolysis
(nmol/μg protein)

LDH
0.03 LDH

Pyruvate
0.02

0.01 Glucose

TCA cycle
0 Ketogenic
Standard Ketogenic diet
diet diet

FIGURE 29.4 Decrease in hippocampal lactate by the ketogenic diet. (A) Lactate concentrations in the hippocampus
are lower in mice fed a ketogenic diet than in those fed a standard diet. **p < .01 (unpaired t-test; n = 11 in each group).
(B) The summary scheme showing the reduction in the release of lactate elicited by the ketogenic diet. Reproduced from
Sada et al. (2015).
286

286 section III: Ketogenic Diet in the Laboratory

neurons. We further reported that seizures in vivo or lactate-to-pyruvate conversion) will be useful
were suppressed by the inhibition of LDH (Sada because these are expected to act on more spe-
et  al., 2015). Microinjection of kainic acid into cific components and functions in the brain. To
the hippocampus is known to produce a mouse date, LDH inhibitors have been synthesized to
model of mesial temporal lobe epilepsy, which create antimalarial compounds (Deck et al., 1998;
exhibits spontaneous paroxysmal discharges with Cameron et  al., 2004). More importantly, LDHA
abnormal morphology in the hippocampus (Riban is known to be involved in antitumor actions
et  al., 2002; Gouder et  al., 2003; Heinrich et  al., based on the Warburg effect (Fantin et al., 2006).
2006). The LDH inhibitor oxamate suppressed Also, LDHA inhibitors have already been syn-
the paroxysmal discharges in the hippocampus thesized and studied as antitumor reagents (Le
of this chronic seizure model (Sada et  al., 2015). et al., 2010; Granchi et al., 2011; Farabegoli et al.,
The in vivo knockdown of lactate dehydrogenase 2012). Drug development for epilepsy by targeting
A  (LDHA), a subunit of LDH, by an antisense LDH enzymes, which is based on the antiepileptic
oligodeoxynucleotide also exerted antiepileptic mechanism of the ketogenic diet, may also lead to
actions in this seizure model. Thus, the inhibition new antitumor drugs based on the Warburg effect.
of LDH suppresses seizures in vivo.

FUTURE DIRECTIONS REFERENCES


T O WA R D   D R U G Bélanger, M., Allaman, I., and Magistretti, P.J. (2011).
DEVELOPMENT Brain energy metabolism:  focus on astrocyte-
The findings of our recent study (Sada et  al., neuron metabolic cooperation. Cell Metab 14,
724–728.
2015) indicate that LDH is a metabolic target for
Bough, K.J., Valiyil, R., Han, F.T., and Eagles, D.A.
epilepsy, and also that the inhibition of LDH mim- (1999). Seizure resistance is dependent upon age
ics the antiepileptic effects of the ketogenic diet. and calorie restriction in rats fed a ketogenic diet.
This metabolic target is clearly different from mol- Epilepsy Res. 35, 21–28.
ecules that have already been identified in previ- Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
ous studies about the ketogenic diet; KATP channels Gawryluk, J.W., Greene, J.G., Shaw, R., Smith,
(Ma et al., 2007; Tanner et al., 2011), adenosine A1 Y., Geiger, J.D., and Dingledine, R.J. (2006).
receptors (Masino et al., 2011), and VGLUTs (Juge Mitochondrial biogenesis in the anticonvulsant
et  al., 2010)  are molecules that directly regulate mechanism of the ketogenic diet. Ann Neurol 60,
electrical currents in neurons. Lactate dehydro- 223–235.
genase as a metabolic target against epilepsy will Bouzier-Sore, A.K., Voisin, P., Bouchaud, V.,
be useful in the development of new antiepileptic Bezancon, E., Franconi, J.M., and Pellerin, L.
drugs. The next step toward drug development is (2006). Competition between glucose and lactate
as oxidative energy substrates in both neurons
to identify chemical compounds that act on LDH.
and astrocytes:  a comparative NMR study. Eur J
Although many antiepileptic drugs are now clini- Neurosci 24, 1687–1694.
cally used, none of antiepileptic drugs have been Cameron, A., Read, J., Tranter, R., Winter, V.J., Sessions,
designed to act on LDH. Antiepileptic drugs to R.B., Brady, R.L., Vivas, L., Easton, A., Kendrick,
act on LDH are also not known. Therefore, we H., Croft, S.L., et  al. (2004). Identification and
explored LDH inhibitors from clinically used anti- activity of a series of azole-based compounds
epileptic drugs, and found that stiripentol, a clini- with lactate dehydrogenase-directed anti-malarial
cal drug for Dravet syndrome (Chiron et al., 2000), activity. J Biol Chem 279, 31429–31439.
is an LDH inhibitor (Sada et al., 2015). By modify- Chiron, C., Marchand, M.C., Tran, A., Rey, E.,
ing its chemical structure, we further identified a d’Athis, P., Vincent, J., Dulac, O., and Pons, G.
stiripentol analog that inhibits LDH and strongly (2000). Stiripentol in severe myoclonic epilepsy
suppresses seizures in vivo (Sada et  al., 2015). in infancy:  a randomised placebo-controlled
Thus, new antiepileptic drugs to mimic the keto- syndrome-dedicated trial. STICLO study group.
Lancet 356, 1638–1642.
genic diet can be developed in the future by target-
Chuquet, J., Quilichini, P., Nimchinsky, E.A., and
ing LDH enzymes with stiripentol derivatives.
Buzsáki G. (2010). Predominant enhancement
In order to develop new antiepileptic drugs, of glucose uptake in astrocytes versus neurons
it will be important to create more powerful and during activation of the somatosensory cortex. J
selective LDH inhibitors. The design and synthe- Neurosci 30, 15298–15303.
sis of selective inhibitors for each LDH subunit D’Ambrosio, R., Wenzel, J., Schwartzkroin, P.A.,
(LDHA or LDHB) and/or for the unidirectional McKhann, G.M., 2nd, and Janigro, D. (1998).
conversion of LDH activities (pyruvate-to-lactate Functional specialization and topographic
287

Chapter 29: Lactate Dehydrogenase 287

segregation of hippocampal astrocytes. J Neurosci Itoh, Y., Esaki, T., Shimoji, K., Cook, M., Law,
18, 4425–4438. M.J., Kaufman, E., and Sokoloff, L. (2003).
Deck, L.M., Royer, R.E., Chamblee, B.B., Hernandez, Dichloroacetate effects on glucose and lactate oxi-
V.M., Malone, R.R., Torres, J.E., Hunsaker, L.A. dation by neurons and astroglia in vitro and on
Piper, R.C., Makler, M.T., and Vander Jagt, D.L. glucose utilization by brain in vivo. Proc Natl Acad
(1998). Selective inhibitors of human lactate dehy- Sci USA 100, 4879–4884.
drogenases and lactate dehydrogenase from the Izumi, Y., Benz, A.M., Katsuki, H., and Zorumski, C.F.
malarial parasite Plasmodium falciparum. J Med (1997). Endogenous monocarboxylates sustain
Chem 41, 3879–3887. hippocampal synaptic function and morphologi-
Fantin, R.F., St-Pierre, J., and Leder, P. (2006). cal integrity during energy deprivation. J Neurosci
Attenuation of LDH-A expression uncovers a link 17, 9448–9457.
between glycolysis, mitochondrial physiology, and Juge, N., Gray, J.A., Omote, H., Miyaji, T., Inoue, T.,
tumor maintenance. Cancer Cell 9, 425–434. Hara, C., Uneyama, H., Edwards, R.H., Nicoll,
Farabegoli, F., Vettraino, M., Manerba, M., Fiume, L., R.A., and Moriyama, Y. (2010). Metabolic con-
Roberti, M., and Di Stefano, G. (2012). Galloflavin, trol of vesicular glutamate transport and release.
a new lactate dehydrogenase inhibitor, induces the Neuron 68, 99–112.
death of human breast cancer cells with different Kawamura, M., Jr., Ruskin, D.N., and Masino, S.A.
glycolytic attitude by affecting distinct signaling (2010). Metabolic autocrine regulation of neurons
pathways. Eur J Pharm Sci 47, 729–738. involves cooperation among pannexin hemichan-
Garriga-Canut, M., Schoenike, B., Qazi, R., Bergendahl, nels, adenosine receptors, and KATP channels. J
K., Daley, T.J., Pfender, R.M., Morrison, J.F., Neurosci 30, 3886–3895.
Ockuly, J., Stafstrom, C., Sutula, T., and Roopra, Kwan, P., and Brodie, M.J. (2000). Early identification
A. (2006). 2-Deoxy-D-glucose reduces epilepsy of refractory epilepsy. N Engl J Med 342, 314–319.
progression by NRSF-CtBP-dependent metabolic Lam, T.K., Gutierrez-Juarez, R., Pocai, A., and Rossetti,
regulation of chromatin structure. Nat Neurosci 9, L. (2005). Regulation of blood glucose by hypotha-
1382–1387. lamic pyruvate metabolism. Science 309, 943–947.
Giménez-Cassina, A., Martínez-François J.R., Fisher, Larrabee, M.G. (1995). Lactate metabolism and its
J.K., Szlyk, B., Polak, K., Wiwczar, J., Tanner, G.R., effects on glucose metabolism in an excised neural
Lutas, A., Yellen, G., and Danial, N.N. (2012). tissue. J Neurochem 64, 1734–1741.
BAD-dependent regulation of fuel metabolism Le, A., Cooper, C.R., Gouw, A.M., Dinavahi, R.,
and KATP channel activity confers resistance to epi- Maitra, A., Deck, L.M., Royer, R.E., Vander Jagt,
leptic seizures. Neuron 74, 719–730. D.L., Semenza, G.L., et al. (2010). Inhibition of lac-
Gouder, N., Fritschy, J.M., and Boison, D. (2003). tate dehydrogenase A induces oxidative stress and
Seizure suppression by adenosine A1 receptor acti- inhibits tumor progression. Proc Natl Acad Sci
vation in a mouse model of pharmacoresistant epi- USA 107, 2037–2042.
lepsy. Epilepsia 44, 877–885. Lutas, A., and Yellen, G. (2013). The ketogenic
Granchi, C., Roy, S., Giacomelli, C., Macchia, M., diet: metabolic influences on brain excitability and
Tuccinardi, T., Martinelli, A., Lanza, M., Betti, L., epilepsy. Trends Neurosci 36, 32–40.
Giannaccini, G., Lucacchini, A., Funel, N., et  al. Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic
(2011). Discovery of N-hydroxyindole-based diet metabolites reduce firing in central neu-
inhibitors of human lactate dehydrogenase iso- rons by opening KATP channels. J Neurosci 27,
form A (LDH-A) as starvation agents against can- 3618–3625.
cer cells. J Med Chem 54, 1599–1612. Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin,
Heinrich, C., Nitta, N., Flubacher, A., Müller, M., D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and
Fahrner, A., Kirsch, M., Freiman, T., Suzuki, F., Boison, D. (2011). A ketogenic diet suppresses sei-
Depaulis, A., Frotscher, M., et  al. (2006). Reelin zures in mice through adenosine A1 receptors. J
deficiency and displacement of mature neurons, Clin Invest 121, 2679–2683.
but not neurogenesis, underlie the formation of Meldrum, B.S., and Rogawski, M.A. (2007). Molecular
granule cell dispersion in the epileptic hippocam- targets for antiepileptic drug development.
pus. J Neurosci 26, 4701–4713. Neurotherapeutics 4, 18–61.
Henneberger, C., Papouin, T., Oliet, S.H., and Rusakov, Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
D.A. (2010). Long-term potentiation depends on Edwards, N., Fitzsimmons, G., Whitney, A., and
release of D-serine from astrocytes. Nature 463, Cross, J.H. (2008). The ketogenic diet for the treat-
232–236. ment of childhood epilepsy:  a randomised con-
Hu, Y., and Wilson, G.S. (1997). A temporary local trolled trial. Lancet Neurol 7, 500–506.
energy pool coupled to neuronal activity:  fluc- Nehlig, A., Wittendorp-Rechenmann, E., and
tuations of extracellular lactate levels in rat brain Lam, C.D. (2004). Selective uptake of [14C]2-
monitored with rapid-response enzyme-based deoxyglucose by neurons and astrocytes: high-
sensor. J Neurochem 69, 1484–1490. resolution microautoradiographic imaging by
288

288 section III: Ketogenic Diet in the Laboratory

cellular 14C-trajectography combined with immu- Sada, N., Lee, S., Katsu, T., Otsuki, T., and Inoue,
nohistochemistry. J Cereb Blood Flow Metab 24, T. (2015). Targeting LDH enzymes with a sti-
1004–1014. ripentol analog to treat epilepsy. Science 347,
Parsons, M.P., and Hirasawa, M. (2010). ATP-sensitive 1362–1367.
potassium channel-mediated lactate effect on Schurr, A., West, C.A., and Rigor, B.M. (1988).
orexin neurons: implications for brain energetics Lactate-supported synaptic function in the rat
during arousal. J Neurosci 30, 8061–8070. hippocampal slice preparation. Science 240,
Rho, J.M., Anderson, G.D., Donevan, S.D., and 1326–1328.
White, H.S. (2002). Acetoacetate, acetone, and Silver, I.A., and Erecińska, M. (1994). Extracellular
dibenzylamine (a contaminant in L-(+)-β- glucose concentration in mammalian brain:  con-
hydroxybutyrate) exhibit direct anticonvulsant tinuous monitoring of changes during increased
actions in vivo. Epilepsia 43, 358–361. neuronal activity and upon limitation in oxygen
Riban, V., Bouilleret, V., Pham-Lê, B.T., Fritschy, J.M. supply in normo-, hypo-, and hyperglycemic ani-
Marescaux, C., and Depaulis, A. (2002). Evolution mals. J Neurosci 14, 5068–5076.
of hippocampal epileptic activity during the devel- Smith, D., Pernet, A., Hallett, W.A., Bingham, E.,
opment of hippocampal sclerosis in a mouse Marsden, P.K., and Amiel, S.A. (2003). Lactate: a
model of temporal lobe epilepsy. Neuroscience preferred fuel for human brain metabolism in vivo.
112, 101–111. J Cereb Blood Flow Metab 23, 658–664.
Rouach, N., Koulakoff, A., Abudara, V., Willecke, K., Tanner, G.R., Lutas, A., Martínez-François, J.R., and
and Giaume, C. (2008). Astroglial metabolic net- Yellen, G. (2011). Single KATP channel opening in
works sustain hippocampal synaptic transmission. response to action potential firing in mouse den-
Science 322, 1551–1555. tate granule neurons. J Neurosci 31, 8689–8696.
289

30
Effects of the Ketogenic Diet on 
the Blood-Brain Barrier
MANOJ BANJARA, PHD AND DAMIR JANIGRO, PHD

THE BLOOD-BRAIN optimum neuronal function. It achieves a function


BARRIER of “transport barrier” by facilitating the uptake
The blood-brain barrier (BBB) is a selectively per- of necessary nutrients, while at the same time
meable cellular boundary that protects the mam- preventing the uptake or actively effluxing other
malian brain from systemic factors (Daneman and molecules. Additionally, the BBB functions as a
Prat, 2015). Even though the microvessel endothe- “metabolic barrier,” possessing intracellular (e.g.,
lial cells (ECs) have a primary role in the formation cytochrome P450) and extracellular (e.g., pepti-
of the BBB, several other cells are equally important dases and nucleotidases) enzymes (Abbott et  al.,
to maintain BBB integrity. Endothelial cells, astro- 2010; Ghosh et  al., 2013; Ghosh et  al., 2011a;
cytes, pericytes, neurons, and other glial cells form Ghosh et al., 2011b). Functions of individual cells
a “neurovascular unit” (Figure 30.1; Abbott et  al., in brain microvessel are discussed in detail in this
2006; Grant and Janigro, 2010; Neuwelt et al., 2011). section
In addition to the layer of capillary ECs, other lay-
ers exist between blood and brain. These include a Endothelial Cells
basement membrane (BM), consisting of type IV Endothelial cells line the interior of blood vessels.
collagen, fibronectin, and laminin that cover capil- The membranes of the capillary ECs are divided
laries completely; pericytes embedded in the BM; into two distinct sides:  luminal (blood side) and
and astrocytes whose processes surround the BM abluminal (brain side) (Daneman and Prat, 2015).
(Figure 30.1). Each of these layers has potential to The ECs form blood vessels, and mural cells sit
restrict the movement of solutes (Hawkins et  al., on the abluminal side of the EC. These brain ECs
2006). Thus, the microenvironment of the BBB acts are less “leaky” than those of the peripheral ves-
in concert with tight junctions to maintain the selec- sels. However, it has been shown that brain ECs
tive permeability of the neurovascular unit barrier. become “leaky” when they are allowed to vascu-
Simply stated, BBB integrity is an essential player larize peripheral tissue, while peripheral ECs form
involved in the maintenance of brain homeostasis. tight junctions resembling the BBB when allowed
Disruption of the BBB occurs in various conditions to vascularize the brain parenchyma (Rubin et al.,
such as neoplasia, trauma, epilepsy, Alzheimer’s 1991). Central nervous system (CNS) micro-
disease, infections (e.g., meningitis), and sterile vascular ECs are 39% thinner than muscle ECs
inflammation (e.g., multiple sclerosis) (Huber et al., (Coomber and Stewart, 1985).
2001; Zlokovic, 2008). Thus, in these conditions it is The type and amount of membrane lipids and
highly desirable to reverse the disruption of the BBB. proteins vary between luminal and abluminal sides
However, a highly restricted BBB is a huge challenge of the blood vessel (Tewes and Galla, 2001). Thus,
for the delivery of drugs into the diseased brain nutrients must transverse two sheaths of mem-
(Gloor et al., 2001). This dual nature of the BBB has brane; their combined characteristics determine
been recently reviewed by Saunders et al. (2014). which particles cross the barrier, and how fast. The
EC transporters are divided into two major catego-
C E L L U L A R A S S O C I AT I O N S ries: efflux transporters and nutrient transporters.
AT   T H E   B B B Efflux transporters are polarized to the luminal
The BBB maintains the proper ionic composition surface and transport a broad variety of lipophilic
of the brain interstitial fluid, which is essential for molecules (Cordon-Cardo et  al., 1989; Thiebaut
290

290 section III: Ketogenic Diet in the Laboratory

(a)
P

E
A E Lumen of blood A
vessels

S P
TJ
BM A: Astrocytes
N
A BM: Basement membrane
E: Endothelial cells
N: Neurons
P: Pericytes
S: Smooth muscle cells
(b) TJ: Tight junction

Blood
LAM
JAM
OP
GLUT1 MCT LAT1 Tfr JAC Z03 Claudin
Z02
Z01

ins
Caten CD
31 Z0-
Pgp BCRP MRP
E Tfr E
Z0-

N-cadherin
3
Ocln Z0-

OP
2

VE - JAC
1

Cad
BM herin
P
Connexin

CNS
Parenchyma

FIGURE 30.1 Schematic diagram of a brain capillary. A. Cell-to-cell interaction at the BBB. The endothelial cells
(E) are joined together by tight junctions (TJ) and surrounded by a basement membrane (BM). Pericytes (P) reside
within the BM and are distributed discontinuously along the length on cerebral capillaries. The cerebral capillaries are
surrounded by astrocytic foot processes or end-feet from astrocytes (A). Axonal projections from neurons (N) containing
vasoactive neurotransmitters and peptide onto arteriolar smooth muscle cells (S). BBB permeability may be regulated by
the molecules released from cells associated with endothelium (e.g., microglia, astrocytes). B. Schematic representation
of structural and transport components at the BBB. GLUT1, glucose transporter 1; MCT, monocarboxylate transporter;
LAT1, L-type amino acid transporter 1; Tfr, transferrin receptor; JAM, junctional adhesion molecules; LAM, leukocyte
adhesion molecules; ZO-1/2/3, zonula occludin-1/2/3; JACOP, junction-associated coiled-coil protein; Ocln, Occludin;
CD31, cluster of differentiation 31; Pgp, p-glycoprotein; BCRP, breast cancer resistant protein; MRP, multidrug
resistance-associated protein.

et al., 1987). Nutrient transporters are specific for enter the CNS (Daneman et al., 2010). Moreover,
the transport of nutrients into, and the removal of these ECs generate a barrier by altering the physical
waste products from, the CNS (Mittapalli et  al., properties of molecules, which then can modulate
2010). The ECs of the BBB contain high numbers their solubility, reactivity, and transport proper-
of mitochondria, which are important to generate ties. These metabolic properties are further regu-
a large amount of adenosine triphosphate (ATP) lated by the flow (Cucullo et al., 2011; Desai et al.,
to maintain ion gradients critical for the function 2002). The combination of a physical barrier (e.g.,
of transporters. In addition, CNS ECs express low tight junctions and low transcytosis), a molecular
levels of leukocyte adhesion molecules, which ulti- barrier (e.g., low leukocyte adhesion molecules,
mately limits the number of immune cells that can efflux transporters, and specific metabolism), and
291

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 291

presence of specific nutrient transporters (e.g., Astrocytes


Glut1; McAllister et  al., 2001)  allows CNS ECs Astrocytes are important glial cells that help in
to firmly regulate CNS homeostasis (Daneman conditioning and developing the brain microves-
and Prat, 2015; Ghosh et  al., 2010). The specific sel ECs. The interactions of ECs and astrocytes
nutrient transporters are probably topographi- are known to regulate the phenotype of the BBB
cally organized for the development or function under (patho)physiological conditions (Prat
of brain-specific region or particular subclass of et  al., 2001). Astrocytes are known to alter the
neurons. A major unanswered question regarding properties of ECs in multiple ways (Grant et al.,
the BBB and ECs is whether they possess unique 1998; Haseloff et al., 2005; Janzer and Raff, 1987;
properties in different brain regions. Lee et  al., 2007; Stanness et  al., 1996; Stanness
The following features distinguish BBB ECs et al., 1997):
from peripheral vasculature (Arcangeli et al., 1999;
Dalvi et al., 2014; Gloor et al., 2001; Hawkins and 1. Tightening the BBB, as evidenced by the
Davis, 2005; Rubin et al., 1991): decreased paracellular permeability of
sucrose.
1. Presence of a large number of tight 2. Elevating transendothelial electrical
junctions to limit the paracellular resistance.
movement of macromolecules (Figures 3. Increasing the activity of barrier-
30.1 and 30.2). related marker enzymes such as
2. Controlled fluid-phase endocytosis alkaline phosphatase and γ-glutamyl
rate to limit transcellular passage of transpeptidase.
macromolecules. 4. Enhancing the number, length, and
3. Presence of highly specific transporters and complexity of tight junctions.
carrier molecules. 5. Increasing the expression of glucose
4. Absence of fenestrations. transporters.
5. Few cytoplasmic vesicles. 6. Secreting angiogenic factors like vascular
6. Increased mitochondrial content. endothelial growth factor (VEGF),

1 2 3 4 6
5
Blood

Endothelial
Cells

Brain Pericyte

Neuron Astrocyte
Microglia

Lipid-soluble molecules 1: Transcellular pathway


2: Paracellular pathway
Water-soluble molecules 3: Transport protein
Tight junctions 4: Receptor-mediated transcytosis
Transport proteins 5: Adsorptive transcytosis
6: Efflux transport
Receptors
Positively charged molecules

FIGURE 30.2 Schematic diagram of transport pathways in cerebrovascular endothelial cells. The transport of diverse
molecules from the circulation across the BBB may include (1) transcellular lipophilic pathway, (2) paracellular aqueous
pathway across tight junctions, (3) transport proteins, (4) receptor-mediated transcytosis, (5) adsorptive transcytosis,
and (6) efflux transporters.
292

292 section III: Ketogenic Diet in the Laboratory

which is essential for the formation and All blood vessels, including those of the brain,
remodeling of embryonic blood vessels, are ensheathed by extracellular matrix layers called
and decreasing vascular stability in adult basement membranes (BMs). The inner vascular
blood vessels. BM and outer parenchymal BM are two BMs that
surround the vascular tube (Daneman and Prat,
Angiotensin-converting enzyme-I produced 2015; Sorokin, 2010). The BM is an extracellular
by astrocytes converts angiotensin I  into angio- matrix secreted specially by ECs and pericytes in
tensin II, which in turn acts on type I angiotensin vascular BM, and by astrocytic processes in paren-
receptors expressed by brain ECs. Angiotensin II chymal BM. Type IV collagens, nidogen, heparin
promotes the recruitment of junctional proteins sulfate proteoglycans, and laminin are some mol-
into the lipid raft, restricting BBB permeability ecules present in BMs. These membranes anchor
(Wosik et  al., 2007). Furthermore, retinoic acid many signaling processes and provide an addi-
(RA) secreted by radial glial cells acts on RA- tional barrier that molecules and cells must cross
receptor β expressed in the developing vascula- prior to accessing neural tissue. The degradation of
ture, which increases transendothelial electrical BMs by matrix metalloproteinases is observed in
resistance by enhancing the expression of vascular different neurological diseases, allowing the infil-
endothelial-cadherin, p-glycoprotein, and zonula tration of leukocytes (Daneman and Prat, 2015;
occludin-1 (Figure 30.1; Mizee et al., 2013). Other Sorokin, 2010).
factors such as transforming growth factor-β,
glial cell-derived neurotrophic factor, and src- M O L E C U L E S AT   T H E
suppressed C-kinase substrate are postulated to BLOOD-BRAIN BARRIER
play a role in maintaining tightness of the BBB The structure and function of the BBB are
(Haseloff et al., 2005). highly specialized by the presence of typical com-
ponents that are discussed in this section.
Pericytes
Pericytes, the mural cells of blood microvessels, Tight Junctions
are specialized cells of mesenchymal lineage that The tight junction (TJ) consists of membrane and
surround the surface of the vascular tube. They cytoplasmic proteins (Figure 30.1). Identification
are on the abluminal side of the microvascular of molecules expressed by CNS ECs led us to under-
endothelial tube and are embedded on the vascu- stand the structural and transport components of
lar basement membrane (Abbott et al., 2010). The the BBB (Ohtsuki et  al., 2014). Claudins, occlu-
CNS microvasculatures have a higher pericyte-to- din, and junctional adhesion molecules (JAM) are
endothelial ratio (1:3 to 1:1) than muscle (1:100) the integral membrane proteins. Cingulin, zonula
(Shepro and Morel, 1993). Pericytes in the BBB occludin proteins (ZO-1, -2, -3), AF6, 7H6 anti-
have several major functions (Armulik et  al., gen, and symplekin are the cytoplasmic accessory
2011; Hall et al., 2014; Lai and Kuo, 2005): proteins that form plaque and function as adap-
tor proteins. The tight junctional complexes are
1. Formation and maintenance of tight dynamic entities that can “bend without break-
junctions. ing,” thereby sustaining structural integrity (Huber
2. Autoregulation of cerebrovascular et al., 2001).
blood flow. Claudins are the largest family of transmem-
3. Secretion of angiopoietin-1 and brain brane phosphoproteins; so far 24 members (clau-
angiogenesis. dins 1–24) have been characterized (Ballabh
4. Initiation of extrinsic blood coagulation et al., 2004). Among these, claudins 1, 3, 5, and
pathway after cerebrovascular injury. 12 have a role in tight-junction formation at
5. Regulation of inflammation by the the BBB. Claudin-1 is an integral component of
secretion of cytokines (e.g., IL-1β and tight junctions, whose absence is associated with
IL-6), leukocyte transmigration, antigen several pathological conditions, such as stroke,
presentation, and T-cell activation. inflammatory diseases, and tumors (Liebner
6. Contraction of capillary diameter by et al., 2000).
contractile proteins in pericytes. Occludin is another transmembrane phospho-
protein, and its subcellular localization parallels
The lack of a pericyte-specific marker, how- that of claudin. In adult brain microvessel ECs,
ever, often leads to misidentification of pericytes occludin expression is higher than the peripheral
as other cells occupying the perivascular space. ECs that interact with claudins. Jointly, they form
293

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 293

channels to regulate the paracellular flow of ions Efflux transporters (e.g., multidrug resistance
and other hydrophilic molecules (Hirase et  al., protein 1, MDR1; breast cancer resistance pro-
1997; Huber et al., 2001; Morita et al., 1999). tein, BCRP; multidrug resistance proteins, MRPs)
The JAMs are members of the immunoglobu- hydrolyze ATP to transport substrates against their
lin superfamily that form homotypic interactions concentration gradient (Ha et  al., 2007). Many
at the endothelial and epithelial cells TJ, which efflux transporters are localized on the luminal
are known to regulate leukocyte extravasation surface in order to transport substrates into the
and paracellular permeability (Aurrand-Lions blood compartment. These transporters facilitate
et al., 2001). the movement of a diverse range of substrates, and
Several cytoplasmic proteins are also essen- provide a barrier to many small lipophilic mol-
tial components of the TJs. Actin is the cyto- ecules, which would otherwise passively diffuse
skeleton protein that plays a major role in through the membrane of ECs. MDR1, also called
maintenance of the TJ. Actin-degrading mol- p-glycoprotein (Pgp), is a well-studied transporter,
ecules (e.g., cytochalasin-D and phalloidin) can which is associated with drug-resistant epilepsy
disrupt the actin cytoskeleton, compromising the (Dombrowski et al., 2001; Marchi et al., 2004) and
TJ (Huber et al., 2001). Zonula occludin proteins tumors (Abbott et  al., 2001). Endogenous sub-
(ZO-1, -2, -3) are also important. These proteins strates of efflux transporters are still not well
make direct contact with claudins, occludins, studied.
and JAMs on one side and the actin cytoskele- Nutrient transporters facilitate the movement
ton on the other (Ballabh et al., 2004). Cingulin of nutrients according to their concentration
is another protein that links the TJ accessory gradients. A wide variety of such transporters are
proteins with the cytoskeleton (Huber et  al., expressed in CNS ECs to deliver specific nutri-
2001). Several intracellular processes involving ents into the brain parenchyma. Most of these
calcium-signaling, phosphorylation, G-proteins, nutrient transporters are in the solute carrier
proteases, and inflammatory cytokine secretion class of facilitated transporters (slc2a1 transports
can modulate TJ proteins (Huber et  al., 2001; glucose, slc16a1 transports lactate and pyruvate,
Kniesel and Wolburg, 2000). SCL7A1 transports cationic amino acids, slc7a5
transports neural amino acids and L-Dopa)
Leukocyte Adhesion Molecules (Zlokovic, 2008). Slc2a1, also called glucose
The CNS of healthy individuals has an extremely transporter 1 (Glut1), is a well-studied nutrient
low immune surveillance, lacking neutrophils transporter that provides glucose to the CNS
and lymphocytes in the parenchyma (Galea et al., (Figure 30.4). It is more frequently expressed by
2007). Entry of leukocytes into the brain paren- brain ECs than nonbrain ECs (Cornford et  al.,
chyma requires multiple steps, including rolling 1994). In humans, Glut1 deficiency leads to an
adhesion, firm adhesion, and extravasation. This epileptic syndrome, which can be treated with a
requires several leukocyte adhesion molecules high ketone diet (De Vivo et  al., 2002; De Vivo
(LAMs) that include selectins (E-, P- selectin) et al., 1991). Most of the transporters in the BBB-
for rolling adhesion and immunoglobulin family ECs provide nutrients to the brain, however, in
members for firm adhesion (Huang et al., 2006). In mild cognitive impairment and Alzheimer’s dis-
a normal brain, the expression of these adhesion ease a receptor (receptor for advanced glycation
molecules is extremely lower in CNS ECs than end products) is thought to assist in transporting
in peripheral ECs. However, LAMs are elevated waste (amyloid β) from blood to brain (Daneman
in neuroinflammatory conditions like stroke and and Prat, 2015).
multiple sclerosis (Daneman et  al., 2010; Huang
et al., 2006). OT H E R C O M P O N E N T S
The rate of transcytosis is lower in brain ECs; how-
TRANSPORTERS ever, it is up-regulated upon BBB dysfunction dur-
The paracellular junctions, “having low perme- ing brain injury and disease. In ECs, transcytosis
ability,” control the transport of molecules and (Figure 30.2) is mediated via caveoline-based ves-
ions between the blood and brain. The ECs of the icle trafficking (Gu et al., 2012). Additionally, the
CNS are highly polarized and have distinct lumi- plasmalemma vesicle-associated protein number
nal and abluminal compartments. As mentioned is lower in the ECs of a healthy BBB than the ECs
earlier, there are two major transporter types that of a BBB following traumatic brain injury (Shue
are expressed in CNS ECs: efflux transporters and et al., 2008). The significance of these changes in
nutrient transporters (Figures 30.1–30.4). the context of disease etiology is unknown.
294

294 section III: Ketogenic Diet in the Laboratory

FUNCTIONS OF THE CNS express a number of energy-dependent efflux


BLOOD-BRAIN BARRIER transporters (Figures 30.1 and 30.2) that actively
extrude neurotoxins out of the brain (Abbott et al.,
Regulation of Transit across 2010). These transporters also prevent neuronal
the Blood-Brain Barrier cell death where neurons are exposed to toxins
The BBB provides a suitable environment for neu- (Marchi et al., 2004).
ral functions because it regulates ionic composi-
tion in the brain. The combination of ion-specific Transport of Nutrients to the Brain
channels and transporters at the BBB regulates the Most of the essential water-soluble nutrients and
composition and quantity of ions in the CNS. The metabolites required for neuronal survival and
concentration of potassium in mammalian brain differentiation are transported into the brain by
cerebrospinal fluid (CSF) and interstitial fluid specific transport systems (Figures  30.2–30.4) at
(ISF) is maintained at around 2.5–2.9 mM, though the BBB. The endothelium begins to differentiate
it is approximately 4.5 mM in plasma. Even though into a barrier layer from the embryonic angiogen-
plasma K+ concentration changes following exer- esis stage, and is maintained in adults by its asso-
cise or meals, it remains within 2.5–2.9 mM range ciation with other cell types, especially the endfeet
in the brain (see Janigro, 1999, 2012). In addition, of astroglial cells. Astrocytic glial cells promote
Ca+, Mg2+, and pH are actively regulated in the BBB the up-regulation of tight junction proteins and
(Abbott et al., 2010; Jeong et al., 2006; Nischwitz et the differential expression of luminal and ablu-
al., 2008; Somjen, 2004). minal membrane-specific transporters (Abbott
The BBB acts as a shield to prevent the entry et al., 2006; Wolburg et al., 2009). Other cell types,
of most macromolecules into the brain. The total namely pericytes, microglia, and nerve terminals
protein content in plasma is much higher than in play supporting roles in barrier induction, mainte-
CSF, and the individual protein composition is nance, and function (Abbott et al., 2006; Nakagawa
distinctly different (Abbott et  al., 2010). Proteins et al., 2009; Shimizu et al., 2008).
that are markedly high in plasma (e.g., albumin, Together with oxygen, glucose is the obligatory
prothrombin, and plasminogen) are detrimental nutrient for the human brain. The BBB regulates
to nervous tissue, as they can trigger cell apoptosis the transport of glucose in the CNS and prevents
(Gingrich and Traynelis, 2000). Factor Xa and tis- equalization of plasma levels (~ 5 mM) to the
sue plasminogen activator are widely expressed in CSF and ISF levels (< 2.5 mM) (McAllister et al.,
the CNS, and can convert prothrombin and plas- 2001). It is used for ATP production, which is
minogen to thrombin and plasmin, respectively. primarily used for ion transport and the mainte-
If present, thrombin and plasmin can initiate cas- nance of the ion gradient (Magistretti and Pellerin,
cades resulting in seizures, glial activation, glial 1995). However, a small fraction is also used for
cell division, and cell death (Abbott et  al., 2010). biosynthetic processes. The diffusion of this vital
Hence, the ingress of these macromolecules upon nutrient across the BBB is facilitated by the glu-
BBB disruption can have serious pathological cose transporter type 1 (Glut1). In the brain, Glut1
consequences. interacts with other Glut1 isoforms that mediate
After taking meals, blood levels of a neuro- glucose transport into neurons and astrocytes
excitatory amino acid, glutamate, are elevated. (Klepper, 2008).
If glutamate is freely transposed into the brain Brain glucose levels in a healthy and normally
ISF, it can be detrimental. For example, in an active person are kept adequate by a complex regu-
ischemic stroke patient, uncontrolled glutamate latory mechanism to ensure an appropriate supply
release into the brain can cause permanent neu- to neurons, in spite of massive drops in plasma lev-
roexcitatory damage of neural tissue. The periph- els. Glucose is controlled by intracellular enzymes
eral and central nervous systems share many of (e.g., hexokinase) and Glut1 proteins that are
the same neurotransmitters; the BBB, however, expressed asymmetrically in the ECs (lower con-
keeps them separate and minimizes the cross-talk centration in abluminal membrane than in lumi-
(Abbott et al., 2010; Abbott et al., 2006; Bernacki nal membrane) (McAllister et  al., 2001). Thus,
et al., 2008). glucose levels in the brain parenchyma and CSF
The BBB is a protective barricade that shields ultimately depend on the “barrier” nature of the
the brain from neurotoxic substances circulat- brain endothelium.
ing in the blood (e.g., endogenous metabolites or Under normal conditions, asymmetrically
proteins, and xenobiotics ingested in the diet or distributed Glut1 in the BBB transports blood
acquired from the environment). The ECs in the glucose into the brain, in which barrier integrity
295

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 295

is maintained by tight junctions, preventing potas- Once KBs cross the BBB, brain cells take up
sium leakage in the brain (Figure 30.5). However, KBs via diffusion or a carrier-mediated process to
under pathological conditions GLUT1 expression support cellular energy requirements. Even though
is altered (Cornford et al., 1998a; Cornford et al., neurons and glial cells express MCTs, since KBs
1998c). Cerebral blood flow also fails to meet the must transverse the BBB first, their role appears
brain metabolic demand when the BBB is damaged more significant at the BBB.
(Bruehl et al., 1998), and in such cases, potassium
leaks across the open tight junctions in the brain Regulation of Cerebral Ketone Uptake
(Cornford et  al., 1998a; Cornford et  al., 1998b; In the absence of glucose, monocarboxylic acids
Cornford et al., 1998c). The elevated extracellular (e.g., ketones, lactate, and pyruvate) generate a
[K+] further increases the metabolic demand due substantial amount of energy for the brain (Pierre
to exaggerated neuronal firing and lost homeosta- and Pellerin, 2005). Prolonged fasting elevates
sis by voltage-dependent channels. normal circulating KB concentrations from ~5.8
However, increased plasma ketone levels pro- mM to 9 mM (White and Venkatesh, 2011). The
duced due to starvation, exercise, or ketogenic plasma concentration of KBs is a major factor
diet (KD) ingestion provide an alternative energy regulating the rate of cerebral uptake. Since KBs
source to fulfill the energy demand in BBB- are hydrophilic compounds in the blood, specific
damaged conditions (Figures 30.3 and 30.5). In transporters are required to facilitate KB diffu-
addition, ketone bodies (KBs) directly enhance sion across the BBB and maintain proper levels of
Glut1 activity (Janigro, 1999). these metabolic products in the brain (Pierre and
Pellerin, 2005). Monocarboxylic acid transporters
K E TO G E N I C   D I E T (e.g., MCT1 and MCT2) involved in transport-
The KD is a strict high-fat, low-carbohydrate, ing monocarboxylic acids (e.g., lactic acid and
and low-protein diet. As a result of this diet, the pyruvate) are thought to facilitate the transport
body receives a minimal dietary source of glu- of KBs. The first monocarboxylic acid transporter
cose, which is required for all metabolic needs identified in brain ECs was MCT1. Additionally,
(Freeman et  al., 2006). Fatty acids become an small amounts of MCT1 were found in astrocytic
obligatory source of cellular energy production, endfeet surrounding the capillaries. Interestingly,
however, they do not readily cross the BBB due MCT2 was found only in BBB ECs and neurons,
to the presence of tight junctions (Mitchell et al., but not in astrocytes. On the other hand, MCT4
2009). Their transport into the brain by diffu- was present exclusively in the astrocytes and glial
sion or via specific protein-mediated transport is cells. Glucose and monocarboxylate transporters
still debated and controversial (Abumrad et  al., expressed in the major brain cells are summarized
1998; Hamilton, 1998). The KD is characterized in a schematic diagram (Figure 30.4). This distri-
by elevated serum levels of circulating KBs:  ace- bution of MCT is associated with the distinctive
toacetate (AcAc), β-hydroxybutyrate (BHB), and functional characteristics. Brain MCT1 and MCT4
acetone (Ac), which are mainly produced by the facilitate the release of monocarboxylates into the
liver. The plasma concentration of KBs increases to extracellular space and allow cells to maintain a
three-to-four times its basal levels (AcAc: 100 µM, high glycolytic rate (Pierre and Pellerin, 2005).
BHB: 200 µM) (Musa-Veloso et al., 2002). The KBs These monocarboxylates produced and released by
are utilized by extrahepatic tissues as an energy astrocytes in the extracellular space are an energy
source. However, Ac is not an energy source and substrate for active neurons; monocarboxylates
is exhaled or excreted as a waste. In the absence are taken up by MCT2 expressed in the dendrites
of glucose, KBs are the preferred source of brain and axons of neurons (Pierre and Pellerin, 2005;
energy. They are transported into the brain by Takimoto and Hamada, 2014).
simple (Ac) or facilitated diffusion (AcAc and At a given arterial concentration, AcAc uptake
BHB) mediated by the monocarboxylate trans- is double that of BHB; however, it is not yet under-
porter (MCT) (Figures 30.3–30.5) (Cremer, 1982; stood how the expression of MCT is regulated
Klepper, 2008). These MCTs are expressed in the (White and Venkatesh, 2011). Prolonged fasting
plasma membrane of choroid plexus and BBB cells increases BBB uptake of KBs by increasing the
(endothelial and epithelial), glia, and neurons, expression of MCT1 as high as eightfold in mice
which ultimately oxidize ketones in the mito- (Leino et  al., 2001). Similarly, another report in
chondrial matrix, releasing acetyl-CoA that enters human subjects showed a 13-fold increase in
into the tricarboxylic acid (TCA) or Krebs cycle cerebral BHB uptake following several days of
(Morris, 2005) (Figures 30.3 and 30.4). fasting (Hasselbalch et  al., 1994). Interestingly, a
296

Ketogenic Diet

Glucose GLUT1 Glucose

Acetone Acetone
Fatty acid
FFA Oxidation Acetyl-
CoA
Acetoacetate MCT Acetoacetate
FFA
-hydroxybutyrate MCT
-hydroxybutyrate
Oxaloacetate

TCA cycle

ix
atr
lM
ria
Stomach
ito
ch
ond
Blood BBB
M

Cytosol
Liver
Brain
BBB: Blood-brain barrier
FFA: Free fatty acid
GLUT1: Glucose transporter 1
MCT: Monocarboxylate transporter
TCA: Tricarboxylic acid

FIGURE  30.3 Schematic diagram of ketogenic diet metabolism and brain uptake. The ketogenic diet is deficient
in carbohydrates, but provides large amounts of long chain free fatty acids (FFA). The acetyl-CoA produced by fatty
acid oxidation goes into the Krebs/tricarboxylic acid (TCA) cycle or is converted to ketone body (KB) acetoacetate
(AcAc), which spontaneously degrades to acetone (Ac). AcAc is further converted to β-hydroxybutyrate (BHB) in a
reversible reaction. These KBs represent alternative energy substrates for the brain. GLUT1, glucose transporter 1; MCT,
monocarboxylate transporter.

Lumen of blood
vessels A

GLUT1 MCT1 A: Astrocyte


GLUT3 MCT2 C: Choroid Plexus
GLUT4 MCT3 E: Endothelial Cell
GLUT5 MCT4 N: Neuron
GLUT6 M: Microglia
GLUT8

FIGURE  30.4 Schematic diagram of glucose and monocarboxylate transporters in the brain. Multiple types of
glucose transporters (GLUTs) and monocarboxylate transporters (MCTs) are responsible to transport glucose and
monocarboxylic acids (ketone bodies, lactate, and pyruvate), respectively.
297

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 297

Normal BBB Damaged BBB Damaged BBB + Ketogenic Diet

A A
Glucose A
Glucose Glucose

KB
KB
P
P KB P
Glucose

E E E
Blood Brain Blood Brain Blood Brain

A: Astrocyte
E: Endothelial Cell
P: Pericytes
GLUT1
MCT
Cx43

FIGURE 30.5 Schematic diagram of cellular and molecular events in normal and BBB damaged tissue. The ketogenic
diet potentially has a role to reestablish physiologic metabolic supply and repair damaged BBB by inducing the expression
levels of monocarboxylate transporter (MCT) and connexin 43 (Cx43), and enhancing glucose transporter 1 (GLUT1)
activity.

rapid increase in plasma ketone levels, following et al., 2009; Ivens et al., 2007; Tomkins et al., 2008),
an intravenous infusion of BHB, does not induce ultimately compromising the BBB integrity. An
cerebral uptake as significantly as prolonged fast- optimum amount of neuroinflammation is con-
ing; this suggests that MCT up-regulation is par- sidered neuroprotective, and this generally occurs
tially dependent on the length of exposure to for a short period of time. Chronic neuroinflam-
increased plasma KBs (Pan et al., 2000; Pan et al., mation is often detrimental, inducing further cell
2001). Thus, a prolonged high-fat diet increases and tissue damage. However, the opposing mecha-
blood ketone concentrations, leading to an ele- nisms of this ostensibly paradoxical exacerbation
vated expression of MCT and cerebral uptake of or amelioration of parenchymal brain injury are
KBs (Figure 30.5). largely unknown (Cederberg and Siesjo, 2010;
Finnie, 2013).
Anti-Inflammatory Effects The therapeutic efficacy of large spectrum anti-
of Ketone Bodies neuroinflammatory drugs is limited by side effects
Neuroinflammation is defined as inflammation after even transient immune suppression. Recent
of CNS that occurs upon invasion of pathogens, studies suggest that the KD could be useful, par-
trauma, and/or neurodegeneration. Cells such as ticularly in patients with recent worsening of epi-
macrophages, astrocytes, and oligodendrocytes lepsy and inflammation (Janigro, 1999; Marchi
and molecules such as cytokines and comple- et al., 2012; Nabbout et al., 2011). The KD has been
ment and pattern-recognition components are used as an effective treatment in patients with
the known contributors to neuroinflammation inflammation-induced epileptic encephalopathies
(Banjara, 2014; de Vries et  al., 2012; Glass et  al., (e.g., fever-induced refractory epileptic encepha-
2010; Marchi et al., 2014). Proinflammatory mol- lopathy in school-aged children) (Nabbout et  al.,
ecules are either generated locally within the CNS 2011). Additionally, polysaturated fatty acids,
or transported from the peripheral system upon mostly eicosapentaenoic acid and docosahexae-
BBB disruption. Moreover, inflammatory media- noic acid, are thought to have anti-inflammatory
tors have been shown to influence tight junctions actions, mediated typically by their hydroxylated
and to activate astrocytes and microglia (David metabolites (Grimble, 1998; Porta et  al., 2009).
298

298 section III: Ketogenic Diet in the Laboratory

Furthermore, KD attenuated thermal nociception immune system, which is primarily responsible for
and decreased peripheral edema, indicating an IL-1β derived autoinflammatory disorders such as
anti-inflammatory effect (Ruskin et al., 2009). gout, obesity, atherosclerosis, and neurodegenera-
A systemic review published by Gibson et al., tive diseases (Levy et al., 2015). And BHB, but not
which analyzed 16 independent studies contain- structurally similar AcAc or butyrate, was shown
ing a total of 733 rodent studies, demonstrated to limit the cytokine production by inhibiting
a significant protective (both pathological and the activation of these NLRP3 inflammasome in
functional) effect of KD or exogenous adminis- human monocytes in vitro (Yasui et  al., 2011).
tration of KBs (p < .001) on outcomes following Since NLRP3 is a common player of sterile neu-
cerebral ischemia. Moreover, regardless of dietary roinflammation, the identification of BHB as an
intervention or administration-induced ketogenic NLRP3 inhibitor provides a rationale to further
state, ketones were found to have beneficial/anti- investigate the effectiveness of KD for the treat-
inflammatory effects on functional/behavioral ment of neuroinflammatory diseases.
tests, lesion volume, and brain water content
(Gibson et al., 2012).
Neuroprotective Effects of Ketone Bodies
In traumatic brain injury (TBI) models, con-
Ketones, an alternative energy source for normal
cussion or mild TBI models, and spinal cord
or injured brain, may also have neuroprotective
injury, KD intervention (pretreatment or admin-
effects (Prins, 2008). Ketones are considered neu-
istration to postinjury animals) revealed improved
roprotective due to the following:
structural and functional outcomes (Prins, 2008;
Prins and Matsumoto, 2014). Within minutes after
1. BHB is a more efficient energy source than
injury, the ionic equilibrium across the neuronal
glucose. It can also stimulate mitochondrial
membrane is disrupted, which requires cellular
biogenesis by up-regulating mitochondrial
energy to reestablish homeostasis. Thus, the cere-
enzymes and genes that stimulate energy
bral glucose uptake is increased in both rodent and
metabolism (Veech et al., 2001).
human within 30 min after fluid percussion and
2. KBs protect cells against glutamate-
within 8 days after TBI, respectively (Bergsneider
mediated apoptosis and necrosis by
et al., 1997). However, this transient “hyperglycol-
attenuating the formation of reactive
ysis” is followed by a prolonged period of reduc-
oxygen species (Ziegler et al., 2003). They
tion in glucose metabolism. Hence, utilizing brain
also attenuate apoptosis by reducing the
ketone metabolism after TBI as a therapeutic
activation of caspase-3 (Gasior et al., 2006).
approach is appealing as it can bypass the early glu-
3. KBs enhance the conversion of glutamate
cose metabolic derangements and also offers mul-
to gamma-aminobutyric acid (GABA)
tiple consequences that are beneficial to the brain
and boost the GABA-mediated inhibition
(Prins and Matsumoto, 2014). Ketone metabo-
(Gasior et al., 2006).
lism is particularly advantageous to TBI patients
4. Cerebral blood flow elevated by ketone
because it decreases the production of free radi-
metabolism is often considered as an effect
cals in mitochondria and cytosol (Sullivan et  al.,
of neuroprotective therapy (Hasselbalch
2004; Ziegler et  al., 2003). In addition, animals
et al., 1996).
on the KD produced less Bcl-2 associated protein,
ultimately decreasing cellular apoptosis and brain
Even though studies so far demonstrated
swelling. Furthermore, KD-induced decrease in
beneficial neuroprotective effects of KD, fur-
mitochondrial release of the cytochrome c into the
ther research is necessary to clarify issues such
cytosol inhibits the stimulation of apoptotic sig-
as dosing, timing, and the route and duration of
naling cascade (Hu et al., 2009a; Hu et al., 2009b).
administration.
More recently, it has been shown that KD can
modulate lipopolysaccharide-induced fever by
decreasing peripheral inflammation and CNS IL- Effects of Ketone Bodies on 
1β expression (Dupuis et al., 2015). These proper- the Blood-Brain Barrier
ties potentially contribute to the beneficial effect Data on the effects of the KD and/or KBs on the
of KD in neuroinflammatory conditions (e.g., BBB are based largely on animal studies. There
epilepsy, Alzheimer’s disease, Parkinson’s disease, is evidence of rapid changes in cerebral blood
multiple sclerosis) and/or brain trauma. flow and cellular transporters (decreased Glut1
The NOD-like receptor P3 (NLRP3) inflam- expression) that favor KBs’ uptake and metabo-
masome is a multiprotein complex of the innate lism in several neurological conditions such as
299

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 299

TBI, hemorrhagic shock, ischemia, and hypoxia pH and sodium concentrations in the brain
(Gasior et al., 2006; Gibson et al., 2012; Prins and (Hiraide et  al., 1991). Thus, hypoglycemia and
Matsumoto, 2014; White and Venkatesh, 2011). dehydration are two predicted side effects of
Prins et al. demonstrated an increase in expres- continuous ketone consumption. The long-term
sion of MCT (MCT1 and MCT2) levels and ketone consequences of reduction in glucose cerebral
transport following TBI in rats (Prins and Giza, metabolism are thought to increase cerebral
2006). Additionally, ketone metabolizing enzyme, blood flow, increase expression levels of MCT,
β-hydroxybutyrate dehydrogenase, is elevated fol- and enhance KB’s uptake and metabolism on the
lowing cerebral injury that converts BHB to AcAc, BBB, however details are not yet known. In con-
which is scarce in the adult brain (Tieu et al., 2003). trast, a report showed that the prolonged intake of
These evidences suggest brain’s improved ability to KD can significantly raise mean blood cholesterol
utilize exogenous KBs. Thus, taking advantage of levels, leading to lipid deposition in blood vessels
improved transport and cellular metabolism of (Freeman et al., 1998).
ketones, brain’s reliance for energy may shift from
glucose to KBs in injured brain. It is beneficial for CONCLUDING REMARKS
the brain, particularly as hyperglycemia is deleteri- It has long been recognized that increased con-
ous to the injured brain (Salim et al., 2009). centration of KBs associated with the KD is effi-
Astrocyte endfeet lacking the gap junction pro- cient fuel for the brain. Dramatic effects of such
tein connexin 43 (Cx43) are inefficient in trans- high-fat dietary treatment were shown in patients
membrane receptor anchoring. This weakens the with multiple neurological disorders and Glut1
BBB, which further fails upon increased hydro- deficiency syndrome. Studies suggest that the
static vascular pressure and shear stress (Ezan ketogenic diet potentially has the ability to rees-
et al., 2012). Interestingly, higher concentrations of tablish physiologic metabolic supply by activating
KBs, either alone or in combination, significantly glucose transporters, enhancing the expression of
up-regulated the expression of the Cx43 at both ketone transporters, and repairing damaged BBB
mRNA and protein levels (Ho et al., 2013) (Figure by inducing the expression levels of a gap junction
30.5). Further, the up-regulation of Cx43 pro- proteins. However, additional studies are essential
tein in ECs and glial cells by KBs accelerated cell to validate these initial observations and explore
migration (Bates et al., 2007). further.
In a separate report, BHB (0.5 mM) has been
shown to promote human endothelial cell prolif- REFERENCES
eration (Cheng et al., 2006). These studies suggest Abbott, N.J., Khan, E.U., Rollinson, C., Reichel, A.,
that the KD plays a role in repairing damaged BBB Janigro, D., Dombrowski, S., Dobbie, M., and
Begley, D.J. (2001). Drug resistance in epilepsy: the
and restoring normal brain function. In contrast,
role of the blood-brain barrier. In Mechanisms
Freeman et al. demonstrated AcAc-induced inhi-
of Drug Resistance in Epilepsy:  Lessons
bition of EC proliferation by elevation of oxida- from Oncology, Bock, G. and Goode, J.A. ed.
tive stress (Freeman et  al., 2006; Freeman et  al., (Chichester, UK: John Wiley), pp. 38–47.
1998). Hence, the response of cell types to dif- Abbott, N.J., Patabendige, A.A., Dolman, D.E., Yusof,
ferent KBs might vary depending on cell-specific S.R., and Begley, D.J. (2010). Structure and func-
characteristics. tion of the blood-brain barrier. Neurobiol Dis
In severely uncontrolled diabetes, KBs are pro- 37, 13–25.
duced in massive quantities, which is described Abbott, N.J., Ronnback, L., and Hansson, E. (2006).
as ketoacidosis; this causes high concentrations Astrocyte-endothelial interactions at the blood-
of protons and overwhelms the buffering system brain barrier. Nat Rev Neurosci 7, 41–53.
of the body by activating a pH sensitive sodium- Abumrad, N., Harmon, C., and Ibrahimi, A. (1998).
proton pump (Bohn and Daneman, 2002). In Membrane transport of long-chain fatty acids: evi-
contrast, during high-fat/low-carbohydrate dence for a facilitated process. J Lipid Res 39,
2309–2318.
intake, ketone bodies are produced in a regulated
Arcangeli, A., Rosati, B., Crociani, O., Cherubini, A.,
manner causing a harmless physiological state Fontana, L., Passani, B., Wanke, E., and Olivotto,
called dietary ketosis. There is minimal data on M. (1999). Modulation of HERG current and herg
the adverse effects of ketogenic diet on the BBB. gene expression during retinoic acid treatment of
Depending on concentrations, acute administra- human neuroblastoma cells: potentiating effects of
tion of KBs alters the pH, sodium level, lipids, BDNF. J Neurobiol 40, 214–225.
and glucose in the blood. The acute intravenous Armulik, A., Genov, G., and Betsholtz, C. (2011).
infusion of BHB also significantly increases the Pericytes:  developmental, physiological, and
300

300 section III: Ketogenic Diet in the Laboratory

pathological perspectives, problems, and prom- Cornford, E.M., Hyman, S., Cornford, M.E., Damian,
ises. Dev Cell 21, 193–215. R.T., and Raleigh, M.J. (1998b). A single glucose
Aurrand-Lions, M., Johnson-Leger, C., Wong, C., Du transporter configuration in normal primate brain
Pasquier, L., and Imhof, B.A. (2001). Heterogeneity endothelium:  comparison with resected human
of endothelial junctions is reflected by differen- brain. J Neuropathol Exp Neurol 57, 699–713.
tial expression and specific subcellular localiza- Cornford, E.M., Hyman, S., Cornford, M.E., Landaw,
tion of the three JAM family members. Blood 98, E.M., and Delgado-Escueta, A.V. (1998c). Interictal
3699–3707. seizure resections show two configurations of endo-
Ballabh, P., Braun, A., and Nedergaard, M. (2004). The thelial Glut1 glucose transporter in the human
blood-brain barrier: an overview: structure, regu- blood-brain barrier. J Cereb Blood Flow Metab
lation, and clinical implications. Neurobiol Dis 18, 26–42.
16, 1–13. Cremer, J.E. (1982). Substrate utilization and brain
Banjara, M. (2014). Lipocalin-2:  a new regulator of development. J Cereb Blood Flow Metab 2, 394–407.
non-pathogen-associated neuroinflammation. Int Cucullo, L., Hossain, M., Puvenna, V., Marchi, N.,
J Clin Exp Neurol 2, 8–15. and Janigro, D. (2011). The role of shear stress in
Bates, D.C., Sin, W.C., Aftab, Q., and Naus, C.C. Blood-Brain Barrier endothelial physiology. BMC
(2007). Connexin43 enhances glioma invasion by Neurosci 12, 40.
a mechanism involving the carboxy terminus. Glia Dalvi, S., On, N., Nguyen, H., Pogorzelec, M., Miller,
55, 1554–1564. D.W., and Hatch, G.M. (2014). e blood brain bar-
Bergsneider, M., Hovda, D.A., Shalmon, E., Kelly, D.F., rier-regulation of fatty acid and drug transport. In
Vespa, P.M., Martin, N.A., Phelps, M.E., McArthur, Neurochemistry, T. Heinbockel, ed. (InTech). DOI:
D.L., Caron, M.J., and Kraus, J.F. (1997). Cerebral 10.5772/57604. Available from: http://www.intecho-
hyperglycolysis following severe traumatic brain pen.com/books/neurochemistry/the-blood-brain-
injury in humans:  a positron emission tomogra- barrier-regulation-of-fatty-acid-and-drug-transport
phy study. J Neurosurg 86, 241–251. Daneman, R., and Prat, A. (2015). The blood-brain
Bernacki, J., Dobrowolska, A., Nierwiska, K., and barrier. Cold Spring Harb Perspect Biol 7, a020412.
Malecki, A. (2008). Physiology and pharmacologi- Daneman, R., Zhou, L., Agalliu, D., Cahoy, J.D.,
cal role of the blood-brain barrier. Pharmacol Rep Kaushal, A., and Barres, B.A. (2010). The mouse
60, 600–622. blood-brain barrier transcriptome: a new resource
Bohn, D., and Daneman, D. (2002). Diabetic ketoaci- for understanding the development and func-
dosis and cerebral edema. Curr Opin Pediatr 14, tion of brain endothelial cells. PLoS One 5,
287–291. e13741–e13741.
Bruehl, C., Hagemann, G., and Witte, O.W. (1998). David, Y., Cacheaux, L.P., Ivens, S., Lapilover, E.,
Uncoupling of blood flow and metabolism in epi- Heinemann, U., Kaufer, D., and Friedman, A.
lepsy. Epilepsia 39, 1235–1242. (2009). Astrocytic dysfunction in epileptogen-
Cederberg, D., and Siesjo, P. (2010). What has inflam- esis:  Consequence of altered potassium and glu-
mation to do with traumatic brain injury? Childs tamate homeostasis? J Neurosci 29, 10588–10599.
Nerv Syst 26, 221–226. De Vivo, D.C., Trifiletti, R.R., Jacobson, R.I., Ronen,
Cheng, S., Chen, G.Q., Leski, M., Zou, B., Wang, Y., and G.M., Behmand, R.A., and Harik, S.I. (1991).
Wu, Q. (2006). The effect of D, L-hydroxybutyric Defective glucose transport across the blood-brain
acid on cell death and proliferation in L929 cells. barrier as a cause of persistent hypoglycorrhachia,
Biomaterials 27, 3758–3765. seizures, and developmental delay. N Engl J Med
Coomber, B.L., and Stewart, P.A. (1985). Morphometric 325, 703–709.
analysis of CNS microvascular endothelium. De Vivo, D.C., Leary, L., and Wang, D.O.N.G. (2002).
Microvasc Res 30, 99–115. Glucose transporter 1 deficiency syndrome and other
Cordon-Cardo, C., O’Brien, J.P., Casals, D., Rittman- glycolytic defects. J Child Neurol 17, 3S15–13S23.
Grauer, L., Biedler, J.L., Melamed, M.R., and de Vries, H.E., Kooij, G., Frenkel, D., Georgopoulos, S.,
Bertino, J.R. (1989). Multidrug-resistance gene (P- Monsonego, A., and Janigro, D. (2012). Inflammatory
glycoprotein) is expressed by endothelial cells at events at blood-brain barrier in neuroinflammatory
blood-brain barrier sites. PNAS 86, 695–698. and neurodegenerative disorders:  Implications for
Cornford, E.M., Hyman, S., and Swartz, B.E. clinical disease. Epilepsia 53, 45–52.
(1994). The human brain GLUT1 glucose trans- Desai, S.Y., Marroni, M., Cucullo, L., Krizanac-Bengez,
porter:  ultrastructural localization to the blood- L., Mayberg, M.R., Hossain, M.T., Grant, G.G., and
brain barrier endothelia. J Cereb Blood Flow Janigro, D. (2002). Mechanisms of endothelial sur-
Metab 14, 106–112. vival under shear stress. Endothelium 9, 89–102.
Cornford, E.M., Hyman, S., Cornford, M.E., and Dombrowski, S.M., Desai, S.Y., Marroni, M., Cucullo,
Damian, R.T. (1998a). Glut1 glucose transporter L., Goodrich, K., Bingaman, W., Mayberg, M.R.,
in the primate choroid plexus endothelium. J Bengez, L., and Janigro, D. (2001). Overexpression
Neuropathol Exp Neurol 57, 404–414. of multiple drug resistance genes in endothelial
301

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 301

cells from patients with refractory epilepsy. cellular permeability control at the blood-brain
Epilepsia 42, 1501–1506. barrier. Brain Res Rev 36, 258–264.
Dupuis, N., Curatolo, N., Benoist, J.F., and Auvin, S. Grant, G.A., Abbott, N.J., and Janigro, D. (1998).
(2015). Ketogenic diet exhibits anti-inflammatory Understanding the physiology of the blood-brain
properties. Epilepsia 56, e95–e98. barrier:  in vitro models. News Physiol Sci 13,
Ezan, P., Andr, P., Cisternino, S., Saubama, B., Boulay, 287–293.
A.C., Doutremer, S., Thomas, M.A., Quenech’du, Grant, G.A., and Janigro, D. (2004). The blood-brain
N., Giaume, C., and Cohen-Salmon, M. (2012). barrier. In Youmans Neurological Surgery, H.R.
Deletion of astroglial connexins weakens the Winn, ed. (Philadelphia, PA:  Saunders), pp.
blood-brain barrier. J Cereb Blood Flow Metab 32, 153–174.
1457–1467. Grimble, R.F. (1998). Nutritional modulation of cyto-
Finnie, J.W. (2013). Neuroinflammation:  benefi- kine biology. Nutrition 14, 634–640.
cial and detrimental effects after traumatic brain Gu, Y., Zheng, G., Xu, M., Li, Y., Chen, X., Zhu, W.,
injury. Inflammopharmacology 21, 309–320. Tong, Y., Chung, S.K., Liu, K.J., and Shen, J. (2012).
Freeman, J.M., Vining, E.P., Pillas, D.J., Pyzik, P.L., and Caveolin-1 regulates nitric oxide-mediated matrix
Casey, J.C. (1998). The efficacy of the ketogenic metalloproteinases activity and blood-brain bar-
diet-1998: a prospective evaluation of intervention rier permeability in focal cerebral ischemia and
in 150 children. Pediatrics 102, 1358–1363. reperfusion injury. J Neurochem 120, 147–156.
Freeman, J., Veggiotti, P., Lanzi, G., Tagliabue, A., Ha, S.N., Hochman, J., and Sheridan, R.P. (2007). Mini
and Perucca, E. (2006). The ketogenic diet:  from review on molecular modeling of P-glycoprotein
molecular mechanisms to clinical effects. Epilepsy (Pgp). Curr Top Med Chem 7, 1525–1529.
Res 68, 145–180. Hall, C.N., Reynell, C., Gesslein, B., Hamilton, N.B.,
Galea, I., Bechmann, I., and Perry, V.H. (2007). What Mishra, A., Sutherland, B.A., Farrell, F.M., Buchan,
is immune privilege (not)? Trends Immunol A.M., Lauritzen, M., and Attwell, D. (2014).
28, 12–18. Capillary pericytes regulate cerebral blood flow in
Gasior, M., Rogawski, M.A., and Hartman, A.L. (2006). health and disease. Nature 508, 55–60.
Neuroprotective and disease-modifying effects of Hamilton, J.A. (1998). Fatty acid transport: difficult or
the ketogenic diet. Behav Pharmacol 17, 431. easy? J Lipid Res 39, 467–481.
Ghosh, C., Gonzalez-Martinez, J., Hossain, M., Haseloff, R.F., Blasig, I.E., Bauer, H.C., and Bauer, H.
Cucullo, L., Fazio, V., Janigro, D., and Marchi, N. (2005). In search of the astrocytic factor (s) modu-
(2010). Pattern of P450 expression at the human lating blood-brain barrier functions in brain capil-
blood-brain barrier:  roles of epileptic condition lary endothelial cells in vitro. Cell Mol Neurobiol
and laminar flow. Epilepsia 51, 1408–1417. 25, 25–39.
Ghosh, C., Marchi, N., Desai, N.K., Puvenna, V., Hasselbalch, S.G., Knudsen, G.M., Jakobsen, J.,
Hossain, M., Gonzalez-Martinez, J., Alexopoulos, Hageman, L.P., Holm, S., and Paulson, O.B.
A.V., and Janigro, D. (2011a). Cellular localiza- (1994). Brain metabolism during short-term star-
tion and functional significance of CYP3A4 in the vation in humans. J Cereb Blood Flow Metab 14,
human epileptic brain. Epilepsia 52, 562–571. 125–131.
Ghosh, C., Puvenna, V., Gonzalez-Martinez, J., Hasselbalch, S.G., Madsen, P.L., Hageman, L.P., Olsen,
Janigro, D., and Marchi, N. (2011b). Blood-brain K.S., Justesen, N., Holm, S., and Paulson, O.B.
barrier P450 enzymes and multidrug transporters (1996). Changes in cerebral blood flow and carbo-
in drug resistance: a synergistic role in neurologi- hydrate metabolism during acute hyperketonemia.
cal diseases. Curr Drug Metab 12, 742–749. Am J Physiol Endocrinol Metab 270, E746–E751.
Ghosh, C., Hossain, M., Puvenna, V., Martinez- Hawkins, B.T., and Davis, T.P. (2005). The blood-brain
Gonzalez, J., Alexopolous, A., Janigro, D., and barrier/neurovascular unit in health and disease.
Marchi, N. (2013). Expression and functional Pharmacol Rev 57, 173–185.
relevance of UGT1A4 in a cohort of human drug- Hawkins, R.A., O’Kane, R.L., Simpson, I.A., and Via,
resistant epileptic brains. Epilepsia 54, 1562–1570. J.R. (2006). Structure of the blood-brain barrier
Gibson, C.L., Murphy, A.N., and Murphy, S.P. (2012). and its role in the transport of amino acids. J Nutr
Stroke outcome in the ketogenic state-systematic 136, 218S–226S.
review of the animal data. J Neurochem 123, 52–57. Hiraide, A., Katayama, M., Sugimoto, H., Yoshioka,
Gingrich, M.B., and Traynelis, S.F. (2000). Serine pro- T., and Sugimoto, T. (1991). Effect of 3-
teases and brain damage is there a link? Trends hydroxybutyrate on posttraumatic metabolism in
Neurosci 23, 399–407. man. Surgery 109, 176–181.
Glass, C.K., Saijo, K., Winner, B., Marchetto, M.C., and Hirase, T., Staddon, J.M., Saitou, M., Ando-Akatsuka,
Gage, F.H. (2010). Mechanisms underlying inflam- Y., Itoh, M., Furuse, M., Fujimoto, K., Tsukita, S.,
mation in neurodegeneration. Cell 140, 918–934. and Rubin, L. (1997). Occludin as a possible deter-
Gloor, S.M., Wachtel, M., Bolliger, M.F., Ishihara, H., minant of tight junction permeability in endothe-
Landmann, R., and Frei, K. (2001). Molecular and lial cells. J Cell Sci 110, 1603–1613.
302

302 section III: Ketogenic Diet in the Laboratory

Ho, C.F., Chan, K.W., Yeh, H.I., Kuo, J., Liu, H.J., and Levy, M., Thaiss, C.A., and Elinav, E. (2015). Taming
Wang, C.Y. (2013). Ketone bodies upregulate the inflammasome. Nat Med 21, 213–215.
endothelial connexin 43 (Cx43) gap junctions. Vet Liebner, S., Fischmann, A., Rascher, G., Duffner,
J 198, 696–701. F., Grote, E.H., Kalbacher, H., and Wolburg, H.
Hu, Z.G., Wang, H.D., Jin, W., and Yin, H.X. (2009a). (2000). Claudin-1 and claudin-5 expression and
Ketogenic diet reduces cytochrome c release and tight junction morphology are altered in blood
cellular apoptosis following traumatic brain injury vessels of human glioblastoma multiforme. Acta
in juvenile rats. Ann Clin Lab Sci 39, 76–83. Neuropathol 100, 323–331.
Hu, Z.G., Wang, H.D., Qiao, L., Yan, W., Tan, Q.F., Magistretti, P.J., and Pellerin, L. (1995). Cellular basis
and Yin, H.X. (2009b). The protective effect of the of brain energy metabolism and their relevance to
ketogenic diet on traumatic brain injury-induced brain imaging-evidence for a prominent role for
cell death in juvenile rats. Brain Inj 23, 459–465. astrocytes. Cereb Cortex 5, 301–306.
Huang, J., Upadhyay, U.M., and Tamargo, R.J. (2006). Marchi, N., Granata, T., Alexopoulos, A., and Janigro,
Inflammation in stroke and focal cerebral isch- D. (2012). The blood-brain barrier hypothesis in
emia. Surg Neurol 66, 232–245. drug resistant epilepsy. Brain 135, e211.
Huber, J.D., Egleton, R.D., and Davis, T.P. (2001). Marchi, N., Granata, T., and Janigro, D. (2014).
Molecular physiology and pathophysiology of Inflammatory pathways of seizure disorders.
tight junctions in the blood-brain barrier. Trends Trends Neurosci 37, 55–65.
Neurosci 24, 719–725. Marchi, N., Hallene, K.L., Kight, K.M., Cucullo, L.,
Ivens, S., Kaufer, D., Flores, L.P., Bechmann, I., Moddel, G., Bingaman, W., Dini, G., Vezzani, A.,
Zumsteg, D., Tomkins, O., Seiffert, E., Heinemann, and Janigro, D. (2004). Significance of MDR1 and
U., and Friedman, A. (2007). TGF-beta receptor- multiple drug resistance in refractory human epi-
mediated albumin uptake into astrocytes is leptic brain. BMC Med 2, 37.
involved in neocortical epileptogenesis. Brain 130, McAllister, M.S., Krizanac-Bengez, L., Macchia,
535–547. F., Naftalin, R.J., Pedley, K.C., Mayberg, M.R.,
Janigro, D. (1999). Blood-brain barrier, ion homeo- Marroni, M., Leaman, S., Stanness, K.A., and
stasis and epilepsy: possible implications towards Janigro, D. (2001). Mechanisms of glucose trans-
the understanding of ketogenic diet mechanisms. port at the blood-brain barrier: an in vitro study.
Epilepsy Res 37, 223–232. Brain Res 904, 20–30.
Janigro, D. (2012). Are you in or out? Leukocyte, ion, Mitchell, R.W., Edmundson, C.L., Miller, D.W., and
and neurotransmitter permeability across the Hatch, G.M. (2009). On the mechanism of oleate
epileptic blood-brain barrier. Epilepsia 53 (Suppl transport across human brain microvessel endo-
1), 26–34. thelial cells. J Neurochem 110, 1049–1057.
Janzer, R.C., and Raff, M.C. (1987). Astrocytes induce Mittapalli, R.K., Manda, V.K., Adkins, C.E.,
blood-brain barrier properties in endothelial cells. Geldenhuys, W.J., and Lockman, P.R. (2010).
Nature 325, 253–257. Exploiting nutrient transporters at the blood-brain
Jeong, S.M., Hahm, K.D., Shin, J.W., Leem, J.G., Lee, barrier to improve brain distribution of small mol-
C., and Han, S.M. (2006). Changes in magnesium ecules. Ther Deliv 1, 775–784.
concentration in the serum and cerebrospinal fluid Mizee, M.R., Wooldrik, D., Lakeman, K.A., van het
of neuropathic rats. Acta Anaesthesiol Scand 50, Hof, B., Drexhage, J.A., Geerts, D., Bugiani, M.,
211–216. Aronica, E., Mebius, R.E., and Prat, A. (2013).
Klepper, J. (2008). Glucose transporter deficiency Retinoic acid induces blood-brain barrier devel-
syndrome (GLUT1DS) and the ketogenic diet. opment. J Neurosci 33, 1660–1671.
Epilepsia 49, 46–49. Morita, K., Furuse, M., Fujimoto, K., and Tsukita, S.
Kniesel, U., and Wolburg, H. (2000). Tight junctions (1999). Claudine multigene family encoding four-
of the blood-brain barrier. Cell Mol Neurobiol transmembrane domain protein components of
20, 57–76. tight junction strands. PNAS 96, 511–516.
Lai, C.H., and Kuo, K.H. (2005). The critical compo- Morris, A.A.M. (2005). Cerebral ketone body metabo-
nent to establish in vitro BBB model:  pericyte. lism. J Inherit Metab Dis 28, 109–121.
Brain Res Rev 50, 258–265. Musa-Veloso, K., Likhodii, S.S., and Cunnane, S.C.
Lee, S., Chen, T.T., Barber, C.L., Jordan, M.C., (2002). Breath acetone is a reliable indicator of
Murdock, J., Desai, S., Ferrara, N., Nagy, A., Roos, ketosis in adults consuming ketogenic meals. Am J
K.P., and Iruela-Arispe, M.L. (2007). Autocrine Clin Nutr 76, 65–70.
VEGF signaling is required for vascular homeo- Nabbout, R., Vezzani, A., Dulac, O., and Chiron, C.
stasis. Cell 130, 691–703. (2011). Acute encephalopathy with inflammation-
Leino, R.L., Gerhart, D.Z., Duelli, R., Enerson, B.E., mediated status epilepticus. Lancet Neurol 10,
and Drewes, L.R. (2001). Diet-induced ketosis 99–108.
increases monocarboxylate transporter (MCT1) Nakagawa, S., Deli, M.A., Kawaguchi, H., Shimizudani,
levels in rat brain. Neurochem Int 38, 519–527. T., Shimono, T., Kittel, A., Tanaka, K., and Niwa,
303

Chapter 30: Effects of the Ketogenic Diet on the Blood-Brain Barrier 303

M. (2009). A new blood-brain barrier model using Ruskin, D.N., Kawamura, M., and Masino, S.A.
primary rat brain endothelial cells, pericytes and (2009). Reduced pain and inflammation in juve-
astrocytes. Neurochem Int 54, 253–263. nile and adult rats fed a ketogenic diet. PLoS One
Neuwelt, E.A., Bauer, B., Fahlke, C., Fricker, G., Iadecola, 4, e8349.
C., Janigro, D., Leybaert, L., Molnar, Z., O’Donnell, Salim, A., Hadjizacharia, P., Dubose, J., Brown, C.,
M.E., Povlishock, J.T., et al. (2011). Engaging neu- Inaba, K., Chan, L.S., and Margulies, D. (2009).
roscience to advance translational research in brain Persistent hyperglycemia in severe traumatic brain
barrier biology. Nat Rev Neurosci 12, 169–182. injury: an independent predictor of outcome. Am
Nischwitz, V., Berthele, A., and Michalke, B. (2008). Surg 75, 25–29.
Speciation analysis of selected metals and deter- Saunders, N.R., Dreifuss, J.J., Dziegielewska, K.M.,
mination of their total contents in paired serum Johansson, P.A., Habgood, M.D., Mllgrd, K., and
and cerebrospinal fluid samples:  an approach to Bauer, H.C. (2014). The rights and wrongs of
investigate the permeability of the human blood- blood-brain barrier permeability studies:  a walk
cerebrospinal fluid-barrier. Anal Chim Acta 627, through 100 years of history. Front Neurosci 8.
258–269. Shepro, D., and Morel, N.M. (1993). Pericyte physiol-
Ohtsuki, S., Hirayama, M., Ito, S., Uchida, Y., Tachikawa, ogy. FASEB Journal 7, 1031–1038.
M., and Terasaki, T. (2014). Quantitative targeted Shimizu, F., Sano, Y., Maeda, T., Abe, M., Nakayama,
proteomics for understanding the blood-brain H., Takahashi, R., Ueda, M., Ohtsuki, S., Terasaki,
barrier: towards pharmacoproteomics. Expert Rev T., and Obinata, M. (2008). Peripheral Nerve peri-
Proteomics 11, 303–313. cytes originating from the blood-nerve barrier
Pan, J.W., Rothman, D.L., Behar, K.L., Stein, D.T., and expresses tight junctional molecules and trans-
Hetherington, H.P. (2000). Human brain [bgr]- porters as barrier-forming cells. J Cell Physiol 217,
hydroxybutyrate and lactate increase in fasting- 388–399.
induced ketosis. J Cereb Blood Flow Metab 20, Shue, E.H., Carson-Walter, E.B., Liu, Y., Winans,
1502–1507. B.N., Ali, Z.S., Chen, J., and Walter, K.A. (2008).
Pan, J.W., Telang, F.W., Lee, J.H., de Graaf, R.A., Plasmalemmal vesicle associated protein-1 (PV-1)
Rothman, D.L., Stein, D.T., and Hetherington, H.P. is a marker of blood-brain barrier disruption in
(2001). Measurement of &beta;-hydroxybutyrate rodent models. BMC Neurosci 9, 29.
in acute hyperketonemia in human brain. J Somjen, G.G. (2004). Ions in the brain: normal func-
Neurochem 79, 539–544. tion, seizures, and stroke (New York, USA: Oxford
Pierre, K., and Pellerin, L. (2005). Monocarboxylate University Press).
transporters in the central nervous system:  dis- Sorokin, L. (2010). The impact of the extracellular
tribution, regulation and function. J Neurochem matrix on inflammation. Nat Rev Immunol 10,
94, 1–14. 712–723.
Porta, N., Valle, L., Boutry, E., Fontaine, M., Dessein, Stanness, K.A., Guatteo, E., and Janigro, D. (1996).
A.F., Joriot, S., Cuisset, J.M., Cuvellier, J.C., and A dynamic model of the blood-brain barrier “in
Auvin, S. (2009). Comparison of seizure reduc- vitro.” Neurotoxicology 17, 481–496.
tion and serum fatty acid levels after receiving the Stanness, K.A., Westrum, L.E., Fornaciari, E.,
ketogenic and modified Atkins diet. Seizure 18, Mascagni, P., Nelson, J.A., Stenglein, S.G., Myers,
359–364. T., and Janigro, D. (1997). Morphological and
Prat, A., Biernacki, K., Wosik, K., and Antel, J.P. (2001). functional characterization of an in vitro blood-
Glial cell influence on the human blood-brain bar- brain barrier model. Brain Res 771, 329–342.
rier. Glia 36, 145–155. Sullivan, P.G., Rippy, N.A., Dorenbos, K., Concepcion,
Prins, M.L. (2008). Cerebral metabolic adaptation R.C., Agarwal, A.K., and Rho, J.M. (2004). The
and ketone metabolism after brain injury. J Cereb ketogenic diet increases mitochondrial uncou-
Blood Flow Metab. 28, 1–16. pling protein levels and activity. Ann Neurol 55,
Prins, M.L., and Giza, C.C. (2006). Induction of mono- 576–580.
carboxylate transporter 2 expression and ketone Takimoto, M., and Hamada, T. (2014). Acute exer-
transport following traumatic brain injury in juve- cise increases brain region-specific expression of
nile and adult rats. Dev Neurosci 28, 447–456. MCT1, MCT2, MCT4, GLUT1, and COX IV pro-
Prins, M.L., and Matsumoto, J.H. (2014). The col- teins. J Appl Physiol 116, 1238–1250.
lective therapeutic potential of cerebral ketone Tewes, B.J., and Galla, H.J. (2001). Lipid polarity in
metabolism in traumatic brain injury. J Lipid Res brain capillary endothelial cells. Endothelium 8,
55, 2450–2457. 207–220.
Rubin, L.L., Hall, D.E., Porter, S., Barbu, K., Cannon, Thiebaut, F., Tsuruo, T., Hamada, H., Gottesman,
C., Horner, H.C., Janatpour, M., Liaw, C.W., M.M., Pastan, I., and Willingham, M.C. (1987).
Manning, K., Morales, J., et al. (1991). A cell cul- Cellular localization of the multidrug resistance
ture model of the blood-brain barrier. J Cell Biol gene product in normal human tissues. PNAS 84,
115, 1725–1735. 7735–7738.
304

304 section III: Ketogenic Diet in the Laboratory

Tieu, K., Perier, C., Caspersen, C., Teismann, P., Wu, Wosik, K., Cayrol, R., Dodelet-Devillers, A.,
D.C., Yan, S.D., Naini, A., Vila, M., Jackson- Berthelet, F., Berna 32rd, M., Moumdjian, R.,
Lewis, V., and Ramasamy, R. (2003). D-beta- Bouthillier, A., Reudelhuber, T.L., and Prat, A.
hydroxybutyrate rescues mitochondrial respiration (2007). Angiotensin II controls occludin function
and mitigates features of Parkinson disease. J Clin and is required for blood-brain barrier mainte-
Invest 112, 892. nance: relevance to multiple sclerosis. J Neurosci
Tomkins, O., Shelef, I., Kaizerman, I., Eliushin, 27, 9032–9042.
A., Afawi, Z., Misk, A., Gidon, M., Cohen, A., Yasui, T., Hirose, J., Tsutsumi, S., Nakamura, K.,
Zumsteg, D., and Friedman, A. (2008). Blood- Aburatani, H., and Tanaka, S. (2011). Epigenetic
brain barrier disruption in post-traumatic epi- regulation of osteoclast differentiation:  possible
lepsy. J Neurol Neurosurg Psychiatry 79, 774–777. involvement of Jmjd3 in the histone demethylation
Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A., of Nfatc1. J Bone Miner Res 26, 2665–2671.
and Cahill, G.F. (2001). Ketone bodies, potential Ziegler, D.R., Ribeiro, L.C., Hagenn, M., Arajo,
therapeutic uses. IUBMB Life 51, 241–247. E., Torres, I.L., Gottfried, C., Netto, C.A., and
White, H., and Venkatesh, B. (2011). Clinical Gonalves, C.A. (2003). Ketogenic diet increases
review: ketones and brain injury. Crit Care 15, 219. glutathione peroxidase activity in rat hippocam-
Wolburg, H., Noell, S., Mack, A., Wolburg-Buchholz, pus. Neurochem Res 28, 1793–1797.
K., and Fallier-Becker, P. (2009). Brain endothelial Zlokovic, B.V. (2008). The blood-brain barrier in
cells and the glio-vascular complex. Cell Tissue health and chronic neurodegenerative disorders.
Res 335, 75–96. Neuron 57, 178–201.
305

SECTION IV
Ketone-Based Metabolism

General Health and Metabolic Alternatives

D O M I N I C P. D ’ AG O S T I N O , P H D , S E C T I O N E D I T O R
306
307

31
Overview of Ketone-Based Metabolism
General Health and Metabolic Alternatives

D O M I N I C P. D ’ AG O S T I N O , P H D

T he ketone bodies β-hydroxybutyrate (BHB)


and acetoacetate (AcAc) are produced
from fatty acids in the liver and serve as alterna-
metabolites as alternative fuels and potent signal-
ing molecules.
Induction of hyperketonemia through fasting
tive energy sources for the brain, heart, skeletal or the KD is known to produce acute and chronic
muscle, and peripheral tissues during prolonged changes in metabolic physiology and molecu-
fasting, calorie restriction, strenuous exercise, lar signaling pathways that provide therapeutic
or adherence to a high-fat, low-carbohydrate effects in varied disease states. Metabolic-based
ketogenic diet (KD) (Cahill and Veech, 2003). mechanisms of ketone therapies include an eleva-
Ketones have historically been labeled as abnor- tion of blood ketones and associated anaplerosis
mal metabolic byproducts of a pathological state with simultaneous suppression of blood glucose,
(VanItallie and Nufert, 2003), particularly due to increased insulin sensitivity, improved mitochon-
their association with diabetic ketoacidosis (DKA) drial efficiency, and suppressed oxidative stress
in uncontrolled type 1 diabetics, but emerging and inflammation. The reported benefits of hyper-
interest has focused on nutritional ketosis as a ketonemia and similar metabolic alternatives
powerful metabolic therapy for general health and have generated significant interest in the science
a growing number of medical conditions in addi- and application of implementing strategies for
tion to drug-resistant epilepsy, where its use is well inducing and sustaining blood levels of specific
established (Stafstrom and Rho, 2012). Nutritional metabolites (Kesl et  al., 2016). Most exogenous
ketosis produces a nonpathological hyperketone- ketone supplements and engineered anapleurotic
mia resulting from decreased glucose availability, agents are currently under investigation for under-
lower insulin, and increased fat oxidation; how- standing their potential benefits for both healthy
ever, long-term maintenance of the KD can be and diseased individuals alike. It is likely that
difficult for some and requires strict adherence. most, if not all, of the conditions that are known
The restrictive nature of the KD has limited the to benefit from the KD would receive some ben-
clinical applicability of therapeutic ketosis due to efit from exogenous ketone supplementation by
practical considerations. Emerging data suggests elevating blood ketones and lowering blood glu-
that many of the benefits of the KD are mecha- cose (Cahill and Veech, 2003). Currently the most
nistically attributable to the ketone bodies or studied type of exogenous ketone would be ketone
specific medium chain triglycerides (MCTs), and esters, which induce a dose-dependent hyperke-
this has motivated investigators to develop strate- tonemia (1–7mM) in rats, mice, dogs, pigs, and
gies to further augment the efficacy of the KD or humans (Birkhahn and Border, 1978; Clarke et al.,
use metabolic-based supplements to circumvent 2012; Desrochers et al., 1995; Pascual et al., 2014).
the need for dietary restriction to improve com- A  recent case study of Alzheimer’s disease high-
pliance and the maintenance of this therapeutic lights its application in humans (Newport et  al.,
state (Newman and Verdin, 2014; Veech, 2004). 2015). There are a growing number of promising
This section, “Ketone-Based Metabolism: General metabolic alternatives to ketone esters, and many
Health and Metabolic Alternatives” includes chap- of these agents and formulas are being evaluated
ters that discuss the expanding medical and per- for their therapeutic efficacy, practical application
formance applications of nutritional ketosis and and potential synergy with the KD (Borges and
the emerging science of ketones and other related Sonnewald, 2012; Kesl et  al., 2016). Importantly,
308

308 section IV: Ketone-Based Metabolism

ketone supplementation and metabolic alterna- a novel metabolic-independent mechanism of


tives provide a tool for achieving ketosis in patients MCTs, which were previously thought only to be
who are unable, unwilling, or uninterested in con- nutritional support enhancing ketogenesis. Also
suming a low-carbohydrate or ketogenic diet. included in this section is a chapter by Borges
Included in this section are chapters that dis- (Chapter  34) on triheptanoin, the triglyceride of
cuss conditions for which ketones and metabolic heptanoate (C7 fatty acid), a metabolic alternative
alternatives have the most potential, including to the KD that is being used to treat patients with
seizure disorders, glucose transporter type 1 defi- rare genetic metabolic disorders (Pascual et  al.,
ciency syndrome (GLUT1DS), Alzheimer’s disease 2014). The compelling animal work associated
(AD), brain and metastatic cancer, insulin resis- with triheptanoin shows that metabolic altera-
tance and type 2 diabetes mellitus (T2DM), weight tions found in brains of rodent seizure models
loss, and performance. GLUT1DS is a rare genetic can be restored by this compound, which appears
disorder caused by a mutation in the SLC2A1 to cross the blood-brain barrier (Borges and
gene, which encodes the glucose transporter pro- Sonnewald, 2012). Preclinical and clinical studies
tein type 1. Patients with GLUT1DS have impaired indicate that triheptanoin is beneficial in numer-
glucose transport into the brain and other tissues, ous neurological and neuro-muscular disorders,
and thus alternative energy substrates in the form making it a promising treatment for a variety of
of ketones or other metabolic alternatives offer a clinical conditions (Borges and Sonnewald, 2012).
means for the metabolic management of this dis- Very little attention has been given to the potential
order (Pascual et al., 2014). A known hallmark of function of protein and specific amino acids in the
AD is impaired brain glucose metabolism, which ketogenic diet. Hartman (Chapter 35) focuses on
is associated with neurodegeneration and rapid this topic by discussing the antiseizure potential
progression (Cunnane et  al., 2016). Preclinical of select amino acids and the cellular and molecu-
data demonstrate that ketones protect against AD lar mechanisms associated with their effects. The
development and slow its progression (Kashiwaya chapter by Stafstrom et al. (Chapter 36) discusses
et al., 2013). Cancer is another disorder potentially inhibition of glycolysis as one way that the keto-
treated with nutritional ketosis due a metabolic genic diet suppresses seizures and how 2-deoxy-
phenotype that is highly glycolytic, has abnormal D-glucose (2DG) is an anti-glycolytic tool that
mitochondrial structure and function, and has a has anti-seizure effects in numerous seizure mod-
deficiency in many ketolytic enzymes (Seyfried els. Westman et al. (Chapter 37) review the ratio-
and Shelton, 2010). Indeed, numerous studies have nale and recent clinical research supporting the
shown that cancer cells lack the capacity to metab- use of a low-carbohydrate, ketogenic diet in indi-
olize BHB for energy, and thus glucose restriction viduals with obesity and T2DM. The last chapter
can be lethal (Maurer et  al., 2011). This section in this section, Hyde et  al. (Chapter  38) discuss
discusses the research showing that ketone ester the many health benefits of nutritional ketosis
administration simultaneously decreases blood that are relevant to athletes, including enhanced
glucose and insulin concentrations, suggesting fat oxidation, reduced inflammation, reduction in
that exogenous ketone supplementation could be blood lipids, and suppression of oxidative stress.
used for insulin resistance and T2DM (Kashiwaya Zupec-Kania et al. (Chapter 39), reviews the gen-
et  al., 1997). The beneficial effects of ketones in esis and ongoing efforts of the Charlie Foundation
T2DM may be via direct regulation of HDAC- and Matthew’s Friends in promoting ketogenic
dependent glucose metabolism and by inducing therapies for epilepsy and more recently as an
resistance to oxidative stress, which helps restore adjuvant for the metabolic management of can-
insulin sensitivity. cer and other neurological and neurodegenerative
Included in this section is a chapter by Poff disorders. We hope the chapters in this section
et. al (Chapter  32) that describes the develop- will help the reader gain an appreciation for the
ment and testing of exogenous ketone supple- science and emerging applications of nutritional
ments, the diseases they are being investigated ketosis, ketone supplementation, and other meta-
for use in, and the potential mechanisms of bolic alternatives that will inevitably be utilized
action of their therapeutic efficacy. The chapter for the treatment and prevention for a broad
by Walker and Williams (Chapter  33) gives an range of disease states.
overview of the acute in vitro and in vivo stud-
ies of the MCT, decanoic acid, which imparts REFERENCES
specific signaling effects by directly inhibiting Birkhahn, R.H., and Border, J.R. (1978). Intravenous
AMPA receptors. Thus, this data demonstrates feeding of the rat with short chain fatty acid
309

Chapter 31: Overview of Ketone-Based Metabolism 309

esters:  II. Monoacetoacetin. Am J Clin Nutr 31, Kesl, S.L., Poff, A.M., Ward, N.P., Fiorelli, T.N., Ari,
436–441. C., Van Putten, A.J., Sherwood, J.W., Arnold, P.,
Borges, K., and Sonnewald, U. (2012). Triheptanoin: a and D’Agostino, D.P. (2016). Effects of exogenous
medium chain triglyceride with odd chain fatty ketone supplementation on blood ketone, glucose,
acids: a new anaplerotic anticonvulsant treatment? triglyceride, and lipoprotein levels in Sprague-
Epilepsy Res 100, 239–244, Dawley rats. Nutr Metab (Lond) 13, 9.
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? Maurer, G., Brucker, D., Bähr, O., Harter, P., Hattingen,
Good medicine? Trans Am Clin Climatol Assoc E., Walenta, S., Mueller-Klieser, W., Steinbach, J.,
114, 149–161; discussion 162–143. and Rieger, J. (2011). Differential utilization of
Clarke, K., Tchabanenko, K., Pawlosky, R., Carter, ketone bodies by neurons and glioma cell lines: a
E., Todd King, M., Musa-Veloso, K., Ho, M., rationale for ketogenic diet as experimental glioma
Roberts, A., Robertson, J., Vanitallie, T.B., et  al. therapy. BMC Cancer 11, 315.
(2012). Kinetics, safety and tolerability of (R)-3- Newman, J.C., and Verdin, E. (2014). Ketone bodies as
hydroxybutyl (R)-3-hydroxybutyrate in healthy signaling metabolites. Trends Endocrinol Metabol
adult subjects. Regul Toxicol Pharmacol 63, 25, 42–52.
401–408, Newport, M.T., VanItallie, T.B., Kashiwaya, Y., King,
Cunnane, S.C., Courchesne-Loyer, A., St-Pierre, M.T., and Veech, R.L. (2015). A new way to pro-
V., Vandenberghe, C., Pierotti, T., Fortier, M., duce hyperketonemia: use of ketone ester in a case
Croteau, E., and Castellano, C.A. (2016). Can of Alzheimer’s disease. Alzheimers Dement11,
ketones compensate for deteriorating brain glu- 99–103.
cose uptake during aging? Implications for the Pascual, J.M., Liu, P., Mao, D., Kelly, D.I., Hernandez,
risk and treatment of Alzheimer’s disease. Ann NY A., Sheng, M., Good, L.B., Ma, Q., Marin-Valencia,
Acad Sci,Mar;1367(1):12–20. I., Zhang, X., et  al. (2014). Triheptanoin for glu-
Desrochers, S., Dubreuil, P., Brunet, J., Jette, M., cose transporter type I deficiency (G1D): modula-
David, F., Landau, B.R., and Brunengraber, H. tion of human ictogenesis, cerebral metabolic rate,
(1995). Metabolism of (R,S)-1,3-butanediol ace- and cognitive indices by a food supplement. JAMA
toacetate esters, potential parenteral and enteral Neurol 71, 1255–1265.
nutrients in conscious pigs. Am J Physiol 268, Seyfried, T.N., and Shelton, L.M. (2010). Cancer as a
E660–667. metabolic disease. Nutr Metab (Lond) 7, 7.
Kashiwaya, Y., King, M.T., and Veech, R.L. (1997). Stafstrom, C.E., and Rho, J.M. (2012). The ketogenic
Substrate signaling by insulin:  a ketone bodies diet as a treatment paradigm for diverse neuro-
ratio mimics insulin action in heart. Am J Cardiol logical disorders. Front Pharmacol 3, 59.
80, 50A–64A. VanItallie, T.B., and Nufert, T.H. (2003). Ketones: metab-
Kashiwaya, Y., Bergman, C., Lee, J.H., Wan, R., King, olism’s ugly duckling. Nutr Rev 61, 327–341.
M.T., Mughal, M.R., Okun, E., Clarke, K., Mattson, Veech, R.L. (2004). The therapeutic implications of
M.P., and Veech, R.L. (2013). A ketone ester diet ketone bodies:  the effects of ketone bodies in
exhibits anxiolytic and cognition-sparing proper- pathological conditions:  ketosis, ketogenic diet,
ties, and lessens amyloid and tau pathologies in a redox states, insulin resistance, and mitochondrial
mouse model of Alzheimer’s disease. Neurobiol metabolism. Prostaglandins Leukot Essent Fatty
Aging 34, 1530–1539. Acids 70, 309–319.
310

32
Ketone Supplementation for Health and Disease
A N G E L A M . P O F F, P H D , S H A N N O N L . K E S L , P H D ,
A N D D O M I N I C P. D ’ AG O S T I N O , P H D

INDUCING THERAPEUTIC and esters that have been shown to elevate blood
K E TO S I S W I T H   K E TO N E ketone levels independent of caloric or carbo-
S U P P L E M E N TAT I O N hydrate restriction. The investigation of natural
Ketones are typically produced in the liver only and synthetically derived ketogenic precursors to
under certain physiological conditions associated establish nutritional ketosis without the need for
with the suppression of the hormone insulin: star- dietary restriction has revealed that each formu-
vation, fasting, calorie restriction, prolonged lation has distinct properties in terms of extent
exercise, or during the consumption of a high- and duration of ketosis as well as metabolic sig-
fat, low-carbohydrate ketogenic diet (KD). The naling properties, including anticonvulsant effects
restrictive nature of these states has limited the (D’Agostino et  al., 2013; Kesl et  al., 2015a; Kesl
clinical applicability of therapeutic ketosis due to et al., 2016). Most of the developed ketone supple-
practical considerations. In an effort to circumvent ments are currently under investigation for safety
this dilemma, researchers have recently developed and efficacy in a number of disease states.
a number of exogenous ketogenic supplements—
ketogenic precursors that are metabolized Medium Chain Triglycerides
to produce a dose-dependent elevation of β- Medium chain triglycerides (MCTs) contain a
hydroxybutyrate (βHB) and acetoacetate (AcAc) glycerol backbone esterified to medium-chain
in the blood (Clarke et al., 2012b; D’Agostino et al., fatty acids (MCFAs), which are fatty acids with
2013; Kesl et  al., 2016; Puchowicz et  al., 2000). hydrocarbon side chains 6 to 12 carbons in length.
These ketone supplements allow for a rapid and The MCFAs include caproic acid (C6:0, hexanoic
controlled induction of physiologic ketosis with- acid), caprylic acid (C8:0, octanoic acid), cap-
out the need for severe dietary restrictions. Since ric acid (C10:0, decanoic acid), and lauric acid
many of the benefits of ketosis are mechanistically (C12:0, dodecanoic acid). The MCTs are natu-
attributable to the ketone bodies, it is possible that rally found in coconut oil (~60%), palm kernel oil
exogenous ketone supplementation could mimic (~30%), cheese (7.3%), whole milk (6.9%), but-
the therapeutic efficacy of the KD for certain dis- ter (6.8%), and full-fat yogurt (6.6%) (Karen and
orders. In addition, the adjuvant use of exogenous Welma, 2015; Koji and Truyoshi, 2010). Compared
ketone supplements for patients consuming a KD to long-chain fatty acids (LCFAs), MCFAs have a
could provide a method for faster adaptation, eas- lower melting point, smaller molecule size, and are
ier sustainment of ketosis, and perhaps help return less calorically dense (8.3 calories per gram ver-
a sense of normalcy to patients and their caregiv- sus 9.2). These distinct physiochemical properties
ers. Furthermore, in some scenarios, an exogenous allow MCTs to be absorbed directly into the blood-
ketone-supplemented KD may be more effective stream through the hepatic portal vein without the
than KD or ketone supplementation alone. need for bile or pancreatic enzymes for degrada-
tion. Additionally, MCTs do not require carnitine
DEVELOPMENT AND to enter the mitochondria, but rather quickly cross
T E S T I N G O F   K E TO N E the mitochondrial matrix, where they are metabo-
SUPPLEMENTS lized to acetyl co-A and subsequently to ketone
There are numerous sources of ketones and keto- bodies. Thus, they are easily and rapidly digested,
genic precursors being developed and tested, transferred to the liver, and used for energy rather
including medium chain triglycerides, diols, salts, than stored as fat. In comparison, LCFAs require
311

Chapter 32: Ketone Supplementation for Health and Disease 311

re-esterification in the small intestine, transport of certain polyester plasticizers, and a humec-
by chylomicrons via the lymphatic and vascular tant for cosmetics, and has been considered as a
systems, and oxidation in the liver for energy or synthetic food for long-duration space missions
storage. The MCTs are metabolized as rapidly as (Budavari, 1989; Dymsza, 1975; Falbe et al., 1985).
glucose but have roughly twice the energy density When ingested orally, BD is metabolized by the
(Bell et  al., 1997). In the early 1980s, Dr.  Vigen liver via alcohol dehydrogenase (ADH) to β-
K. Babayan of the Nutrition Laboratory at Harvard hydroxybutyraldehyde, which is rapidly oxidized
University developed a method to produce MCTs to βHB by aldehyde dehydrogenase (Tate et  al.,
in large quantities (Bach and Babayan, 1982). 1971). Further, BD contributes approximately 6
These commercialized MCTs are acquired through kcal/g of energy and can produce dose-dependent
lipid fractionation from natural fats such as coco- millimolar concentrations of ketones in the blood
nut oil and milk and are predominantly composed in a ratio of 6:1 of βHB to AcAc (D’Agostino et al.,
of C:8 and C:10 MCTs (Babayan, 1987; Hashim 2013; Desrochers et  al., 1992; Drackley et  al.,
and Tantibhedyangkul, 1987). 1990; Tobin et  al., 1972). Extensive toxicology
Since the 1970s, MCTs have been used in a studies have concluded that BD is safe with very
modification of the classical KD as an alternative few adverse health effects in humans or animals
fat source (Huttenlocher et  al., 1971). The keto- (Dymsza, 1975; Hess et al, 1981; Opitz, 1958; Scala
genic properties of MCTs allow patients to eat less and Paynter, 1967).
total fat in their diet and include more carbohy- In a 28-day study, a daily 5-g/kg dose of BD
drate and protein without sacrificing their nutri- administered via intragastric gavage in Sprague-
tional ketosis. Although MCTs have the potential Dawley rats showed a significant elevation of
to be an efficient and beneficial ketogenic fat, blood ketones (1mM βHB, <30 min) without an
they are currently limited in clinical usage due to effect on blood glucose, triglyceride, or lipoprotein
gastrointestinal (GI) side effects stimulated by levels (Kesl et  al., 2016). Recently, BD has been
the large dose needed to induce ketonemia (typi- investigated as a backbone of ketone mono- and
cally greater than 40 g/day) (Bell et al., 1997). The diesters, which are discussed later in this chapter.
original modified KD with MCT allowed 60% of
its energy to be derived from MCTs; however, this Ketone Salts
caused GI distress in some children (Huttenlocher, Originally, researchers attempted to administer
1976; Mak et  al., 1999; Sills et  al., 1986; Trauner, βHB or AcAc in their free acid forms; however,
1985). For this reason, an additional modified this was shown to be too expensive and ineffec-
KD with MCT was developed using only 30% of tive at producing sustained ketosis. Subsequently,
its energy from MCTs; however, it induced much it was suggested to buffer the free acid of βHB
lower levels of ketosis (Elizabeth et al., 2009; Ruby with sodium, but this causes potentially harmful
et  al., 1989). In a recent study, juvenile Sprague- sodium overload and mineral imbalance at thera-
Dawley rats were given a daily intragastric bolus peutic levels of ketosis, and is it has been shown
of MCT oil (10 g/kg) which rapidly elevated blood that βHB alone is largely ineffective at preventing
βHB levels (4mM βHB, <30 min) and remained seizures in animal models (Bough and Rho, 2007).
significantly elevated for up to 12 hours (Kesl A  study showed that oral administration of Na+/
et al., 2016). In addition to their metabolic effects, βHB in doses from 80 to 900 mg/kg/day elevated
emerging data suggests that octanoic acid (C8) has blood ketone levels to 0.19–0.36 mM, which was
specific anticonvulsant and neuroprotective prop- therapeutic in children with acyl CoA dehydro-
erties independent of ketones (Chang et al., 2015). genase deficiency (Hove et  al, 2003). However,
Research and development is currently being con- for a 70-kg man to achieve these levels of ketosis
ducted to formulate MCT oil into a powder with would require ingesting between 5.6 and 6.3 g/day.
soluble fiber compounds, which may delay gastric Considering the potential effects of such a large
emptying and enhance absorption to improve tol- sodium load, the costs of the administration of Na+/
erability and ketogenic potential. βHB salts to achieve ketosis made this approach
unrealistic (Veech, 2004).
1,3-Butanediol More recently, ketone salts with a balanced
1,3-Butanediol (BD; also known as 1,3-butylene electrolyte formulation to prevent sodium over-
glycol) is an FDA-approved organic di-alcohol load have been developed and tested and can
(diol), naturally present in some species of pepper include potassium, calcium, magnesium, lithium,
(Capsicum annuum), and is used as a food flavor- arginine, lysine, histidine, ornithine, creatine,
ing solvent, an intermediate in the manufacture agmatine, or citrulline. Maintaining a balanced
312

312 section IV: Ketone-Based Metabolism

electrolyte ratio should help offset any potential Formulating in this way allows for rapid and sus-
adverse effects of sodium on blood pressure. It is tained elevation of ketosis by delivering exogenous
speculated that this formulation will be especially ketones while simultaneously stimulating endog-
beneficial for elderly patients most susceptible enous ketogenesis with MCTs. In addition, the
to sodium-induced hypertension (Veech, 2004). combination formulation allows for a lower dosing
Furthermore, when first attempting to follow a of the components as compared to administering
KD, many people experience headaches and leth- the individual compounds, thus reducing poten-
argy during the initial stages of “keto-adaptation,” tial for side effects (gastric hyperosmolality) and
a term that describes the adaptive changes in resulting in a distinct blood ketone profile that is
metabolic physiology associated with transition- sustained over a longer period of time (D’Agostino
ing more toward fat and ketone metabolism. The et al., 2014).
symptoms are largely a result of reduced glucose In a 28-day study, the combination of a 50%
availability to the brain and a transient depletion Na+/K+ βHB salt mixed in a 1:1 solution with
of minerals, especially sodium, potassium, and MCT oil (BMS+MCT) significantly elevated and
magnesium in the plasma (Zhang et  al., 2013b). sustained blood ketone levels and reduced blood
These symptoms can be attenuated or reversed glucose levels in a dose-dependent manner (Kesl
with sufficient supplementation of sodium, potas- et al., 2016). Additionally, the study demonstrated
sium, calcium, and magnesium, and thus a ketone a significant correlation between elevated blood
supplement that delivers ketones with these elec- ketone levels and reduced blood glucose levels post
trolytes would be favorable. intragastric gavage administration with no effect
Several mineral ketone salt combinations are on blood triglyceride or lipoprotein levels. A note-
currently being tested for safety and efficacy. In a worthy observation from the study revealed that
recent study, neither a 5-g/kg nor a 10-g/kg intra- in rats, MCT caused a rise in ketones (>3mM) that
gastric gavage of Na+/K+ βHB salts significantly exceeds what is possible in humans due to gastro-
elevated blood ketone levels or reduced blood glu- intestinal intolerance. This is likely due to the fact
cose levels in juvenile Sprague-Dawley rats (Kesl that rats have a higher rates of absorption for MCT
et  al., 2016). However, there appears to be inter- and higher rates of hepatic fat metabolism, which
species variability in absorption, as in a recent case stimulates greater ketone production. Considering
study a 100-kg male subject was administered a these results it is important to take into account
4% Na+/K+ βHB salt solution (containing 11 grams interspecies variability in the metabolic response to
of sodium and 7.1 grams βHB) and his plasma ketone supplements, which will need to be further
levels of βHB were significantly elevated at 30–60 characterized to fully understand how we could
minutes after administration. After receiving the extrapolate findings and translate into human dos-
same dose for 3  days, the patient had sustained ing equivalents. In the rats, the BMS+MCT sup-
elevated blood ketone levels from 15 to 120 min- plement elevated blood ketones similar to that of
utes after administration. Similar results were seen MCT alone; however, the gastric side effects were
in a 70-kg male who fasted 3 days prior to admin- not observed, suggesting a potential method for
istration of the ketone salt supplement (D’Agostino avoiding this unwanted adverse effect (Kesl et al.,
et al., 2014). In a 15-week study, Sprague-Dawley 2016). In a case study, a 100-kg male was admin-
rats were administered Na+/Ca+2 ketone salt 20% istered a combination of a 4% Na+/K+ βHB salt
by weight (~25 g/kg/day) in their food fed ad libi- solution (containing 11 grams of sodium and 7.1
tum. The Na+/Ca+2 βHB–supplemented rats exhib- grams βHB) + 20 mL MCT oil. This combination
ited sustained elevated blood ketone levels (1 mM demonstrated higher elevation of blood ketone
βHB) at 1, 4, 8, 10, and 13 weeks of chronic feed- levels than either βHB salts or MCT oil alone,
ing, which did not affect blood glucose levels com- starting at 15 minutes post consumption and last-
pared with controls (Poff et al., 2016). ing for 4 hours. Similar results were observed in
a 70-kg male who fasted 3  days prior to admin-
Combination βHB Mineral Salt istration; however elevated blood ketone levels
and Medium Chain Triglyceride were observed sooner after supplementation and
Considering the variety of ketogenic precursors were sustained for a considerably longer time after
available, researchers are investigating unique administration (8 hours) (D’Agostino et al., 2015).
combinations of the individual supplements in This effect was accompanied by a reduction in
hopes of optimizing their benefits. A  combina- blood glucose, and a lower starting blood glucose
tion of βHB mineral salts (BMS) and MCT oil has concentration on each subsequent day of supple-
been administered in ratios of 1:1 to 1:2 mixtures. mentation (D’Agostino et al., 2014). In a 15-week
313

Chapter 32: Ketone Supplementation for Health and Disease 313

study, Sprague-Dawley rats were administered case study of Alzheimer’s disease (AD; Clarke et al.,
a 1:1 mixture of Na+/Ca+2 ketone salt + MCT oil 2012a; Clarke et al., 2012b; Newport et al., 2015).
(20% by weight, ~25 g/kg/day) in their food fed ad Recently, the ketone ester R,S-1,3-butanediol ace-
libitum. The combination-supplemented rats had toacetate diester (BD-AcAc2), given as an intragas-
significantly sustained and elevated blood ketone tric gavage, elevated both AcAc and βHB blood
levels at weeks 3, 4, 8, 10, and 13 without signifi- levels to >3mM in rats (D’Agostino et  al., 2013).
cantly affecting blood glucose levels during the The induction of therapeutic ketosis was rapid
study (Kesl et al., 2014). (within 30 minutes) and sustained at high levels
for over 4 hours. In a 28-day study, a daily intra-
Ketone Esters gastric gavage (5 g/kg body weight) of BD-AcAc2
Researchers have developed and investigated sev- induced significantly elevated blood ketone lev-
eral synthetic ketone mono- and di-esters to induce els and significantly reduced blood glucose levels
nutritional ketosis independent of dietary calorie without significantly altering blood triglyceride or
or carbohydrate restriction. When ketone esters lipoprotein levels (Kesl et al., 2016). In a 15-week
are administered, gastric esterases liberate ketones chronic feeding study, the BD-AcAc2 was admin-
(βHB and AcAc) as a free acid from a backbone istered to Sprague-Dawley rats in a low dose (5%
molecule, which varies depending on the specific food weight, 10 g/kg/day) (LKE) and a high dose
formulation, but is favorably a ketogenic precur- (20% food weight, 25 g/kg/day) (HKE) ad libitum.
sor such as BD. As previously discussed, BD is Both doses significantly elevated blood ketone lev-
subsequently metabolized by the liver to produce els without reducing blood glucose levels. Serum
βHB (D’Agostino et  al., 2013). Thus, the ketone clinical chemistry of both LKE and HKE did not
esters currently available are unique among the reveal any alterations in markers of kidney and
aforementioned ketone supplements in that they liver function compared with rats fed standard
can directly elevate ketones and supply ketogenic chow (Poff et al., 2016).
precursors that can further sustain ketogenesis.
Additionally, synthetically derived ketone esters P OT E N T I A L T H E R A P E U T I C
are currently the most potent form of exogenous M E C H A N I S M S O F   K E TO N E
ketones available, but their potency also necessi- S U P P L E M E N TAT I O N
tates a thorough investigation of their long-term Induction of hyperketonemia produces acute and
safety and toxicity. chronic changes in metabolic physiology and
In the late 1970s, Birkhahn et  al. were the molecular signaling pathways that provide thera-
first to synthesize a monoester of glycerol and peutic effects in varied disease states. Metabolic-
AcAc (monoacetoacetin) for parenteral nutri- based mechanisms of ketone therapies include
tion. These studies demonstrated that monoace- an elevation of blood ketones and associated
toacetin induced hyperketonemia comparable to anaplerosis with simultaneous suppression of
fasted rats at a dose of 50 g/kg per day (Birkhahn blood glucose, enhancement of insulin sensitivity,
and Border, 1978; Birkhahn et al., 1977; Birkhahn mitochondrial efficiency, suppression of specific
et  al., 1979). In attempts to increase the caloric inflammatory mediators and inhibition of oxi-
density of monoacetoacetin, they synthesized dative stress and preservation of mitochondrial
both a monoester and triester of glycerol and βHB. health and function.
These esters are hydrolyzed to release free βHB in a
way that elevates and sustains blood ketones. Later, Suppression of Blood Glucose and
Desrochers and colleagues synthesized mono- Enhancement of Insulin Sensitivity
and diesters of AcAc with BD that elevated both Hyperglycemia and hyperinsulinemia are patho-
AcAc and βHB (Desrochers et  al., 1995b). These logically linked to numerous disorders, includ-
and other ketone esters developed by or in col- ing cancer, cardiovascular disease, obesity, type
laboration with Henri Brunengraber and Richard 2 diabetes, impaired wound healing, and neu-
Veech have demonstrated an ability to induce a rodegenerative diseases, among others (Laakso
dose-dependent hyperketonemia (1–7 mM) in and Kuusisto, 2014; Ryu et al., 2014). These states
rats, mice, dogs, pigs, and humans (Brunengraber, are associated with chronic systemic inflamma-
1997; Ciraolo et al., 1995; Desrochers et al., 1995a; tion, oxidative stress, impairment of the immune
Puchowicz et  al., 2000; Srivastava et  al., 2012; system, and vascular and metabolic dysfunction
Sylvain et  al., 1995). Clarke and colleagues dem- (Bornfeldt and Tabas, 2011; de Carvalho Vidigal
onstrated the safety of a ketone ester in rats and et al., 2012; Turina et al., 2005). Exogenous ketone
humans that has also been documented in a recent supplements may provide therapeutic benefits
314

314 section IV: Ketone-Based Metabolism

in diseases characterized by hyperglycemia or supplements and lasted anywhere from 30 minutes


hyperinsulinemia as reports have demonstrated to 12 hours, suggesting that ketone supplementa-
that ketone administration lowers blood glucose tion could potentially provide a novel method of
by increasing insulin sensitivity. To demonstrate glycemic control.
this, male rats were fed a standard diet with 30% of
calories replaced with the R-3-hydroxybutyrate-R- Enhanced Metabolic Efficiency
1,3-butanediol monoester for 14 days. The ketone The superior metabolic efficiency of ketone bod-
ester-supplemented diet induced nutritional keto- ies has been known since the 1940s, when Henry
sis (3.5 mM βHB), and both plasma glucose and Lardy compared the energetic efficiency of 16
insulin were decreased by approximately 50% major carbohydrate, lipid, and intermediary
(Srivastava et  al., 2012). Glucose was decreased metabolites (Lardy and Phillips, 1945). He dem-
from 5 mM to 2.8 mM, and insulin was decreased onstrated that βHB and AcAc were unique among
from 0.54 ng/mL to 0.26 ng/mL. In a similar study the panel of metabolites tested in their ability to
by the same group, mice receiving a KE diet exhib- increase bull sperm mobility while simultaneously
ited a 73% increase in the Quantitative Insulin- decreasing oxygen consumption. Nearly 50  years
Sensitivity Check Index (QUICKI), a surrogate later, Richard Veech and colleagues confirmed
marker of insulin sensitivity, compared with con- Lardy’s observation and elucidated the molecular
trol, calorie-matched mice (Srivastava et al., 2012). mechanisms underlying the phenomenon in the
Fasting plasma glucose levels were not altered in working perfused rat heart (Kashiwaya et al., 1994).
these mice, but fasting plasma insulin levels were They demonstrated that supplementation of 5 mM
reduced by approximately 85% in the KE-fed mice ketones (4 mM βHB, 1 mM AcAc) to glucose-
compared with controls, demonstrating that exog- containing perfusate (10 mM glucose) increased
enous ketones enhance insulin sensitivity. cardiac hydraulic work by approximately 25%
In a study examining the potential use of while simultaneously reducing oxygen consump-
ketogenic supplements as a cancer therapy, mice tion (Kashiwaya et al., 1994). Their study revealed
consuming a standard high carbohydrate diet that this effect was mediated by a reduction of the
mixed with the R,S-1,3-butanediol diacetoacetate mitochondrial NAD couple and an oxidation of the
ester (BD-AcAc2) at 20% by weight had approxi- coenzyme Q couple, which increases the energy of
mately 30% lower blood glucose than control the redox span between these sites. This results in
animals (Poff et  al., 2014). The administration of an increase in energy released by electrons in the
BD did not significantly elevate blood ketones in ETC, causing more protons to be pumped into
this study, nor did it decrease blood glucose. This the inner mitochondrial space, thus enhancing the
result is likely due in part to the comparatively electrochemical gradient established there and
small dose of BD being consumed at any point in increasing the energy of ATP hydrolysis. Indeed,
time according to the route of administration, as thermodynamic tables for heat of combustion, cal-
BD in higher doses is known to elevate ketones. culated with bomb calorimeter experiments, show
Insulin levels were not investigated in this study. that βHB produces more energy than glucose per
In a study designed to assess the dose-dependent 2-carbon moiety (Cahill and Veech, 2003). These
effects of exogenous ketone supplements on blood results are supported by human studies that dem-
glucose, ketones, and lipids, healthy male rats onstrated a reduction in blood flow and oxygen
were administered one of five ketogenic agents consumption in the brains of fasted obese sub-
daily via intragastric gavage (Kesl et al., 2016). The jects in ketosis (McHenry, 1966). Thus, ketones
ketogenic agents investigated included:  BD, BD- appear to be a superior fuel for ATP production
AcAc2, a sodium/potassium β-hydroxybutyrate per unit oxygen compared to glucose, and can be
mineral salt (BMS), medium chain triglyceride oil considered among the most metabolically efficient
(MCT oil), BMS+MCT 1:1 mixture (BMS+MCT), energy metabolites (Figure 32.1).
and the R,S-1,3 butanediol diacetoacetate ester
(BD-AcAc2). BD-AcAc2, BMS, MCT oil, and Anti-Inflammatory Effects
BMS+MCT-treated rats demonstrated a decrease The physiological state of ketosis has known
in blood glucose following ketone supplement anti-inflammatory properties, as several stud-
administration given as an acute bolus. Similarly to ies have demonstrated that the KD reduces cir-
the previously mentioned mouse study, BD did not culating inflammatory markers in animals and
lower blood glucose in these animals, although it is in humans (Cassandra et  al., 2007; Sharman and
known to be a hypoglycemic agent. The duration Volek, 2004; Torres-Gonzalez et al., 2008). Recent
of the reduction in blood glucose varied between evidence suggests that this effect is, at least in
315

Chapter 32: Ketone Supplementation for Health and Disease 315

Interstitial 3Na+
space

Cytosol ADP+Pi ATP


2K+
Glucose ADP+Pi ATP
Major Energy Glucose ADP+Pi
∆G ATP
Source NAD+ ATP synthase
ATP Mitochondrion
NADH
Pyruvate– Pyruvate–
PDH
3H+ Cytochrome
Glycolysis oxidase 1/2O2
4H+
Cyt aa3
Acetyl CoA Electron 2e– H2O
transport Cyt C
system
CoQH2-CytC
Reductase
D-B-hydroxybutyrate
– TCA cycle 4H+
CoQ
CoQH2
2e–
Alternative Energy NAD+ NADH DH
Source
NAD+
Acetoacetyl CoA NADH 4H+
NADH
Acetoacetate–
– –
BHB BHB

FIGURE 32.1 Ketone bodies added in the presence of glucose fundamentally enhance mitochondrial metabolism.
When added to glucose, a physiological level of ketone bodies reduces the mitochondrial NAD couple, oxidizes the
coenzyme Q couple and increases the ΔG0 of ATP hydrolysis. These changes are shown on the right side of the figure.
The arrows illustrate the effects of ketone bodies compared with glucose alone. Ketone bodies provide an alternative
metabolic fuel, which can preserve ATP production during the impairment of glycolysis, as occurs in diabetes or
insulin resistance, or during inhibition of pyruvate dehydrogenase complex (PDH) as occurs in the presence of amyloid.
Oxidation of the coenzyme Q couple decreases superoxide anion production. Increases in the ΔG0 of ATP hydrolysis
widen the extra/intracellular ionic gradients leading to hyperpolarization of cells, which may play a role in treating
certain forms of epilepsy. The actions of ketone bodies mimic the acute effects of insulin in insulin-sensitive tissue and
tissues with high metabolic demands, including heart and brain. Figure taken with permission from Veech, et al.: Ketone
bodies—potential therapeutic uses.

part, directly mediated by the ketone bodies, (ROS), glycolytic inhibition, UCP, or SIRT2 signal-
indicating that exogenous ketone supplementa- ing, further validating its function as a signaling
tion could be used to suppress inflammation. metabolite. To investigate the in vivo translatabil-
Dixit and colleagues reported that βHB inhibits ity of this finding, researchers administered the
the NLRP3 inflammasome, an important compo- BD-AcAc2 to a familial cold anti-inflammatory
nent of the innate immune system that controls syndrome (FCAS) mouse model with an induced
activation of caspase-1 and production of the missense mutation in NLRP3. The ketone ester
pro-inflammatory cytokines IL-1β and IL-18 by protected the mice from neutrophilia and hyper-
macrophages (Youm et  al., 2015). This effect was glycemia, and did not affect infiltration of perito-
mediated specifically by βHB, as neither AcAc nor neal macrophages or overall frequency of splenic
the structurally related fatty acids butyrate and T cells, macrophages, or neutrophils, leading the
acetate elicited this response. The βHB-induced authors to conclude that elevating blood ketones
inhibition of the NLRP3 inflammasome occurs could be a therapeutic option for patients with
without its being oxidized in the TCA cycle, and NLRP3-mediated chronic inflammatory diseases.
is thus independent of its function as an energy Indeed, other research has shown that inhibition
metabolite. Furthermore, the anti-inflammatory of the NLRP3 inflammasome mitigates the sever-
changes associated with βHB were not dependent ity of numerous inflammatory diseases, includ-
on alterations in AMPK, reactive oxygen species ing atherosclerosis, type 2 diabetes, AD, and gout,
316

316 section IV: Ketone-Based Metabolism

among others (Duewell et al., 2010; Heneka et al., and AcAc decreases neuronal ROS production
2013; Martinon et al., 2006; Vandanmagsar et al., following glutamate exposure (Maalouf et  al.,
2011; Youm et al., 2013; Youm et al., 2015). 2007)  and inhibits apoptosis in cortical slices
In recent studies rats were fed one of three exposed to hydrogen peroxide (H2O2) (Kim do
exogenous ketone supplements (BD-AcAc2, BMS, et al., 2007).
or a 1:1 mixture of BMS:MCT mixed into stan- Eric Verdin and colleagues recently demon-
dard rodent chow at 18% by weight for 15 weeks. strated that βHB functions as an endogenous his-
Inflammatory profiling was performed on serum tone deacetylase inhibitor (HDACI) in vitro and
collected from the animals at the end of the in vivo at physiologic concentrations easily achiev-
chronic feeding study and revealed decreases in able with exogenous ketone supplementation
several pro-inflammatory cytokines including (Shimazu et al., 2013). Fasting, calorie restriction,
IL-1β, IL-6, IFN-γ, MCP-1, and RANTES (Poff and exogenous βHB administration all increased
et al., 2016). global histone acetylation in mice and induced the
transcriptional activation of the oxidative stress
Inhibition of Oxidative Stress resistance factors FOXO3A and MT2. This effect
The ketogenic diet has been reported to reduce was found to be mediated directly by inhibition of
oxidative stress in vivo in a number of preclini- class 1 and 2 HDACs. Furthermore, the authors
cal and clinical reports (Jarrett et  al., 2008). demonstrated that exogenous ketone supplemen-
Studies suggest that this effect is mediated by tation could prevent oxidative stress. Mice were
the ketone bodies themselves, and therefore the administered βHB via a subcutaneous pump for
effect would likely be recapitulated with exog- 24 hours prior to receiving an injection of para-
enous ketone supplementation. Some important quat, which induces the production and accumula-
molecular mechanisms underlying the effects tion of ROS. Protein carbonylation was suppressed
of ketone metabolism on oxidative stress were by 54%, and lipid peroxidation was completely
delineated by studies investigating the bioener- suppressed, in the kidneys of βHB pretreated mice.
getic efficiency and mitochondrial respiration in Immunoblotting of kidney tissue from these ani-
the working perfused rat heart following admin- mals revealed a significant increase in the mito-
istering of a glucose-containing perfusate supple- chondrial superoxide dismutase (MnSOD) and
mented with exogenous 5 mM βHB (Kashiwaya catalase (CAT) endogenous antioxidant systems.
et  al., 1994). As described previously, ketone This study strongly supports the feasibility and
metabolism increases the oxidation of ubiqui- applicability of exogenous ketone supplements for
nol (Q) in the electron transport chain, reducing the prevention of oxidative stress.
semiquinone radical (Q∙), an intermediate in the
reduction of ubiquinone that is sensitive to oxida- Preservation of Mitochondrial
tion by molecular oxygen to produce superoxide Health and Function
anion (O2−∙). O2−∙ is an important precursor for Ketone metabolism is generally recognized to
the generation of many ROS; therefore, ketone support or enhance mitochondrial health (Veech,
metabolism suppresses mitochondrial ROS pro- 2004). As previously described, ketones suppress
duction (Kashiwaya et al., 1994). Simultaneously, mitochondrial ROS production and enhance
ketone metabolism suppresses oxidative stress endogenous antioxidant systems. The resultant
by enhancing endogenous antioxidant capac- reduction in oxidative stress protects the mito-
ity. Ketone metabolism induces reduction of the chondrial DNA and membranes from damage
mitochondrial NAD and cytoplasmic NADP cou- that would impair respiratory and mitochondrial
ples. And NADH and NADPH are necessary for function. Interestingly, exogenous ketone supple-
the regeneration of reduced glutathione (GSH), mentation with a ketone ester has been shown to
which is required for the neutralization of ROS induce mitochondrial biogenesis (Srivastava et al.,
by glutathione peroxidase, an important endog- 2012). Mice in this study were fed a diet from
enous antioxidant enzyme. These molecular which approximately 30% of calories were derived
effects are observed in vivo, as the KD has been from the ketone ester D-β-hydroxybutyrate-R-
shown to increase the ratio of reduced to oxidized 1,3-butanediol monoester for one month. The
mitochondrial glutathione (GSH:GSSG) in rat mitochondrial content and expression of elec-
brains (Jarrett et  al., 2008). These effects appear tron transport chain proteins were significantly
to be ubiquitous in various tissues; however, the increased in the intrascapular brown adipose tis-
brain has been the most well characterized in this sue as compared with control mice, although calo-
regard. For example, in vitro treatment with βHB rie intake was matched between the two groups.
317

Chapter 32: Ketone Supplementation for Health and Disease 317

CONDITIONS WHERE preliminary investigation into preventing or


K E T O N E S U P P L E M E N TAT I O N delaying seizures with ketogenic supplements
I S L I K E LY B E N E F I C I A L in a variety of transgenic rodent and chemical-
Because of the previously described therapeutic induced seizure models. Pentylenetetrazole (PTZ)
mechanisms of ketone metabolism, exogenous is a GABA antagonist and epileptogenic agent
ketone supplementation is being investigated as that is used to induce seizures in rodents for pre-
a potential treatment for a number of disorders. clinical analysis of anticonvulsant therapies. In
Here we discuss some conditions for which ketone a recent study by Coppola and colleagues, the
supplements have been most well demonstrated to dosage threshold for seizure induction of PTZ
be useful, including epilepsy and seizure disorders, was assessed in control (water) and ketone ester-
glucose transporter type 1 deficiency syndrome, treated rats (Viggiano et  al., 2015). A  single oral
AD, cancer, insulin resistance and type 2 diabetes dose (4 g/kg body weight) of BD-AcAc2 elevated
mellitus, and weight loss. blood βHB (2.7mM) and increased the thresh-
old of PTZ seizure from 122±6 mg/kg to 140±11
Epilepsy/Seizure Disorders mg/kg. Unpublished data from preliminary stud-
The KD is a proven, effective therapy for epilepsy ies also suggest an anticonvulsant effect of ketone
in children and adults (Klein et al., 2014; Li et al., ester treatment in the WAG/Rij rat model of
2013). In many cases, it is more effective than anti- absent epilepsy, the Ube3a m-/p+ mouse model of
epileptic drugs (AEDs) and is therefore routinely Angelman syndrome, and the kainic acid–induced
used as a front-line treatment for children with mouse seizure model.
retractable (drug-resistant) epilepsy (Levy et  al.,
2011). Although the mechanisms of KD therapy Glucose Transporter Type 1
are largely unknown, achieving and sustaining Deficiency Syndrome
therapeutic ketonemia (>1 mM blood ketones) or Glucose transporter type 1 deficiency syndrome
ketonuria (>40 mg/dL) is generally necessary for (GLUT1 DS) is a rare genetic disorder caused by
antiseizure efficacy. Despite its success, the dietary a mutation in the SLC2A1 gene, which encodes
restrictions of the KD can be unpalatable for some the glucose transporter protein type 1 (GLUT1).
patients and difficult for caregivers, a contributing This mutation results in a glucose deficiency in the
factor for cessation of treatment (Klein et al., 2014; brain, which causes seizures as well as cognitive
Levy et  al., 2011). Exogenous ketogenic supple- and physical developmental delay. In a seminal
mentation mimics the metabolic and physiologic study in 1967, Cahill and colleagues discovered
effects of the KD, including enhancing mito- that ketones replace glucose as the predominant
chondrial biogenesis, anaplerosis, suppression energy substrate for the brain during prolonged
of glycolysis, and increasing ATP and adenosine fasting and starvation (Cahill, 2006). GLUT1 DS
production, all thought to mediate the therapeu- is treated with the KD, which circumvents the
tic effects of KD in epilepsy (Kesl et al., 2014; Kesl metabolic blockade by inducing ketosis and is
et al., 2016; Kovac et al., 2013; Masino and Geiger, effective at suppressing seizures and enhancing
2009; Srivastava et al., 2012; Stafstrom et al., 2009; cognitive and motor development in most patients.
Stafstrom et al., 2008). Maintaining therapeutic levels of ketosis is criti-
One of the earliest reports of the antiseizure cal to support the development of children with
efficacy of exogenous ketogenic supplementation this disorder. Thus, it is clear how an exogenous
was performed in a unique seizure model that uses ketone supplement could be useful in this patient
hyperbaric hyperoxia (HBO) to reliably induce population. Triheptanoin is a triglyceride contain-
epileptic-like (tonic-clonic) seizures in wild-type ing three heptanoates, a 7-carbon fatty acid whose
rats, a condition known as central nervous system metabolism produces the 5-carbon ketone bod-
oxygen toxicity (CNS-OT). A  single oral dose of ies β-ketopentanoate and β-hydroxypentanoate.
the ketone ester BD-AcAc2 induced rapid (within Because it possesses odd carbon FAs, heptanoate
30 minutes) and sustained (>4 hours) ketosis is metabolized through β-oxidation to produce
(>3mM βHB and >3mM AcAc) and prolonged propionyl-CoA, which can be subsequently car-
the latency to seizure by 574% (D’Agostino et al., boxylated to succinyl-CoA, replenishing the TCA
2013). An elevation in AcAc and acetone appear to cycle via anaplerosis (Borges and Sonnewald,
be required for the anticonvulsant effects of keto- 2012). Triheptanoin has shown therapeutic efficacy
sis. BD elevated blood βHB (>5mM) but did not in children and adults with GLUT1 DS, reducing
elevate AcAc or acetone nor did it affect latency seizure activity and improving neuropsychological
to seizure. This encouraging response prompted performance and cerebral metabolic rate (Pascual
318

318 section IV: Ketone-Based Metabolism

et al., 2014). There are ongoing studies investigat- control mice. Also, MRI analysis revealed that
ing the therapeutic effects of ketone salts and esters HBME prevented the development of asymmetri-
in a GLUT1 DS mouse model and may provide an cal ventricle morphology, which was observed
alternative or adjunctive treatment to the KD. in the untreated AD mice. Following 2.5 months
of treatment, the brains of the animals were ana-
Alzheimer’s Disease lyzed via immunohistochemistry. The authors also
In the early stages of AD, the brain exhibits a defi- performed in vitro studies to further investigate
ciency in glucose metabolism, which contributes mechanism of protection. The HBME-treated
to the neurodegeneration and progression of the mice exhibited reduced Aβ plaque deposition in
disorder (Chu and Jiao, 2015; de la Monte, 2012). the cortex and hippocampus. The authors reported
Patients with preclinical and clinical AD have that HBME supported neuronal survival following
decreased cerebral glucose metabolism as visu- glucose deprivation and prevented NaN3-induced
alized by fluorodeoxyglucose positron emission mitochondrial dysfunction by rescuing expres-
tomography (FDG-PET) (Mosconi et al., 2011). It sion of the respiratory complexes, reducing ROS
is thought that this decrease in glucose metabolism production, and maintaining mitochondrial mem-
is associated with brain insulin resistance (Talbot brane potential.
et  al., 2012). As ketones are the principal alter- In an encouraging case report by Newport
native fuel for the brain during fasting or starva- and Veech, supplementation with medium chain
tion, elevating blood ketone levels in AD patients fatty acids (MCFA) and (R)-3-hydroxybutyl (R)-
would theoretically bypass the deficiencies in glu- 3-hydroxybutyrate ketone monoester (KME)
cose metabolism and provide energy to the starv- was investigated as a potential therapy for AD
ing neurons. Data to support this hypothesis was (Newport et al., 2015). The patient described was
recently reported by Cunnane and colleagues, who an APOE4-positive, 63-year-old Caucasian male
demonstrated that while brain glucose uptake is with early-onset, sporadic AD diagnosed 12 years
impaired in AD, ketone uptake remains unaf- prior to the publication of the report. His disease
fected. Indeed, the authors conclude that supply- rapidly progressed prior to initiation of ketosis
ing ketones to the brains of AD patients could therapy in 2008, characterized by increasingly
restore the brain fuel supply to serve as a potential severe memory loss and an inability to carry out
therapeutic (Cunnane et  al., 2016). Thus, exog- normal activities of daily living. His Mini-Mental
enous ketone supplementation could be a useful State Examination (MMSE) score declined from
tool for supporting cerebral energy metabolism in 23 to 12 between 2004 and 2008, and his MRI
this regard. scans revealed diffuse involutional changes of
There is preclinical data to suggest that his frontal and parietal lobes with atrophy of the
ketones could protect against AD development amygdala and hippocampus. The patient initially
or slow its progression. The ability of a βHB- and began ketosis treatment in May 2008 by consum-
BD-containing ketone ester to suppress AD pro- ing sources of MCFAs—MCT oil and coconut oil
gression was evaluated in a triple transgenic AD (CO). His dose and regimen of administration was
mouse model (3xTgAD) (Kashiwaya et al., 2013). optimized, and eventually reached 165 mL of a
Presymptomatic mice were fed a diet with approxi- 4:3 mixture of MCT:CO divided into 3–4 servings
mately 20% kcal from the ketone ester and com- over the course of the day. Within 75 days of treat-
pared with isocaloric standard diet–fed control ment, the patient’s MMSE score improved from
animals. Ketone ester-treated animals exhibited 12 to 20. The patient continued MCFA treatment
less anxiety and improved performance on learn- for 20  months, and during that time he exhib-
ing and memory tests at 4 and 7 months after ini- ited remarkable cognitive and physical improve-
tiation of the diet. Immunohistochemical analysis ments. His Alzheimer’s Disease Assessment
of the brain revealed that the ketone ester-fed mice Scale-Cognitive (ADAS-Cog) rose 6 points and
had less Aβ and hyperphosphorylated tau deposi- his activities of daily living (ADLs) score rose 14
tion in the cortex, hippocampus, and amygdala. points over that time period. Further, MRI scans
In a similar study, 3-hydroxybutyrate methyl ester revealed no changes in brain atrophy from June
(HBME), a derivative of βHB, was assessed for its 2008 to April 2010, suggesting stabilization of the
therapeutic efficacy in a double transgenic mouse disease process. In 2010, the patient also began
model of AD (Zhang et  al., 2013a). The HBME- taking the KME (28.7 g KME, three times daily).
treated mice (40 mg/kg/d via intragastric gavage) The patient exhibited many improvements follow-
exhibited improved spatial learning and working ing KME treatment. Within a few days of initiating
memory and decreased anxiety compared with KME therapy, the patient regained the ability to
319

Chapter 32: Ketone Supplementation for Health and Disease 319

recite and write out the alphabet, and dress himself. observed in the patient over this 2-year study, sug-
He began improving in a number of ADLs such as gesting that prolonged hyperketonemia is likely
showering, shaving, and putting away dishes, and safe. The authors note that not all patients may
demonstrated improvements in abstract thinking, respond to such therapy in a similar manner, but
insight, and sense of humor. The patient himself that appropriately designed trials should be con-
reported feeling happier and more energetic after ducted to evaluate the percentage of Alzheimer’s
beginning treatment with KME. Over time, he patients responsive to ketone ester therapy.
exhibited significant improvements in memory An oral ketogenic compound and prescrip-
retrieval and regained the ability to perform com- tion medical food called AC-1202 (trade name
plex tasks such as vacuuming and yard work. Both Axona) was developed by Accera, Inc., as an AD
MCFA and KME treatment significantly elevated therapy. AC-1202 elevates blood ketone levels, as it
blood ketones, even while the patient continued to contains MCFAs, which are natural ketogenic pre-
consume a normal diet. cursors. AC-1202 was evaluated in a randomized,
The KME was particularly effective at induc- double-blind, placebo-controlled, multicenter trial
ing ketosis, elevating blood ketones up to 7 mM in patients with mild to moderate AD (Henderson
within 1 hour of administration (Figure 32.2), et  al., 2009). AC-1202 induced a mild level of
a level approximately 5–10 times greater than ketosis in patients (up to 0.3–0.4 mM), which
is possible with the classical ketogenic diet or was significantly higher than placebo controls.
MCFA consumption. The patient’s caregiver, a AC-1202 treatment induced small improvements
physician, noted that his improved cognitive and in ADAS-Cog, MMSE, and ADCS-CGIC (AD
physical performance seemed to track his plasma Cooperative Study—Clinical Global Impression
ketone concentrations, with the greatest improve- of Change) scores compared with placebo in
ments seen during peak elevation in blood some subgroups of AD patients tested. In many
ketones. Importantly, there were no adverse effects of the patient subgroups, AC-1202 did not affect

Responses of whole blood ß-hydroxybutyrate levels to


different doses of ketone monoester (KME)
7
25 grams
35 grams
6 50 grams

5
ß-hydroxybutyrate mM

0
1 2 3 4 5 6
Hours after KME ingestion

FIGURE 32.2 β-Hydroxybutyrate (βHB) concentrations rose to 3 to 7 mM 1 hour after ingestion of ketone monoester
(KME) in three different doses, 25, 35, and 50 g, taken on separate days. The peak levels measured are in the range of
those obtained during adherence to the classical ketogenic diet and are about 10-fold the concentrations achievable by
MCFA administration. The findings suggest that therapeutic ketosis can be maintained throughout the day if KME is
taken every 3 to 4 hours. Precision Xtra Glucose and Ketone Monitoring System (Abbott Diabetes Care, Inc., Alameda,
CA, USA) was used to measure βHB levels in capillary blood samples. Acetoacetate (AcAc) was not measured. This
figure was reprinted with permission from Newport et al. (2015).
320

320 section IV: Ketone-Based Metabolism

performance of APOE4 negative patients on these late-stage, metastatic cancer (Fine et  al., 2012).
tests. The modest improvements observed in this Prior to beginning the dietary intervention, all
study may potentially be due to the comparatively patients exhibited progressive disease. Following
low level of ketosis induced by AC-1202, consider- 1 month of treatment, over 50% of patients showed
ing what is attainable with KME or similar keto- stable disease or partial remission. Interestingly,
genic supplements. Furthermore, the lack of effect there was no significant drop in blood glucose in
observed in APOE4 positive patients could also the patients over the course of the diet, but rather,
be a result of the mild level of ketosis induced, or patient response was most strongly correlated with
alternatively because the study may have lacked degree of ketosis relative to baseline. In vitro and
the statistical power necessary to reveal a signifi- preclinical studies have confirmed the hypoth-
cant effect in this subpopulation (Newport et  al., esis that ketones are damaging to cancer. In 1979,
2015). Indeed, the case report by Newport and Magee et  al. demonstrated that βHB inhibited
colleagues suggests that ketosis can be an effective proliferation in a dose-dependent manner up to
therapy for AD in some APOE4 positive patients. 20 mM in multiple cancer cell lines of varied ori-
Taken together, these reports clearly demonstrate gin (Magee et al., 1979). Similarly, both AcAc and
the potential utility of exogenous ketone supple- βHB inhibited viability and induced apoptosis in
mentations to confer the therapeutic benefits of neuroblastoma cells, but had no effect on control
ketosis in a population of patients for which severe fibroblasts (Skinner et  al., 2009). Another study
dietary restrictions would be extremely difficult, if demonstrated that 5 mM βHB slows proliferation
not impossible. and decreases viability in VM-M3 glioma cells,
even in the presence of excess (25 mM) glucose.
Cancer Exogenous ketone supplementation with BD or
Unlike healthy tissues, many cancers do not appear the BD-AcAc2 elicited potent anticancer effects in
capable of efficiently metabolizing ketone bod- the VM-M3 model of metastatic cancer, slowing
ies for energy. Cancer cells often lack expression tumor growth and prolonging survival by 51% and
of the ketone utilization enzymes, like succinyl- 69%, respectively (Poff et al., 2014). These obser-
coenzyme A:3-oxoacid coenzyme A  transferase vations strongly suggest that exogenous ketone
(SCOT) (Chang et  al., 2013; Sawai et  al., 2004; supplements could be used as an effective adjuvant
Skinner et  al., 2009; Tisdale and Brennan, 1983). therapy for cancer.
Abnormal mitochondrial function and impaired There are multiple mechanisms by
OXPHOS capacity is another ubiquitous feature which ketones may be damaging to cancer
of cancers which likely limits the use of ketones, cells: (1) Cancer is particularly reliant on the gly-
which are metabolized exclusively within the colytic pathway for energy production and bio-
mitochondria, as a fuel for cancer. Indeed, a study synthesis (Gillies et al., 2008), and βHB inhibits
on five different brain cancer cell lines concluded the first and third enzymatic reactions of glycoly-
that glioma cells lack the capacity to metabolize sis (Randle et al., 1964). (2) Excess fermentation
βHB during glucose restriction, unlike healthy in cancer cells causes lactate production and a
brain cells (Fredericks, and Ramsey, 1978; Maurer subsequent acidification of the tumor microenvi-
et al., 2011). ronment, which promotes malignancy. Both lac-
The ketogenic diet, fasting, and calorie restric- tate and the ketone bodies are transported across
tion are dietary regimens that have been shown to the plasma membrane by the monocarboxylic
inhibit cancer progression in both preclinical and transporters family of transporters (Halestrap
clinical studies (Fine et al., 2012; Freedland et al., and Price, 1999). And βHB has been shown to
2008; Mavropoulos et  al., 2009; Nebeling et  al., inhibit lactate export from isolated rat hepato-
1995; Poff et  al., 2013; Rossifanelli et  al., 1991; cytes in vitro (Metcalfe et al., 1986). It is possible
Shelton et al., 2010; Wheatley et al., 2008; Zuccoli that ketones may damage cancer cells by inhibit-
et al., 2010). Until recently, the generally accepted ing lactate export through competitive inhibition
mechanism of action of these therapies was of monocarboxylic transporters, subsequently
decreasing glucose availability to the tumor and inducing intracellular acidification and prevent-
suppressing the insulin and IGF signaling path- ing the tumor-promoting effects of lactate in
ways. However, all three of these therapies also the tumor microenvironment. (3)  Inflammation
elevate blood ketones, and recent evidence sug- and oxidative stress are both known to promote
gests that ketones themselves may possess inher- cancer development and progression (Coussens
ent anticancer properties. One small clinical trial and Werb, 2002; Wang and Yi, 2008); thus, the
studied the effects of a 28-day KD in patients with inhibitory effects of ketone metabolism on both
321

Chapter 32: Ketone Supplementation for Health and Disease 321

of these pathways could contribute to its antican- hypothesized that ketones could be therapeutic by
cer efficacy. (4)  Cancers exhibit widespread dif- correcting metabolic defects of acute insulin defi-
ferences in such epigenetic patterns compared ciency or in the insulin-resistant state (Kashiwaya
to their normal tissue counterparts, allowing et al., 1997).
them to increase expression of oncogenes and The HDACI activity of βHB could also be
inhibit the expression of tumor suppressor genes beneficial in T2DM by altering the direct regula-
(Hassler and Egger, 2012). Histone deacetylase tion of HDAC-dependent glucose metabolism
inhibitors (HDACI) are being investigated for and by inducing resistance to oxidative stress. The
their use as antineoplastic agents, and have been HDACs regulate the expression of genes encoding
shown to elicit a plethora of anticancer effects in many metabolic enzymes, and HDAC3 knock-
vitro including activation of apoptosis, induction out animals exhibit reduced glucose and insu-
of ROS generation and DNA damage, and inhibi- lin. Suberoylanilide hydroxamic acid (SAHA), a
tion of DNA repair (Bose et al., 2014). As men- class I HDAC inhibitor, has been shown to improve
tioned, βHB functions as an endogenous HDACI, insulin sensitivity and increase oxidative metabo-
a mechanism that may underlie its potential use lism and metabolic rate in a mouse model of diabe-
as a cancer treatment. (5) Finally, mitochondrial tes (Galmozzi et al., 2013). Butyrate, a short-chain
transfer studies have demonstrated that healthy fatty acid that is structurally similar to βHB and
mitochondria act as a tumor suppressor (Seyfried, also acts as a HDACI, lowers blood glucose and
2012). The potential for ketone metabolism to insulin levels and improves glucose tolerance and
support or enhance mitochondrial health could respiratory efficiency (Gao et al., 2009). The vascu-
also account for its therapeutic effects and its lar dysfunction in T2DM is thought to be caused
potential for preventing carcinogenesis. by oxidative stress (Giacco and Brownlee, 2010).
And HDAC inhibition prevents renal damage in
Insulin Resistance/ Type 2 mouse models of diabetic nephropathy through
Diabetes Mellitus modulation of redox mechanisms (Advani et  al.,
As previously described, ketone ester adminis- 2011). Therefore, βHB suppression of oxidative
tration simultaneously decreases blood glucose stress through HDAC inhibition may help restore
and blood insulin concentrations (Srivastava insulin sensitivity and manage complications of
et al., 2012). Thus, less insulin is required to pro- diabetes.
mote peripheral glucose uptake, exemplifying an
enhancement in insulin sensitivity. These results Weight Loss
suggest a potential therapeutic use of exogenous The KD is hypothesized to induce weight loss by
ketone supplementation for insulin resistance and reducing appetite through the satiety effect of
type 2 diabetes mellitus (T2DM). Indeed, exog- protein and ketone bodies, reducing de novo lipo-
enous ketones qualitatively mimic the acute meta- genesis and increasing lipolysis, and enhancing
bolic effects of insulin (Kashiwaya et  al., 1997). metabolic efficiency with fat and ketone metabo-
It is known that insulin activates pyruvate dehy- lism (Paoli, 2014). However, the KD can be difficult
drogenase (PDH) to increase the production of to maintain long-term for many individuals, and
acetyl CoA. The administration of 5 mM ketones many people regain weight rapidly upon returning
mimicked this effect, increasing acetyl CoA pro- to a standard diet (Paoli, 2014). Recently, prelimi-
duction 15-fold in the glucose-perfused isolated nary studies have shown that exogenous ketone
rat heart (Kashiwaya et al., 1994). Furthermore, in supplementation can also induce weight loss.
this model, ketones and insulin increased cardiac It should be noted that ketone supplements are
hydraulic efficiency to a similar degree, approxi- sources of calories, with each ketone supplement
mately 25%–35% (Kashiwaya et  al., 1994). In providing on average 5–8 kcal/gram ingested;
another study by Srivastava et al., mice that were therefore, patients would need to decrease dietary
fed a diet formulated with ketone ester (30% kcal) caloric intake in order to prevent weight gain. The
had a 73% increase in the Quantitative Insulin- reported satiating effect of ketosis may aid in this
Sensitivity Check Index (QUICKI), a surrogate adjustment.
marker of insulin sensitivity, compared with con- The administration of both βHB and BD has
trol, calorie-matched mice (Srivastava et al., 2012). been shown to decrease food intake in rats and
Fasting plasma glucose levels were not altered in pigmy goats (Arase et  al., 1988; Carpenter and
these mice, but fasting plasma insulin levels were Grossman, 1983; Davis et  al., 1981; Langhans
reduced by approximately 85% in the KE-fed mice et al., 1983; Rossi et al., 2000). Similarly, it is sug-
compared with controls. The authors therefore gested that MCTs increase satiety, resulting in
322

322 section IV: Ketone-Based Metabolism

reduced food intake and weight loss as a con- ketones and lowering blood glucose. Importantly,
sequence of their rapid oxidation into ketone ketone supplementation provides a tool for achiev-
bodies (Krotkiewski, 2001; Poppitt et  al., 2010; ing ketosis in patients who are unable, unwilling,
Wymelbeke and Himaya, 1998). The MCTs may or uninterested in consuming a low carbohydrate
further counteract fat deposition in adipocytes by or ketogenic diet. It may also help circumvent
increasing thermogenesis (Dulloo, 2011). Several some of the difficulties associated with KD therapy,
studies in animals and humans have revealed as it allows for a rapid dose-dependent induction
increased energy expenditure and lipid oxida- of ketosis, which can be sustained with prolonged
tion with MCTs compared with LCTs (Baba et al., consumption and monitored precisely with com-
1982; Bach and Babayan, 1982; Crozier et  al., mercially available technologies (e.g., blood ketone
1987; Dulloo et  al., 1996; Ferreira et  al., 2014; meters). Simultaneously, it could provide patients
Karen and Welma, 2015; Scalfi et al., 1991; Seaton with the opportunity to reap the benefits of keto-
et al., 1986; St-Onge et al., 2003a, 2003b). Ketone sis without the practical and social difficulties of a
esters have also been shown to affect weight in highly restrictive diet. Further research is needed
mice, rats, and humans. Mice administered a to fully investigate the clinical utility and feasibility
diet supplemented with ketone ester resulted in of exogenous ketone supplements as a method of
reduced voluntary food intake, increased insulin inducing therapeutic ketosis.
sensitivity, increased resting energy expenditure,
and increased brown fat activity, demonstrat-
REFERENCES
ing the potential utility of the ketone ester as an
Advani, A., Huang, Q., Thai, K., Advani, S.L., White,
antiobesity compound (Srivastava et  al., 2012). K.E., Kelly, D.J., Yuen, D.A., Connelly, K.A.,
In another study, mice fed a ketone ester diet Marsden, P.A., and Gilbert, R.E. (2011). Long-
maintained a body weight of approximately 10%– term administration of the histone deacetylase
12% less than control, standard diet–fed animals inhibitor vorinostat attenuates renal injury in
(Kashiwaya et  al., 2013). Kashiwaya et  al. sug- experimental diabetes through an endothelial
gested that the ketone ester–supplemented diet nitric oxide synthase-dependent mechanism. Am
may benefit obesity as it decreases brain malonyl J Pathol 178, 2205–2214.
CoA, an important metabolic mediator of appetite Arase, K., Fisler, J.S., Shargill, N.S., York, D.A., and
(Kashiwaya et al., 2010). In a 28-day oral gavage Bray, G.A. (1988). Intracerebroventricular infu-
study, all five ketogenic supplements tested (BD, sions of 3-OHB and insulin in a rat model of
BD-AcAc2, MCT, BMS, BMS+MCT) caused a sig- dietary obesity. Am J Physiol 255, 81.
nificant reduction in weight gain compared with Baba, N., Bracco, E.F., and Hashim, S.A. (1982).
Enhanced thermogenesis and diminished depo-
control animals (Kesl et al., 2016). Similarly, a 15-
sition of fat in response to overfeeding with diet
week chronic feeding study in which BD-AcAc2, containing medium chain triglyceride. Am J Clin
BMS, or BMS+MCT replaced approximately 20% Apr;35(4):678–82. .
of the diet by weight, fed ad libitum, led to sig- Babayan, V.K. (1987). Medium chain triglycerides and
nificant reduction in weight gain compared with structured lipids. Lipids Jun;22(6):417–20.
control animals (unpublished data). In a 7-day Bach, A.C., and Babayan, V.K. (1982). Medium-chain
study, eight humans with T2DM consumed three triglycerides:  an update. Am J Clin Nutr 36,
KE-supplemented drinks a day in conjunction 950–962.
with their normal diet. The group lost an aver- Bell, S.J., Bradley, D., Forse, R.A., and Bistrian, B.R.
age of 2% of their body weight (Hashim and (1997). The new dietary fats in health and disease.
VanItallie, 2014). J Am Diet Assoc. 97, 280–286.
Birkhahn, R.H., and Border, J.R. (1978). Intravenous
CONCLUSION feeding of the rat with short chain fatty acid esters.
II. Monoacetoacetin. Am J Clin Nutr 31, 436–441.
Exogenous ketone supplements are being devel-
Birkhahn, R.H., McMenamy, R.H., and Border, J.R.
oped as an alternative or adjuvant method of
(1977). Intravenous feeding of the rat with short
inducing therapeutic ketosis aside from the clas- chain fatty acid esters:  I. Glycerol monobutyrate.
sic ketogenic diet. Emerging evidence has dem- Am J Clin Nutr 30, 2078–2082.
onstrated that these novel compounds have the Birkhahn, R.H., McMenamy, R.H., and Border, J.R.
potential to offer benefits for both healthy and dis- (1979). Monoglyceryl acetoacetate: a ketone body-
eased individuals alike. It is likely that most, if not carbohydrate substrate for parenteral feeding of
all, of the conditions that are known to benefit from the rat. J Nutr 109, 1168–1174.
the KD would receive some benefit from exog- Borges, K., and Sonnewald, U. (2012). Triheptanoin: a
enous ketone supplementation by elevating blood medium chain triglyceride with odd chain fatty
323

Chapter 32: Ketone Supplementation for Health and Disease 323

acids: a new anaplerotic anticonvulsant treatment? A., Robertson, J., Vanitallie, T., and Veech, R.
Epilepsy Res 100, 239–244. (2012b). Kinetics, safety and tolerability of (R)-
Bornfeldt, K.E., and Tabas, I. (2011). Insulin resis- 3-hydroxybutyl (R)-3-hydroxybutyrate in healthy
tance, hyperglycemia, and atherosclerosis. Cell adult subjects. Regul Toxicol Pharmacol: RTP 63,
Metabol 14, 575–585. 401–408.
Bose, P., Dai, Y., and Grant, S. (2014). Histone deacetylase Coussens, L.M., and Werb, Z. (2002). Inflammation
inhibitor (HDACI) mechanisms of action:  emerg- and cancer. Nature 420, 860–867.
ing insights. Pharmacol Ther Sep;143(3):323–36. Crozier, G., Bois-Joyeux, B., Chanez, M., Girard, J.,
Bough, K.J., and Rho, J.M. (2007). Anticonvulsant and Peret, J. (1987). Metabolic effects induced by
mechanisms of the ketogenic diet. Epilepsia long-term feeding of medium-chain triglycerides
48, 43–58. in the rat. Metabolism.1987 Aug;36(8):807–14
Brunengraber, H. (1997). Potential of ketone body Cunnane, S.C., Courchesne-Loyer, A., St-Pierre, V.,
esters for parenteral and oral nutrition. Nutrition Vandenberghe, C., Pierotti, T., Fortier, M., Croteau,
(Burbank, Los Angeles County, Calif) 13, 233–235. E., and Castellano, C.A. (2016). Can ketones com-
Budavari, S.E. (1989). The Merck index: An encyclope- pensate for deteriorating brain glucose uptake
dia of chemicals, drugs, and biologicals. Rahway, during aging? Implications for the risk and treat-
N.J: Merck. ment of Alzheimer’s disease. Ann NY Acad Sci.
Cahill, G. (2006). Fuel metabolism in starvation. D’Agostino, D., Arnold, P., and Kesl, S.  (2014).
Annual review of nutrition 26, 1–22. Compositions and methods for producing ele-
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? vated and sustained ketosis. In: Compositions and
Good medicine? Trans Am Clin Climatol Assoc Methods for Producing Elevated and Sustained
114, 149–161; discussion 162–143. Ketosis: US Patent: WO2014153416 A1; University
Carpenter, R.G., and Grossman, S.P. (1983). Plasma fat of South Florida):  http://www.google.com/pat-
metabolites and hunger. Physiol Behav 30, 57–63. ents/WO2014153416A1?cl=en.
Cassandra, E.F., Stephen, D.P., Maria Luz, F., Erin, D’Agostino, D.P., Pilla, R., Held, H.E., Landon, C.S.,
E.Q., Richard, J.W., Doug, M.B., William, J.K., Puchowicz, M., Brunengraber, H., Ari, C., Arnold,
Richard, D.F., and Jeff, S.V. (2007). Comparison of P., and Dean, J.B. (2013). Therapeutic ketosis with
low fat and low carbohydrate diets on circulating ketone ester delays central nervous system oxygen
fatty acid composition and markers of inflamma- toxicity seizures in rats. Am J Physiol Regul Integr
tion. Lipids 43(1):65–77 Comp Physiol 304, R829–836.
Chang, H.T., Olson, L.K., and Schwartz, K.A. (2013). Davis, R.J., Brand, M.D., and Martin, B.R. (1981).
Ketolytic and glycolytic enzymatic expression pro- The effect of insulin on plasma-membrane and
files in malignant gliomas:  implication for keto- mitochondrial-membrane potentials in isolated
genic diet therapy. Nutr Metab (Lond) 10, 47. fat-cells. Biochem J 196, 133–147.
Chang, P., Zuckermann, A.M., Williams, S., Close, A.J., de Carvalho Vidigal, F., Guedes Cocate, P., Goncalves
Cano-Jaimez, M., McEvoy, J.P., Spencer, J., Walker, Pereira, L., and de Cassia Goncalves Alfenas, R.
M.C., and Williams, R.S. (2015). Seizure control by (2012). The role of hyperglycemia in the induction
derivatives of medium chain fatty acids associated of oxidative stress and inflammatory process. Nutr
with the ketogenic diet show novel branching- Hosp 27, 1391–1398.
point structure for enhanced potency. J Pharmacol de la Monte, S.M. (2012). Brain insulin resistance and
Exp Ther 352, 43–52. deficiency as therapeutic targets in Alzheimer’s
Chu, C.B., and Jiao, L.D. (2015). (R)-3-oxobutyl 3- disease. Curr Alzheimer Res 9, 35–66.
hydroxybutanoate (OBHB) induces hyperketone- Desrochers, S., David, F., Garneau, M., Jetté, M., and
mia in Alzheimer’s disease. Int J Clin Exp Med 8, Brunengraber, H. (1992). Metabolism of R- and
7684–7688. S-1,3-butanediol in perfused livers from meal-fed
Ciraolo, S.T., Previs, S.F., Fernandez, C.A., Agarwal, and starved rats. Biochem J 285 (Pt 2), 647–653.
K.C., David, F., Koshy, J., Lucas, D., Tammaro, A., Desrochers, S., Dubreuil, P., Brunet, J., Jetté, M., David,
Stevens, M.P., and Tserng, K.Y. (1995). Model of F., Landau, B.R., and Brunengraber, H. (1995a).
extreme hypoglycemia in dogs made ketotic with Metabolism of (R,S)-1,3-butanediol acetoacetate
(R,S)-1,3-butanediol acetoacetate esters. Am J esters, potential parenteral and enteral nutrients in
Physiol 269, 75. conscious pigs. Am J Physiol 268, 7.
Clarke, K., Tchabanenko, K., Pawlosky, R., Carter, Desrochers, S., Quinze, K., Dugas, H., Dubreuil, P.,
E., Knight, N., Murray, A., Cochlin, L., King, M., Bomont, C., David, F., Agarwal, K.C., Kumar, A.,
Wong, A., Roberts, A., et  al. (2012a). Oral 28- Soloviev, M.V., Powers, L., et al. (1995b). R,S-1,3-
day and developmental toxicity studies of (R)- butanediol acetoacetate esters, potential alternates
3-hydroxybutyl (R)-3-hydroxybutyrate. Regul to lipid emulsions for total parenteral-nutrition. J
Toxicol Pharmacol: RTP 63, 196–208. Nutr Biochem 6, 111–118.
Clarke, K., Tchabanenko, K., Pawlosky, R., Carter, E., Drackley, J.K., Richard, M.J., and Young, J.W. (1990).
Todd King, M., Musa-Veloso, K., Ho., M., Roberts, In vitro production of beta-hydroxybutyrate from
324

324 section IV: Ketone-Based Metabolism

1,3-butanediol by bovine liver, rumen mucosa, Giacco, F., and Brownlee, M. (2010). Oxidative
and kidney. J Dairy Sci 73, 679–682. stress and diabetic complications. Circ Res 107,
Duewell, P., Kono, H., Rayner, K.J., Sirois, C.M., 1058–1070.
Vladimer, G., Bauernfeind, F.G., Abela, G.S., Gillies, R., Robey, I., and Gatenby, R. (2008). Causes
Franchi, L., Nunez, G., Schnurr, M., et al. (2010). and consequences of increased glucose metabo-
NLRP3 inflammasomes are required for ath- lism of cancers. J Nucl Med 49 (Suppl 2): 24S–42S.
erogenesis and activated by cholesterol crystals. Halestrap, A., and Price, N. (1999). The proton-
Nature 464, 1357–1361. linked monocarboxylate transporter (MCT) fam-
Dulloo, A.G. (2011). The search for compounds that ily: structure, function and regulation. Biochem J
stimulate thermogenesis in obesity manage- 343 Pt 2, 281–380.
ment:  from pharmaceuticals to functional food Hashim, S.A., and Tantibhedyangkul, P. (1987).
ingredients. Obes Rev 12, 866–883. Medium chain triglyceride in early life: effects on
Dulloo, A.G., Fathi, M., and Mensi, N. (1996). Twenty- growth of adipose tissue. Lipids 22(6):429–434
four-hour energy expenditure and urinary Hashim SA and VanItallie, T.B. (2014). Ketone body
catecholamines of humans consuming low-to- therapy: from the ketogenic diet to the oral admin-
moderate amounts of medium-chain triglycer- istration of ketone ester. J Lipid Res 55, 1818–1826.
ides: a dose-response. Eur J Clin 50(3):152–158. Hassler, M.R., and Egger, G. (2012). Epigenomics of
Dymsza, H.A. (1975). Nutritional application and impli- cancer:  emerging new concepts. Biochimie 94,
cation of 1,3-butanediol. Fed Proc 34, 2167–2170. 2219–2230.
Elizabeth, G.N., Hannah, C., Ruby, H.S., Margaret, Henderson, S.T., Vogel, J.L., Barr, L.J., Garvin, F.,
S.L., Nicole, E., Georgiana, F., Andrea, W., and Jones, J.J., and Costantini, L.C. (2009). Study of
Cross, J.H. (2009). A randomized trial of classical the ketogenic agent AC-1202 in mild to moderate
and medium-chain triglyceride ketogenic diets in Alzheimer’s disease:  a randomized, double-blind,
the treatment of childhood epilepsy. Epilepsia 50, placebo-controlled, multicenter trial. Nutr Metab
1109–1117. (Lond) 6, 31.
Falbe, J., Bahrmann, H., and Lipps, W. (1985). Alcohols, Heneka, M.T., Kummer, M.P., Stutz, A., Delekate, A.,
aliphatic. Ullmann’s Encyclopedia.http://onlineli- Schwartz, S., Vieira-Saecker, A., Griep, A., Axt,
brary.wiley.com/doi/10.1002/14356007.a01_279/ D., Remus, A., Tzeng, T.C., et al. (2013). NLRP3 is
abstract activated in Alzheimer’s disease and contributes to
Ferreira, L., Lisenko, K., and Barros, B. (2014). pathology in APP/PS1 mice. Nature 493, 674–678.
Influence of medium‐chain triglycerides on con- Hess, F.G., Cox, G.E., Bailey, D.E., Parent, R.A., and
sumption and weight gain in rats:  a systematic Becci, P.J. (1981). Reproduction and teratology
review. J Anim Physiol Anim Nutr (Berl) study of 1,3-butanediol in rats. J Appl Toxicol 1,
Fine, E., Segal-Isaacson, C., Feinman, R., Herszkopf, S., 202–209.
Romano, M., Tomuta, N., Bontempo, A., Negassa, Hove JLK, Grünewald, S., Jaeken, J., and Demaerel,
A., and Sparano, J. (2012). Targeting insulin inhi- P. (2003). D, L-3-hydroxybutyrate treatment of
bition as a metabolic therapy in advanced can- multiple acyl-CoA dehydrogenase deficiency
cer: a pilot safety and feasibility dietary trial in 10 (MADD). Lancet 361(9367):1433–5
patients. Nutrition 28, 1028–1035. Huttenlocher, P.R. (1976). Ketonemia and sei-
Fredericks, M., and Ramsey, R.B. (1978). 3-Oxo acid zures: metabolic and anticonvulsant effects of two
coenzyme A  transferase activity in brain and ketogenic diets in childhood epilepsy. Pediat Res
tumors of the nervous system. J Neurochem 31, 10, 536–540.
1529–1531. Huttenlocher, P.R., Wilbourn, A.J., and Signore, J.M.
Freedland, S., Mavropoulos, J., Wang, A., Darshan, (1971). Medium-chain triglycerides as a therapy
M., Demark-Wahnefried, W., Aronson, W., for intractable childhood epilepsy. Neurology 21,
Cohen, P., Hwang, D., Peterson, B., et  al. (2008). 1097–1103.
Carbohydrate restriction, prostate cancer growth, Jarrett, S.G., Milder, J.B., Liang, L.P., and Patel, M.
and the insulin-like growth factor axis. The (2008). The ketogenic diet increases mitochondrial
Prostate 68, 11–19. glutathione levels. J Neurochem 106, 1044–1051.
Galmozzi, A., Mitro, N., Ferrari, A., Gers, E., Gilardi, F., Karen, M., and Welma, S. (2015). Effects of medium-
Godio, C., Cermenati, G., Gualerzi, A., Donetti, E., chain triglycerides on weight loss and body com-
Rotili, D., et al. (2013). Inhibition of class I histone position: a meta-analysis of randomized controlled
deacetylases unveils a mitochondrial signature and trials. J Acad Nutr Diet 115.
enhances oxidative metabolism in skeletal muscle Kashiwaya, Y., Sato, K., Tsuchiya, N., Thomas, S., Fell,
and adipose tissue. Diabetes 62, 732–742. D.A., Veech, R.L., and Passonneau, J.V. (1994).
Gao, Z., Yin, J., Zhang, J., Ward, R.E., Martin, R.J., Control of glucose utilization in working perfused
Lefevre, M., Cefalu, W.T., and Ye, J. (2009). Butyrate rat heart. J Biol Chem 269, 25502–25514.
improves insulin sensitivity and increases energy Kashiwaya, Y., King, M.T., and Veech, R.L. (1997).
expenditure in mice. Diabetes 58, 1509–1517. Substrate signaling by insulin:  a ketone bodies
325

Chapter 32: Ketone Supplementation for Health and Disease 325

ratio mimics insulin action in heart. Am J Cardiol Langhans, W., Wiesenreiter, F., and Scharrer, E.
80, 50A–64A. (1983). Different effects of subcutaneous D,L-3-
Kashiwaya, Y., Pawlosky, R., Markis, W., King, M.T., hydroxybutyrate and acetoacetate injections on
Bergman, C., Srivastava, S., Murray, A., Clarke, food intake in rats. Physiol Behav 31, 483–486.
K., and Veech, R.L. (2010). A ketone ester diet Lardy, H.A., and Phillips, P.H. (1945). Studies of fat
increases brain malonyl-CoA and Uncoupling pro- and carbohydrate oxidation in mammalian sper-
teins 4 and 5 while decreasing food intake in the matozoa. Arch Biochem 6, 53–61.
normal Wistar Rat. J Biol Chem 285, 25950–25956. Levy, R.G., Cooper, P.N., and Giri, P. (2011). Ketogenic
Kashiwaya, Y., Bergman, C., Lee, J.H., Wan, R., King, diet and other dietary treatments for epilepsy.
M.T., Mughal, M.R., Okun, E., Clarke, K., Mattson, Cochrane Database Syst Rev 3.
M.P., and Veech, R.L. (2013). A ketone ester diet Li, H.-F.F., Zou, Y., and Ding, G. (2013). Therapeutic
exhibits anxiolytic and cognition-sparing proper- success of the ketogenic diet as a treatment option
ties, and lessens amyloid and tau pathologies in a for epilepsy: a meta-analysis. Iranian J Pediatr 23,
mouse model of Alzheimer’s disease. Neurobiol 613–620.
Aging 34, 1530–1539. Maalouf, M., Sullivan, P.G., Davis, L., Kim, D.Y., and
Kesl, S., Poff, A., Ward, N., Fiorelli, T., Ari, C., Rho, J.M. (2007). Ketones inhibit mitochondrial
D’Agostino, D. (2014). Methods of Sustaining production of reactive oxygen species production
Dietary Ketosis in Sprague-Dawley Rats. (San following glutamate excitotoxicity by increasing
Diego, CA:  Federation of the American Societies NADH oxidation. Neuroscience 145, 256–264.
for Experimental Biology). Magee, B.A., Potezny, N., Rofe, A.M., and Conyers,
Kesl, S., Poff, A., Ward, N., Fiorelli, T., Ari, C., Van R.A. (1979). The inhibition of malignant cell
Putten, A., Sherwood, J., Arnold, P., and D’Agostino, growth by ketone bodies. Aust J Exp Biol Med Sci
D. (2015a). Effect of Sustaining Dietary Ketosis 57, 529–539.
on the Hippocampal and Serum Metabolome of Mak, S.C., Chi, C.S., and Wan, C.J. (1999). Clinical expe-
Sprague-Dawley Rats. (Boston, MA: Federation of rience of ketogenic diet on children with refrac-
American Societies of Experimental Biology). tory epilepsy. Acta paediatrica Taiwanica = Taiwan
Kesl, S.L., Poff, A.M., Ward, N.P., Fiorelli, T.N., Ari, er ke yi xue hui za zhi 40, 97–100.
C., Van Putten, A.J., Sherwood, J.W., Arnold, P., Martinon, F., Petrilli, V., Mayor, A., Tardivel, A., and
and D’Agostino, D.P. (2016). Effects of exogenous Tschopp, J. (2006). Gout-associated uric acid crys-
ketone supplementation on blood ketone, glucose, tals activate the NALP3 inflammasome. Nature
triglyceride, and lipoprotein levels in Sprague- 440, 237–241.
Dawley rats. Nutr Metab (Lond) 13, 9. Masino, S.A., and Geiger, J.D. (2009). The ketogenic
Kim do, Y., Davis, L.M., Sullivan, P.G., Maalouf, M., diet and epilepsy:  is adenosine the missing link?
Simeone, T.A., van Brederode, J., and Rho, J.M. Epilepsia 50, 332–333.
(2007). Ketone bodies are protective against oxi- Maurer, G., Brucker, D., Bähr, O., Harter, P., Hattingen,
dative stress in neocortical neurons. J Neurochem E., Walenta, S., Mueller-Klieser, W., Steinbach, J.,
101, 1316–1326. and Rieger, J. (2011). Differential utilization of
Klein, P., Tyrlikova, I., and Mathews, G.C. (2014). ketone bodies by neurons and glioma cell lines: a
Dietary treatment in adults with refractory epi- rationale for ketogenic diet as experimental glioma
lepsy: a review. Neurology 83, 1978–1985. therapy. BMC Cancer 11, 315.
Koji, N., and Teruyoshi, Y. (2010). Medium-chain fatty Mavropoulos, J., Buschemeyer, W., Tewari, A.,
acids:  Functional lipids for the prevention and Rokhfeld, D., Pollak, M., Zhao, Y., Febbo, P.,
treatment of the metabolic syndrome. Pharmacol Cohen, P., Hwang, D., Devi, G., et al. (2009). The
Res. 61(3), 208–212 effects of varying dietary carbohydrate and fat con-
Kovac, S., Abramov, A.Y., and Walker, M.C. (2013). tent on survival in a murine LNCaP prostate can-
Energy depletion in seizures: anaplerosis as a strat- cer xenograft model. Cancer Prev Res 2, 557–565.
egy for future therapies. Neuropharmacology 69, McHenry LC, Jr. (1966). Cerebral blood flow. N Engl J
96–104. Med 274, 82–91.
Krotkiewski, M. (2001). Value of VLCD supplementa- Metcalfe, H.K., Monson, J.P., Welch, S.G., and Cohen,
tion with medium chain triglycerides. Int J Obes R.D. (1986). Inhibition of lactate removal by
Relat Metab Disord. 25(9), 1393–1400. ketone bodies in rat liver:  evidence for a quanti-
Mosconi, L., McHugh, P., de Leon, M.J., Berti, V., and tatively important role of the plasma membrane
Pupi, A. (2011). A Tale of Two Tracers:  Glucose lactate transporter in lactate metabolism. J Clin
Metabolism and Amyloid Positron Emission Invest 78, 743–747.
Tomography Imaging in Alzheimer’s Disease. Nebeling, L.C., Miraldi, F., Shurin, S.B., and Lerner, E.
(Amsterdam: IOS Press), 219–234. (1995). Effects of a ketogenic diet on tumor metab-
Laakso, M., and Kuusisto, J. (2014). Insulin resistance olism and nutritional status in pediatric oncology
and hyperglycaemia in cardiovascular disease patients:  two case reports. J Am Coll Nutr 14,
development. Nat Rev Endocrinol 10, 293–302. 202–208.
326

326 section IV: Ketone-Based Metabolism

Newport, M.T., VanItallie, T.B., Kashiwaya, Y., King, epilepsy:  short-term clinical effects. Dev Med
M.T., and Veech, R.L. (2015). A new way to pro- Child Neurol 31, 145–151.
duce hyperketonemia: use of ketone ester in a case Ryu, T.Y., Park, J., and Scherer, P.E. (2014).
of Alzheimer’s disease. Alzheimers Dement 11, Hyperglycemia as a risk factor for cancer progres-
99–103. sion. Diabetes Metab J 38, 330–336.
Opitz, K. (1958). Über die glykogenbildende Wirkung Sawai, M., Yashiro, M., Nishiguchi, Y., Ohira, M., and
von Glykolen. Naunyn Schmiedebergs Arch Hirakawa, K. (2004). Growth-inhibitory effects
Pharmacol 234(5), 448–454 of the ketone body, monoacetoacetin, on human
Paoli, A. (2014). Ketogenic diet for obesity: friend or gastric cancer cells with succinyl-CoA, 3-oxoacid
foe? Int J Environ Res Publ Health 11, 2092–2107. CoA-transferase (SCOT) deficiency. Anticancer
Pascual, J.M., Liu, P., Mao, D., Kelly, D.I., Hernandez, Res 24, 2213–2217.
A., Sheng, M., Good, L.B., Ma, Q., Marin-Valencia, Scala RA and Paynter, O.E. (1967). Chronic oral tox-
I., Zhang, X., et  al. (2014). Triheptanoin for glu- icity of 1, 3-butanediol. Toxicol Appl Pharmacol
cose transporter type I deficiency (G1D): modula- 10(1),160–164
tion of human ictogenesis, cerebral metabolic rate, Scalfi, L., Coltorti, A., and Contaldo, F. (1991).
and cognitive indices by a food supplement. JAMA Postprandial thermogenesis in lean and obese
Neurol 71, 1255–1265. subjects after meals supplemented with medium-
Poff, A.M., Ari, C., Seyfried, T.N., and D’Agostino, D.P. chain and long-chain triglycerides. Am J Clin Nutr
(2013). The ketogenic diet and hyperbaric oxygen 53(5),1130–1133.
therapy prolong survival in mice with systemic Seaton, T.B., Welle, S.L., and Warenko, M.K. (1986).
metastatic cancer. PLoS One 8: e65522. Thermic effect of medium-chain and long-chain
Poff, A.M., Ari, C., Arnold, P., Seyfried, T.N., and triglycerides in man. Am J Clin Nutr. 44(5),
D’Agostino, D.P. (2014). Ketone supplementation 630–634
decreases tumor cell viability and prolongs sur- Seyfried, T.N. (2012). Cancer as a Metabolic
vival of mice with metastatic cancer. Int J Cancer Disease:  On the Origin, Management, and
1711–1720. Prevention of Cancer. (Hoboken, NJ: Wiley).
Poff, A., Kesl, S.,Ward, N., D’Agostino, D.  (2016). Sharman, M., and Volek, J. (2004). Weight loss leads
Metabolic effects of exogenous ketone to reductions in inflammatory biomarkers after a
supplementation—an alternative or adjuvant to very-low-carbohydrate diet and a low-fat diet in
the ketogenic diet as a cancer therapy? Keystone overweight men. Clinical Science 107, 365–369.
Symposia—New Frontiers in Tumor Metabolism, Shelton, L.M., Huysentruyt, L.C., Mukherjee, P., and
Banff, Alberta, Canada. Seyfried, T.N. (2010). Calorie restriction as an
Poppitt, S.D., Strik, C.M., MacGibbon AKH, McArdle, anti-invasive therapy for malignant brain cancer
B.H., Budgett, S.C., and McGill, A.T. (2010). Fatty in the VM mouse. ASN Neuro 2: e00038.
acid chain length, postprandial satiety and food Shimazu, T., Hirschey, M.D., Newman, J., He, W.,
intake in lean men. Physiol Behav 101, 161–167. Shirakawa, K., Le Moan, N., Grueter, C.A.,
Puchowicz, M.A., Smith, C.L., Bomont, C., Koshy, J., Lim, H., Saunders, L.R., Stevens, R.D., et  al.
David, F., and Brunengraber, H. (2000). Dog model (2013). Suppression of oxidative stress by beta-
of therapeutic ketosis induced by oral administra- hydroxybutyrate, an endogenous histone deacety-
tion of R,S-1,3-butanediol diacetoacetate. J Nutr lase inhibitor. Science 339, 211–214.
Biochem 11, 281–287. Sills, M.A., Forsythe, W.I., Haidukewych, D.,
Randle, P.J., Newsholme, E.A., and Garland, P.B. MacDonald, A., and Robinson, M. (1986). The
(1964). Regulation of glucose uptake by muscle. medium chain triglyceride diet and intractable
8.  Effects of fatty acids, ketone bodies and pyru- epilepsy. Archives of Disease in Childhood 61,
vate, and of alloxan-diabetes and starvation, on the 1168–1172.
uptake and metabolic fate of glucose in rat heart Skinner, R., Trujillo, A., Ma, X., and Beierle, E. (2009).
and diaphragm muscles. Biochem J 93, 652–665. Ketone bodies inhibit the viability of human neu-
Rossi, R., Dörig, S., Del Prete, E., and Scharrer, E. roblastoma cells. Journal of Pediatric Surgery
(2000). Suppression of feed intake after paren- 44, 212.
teral administration of D-beta-hydroxybutyrate in Srivastava, S., Kashiwaya, Y., King, M., Baxa, U., Tam, J.,
pygmy goats. J Vet Med A, Physiol, Pathol, Clinic Niu, G., Chen, X., Clarke, K., and Veech, R. (2012).
Med 47, 9–16. Mitochondrial biogenesis and increased uncou-
Rossifanelli, F., Franchi, F., Mulieri, M., Cangiano, C., pling protein 1 in brown adipose tissue of mice fed
Cascino, A., Ceci, F., Muscaritoli, M., Seminara, P., a ketone ester diet. FASEB Journal: Official publi-
and Bonomo, L. (1991). Effect of energy substrate cation of the Federation of American Societies for
manipulation on tumor-cell proliferation in paren- Experimental Biology 26, 2351–2362.
terally fed cancer-patients. Clin Nutr 10, 228–232. St-Onge, M.P., Bourque, C., Jones PJH, and Ross, R.
Ruby, H.S., Jane, E., Bower, B.D., and Aynsley-Green, (2003a). Medium-versus long-chain triglycerides
A. (1989). Ketogenic diets in the treatment of for 27  days increases fat oxidation and energy
327

Chapter 32: Ketone Supplementation for Health and Disease 327

expenditure without resulting in changes in body Veech, R. (2004). The therapeutic implications of ketone
composition in overweight … . Int J Obes Relat bodies: the effects of ketone bodies in pathological
Metab Disord 27(1), 95–102. conditions:  ketosis, ketogenic diet, redox states,
St‐Onge, M.P., Ross, R., and Parsons, W.D. (2003b). insulin resistance, and mitochondrial metabo-
Medium‐chain triglycerides increase energy lism. Prostaglandins Leukot Essent Fatty Acids 70,
expenditure and decrease adiposity in overweight 309–319.
men. Obes Res 11(3), 395–402. Veech RL, Chance B, Kashiwaya Y, et al: Ketone bod-
Stafstrom, C.E., Roopra, A., and Sutula, T.P. (2008). ies, potential therapeutic uses. IUBMB life 51:
Seizure suppression via glycolysis inhibition with 241–7, 2001
2-deoxy-D-glucose (2DG). Epilepsia 49(Suppl 8), Veech, R.L. (2014). Ketone ester effects on metabolism
97–100. and transcription. J Lipid Res 55, 2004–2006.
Stafstrom, C.E., Ockuly, J.C., Murphree, L., Valley, M.T., Viggiano, A., Pilla, R., Arnold, P., Monda, M.,
Roopra, A., and Sutula, T.P. (2009). Anticonvulsant D’Agostino, D., and Coppola, G. (2015).
and antiepileptic actions of 2-deoxy-D-glucose in Anticonvulsant properties of an oral ketone ester
epilepsy models. Ann Neurol 65, 435–447. in a pentylenetetrazole-model of seizure. Brain
Sylvain, D., Khadijah, Q., Hermann, D., Pascal, D., Res 1618, 50–54.
Catherine, B., France, D., Kamlesh, C.A., Alok, Wang, J., and, Yi, J. (2008). Cancer cell killing via
K., Maxim, V.S., Lisa, P., Bernard, R.L., and Henri, ROS: to increase or decrease, that is the question.
B. (1995). R,S-1,3-butanediol acetoacetate esters, Cancer Biol Ther 7, 1875–1884.
potential alternates to lipid emulsions for total Wheatley, K.E., Williams, E.A., Smith, N.C., Dillard, A.,
parenteral nutrition. J Nutr Biochem 6. Park, E.Y., Nunez, N.P., Hursting, S.D., and Lane,
Talbot, K., Wang, H.Y., Kazi, H., Han, L.Y., Bakshi, M.A. (2008). Low-carbohydrate diet versus caloric
K.P., Stucky, A., Fuino, R.L., Kawaguchi, K.R., restriction:  effects on weight loss, hormones, and
Samoyedny, A.J., Wilson, R.S., et  al. (2012). colon tumor growth in obese mice. Nutr Cancer
Demonstrated brain insulin resistance in 60, 61–68.
Alzheimer’s disease patients is associated with Wymelbeke, V.V., and Himaya, A. (1998). Influence of
IGF-1 resistance, IRS-1 dysregulation, and cogni- medium-chain and long-chain triacylglycerols on
tive decline. J Clin Invest 122, 1316–1338. the control of food intake in men. Am J Clin Nutr
Tate, R.L., Mehlman, M.A., and Tobin, R.B. (1971). 68(2), 226–234.
Metabolic fate of 1,3-butanediol in the rat:  con- Youm, Y.H., Grant, R.W., McCabe, L.R., Albarado,
version to -hydroxybutyrate. J Nutr 101, D.C., Nguyen, K.Y., Ravussin, A., Pistell, P.,
1719–1726. Newman, S., Carter, R., Laque, A., et  al. (2013).
Tisdale, M., and Brennan, R. (1983). Loss of acetoac- Canonical Nlrp3 inflammasome links systemic
etate coenzyme A transferase activity in tumours low-grade inflammation to functional decline in
of peripheral tissues. Br J Cancer 47, 293–297. aging. Cell Metabol 18, 519–532.
Tobin, R.B., Mehlman, M.A., and Parker, M. (1972). Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg, E.L.,
Effect of 1,3-butanediol and propionic acid on Bodogai, M., Kim, D., D’Agostino, D., Planavsky,
blood ketones, lipids and metal ions in rats. J Nutr N., Lupfer, C., Kanneganti, T.D., et al. (2015). The
102, 1001–1008. ketone metabolite beta-hydroxybutyrate blocks
Torres-Gonzalez, M., Volek, J.S., Leite, J.O., Fraser, NLRP3 inflammasome-mediated inflammatory
H., and Luz Fernandez, M. (2008). Carbohydrate disease. Nat Med 21(3), 263–269
restriction reduces lipids and inflammation Zhang, J., Cao, Q., Li, S., Lu, X., Zhao, Y., Guan, J.S.,
and prevents atherosclerosis in Guinea pigs. J Chen, J.C., Wu, Q., and Chen, G.Q. (2013a). 3-
Atheroscler Thromb 15, 235–243. Hydroxybutyrate methyl ester as a potential
Trauner, D.A. (1985). Medium-chain triglycer- drug against Alzheimer’s disease via mitochon-
ide (MCT) diet in intractable seizure disorders. dria protection mechanism. Biomaterials 34,
Neurology 35, 237–238. 7552–7562.
Turina, M., Fry, D.E., and Polk, H.C., Jr. (2005). Zhang, Y., Kuang, Y., LaManna, J., and Puchowicz, M.
Acute hyperglycemia and the innate immune sys- (2013b). Contribution of brain glucose and ketone
tem: clinical, cellular, and molecular aspects. Critic bodies to oxidative metabolism. Adv Exp Med Biol
Care Med 33, 1624–1633. 765, 365–370.
Vandanmagsar, B., Youm, Y.H., Ravussin, A., Galgani, Zuccoli, G., Marcello, N., Pisanello, A., Servadei, F.,
J.E., Stadler, K., Mynatt, R.L., Ravussin, E., Stephens, Vaccaro, S., Mukherjee, P., and Seyfried, T.N. (2010).
J.M., and Dixit, V.D. (2011). The NLRP3 inflamma- Metabolic management of glioblastoma multiforme
some instigates obesity-induced inflammation and using standard therapy together with a restricted
insulin resistance. Nat Med 17, 179–188. ketogenic diet: Case Report Nutr Metab (Lond) 7, 33.
328

33
Identifying the Molecular Mechanism of the Medium
Chain Triglyceride (Ketogenic) Diet
M AT T H E W C . WA L K E R , F R C P, P H D A N D R O B I N S . B . W I L L I A M S , P H D

INTRODUCTION in a high attrition rate (Levy et al., 2012). Due to


The medium chain triglyceride (MCT) ketogenic these adverse effects, and a desire to increase the
diet was first introduced as a more palatable alter- efficacy of the diet, many studies have sought to
native to the classical ketogenic diet for the treat- identify the therapeutic mechanism of the diet. As
ment of refractory epilepsy by Huttenlocher et al. the diet was based on the classical ketogenic diet,
(1971). It now provides a key therapeutic approach it was considered to act through the generation of
for the treatment of children with drug-resistant ketones (Bough and Rho, 2007; Rho and Stafstrom,
epilepsy (Levy et  al., 2012; Liu, 2008; Neal and 2011). However, the presence of these ketones
Cross, 2010; Neal et  al., 2009). The diet involves poorly correlates with anticonvulsant efficacy, and
a stringent reduction in carbohydrate intake, with this ketone-based mechanism has not been widely
an elevated consumption of medium chain fatty supported in animal model studies (Likhodii et al.,
acids within a triglyceride backbone. Typically, 2000; Thavendiranathan et al., 2000). Furthermore,
the fats in this diet provide 65% to 75% of total ketones do not directly alter hippocampal synaptic
daily energy requirement. These fats comprise transmission (Thio et al., 2000) nor do they affect
two medium, straight chain fats:  octanoic acid epileptiform activity induced by 4-aminopyridine
(comprising eight carbons) and decanoic acid in ex vivo seizure models (Thio et al., 2000). Thus
(comprising ten carbons); 60%–80% of the MCT the therapeutic mechanism of the MCT ketogenic
intake is octanoic acid and the remainder is mostly diet has, until recently, been unclear.
decanoic acid (Sills et  al., 1986b). Both fats are
rapidly hydrolyzed off the triglyceride backbone M E D I U M C H A I N FAT T Y
in the gut, and absorbed as free fatty acids (Bach ACIDS IN SEIZURE CONTROL
and Babayan, 1982) and metabolized into ketones A clinical role for medium chain fatty acids in
(β-hydroxybutyrate, acetone, and acetoacetate) direct seizure control was proposed over 30 years
and carbon dioxide, or catabolized into long chain ago (Dean et al., 1989; Haidukewych et al., 1982;
fatty acids. This diet results in an elevation of the Sills et al., 1986a), but due to limited study sizes, a
concentration of both fatty acids in peripheral direct correlation between plasma concentrations
blood:  octanoic acid to 104–859 µM and averag- and seizure control was not established. Recently,
ing around 306 µM; and decanoic acid to 87–552 however, an unbiased screen for medium and
µM with an average of 157 µM (Dean et al., 1989; short chain fatty acids related to valproate (Chang
Haidukewych et  al., 1982; Sills et  al., 1986a). In et al., 2012) in a simple cellular model identified
animal models, decanoic acid has been found to a number of fatty acids as potential seizure-con-
penetrate the blood-brain barrier and to be pres- trol treatments. In this study, a simple non-animal
ent in brain at 60%–80% of serum levels (Wlaz model, in which valproate has been shown to reg-
et al., 2012). ulate phosphoinositide turnover (Xu et al., 2007),
The dietary restrictions and high fat intake was used to identify potential anti-seizure com-
of the MCT ketogenic diet can cause a variety pounds (Cunliffe et al., 2015). In this study, over 60
of gastrointestinal-related side effects, such as compounds were screened for an inhibitory effect
cramps, bloating, diarrhea, and vomiting (Liu, on rapid phosphoinositide turnover. Interestingly,
2008). Use of the diet has also been limited by this inhibitory effect has since been confirmed as
poor tolerability, especially in adults, resulting a therapeutic mechanism for valproate in in vitro
329

Chapter 33: Molecular Mechanism of the Medium Chain Triglyceride (Ketogenic) Diet 329

and in vivo animal seizure models (Chang et al., seizure control and related effects (Chang et al.,
2014). Thus, using this phosphoinositide screen, 2013). Here, decanoic acid and nonanoic acid
Chang et al. (2012) were able to investigate a wide were shown to provide protection against an ex
range of fatty acids and related compounds, with vivo model of epileptiform activity generated by
some structures producing little effect, and some decreasing GABAergic inhibition (pentylenetet-
structures showing enhanced activity over valpro- razol, PTZ; Figure 33.1). In these studies, deca-
ate. These potent compounds included decanoic noic acid completely blocked seizure activity 35
acid, nonanoic acid, and the branched 8-carbon minutes post addition. Interestingly, octanoic
backbone 4-methyloctanoic acid. However, these acid, also prescribed in the MCT ketogenic diet,
compounds did not act through regulating inosi- did not reduce seizure activity in this model.
tol levels in this simple model that was previously However, branched derivatives of octanoic acid
suggested as a mechanism of valproate (Shaltiel et showed variable efficacy, with some compounds
al., 2007; Vaden et al., 2001; Williams et al., 2002; providing strong seizure control (e.g., 4-methy-
Williams, 2005). These compounds were further loctanoic acid) and some showing no activ-
tested in a well-established ex vivo mammalian ity (3,7-dimethyloctanoic acid). These results
model for drug-resistant epilepsy, the low mag- confirmed earlier studies (Chang et al., 2012)
nesium hippocampal-entorhinal cortex model but extended these findings to establish a wide
(Chang et al., 2012). Here, brain slices were kept chemical space showing potential efficacy in
“alive” in a bath perfused with oxygenated arti- seizure control. This study also examined these
ficial cerebrospinal fluid, and seizure-like activ- chemicals for inhibition of histone deacetylase
ity was induced by reducing magnesium levels, activity (HDAC), an effect associated with tera-
so enhancing NMDA receptor currents. In these togenicity (Gottlicher et al., 2001; Gurvich et al.,
experiments, equimolar concentrations of a range 2004; Phiel et al., 2001), and thus limiting their
of fatty acids were more potent than valproate, use during pregnancy (Jentink et al., 2010; Koren
within 10 minutes of compound addition (the time et al., 2006). Of the compounds analyzed in this
for the compounds to perfuse the bath and pen- study, only valproate and 2-proplyoctanoic acid
etrate the slice), thus significant levels of ketosis in caused significant inhibition of HDAC activity
these experiments is unlikely. These experiments at 1 mM, a concentration considerably higher
therefore suggested a direct role for medium chain than that of medium chain fatty acids found
fatty acids in seizure control. during MCT ketogenic diet treatment (Sills et
Further studies pursued the analysis of the al., 1986a). This suggests that medium chain
activity of these medium chain fatty acids in fatty acid are unlikely to exhibit the teratogenic

Treatment (1 mM)
120 Control
discharge (100 % of baseline)

O
Frequency of epileptiform

100 Octanoic acid HO


O

80 Valproate HO
O

4-methyloctanoic acid HO
60 O
Nonanoic acid
40 HO
O
4-ethyloctanoic acid
20 HO
O
Decanoic acid
0 O
HO

0 10 20 30 40 50 60 4-BCCA HO
Time (minutes)

FIGURE 33.1 Structurally specific medium chain fatty acids strongly reduce frequency of in vitro epileptiform activity.
In these experiments, an ex vivo hippocampal slice model was used, with seizure-like activity induced by application of
PTZ. Compounds were added (gray box) at 1 mM, and removed after 40 minutes. The frequency of epileptiform activity
is plotted against time following control (DMSO). Straight medium chain fatty acids decanoic (10 carbon) and nonanoic
acid (9 carbon), two branched chain derivatives (4-methyloctanoic acid and 4-ethyloctanoic acid), and a cyclic congener
(trans-4-butylcyclocarboxylic acid; 4-BCCA) were able to strongly reduce seizure-like activity. In contrast, octanoic acid
showed no inhibitory activity. The widely used, established epilepsy treatment, valproate, showed weak activity in this
seizure model. Data derived from Chang et al. (2014), Chang et al. (2015), and Chang et al. (2016).
330

330 section IV: Ketone-Based Metabolism

effects found following valproate treatment. This most potent compounds branched around the
study also showed that medium chain fatty acids fifth carbon. This study also examined these com-
(4-methyloctanoic acid and nonanoic acid) pro- pounds for protection against excitotoxic cell
vide control against self-sustaining status epi- death, an effect that is similar to that resulting
lepticus (SSSE), induced by perforant pathway from status epilepticus (DeLorenzo et al., 2005).
stimulation, in an in vivo rat model (Walker and In these experiments, exposure of hippocampal
Williams, 2015). Analysis of neuronal cell death neurons in culture to low magnesium for 4 hours
in these animals as a result of SSSE showed that triggered cell death (Deshpande et al., 2008),
2 months after seizure induction, nonanoic acid and consistent with the seizure control experi-
significantly reduced cell death in the hilus of ments, neuroprotection was seen with the addi-
the hippocampus, suggesting a neuroprotective tion of compounds with methyl branching from
effect. This therapeutic effect is also unlikely to the fourth to the seventh carbon (Chang et al.,
be related to ketosis, since these compounds rap- 2015). Based on these results, three related com-
idly reduced seizure activity within 10 minutes pounds were then examined: 4-ethyloctanoic
of treatment. For nonanoic acid, these effects acid, containing a longer side chain; 4-methyl-
were also unlikely to occur due to sedation, since nonanoic acid, containing a longer backbone,
sedative effects were not shown even at high con- and trans-4-butylcyclohexane carboxylic acid
centrations (600 mg/kg). Together these studies (4-BCCA), a related cyclic compound. All three
provide strong evidence for the direct activity of compounds also showed strong seizure control
a range of medium chain fatty acids in seizure in the PTZ model (Figure 33.1), and were not
control and neuroprotection. active against HDAC activity. These compounds
One further study has been reported, investi- were further screened in a range of in vivo seizure
gating the breadth of chemical space for medium models (Table 33.1), providing promising activity
chain fatty acids in seizure control (Chang et al., for trans-4-butylcyclohexane carboxylic acid in
2015). This study specifically investigated octanoic multiple models. In a related study, mice treated
acid–related compounds, including a systematic with decanoic acid by gastric gavage (30 mmol/
analysis of methyloctanoic acid derivatives. Using kg) also showed seizure control in the 6-Hz sei-
an ex vivo model of seizure activity, induced in zure and maximal electroshock seizure threshold
rat hippocampal slices by the application of PTZ, (MEST) models (Wlaz et al., 2012). These data
a clear structure activity relationship was seen provide evidence that medium chain fatty acids,
in seizure control, provided by the position of of defined structure, show activity in a range of in
branching on the octanoic acid backbone. The vivo seizure models.

TABLE 33.1 IN VIVO SEIZURE CONTROL DATA FOR ACTIVE COMPOUNDS

Compound Species Seizure Dose Animals Animals ED50


model (mg/kg) (protected/tested) (Toxic/tested) (mg/kg)

4-EOAa Mice 6Hz 150 7/8 — 110


Rat MES 125 12/16 4/15† 100
Mice scMET 200 8/8 4/8¥ 142
Mice CKM 110 6/8 0/8 71
4-BCCAa Mice 6Hz 100 3/4 0/4 81
Rat MES 100 4/8 1/8† ~100*
Mice scMET 150 4/8 0/8 ~150*
Mice CKM 80 8/8 0/8 44
VPA Mice 6Hz 263b
Rat MES 485c
Mice scMET 191d
Mice CKM 174e
Summary of in vivo seizure control in multiple models, with data provided by collaborative research with (a) NINDS, or previously deter-
mined by (b) Barton et al. (2001), (c) Loscher (1999), (d) Rowley and White (2010), or (e) NINDS.
* based on single dose data; — not determined; † unable to grasp rotorod; ¥ ataxia/loss of righting reflex
Source: Partially reproduced from Chang et al. (2015), with permission.
331

Chapter 33: Molecular Mechanism of the Medium Chain Triglyceride (Ketogenic) Diet 331

M E D I U M C H A I N FAT T Y This study also investigated some kinetic


A C I D S A R E A M PA aspects of medium chain fatty acid-dependent
R E C E P TO R AG O N I S T S AMPA receptor inhibition (Chang et al., 2016).
The molecular mechanism of decanoic acid in First, since during seizure activity, membrane
direct seizure control has recently been estab- depolarization occurs, increasing excitability,
lished (Chang et  al., 2016). In this study, deca- decanoic acid was analyzed at varying membrane
noic acid (1 mM) was shown to abolish seizure potentials. These studies identified that under
activity in vitro in both the PTZ- and low- depolarized membrane potentials, decanoic acid
magnesium-induced drug-resistant seizure showed enhanced inhibitory activity, thus poten-
models. As previously described, decanoic acid tially providing more potent inhibition during
blocks seizure activity within 15 minutes of addi- seizure activity. Second, since AMPA receptors
tion to each model, strongly suggesting that the are activated by glutamate, and glutamate levels
effect is directly related to the fatty acid, not to increase during seizure activity (Van Den Pol et
ketone generation. Moreover acetone and β- al., 1996), competition assays were used to explore
hydroxybutyrate (two ketones) have no effect in whether altered glutamate levels modulate deca-
either model at high concentrations (10 mM). noic acid-dependent AMPA receptor inhibition.
The mechanism of decanoic acid for this effect These experiments indicate that decanoic acid is a
was then shown using whole-cell patch clamp noncompetitive AMPA receptor antagonist (Table
recordings of evoked AMPA receptor–mediated 33.2), thus its inhibitory activity is independent of
excitatory postsynaptic currents (EPSCs) from glutamate concentrations.
CA1 pyramidal neurons, where decanoic acid These studies have suggested that medium
reduced EPSC amplitude by 17.0 ± 5.6% at a con- chain fatty acids function in seizure control
centration comparable to its steady state level in through inhibition of AMPA receptors. However,
children on the MCT diet (100 µM) (Sills et al., it remained possible that these compounds could
1986a). These data, for this first time, provided give rise to seizure control through alternative
evidence of a direct mechanism of the MCT keto- mechanisms. To address this, Chang et al. (2016)
genic diet in seizure control through inhibition of employed the hippocampal/PTZ seizure model,
AMPA receptor currents. treated with the well-characterized AMPA recep-
To investigate a direct effect of decanoic acid tor antagonist GYKI 52466 (Arai, 2001), to pro-
on AMPA receptor activity, a range of experi- vide a similar level of AMPA receptor inhibition
ments were carried out using a Xenopus heter- as decanoic acid (1 mM). Under these conditions,
ologous expression system (Chang et  al., 2016).
Here, AMPA receptor subunits (GluA1-3) were
expressed in oocytes individually or in pairs to TABLE 33.2 SUMMARY OF MEDIUM
produce homotetramers (GluA1) or heterotetra- CHAIN FAT T Y ACIDS INHIBITORY
mers (GluA1/2 and GluA2/3), and glutamate was
EFFECTS ON AMPA RECEPTORS
applied, resulting in inward currents, on which
direct application of decanoic acid and related GluA2/3 GluA1/2 GluA1
compounds was tested. This approach allowed the
quantification of the direct inhibition of AMPA Subunit specificity: IC50 (mM)
receptor–mediated currents by medium chain Decanoic acid 0.52 1.16 2.09
fatty acids (Table 33.2). From these experiments, Nonanoic acid 1.48 1.79
decanoic acid was shown to be an AMPA receptor 4-methyloctanoic 0.84
antagonist, with efficacy across all subunit com- acid
position, and greatest potency against GluA2/3 Octanoic acid 3.82
heterotetramers (IC50 = 0.52 mM). Decreasing the
Voltage dependence (IC50 mM, Decanoic acid, GluA2/3)
length of the backbone reduced potency, such that
nonanoic acid and octanoic acid were increasingly –80 mV 1.11
less potent (IC50 = 1.48 and 3.82 mM, respectively), –40 mV 0.43
although addition of a side chain to octanoic acid Kinetics
to produce 4-methyloctanoic acid enhanced
potency (IC50  =  0.84 mM). These data indicate a Decanoic acid is noncompetitive toward glutamate
direct action of decanoic acid and specific related Data was derived from heterologous expression of rat glutamate
structures on inhibition of AMPA receptors to receptors in a Xenopus oocyte model, activated with 100 μM
glutamate.
regulate neuronal function. Source: Data derived from Chang et al. (2016).
332

332 section IV: Ketone-Based Metabolism

GYKI 52466 blocked seizure activity. These transcriptional regulation relevant to a putative
results indicate that direct AMPA inhibition disease-modifying role (Kim et  al., 2015; Masino
is sufficient to explain decanoic acid’s antisei- et  al., 2011), and to have indirect effects on ion
zure effect in in vitro seizure models. Moreover, channels (Sada et al., 2015). Thus ketones may still
AMPA receptors are widely recognized as tar- provide some beneficial effects related to specific
gets for seizure control (Meldrum and Rogawski, causes of epilepsy and in protection against long-
2007; Russo et al., 2012; Szenasi et al., 2008). term epileptogenic changes.
Perampanel, a selective AMPA receptor antago-
nist, has also recently been marketed for the I M P L I C AT I O N S
treatment of refractory partial epilepsy (Rektor, Understanding the mechanism of action of the
2013), confirming the relevance of this mecha- ketogenic diet is essential for the development of
nism to patient treatment. Chang et al. (2016) diets or treatments that are less restrictive, and
also showed that inhibition of AMPA receptors that do not produce the same diet-associated side
by decanoic acid at therapeutically relevant con- effects. The recent data outlined here provide a
centrations provided similar inhibitory effects strong case for the antiseizure effects of the MCT
as Perampanel at levels found during clinical ketogenic diet resulting from increases in plasma
use (Rogawski and Hanada, 2013; Ceolin et al., fatty acid concentrations rather than through the
2012). It is interesting to note however, that the production of ketones. Thus, due to the burgeon-
use of Perampanel is limited by neuropsychiatric ing evidence that fatty acids play a primary role
side effects, in particular aggression (Rugg-Gunn, in the diet’s therapeutic effect and despite the
2014; Steinhoff et al., 2014). This effect is not seen potential beneficial roles of ketones, it would per-
during the MCT ketogenic diet, suggesting that haps be more appropriate to rename the diet, the
these side effects may be peculiar to Perampanel MCT diet.
rather than a class effect of drugs that act at Decanoic acid may have beneficial effects
AMPA receptors; thus the development of phar- beyond its action at AMPA receptors. A  recent
maceutical reagents based on the mechanisms of study has identified a role for decanoic acid in
decanoic acid will hopefully not cause this side regulating mitochondrial proliferation (Hughes
effect. et al., 2014). Here, decanoic acid acts through reg-
Finally, the binding site for decanoic acid on ulation of a fatty acid receptor, PPARγ (Malapaka
AMPA receptors was investigated by in silico et al., 2012; Zuckermann et al., 2015), at therapeu-
modeling (Chang et  al., 2016). Using the trans- tically relevant concentrations (250 μM) over a 6-
membrane domains of GluA2, a putative deca- day period in a neuronal cell line and in human
noic acid–binding region was identified in the fibroblasts. Interestingly, this effect was not shown
M3 helix, thought to be involved in regulating by octanoic acid. The consequent increase in
and gating inward currents. This site is distinct mitochondrial load has been suggested to protect
from that reported for Perampanel (Szenasi et al., against seizure induction (Bough et  al., 2006),
2008), suggesting potential differences in the and to protect against mitochondrial dysfunction
effects of decanoic acid and Perampanel on AMPA in epilepsy that has been clearly demonstrated in
receptor function. This binding site is consistent animal models of status epilepticus (Cock et  al.,
with results from electrophysiology experiments 2002) and in patients (Kunz et al., 2000).
described here, although further studies will need Medium chain fatty acids are rapidly metabo-
to confirm this site. lized in vivo (Bach and Babayan, 1982). It remains,
therefore, a key consideration in the design and
IS THERE STILL A ROLE management of seizure-control treatments based
F O R   K E TO N E S on decanoic acid, to ensure maintenance of thera-
IN SEIZURE CONTROL? peutic fatty acid levels. Early studies of medium
Over the last 30  years, many studies have sug- chain fatty acids show considerable variation in
gested a role for ketones in seizure control and plasma fatty acid levels over a 24-hour period (Sills
neuroprotection (Bough and Rho, 2007; Rho et al., 1986b). It remains unclear if a reduced car-
and Stafstrom, 2011). In some types of epilepsy, bohydrate load with the diet is necessary to main-
ketones may provide an alternative energy source tain decanoic acid levels at an effective level, and
for the brain (Kim et al., 2015; Masino et al., 2011), further studies will need to address this in greater
for example in Glut1 deficiency when glucose is detail. However, a corollary of this is that clinicians
poorly transported across the blood-brain bar- using the MCT diet should focus on monitoring
rier. Ketones have also been demonstrated to alter blood fatty acids rather than just ketone levels.
333

Chapter 33: Molecular Mechanism of the Medium Chain Triglyceride (Ketogenic) Diet 333

The discovery of a molecular target for the MCT Chang, P., Walker, M.C., and Williams, R.S. (2014).
diet  also opens the possibility of a pharmaceutic Seizure-induced reduction in PIP3 levels contrib-
approach to replace the diet. Several considerations utes to seizure-activity and is rescued by valproic
are necessary here. Due to the rapid metabolism acid. Neurobiol Dis 62, 296–306.
of decanoic acid, a chemical that provides similar Chang, P., Zuckermann, A.M., Williams, S., Close, A.J.,
Cano-Jaimez, M., McEvoy, J.P., Spencer, J., Walker,
inhibitory activity against AMPA receptors but is
M.C., and Williams, R.S. (2015). Seizure control by
resistant to metabolic degradation may overcome
derivatives of medium chain fatty acids associated
the stringent dietary regime. Developing novel with the ketogenic diet show novel branching-
chemical structures that show enhanced binding to point structure for enhanced potency. J Pharmacol
the target site on the receptor may also reduce the Exp Ther 352, 43–52.
necessary dose of the compound. This would addi- Chang, P., Augustin, A.M., Boddum, K., Williams, S.,
tionally help to reduce the side effects associated Sun, M., Terschak, J.A., Hardege, J.D., Chen, P.E.,
with high fatty acid intake, in particular gastric irri- Walker, M.C., and Williams, R.S.B. (2016). Seizure
tation. Studies outlined here have provided some control by decanoic acid through direct AMPA
evidence for the efficacy of decanoic acid congeners receptor inhibition. Brain 139(Pt 2):431–443. In
in seizure control and in direct inhibition of AMPA press, online release 25 Nov 2015.
receptors. Further studies will be necessary to iden- Cock, H.R., Tong, X., Hargreaves, I.P., Heales, S.J.,
tify suitable candidates for clinical testing, but may Clark, J.B., Patsalos, P.N., Thom, M., Groves,
M., Schapira, A.H., Shorvon, S.D., et  al. (2002).
result in the “diet in a pill.”
Mitochondrial dysfunction associated with neu-
ronal death following status epilepticus in rat.
REFERENCES Epilepsy Res 48, 157–168.
Arai, A.C. (2001). GYKI 52466 has positive modula- Cunliffe, V.T., Baines, R.A., Giachello, C.N., Lin, W.H.,
tory effects on AMPA receptors. Brain Res 892, Morgan, A., Reuber, M., Russell, C., Walker, M.C.,
396–400. and Williams, R.S. (2015). Epilepsy research meth-
Bach, A.C., and Babayan, V.K. (1982). Medium- ods update:  understanding the causes of epileptic
chain triglycerides: an update. Am J Clin Nutr 36, seizures and identifying new treatments using non-
950–962. mammalian model organisms. Seizure 24, 44–51.
Barton, M.E., Klein, B.D., Wolf, H.H., and White, H.S. Dean, H.G., Bonser, J.C., and Gent, J.P. (1989). HPLC
(2001). Pharmacological characterization of the 6 analysis of brain and plasma for octanoic and dec-
Hz psychomotor seizure model of partial epilepsy. anoic acids. Clin Chem 35, 1945–1948.
Epilepsy Res 47, 217–227. DeLorenzo, R.J., Sun, D.A., and Deshpande, L.S.
Bough, K.J., and Rho, J.M. (2007). Anticonvulsant (2005). Cellular mechanisms underlying acquired
mechanisms of the ketogenic diet. Epilepsia epilepsy: the calcium hypothesis of the induction
48, 43–58. and maintainance of epilepsy. Pharmacol Ther
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., 105, 229–266.
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Deshpande, L.S., Lou, J.K., Mian, A., Blair, R.E.,
Y., Geiger, J.D., and Dingledine, R.J. (2006). Sombati, S., Attkisson, E., and DeLorenzo, R.J.
Mitochondrial biogenesis in the anticonvulsant (2008). Time course and mechanism of hippocam-
mechanism of the ketogenic diet. Ann Neurol 60, pal neuronal death in an in vitro model of status
223–235. epilepticus: role of NMDA receptor activation and
Ceolin, L., Bortolotto, Z.A., Bannister, N., Collingridge, NMDA dependent calcium entry. Eur J Pharmacol
G.L., Lodge, D., and Volianskis, A. (2012). A novel 583, 73–83.
anti-epileptic agent, perampanel, selectively inhib- Gottlicher, M., Minucci, S., Zhu, P., Kramer, O.H.,
its AMPA receptor-mediated synaptic transmis- Schimpf, A., Giavara, S., Sleeman, J.P., Lo, C.F.,
sion in the hippocampus. Neurochem Int 61, Nervi, C., Pelicci, P.G., et al. (2001). Valproic acid
517–522. defines a novel class of HDAC inhibitors inducing
Chang, P., Orabi, B., Deranieh, R.M., Dham, M., differentiation of transformed cells. EMBO J 20,
Hoeller, O., Shimshoni, J.A., Yagen, B., Bialer, 6969–6978.
M., Greenberg, M.L., Walker, M.C., et  al. (2012). Gurvich, N., Tsygankova, O.M., Meinkoth, J.L., and
The antiepileptic drug valproic acid and other Klein, P.S. (2004). Histone deacetylase is a target
medium-chain fatty acids acutely reduce phos- of valproic acid-mediated cellular differentiation.
phoinositide levels independently of inositol in Cancer Res 64, 1079–1086.
Dictyostelium. Dis Model Mech 5, 115–124. Haidukewych, D., Forsythe, W.I., and Sills, M. (1982).
Chang, P., Terbach, N., Plant, N., Chen, P.E., Walker, Monitoring octanoic and decanoic acids in plasma
M.C., and Williams, R.S. (2013). Seizure control from children with intractable epilepsy treated
by ketogenic diet-associated medium chain fatty with medium-chain triglyceride diet. Clin Chem
acids. Neuropharmacology 69, 105–114. 28, 642–645.
334

334 section IV: Ketone-Based Metabolism

Hughes, S.D., Kanabus, M., Anderson, G., Hargreaves, Cross, J.H. (2009). A randomized trial of classical
I.P., Rutherford, T., O’Donnell, M., Cross, J.H., and medium-chain triglyceride ketogenic diets in
Rahman, S., Eaton, S., and Heales, S.J. (2014). the treatment of childhood epilepsy. Epilepsia 50,
The ketogenic diet component decanoic acid 1109–1117.
increases mitochondrial citrate synthase and com- Neal, E.G., and Cross, J.H. (2010). Efficacy of dietary
plex I activity in neuronal cells. J Neurochem 129, treatments for epilepsy. J Hum Nutr Diet 23,
426–433. 113–119.
Huttenlocher, P.R., Wilbourn, A.J., and Signore, J.M. Phiel, C.J., Zhang, F., Huang, E.Y., Guenther, M.G.,
(1971). Medium-chain triglycerides as a therapy Lazar, M.A., and Klein, P.S. (2001). Histone
for intractable childhood epilepsy. Neurology 21, deacetylase is a direct target of valproic acid, a
1097–1103. potent anticonvulsant, mood stabilizer, and terato-
Jentink, J., Loane, M.A., Dolk, H., Barisic, I., Garne, E., gen. J Biol Chem 276, 36734–36741.
Morris, J.K., and de Jong-van den Berg LT (2010). Rektor, I. (2013). Perampanel, a novel, non-
Valproic acid monotherapy in pregnancy and competitive, selective AMPA receptor antago-
major congenital malformations. N Engl J Med nist as adjunctive therapy for treatment-resistant
362, 2185–2193. partial-onset seizures. Expert Opin Pharmacother
Kim, D.Y., Simeone, K.A., Simeone, T.A., Pandya, J.D., 14, 225–235.
Wilke, J.C., Ahn, Y., Geddes, J.W., Sullivan, P.G., Rho, J.M., and Stafstrom, C.E. (2011). The ketogenic
and Rho, J.M. (2015). Ketone bodies mediate anti- diet: what has science taught us? Epilepsy Res
seizure effects through mitochondrial permeabil- 100(3), 210–217.
ity transition. Ann Neurol 78(1), 77–87. Rogawski, M.A., and Hanada, T. (2013). Preclinical
Koren, G., Nava-Ocampo, A.A., Moretti, M.E., pharmacology of perampanel, a selective non-
Sussman, R., and Nulman, I. (2006). Major mal- competitive AMPA receptor antagonist. Acta
formations with valproic acid. Can Fam Physician Neurol Scand Suppl (197), 19–24.
52, 441–442, 444, 447. Rowley, N.M., and White, H.S. (2010). Comparative
Kunz, W.S., Kudin, A.P., Vielhaber, S., Blumcke, I., anticonvulsant efficacy in the corneal kindled
Zuschratter, W., Schramm, J., Beck, H., and Elger, mouse model of partial epilepsy: correlation with
C.E. (2000). Mitochondrial complex I  deficiency other seizure and epilepsy models. Epilepsy Res
in the epileptic focus of patients with temporal 92, 163–169.
lobe epilepsy. Ann Neurol 48, 766–773. Rugg-Gunn, F. (2014). Adverse effects and safety
Levy, R.G., Cooper, P.N., and Giri, P. (2012). Ketogenic profile of perampanel:  a review of pooled data.
diet and other dietary treatments for epilepsy. Epilepsia 55 (Suppl 1), 13–15.
Cochrane Database Syst Rev 3, CD001903. Russo, E., Gitto, R., Citraro, R., Chimirri, A., and De,
Likhodii, S.S., Musa, K., Mendonca, A., Dell, C., S.G. (2012). New AMPA antagonists in epilepsy.
Burnham, W.M., and Cunnane, S.C. (2000). Expert Opin Investig Drugs 21, 1371–1389.
Dietary fat, ketosis, and seizure resistance in rats Sada, N., Lee, S., Katsu, T., Otsuki, T., and Inoue,
on the ketogenic diet. Epilepsia 41, 1400–1410. T. (2015). Epilepsy treatment:  targeting LDH
Liu, Y.M. (2008). Medium-chain triglyceride (MCT) enzymes with a stiripentol analog to treat epilepsy.
ketogenic therapy. Epilepsia 49 (Suppl 8), 33–36. Science 347, 1362–1367.
Loscher, W. (1999). Animal models of epilepsy and epi- Shaltiel, G., Mark, S., Kofman, O., Belmaker, R.H.,
leptic seizures. In Antiepileptic Drugs, Pharmacology and Agam, G. (2007). Effect of valproate deriva-
and Therapeutics, M.J. Eadie and F.J.E. Vajda, eds. tives on human brain myo-inositol-1-phosphate
(Berlin: Springer-Verlag), pp. 19–62. (MIP) synthase activity and amphetamine-
Malapaka, R.R., Khoo, S., Zhang, J., Choi, J.H., Zhou, induced rearing. Pharmacol Rep 59, 402–407.
X.E., Xu, Y., Gong, Y., Li, J., Yong, E.L., Chalmers, Sills, M.A., Forsythe, W.I., and Haidukewych, D.
M.J., et  al. (2012). Identification and mechanism (1986a). Role of octanoic and decanoic acids in the
of 10-carbon fatty acid as modulating ligand of control of seizures. Arch Dis Child 61, 1173–1177.
peroxisome proliferator-activated receptors. J Biol Sills, M.A., Forsythe, W.I., Haidukewych, D.,
Chem 287, 183–195. MacDonald, A., and Robinson, M. (1986b). The
Masino, S.A., Li, T., Theofilas, P., Sandau, U.S., Ruskin, medium chain triglyceride diet and intractable
D.N., Fredholm, B.B., Geiger, J.D., Aronica, E., and epilepsy. Arch. Dis. Child 61, 1168–1172.
Boison, D. (2011). A ketogenic diet suppresses sei- Steinhoff, B.J., Hamer, H., Trinka, E., Schulze-
zures in mice through adenosine A(1) receptors. J Bonhage, A., Bien, C., Mayer, T., Baumgartner, C.,
Clin Invest 121, 2679–2683. Lerche, H., and Noachtar, S. (2014). A multicenter
Meldrum, B.S., and Rogawski, M.A. (2007). Molecular survey of clinical experiences with perampanel in
targets for antiepileptic drug development. real life in Germany and Austria. Epilepsy Res 108,
Neurotherapeutics 4, 18–61. 986–988.
Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S., Szenasi, G., Vegh, M., Szabo, G., Kertesz, S., Kapus, G.,
Edwards, N., Fitzsimmons, G., Whitney, A., and Albert, M., Greff, Z., Ling, I., Barkoczy, J., Simig,
335

Chapter 33: Molecular Mechanism of the Medium Chain Triglyceride (Ketogenic) Diet 335

G., et  al. (2008). 2,3-benzodiazepine-type AMPA Williams, R.S.B. (2005). Pharmacogenetics in
receptor antagonists and their neuroprotective model systems:  Defining a common mecha-
effects. Neurochem Int 52, 166–183. nism of action for mood stabilisers. Prog
Thavendiranathan, P., Mendonca, A., Dell, C., Neuropsychopharmacol Biol Psychiatry 29,
Likhodii, S.S., Musa, K., Iracleous, C., Cunnane, 1029–1037.
S.C., and Burnham, W.M. (2000). The MCT keto- Williams, R.S.B., Cheng, L., Mudge, A.W., and
genic diet:  effects on animal seizure models. Exp Harwood, A.J. (2002). A common mechanism of
Neurol 161, 696–703. action for three mood-stabilizing drugs. Nature
Thio, L.L., Wong, M., and Yamada, K.A. (2000). Ketone 417, 292–295.
bodies do not directly alter excitatory or inhibitory Wlaz, P., Socala, K., Nieoczym, D., Luszczki, J.J.,
hippocampal synaptic transmission. Neurology Zarnowska, I., Zarnowski, T., Czuczwar, S.J.,
54, 325–331. and Gasior, M. (2012). Anticonvulsant profile of
Vaden, D.L., Ding, D., Peterson, B., and Greenberg, caprylic acid, a main constituent of the medium-
M.L. (2001). Lithium and valproate decrease inosi- chain triglyceride (MCT) ketogenic diet, in mice.
tol mass and increase expression of the yeast INO1 Neuropharmacology 62, 1882–1889.
and INO2 genes for inositol biosynthesis. J Biol Xu, X., Muller-Taubenberger, A., Adley, K.E.,
Chem 276, 15466–15471. Pawolleck, N., Lee, V.W., Wiedemann, C., Sihra,
Van Den Pol, A.N., Obrietan, K., and Belousov, A. T.S., Maniak, M., Jin, T., and Williams, R.S. (2007).
(1996). Glutamate hyperexcitability and seizure- Attenuation of phospholipid signaling provides a
like activity throughout the brain and spinal cord novel mechanism for the action of valproic acid.
upon relief from chronic glutamate receptor block- Eukaryot Cell 6, 899–906.
ade in culture. Neuroscience 74, 653–674. Zuckermann, A.M., La Ragione, R.M., Baines, D.L.,
Walker, M.C., and Williams, R.S. (2015). New and Williams, R.S. (2015). Valproic acid protects
experimental therapies for status epilepticus against haemorrhagic shock-induced signalling
in preclinical development. Epilepsy Behav 49, changes via PPARgamma activation in an in vitro
290–293. model. Br J Pharmacol 172(22), 5306–5317.
336

34
Triheptanoin in Epilepsy and Beyond
KARIN BORGES, PHD

INTRODUCTION metabolites, for example, epilepsy (Alvestad et


TO   A NA P L E RO S I S A N D al., 2008; Melo et al., 2005; Smeland et al., 2013;
T R I H E P TA N O I N Willis et al., 2010), stroke (Haberg et al., 2009), and
Glucose, under nonfasting conditions, is usu- amyotrophic lateral sclerosis (ALS) (Niessen et al.,
ally the main fuel for the CNS. Glycolysis, which 2007). This is expected to lead to low oxaloacetate
occurs in cytosol, is the main pathway of metabolic levels, which then can reduce the binding of ace-
breakdown of glucose into pyruvate. When there tyl-CoA by citrate synthase, its oxidation, and ulti-
is limited oxygen, glycolysis produces lactate, oth- mately decrease ATP production.
erwise acetyl-CoA is produced via the pyruvate Various enzymes partake in the refilling of the
dehydrogenase pathway for further oxidation in TCA cycle. In the CNS, carboxylation of pyru-
the tricarboxylic acid (TCA) cycle in mitochon- vate to oxaloacetate (number 1 in Figure 34.2)
dria (Figure 34.2). In aerobic metabolism, after is the main refilling pathway, thought to be tak-
citrate synthase transfers the acetyl-group onto ing place in astrocytes (Patel, 1974). Also, several
oxaloacetate forming citrate, the TCA cycle oxi- enzymes facilitate the formation of 2-oxoglutarate
dizes acetyl-CoA to two carbon dioxide molecules (also called α-ketoglutarate) from glutamate.
in a series of chemical reactions. At the same This includes glutamate dehydrogenase, which
time, the TCA cycle provides the electron trans- forms 2-oxoglutarate and ammonia from gluta-
port chain with reducing equivalents, which then mate and vice versa (2 in Figure 34.2). The reac-
produces the majority of ATP during aerobic con- tion catalyzed by glutamic pyruvic transaminases
ditions (Figure 34.1). The TCA cycle is also part (alanine aminotransferases), pyruvate + glutamate
of many other key metabolic pathways. The TCA ↔ 2-oxoglutarate + alanine refills the cycle with
cycle intermediates containing four or five carbons 2-oxoglutarate (3 in Figure 34.2). In addition,
not only are part of the cycle involved in energy aspartate transaminase (also called glutamic oxa-
production but also are used for other metabolic loacetic transaminase) can bypass part of the TCA
pathways, for example, synthesis of various amino cycle to produce oxaloacetate from 2-oxoglutarate
acids and neurotransmitters, such as glutamate in the reaction: Asp + 2-OG ↔ OAA + Glu (4 in
and γ-aminobutyric acid (GABA). It is therefore Figure 34.2).
important that C4 intermediates of the TCA cycle Another anaplerotic pathway, the propionyl-
get “refilled” (anaplerosis) (Brunengraber and Roe, CoA carboxylation pathway (5 in Figure 34.2), has
2006; Hassel, 2000; Kornberg, 1966). Anaplerosis largely been studied in peripheral tissues (Deng
is required for TCA cycling to occur, as it enhances et  al., 2009; Martini et  al., 2003; Nuutinen et  al.,
acetyl-CoA entry into the TCA cycle by providing 1981; Owen et  al., 2002; Reszko et  al., 2003). It
oxaloacetate, thus increasing TCA cycling and ATP involves the carboxylation of propionyl-CoA to
production. Moreover with increased amounts of methylmalonyl-CoA by propionyl-CoA carboxyl-
TCA cycle metabolites, more substrate for com- ase (EC 6.4.1.3). Methylmalonyl-CoA epimerase
plex II can be produced and reduction of NADH+ (EC 5.1.99.1) and methylmalonyl-CoA mutase (EC
will be enhanced, which again will contribute to 5.4.99.2) then produce succinyl-CoA. The branched
ATP production. Please note that the TCA cycle chain amino acids, isoleucine and valine, as well
can also be short-circuited and still produce ATP, as uneven fatty acids are anaplerotic via this path-
for example with substrate entry as 2-oxoglutarate way, all providing propionyl-CoA (Figure 34.1).
or succinate. In many different models of disease Treatment with triheptanoin, the triglyceride of
there is evidence of reduced amounts of TCA cycle heptanoate (C7 fatty acid), appears to currently be
337

Chapter 34: Triheptanoin in Epilepsy and Beyond 337

Glucose Triheptanoin

Acetyl~CoA Heptanoate

Citrate

OAA Propionyl~CoA
2-OG
NADH
Succinyl~CoA

Fumarate Succinate
ATP
O2 H2O ADP
II
I III IV V

FIGURE 34.1 Boosting the TCA cycling capacity when in need via the propionyl-CoA carboxylation pathway can
increase ATP production. This schematic includes the reduction of NAD+ to NADH by the TCA cycle, which then feeds
electrons into complex I  of the mitochondrial electron transport chain, while succinate directly feeds electrons into
complex II. Ultimately ATP is produced by ATP synthase.

the least toxic treatment when fueling this pathway are metabolized to their CoA adducts via medium
to a large extent. Unlike the branched chain amino chain acyl-CoA synthetases or 3-oxoacid CoA
acids, triheptanoin does not overload the body transferase, respectively, and then converted into
with nitrogen. In addition, providing the uneven the main TCA cycle fuel acetyl-CoA and propio-
medium chain fat as a triglyceride avoids excessive nyl-CoA. After carboxylation the latter can refill
levels of sodium or acid, which otherwise could the TCA cycle with succinyl-CoA (see above) and
challenge physiological homeostasis. thus promote acetyl-CoA oxidation and ATP pro-
Triheptanoin is a tasteless oil and can be mixed duction (Figure 34.2).
with various foods or made into an emulsion. As a
medium chain triglyceride, it is hydrolyzed in the E N E R G Y M E TA B O L I S M
gastrointestinal tract, and due to its lipophilicity IN EPILEPSY
the free medium chain heptanoate is thought to An epileptic seizure or hypersynchronous acti-
diffuse directly into blood and mitochondria of all vation of neuronal networks can be initiated by
tissues. This is unlike long chain fatty acids, which imbalances between excitation and inhibition. In
are much more slowly metabolized, because they addition, impairments in energy metabolism can
first enter the lymph and require various trans- also cause seizures and/or contribute to epilepsy.
port proteins in the blood and for final transport Numerous studies have attempted to shed light
into mitochondria for β-oxidation. Similar to even on energy metabolism in brains of patients with
medium chain fats, heptanoate is converted by the epilepsy and rodent epilepsy models. Assessments
liver to “ketones.” While even chain fats octanoate of metabolic functions or metabolite levels have
and decanoate (C8 and C10) are turned into “C4 revealed dysfunction in energy metabolism in
ketones,” β-hydroxybutyrate and acetoacetate, hep- patients with temporal lobe and extra-temporal
tanoate is metabolized to “C5 ketones,” β-hydroxy- lobe epilepsy (reviewed in Li et al., 2000; Pan et al.,
pentanoate and β-ketopentanoate (Brunengraber 2005; Pan et al., 2008). Similar changes have been
and Roe, 2006; Gu et al., 2010; Kinman et al., found in rodent epilepsy models (Alvestad et  al.,
2006; Roe and Mochel, 2006; Roe et al., 2002). 2008; Melo et al., 2010; Melo et al., 2005; Smeland
After release into the blood, C4 and C5 ketones are et al., 2013; Willis et al., 2010) (Figure 34.3).
taken up into cells by monocarboxylate transport- Furthermore, mitochondrial dysfunction and
ers. Regarding the crossing of the blood-brain bar- mutations within mitochondrial constituents have
rier, monocarboxylate transporters of the MCT1 been described (Kann and Kovacs, 2007; Kudin
type (Broer et al., 1998; Meredith and Christian, et al., 2009; Waldbaum and Patel, 2010), which can
2008) are thought to transport C5 ketones into also contribute to energetic imbalances contribut-
the brain, while heptanoate is more likely to enter ing to seizures.
the brain via diffusion (Oldendorf, 1973). Once in Many studies in the literature discuss interictal
cells, both heptanoate and C5 ketone molecules glucose hypometabolism in patients with temporal
338

Glucose

Pyruvate

CO2 1 Acetyl~CoA
Glutamine
Ala
3
Citrate
Glutamate
OAA 2
Aspartate GABA
2-OG OAA
4

Malate Aspartate

Fumarate Succinyl~CoA Heptanoate,


C5 ketones,
Succinate Ile, Val
Methylmalonyl~CoA
5 CO2
Propionyl~CoA

FIGURE 34.2 The TCA cycle and anaplerotic pathways. The numbers indicate five different anaplerotic pathways.
1 Pyruvate carboxylase forms oxaloactetate by carboxylastion of pyruvate. 2 denotes the activity of glutamate
dehydrogenase, which metabolizes glutamate into 2-oxoglutarate and ammonia and vice versa. 3 shows the reversible
reaction catalyzed by glutamic pyruvic transaminases (also called alanine aminotransferases):  pyruvate + glutamate
<=>2-oxoglutarate + alanine. 4 aspartate transaminase (also called glutamic oxaloacetic transaminase) produces
oxaloacetate from aspartate, while transferring the amino group onto 2-oxoglutarate forming glutamate and vice versa.
5 denotes the activity of the propionyl-CoA carboxylation pathway forming succinyl-CoA. 2-OG—2-oxoglutarate.

(a) MEST test (b) 2nd hit PTZ


18 90
*
**
P TZ threshold (mg/kg i.v.)

80
16
70
CC50 (mA)

14
60

12 50

0 0
CON 35% Trih CON 35% Trih
6 weeks 3 weeks
(c)
Absence seizure model: GABAAR φ 2 (R43Q)

CON

0.5 mV

Trih 2s

FIGURE  34.3 Anticonvulsant effects of oral triheptanoin in three models. (a) Triheptanoin increased the critical
current at which 50% of mice seize (CC50) in the maximal electroshock threshold test (Thomas et al., 2012). (b) It also
increased the threshold to PTZ-induced tonic seizures in chronically epileptic mice (Willis et al., 2010) and (c) decreased
spike wave discharges in a mouse model for absence seizures (Kim et al., 2013).
339

Chapter 34: Triheptanoin in Epilepsy and Beyond 339

lobe epilepsy (TLE), which is a common epilepsy in the brain, is a biochemically valid approach to
type in adults that is largely treatment resistant. improve energy metabolism in “epileptic” brains
In TLE, results from positron emission tomogra- (Brunengraber and Roe, 2006). Thus, we tested
phy with 18F-labeled fluorodeoxyglucose (18FDG) the metabolic effects and anticonvulsant profile of
have been interpreted to show interictal glucose triheptanoin treatment as an anaplerotic approach,
hypometabolism (Arnold et al., 1996; Henry et al., which at that time had already been explored in
1993; Henry et al., 1990). However, care needs to a few animal models and in patients with various
be taken when evaluating these studies, as results genetic metabolic disorders. Please also note two
only revealed decreased 18FDG amounts in cer- reviews on the effects of other approaches to sup-
tain brain areas, which reflects enhanced uptake of plement TCA cycle substrates (Kovac et al., 2013;
18
FDG. Levels of 18FDG cannot be used to assess Tan et al., 2015).
glucose metabolism, because after it is metabo-
lized by hexokinase 18FDG cannot be further T R I H E P TA N O I N
metabolized via glycolysis. Similar to these stud- A LT E R S B R A I N E N E R G Y
ies in epilepsy patients, decreased uptake of 14C- M E TA B O L I S M I N   VA R I O U S
deoxyglucose was found in the chronic phase of SETTINGS, INCLUDING
the lithium-pilocarpine adult rat model of TLE EPILEPSY MODELS
(Dube et al., 2001). Higher total glucose amounts Few publications shed light on the metabolic
were found in extracts from brain areas involved effects of triheptanoin or its metabolite heptanoate
in seizure activity compared to tissue from con- on the brain.
trol rats (Melo et al., 2005). However, total glucose An elegant study by the group of Professor
amounts by themselves are difficult to interpret, as Pascual (Marin-Valencia et  al., 2013), infused
they depend on uptake and rates of metabolism 13
C-labeled 5,6,7-heptanoate into the jugular vein
and no alterations in total glucose amounts were of mice. The 13C-carbons were largely found in
found in the chronic stage of the mouse pilocar- brain glutamine, but not glutamate, indicating that
pine model (Smeland et  al., 2013). In summary, astrocytes primarily metabolize heptanoate and its
decreased glucose uptake appears to be common C5 ketone metabolites.
in some “epileptic” tissues, but there is little knowl- McDonald et  al. (2014) compared the
edge about the metabolism of this major fuel. amounts of various hippocampal metabolites
Given the anticonvulsant efficacy of dietary treat- within energy metabolism pathways (adeno-
ments, such as the ketogenic and modified Atkins sine nucleotides, NAD+, NADH, and NADPH),
diets, there is a critical need for more knowledge glycolysis, the pentose phosphate pathway, and
about glycolysis in epilepsy. the TCA cycle in healthy mice after feeding tri-
Intermediates of the TCA cycle, such as heptanoin and trioctanoin (the triglyceride of
2-oxogluarate and oxaloacetate, are precursors of octanoate) for 3 weeks. While the even medium
the amino acids and neurotransmitters glutamate, chain triglyceride altered the levels of various
GABA, and aspartate. Decreased levels of TCA metabolites, no significant changes were found
intermediates and amino acids have been found in with triheptanoin, indicating that it would have
the chronic seizure stage of rat and mouse epilepsy few metabolic side effects.
models, which display recurrent spontaneous A recent study in patients with Huntington’s
seizures (Alvestad et  al., 2008; Melo et  al., 2005; disease patients found that 1-month triheptanoin
Smeland et al., 2013; Willis et al., 2010). For exam- treatment corrected the energetic response to
ple, in the mouse pilocarpine epilepsy model, we brain activation (Adanyeguh et  al., 2015). In
found lower forebrain levels of malate, aspartate, untreated Huntington’s disease patients, a visual
and acetyl- and propionyl-CoA during the chronic stimulus fails to alter the ratio of inorganic phos-
epileptic stage compared with mice without sei- phate to phosphocreatine, while healthy control
zures (Willis et  al., 2010). While several other and treated patients showed an increase in this
TCA cycle intermediates were not quantified, ratio, indicating that triheptanoin could restore
TCA intermediate levels seemed to be decreased metabolism of high-energy phosphates.
overall, which can result in decreased oxidative Two studies (Hadera et al., 2014; Willis et
phosphorylation. al., 2010) aimed to increase the understanding
Based on this knowledge, providing additional of the metabolic effects of triheptanoin treat-
alternative anaplerotic fuel, which can increase ment in mouse “epileptic” brain tissue. In the
the amounts of C4 intermediates of the TCA cycle pilocarpine model, mice that experience status
340

340 section IV: Ketone-Based Metabolism

epilepticus (SE) show one motor seizure per day seizures (Tan et al., 2007), namely mice with a
on average in the chronic stage of the model, missense (R43Q) mutation in the GABAA recep-
while mice that did not get SE (no SE) do not tor γ2 subunit, triheptanoin decreased the num-
show any seizure activity or neuropathological ber and duration of spike wave discharges (SWD),
changes (Benson et al., 2015; Borges et al., 2003). resulting in a halved time with seizures (Kim et
Willis et al. (2010) compared the levels of various al., 2013) (Figure 34.3C). In chronically epileptic
forebrain metabolites related to the TCA cycle in mice after pilocarpine-induced SE, triheptanoin
the chronic stage of this model. Forebrain lev- reproducibly increased the pentylenetetrazole
els of malate and propionyl-CoA were reduced (PTZ) seizure threshold (Figure 34.3B). Efficacy
in SE versus no SE mice on control diet, but this in this model suggests efficacy against drug-
was prevented by triheptanoin feeding, indicat- resistant seizures, based on the finding that a sim-
ing that triheptanoin can improve a low capacity ilar second hit rat model is resistant to valproate,
of the TCA cycle. Hadera et al. (2014) followed phenytoin, and phenobarbital (Blanco et al., 2009;
the metabolism of [1,2-13C]glucose in the same Borges and Sonnewald, 2011). In the acute max-
mouse model to be able to evaluate the activities imal electroshock threshold test (MEST), a test
of pyruvate dehydrogenase and pyruvate carbox- for the efficacy against generalized seizures, we
ylase, the latter of which provides C4 carbons to observed a reproducible increase of the critical
the TCA cycle. The analyses of 13C label incor- current at which 50% of mice seize (Thomas et al.,
poration into TCA cycle intermediates showed 2012) (CC50, Fig 34.3A). Effects in other acute
that the percentage of enrichment for two 13C seizure models have been variable, for example,
atoms in malate, citrate, succinate, and GABA in the 6-Hz model the threshold to motor sei-
was reduced in control treated SE mice versus no zures was only elevated in some experiments, but
SE mice. Except for succinate, triheptanoin feed- not others (McDonald et al., 2014; Thomas et al.,
ing alleviated these reductions in SE mice, which 2012). Similarly, anticonvulsant effects in models
provides additional evidence that triheptanoin using the GABAA receptor channel blockers, fluo-
can increase the metabolism of glucose via the rothyl or PTZ, have been inconsistent (Thomas et
TCA cycle. The author believes that an adequate al., 2012). This lack of consistent effects in acute
ATP supply is very important in “epileptic” tis- mouse models is not surprising, because energy
sue to keep neuronal membrane potentials stable metabolism in healthy mice is likely to be opti-
and prevent seizure generation. Taken together, mal and unlikely to require additional fuel and/
all studies are consistent with the brain being or anaplerosis. In summary, triheptanoin’s anti-
able to metabolize heptanoate or “C5 ketones,” convulsant profile suggests efficacy against a vari-
and improvements in cases of impaired energy ety of seizure types, including focal and general
metabolism could be detected. motor seizures, absence seizures, and possibly
pharmacoresistant motor seizures (Smith et al.,
ANTICONVULSANT EFFECTS 2007; White, 2003).
O F   T R I H E P TA N O I N A N D Currently, there are three clinical trials of tri-
CLINICAL TRIALS heptanoin in adult and children with medically
Triheptanoin shows a unique anticonvulsant pro- refractory epilepsy in Australia (http://www.
file. It was found to be anticonvulsant in various anzctr.org.au/). The first phase IIa randomized
mouse seizure models (McDonald et al., 2014; double-blind placebo controlled study to evaluate
Thomas et al., 2012; Willis et al., 2010) (Figure the safety and tolerability of oral triheptanoin as an
34.3), with efficacy in three chronic mouse sei- add-on treatment to adolescent and adult patients
zure models. In the chronic corneal kindling with medically refractory epilepsy took place with
model we found a reproducible delay in the kin- Professor O’Brien as the lead doctor at the Royal
dling process in CF1 mice (Willis et al., 2010), Melbourne Hospital. Results are currently being
which is similar to effects found with established analyzed. Patients who took part in this trial have
anticonvulsant drugs in the rat kindling model, been invited to continue in an open-label exten-
namely phenobarbital and low concentrations sion study of oral triheptanoin as an add-on treat-
of valproate (Brandt et al., 2006; Matagne et al., ment. This study recruited patients in 2015/2016
2008; Silver et al., 1991). Protective effects of var- and is going well. In addition, a trial in Brisbane is
ious compounds in kindling models correlate evaluating the effects of triheptanoin in children
well with efficacy in humans against absence sei- ages 3–18 with medically refractory epilepsy and
zures. Also, in a mouse model for genetic absence has stopped recruiting.
341

Chapter 34: Triheptanoin in Epilepsy and Beyond 341

T R I H E P TA N O I N A N D The pediatric leukodystrophy Canavan dis-


OT H E R D I S O R D E R S ease is caused by mutations in aspartoacylase, an
enzyme that is largely found in oligodendrocytes
Metabolic Disorders: Glucose Transporter and catalyzes the hydrolysis of neuronally derived
Type 1 Deficiency, Long Chain Fatty N-acetylaspartate to provide acetyl groups for lipid
Acid Oxidation Disorders, Pyruvate synthesis. In nur7 mutant mice containing a non-
Decarboxylase Deficiency sense mutation in the aspartoacylase gene, early
While this review focused on epilepsies of vari- triheptanoin treatment prevented the loss of oli-
ous or unknown etiologies, triheptanoin is also godendrocytes, dysmyelination, and impairments
being developed as a new treatment for D-glu- in motor function (Francis et al., 2014). The early
cose transporter type 1 deficiency (Pascual et al., loss of various brain metabolites in this model,
2014). In this genetic disorder glucose cannot be such as ATP and acetyl-, malonyl-, and propionyl-
taken up into the brain, which results in epilep- CoA, was largely prevented, indicating that tri-
tic seizures around age 3–4 and later in paroxys- heptanoin can rescue metabolic deficits also in
mal exercise-induced dyskinesias, which can be oligodendrocytes.
very disabling (Klepper and Leiendecker, 2007; The brain infarct area after middle cere-
Suls et al., 2009). The treatments for this disor- bral artery occlusion (MCAO), a mouse model
der are discussed in an other chapter in this book of stroke, was smaller when mice were fed tri-
(Klepper, chapter 5). heptanoin before the insult (Schwarzkopf et  al.,
Originally, triheptanoin was given to patients 2015). In addition, mitochondrial functions were
with rare inborn metabolic disorders, including preserved in mitochondria isolated from brains
long chain fatty acid oxidation disorders due to 1 hour after onset of MCAO (Schwarzkopf et al.,
deficiencies in different enzymes, namely car- 2015), indicating that improved mitochondrial
nitine palmitoyltransferase I  or II, very long- energetic functions contribute to triheptanoin’s
chain acyl-CoA dehydrogenase, L-3-hydroxy neuroprotective effects.
acyl-CoA dehydrogenase, and mitochondrial Similarly, in the ALS mouse model overex-
trifunctional protein (Roe et al., 2002; Roe et al., pressing the human SOD1 G93A mutation, tri-
2008). The use of dietary or body fat as energy heptanoin treatment resulted in 33% less motor
sources is preempted in these disorders, which neuron loss in the L4-L5 spinal cord, and loss of
can result in severe clinical problems, such as motor functions was significantly delayed (Tefera
cardiomyopathy, intermittent rhabdomyolysis, et al. PLOS One, in press).
hypoglycemia, and sudden death. Treatment with A study in an Alzheimer’s disease model indi-
traditional medium chain triglycerides has been cated that triheptanoin in the context of a keto-
unsatisfactory in the past, while triheptanoin as genic diet increased the expression of the mRNA
an alternatively medium chain and anaplerotic levels of Sirt1, Pparg, Sod1, and Sod2 (Aso et  al.,
fuel, which is also gluconeogenic, can decrease 2013). As sirtuin 1 and PPARγ are involved in the
hospitalizations due to major clinical manifesta- regulation of lipid and glucose metabolism as well
tions and incidences in hypoglycemia (Roe and as mitochondrial respiration and oxidative stress,
Brunengraber, 2015; Roe and Mochel, 2006; Roe this suggests that the neuroprotective effects of
et al., 2002; Vockley et al., 2015). Another meta- triheptanoin may also include other mechanisms
bolic disorder that may benefit from triheptanoin that can improve energy metabolism, such as
is pyruvate carboxylase deficiency (Breen et  al., reduced oxidative stress and preserved mitochon-
2014; Mochel et al., 2005). drial functions.
Adult polyglucosan body disease (APBD) is a
Other Disorders: Neurological rare autosomal recessive disorder due to partial
Conditions, Muscle Disorders, deficiency of the glycogen brancher enzyme (GBE)
Cardiac Hypertrophy and leads to deposition of polyglucosan bodies in
Triheptanoin appears to be a promising treat- neurons and glia. Similar to other glycogenoses,
ment for other neurological disorders, including for example, Pompe’s disease, in which glycogen
Canavan disease, stroke, ALS, Alzheimer’s and cannot be metabolized, it is thought that the pro-
Huntington’s disease, autism, and glycogenoses. In gressive deposition of polysaccharides causes cells
three neurological disorders models, oral trihep- to be disrupted. The APBD patients show gradual
tanoin feeding was found to prevent neuronal cell progression with difficulty walking, impaired bal-
death, and it is conceivable that increased energy ance, neurogenic bladder, weakness, and, in about
levels can protect cells from degeneration. half of the cases, dementia. Five APBD patients
342

342 section IV: Ketone-Based Metabolism

treated with triheptanoin experienced stabiliza- rare metabolic enzyme deficiencies, and many
tion of disease progression or some functional patients with fatty acid oxidation disorders depend
improvement (Roe et al., 2010). on its availability. Within the last decade new
There is increasing hope that Huntington’s research has shown promising effects of trihep-
disease patients might benefit from long-term tanoin in a variety of human disorders and their
triheptanoin therapy, as there is evidence that tri- animal models, including epilepsy. Larger-scale
heptanoin improves energy metabolism in patients controlled clinical trials are now needed to prove
in the CNS and also skeletal muscle (Adanyeguh triheptanoin’s safety and tolerability, and there is
et  al., 2015; Mochel et  al., 2010). Moreover, hope that a variety of beneficial effects reported so
improvements in muscle function were found in far can be confirmed.
skeletal muscle in a Rett syndrome model (Park
et  al., 2014)  and rat heart (Nguyen et  al., 2015). DISCLOSURE/CONFLICT
In Sydney, a pilot clinical study is currently taking OF INTEREST
place with patients with inclusion body myositis KB applied for two full US patents on the use of
and Pompe’s disease (http://www.anzctr.org.au/; triheptanoin for epilepsy and ALS.
Corbett et al., 2015).
Among the autism spectrum disorders, Rett AC K N OW L E D G M E N T S
syndrome is a genetic disorder largely caused I am grateful for funding by the American
by mutations in the X-linked gene for the tran- Epilepsy Foundation, Parents against Childhood
scription factor methyl-CpG binding protein 2 Epilepsy, UniQuest, the Thrasher Research Fund,
(MeCP2), resulting in severe disability regarding and Ultragenyx Pharmaceuticals, Inc., for funding
cognitive and motor function. In male MeCP2- ongoing clinical trials of triheptanoin in adults and
deficient mice triheptanoin increased life span and children with treatment resistant epilepsy. I  also
improved social interaction and motor function thank the Australian National Health and Medical
as well as mitochondrial morphology in skeletal Research Council (grant 1044407) for funding my
muscle (Park et  al., 2014). The mutant mice also laboratory research.
showed increased adiposity and lower glucose tol-
erance and insulin sensitivity, which all improved REFERENCES
with triheptanoin treatment, indicating that this Adanyeguh, I.M., Rinaldi, D., Henry, P.G., Caillet, S.,
medium chain triglyceride can counteract obesity Valabregue, R., Durr, A., and Mochel, F. (2015).
and normalize glucose metabolism. Triheptanoin improves brain energy metabolism
It had been known for some time that propio- in patients with Huntington disease. Neurology
nate is an anaplerotic substrate for the heart, for 84, 490–495.
example (Martini et al., 2003). Recently, the effects Alvestad, S., Hammer, J., Eyjolfsson, E., Qu, H.,
Ottersen, O.P., and Sonnewald, U. (2008). Limbic
of triheptanoin in a rat model of cardiac hypertro-
structures show altered glial-neuronal metabolism
phy were assessed. A 30% triheptanoin diet reduced in the chronic phase of kainate induced epilepsy.
left ventricular hypertrophy and improved diastolic Neurochem Res 33, 257–266.
function and myocardial glucose oxidation (Nguyen Arnold, S., Schlaug, G., Niemann, H., Ebner, A.,
et  al., 2015), indicating that anaplerotic actions of Luders, H., Witte, O.W., and Seitz, R.J. (1996).
heptanoate or C5 ketones can benefit the ailing heart. Topography of interictal glucose hypometabolism
Studies on the effect of triheptanoin on altera- in unilateral mesiotemporal epilepsy. Neurology
tions in fatty acid composition have just begun. 46, 1422–1430.
Traditionally, it was thought that medium chain Aso, E., Semakova, J., Joda, L., Semak, V., Halbaut, L.,
fats do not get elongated and subsequently stored. Calpena, A., Escolano, C., Perales, J.C., and Ferrer,
However, in mice, feeding of even and uneven I. (2013). Triheptanoin supplementation to keto-
MCT for 1 year altered the fatty acid profile of liver genic diet curbs cognitive impairment in APP/PS1
and heart significantly, with unusual fatty acids mice used as a model of familial Alzheimer’s dis-
ease. Curr Alzheimer Res 10, 290–297.
becoming detectable, for example, C15, C17:1, and
Benson, M.J., Thomas, N.K., Talwar, S., Hodson, M.P.,
C22:1 with triheptanoin and C22:5n3 with trioc- Lynch, J.W., Woodruff, T.M., and Borges, K. (2015).
tanoin (Tucci et  al., 2015). The potential impacts A novel anticonvulsant mechanism via inhibition
of these changes remain to be explored. of complement receptor C5ar1 in murine epilepsy
models. Neurobiol Dis 76, 87–97.
CONCLUSIONS Blanco, M.M., dos Santos, J.G., Jr., Perez-Mendes,
Triheptanoin was first used as a novel alternative P., Kohek, S.R., Cavarsan, C.F., Hummel, M.,
approach to satisfy energy needs in patients with Albuquerque, C., and Mello, L.E. (2009).
343

Chapter 34: Triheptanoin in Epilepsy and Beyond 343

Assessment of seizure susceptibility in pilocarpine cerebellum and contralateral hemisphere fol-


epileptic and nonepileptic Wistar rats and of sei- lowing middle cerebral artery occlusion in rat. J
zure reinduction with pentylenetetrazole and elec- Neurochem 109 (Suppl 1), 174–181.
troshock models. Epilepsia 50, 824–831. Hadera, M.G., Smeland, O.B., McDonald, T.S., Tan,
Borges, K., Gearing, M., McDermott, D.L., Smith, A.B., K.N., Sonnewald, U., and Borges, K. (2014).
Almonte, A.G., Wainer, B.H., and Dingledine, R. Triheptanoin partially restores levels of tricar-
(2003). Neuronal and glial pathological changes boxylic acid cycle intermediates in the mouse
during epileptogenesis in the mouse pilocarpine pilocarpine model of epilepsy. J Neurochem 129,
model. Exp Neurol 182, 21–34. 107–119.
Borges, K., and Sonnewald, U. (2011). Triheptanoin- Hassel, B. (2000). Carboxylation and anaplerosis in
A medium chain triglyceride with odd chain fatty neurons and glia. MolNeurobiol 22, 21–40.
acids:  A  new anaplerotic anticonvulsant treat- Henry, T.R., Mazziotta, J.C., and Engel, J., Jr. (1993).
ment? Epilepsy Res. 100, 239–44. Interictal metabolic anatomy of mesial temporal
Brandt, C., Heile, A., Potschka, H., Stoehr, T., and lobe epilepsy. Arch Neurol 50, 582–589.
Loscher, W. (2006). Effects of the novel antiepilep- Henry, T.R., Mazziotta, J.C., Engel, J., Jr., Christenson,
tic drug lacosamide on the development of amyg- P.D., Zhang, J.X., Phelps, M.E., and Kuhl, D.E.
dala kindling in rats. Epilepsia 47, 1803–1809. (1990). Quantifying interictal metabolic activity in
Breen, C., White, F.J., Scott, C.A., Heptinstall, L., human temporal lobe epilepsy. J Cereb Blood Flow
Walter, J.H., Jones, S.A., and Morris, A.A. (2014). Metab 10, 748–757.
Unsuccessful treatment of severe pyruvate carbox- Kann, O., and Kovacs, R. (2007). Mitochondria and
ylase deficiency with triheptanoin. Eur J Pediatr neuronal activity. Am J PhysiolCell Physiol 292,
173, 361–366. C641–657.
Broer, S., Schneider, H.P., Broer, A., Rahman, Kim, T.H., Borges, K., Petrou, S., and Reid, C.A.
B., Hamprecht, B., and Deitmer, J.W. (1998). (2013). Triheptanoin reduces seizure susceptibility
Characterization of the monocarboxylate trans- in a syndrome-specific mouse model of general-
porter 1 expressed in Xenopus laevis oocytes by ized epilepsy. Epilepsy Res 103, 101–105.
changes in cytosolic pH. Biochem J 333 (Pt 1), Kinman, R.P., Kasumov, T., Jobbins, K.A., Thomas,
167–174. K.R., Adams, J.E., Brunengraber, L.N., Kutz, G.,
Brunengraber, H., and Roe, C.R. (2006). Anaplerotic Brewer, W.U., Roe, C.R., and Brunengraber, H.
molecules: current and future. J Inherit Metab Dis (2006). Parenteral and enteral metabolism of
29, 327–331. anaplerotic triheptanoin in normal rats. Am J
Corbett, A., Garg, N., Crosbie, A., and Borges, K. Physiol Endocrinol Metab 291, E860–866.
(2015). A pilot study of triheptanoin treatment Klepper, J., and Leiendecker, B. (2007). GLUT1 defi-
in sporadic inclusion body myositis (sIBM). Ann ciency syndrome--2007 update. Dev Med Child
Neurol 78, S102. Neurol 49, 707–716.
Deng, S., Zhang, G.F., Kasumov, T., Roe, C.R., and Kornberg, H.L. (1966). Anaplerotic sequences and
Brunengraber, H. (2009). Interrelations between their role in metabolism. Essays Biochem 2, 1–31.
C4 ketogenesis, C5 ketogenesis, and anaplero- Kovac, S., Abramov, A.Y., and Walker, M.C. (2013).
sis in the perfused rat liver. J Biol Chem 284, Energy depletion in seizures: anaplerosis as a strat-
27799–27807. egy for future therapies. Neuropharmacology 69,
Dube, C., Boyet, S., Marescaux, C., and Nehlig, A. (2001). 96–104.
Relationship between neuronal loss and interictal Kudin, A.P., Zsurka, G., Elger, C.E., and Kunz, W.S.
glucose metabolism during the chronic phase of the (2009). Mitochondrial involvement in temporal
lithium-pilocarpine model of epilepsy in the imma- lobe epilepsy. Exp Neurol 218, 326–332.
ture and adult rat. Exp Neurol 167, 227–241. Li, L.M., Cendes, F., Andermann, F., Dubeau, F.,
Francis, J.S., Markov, V., and Leone, P. (2014). Dietary and Arnold, D.L. (2000). Spatial extent of neu-
triheptanoin rescues oligodendrocyte loss, dysmy- ronal metabolic dysfunction measured by pro-
elination and motor function in the nur7 mouse ton MR spectroscopic imaging in patients
model of Canavan disease. J Inherit Metab Dis 37, with localization-related epilepsy. Epilepsia 41,
369–381. 666–674.
Gu, L., Zhang, G.F., Kombu, R.S., Allen, F., Kutz, G., Marin-Valencia, I., Good, L.B., Ma, Q., Malloy, C.R.,
Brewer, W.U., Roe, C.R., and Brunengraber, H. and Pascual, J.M. (2013). Heptanoate as a neural
(2010). Parenteral and enteral metabolism of fuel:  energetic and neurotransmitter precursors
anaplerotic triheptanoin in normal rats: II. Effects in normal and glucose transporter I-deficient
on lipolysis, glucose production, and liver acyl- (G1D) brain. J Cereb Blood Flow Metab 33,
CoA profile. Am J Physiol Lung Cell Mol Physiol 175–182.
298, E362–371. Martini, W.Z., Stanley, W.C., Huang, H., Rosiers,
Haberg, A.K., Qu, H., and Sonnewald, U. (2009). C.D., Hoppel, C.L., and Brunengraber, H. (2003).
Acute changes in intermediary metabolism in Quantitative assessment of anaplerosis from
344

344 section IV: Ketone-Based Metabolism

propionate in pig heart in vivo. Am J Physiol Owen, O.E., Kalhan, S.C., and Hanson, R.W. (2002).
Endocrinol Metab 284, E351–356. The key role of anaplerosis and cataplerosis for
Matagne, A., Margineanu, D.G., Kenda, B., Michel, citric acid cycle function. J Biol Chem 277,
P., and Klitgaard, H. (2008). Anti-convulsive and 30409–30412.
anti-epileptic properties of brivaracetam (ucb Pan, J.W., Kim, J.H., Cohen-Gadol, A., Pan, C.,
34714), a high-affinity ligand for the synaptic vesi- Spencer, D.D., and Hetherington, H.P. (2005).
cle protein, SV2A. Br J Pharmacol 154, 1662–1671. Regional energetic dysfunction in hippocampal
McDonald, T.S., Tan, K.N., Hodson, M.P., and Borges, epilepsy. Acta Neurol Scand 111, 218–224.
K. (2014). Alterations of hippocampal glucose Pan, J.W., Williamson, A., Cavus, I., Hetherington,
metabolism by even versus uneven medium H.P., Zaveri, H., Petroff, O.A., and Spencer, D.D.
chain triglycerides. J Cereb Blood Flow Metab 34, (2008). Neurometabolism in human epilepsy.
153–160. Epilepsia 49 (Suppl 3), 31–41.
Melo, T., Bigini, P., Sonnewald, U., Balosso, S., Park, M.J., Aja, S., Li, Q., Degano, A.L., Penati, J.,
Cagnotto, A., Barbera, S., Uboldi, S., Vezzani, A., Zhuo, J., Roe, C.R., and Ronnett, G.V. (2014).
and Mennini, T. (2010). Neuronal hyperexcit- Anaplerotic triheptanoin diet enhances mitochon-
ability and seizures are associated with changes in drial substrate use to remodel the metabolome and
glial-neuronal interactions in the hippocampus of improve lifespan, motor function, and sociability
a mouse model of epilepsy with mental retarda- in MeCP2-null mice. PloS One 9, e109527.
tion. J Neurochem 115, 1445–1454. Pascual, J.M., Liu, P., Mao, D., Kelly, D.I., Hernandez,
Melo, T.M., Nehlig, A., and Sonnewald, U. (2005). A., Sheng, M., Good, L.B., Ma, Q., Marin-Valencia,
Metabolism is normal in astrocytes in chronically I., Zhang, X., et  al. (2014). Triheptanoin for glu-
epileptic rats: a (13)C NMR study of neuronal-glial cose transporter type I deficiency (G1D): modula-
interactions in a model of temporal lobe epilepsy. J tion of human ictogenesis, cerebral metabolic rate,
Cereb Blood Flow Metab 25, 1254–1264. and cognitive indices by a food supplement. JAMA
Meredith, D., and Christian, H.C. (2008). The SLC16 Neurology 71, 1255–1265.
monocarboxylate transporter family. Xenobiotica Patel, M.S. (1974). The relative significance of CO2-
38, 1072–1106. fixing enzymes in the metabolism of rat brain. J
Mochel, F., DeLonlay, P., Touati, G., Brunengraber, H., Neurochem 22, 717–724.
Kinman, R.P., Rabier, D., Roe, C.R., and Saudubray, Reszko, A.E., Kasumov, T., Pierce, B.A., David, F.,
J.M. (2005). Pyruvate carboxylase deficiency: clin- Hoppel, C.L., Stanley, W.C., Des Rosiers, C., and
ical and biochemical response to anaplerotic diet Brunengraber, H. (2003). Assessing the reversibil-
therapy. Mol Gen Metab 84, 305–312. ity of the anaplerotic reactions of the propionyl-
Mochel, F., Duteil, S., Marelli, C., Jauffret, C., Barles, CoA pathway in heart and liver. J Biol Chem 278,
A., Holm, J., Sweetman, L., Benoist, J.F., Rabier, 34959–34965.
D., Carlier, P.G., et al. (2010). Dietary anaplerotic Roe, C.R., Bottiglieri, T., Wallace, M., Arning, E.,
therapy improves peripheral tissue energy metab- and Martin, A. (2010). Adult polyglucosan
olism in patients with Huntington’s disease. Eur J body disease (APBD):  anaplerotic diet therapy
Hum Genet 18, 1057–1060. (triheptanoin) and demonstration of defective
Nguyen, T.D., Shingu, Y., Amorim, P.A., Schwarzer, methylation pathways. Mol Gen Metabol 101,
M., and Doenst, T. (2015). Triheptanoin alleviates 246–252.
ventricular hypertrophy and improves myocardial Roe, C.R., and Brunengraber, H. (2015). Anaplerotic
glucose oxidation in rats with pressure overload. J treatment of long-chain fat oxidation disorders
Card Fail. 21, 906–15. with triheptanoin: Review of 15 years experience.
Niessen, H.G., Debska-Vielhaber, G., Sander, K., Mol Gen Metabol. 116, 260–8.
Angenstein, F., Ludolph, A.C., Hilfert, L., Willker, Roe, C.R., and Mochel, F. (2006). Anaplerotic diet
W., Leibfritz, D., Heinze, H.J., Kunz, W.S., et  al. therapy in inherited metabolic disease: therapeu-
(2007). Metabolic progression markers of neu- tic potential. J Inherit Metab Dis 29, 332–340.
rodegeneration in the transgenic G93A-SOD1 Roe, C.R., Sweetman, L., Roe, D.S., David, F., and
mouse model of amyotrophic lateral sclerosis. Eur Brunengraber, H. (2002). Treatment of cardiomy-
J Neurosci 25, 1669–1677. opathy and rhabdomyolysis in long-chain fat oxi-
Nuutinen, E.M., Peuhkurinen, K.J., Pietilainen, dation disorders using an anaplerotic odd-chain
E.P., Hiltunen, J.K., and Hassinen, I.E. (1981). triglyceride. J Clin Invest 110, 259–269.
Elimination and replenishment of tricarboxylic Roe, C.R., Yang, B.Z., Brunengraber, H., Roe, D.S.,
acid-cycle intermediates in myocardium. Biochem Wallace, M., and Garritson, B.K. (2008). Carnitine
J 194, 867–875. palmitoyltransferase II deficiency:  successful
Oldendorf, W.H. (1973). Carrier-mediated blood- anaplerotic diet therapy. Neurology 71, 260–264.
brain barrier transport of short-chain mono- Schwarzkopf, T.M., Koch, K., and Klein, J. (2015).
carboxylic organic acids. Am J Physiol 224, Reduced severity of ischemic stroke and improve-
1450–1453. ment of mitochondrial function after dietary
345

Chapter 34: Triheptanoin in Epilepsy and Beyond 345

treatment with the anaplerotic substance trihep- metabolic treatment approaches. In Bioactive
tanoin. Neuroscience 300, 201–209. Nutraceuticals and Dietary Supplements in
Silver, J.M., Shin, C., and McNamara, J.O. (1991). Neurological and Brain Disease:  Prevention
Antiepileptogenic effects of conventional anticon- and Therapy, R.R. Watson and V.R. Preedy, eds.
vulsants in the kindling model of epilepsy. Ann (Amsterdam: Elsevier), pp. 461–470.
Neurol 29, 356–363. Thomas, N.K., Willis, S., Sweetman, L., and Borges, K.
Smeland, O.B., Hadera, M.G., McDonald, T.S., (2012). Triheptanoin in acute mouse seizure mod-
Sonnewald, U., and Borges, K. (2013). Brain mito- els. Epilepsy Res 99, 312–317.
chondrial metabolic dysfunction and glutamate Tucci, S., Behringer, S., and Spiekerkoetter, U. (2015).
level reduction in the pilocarpine model of tem- De novo fatty acid biosynthesis and elongation
poral lobe epilepsy in mice. J Cereb Blood Flow in very long-chain acyl-CoA dehydrogenase-
Metab 33, 1090–1097. deficient mice supplemented with odd or even
Smith, M., Wilcox, K.S., and White, H.S. medium-chain fatty acids. FEBS J 282, 4242–4253.
(2007). Discovery of antiepileptic drugs. Vockley, J., Marsden, D., McCracken, E., DeWard, S.,
Neurotherapeutics 4, 12–17. Barone, A., Hsu, K., and Kakkis, E. (2015). Long-
Suls, A., Mullen, S.A., Weber, Y.G., Verhaert, K., term major clinical outcomes in patients with long
Ceulemans, B., Guerrini, R., Wuttke, T.V., Salvo- chain fatty acid oxidation disorders before and
Vargas, A., Deprez, L., Claes, L.R., et  al. (2009). after transition to triheptanoin treatment-A retro-
Early-onset absence epilepsy caused by mutations spective chart review. Mol Gen Metab. 116, 53–60
in the glucose transporter GLUT1. Ann Neurol 66, Waldbaum, S., and Patel, M. (2010). Mitochondria,
415–419. oxidative stress, and temporal lobe epilepsy.
Tan, H.O., Reid, C.A., Single, F.N., Davies, P.J., Chiu, Epilepsy Res 88, 23–45.
C., Murphy, S., Clarke, A.L., Dibbens, L., Krestel, White, H.S. (2003). Preclinical development of antiep-
H., Mulley, J.C., et  al. (2007). Reduced cortical ileptic drugs: past, present, and future directions.
inhibition in a mouse model of familial childhood Epilepsia 44 (Suppl 7), 2–8.
absence epilepsy. Proc Natl Acad Sci USA 104, Willis, S., Stoll, J., Sweetman, L., and Borges, K. (2010).
17536–17541. Anticonvulsant effects of a triheptanoin diet in two
Tan, K.N., McDonald, T.S., and Borges, K. (2015). mouse chronic seizure models. Neurobiol Dis 40,
Metabolic dysfunctions in epilepsy and novel 565–572.
346

35
Amino Acids in the Treatment
of Neurological Disorders
A D A M L . H A R T M A N, M D

INTRODUCTION inactive against PTZ. Also, each of the branched-


Studies of the therapeutic mechanisms of nutrient- chain amino acids (L-leucine, L-isoleucine, and
based therapies mainly focus on fats and carbo- L-valine) increase the latency to onset of seizures
hydrates (Masino and Rho, 2012). The role of induced by another GABA-A receptor antagonist,
protein, the third major diet component, has not picrotoxin (Skeie et al., 1994).
been studied as thoroughly as fats and carbohy- Using a different seizure test and dosing para-
drate, but its relevance was demonstrated by the digm, we showed that pretreatment with L-leucine
finding that protein must be restricted for systemic terminates seizures induced by the excitotoxin
ketosis to occur in the ketogenic diet (Laeger et al., kainic acid; L-valine was ineffective, suggest-
2014; Yudkoff et  al., 2007). The building blocks ing that in this test, there is not a class effect of
of proteins, amino acids, have been studied in a branched-chain amino acids (Hartman et  al.,
variety of neurological disorders. This chapter 2015). Interestingly, L-leucine was ineffective in
focuses on the therapeutic use of naturally occur- terminating kainic acid-induced seizures when it
ring proteinogenic amino acids (and some of their was injected after seizure onset.
D-enantiomers) for two model neurological disor- Glycine is somewhat unique among amino
ders, epilepsy and traumatic brain injury. Organic acids in that it can have either excitatory or
acids (e.g., taurine) and modified/synthetic amino inhibitory effects, depending on which receptor
acids are not discussed. is involved:  generally speaking, glycine binding
to NMDA receptors in the brain is excitatory but
SEIZURES AND EPILEPSY binding to glycine receptors in the spinal cord is
Seizures and epilepsy (the propensity for recurrent inhibitory (Johnson and Ascher, 1987; Werman
unprovoked seizures) represent one of the most et al., 1968), although there are exceptions to this
common neurological disorders in all age groups. general rule. Inhibition of the glycine transporter
Current treatments still leave 20-30% of patients (GlyT1) protects against acute and chronic sei-
without adequate seizure control. This has fueled zures in rodents (Kalinichev et  al., 2010b; Shen
interest in drug discovery for epilepsy, including et al., 2015). However, binding of glycine to the N-
amino acids. methyl-D-aspartate (NMDA) receptor likely does
not mediate this effect (Zellinger et al., 2014), and
L-Amino Acids in fact, inhibitors of this site have acute antiseizure
L-leucine and L-isoleucine (300 mg/kg, a high effects (Bristow et al., 1996; Nichols and Yielding,
dose) protect against seizure onset by prolong- 1998). The exact role of glycine in seizure initiation
ing the latency to onset of spike-wave discharges versus the progression or propagation of seizure
and clinical tonic-clonic seizures induced by the activity remains unclear.
GABA-A receptor antagonist pentylenetetrazol These data suggest a number of potential mech-
(PTZ); L-leucine also decreases the duration of anisms for an antiseizure effect. First, the amino
spike-wave discharges, but neither amino acid acids may be working through either cell surface
affects seizure duration (Dufour et  al., 1999). transporters or receptors, as in the case of glycine
Interestingly, α-ketoisocaproic acid, a metabo- and the GlyT1 transporter. Alternatively, they
lite of L-leucine that results from a transamina- may activate intracellular amino acid “sensors,”
tion reaction that also produces L-glutamate, is with downstream effects on signaling pathways.
347

Chapter 35: Amino Acids in the Treatment of Neurological Disorders 347

L-leucine activates the master integrator of cellu-


lar metabolism, mammalian target of rapamycin BOX 35.1
complex 1 (mTORC1) via Sestrin2 (Bar-Peled and NEUROLOGICALLY IMPORTANT
Sabatini, 2014; Cota et al., 2006; Sancak et al., 2008; L- LEUCINE TRANSPORTERS
Wolfson et  al., 2016). However, data suggest that
mTORC1 activation is not the mechanism for sei- (NUMERICAL ORDER, MODIFIED
zure control of L-leucine, because pathologically FROM GALLUS.REACTOME.ORG)
increased mTORC1 activity has been measured
in both humans and rodent models of tuberous SLC3A2 (ATA2)
sclerosis complex (Orlova and Crino, 2010; Zeng SLC6A14
et  al., 2008)  as well as some chemoconvulsant SLC6A15
rodent models of temporal lobe epilepsy; similarly,
SLC7A5 (LAT1)
pharmacological inhibition of mTORC1 decreases
seizures in some models (Buckmaster and Lew,
SLC7A6 (y+LAT2)
2011; Huang et al., 2010; Sosanya et al., 2015; Zeng SLC7A7 (y+LAT1)
et  al., 2009). Thus, it is unlikely that L-leucine, SLC7A9
an mTORC1 activator, protects against seizures
by increasing activity in this particular pathway.
Nonetheless, the role of L-leucine in mTORC1-
related disorders is unknown. This mechanism has shown mixed results in seizure and epilepsy
also does not explain the antiseizure effect of L- studies (Banach et  al., 2011). Not surprisingly,
isoleucine and L-valine. similarly mixed results have been shown with L-
Another potential mechanism for the action arginine. In an electrical kindling paradigm of
of L-leucine (and other amino acids) on seizure epilepsy, L-arginine did not affect acquisition of
activity is activation via amino acid transport- kindling or seizure severity in rats (Herberg et al.,
ers. Examples include LAT1/SLC7A5, which has 1995). In a developmental model of seizures, L-
implications for transport of other small molecules arginine prolonged PTZ-induced EEG-recorded
into the brain (Geier et  al., 2013), and B0AT2/ seizure duration in postnatal day #10 (P10) rat
SLC6A15 (Broer et  al., 2006; Hagglund et  al., pups, although somewhat paradoxically, P21 rats
2013). Interestingly, LAT1/SLC7A5 is expressed in had increased survival (with no change in seizure
pathological balloon cells in tissue from patients duration) (de Vasconcelos et al., 2000). The type of
with tuberous sclerosis (which supports the role of seizure (i.e., tonic vs. tonic-clonic) also was differ-
this transporter in cell growth, one of the cardi- ent in L-arginine-treated P21 pups compared with
nal abnormal features in this disease) (Lim et al., controls, suggesting a form of neuromodulation
2011). An antiseizure mechanism may involve that needs further clarification (and as the authors
countertransport of other amino acids, which note, this may have an impact on survival).
would in turn lead to decreased synaptic concen-
tration of excitatory amino acids and a resulting D-Amino Acids
decrease in seizure activity (Yudkoff et  al., 2007; Our studies of L-leucine led us to consider
Yudkoff et  al., 2005). A  number of other amino transporter-mediated effects or contaminants in
acid transporters also may transport leucine commercial preparations of L-leucine. The most
(Box 35.1). Further testing of these transporters abundant other amino acid “species” in this prep-
(using either pharmacological inhibitors or genet- aration was the enantiomer, D-leucine, which
ically modified organisms) is needed to determine represented up to 0.5% of the L-leucine in our com-
their importance in chronic epilepsy. mercial source (Sigma-Aldrich technical informa-
L-serine is less well known for a role in neuro- tion). Surprisingly, D-leucine terminates the
transmission than its D-enantiomer. Mice lacking behavioral manifestations of kainic acid–induced
the ASC1 transporter (alanine-serine-cysteine) seizures, even when administered after seizure
have seizures (Xie et al., 2005). Indicating stereo- onset (Hartman et  al., 2015). As mentioned pre-
specific requirements, L-serine does not potentiate viously, L-leucine was ineffective when given after
NMDA-induced seizures, although D-serine, an seizure onset in this test. D-leucine (administered
endogenous ligand of this receptor, does (Singh in drinking water for 14 days) also protects against
et al., 1990a). D-serine is discussed below. seizures in the 6-Hz electroshock test, which mod-
L-arginine gained attention because it is the els focal-onset seizures, demonstrating seizure
metabolic precursor of nitric oxide, which itself protection in a different assay. Although D-leucine
348

348 section IV: Ketone-Based Metabolism

is more ketogenic than L-leucine (i.e., its degra- (Harai et al., 2012; Singh et al., 1990a). Similarly,
dation results in the production of ketone bodies exogenously administered D-serine potentiates
but not glucose) (Embden, 1908), D-leucine treat- seizures induced by PTZ (Singh et al., 1990a). D-
ment does not induce systemic ketosis (Hartman serine does not have an effect on spike wave dis-
et al., 2015). D-leucine does not bind to the kainic charges in GAERS (Genetic Absence Epilepsy Rats
acid receptor (thus eliminating competition on the from Strasbourg) Wistar rats (Koerner et al., 1996).
receptor as a mechanism of seizure protection), D-serine potentiates seizures induced by NMDA,
nor does it bind to a panel of other CNS receptors as might be expected (Singh et al., 1990a). In stud-
or transporters. D-leucine is a known ligand of the ies designed to test the efficacy of antiseizure med-
taste receptors Tas1R2/R3 (which are expressed icines at the D-serine/glycine binding site on the
in the hippocampus, a major seizure-generating NMDA receptor, D-serine decreases the antisei-
region of the brain), but it is unclear whether this zure effect of the clinical medicine felbamate, the
represents the mechanism of D-leucine antiseizure experimental compound L-687,414, and the opioid
action (Bassoli et al., 2014; Shin et al., 2010). Other kappa-receptor agonist CI-977, among others (De
ligands of this receptor have been shown to have Sarro et al., 1994; Singh et al., 1990b; Tricklebank
limited antiseizure activity in the maximal electro- et al., 1994; White et al., 1995). Notably, the latter
shock test (Talevi et al., 2012), supporting a poten- experiments were not designed to directly test the
tial role for this receptor in epilepsy. antiseizure efficacy of D-serine but rather used it
Produced by bacteria, D-leucine is found as a coactivator of NMDA receptors.
in food products, particularly those that are D-alanine binds to the D-serine/glycine site on
plant-based (Ekborg-Ott and Armstrong, 1996; the NMDA receptor and may play a role in circa-
Mutaguchi et  al., 2013). D-leucine has been iso- dian endocrine function (Kleckner and Dingledine,
lated from rat and mouse hippocampus, mouse 1988; Morikawa et  al., 2008). Treatment with D-
neocortex, and other areas of the brain at lower alanine does not change seizure-related param-
concentrations (Hamase et al., 1997; Hamase et al., eters in fully amygdala-kindled rats (Croucher
2001). Newly-synthesized mammalian proteins do and Bradford, 1991). A  high dose (1,400 mg/kg)
not include D-leucine (Fox et al., 1998). The only of exogenously administered D-arginine has CNS
other data on therapeutic use of D-leucine were in depressant effects and increases the latency to
analgesia studies, some of which also included use onset of PTZ-induced convulsions (but 700 mg/kg
of D-phenylalanine in combination with D-leucine was ineffective) (Navarro et al., 2005).
(Cheng and Pomeranz, 1980; McKibbin and D-amino acids are considered by some to be
Cheng, 1982; Ninomiya et al., 1990). Importantly, “unnatural,” but the extant literature shows that
the doses required of each amino acid in the anal- other endogenous D-amino acids have biological
gesia studies were ~80x greater than the lowest functions. These are discussed briefly here for the
effective dose in our seizure studies, suggesting a sake of interest and completeness, although they
different mechanism for D-leucine in terminating do not have a reported direct role in epilepsy. D-
seizures. aspartate has neurotransmitter properties and may
Other D-amino acids may play a role in seizure play a role in neurodevelopment, learning, and
activity, although the literature is mixed in terms neuroendocrine function (D’Aniello et  al., 2000;
of efficacy. D-serine binds to the glycine site on the D’Aniello et al., 2011); it also decreases chemically
NMDA receptor, and decreased endogenous con- induced schizophrenia-like symptoms in rodents
centrations have been shown to be partly responsi- (Errico et al., 2011; Errico et al., 2008). The brain
ble for cognitive dysfunction in rats with epilepsy concentration of D-proline is highest in pineal
induced by pilocarpine status epilepticus (Klatte and pituitary tissue, but its endogenous func-
et  al., 2013; Schell et  al., 1995). Results of stud- tion has not been reported (Hamase et al., 2005).
ies on the anticonvulsant effects of D-serine have D-glutamate has been detected in subsets of
been mixed. D-serine protects weakly in the max- neuronal cell bodies in the mesencephalon and
imal electroshock test, where it also potentiates thalamus by immunohistochemical techniques,
the effects of some antiseizure drugs (Kalinichev but its role is unknown (Mangas et  al., 2007).
et  al., 2010a; Peterson, 1991). D-serine increases D-methionine (1 mM) protects cultured auditory
afterdischarge thresholds in amygdala-kindled rats neurons against cisplatin-induced neurotoxicity
(Loscher et al., 1994). Conversely, serine racemace (Gopal et al., 2012). A role for these D-amino acids
knockout mice, which have decreased extracellu- in seizures has not been reported. Interestingly,
lar levels of D-serine in the dentate gyrus, are rel- plasma concentrations of D-serine, D-aspartate,
atively protected against seizures induced by PTZ D-alanine, D-leucine, and D-proline are reduced
349

Chapter 35: Amino Acids in the Treatment of Neurological Disorders 349

CONCLUSIONS
BOX 35.2 Select amino acids have been shown to protect
D- AMINO ACIDS against seizure activity and the sequelae of TBI.
WITH ANTISEIZURE EFFECTS Interestingly, these compounds have a variety of
different mechanisms in these disorders, including
receptor binding and transporter effects, and as
D-leucine metabolic intermediates for other signaling mol-
D-serine ecules. They also may affect intracellular signaling
D-arginine pathways, although evidence for the latter is scant
at this point. Further research will elucidate addi-
tional roles for these amino acids in the treatment
of neurological disorders.
in the rat beta-amyloid hippocampus injection
model of Alzheimer’s disease (Xing et  al., 2016). REFERENCES
Further work should identify whether D-amino Aquilani, R., Boselli, M., Boschi, F., Viglio, S., Iadarola,
acid detection will be useful as a biomarker for P., Dossena, M., Pastoris, O., and Verri, M. (2008).
either identifying the disease or monitoring dis- Branched-chain amino acids may improve recov-
ease progression (Box 35.2). ery from a vegetative or minimally conscious state
in patients with traumatic brain injury:  a pilot
T R AU M AT I C B R A I N study. Arch Phys Med Rehab 89, 1642–1647.
I N J U RY   ( T B I ) Aquilani, R., Iadarola, P., Contardi, A., Boselli, M.,
Traumatic brain injury represents one of the major Verri, M., Pastoris, O., Boschi, F., Arcidiaco, P.,
causes of injury and disability in all age groups and Viglio, S. (2005). Branched-chain amino acids
enhance the cognitive recovery of patients with
(although the causes vary between age groups).
severe traumatic brain injury. Arch Phys Med
Branched chain amino acids (particularly leucine)
Rehab 86, 1729–1735.
represent a major source of glutamate in the cen- Banach, M., Piskorska, B., Czuczwar, S.J., and
tral nervous system (Sakai et al., 2004). Glutamate Borowicz, K.K. (2011). Nitric oxide, epileptic
serves as an excitatory neurotransmitter, an exci- seizures, and action of antiepileptic drugs. CNS
totoxin, and an energy store (used indirectly for Neurol Disord-Dr 10, 808–819.
generation of ATP). In the mouse lateral fluid Bar-Peled, L., and Sabatini, D.M. (2014). Regulation
percussion model of traumatic brain injury, hip- of mTORC1 by amino acids. Trends Cell Biol 24,
pocampal concentrations of each of the branched 400–406.
chain amino acids (total levels, i.e., L- and D- Bassoli, A., Borgonovo, G., Caremoli, F., and Mancuso,
amino acids) are decreased; supplementation of G. (2014). The taste of D- and L-amino acids: IN
these amino acids for 5 or 10 days (but not shorter vitro binding assays with cloned human bitter
durations) reverses injury-induced deficits in (TAS2Rs) and sweet (TAS1R2/TAS1R3) receptors.
Food Chem 150, 27–33.
both anterograde and retrograde memory (Cole
Bristow, L.J., Hutson, P.H., Kulagowski, J.J., Leeson,
et  al., 2010; Elkind et  al., 2015). In human tri-
P.D., Matheson, S., Murray, F., Rathbone, D.,
als, supplementation of all three branched chain Saywell, K.L., Thorn, L., Watt, A.P., et  al. (1996).
amino acids led to a substantial improvement Anticonvulsant and behavioral profile of L-
in Disability Rating Scale scores (which evalu- 701,324, a potent, orally active antagonist at the
ate factors such as self-care and employability) glycine modulatory site on the N-methyl-D-
in patients with severe TBI or in a posttraumatic aspartate receptor complex. J Pharmacol Exp Ther
vegetative state (in both studies, mean Glasgow 279, 492–501.
Coma scores were in the 5.5–5.9 range, represent- Broer, A., Tietze, N., Kowalczuk, S., Chubb, S.,
ing significant impairment) (Aquilani et al., 2008; Munzinger, M., Bak, L.K., and Broer, S. (2006).
Aquilani et al., 2005). The orphan transporter v7-3 (slc6a15) is a Na+-
L-serine is neuroprotective in a mouse weight- dependent neutral amino acid transporter (B(0)
drop model in the acute phase of TBI (measured AT2). Biochem J 393, 421–430.
Buckmaster, P.S., and Lew, F.H. (2011). Rapamycin
morphologically), and the mechanism may involve
suppresses mossy fiber sprouting but not seizure
activation of glycine receptors (Zhai et al., 2015).
frequency in a mouse model of temporal lobe epi-
L-serine also decreases astrocytosis, microglial lepsy. J Neurosci 31, 2337–2347.
activation, and concentrations of inflammatory Cheng, R.S., and Pomeranz, B. (1980). A combined
cytokines in the later phases of this model of TBI treatment with D-amino acids and electroacu-
(Zhai et al., 2015). puncture produces a greater analgesia than either
350

350 section IV: Ketone-Based Metabolism

treatment alone; naloxone reverses these effects. Errico, F., Rossi, S., Napolitano, F., Catuogno, V.,
Pain 8, 231–236. Topo, E., Fisone, G., D’Aniello, A., Centonze, D.,
Cole, J.T., Mitala, C.M., Kundu, S., Verma, A., Elkind, and Usiello, A. (2008). D-aspartate prevents cor-
J.A., Nissim, I., and Cohen, A.S. (2010). Dietary ticostriatal long-term depression and attenuates
branched chain amino acids ameliorate injury- schizophrenia-like symptoms induced by amphet-
induced cognitive impairment. Proc Natl Acad Sci amine and MK-801. J Neurosci 28, 10404–10414.
USA 107, 366–371. Fox, H.L., Pham, P.T., Kimball, S.R., Jefferson, L.S., and
Cota, D., Proulx, K., Smith, K.A., Kozma, S.C., Lynch, C.J. (1998). Amino acid effects on transla-
Thomas, G., Woods, S.C., and Seeley, R.J. (2006). tional repressor 4E-BP1 are mediated primarily by
Hypothalamic mTOR signaling regulates food L-leucine in isolated adipocytes. Am J Physiol-Cell
intake. Science 312, 927–930. Ph 275, C1232–C1238.
Croucher, M.J., and Bradford, H.F. (1991). The influ- Geier, E.G., Schlessinger, A., Fan, H., Gable, J.E.,
ence of strychnine-insensitive glycine receptor Irwin, J.J., Sali, A., and Giacomini, K.M. (2013).
agonists and antagonists on generalized seizure Structure-based ligand discovery for the Large-
thresholds. Brain Res 543, 91–96. neutral Amino Acid Transporter 1, LAT-1. Proc
D’Aniello, A., Di Fiore, M.M., Fisher, G.H., Milone, A., Natl Acad Sci USA 110, 5480–5485.
Seleni, A., D’Aniello, S., Perna, A.F., and Ingrosso, Gopal, K.V., Wu, C., Shrestha, B., Campbell, K.C.,
D. (2000). Occurrence of D-aspartic acid and N- Moore, E.J., and Gross, G.W. (2012). d-Methionine
methyl-D-aspartic acid in rat neuroendocrine tis- protects against cisplatin-induced neurotoxic-
sues and their role in the modulation of luteinizing ity in cortical networks. Neurotoxicol Teratol 34,
hormone and growth hormone release. FASEB J 495–504.
14, 699–714. Hagglund, M.G.A., Roshanbin, S., Lofqvist, E.,
D’Aniello, S., Somorjai, I., Garcia-Fernandez, J., Topo, Hellsten, S.V., Nilsson, V.C.O., Todkar, A., Zhu,
E., and D’Aniello, A. (2011). D-Aspartic acid is a Y.N., Stephansson, O., Drgonova, J., Uhl, G.R.,
novel endogenous neurotransmitter. FASEB J 25, et  al. (2013). B(0)AT2 (SLC6A15) is local-
1014–1027. ized to neurons and astrocytes, and is involved
De Sarro, G., Ongini, E., Bertorelli, R., Aguglia, U., in mediating the effect of leucine in the brain.
and De Sarro, A. (1994). Excitatory amino acid PLoS One 8.
neurotransmission through both NMDA and Hamase, K., Homma, H., Takigawa, Y., Fukushima, T.,
non-NMDA receptors is involved in the anticon- Santa, T., and Imai, K. (1997). Regional distribu-
vulsant activity of felbamate in DBA/2 mice. Eur J tion and postnatal changes of D-amino acids in rat
Pharmacol 262, 11–19. brain. Biochim Biophys Acta 1334, 214–222.
de Vasconcelos, A.P., Gizard, F., Marescaux, C., Hamase, K., Inoue, T., Morikawa, A., Konno, R., and
and Nehlig, A. (2000). Role of nitric oxide Zaitsu, K. (2001). Determination of free D-proline
in pentylenetetrazol-induced seizures:  age- and D-leucine in the brains of mutant mice lack-
dependent effects in the immature rat. Epilepsia ing D-amino acid oxidase activity. Anal Biochem
41, 363–371. 298, 253–258.
Dufour, F., Nalecz, K.A., Nalecz, M.J., and Nehlig, A. Hamase, K., Konno, R., Morikawa, A., and Zaitsu, K.
(1999). Modulation of pentylenetetrazol-induced (2005). Sensitive determination of D-amino acids
seizure activity by branched-chain amino acids and in mammals and the effect of D-amino-acid oxi-
alpha-ketoisocaproate. Brain Res 815, 400–404. dase activity on their amounts. Biol Pharm Bull 28,
Ekborg-Ott, K.H., and Armstrong, D.W. (1996). 1578–1584.
Evaluation of the concentration and enantiomeric Harai, T., Inoue, R., Fujita, Y., Tanaka, A., Horio, M.,
purity of selected free amino acids in fermented Hashimoto, K., Hongou, K., Miyawaki, T., and
malt beverages (beers). Chirality 8, 49–57. Mori, H. (2012). Decreased susceptibility to sei-
Elkind, J.A., Lim, M.M., Johnson, B.N., Palmer, C.P., zures induced by pentylenetetrazole in serine race-
Putnam, B.J., Kirschen, M.P., and Cohen, A.S. mase knockout mice. Epilepsy Res 102, 180–187.
(2015). Efficacy, dosage, and duration of action of Hartman, A.L., Santos, P., O’Riordan, K.J., Stafstrom,
branched chain amino acid therapy for traumatic C.E., and Marie Hardwick, J. (2015). Potent
brain injury. Front Neurol 6, 73. anti-seizure effects of D-leucine. Neurobiol Dis
Embden, G. (1908). Uber das Verhalten der optish- 82, 46–53.
isomeren Leucine in der Leber. Beitr z Chem Herberg, L.J., Grottick, A., and Rose, I.C. (1995).
Physiol u Path 11, 348–355. Nitric-oxide synthesis, epileptic seizures and kin-
Errico, F., Nistico, R., Napolitano, F., Mazzola, C., dling. Psychopharmacology 119, 115–123.
Astone, D., Pisapia, T., Giustizieri, M., D’Aniello, Huang, X., Zhang, H., Yang, J., Wu, J., McMahon, J.,
A., Mercuri, N.B., and Usiello, A. (2011). Increased Lin, Y., Cao, Z., Gruenthal, M., and Huang, Y.
D-aspartate brain content rescues hippocampal (2010). Pharmacological inhibition of the mam-
age-related synaptic plasticity deterioration of malian target of rapamycin pathway suppresses
mice. Neurobiol Aging 32, 2229–2243. acquired epilepsy. Neurobiol Dis 40, 193–199.
351

Chapter 35: Amino Acids in the Treatment of Neurological Disorders 351

Johnson, J.W., and Ascher, P. (1987). Glycine potenti- of D-alanine and related compounds in rats and
ates the NMDA response in cultured mouse-brain the effect of restricted feeding on their amounts.
neurons. Nature 325, 529–531. J Chromatogr B Analyt Technol Biomed Life Sci
Kalinichev, M., Starr, K.R., Teague, S., Bradford, A.M., 875, 168–173.
Porter, R.A., and Herdon, H.J. (2010a). Glycine Mutaguchi, Y., Ohmori, T., Wakamatsu, T., Doi, K., and
transporter 1 (GlyT1) inhibitors exhibit anticon- Ohshima, T. (2013). Identification, purification,
vulsant properties in the rat maximal electroshock and characterization of a novel amino acid race-
threshold (MEST) test. Brain Res 1331, 105–113. mase, isoleucine 2-epimerase, from Lactobacillus
Kalinichev, M., Starr, K.R., Teague, S., Bradford, A.M., species. J Bacteriol 195, 5207–5215.
Porter, R.A., and Herdon, H.J. (2010b). Glycine Navarro, E., Alonso, S.J., Martin, F.A., and Castellano,
transporter 1 (GlyT1) inhibitors exhibit anticon- M.A. (2005). Toxicological and pharmacologi-
vulsant properties in the rat maximal electroshock cal effects of D-arginine. Basic Clin Pharmacol
threshold (MEST) test. Brain Research 1331, Toxicol 97, 149–154.
105–113. Nichols, A.C., and Yielding, K.L. (1998). Anticonvulsant
Klatte, K., Kirschstein, T., Otte, D., Pothmann, L., activity of 4-urea-5,7-dichlorokynurenic acid
Muller, L., Tokay, T., Kober, M., Uebachs, M., derivatives that are antagonists at the NMDA-
Zimmer, A., and Beck, H. (2013). Impaired D- associated glycine binding site. Mol Chem
serine-mediated cotransmission mediates cog- Neuropathol 35, 1–12.
nitive dysfunction in epilepsy. J Neurosci 33, Ninomiya, Y., Kawamura, H., Nomura, T., Uebayashi,
13066–13080. H., Sabashi, K., and Funakoshi, M. (1990).
Kleckner, N.W., and Dingledine, R. (1988). Analgesic effects of D-amino acids in four inbred
Requirement for glycine in activation of NMDA- strains of mice. Comp Biochem Physiol C 97,
receptors expressed in Xenopus oocytes. Science 341–343.
241, 835–837. Orlova, K.A., and Crino, P.B. (2010). The tuberous
Koerner, C., Danober, L., Boehrer, A., Marescaux, C., sclerosis complex. Ann NY Acad Sci 1184, 87–105.
and Vergnes, M. (1996). Thalamic NMDA trans- Peterson, S.L. (1991). Anticonvulsant drug potentia-
mission in a genetic model of absence epilepsy in tion by glycine in maximal electroshock seizures
rats. Epilepsy Res 25, 11–19. is mimicked by D-serine and antagonized by
Laeger, T., Henagan, T.M., Albarado, D.C., Redman, 7-chlorokynurenic acid. Eur J Pharmacol 199,
L.M., Bray, G.A., Noland, R.C., Munzberg, H., 341–348.
Hutson, S.M., Gettys, T.W., Schwartz, M.W., et al. Sakai, R., Cohen, D.M., Henry, J.F., Burrin, D.G., and
(2014). FGF21 is an endocrine signal of protein Reeds, P.J. (2004). Leucine-nitrogen metabolism
restriction. J Clin Invest 124, 3913–3922. in the brain of conscious rats:  its role as a nitro-
Lim, B.C., Cho, K.Y., Lim, J.S., Lee, R.S., Kim, H.S., gen carrier in glutamate synthesis in glial and
Kim, M.K., Kim, J.H., Woo, Y.J., Kim, J.K., Kim, neuronal metabolic compartments. Journal of
D.K., et al. (2011). Increased expression of l-amino Neurochemistry 88, 612–622.
acid transporters in balloon cells of tuberous scle- Sancak, Y., Peterson, T.R., Shaul, Y.D., Lindquist, R.A.,
rosis. Child Nerv Syst 27, 63–70. Thoreen, C.C., Bar-Peled, L., and Sabatini, D.M.
Loscher, W., Wlaz, P., Rundfeldt, C., Baran, H., and (2008). The Rag GTPases bind raptor and medi-
Honack, D. (1994). Anticonvulsant effects of the ate amino acid signaling to mTORC1. Science 320,
glycine/NMDA receptor ligands D-cycloserine 1496–1501.
and D-serine but not R-(+)-HA-966 in amygdala- Schell, M.J., Molliver, M.E., and Snyder, S.H. (1995).
kindled rats. Br J Pharmacol 112, 97–106. D-serine, an endogenous synaptic modula-
Mangas, A., Covenas, R., Bodet, D., Geffard, M., Aguilar, tor:  localization to astrocytes and glutamate-
L.A., and Yajeya, J. (2007). Immunocytochemical stimulated release. Proc Natl Acad Sci USA 92,
visualization of D-glutamate in the rat brain. 3948–3952.
Neuroscience 144, 654–664. Shen, H.Y., van Vliet, E.A., Bright, K.A., Hanthorn,
Masino, S.A., and Rho, J.M. (2012). Mechanisms of M., Lytle, N.K., Gorter, J., Aronica, E., and Boison,
ketogenic diet action. In Jasper’s Basic Mechanisms D. (2015). Glycine transporter 1 is a target for the
of the Epilepsies, J.L. Noebels, M. Avoli, M.A. treatment of epilepsy. Neuropharmacology 99,
Rogawski, O.R. W., and A.V. Delgado-Escueta, 554–565.
eds. (Bethesda: National Center for Biotechnology Shin, Y.J., Park, J.H., Choi, J.S., Chun, M.H., Moon,
Information), pp. 1001–1022. Y.W., and Lee, M.Y. (2010). Enhanced expression
McKibbin, L.S., and Cheng, R.S. (1982). Systemic d- of the sweet taste receptors and alpha-gustducin
phenylalanine and d-leucine for effective treat- in reactive astrocytes of the rat hippocampus
ment of pain in the horse. Canadian VetJ/La Revue following ischemic injury. Neurochem Res 35,
Veterinaire Canadienne 23, 39–40. 1628–1634.
Morikawa, A., Hamase, K., Miyoshi, Y., Koyanagi, S., Singh, L., Oles, R.J., and Tricklebank, M.D. (1990a).
Ohdo, S., and Zaitsu, K. (2008). Circadian changes Modulation of seizure susceptibility in the mouse
352

352 section IV: Ketone-Based Metabolism

by the strychnine-insensitive glycine recognition Xie, X.M., Dumas, T., Tang, L., Brennan, T., Reeder,
site of the NMDA receptor/ion channel complex. T., Thomas, W., Klein, R.D., Flores, J., O’Hara, B.F.,
Br J Pharmacol 99, 285–288. Heller, H.C., et  al. (2005). Lack of the alanine-
Singh, L., Vass, C.A., Hunter, J.C., Woodruff, G.N., serine-cysteine transporter 1 causes tremors, sei-
and Hughes, J. (1990b). The anticonvulsant zures, and early postnatal death in mice. Brain Res
action of CI-977, a selective kappa-opioid recep- 1052, 212–221.
tor agonist:  a possible involvement of the gly- Xing, Y., Li, X., Guo, X., and Cui, Y. (2016).
cine/NMDA receptor complex. Eur J Pharmacol Simultaneous determination of 18 D-amino
191, 477–480. acids in rat plasma by an ultrahigh-performance
Skeie, B., Petersen, A.J., Manner, T., Askanazi, J., and liquid chromatography-tandem mass spectrom-
Steen, P.A. (1994). Effects of valine, leucine, isoleu- etry method:  application to explore the potential
cine, and a balanced amino acid solution on the relationship between Alzheimer’s disease and D-
seizure threshold to picrotoxin in rats. Pharmacol amino acid level alterations. Anal Bioanal Chem
Biochem Behav 48, 101–103. 408, 141–150.
Sosanya, N.M., Brager, D.H., Wolfe, S., Niere, F., and Yudkoff, M., Daikhin, Y., Melo, T.M., Nissim, I., and
Raab-Graham, K.F. (2015). Rapamycin reveals an Sonnewald, U. (2007). The ketogenic diet and
mTOR-independent repression of Kv1.1 expres- brain metabolism of amino acids:  relationship
sion during epileptogenesis. Neurobiol Dis 73, to the anticonvulsant effect. Annu Rev Nutr 27,
96–105. 415–430.
Talevi, A., Enrique, A.V., and Bruno-Blanch, L.E. Yudkoff, M., Daikhin, Y., Nissim, I., Horyn, O.,
(2012). Anticonvulsant activity of artificial sweet- Lazarow, A., Luhovyy, B., and Wehrli, S. (2005).
eners: a structural link between sweet-taste recep- Response of brain amino acid metabolism to keto-
tor T1R3 and brain glutamate receptors. Bioorg sis. Neurochem Int 47, 119–128.
Med Chem Lett 22, 4072–4074. Zellinger, C., Salvarnoser, J.D., Soerensen, J., van
Tricklebank, M.D., Bristow, L.J., Hutson, P.H., Leeson, Vliet, E.A., Aronica, E., Gorter, J., and Potschka,
P.D., Rowley, M., Saywell, K., Singh, L., Tattersall, H. (2014). Pre-treatment with the NMDA recep-
F.D., Thorn, L., and Williams, B.J. (1994). The anti- tor glycine-binding site antagonist L-701,324
convulsant and behavioural profile of L-687,414, improves pharmacosensitivity in a mouse kindling
a partial agonist acting at the glycine modulatory model. Epilepsy Res 108, 634–643.
site on the N-methyl-D-aspartate (NMDA) recep- Zeng, L.H., Xu, L., Gutmann, D.H., and Wong, M.
tor complex. Br J Pharmacol 113, 729–736. (2008). Rapamycin prevents epilepsy in a mouse
Werman, R., Davidoff, R.A., and Aprison, M.H. model of tuberous sclerosis complex. Ann Neurol
(1968). Inhibitory action of glycine on spinal neu- 63, 444–453.
rons in cat. J Neurophysiol 31, 81–95. Zeng, L.H., Rensing, N.R., and Wong, M. (2009). The
White, H.S., Harmsworth, W.L., Sofia, R.D., and mammalian target of rapamycin signaling path-
Wolf, H.H. (1995). Felbamate modulates the way mediates epileptogenesis in a model of tem-
strychnine-insensitive glycine receptor. Epilepsy poral lobe epilepsy. J Neurosci 29, 6964–6972.
Res 20, 41–48. Zhai, P.P., Xu, L.H., Yang, J.J., Jiang, Z.L., Zhao, G.W.,
Wolfson, R.L., Chantranupong, L., Saxton, R.A., Sun, L., Wang, G.H., and Li, X. (2015). Reduction
Shen, K., Scaria, S.M., Cantor, J.R., and Sabatini, of inflammatory responses by L-serine treatment
D.M. (2016). Sestrin2 is a leucine sensor for the leads to neuroprotection in mice after traumatic
mTORC1 pathway. Science 351, 43–48. brain injury. Neuropharmacology 95, 1–11.
353

36
2-Deoxyglucose
Metabolic Control of Seizures through Inhibition of Glycolysis

C A R L E . S TA F S T R O M , M D , P H D A N D T H O M A S P. S U T U L A , M D , P H D

INTRODUCTION glycolysis (Figure 36.1). This effect and the obser-


Conventional anticonvulsant medications reduce vation that minimal carbohydrate intake can abol-
neuronal excitability through effects on ion chan- ish seizure control achieved by the KD suggests
nels or synaptic function. In recent years, it has that inhibitors of glycolysis may mimic some of the
become clear that metabolic factors also play a role favorable therapeutic effects of the diet. Among
in the modulation of neuronal excitability (Reid compounds that inhibit glycolysis, the glucose
et al., 2014). This volume contains many examples analog 2-deoxy-D-glucose (2DG) is a promising
of potentially beneficial metabolic treatments for novel agent for seizure protection. 2DG differs
epilepsy and other neurological disorders. In par- from glucose by removal of a single oxygen atom
ticular, the high-fat, low-carbohydrate ketogenic from the 2-position (Figure 36.2A). 2DG is taken
diet (KD) is effective in controlling seizures in many up by cells and undergoes phosphorylation at the
children whose seizures are refractory to anticon- 6-position to 2DG-6P, but glycolytic flux is reduced
vulsant medications (Neal et al., 2008). However, because 2DG-6P cannot undergo isomerization by
the mechanisms of action of the KD and its vari- phosphoglucose isomerase (Figure 36.2B). Uptake
ants (e.g., medium chain triglyceride diet, modi- of glucose and 2DG is enhanced in energetically
fied Atkins diet, low glycemic index treatment) active cells. 2DG has been used for decades as a
are very complex and are not fully characterized fluorinated positron emitted tracer (F18-2DG) for
(Rho and Stafstrom, 2012; Lutas and Yellen, 2013; measurement and imaging of regional glucose uti-
Gano et  al., 2014). Possible mechanisms include lization by positron emission tomography (PET)
reduction of excitability by ketone bodies or fatty (Wree, 1990). 2DG has also been investigated as
acids, altered neurotransmitter synthesis or action, an adjuvant chemotherapeutic agent for several
improved mitochondrial function, or a combina- types of cancer, since rapidly dividing, metaboli-
tion of these or other factors. The key observation cally active neoplastic cells with enhanced glucose
that ingestion of small amounts of carbohydrate by uptake are vulnerable to glycolytic inhibition by
children on the KD results in loss of seizure con- 2DG (Pelicano et al., 2006; Cheong et al., 2011).
trol (Huttenlocher, 1976) led to the idea that car-
bohydrate restriction could exert a protective effect ANTICONVULSANT
against seizures (Greene et al., 2003). In addition ACTIONS OF 2DG
to limiting carbohydrate intake, restricting calorie We tested the effects of 2DG in several models
intake also suppresses seizures and affords neuro- of acute seizures in vivo and in vitro (Stafstrom
protection (Greene et al., 2003; Ingram and Roth, et  al., 2009). When exposed to elevated extracel-
2011; Pani, 2015; Yuen and Sander, 2014), and in lular potassium (Ko+, 7.5 mM), hippocampal CA3
fact, the KD was initially formulated to mimic the neurons in slices from adult rats developed high-
physiological effects of fasting. Although some frequency interictal (epileptiform) bursts at a fre-
data supports intermittent fasting for seizure con- quency of about 30 per minute. Addition of 2DG
trol (Hartman et al., 2013), fasting is not a feasible (10 mM) to the bathing medium reversibly reduced
long-term treatment option. the burst frequency by about 50% (Stafstrom
As an alternative to fasting, the KD restricts et al., 2009). Similarly, 2DG reduced epileptiform
dietary carbohydrates and generates ketone bodies bursts induced by bath application of the chemi-
as the proximate energy source, thereby reducing cal convulsants bicuculline, a γ-aminobutyric acid
354

354 section IV: Ketone-Based Metabolism

KD

Ketone bodies Anaplerosis


(β-OHB, AcAc)
2DG

Glycolysis
PDH
Glucose Pyruvate ACoA TCA

Gluconeogenesis

FDP

PPP Lactate

FIGURE  36.1 Glucose metabolism and points at which interventions could affect neuronal excitability and
seizure control. Glucose can be diverted to the pentose phosphate shunt (PPP) via fructose-1,6-diphosphate (FDP).
2-Deoxyglucose (2DG) inhibits glycolysis by blocking the phosphoglucose isomerase step. The ketogenic diet (KD), via
ketone bodies, bypasses glycolysis by providing acetyl-CoA (ACoA) to the TCA (tricarboxylic acid cycle) after glycolysis.
Anaplerosis refers to the “refilling” of intermediate compounds depleted from the TCA cycle. Other abbreviations: β-
OHB, beta-hydroxybutyrate; AcAc, acetoacetate; PDH, pyruvate dehydrogenase. Reprinted with permission from Rho
and Stafstrom, 2012.

(GABA) receptor antagonist, or 4-aminopyridine, seconds; 2DG decreased the occurrence of these
a potassium channel blocker. In slices from juve- ictal bursts. Therefore, 2DG has an anticonvul-
nile animals (P10-13), 7.5 mM Ko+ induced pro- sant effect on both interictal and ictal epileptiform
longed ictal discharges in area CA3 for 10–30 activity in the CA3 region of hippocampus.

(a) (b)
CH2OH CH2OPO32– Glucose
3–O CH2OH
H OH H OH 3POH2C
H H O
OH H OH H HHO Glucose-6-P
HO OH HO OH H OH
GPI 2DG
H OH H OH HO H
Glucose Glucose-6P Fructose-6P Fructose-6-P

CH2OH CH2OPO32– PFK Pyruvate


3–O CH2OH
H OH H OH 3PCH2C
H H O Fructose-1,6-P
OH H OH H HHO NAD
HO OH HO OH H OH
H H H H HO H NADH

2DG 2DG-6P TCA cycle

FIGURE 36.2 Glucose, 2DG, and the glycolytic pathway. (a) Chemical structures of glucose, 2-deoxyglucose (2DG),
and intermediates of the initial steps of glycolysis. Phosphorylation of 2DG yields 2DG-6P, which cannot undergo
isomerization by glucose-6-phosphate (glucose-6-P) isomerase (GPI) to fructose-6-phosphate (fructose-6-P), thereby
preventing subsequent steps of glycolysis. (b) Schematic diagram of key steps of glycolysis illustrating the rate-limiting
step involving phosphofructokinase, which is inhibited by pyruvate, the end product of the pathway. Oxidation of
phosphoenol-pyruvate (structure not shown) to pyruvate generates nicotinamide adenine dinucleotide (NADH) before
entry into the tricarboxylic acid (TCA) cycle. Reprinted with permission from Stafstrom et al., 2009.
355

Chapter 36: 2-Deoxyglucose 355

An anticonvulsant effect was also seen in in It is worth mentioning that the KD and 2DG
vivo seizure models. Kindling refers to the pro- both suppress seizure activity, but our studies with
gressive decrease in seizure threshold in response 2DG were not intended to investigate KD mecha-
to repeated subconvulsive electrical stimuli. nisms. For instance, 2DG does not cause ketosis.
Kindled seizures can be elicited by stimulation of Both 2DG and the KD suppress seizures in Fring’s
many brain pathways. We assessed the effect of mice and in the 6-Hz model (Hartman et  al.,
2DG on kindling development by stimulation of 2008), but the KD is effective in the MES model
the perforant path or the olfactory bulb (Garriga- while 2DG is not. The former two models are used
Canut et al., 2006; Stafstrom et al., 2009). 2DG was to model focal onset seizures, whereas the MES is
administered at a dose of 250 mg/kg intraperitone- considered to mimic generalized tonic-clonic sei-
ally (i.p.) 30 minutes prior to daily kindling stimu- zures. Kindling is abrogated by both the KD and
lation of either pathway. In response to kindling 2DG, though exact kindling protocols differ widely
of the perforant path, but not the olfactory bulb, among studies (Hori et  al., 1997; Garriga-Canut
2DG-pretreated rats displayed an increase in mean et al., 2006; Stafstrom et al., 2009). Therefore, 2DG
afterdischarge (AD) threshold over time, defining and the KD are not comparable directly, though
a region-specific anticonvulsant effect. both modify seizure susceptibility by altering met-
In addition, an anticonvulsant effect of 2DG abolic pathways involved in energy regulation.
was seen in two other acute seizure models in ani- Therefore, potentially useful clinical information
mals. First, in the 6 Hz stimulation model (Barton can be gained from studies of both compounds.
et al., 2001), psychomotor seizures were induced by
corneal stimulation using electrical pulses at a fre-
quency of 6 Hz. Pretreatment with 2DG resulted in ANTIEPILEPTIC
seizure protection in 75% of rats tested (Stafstrom ACTIONS OF 2DG
et  al., 2009). Second, in Fring’s mice with audio- An antiepileptic (or antiepileptogenic) effect is
genic seizures, 50% of mice were protected from defined as the slowing or prevention of epilepsy
sound-induced seizures after pretreatment with development. Antiepileptogenesis is “disease-
2DG (Stafstrom et  al., 2009). However, 2DG did modifying,” meaning that the process of epilepsy
not protect against pentylenetetrazole- or maximal development is hindered. Using the kindling
electroshock-induced seizures in rats. Therefore, model, with stimulation of either the olfactory bulb
anticonvulsant effects of 2DG were demonstrated or perforant path, we investigated whether 2DG
in several but not all in vivo seizure models, with mediates an antiepileptogenic effect. Kindling
a pattern of effectiveness unlike that afforded by progression was quantified by the number of ADs
any currently available antiseizure medicine. For required to elicit specific stages of seizure sever-
example, no conventional antiseizure medication ity, using the Racine scoring scale (Racine, 1972).
exhibits region-specific activity, as indicated by Pretreatment with 2DG at doses ranging from 37.5
2DG’s differential action on perforant path versus mg/kg intraperitoneally (i.p.) (we found this to be
olfactory bulb. Thus, 2DG’s mechanisms are likely the minimally effective dose) up to 250 mg/kg i.p.,
to be broad, affecting seizure-induced plasticity given 30 minutes before each kindling stimulation,
across many seizure types and syndromes, but an resulted in an ~2-fold increase in the number of
exact correlation with human epilepsies is not yet ADs required to achieve class III, IV, or V kindled
possible (Holmes and Zhao, 2008). seizures in both regions (Garriga-Canut et  al.,
Not all studies have confirmed that 2DG exerts 2006; Sutula and Franzoso, 2008; Stafstrom et al.,
an anticonvulsant effect. In the presence of 2DG, 2009). These results indicate a disease-modifying
latency to seizure onset was slightly decreased in antiepileptic action. Note that the olfactory bulb
mice when chemoconvulsants (pentylenetetrazole, kindling experiments described in the preced-
kainic acid) were coadministered intravenously ing paragraph, testing the anticonvulsant effects
with 2DG (Gasior et al., 2010). While this outcome of 2DG, showed no significant effect of this com-
has been interpreted as a possible proconvulsant pound on kindled seizure threshold. Therefore,
effect, an alternative explanation is that the well- 2DG has different anticonvulsant effects in differ-
documented acute increase in cerebral blood flow ent brain regions, whereas its antiepileptic effects
induced by 2DG enhances brain delivery of par- were similar in hippocampus and olfactory bulb.
enteral convulsants such as pentylenetetrazole and Furthermore, 2DG retarded kindled seizure pro-
kainic acid, thereby shortening the latency to ini- gression when administered immediately after
tial seizure manifestations as a threshold outcome or 10 minutes after a kindled seizure, raising the
measure. possibility that 2DG can be used as a “postseizure”
356

356 section IV: Ketone-Based Metabolism

treatment (e.g., for clusters of seizures or even continued to afford protection against kindling
status epilepticus) (Sutula and Franzoso, 2008; progression in these transgenic animals.
Stafstrom et al., in preparation). Others have reported evidence that the effect
2DG exerts neuroprotective actions in several of 2DG is mediated through up-regulation of KATP
other models as well. In hippocampal cell cultures, channel subunits Kir6.1 and Kir6.2 (Yang et  al.,
2DG increased neuronal resistance to oxidative 2013). KATP channels are closed in the presence
and metabolic insults by inducing stress proteins, of intracellular ATP and open when intracellular
and in rats, 2DG allayed kainic acid seizure- ATP is depleted. Open KATP channels efflux K+,
induced memory deficits and hippocampal neu- hyperpolarizing the cell and decreasing its excit-
ron loss (Lee et al., 1999). In another model, 2DG ability. It has been hypothesized that ketones,
pretreatment of mice undergoing bilateral carotid by decreasing glycolysis and thus ATP produc-
artery occlusion protected against seizures that tion, may lower cellular excitability (Ma et  al.,
typically occur in this protocol (Redjak et  al., 2007). Decreased ATP levels, perhaps restricted
2001). Numerous mechanisms of 2DG-mediated to submembrane compartments adjacent to KATP
neuroprotection have been hypothesized, includ- channels, might lead to enhanced K+ efflux and
ing activation of adenosine monophosphate hyperpolarization. Such effects could be limited to
(AMP)-activated protein kinase (Park et al., 2009), certain neuron types (e.g., substantia nigra, den-
reduction in oxidative stress (Yao et  al., 2011), tate gyrus) that function as a gate for pathologi-
and disruption of glycosylation causing protein cal discharges (Ma et al., 2007; Tanner et al., 2011;
unfolding (Zhang et al., 2014), to name a few. Lutas and Yellen, 2013).
The rapid onset of anticonvulsant effects
POS SIBLE MECHANISMS of 2DG in vitro and in vivo suggests that 2DG
OF 2DG EFFECTS may be exerting direct actions at the synaptic or
The acute and chronic actions of 2DG probably membrane levels. The effects of 2DG on excit-
involve different cellular and molecular mecha- atory synaptic transmission were investigated
nisms. The chronic antiepileptic effects of 2DG in pilot experiments using hippocampal slices
have been associated with decreased expression (Pan et  al., 2008; Pan et  al., 2014; Rutecki et  al.,
of brain derived neurotrophic factor (BDNF) and in preparation). In hippocampal area CA3, there
its receptor, tyrosine kinase B (trkB), which are was no effect of 10 mM 2DG on the frequency or
required for kindling progression (He et al., 2004). amplitude of spontaneous excitatory postsynaptic
2DG suppression of seizure-induced increases currents (sEPSCs). However, after the induction
in BDNF and trkB is mediated by the transcrip- of epileptiform bursting in this region by appli-
tional repressor neuron restrictive silencing factor cation of elevated (7.5 mM) Ko+, 2DG reduced
(NRSF) and its nicotinamide adenine dinucle- sEPSC frequency and amplitude, suggesting that
otide hydride (NADH)–sensitive corepressor the effects of 2DG are activity-dependent—2DG
carboxy-terminal binding protein (CtBP) acting is taken up by actively firing cells and works pref-
at the promoter regions of BDNF and trkB genes. erentially in conditions of intense neuronal activ-
In pathological conditions such as seizures, glycol- ity such as seizures. The same dose of 2DG had no
ysis is enhanced to meet the energy demands of effect on miniature EPSCs isolated by exposure to
activated neurons. The increase in glycolysis ele- tetrodotoxin, which blocks sodium channels and
vates NADH, which in turn causes dissociation of thus reduces neuronal activity-mediated synap-
CtBP from NRSF, thus decreasing transcriptional tic release of neurotransmitters, but miniature
repression and resulting in increased expression of EPSCs were significantly reduced by 10 mM 2DG
BDNF and trkB. In the presence of 2DG, which in the elevated Ko+ condition. These effects are not
reduces NADH levels as a consequence of glyco- a general consequence of glycolysis inhibition,
lytic inhibition, the NRSF-CtBP complex main- since other glycolysis blockers depress sEPSCs in
tains repression of BDNF and trkB, and kindling both normal and epileptic slices (Pan et al., 2009;
progression is slowed (Garriga-Canut et al., 2006; Devinney et  al., 2009). 2DG also reduces spon-
Huang and McNamara, 2006). taneous inhibitory postsynaptic current (sIPSC)
Compared to its pivotal role in mediating frequency in conditions of elevated Ko+ (7.5
chronic 2DG effects, NRSF is not required for the mM), but to a lesser extent than sEPSCs, result-
antiepileptic effect of the KD (Hu et al., 2011). The ing in a net reduction in excitatory transmission
antiepileptic effect of 2DG was abolished in mice (Pan et al., 2015). 2DG has no significant effects
with conditional knockout of NRSF, but the KD on intrinsic membrane properties, and its acute
effects on synaptic transmission appear to be
357

Chapter 36: 2-Deoxyglucose 357

presynaptic based on analysis of miniature EPSCs spatial learning and memory in the Morris water
(Pan et al, 2015). maze (Ockuly et al., 2012). For acute testing, 2DG
Therefore, one of the unique features about was injected 15 minutes prior to the water maze
2DG is its use-dependence. That is, 2DG is taken trial each testing day. For chronic testing, 2DG (250
up only by neurons that are metabolically active, as mg/kg or 500 mg/kg i.p., twice daily) was injected
occurs in areas of circuitry involved in seizure activ- daily for 14 days before water maze testing began.
ity. This represents a distinct advantage when con- Neither protocol caused a difference in the latency
sidering the goal of a medication in targeting only to platform acquisition (spatial learning) or reten-
brain areas displaying pathological seizure activity. tion of platform location (probe test) by either dose
The acute anticonvulsant effects of 2DG may of 2DG, suggesting that 2DG has no obvious delete-
be influenced by other, as yet undetermined met- rious effect on spatial memory or learning.
abolic or electrophysiological consequences of Rats were also tested on the open field test,
glycolytic inhibition. For example, 2DG’s effects which assesses exploratory activity and has also
might be spatially limited to certain submem- been regarded as a measure of anxiety (Ockuly
brane compartments, it might alter systemic lipid et  al., 2012). Rats were pretreated (30 min prior
metabolism, or it might modify mitochondrial to open field testing) i.p. with saline, 50 mg/kg
metabolism in a manner that subsequently influ- 2DG, or 250 mg/kg 2DG in a crossover design.
ences neuronal excitability. The exploratory activity (number of lines crossed
Another compound in the glycolytic pathway, in the open field arena) did not differ between the
fructose-1,6-diphosphate (FDP), has been shown saline- and 50 mg/kg 2DG-treated groups, but
to exert acute anticonvulsant activity in several the rats receiving 250 mg/kg 2DG had decreased
seizure models in adult rats including kainic acid, motor activity (fewer lines crossed). When the
pilocarpine, and pentylenetetrazole (Lian et  al., saline group was crossed over to receive 2DG 250
2007). In that study, the effectiveness of FDP as mg/kg and the prior 250 mg/kg group was crossed
an anticonvulsant surpassed that of 2DG, KD, over to saline, the results reversed—rats receiving
and valproate. The mechanism of FDP’s anticon- 250 mg/kg 2DG had diminished open field activity
vulsant effect is unclear. Fructose-1,6-diphosphate and the group now receiving saline had increased
increases glucose flux from glycolysis into the pen- motor activity. Therefore, the 250 mg/kg dose of
tose phosphate pathway (PPP) (Figure 36.1), and 2DG caused a transient, reversible decrease in
NADPH generated in the PPP reduces glutathione, exploratory activity. Taken together, these find-
resulting in an anticonvulsant action. Therefore, ings suggest that 2DG has no permanent adverse
FDP may exert an endogenous anticonvulsant behavioral effects on spatial learning and memory,
action (Stringer and Xu, 2008). Subsequent stud- exploratory activity, or anxiety at doses that sup-
ies have established that FDP retards kindling pro- press seizures and retard epilepsy progression,
gression by attenuating BDNF and trkB expression supporting its potential for clinical use.
(Ding et al., 2010), like 2DG. Furthermore, during Some animal studies have shown that 2DG at
the kindling process, FDP inhibits the kindling- high doses is associated with adverse cardiac effects.
induced down-regulation of the expression of Oral or intravenous doses of 2DG 250–2,000
potassium-chloride cotransporter 2 (KCC2) and mg/kg given to rats or mice over 7 days caused a
decreases the expression of sodium-potassium- dose-dependent fall in mean arterial pressure,
chloride cotransporter 1 (NKCC1), suggesting that decreased respiratory rate, and increased mortality
FDP might alter the switch between GABAergic (Vijayaraghavan et al., 2006). Detailed pathological
excitation and inhibition (Ding et al., 2013). These evaluation of cardiac tissue after chronic oral 2DG
findings remain to be verified, but together, results ingestion in two rat strains revealed cardiotoxic
from several laboratories have identified modifica- effects with vacuolization of cardiac myocytes,
tion of glycolysis as a possible novel mechanism increased incidence of pheochromocytomas, and
for treatment of seizures. reduced life spans (Minor et  al., 2010). Cardiac
effects of 2DG in rats had features consistent with
2 D G A S   A P OT E N T I A L autophagy, a process by which a cell degrades
CLINICAL AGENT unnecessary cellular components in response to
Preclinical studies to evaluate safety and toxicity of nutrient stress. Comprehensive preclinical safety
2DG have demonstrated that 2DG is well tolerated and toxicology studies have subsequently dem-
in rats and dogs at doses associated with anticon- onstrated reversible species-specific cardiac tox-
vulsant and antiepileptic effects. We used both acute icity at high doses in F344 rats but not in Beagle
and chronic protocols to test the effects of 2DG on dogs; this toxicity was detectable by monitoring
358

358 section IV: Ketone-Based Metabolism

TABLE 36.1 EFFECTS OF 2DG IN MODELS OF SEIZURES AND EPILEPSY

Anticonvulsant Effects

In vitro
High Ko+ Decreased interictal and ictal bursting (CA3) Stafstrom et al., 2009
Bicuculline Decreased interictal bursting (CA3) Stafstrom et al., 2009
4-Aminopyridine Decreased interictal bursting (CA3) Stafstrom et al., 2009
In vivo
6-Hz corneal stimulation Seizure protection Stafstrom et al., 2009;
in mice Gasior et al., 2010
Audiogenic seizures in Seizure protection Stafstrom et al., 2009
Fring’s mice
Kindling of perforant path Increased afterdischarge threshold Garriga-Canut et al., 2006
Pilocarpine Decreased seizure severity and duration; Lian et al., 2007
increased seizure latency
Kainic acid i.p. No effect Lian et al., 2007
Kainic acid i.v. Decreased seizure threshold Gasior et al., 2010
Pentylenetetrazole i.v. or s.c. Decreased seizure threshold Gasior et al., 2010
Shortened seizures Lian et al., 2007
No effect Stafstrom et al., 2009
Maximum electroshock No effect Stafstrom et al., 2009
Decreased seizure threshold Gasior et al., 2010

Antiepileptic Effects

Kindling of perforant path Greater number of afterdischarges required Garriga-Canut et al., 2006


to reach class III, IV and V seizures
Kindling of olfactory bulb Greater number of afterdischarges required Stafstrom et al., 2009
to reach class III, IV and V seizures
Kindling of amygdala No effect Gasior et al., 2010

Behavioral Effects

Morris water maze No adverse effect on spatial learning or  Ockuly et al., 2012


memory
Open field test No lasting effect on exploratory activity Ockuly et al., 2012

Legend: Ko+, extracellular potassium; i.p., intraperitoneal; i.v., intravenous; s.c., subcutaneous

plasma N-terminal probrain natriuretic peptide (Maschek et al., 2004). In patients with advanced
(Nt-proBNP) (Terse et al., 2016). prostate cancer, a 2-week dose-escalating study
Experience regarding human safety of 2DG revealed dose-limiting toxicity of grade 3 asymp-
has been obtained in cancer trials (Pelicano et al., tomatic QTc prolongation at a dose of 60 mg/kg,
2006). Tumor cells are dependent on glycolysis to but doses of 45 mg/kg were well tolerated (Stein
support their metabolic requirements. By reducing et al., 2010).
glycolysis, 2DG deprives rapidly growing cells of
their required cellular fuel (Cheong et  al., 2011). CONCLUSIONS
For example, 2DG inhibits breast cancer cell The inhibition of glycolysis with compounds such
growth in a dose-dependent manner and causes as 2DG represents a novel therapeutic approach to
cell death through apoptosis (Aft et  al., 2002). epilepsy. The effects of 2DG in animal models are
In combination with adriamycin, 2DG slows the summarized in Table 36.1. 2DG exerts acute anti-
growth of solid tumors such as osteosarcoma convulsant effects in vitro that are independent of
359

Chapter 36: 2-Deoxyglucose 359

the method of seizure induction. 2DG has acute Ockuly, J., Stafstrom, C., Sutula, T., et  al. (2006).
anticonvulsant effects in vivo in several models of 2-Deoxy-D-glucose reduces epilepsy progres-
seizure induction and also possesses novel chronic sion by NRSF-CtBP-dependent metabolic regu-
antiepileptic effects against progression of seizures lation of chromatin structure. Nature Neurosci 9,
and the adverse consequences of seizure-induced 1382–1387.
Gasior, M., Yankura, J., Hartman, A.L., French, A.,
plasticity that are associated with alterations in
and Rogawski, M.A. (2010). Anticonvulsant and
neuronal gene expression. Finally, 2DG has a
proconvulsant actions of 2-deoxy-D-glucose.
favorable preliminary toxicity profile. Together, Epilepsia 5, 1385–1394.
these observations support 2DG or similar agents Greene, A.E., Todorova, M.T., and Seyfried, T.N.
as feasible for treating epilepsy in patients. The (2003). Perspectives on the metabolic manage-
detailed anticonvulsant and antiepileptic mecha- ment of epilepsy through dietary reduction of glu-
nisms of 2DG actions remain to be clarified. cose and elevation of ketone bodies. J Neurochem
86, 529–537.
AC K N OW L E D G M E N T S Hartman, A. L., Lyle, M., Rogawski, M. A., and Gasior,
This work was supported by The Charlie M. (2008). Efficacy of the ketogenic diet in the 6-
Foundation (CES), NIH RO1 25020 (TPS), the Hz seizure test. Epilepsia 49, 334–339.
Epilepsy Research Foundation New Therapy Hartman, A.L., Rubenstein, J.E., and Kossoff, E.H.
Development Project, and the Wisconsin Alumni (2013). Intermittent fasting:  a “new” historical
strategy for controlling seizures? Epilepsy Res 104,
Research Foundation.
275–279.
He, X.P., Kotloski, R., Nef, S., Luikart, B.W., Parada,
REFERENCES L.F., and McNamara, J.O. (2004). Conditional
Aft, R.L., Zhang, F.W., and Gius, D. (2002). Evaluation deletion of TrkB but not BDNF prevents epi-
of 2-deoxy-D-glucose as a chemotherapeutic leptogenesis in the kindling model. Neuron
agent: mechanism of cell death. Brit J Cancer 87, 43, 31–42.
805–812. Holmes, G. L., and Zhao, Q. (2008). Choosing the cor-
Barton, M.E., Klein, B.D., Wolf, H.H., and White, H.S. rect antiepileptic drugs: from animal studies to the
(2001). Pharmacological characterization of the 6 clinic. Pediatr Neurol 38, 151–162.
Hz psychomotor seizure model of partial epilepsy. Hori, A., Tandon, P., Holmes, G. L., and Stafstrom,
Epilepsy Res 47, 217–227. C. E. (1997). Ketogenic diet:  effects on expres-
Cheong, J.H., Park, E.S., Liang, J., Dennison, J.B., sion of kindled seizures and behavior in adult rats.
Tsavachidou, D., Nguyen-Charles, C., WaCheng, Epilepsia 38, 750–758.
K., Hall, H., Zhang, D., Lu, Y., et  al. (2011). Hu, X.L., Cheng, X., Fei, J., and Xiong, Z.Q. (2011).
Dual inhibition of tumor energy pathway by 2- Neuron-restrictive silencer factor is not required
deoxyglucose and metformin is effective against a for the antiepileptic effect of the ketogenic diet.
broad spectrum of preclinical cancer models. Mol Epilepsia 52, 1609–1616.
Cancer Ther 10, 2350–2362. Huang, Y.Z. and McNamara, J.O. (2006). Inhibiting
Devinney, M.J., Pan, Y.-Z., Rutecki, P.A., and Sutula, glycolysis to reduce seizures:  how it might work.
T.P. (2009). Differential effects of glycolytic and Nature Neurosci 9, 1351–1352.
mitochondrial metabolism on epileptic network Huttenlocher, P.R. (1976). Ketonemia and sei-
synchronization in the CA3 subfield of the hippo- zures: metabolic and anticonvulsant effects of two
campus. Soc Neurosci Abstr 331.9/I5. ketogenic diets in childhood epilepsy. Pediatr Res
Ding, Y., Wang, S., Jiang, Y., Yang, Y., Zhang, M., Guo, 10, 536–540.
Y., Wang, S., and Ding, M.P. (2013). Fructose-1,6- Ingram, D.K., and Roth, G.S. (2011). Glycolytic inhibi-
diphosphate protects against epileptogenesis by tion as a strategy for developing calorie restriction
modifying cation-chloride co-transporters in a mimetics. Exp Gerontol 46, 148–154.
model of amygdaloid-kindling temporal epilepti- Lee, J., Bruce-Keller, A.J., Kruman, Y., Chan, S.L., and
cus. Brain Res 1539, 87–94. Mattson, M.P. (1999). 2-Deoxy-D-glucose pro-
Ding, Y., Wang, S., Zhang, M.M., Guo, Y., Yang, Y., tects hippocampal neurons against excitotoxic and
Weng, S.Q., Wu, J.M., Qiu, X., and Ding, M.P. oxidative injury:  evidence for the involvement of
(2010). Fructose-1,6-diphosphate inhibits seizure stress proteins. J Neurosci Res 57, 48–61.
acquisition in fast hippocampal kindling. Neurosci Lian, X.-Y., Khan, F.A., and Stringer, J.L. (2007).
Lett 477, 33–36. Fructose-1,6-biphosphate has anticonvulsant
Gano, L.B., Patel, M., and Rho, J.M. (2014). Ketogenic activity in models of acute seizures in adult rats. J
diets, mitochondria, and neurological diseases. J Neurosci 27, 12007–12011.
Lipid Res 55, 2211–2228. Lutas, A., and Yellen, G. (2013). The ketogenic
Garriga-Canut, M., Schoenike, B., Qazi, R., Bergendahl, diet: metabolic influences on brain excitability and
K., Daley, T.J., Pfender, R.M., Morrison, J.F., epilepsy. Trends Neurosci 36, 32–40.
360

360 section IV: Ketone-Based Metabolism

Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic Redjak, K., Redjak, R., Sieklucka-Dziuba, M.,
diet metabolites reduce firing in central neurons Stelmasiak, Z., and Grieb, P. (2001). 2-
by opening K(ATP) channels. J Neurosci 27, Deoxyglucose enhances epileptic tolerance evoked
3618–3625. by transient incomplete brain ischemia in mice.
Maschek, G., Savaraj, N., Priebe, W., Braunschweiger, Epilepsy Res 43, 271–278.
P., Hamilton, K., Tidmarsh, G.F., DeYoung, L.R., Reid, C.A., Mullen, S., Kim, T.H., and Petrou, S.
and Lampidis, T.J. (2004). 2-Deoxy-D-glucose (2014). Epilepsy, energy deficiency and new thera-
increases the efficacy of adriamycin and paclitaxel peutic approaches including diet. Pharmacol Ther
in human osteosarcoma and non-small cell lung 144, 192–201.
cancers in vivo. Cancer Res 64, 31–34. Rho, J.M., and Stafstrom, C.E. (2012). The ketogenic
Minor, R.K., Smith Jr, D.L., Sossong, A.M., Kaushik, diet: what has science taught us? Epilepsy Res 100,
S., Poosala, S., Spangler, E.L., Roth, G.S., Lane, M., 210–217.
Allison, D.B., deCabo, R., et  al. (2010). Chronic Stafstrom, C.E., Ockuly, J.C., Murphree, L., Valley,
ingestion of 2-deoxy-D-glucose induces cardiac M.T., Roopra, A., and Sutula, T.P. (2009).
vacuolization and increases mortality in rats. Anticonvulsant and antiepileptic actions of 2-
Toxicol Appl Pharmacol 243, 332–339. deoxy-D-glucose in epilepsy models. Ann Neurol
Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S., 65, 435–447.
Edwards, N., Fitzsimmons, G., Whitney, A., and Stein, M., Lin, H., Jeyamohan, C., Dvorzhinski, D.,
Cross, J.H. (2008). The ketogenic diet for the treat- Gounder, M., Bray, K., Eddy, S., Goodin, S.,
ment of childhood epilepsy:  a randomised con- White, E., DiPaola. R.S. (2010). Targeting tumor
trolled trial. Lancet Neurol 7, 500–506. metabolism with 2-Deoxyglucose in patients with
Ockuly, J.C., Gielissen, J.M., Levenick, C.V., Zeal, C., castrate-resistant prostate cancer and advanced
Groble, K., Munsey, K., Sutula, T.P., and Stafstrom, malignancies. Prostate 70, 1388–1394.
C.E. (2012). Behavioral, cognitive, and safety Stringer, J.L., and Xu, K. (2008). Possible mechanisms
profile of 2-deoxy-2-glucose (2DG) in adult rats. for the anticonvulsant activity of fructose-1,6-
Epilepsy Res 101, 246–252. diphosphate. Epilepsia 49 (Suppl. 8), 101–103.
Pan, Y.Z., Ockuly, J.C., Rutecki, P.A., and Sutula, T.P. Sutula, T., and Franzoso, S. (2008). Dose-response and
(2008). Activity-dependent effects of 2DG influ- time course of action of disease-modifying effects
encing synaptic transmission, LTP, LTD, and epi- of 2DG on kindling progression:  effectiveness of
leptogenesis. Soc Neurosci Abstr, 147.146/W129. administration after seizures. Epilepsia 49 (Suppl
Pan, Y.Z., Devinney, M.J., Rutecki, P.A., and Sutula, 7), 382.
T.P. (2009). Effect of glycolytic metabolism on Tanner, G.R., Lutas, A., Martínez-François, J.R.,
synaptic currents in CA3 pyramidal neurons. Soc and Yellen, G. (2011). Single K ATP channel
Neurosci Abstr 331.10/16. opening in response to action potential firing in
Pan, Y.Z., Sutula, T., and Rutecki, P.A. (2014). Evidence mouse dentate granule neurons. J Neurosci 31,
for use-dependent and presynaptic actions of 2DG 8689–8696.
on abnormal synaptic network activity in the CA3 Terse, P.S., Joshi, P.S., Bordelon, N.R., Brys, A.M.,
region of the hippocampus. Soc Neurosci Abstr Patton, K.M., Arndt, T.P., and Sutula, T.P. (2016).
485.01. 2-Deoxy–D-Glucose (2-DG) induced cardiac
Pan Y.Z., Sutula, T.P., Rutecki, P.A. (2015). 2DG toxicity in rat: NT-proBNP and BNP as potential
reduces abnormal hippocampal network excit- early cardiac safety biomarkers. Int J Toxicol 35,
ability without changing membrane properties. 284–293.
Abstr #3.247, American Epilepsy Society Annual Vijayaraghavan, R., Kumar, D., Dube, S.N., Singh, R.,
Meeting (www.aesnet.org). Pandey, K.S., Bag, B.C., Kaushik, M.P., Sekhar, K.,
Pani, G. (2015). Neuroprotective effects of dietary Dwarakanath, B.S., and Ravindranath, T. (2006).
restriction: evidence and mechanisms. Semin Cell Acute toxicity and cardio-respiratory effects of
Dev Biol 40, 106–114. 2-deoxy-2-glucose:  a promising radio sensitiser.
Park, M., Song, K.S., Kim, H.K., Park, Y.J., Kim, H.S., Biomed Environ Sci 19, 96–103.
Bae, M.I., and Lee, J. (2009). 2-Deoxy-d-glucose Wree, A. (1990). Principles of the 2-deoxyglucose
protects neural progenitor cells against oxidative method for the determination of the local cerebral
stress through the activation of AMP-activated glucose utilization. Eur J Morphol 28, 132–138.
protein kinase. Neurosci Lett 449, 201–206. Yang, H., Guo, R., Wu, J., Peng, Y., Xie, D., Zheng, W.,
Pelicano, H., Martin, D.S., Xu, R.-H., and Huang, P. Huang, X., Liu, D., Liu, W., Huang, L., et al. (2013).
(2006). Glycolysis inhibition for anticancer treat- The antiepileptic effect of the glycolytic inhibitor
ment. Oncogene 25, 4633–4646. 2-deoxy-D-glucose is mediated by upregulation
Racine, R.J. (1972). Modification of seizure activ- of K(ATP) channel subunits Kir6.1 and Kir6.2.
ity by electrical stimulation:  II. Motor seizure. Neurochem Res 38, 677–685.
Electroencephalogr Clin Neurophysiol 32, Yao, J., Chen, S., Mao, Z., Cadenas, E., and Brinton,
281–294. R.D. (2011). 2-Deoxy-D-glucose treatment
361

Chapter 36: 2-Deoxyglucose 361

induces ketogenesis, sustains mitochondrial treatment for drug resistant epilepsy. Epilepsy
function, and reduces pathology in female Behav 33, 110–114.
mouse model of Alzheimer’s disease. PLoS One Zhang, D., Li, J., Wang, F., Hu, J., Wang, S., and Sun,
6, e21788. Y. (2014). 2-Deoxy-D-glucose targeting of glucose
Yuen, A.W.C., and Sander, J.W. (2014). Rationale for metabolism in cancer cells as a potential therapy.
using intermittent calorie restriction as a dietary Cancer Lett 355, 176–183.
362

37
Ketogenic Diets as Highly Effective Treatments
for Diabetes Mellitus and Obesity
E R I C C . W E S T M A N , M D , M H S , E M I LY M AG U I R E , M S C ,
A N D W I L L I A M S . YA N C Y J R . , M D , M H S

INTRODUCTION T2DM brings with it many indirect costs. These


The prevalence of overweight and obesity has dra- include loss of productivity for employers and loss
matically increased in the past 40 years, with levels of income for both employer and employee due
now reaching epidemic proportions in both devel- to chronic sickness and absence from work, early
oped and developing countries (Lobstein and Baur, retirement, or premature mortality (Bottomley
2005; Mastorakos et al., 2010). Obesity brings with and Raymond 2007).
it an increased risk for premature mortality and Type 2 diabetes mellitus has long been
development of comorbidities including hyperten- regarded as a chronic progressive condition that
sion, dyslipidemia, and type 2 diabetes mellitus is heterogeneous in nature (Lim et  al., 2011;
(T2DM) (Stein and Colditz, 2004). Obesity has Nyenwe, 2011). It is a complex disorder in which
been shown to be one of the main etiological fac- the interaction of both the environment and an
tors in the development of T2DM, and the current individual’s genetics results in the development
epidemic of the disease is believed to be largely of insulin resistance (IR) and, eventually, β-cell
attributable to the increased incidence of obe- dysfunction (Leahy, 2005). The β-cell function
sity (The Look AHEAD Research Group, 2003). declines over a period of time, and the develop-
Epidemiological studies have shown that the risk ment of IR to T2DM is progressive, with some
of developing T2DM increases exponentially with evidence showing it can take up to 12  years for
a body mass index (BMI) >28kg/m² (Clark, 2004). the disease to develop (Nyenwe, 2011). Obesity
And T2DM is fast becoming a serious public worsens IR, as adipose tissue releases increased
health problem all over the world—the disease is amounts of hormones (e.g., adiponectin), pro-
expected to affect 380  million people globally by inflammatory cytokines (e.g., tumor-necrosis-α),
2025 (Tahrani et al., 2010). and other factors that are involved in the develop-
Once developed, T2DM brings with it many ment of IR (Kahn et al. 2000). As obesity becomes
health, social, and economic burdens (Huang a chronic state, there is an increase in the release
et  al., 2010). In particular, individuals with the of nonesterified fatty acids (NEFAs), particularly
disease can experience a life expectancy shortened from intra-abdominal fat (Leahy, 2005). This
by up to 10 years when compared with individu- is believed to be one of the crucial factors link-
als without the disease, due to an increased risk ing obesity, specifically central obesity, to IR and
of both macrovascular and microvascular com- eventually T2DM (Stumvoll et al., 2005). Insulin
plications (Bottomley and Raymond, 2007). As resistance is thought to impair liver and pancre-
the worldwide prevalence of the disease increases, atic β-cell function, consequently reducing the
so too does the demand for and cost of medi- target tissues’ sensitivity to insulin (Tahrani et al.,
cal care in order to treat this progressive disease 2010). In addition to defective insulin action and
(Brandle et  al., 2003; Bottomley and Raymond, secretion, nonsuppressible glucagon secretion
2007). Reports show that effective glycemic con- after a meal is also evident in individuals with
trol is one of the strongest predictors in decreasing T2DM (Nyenwe, 2011).
annual healthcare costs among individuals with With the significant health, social, and eco-
T2DM (Hansen et  al., 2010). As well as having nomic burdens associated with T2DM, it is clear
direct costs to society and to an individual’s health, that devising effective interventions for treating
363

Chapter 37: Ketogenic Diets as Highly Effective Treatments 363

and, most importantly, preventing the disease an increase in blood or urine ketone bodies (typ-
are imperative. A  fundamental approach for the ically < 20 grams of dietary carbohydrate/day).
effective prevention and treatment of T2DM is These recent studies for the treatment of obesity
weight management (Miles et  al., 2002; Nyenwe, have been summarized in several meta-analy-
2011). Currently, it is understood that a moderate ses (Nordmann et al., 2006, Hession et al., 2008,
weight loss of between 5% and 10% of body weight Santos 2012, Krieger 2006, Hu 2012, Johnston
has been shown to offer improvements in glyce- 2014). These studies are notable in that, despite
mic control, blood pressure, and dyslipidemia in the higher fat content of the low-carbohydrate
patients with T2DM. diet, the adverse changes in serum cholesterol
that were predicted from other diet studies did
ORIGINS OF THE USE not occur. The low-carbohydrate diet improves
O F   K E TO G E N I C D I E T S the cardiometabolic risk factors by lowering
T O   T R E AT O B E S I T Y serum triglyceride and raising HDL-cholesterol,
One of the first descriptions of a low-carbohydrate both components of the metabolic syndrome
weight-loss program was written in 1863 by (Table 37.1; Volek and Feinman, 2005). There are
William Banting, called A Letter on Corpulence very few dietary disease endpoint outcome stud-
(Banting, 1863). As a result of Banting’s letter, a ies, but a low-carbohydrate diet was an inter-
low-carbohydrate, high-fat diet was a widely rec- vention arm in a study assessing carotid intimal
ommended diet for obesity until the mid-20th thickness after 2 years (Shai et al, 2010). In this
century. John Yudkin, a professor of nutrition in study, all diet interventions, including the low-
the United Kingdom, brought attention to carbo- carbohydrate diet led to significant regression of
hydrate restriction as a method of losing weight, measurable carotid intimal thickness. The effect
and suspected that sugar was a culprit in modern appeared to be mediated by weight loss–induced
illnesses (Yudkin, 1959). decline in blood pressure.

RANDOMIZED, ORIGINS OF THE USE


CONTROLLED TRIALS O F   K E TO G E N I C D I E T S
O F   C A R B O H Y D R AT E T O   T R E AT D I A B E T E S
RESTRICTION FOR OBESITY MELLITUS
Marginalized as a “fad diet,” the low-carbohy- The discovery of insulin and dissemination of
drate diet was forgotten as a mainstream treat- its use in the early 1920s was truly miraculous
ment for obesity in the last half of the 20th for individuals with type 1 diabetes mellitus
century. Over the last 15 years, several indepen- (T1DM), who had no endogenous insulin pro-
dent groups have reexamined low-carbohydrate duction. Insulin treatment was quickly incor-
diets and given them the new name of “carbohy- porated into clinical practice (Bliss, 1982). The
drate-restricted diets,” or “ketogenic diets” if the widespread use of insulin and subsequently
dietary carbohydrate is sufficiently low to cause developed antiglycemic medications for T2DM,

TABLE 37.1 OUTPATIENT RANDOMIZED, CONTROLLED TRIALS OF CARB OHYDRATE


VERSUS FAT RESTRICTION FOR OBESIT Y: RESULTS

Fat Restricted Diet Carbohydrate Restricted Diet

Reference n Weight LDL Trig HDL Weight LDL Trig HDL Duration

Sondike 30 –4.1 kg –17%* –6% +2% –9.9 kg* +4% –48%* +4% 3 months
Brehm 42 –3.9 kg –5% +2% +8% –8.5 kg* 0% –23%* +13% 6 months
Samaha 132 –3.1 kg –3% +2% –12% –5.1 kg* +6% –29%* –2%* 12 months
Foster 63 –4.5 kg –6% +1% +3% –7.3 kg +1% –28%* +18%* 12 months
Yancy 119 –6.5 kg –3% –15% –1% –12.0 kg* +2% –42%* +13%* 6 months
Brinkworth 40 –11.5 kg +3%* –12%* 0%* –14.5 kg +3%* –35%* +21%* 12 months
Shai** 211 –2.9 kg* –0.1% –2%* +17%* –4.7 kg* –3% –13%* +22%* 24 months
* p < .05 for between group comparison
** Study also included a third Mediterranean diet intervention (not shown here)
364

364 section IV: Ketone-Based Metabolism

however, have not allowed individuals with dia- cucumbers, spinach, asparagus, rhubarb, endive,
betes to live entirely normal lives. Like T1DM, dandelion greens, swiss chard, celery, and mush-
T2DM is known as a chronic disease with high rooms. Vegetables with carbohydrate content from
rates of retinopathy, nephropathy, and vas- 3% to 5% were tomatoes, brussels sprouts, water-
culopathy (Feudtner, 2003). Importantly, the cress, sea kale, okra, cauliflower, eggplant, cabbage,
pathophysiology of the majority (95%) of indi- canned string beans, broccoli, and canned arti-
viduals with diabetes today is related to insulin chokes. Non-carbohydrate-containing foods were
resistance (T2DM), not to insulin deficiency not limited (meat, eggs, fish).
(T1DM). Today, individuals with either T1DM So, before the modern medical therapies of
or T2DM are instructed to self-monitor their medication and insulin, the latter being discov-
blood glucose and self-administer subcutane- ered in 1921, the leaders in diabetes used a low-
ous insulin to achieve normoglycemia. Despite carbohydrate, high-fat diet for the treatment of
intensive management with insulin and oral diabetes.
medications, normoglycemia remains an elusive
goal (DCCT 1993, UKPDS 1998). D I E TA RY
In hindsight, the rapid adoption of insulin ther- R E C O M M E N D AT I O N S
apy and the hope that insulin would “cure” T2DM FOR DIABETES IN THE
may have led to a premature departure from suc- P O S T- I N S U L I N   E R A
cessful treatments for T2DM prior to the discovery The discovery and therapeutic use of insulin
of insulin. In the pre-insulin era, the pharmaco- changed the management of T1DM and T2DM,
logical treatment of diabetes mellitus included including its dietary management (Franz et  al.,
a combination of modalities including alcohol, 2002). Sansum achieved the absence of glycosu-
opioids, arsenic, and potassium bromide. More ria with increased levels of dietary carbohydrate
potent than medication, though, was the high-fat, by increasing the dose of insulin (Sansum, 1928).
low-carbohydrate diet. From a textbook written in From the calculated means of the cases reported,
1877: “There are few diseases which present to the an increase in dietary carbohydrate from 41.9 g
practitioner so clear an indication of what is to be to 196 g could be made with an increase of insu-
done … a Diabetic should exclude all saccharine lin from 20.4 units to 85.6 units/day on average.
[sugary] and farinaceous [starchy] materials from It was also observed that in patients already on
his diet” (Morgan, 1877). insulin, glycosuria remained absent and carbohy-
The leading authorities in the early 1900s, drate could be increased without increasing the
Frederick Madison Allen and Elliott P. Joslin, pub- insulin dosage if the caloric intake was reduced
lished several studies supporting the use of a low- (Richardson, 1929; Rabinowitch, 1930). From
carbohydrate, high-fat diet for diabetes. Allen used the calculated means of the cases reported, an
a pancreatectomized dog model of diabetes and increase from 73g to 144g of carbohydrate could
observed that feeding a high-carbohydrate diet be made by reducing the caloric intake from 1,788
led to glycosuria. If, however, the dogs were fed a kcal to 1,427 kcal on average. However, it is inter-
high-fat, low-carbohydrate diet, the glycosuria was esting to note that Richardson studied individu-
no longer detectable. In the treatment of diabetes als with T1DM. In regard to T2DM, Richardson
in humans, Allen employed fasting, then a step- wrote, “it has been evident that with patients who
wise reintroduction of macronutrients to find the are fat or markedly overweight it is not possible
threshold at which glycosuria developed (Allen, to make this change in the fat and carbohydrate
1914, 1915a, 1915b, 1920). Using this method, the of the diet. The tolerance has been exceeded and
average diet recommendation for diabetes was a sugar in the urine has resulted in all of those cases
diet containing 70% fat, 18% protein, 4% alcohol, which we have tried” (Richardson, 1929). Higher
and only 8% of calories from carbohydrate (Allen carbohydrate intakes could be consumed, but
et al., 1919; Westman et al., 2006). glycosuria would appear unless the insulin dos-
Like Allen, Joslin recommended a 70% fat, age was increased, or the total caloric intake was
10% carbohydrate diet for the treatment of diabe- decreased.
tes (Joslin, 1928). Joslin categorized carbohydrate- Note that these observations were made
containing foods by their carbohydrate content, using glycosuria as the measure of glycemic con-
and advised his patients to eat vegetables with trol. While individual variability exists, glucose
less than 5% carbohydrate content (Joslin, 1919). does not typically appear in the urine until the
According to his classification, vegetables with car- serum level is greater than 180 mg/dL (Buse
bohydrate content from 1% to 3% included lettuce, et al., 2003).
365

Chapter 37: Ketogenic Diets as Highly Effective Treatments 365

E VO L U T I O N Clearly, the discovery and clinical application


O F   T H E D I E TA RY of insulin therapy revolutionized the treatment of
R E C O M M E N D AT I O N S diabetes mellitus due to insulin deficiency, and no
FOR TYPE 2 DIABETES one would question the use of insulin for T1DM.
After the use of insulin became routine, the For diabetes mellitus related to insulin resistance,
amount of carbohydrate in the “diabetic diet” however, it is not clear that medication therapy
was gradually increased. Although carbohydrate (including insulin) is superior to a high-fat, low-
counting was still an important part of the diabe- carbohydrate diet for glycemic control and avoid-
tic diet recommended by the American Dietetic ance of long-term complications. This study was
Association as late as 1950, by 1971 the guidelines never done. It can be argued that because the path-
read “Important dietary concepts have developed ophysiology of T2DM involves insulin resistance
in the last decade which require some alteration in and hyperinsulinemia, using insulin therapy is
long-held precepts. There no longer appears to be counterintuitive.
any need to restrict disproportionately the intake Several limitations temper our ability to
of carbohydrate” (Bierman et al., 1971). (It is inter- directly apply this historical information today.
esting in retrospect that there are no references Due to the possibility of spectrum bias, it is dif-
included in this 1971 position paper.) ficult to ascertain the severity of diabetes that was
It appears that the change in guidelines to treated by Allen and Joslin. Their patients may
soften the limitation of dietary carbohydrate was have had less severe or progressed T2DM and
based on a few small studies. In 1963, a study of therefore might not be representative of the dia-
insulin-dependent diabetic patients compared two betic population of that time, since individuals
2,200-kcal eucaloric diets containing two relatively with more severe disease are likely to have died.
high levels of carbohydrate (41% vs. 64%), and There was also no formal distinction between type
found that both diets led to similar glucose control 1 and type 2 diabetes mellitus at the time, though
(Stone and Connor, 1963). Blood glucose meas- it was noted that children and young adults pre-
urement was not used, however, and good glucose sented with weight loss (probably type 1), while
control was defined as no glycosuria and few hypo- older patients were often obese (probably type 2).
glycemic episodes. According to the authors, “In In the sample of Allen’s patients, with a mean age
practical terms, this meant the avoidance of more of 40, most of these patients probably had type 2
than minimal glycosuria, the avoidance of more diabetes.
than 10 gm. of glucose per twenty-four-hour spec- In summary, one of the widely recommended
imen of urine and of more than one mild hypogly- treatments of diabetes mellitus in the early 1900s
cemic episode every two weeks.” before the introduction of medication therapy was
In 1971, a paper was published involving 13 a high-fat, low-carbohydrate diet.
patients with mild diabetes who were given liq-
uid formula diets containing 85%CHO/15%PRO/ NUTRITION THERAPY
0%FAT or 45%CHO/15%PRO/40%FAT diet for F O R   D I A B E T E S :   L OW
a period of 8–10 days (Brunzell et al., 1971). The G LY C E M I C D I E TA RY
carbohydrate was either dextrose or a mixture of PAT T E R N S
dextrins and maltose; calories were adjusted to The amount of glucose in the human bloodstream
maintain a constant body weight. On the 85% car- is regulated very closely, and chronic, small eleva-
bohydrate diet, the fasting blood glucose was 91 tions of serum glucose lead to glucose intolerance,
mg/dL and the fasting insulin was 16.2, compared T2DM, and the associated complications. The
with 100.2 mg/dL and 20.8, respectively, on the amount of serum glucose in an adult with a serum
lower carbohydrate diet. The conclusion was that glucose of 100 mg/dL and 5–7 liters of blood is
the glucose control was similar for a moderate or about 5–7 grams (about the amount contained in a
high carbohydrate diet. heaping teaspoon of table sugar or a few medium-
It appears that the recommendation of a 55% sized strawberries). Normal serum glucose ranges
carbohydrate diet was based on extremely small from 80 to 99 mg/dL, and when a fasting serum
studies that used either glycosuria or hypoglyce- glucose is elevated to 100–124 mg/dL, the diagno-
mia as the measure of diabetes control (not precise sis of impaired fasting glucose is made. When the
enough by today’s standards) and compared rela- fasting glucose is above 124 mg/dL or a random
tively high levels of carbohydrate intake, which does glucose is elevated above 200 mg/dL with com-
not take into account the clinical experience of low- mon symptoms such as polydipsia or polyuria, the
carbohydrate diets prior to the discovery of insulin. diagnosis of T2DM is made (American Diabetes
366

366 section IV: Ketone-Based Metabolism

Association, 2010). These small changes in concen- of a meal is a function of not only the glycemic
tration in serum glucose represent changes in the index of  the carbohydrate, but also of  the other
amount of glucose in the blood of only a few grams. macronutrients and foods ingested at  the same
The “glycemic index” is a concept that cat- time. Despite these criticisms, low glycemic index
egorizes single foods containing carbohydrate diets can lead to improvement in diabetic control
on the basis of the rise in blood glucose after the (Brand-Miller et al., 2003). However, the magni-
ingestion of  a standard amount of  carbohydrate tude of this effect on glycemic control in patients
from the particular food (Jenkins et al., 1981; Holt with  T2DM is modest, with  a typical response
et al., 1997; Ludwig, 2002). This glucose response of  0.43% reduction compared with  higher gly-
is then compared with the glucose response to a cemic diets. Because the  “low glycemic” diets
standard weight of  white bread or glucose. The in these studies have contained from 40% to 60%
“glycemic load” is a variation of  the “glycemic of  calories from  carbohydrate, it is possible that
index” in that the usual serving size of the food a more potent effect on  lowering blood glucose
is the unit of comparison, in the attempt to make may be observed with a reduction in the percent-
the construct more clinically useful. One problem age of  carbohydrate and not just the  glycemic
with  these constructs is that the  glycemic effect index.

Glucose response
900
Net Areas 50 Total Areas 500
a 8000
600 400
25

mmol • h/L
mmol • h/L
mg • h/dL

mg • h/dL

300 6000 a 300


0 0
4000 b 200
–300 c
–25
b 2000 100
–600 b
–900 –50 0 0 20
350 Standard CHO Fasting Standard CHO Fasting
Diet Free Diet Free 18
300
16

250 14
mmol/L
12
mg/dL

200
10

150 8

6
100
Fasting 4
50 CHO Free
Standard Diet 2
B L D
0 0
0 2 4 6 8 10 12 14 16 18 20 22 24
Time (Hours)

FIGURE  37.1 Fasting and Postprandial Glucose Response of a Ketogenic Diet 24-h glucose response. The open
circle–solid line represents the mean glucose concentration at several time points during the first 24 h of both days
during which the standard diet was ingested (i.e. day 1 of each arm of the study). The triangle–dotted line represents
the mean glucose concentration during the last 24 h on a carbohydrate-free diet. The closed circle–solid line represents
the mean glucose concentration during the last 24 h of the fast (energy-free) diet. B, L, D, indicate the times at which
breakfast, lunch, and dinner were ingested.The net area response (Left Insert) indicates the area under the curve using
the fasting concentration as baseline. Different letters on bars indicate statistically significant differences (Friedman
P = <0.0012).The total area response (Right Insert) indicates the area under the curve, using zero as baseline. Different
letters on bars indicate statistically significant differences (Friedman: P = <0.0001) (Nuttall, Almokayyad, Gannon, 2015).
367

Chapter 37: Ketogenic Diets as Highly Effective Treatments 367

G LY C E M I C E F F E C T O F individuals with T2DM. In this study, an ad libi-


D I E TA RY C A R B O H Y D R AT E tum low-carbohydrate, ketogenic diet (<20 g/day)
In controlled studies, consuming foods of lower gly- led to a spontaneous reduction in caloric intake
cemic index versus foods of higher glycemic index and improvement in insulin sensitivity measured
leads to a reduction in postprandial glucose and insu- by insulin clamp technique (Boden et  al., 2005).
lin. When the glycemic concept is taken further and Additionally, serum glucose levels improved substan-
applied to the reduction of the amount of carbohy- tially and consistently enough in the 10 subjects that
drate to less than 20–40 g/d—all of the carbohydrate- hemoglobin A1c improved significantly from base-
containing foods are “very low” to “no” glycemic line after only 14 days on the low-carbohydrate diet.
index foods. (Fat has a glycemic index of 0 because
the consumption of fat does not raise the blood glu- RANDOMIZED,
cose.) When the carbohydrate intake is this low, the CONTROLLED TRIALS
glucose and insulin responses to the meals are even O F   C A R B O H Y D R AT E
lower. In fact, the fasting and postprandial glucose RESTRICTION FOR TYPE 2
and insulin levels after carbohydrate-deficient meals DIABETES MELLITUS
are almost as low as total fasting (not eating any- There has been a recent interest in studying the
thing at all) (Figure 37.1; Nuttall et  al., 2015). The use of low-carbohydrate diets for the treatment of
lack of postprandial rise in glucose and insulin for an T2DM. Several of the recent studies have limited
extremely low carbohydrate diet (<20 g/day) has been carbohydrate intake to the degree that was used by
replicated in two other studies in subjects without Allen and Joslin (Table 37.3, Daly 2006, Westman
diabetes (Bisschop et al., 2000; Noakes et al., 2006). 2008, Gulbrand 2012, Saslow 2014, Tay 2014,
In summary, lowering the glycemic index and Yamada 2014, Mayer 2014). The consistent find-
the absolute amount of carbohydrate in the diet ing is that the lower carbohydrate, even ketogenic,
can have a profound effect on lowering the blood diets lead to a greater improvement in T2DM than
glucose; dietary protein has a small effect on blood the diets containing higher levels of carbohydrate.
glucose; dietary fat has none. This is the rationale Lowering dietary carbohydrate intake demon-
for use of a low-carbohydrate diet for the treat- strated benefits on glycemic control beyond its
ment and prevention of diabetes. weight loss effects in some of the studies, while at
the same time lowering antiglycemic medication
C O N T R O L L E D O U T PAT I E N T requirements.Two meta-analyses have also sum-
S T U D I E S O F   L OW E R marized these studies (Kirk 2008, Dyson 2008).
C A R B O H Y D R AT E D I E T S
FOR TYPE 2 DIABETES P I L OT S T U DY O F   A
MELLITUS K E TO G E N I C D I E T
From 1998 to 2004, several controlled studies FOR WEIGHT LOS S
showed that lowering the dietary carbohydrate can IN OBESITY VERSUS TYPE 2
lead to a reduction in fasting blood glucose and DIABETES MELLITUS
HbA1c, and that this effect can be independent of While clinical experience suggests that weight loss
weight loss (Table 37.2). However, none of these is slower for individuals with T2DM than obesity
studies lowered the carbohydrate intake to the lev- alone, there are few studies directly comparing this
els of the ketogenic diet, which had been employed clinical observation for ketogenic diets. One of this
clinically in the early 1900s, as discussed earlier. chapter’s authors (EM) conducted a retrospective
A review of existing studies in 2008 concluded that cohort study to assess the rate of weight change that
a low-carbohydrate diet would be useful for the occurred between a group of overweight or obese
treatment of T2DM (Dyson, 2008). type 2 diabetics compared with age-, weight-, and
gender-matched control groups of overweight or
M E TA B O L I C WA R D S T U D Y obese individuals (unpublished data).
O F   T H E K E TO G E N I C D I E T The intervention was the Go Lower Low
FOR TYPE 2 DIABETES Carbohydrate Diet, which provided around 10%
MELLITUS of total energy from carbohydrates, 30% of total
A major limitation of out-patient clinical trials is energy from protein, and 60% of total energy from
that the dietary intake is estimated by self-reported fat. All meals were delivered to individuals, pro-
intake questionnaires and may not reflect actual viding them with breakfast, lunch, dinner, and up
dietary intake. This limitation was overcome in a to 2 snacks per day. Weekly support was provided
carefully controlled metabolic ward study among to participants with additional nutrition support
368
TABLE 37.2 CONTROLLED STUDIES OF LOWER CARB OHYDRATE DIETS FOR T YPE 2 DIABETES MELLITUS

Reference N BMI KCAL CHO PRO FAT Duration Post Pre Post Weight
Glucose HgbA1c HgbAIc Change
kg/m2 % % % mg/dL % % kg

Gutierrez 19 27.9 Varied 55 20 25 12 weeks 183 8.1 8.9 –0.8


1998 25 45 30 8 weeks 192 9.9 8.1 –1.4
Heilbronn 35 33 1541 73 17 10 12 weeks 131 8.5 7.0 –6.6
1999 1596 50 18 32 MUF 119 7.8 6.8 –6.6
1613 50 17 33 SF 122 7.5 6.6 –6.6
Heilbronn 45 22 1440 60 HGI 22 18 12 weeks 110 6.7 6.1 –4.8
2002 1440 60 LGI 22 18 116 6.7 6.0 –4.4
Gannon 12 31 2250 55 15 34 5 weeks 114 8.0 7.7 0
2003 2235 40 30 30 114 8.1 7.3 0
Rizkalla 12 31 2291 38 HGI 20 37 4 weeks 176 7.5 7.6 –0.6
2004 2222 36 LGI 21 37 165 7.6 7.2 –0.6
Gannon 8 31 2825 55 15 30 5 weeks 198 9.8 9.8 –1.8
2004 2835 20 30 50 126 9.8 7.6 –1.8

BMI = body mass index, HGI = high glycemic index, LGI = low glycemic index, MUF = high monounsaturated fat, SF = high saturated fat
369
TABLE 37.3 OUTPATIENT RANDOMIZED, CONTROLLED TRIALS OF CARB OHYDRATE
RESTRICTION FOR T YPE 2 DIABETES MELLITUS

Reference N BMI Carbohydrates Duration Start End Difference Difference


HgbA1c HgbA1c HgbA1c Weight
kg/m2 grams/day months percent percent Percent kilograms

Daly 102 36 170 3 9.0 8.8 –0.2 –0.9


2006 110 9.0 8.5 –0.5 –3.6
Westman 49 38 100 6 8.3 7.8 –0.5* –6.9
2008 20 8.8 7.3 –1.5* –11.0
Guldbrand 61 32 225 24 7.2 7.4 +0.2 –4.0
2012 90 7.5 7.5 –0.0 –4.0
Saslow 34 36 138 3 6.9 6.9 0* –2.6
2014 58 6.6 6.0 –0.6* –5.5
Tay 115 34 205 6 7.3 6.6 –0.7* –12
2014 57 7.4 6.2 –0.6* –12
Yamada 24 25 203 6 7.7 7.5 –0.2* –1.4
2014 126 7.6 7.0 –0.6* –2.6*
Mayer 46 40 156 + O 12 7.6 7.7 +0.1* –8.1
2014 76 7.6 6.9 –0.7* –7.5
*p < .05 for between group comparison.
n = sample size, O = Orlistat
370

370 section IV: Ketone-Based Metabolism

provided by qualified nutritionists. Between weeks intervention trial at the Durham Veterans Affairs
9 and 12, only breakfast and snacks were provided, Medical Center (Yancy et al., 2003). At diet ini-
which required individuals to prepare two meals tiation, diabetic medication was reduced to avoid
per day. hypoglycemia. At baseline, the mean age was 56.3
A sample of 20 participants (10 obese with dia- years and the mean BMI was 42.2 kg/m2. After
betes and 10 obese without diabetes) was identified 16 weeks, the mean hemoglobin A1c decreased
for inclusion in this study. At baseline, the mean by 15% from baseline (7.4% to 6.3%, p < .001).
age was 55.5 years, mean BMI was 36.0 kg/m2, Medications for diabetes were discontinued in 7
and 70% were female, and the mean HgbA1c was subjects, reduced in 10 subjects, and unchanged
12.3%. After 3 months of following the dietary in 5 subjects. The mean body weight decreased by
protocol, obese nondiabetic participants had lost 6.5% from 130.9 kg to 122.4 kg (p < .001). Fasting
significantly more weight than obese type 2 dia- serum triglyceride decreased 40% (2.61 mmol/L to
betic participants (-20.56 ± 8.8 kg versus -12.84 ± 1.54 mmol/L, p < .001), while other serum lipid
8.1 kg; p = .05). However, there was no significant measurements did not change significantly.
difference between these groups in mean weight
change at 12 months. These results are in line with CASE REPORT
other previous studies that have demonstrated that OF REMIS SION OF SEVERE
patients with type 2 diabetes do not lose as much TYPE 2 DIABETES MELLITUS
weight as their nondiabetic counterparts (Khan et W I T H   K E TO G E N I C   D I E T
al., 2000; Miles et al., 2002). One of the authors (ECW) recently treated an oth-
erwise healthy 60-year-old Caucasian male with a
NONRANDOMIZED STUDIES BMI of 26.9 kg/m2 who had a new onset of T2DM.
U S I N G T H E   K E TO G E N I C The initial hemoglobin A1C was 10.5% on routine
DIET IN TYPE 2 DIABETES annual testing, and serum glucose levels were
MELLITUS also reflective of hyperglycemia. He was given
In order to assess the effect of the ketogenic diet instruction on how to follow a low-carbohydrate,
on T2DM in clinical practice, one of the authors ketogenic diet (<20 grams of carbohydrate/day),
(ECW) performed chart reviews of physicians and the repeat hemoglobin A1C at 1, 5, 12, and
who had used carbohydrate restriction for many 24 months was 6.4%, 5.5%, 5.6%, and 5.5% respec-
years in their clinical practices (Hickey et al., 2003; tively (Figure 37.2). The current recommended
O’Neill et  al., 2003; Vernon et  al., 2003; Vernon approach to manage severe diabetes mellitus is
et al., 2004) (Table 37.4). Nielsen also reported the to start chronic insulin therapy. This case report
results of long-term follow-up in a clinical setting suggests that potent lifestyle approaches like the
(Nielsen and Joensson, 2008). These cases show ketogenic diet may be useful instead of medication
that reversal of T2DM is possible in the context of therapy.
selection of highly motivated individuals, or as a
result of programs that have excellent adherence. CONCLUSION
One of these research projects was a prospec- In summary, lifestyle modification that reduces
tive study which enrolled 28 overweight sub- carbohydrate intake is effective in the treatment
jects with T2DM into a 16-week single-arm diet of obesity, T2DM, and metabolic syndrome.

TABLE 37.4 CLINICAL PRACTICE AND NONRANDOMIZED STUDIES


OF CARBOHYDRATE RESTRICTION FOR T YPE 2 DIABETES MELLITUS

Reference N Baseline Carbohydrates Duration Pre Post Weight


Weight (kg) Pct of Calories/day Months HbA1c HBA1c Difference
% % %

Vernon 2003 14 123.2 10% 8 10.0 5.9 –9.7


O’Neill 2003 20 82 10% 10 8.4 5.8 –6.7
Nielsen 2005 16 100 20% 12 8.0 6.6 –11.9
Yancy 2005 28 131.4 7% 4 7.5 6.3 –6.6
371

Chapter 37: Ketogenic Diets as Highly Effective Treatments 371

64 y/o Male, BMI 29.1, New Onset Diabetes


205 12
200 11
10.5
195 198 10
190 9
8
185 189
7
180 6.3
5.9 6
175 5.5
5
170
Weight (pounds) 4
165 170 169 169 3
HgbA1c (%)
160 2
155 1
150 0
Oct-10
Jan-11
Apr-11
Jul-11
Oct-11
Jan-12
Apr-12
Jul-12
Oct-12
Jan-13
Apr-13
Jul-13
Oct-13
Jan-14
Apr-14
Jul-14
Oct-14
Jan-15
Low Carbohydrate Ketogenic Diet
(No medications)

FIGURE 37.2 Treatment of New Onset Type 2 Diabetes Mellitus with a Ketogenic Diet

A low-carbohydrate, ketogenic diet combines two D.B., Larson, G., Lustig, R.H., et al. (2008). Dietary
approaches that, on their own, improve blood glu- carbohydrate restriction in type 2 diabetes mel-
cose control: weight loss and reduction of glycemic litus and metabolic syndrome:  time for a critical
index/load diet response to dietary intake. appraisal. Nutr Metab (Lond) 5, 9.
As for most ongoing lifestyle change programs, Allen, F.M. (1914). Studies concerning diabetes. JAMA
63, 939–943.
research efforts to improve dietary adherence are
Allen, F.M. (1915a). Prolonged fasting in diabetes.
needed. We have found that introducing the con-
Trans Assoc Am Physicians 30, 323–329.
cept that T2DM is possibly reversible, and that Allen, F.M. (1915b). The treatment of diabetes. Boston
individuals may be able to have normal blood Med Surg J 172, 241–247.
glucose off medications is highly motivating, and Allen, F.M., Stillman, E., and Fitz, R. (1919). Total
the reduction or elimination of medications may dietary regulation in the treatment of diabetes.
improve long-term adherence. Most individuals Monograph No. 11. (New  York:  The Rockefeller
with T2DM have been told repeatedly that they Institute for Medical Research).
will always have diabetes and always require Allen, F.M. (1920). Protein diets and undernutrition in
medication—but this is clearly not the case in treatment of diabetes. JAMA 74, 571–577.
some individuals with amelioration of the under- American Diabetes Association. (2010). Diagnosis
lying insulin resistance. and classification of diabetes mellitus. Diabetes
The underlying principle of carbohydrate Care 33 (Suppl 1), S62.
restriction and the history of using the low- Banting, W. (2005) [1863]. Letter on Corpulence.
(New York: Cosimo Classics).
carbohydrate diet for T2DM suggest that the
Bierman, E.L., Albrink, M.J., and Arky, R.A. (1971).
low-carbohydrate approach may play an impor- Special report: principles of nutrition and dietary
tant nonpharmacologic role in reversing the recommendations for patients with diabetes mel-
current epidemic of “diabesity” (Accurso et  al., litus: 1971. Diabetes 20, 633–634.
2008; Feinman et  al., 2015; Khazrai et  al., 2014). Bisschop, P.H., Pereira Arias, A.M., Ackermans,
Furthermore, it suggests that the common M.T., Endert, E., Pijl, H., Kuipers, F., Meijer, A.J.,
approach of antiglycemic medication + higher car- Sauerwein, H.P., and Romjin, J.A. (2000). The
bohydrate diets should be more rigorously tested effects of carbohydrate variation in isocaloric diets
against the low-carbohydrate, ketogenic approach on glycogenolysis and gluconeogenesis in healthy
for the treatment of T2DM. men. J Clin Endocrinol Metab 85, 1963–1967.
Bliss, M. (1982). The Discovery of Insulin.
REFERENCES (Chicago: University of Chicago Press).
Accurso, A., Bernstein, R.K., Dahlqvist, A., Draznin, Boden, G., Sargrad, K., Homko, C., Mozzoli, M., and
B., Feinman, R.D., Fine, E.J., Gleed, A., Jacobs, Stein, T.P. (2005). Effect of a low-carbohydrate diet
372

372 section IV: Ketone-Based Metabolism

on appetite, blood glucose levels, and insulin resis- (2015). Dietary carbohydrate restriction as the first
tance in obese patients with type 2 diabetes. Ann approach in diabetes management: critical review
Intern Med 142, 403–411. and evidence base. Nutrition 31, 1–13.
Bottomley, J.M., and Raymond, F.D. (2007). Pharmaco- Feudtner, C. (2003). Bittersweet:  Diabetes, Insulin,
economic issues for diabetes therapy. Best Pract and the Transformation of Illness. (Chapel
Res Clin Endocrinol Metab 21, 657–685. Hill: University of North Carolina Press).
Brandle, M., Zhou, H., Smith, B.R., Marriott, D., Foster, G.D., Wyatt, H.R., Hill, J.O., McGuckin, B.G.,
Burke, R., Tabaei, B.P., Brown, M.B., and Herman, Brill, C., Mohammed, B.S., Szapary, P.O., Rader,
W.H. (2003). The direct medical cost of type 2 dia- D.J., Edman, J.S., and Klein, S. (2003). A random-
betes. Diabetes Care 26, 2300–2304. ized trial of a low-carbohydrate diet for obesity. N
Brand-Miller, J., Hayne, S., Petocz, P., and Colagiuri, S. Engl J Med 348, 2082–2090.
(2003). Low-glycemic index diets in the manage- Foster, G.D., Wyatt H.R., Hill, J.O., Makris, A.P.,
ment of diabetes:  a meta-analysis of randomized Rosenbaum, D.L., Brill, C., Stein, R.I., Mohammed,
controlled trials. Diabetes Care 26, 2261–2267. B.S., Miller, B., Rader, D.J., et  al. (2010). Weight
Brehm, B.J., Seeley, R.J., Daniels, S.R., and D’Alessio, and metabolic outcomes after 2  years on a low-
D.A. (2003). A randomized trial comparing a very carbohydrate versus low fat diet. Ann Intern Med
low carbohydrate diet and a calorie-restricted 153, 147–157.
low fat diet on body weight and cardiovascular Franz, M.J., Bantle, J.P., Beebe, C.A., Brunzell, J.D.,
risk factors in healthy women. J Clin Endocrinol Chiasson, J.L., Garg, A., Holzmeister, L.A.,
Metab 88, 1617–1623. Hoogwerf, B., Mayer-Davis, E., Mooradian, A.D.,
Brinkworth, G.D., Noakes, M., Buckley, J.D., Keogh, et  al. (2002). Evidence-based nutrition principles
J.B., and Clifton, P.M. (2009). Long-term effects and recommendations for the treatment and pre-
of a very-low-carbohydrate weight loss diet com- vention of diabetes and related complications.
pared with an isocaloric low-fat diet after 12 mo. Diabetes Care 25, 148–198.
Am J Clin Nutr 90, 23–32. Gannon, M.C., and Nuttall, F.Q. (2004). Effect of a
Brunzell, J.D., Lerner, R.L., Hazzard, W.R., Porte, D., high-protein, low-carbohydrate diet on blood
Jr., and Bierman, E.L. (1971). Improved glucose glucose control in people with type 2 diabetes.
tolerance with high carbohydrate feeding in mild Diabetes 53, 2375–2382.
diabetes. N Engl J Med 284, 521–524. Gannon, M.C., Nuttall, F.Q., Saeed, A., Jordan, K.,
Buse, J.B., Polonsky, K.S., and Burant, C.F. (2003). and Hoover, H. (2003). An increase in dietary
Type 2 Diabetes Mellitus. In Williams Textbook protein improves the blood glucose response in
of Endocrinology, 10th ed., P.R. Larsen, H.M. persons with type 2 diabetes. Am J Clin Nutr 78,
Kronenberg, S.  Melmed, and K.S. Polonsky, eds. 734–741.
(Philadelphia: Saunders), p. 1457. Garg, A., Bonanome, A., Grundy, S.M., Zhang, Z.J.,
Clark, M. (2004). Is weight loss a realistic goal of and Unger, R.H. (1988). Comparison of a high-
treatment in type 2 diabetes? The implications of carbohydrate diet with a high-monounsaturated-
restraint theory. Patient Educ Couns, 53, 277–283. fat diet in patients with non-insulin-dependent
DCCT. (1993). The effect of intensive treatment of dia- diabetes mellitus. N Engl J Med 319, 829–834.
betes on the development and progression of long- Garg, A., Bantle, J.P., Henry, R.R., Coulston, A.M.,
term complications in insulin-dependent diabetes Griver, K.A., Raatz, S.K., Brinkley, L., Chen, Y.D.,
mellitus. The Diabetes Control and Complications Grundy, S.M., and Huet, B.A. (1994). Effects of
Trial Research Group. N Engl J Med 329, 977–986. varying carbohydrate content of diet in patients
Daly, M.E., Paisey, R., Paisey, R., Milward, B.A., Eccles, with non-insulin-dependent diabetes mellitus.
C. Williams, K., Hammersley, S., MacLeod, K.M., JAMA 271, 1421–1428.
and Gale, T.J. (2006). Short-term effects of severe Gutierrez, M., Akhavan, M., Jovanovic, L., and
dietary carbohydrate-restriction advice in Type 2 Peterson, C.M. (1998). Utility of a short-term 25%
diabetes—a randomized controlled trial. Diabet carbohydrate diet on improving glycemic control
Med 23, 15–20. in type 2 diabetes mellitus. J Am Coll Nutr 17,
Dashti, H.M., Al-Zaid, N.S., Mathew, T.W., Al- 595–600.
Mousawi, M., Talib, H., Asfar, S.K., and Behbahani, Hansen, R.A., Farley, J.F., Droege, M., and Maciejewski,
A.I. (2006). Long-term effects of ketogenic diet in M.L. (2010). A Retrospective cohort study of eco-
obese subjects with high cholesterol level. Mol Cell nomic outcomes and adherence to monotherapy
Biochem 286, 1–9. with metformin, pioglitazone, or a sulfonylurea
Dyson, P.A. (2008). A review of low and reduced car- among patients with Type 2 Diabetes Mellitus in
bohydrate diets and weight loss in type 2 diabetes. the United States From 2003 to 2005. Clin Ther 32,
J Hum Nutr Diet 21(6), 530–538. 1308–1319.
Feinman, R.D., Pogozelski, W.D., Astrup, A., Heilbronn, L.K., Noakes, M., and Clifton, P.M. (1999).
Bernstein, R.K., Fine, E.J., Westman, E.C. Accurso, Effect of energy restriction, weight loss, and diet
A., Frassetto, L., Gower, B.A., McFarlane, S.I., et al. composition on plasma lipids and glucose in
373

Chapter 37: Ketogenic Diets as Highly Effective Treatments 373

patients with type 2 diabetes. Diabetes Care 22, diabetes:  a meta-analysis. J Am Diet Assoc 108,
889–895. 91–100.
Heilbronn, L.K., Noakes, M., and Clifton, P.M. (2002). Krieger, J.W., Sitren, H.S., Daniels, M.J., and
The effect of a high- and low-glycemic index Langkamp-Henken, B. (2006). Effects of varia-
energy restricted diets on plasma lipid and glucose tion in protein and carbohydrate intake on body
profiles in type 2 diabetic subjects with varying mass and composition during energy restriction: a
glycemic control. J Am Coll Nutr 21, 120–127. meta-regression. Am J Clin Nutr 83, 260–274.
Hession, M., Rolland, C., Kulkarni, U., Wise, A., and Leahy, J.L. (2005). Pathogenesis of type 2 diabetes mel-
Broom, J. (2009). A systematic review of random- litus. Arch Med Res 36, 197–209.
ized controlled trials of low-carbohydrate vs. low- Lim, E.L., Hollingsworth, K.G., Aribisala, B.S., Chen,
fat/low-calorie diets in the management of obesity M.J., Mathers, J.C. and Taylor, R. (2011). Reversal
and its comorbidities. Obes Rev, 10, 36–50. of type 2 diabetes: normalisation of beta cell func-
Hickey, J.T., Hickey, L., Yancy Jr., W.S., Hepburn, J., tion in association with decreased pancreas and
and Westman, E.C. (2003). Clinical use of a car- liver triacylglycerol. Diabetologia, 54, 2506–2514.
bohydrate-restricted diet to treat the dyslipidemia Ludwig, D.S. (2002). The glycemic index: physiologi-
of the metabolic syndrome. Metab Syndr Relat cal mechanisms relating to obesity, diabetes, and
Disord 1, 227–232. cardiovascular disease. JAMA 287, 2414–2423.
Holt, S.H., Brand-Miller, J.C., and Petocz, P. (1997). An Mayer, S.B., Jeffreys, A.S., Olsen, M.K., McDuffie, J.R.,
insulin index of foods: the insulin demand gener- Feinglos, M.N., and Yancy Jr., W.S., (2014). Two
ated by 1000-kJ portions of common foods. Am J diets with different haemoglobin A1c and antigly-
Clin Nutr 66, 1264–1276. caemic medication effects despite similar weight
Hu, T., Mills, K.T., Demanelis, K., Eloustaz, M., Yancy loss in type 2 diabetes. Diabetes Obes Metab 16,
Jr., W.S., Kelly, T.N., He, J., and Bazzano, L.A. 90–93.
(2012). Effects of low-carbohdyrate diets versus Miles, J.M., Leiter, J., Hollander, P., Wadden, T.,
low-fat diets on metabolic risk factors: a meta- Anderson, J.W., Doyle, M., Foreyt, J., Aronne, L.,
analysis of randomized controlled clinical trials. and Klein S. (2002). Effect of orlistat in overweight
Am J Epidemiol 176, S44–S54. and obese patients with type 2 diabetes treated
Huang, M.F., Courtney, M., Edwards, H., and with metformin. Diabetes Care 25, 1123–1128.
McDowell, J. (2010). Factors that affect health Morgan, W. (1877). Diabetes Mellitus:  Its History,
outcomes in adults with type 2 diabetes:  a cross- Chemistry, Anatomy, Pathology, Physiology, and
sectional study. Int J Nurs Stud 47, 542–549. Treatment. (London:  Homeopathic Publishing
Jenkins, D.J., Wolever, T.M., Taylor, R.H., Barker, Company).
H., Fielden, H., Baldwin, J.M., Bowling, A.C., Nielsen, J.V., and Joensson, E.A. (2008). Low-
Newman, H.C., Jenkins, A.L., and Goff, D.V. carbohydrate diet in type 2 diabetes:  stable
(1981). Glycemic index of foods:  a physiological improvement of bodyweight and glycaemic con-
basis for carbohydrate exchange. Am J Clin Nutr trol during 44  months follow-up. Nutr Metab
34, 362–366. (Lond) 5, 14.
Johnston, B.C., Kanters, S., Bandayrel, K., Naji, Noakes, M., Foster, P.R., Keogh, J.B., James, A.P.,
F., Siemieniuk, R.A., Ball, G.D.C., Busse, J.W., Mamo, J.C., and Clifton, P.M. (2006). Comparison
Thorlund, K., Guyatt, G., Jansen, J.P., and Mills, E.J. of isocaloric very low carbohydrate/high saturated
(2014). Comparison of weight loss among named fat and high carbohydrate/low saturated fat diets
diet programs in overweight and obese adults. A on body composition and cardiovascular risk.
meta-analysis. JAMA 312:923-933. Nutr Metab (Lond). 3, 7.
Joslin, E.P. (1928). Ideals in the treatment of diabetes Nordmann, A.J., Nordmann, A., Briel, M., Keller,
and methods for their realization. N Engl J Med U., Yancy Jr., W.S., Brehm, B.J., and Bucher, H.C.
198, 379–382. (2006). Effects of low-carbohydrate vs low-fat diets
Joslin, E.P. (1919). A Diabetic Manual for the Mutual on weight loss and cardiovascular risk factors.
Use of Doctor and Patient. (Philadelphia:  Lea & Arch Intern Med 166, 285–293.
Febiger). Nuttall, F.Q., Almokayyad, R.M., and Gannon, M.C.
Khazrai, Y.M., Defeudis, G., and Pozzilli, P. (2014). (2015). Comparison of a carbohydrate-free diet vs.
Effect of diet on type 2 diabetes mellitus: a review. fasting on plasma glucose, insulin and glucagon in
Diabetes Metab Res Rev 30 (Suppl 1), 24–33. type 2 diabetes. Metabolism 64, 253–262.
Khan, M.A., St Peter, J.V., Breen, G.A., Hartley, G.G., Nyenwe, E.A. (2011). Management of type 2 diabe-
and Vessey, J.T. (2000). Diabetes disease stage pre- tes: evolving strategies for the treatment of patients
dicts weight loss outcomes with long-term appetite with type 2 diabetes. Metabolism 60, 1–23.
suppressants. Obesity Res 8, 43–48. O’Neill, D.F., Westman, E.C., and Bernstein, R.K.
Kirk, J.K., Graves, D.E., Craven, T.E., Lipkin, E.W., (2003). The effects of a low-carbohydrate regimen
Austin, M., and Margolis, K.L. (2008). Restricted- on glycemic control and serum lipids in diabetes
carbohydrate diets in patients with type 2 mellitus. Metab Syndr Relat Disord 1, 291–298.
374

374 section IV: Ketone-Based Metabolism

Rabinowitch, I.M. (1930). Experiences with a high Tahrani, A.A., Piya, M.K., Kennedy, A., and Barnett,
carbohydrate-low calorie diet for the treatment of A.H. (2010). Glycaemic control in type 2 diabe-
diabetes mellitus. CMAJ 23, 489–498. tes:  targets and new therapies. Pharmacol Ther
Richardson, R. (1929). High-carbohydrate diets in dia- 125, 328–361.
betes mellitus. Am J Med Sci 177, 426–430. Tay, J., Luscombe-Marsh, N.D., Thompson, C.H.,
Rizkalla, S.W., Taghrid, L., Laromiguiere, M., Huet, D., Noakes, M., Buckley, J.D., Wittert, G., A., Yancy,
Boillot, J., Rogoir, A., Elgrably, F., and Slama, G. W.S., and Brinkworth, G.D. (2014). A very low-
(2004). Improved plasma glucose control, whole- carbohydrate, low-saturated fat diet for type 2 dia-
body glucose utilization, and lipid profile on a betes management:  a randomized trial. Diabetes
low-glycemic index diet in type 2 diabetic men: a Care 37, 2909–2918.
randomized controlled trial. Diabetes Care 27, The Look Ahead Research Group. (2003). Look
1866–1872. AHEAD (Action for Health in Diabetes):  design
Samaha, F.F., Iqbal, N., Seshadri, P., Chicano, K.L., and methods for a clinical trial of weight loss for
Daily, D.A., McGrory, J., Williams, T., Williams, the prevention of cardiovascular disease in type 2
M., Gracely, E.J., and Stern, L. (2003). A low- diabetes. Control Clin Trials 24, 610–628.
carbohydrate as compared with a low-fat diet in UKPDS. United Kingdom Prospective Diabetes Study
severe obesity. N Engl J Med 348, 2074–2081. 33. (1998). Intensive blood-glucose control with
Sansum, W.D., Blatherwick, N.R., and Bowden, R. sulphonylurea or insulin compared with con-
(1928). The use of high carbohydrate diets in ventional treatment and risk of complications in
the treatment of diabetes mellitus. JAMA 86, patients with type 2 diabetes. Lancet 352, 837–853.
178–181. UKPDS. United Kingdom Prospective Diabetes Study
Santos, F.L., Esteves, S.S., da Costa Pereira, A., Yancy, 34. (1998). Effect of intensive blood-glucose control
W.S.Jr., and Nunes, J.P.L. (2012). Systematic review with metformin on complications in overweight
and meta-analysis of clinical trials of the effects of patients with type II diabetes. Lancet 352, 854–865.
low carbohydrate diets on cardiovascular risk fac- Vernon, M.C., Mavropoulos, J., Transue, M., Yancy Jr.,
tors. Obes Rev 13(11), 1048-1066. W.S., and Westman, E.C. (2003). Clinical experi-
Saslow, L.R., Kim, S., Daubenmier, J.J., Moskowitz, ence of a carbohydrate-restricted diet: effect on
J.T., Phinney, S.D., Goldman, V., Murphy, E.J., diabetes mellitus. Metab Syndr Relat Disord 1,
Cox, R.M., Moran, P., and Hecht, F.M. (2014). 233–237.
A randomized pilot trial of a moderate carbohy- Vernon, M.C., Kueser, B., Transue, M., Yates, H.E.,
drate diet compared to a very low carbohydrate Yancy Jr., W.S., and Westman, E.C. (2004). Clinical
diet in overweight or obese individuals with type experience of a carbohydrate-restricted diet for the
2 diabetes mellitus or prediabetes. PLoS One 9, metabolic syndrome. Metab Syndr Relat Disord 2,
e91027. 180–186.
Shai, I., Schwarzfuchs, D., Henkin, Y., Shahar, D.R., Volek, J.S., and Feinman, R.D. (2005). Carbohydrate
Witkow, S., Greenberg, I., Golan, R., Fraser, D., restriction improves the features of metabolic syn-
Bolotin, A., Vardi, H., et  al. (2008). Weight loss drome: metabolic syndrome may be defined by the
with a low-carbohydrate, Mediterranean, or low- response to carbohydrate restriction. Nutr Metab
fat diet. N Engl J Med 359, 229–241. (Lond) 2, 31.
Shai, I., Spence, D., Schwarzfuchs, D., Henkin, Y., Westman, E.C., Yancy, W.S., Jr., and Humphreys, M.
Parraga, G., Rudich, A., Fenster, A., Mallett, C., (2006). Dietary treatment of diabetes mellitus in
Liel-Cohen, N., Tirosh, A., et  al. (2010). Dietary the pre-insulin era (1914–1922). Perspect Biol
intervention to reverse carotid atherosclerosis. Med 49, 77–83.
Circulation, 121, 1200–1208. Yamada, Y., Uchida, J., Izumi, H., Tsukamoto, Y., Inoue,
Sondike, S.B., Copperman, N., and Jacobson, M.S. G., Watanabe, Y., Irie, J., and Yamada, S. (2014).
(2003). Effects of a low-carbohdyrate diet on A non-calorie-restricted low-carbohydrate diet is
weight loss and cardiovascular risk factor in over- effective as an alternative therapy for patients with
weight adolescents. J Pediatr 142, 253–258. type 2 diabetes. Intern Med 53, 13–19.
Stein, C.J., and Colditz, G.A. (2004). The epidemic of Yancy Jr., W.S., Vernon, M.C., and Westman, E.C.
obesity. J Clin Endocrinol Metab 89, 2522–2525. (2003). A pilot trial of a low-carbohydrate keto-
Stone, D.B., and Connor, W.E. (1963). The prolonged genic diet in patients with Type 2 diabetes. Metab
effects of a low cholesterol, high carbohydrate Syndr Rel Dis 1, 239–244.
diet upon the serum lipids in diabetic patients. Yancy Jr., W.S., Olsen, M.K., Guyton, J.R., Bakst, R.P.,
Diabetes 12, 127–132. and Westman, E.C. (2004). A low-carbohydrate,
Stumvoll, M., Goldstein, B.J., and Van Heaften T.W. ketogenic diet versus a low-fat diet to treat obe-
(2005). Type 2 diabetes: principles of pathogenesis sity and hyperlipidemia: a randomized, controlled
and therapy. Lancet 365, 1333–1346. trial. Ann Intern Med 140, 769–777.
375

Chapter 37: Ketogenic Diets as Highly Effective Treatments 375

Yancy Jr., W.S., Foy, M., Vernon, M.C., and Westman, and Oddone, E.Z. (2010). A randomized trial of
E.C. (2005). A low-carbohydrate ketogenic diet a low-carbohydrate diet vs orlistat plus a low-
to treat Type 2 Diabetes. Nutr Metab (London) fat diet for weight loss. Arch Intern Med 170,
2, 34. 136–145.
Yancy Jr., W.S., Westman, E.C., McDuffie, J.R., Yudkin, J. (1959). The causes and cure of obesity.
Grambow, S.C., Jeffreys, A.S., Bolton, J., Chalecki, A., Lancet 274, 1135–1138.
376

38
Keto-Adaptation in Health and Fitness
PA R K E R H Y D E , C S C S , C I S S N , V I N C E N T J. M I L L E R , M S ,
AND JEFF S. VOLEK, PHD, RD

INTRODUCTION evidenced by the fact that half the adults in the


Not completely dissimilar from the gravitational United States are prediabetic or diabetic (Menke
potential energy of a ball sitting atop a tower, our et al., 2015).
bodies have an abundance of metabolic potential The parallel increase in dietary carbohydrate
energy (MPE). This MPE can be found in two and the sharp rise in obesity and diabetes in the
primary storage forms, carbohydrate and fats. United States is prima facie evidence that carbo-
Carbohydrates stored predominantly as glycogen hydrate intake has an important effect on health
in skeletal muscle and liver represent approxi- status. The mechanistic links between dietary
mately 2,000 kcals of energy, whereas fat stores in carbohydrate, oversecretion of insulin, and exac-
adipose tissue are at least an order of magnitude erbation of insulin resistance make the over-
greater. Thus, from an evolutionary standpoint, consumption of sugars and starches the most
dietary carbohydrate more aptly represents a obvious driver of the obesity and diabetes epidem-
single matchbook of MPE, better suited for tran- ics. Intuitively, decreasing carbohydrate is an obvi-
sient fight-or-flight responses and not the pri- ous solution. To what extent should carbohydrate
mary fuel source. Since storage is limited, when be decreased? The answer is not simple and varies
carbohydrate is consumed it must be oxidized or from person to person and even within an indi-
converted to something else. The glucose-insulin vidual over time. For many with insulin resistance
axis supports this hierarchy of fuel use, as acute (i.e., carbohydrate intolerance), a modest reduc-
elevations in insulin following consumption of tion in dietary carbohydrate may be all that is
carbohydrate promptly inhibit both adipose tissue necessary to maintain metabolic health. For others
lipolysis and systemic fat oxidation (Bonadonna with a higher degree of insulin resistance, effective
et al., 1990; Jensen et al., 1989). Thus, when energy metabolic correction requires keeping carbohy-
expenditure is high there is a constant need to sup- drates under 40–50 g/day for an extended period
ply a steady source of carbohydrate-based fuel. As of time, resulting in keto-adaptation.
exemplified by the metaphor “hitting the wall,” Keto-adaptation is a process that sustains
when carbohydrate stores are near depletion dur- optimum fuel flow to all organs through use of
ing exercise, the body cannot simply transition to metabolic pathways we have acquired over 2 mil-
using fat efficiently, unless, the person has under- lion years as hunters/gatherers/herders. The most
gone a very low-carbohydrate diet for several important aspect of keto-adaptation is the ability
weeks, which results in a complex set of coordi- of the brain to displace glucose for ketones as the
nated adaptations that dramatically accelerate the primary fuel source. Pioneering work in the 1960s
body’s ability to access and use lipid-based fuels. and 1970s examining starvation ketosis showed
The rest of this chapter discusses the concept that that the brain will switch to preferential use of
humans function more efficiently when they are ketone oxidation (~60%) for derivation of energy
utilizing predominantly fat for fuel. (Cahill et al., 1970; Cahill and Owen, 1968; Owen
Modern diets emphasize carbohydrate-dense et al., 1967). This preferential use of ketone bodies
foods, and therefore bias metabolism toward as a primary fuel source provides both an energy-
carbohydrate oxidation while impairing fat oxi- and neuroprotective effect (Cahill et  al., 1970).
dation. Whereas some individuals can maintain Similar effects are observed in non-starvation-
weight and metabolic health consuming low-fat/ induced ketosis, when carbohydrate is restricted
high-carbohydrate diets, the majority cannot, as and dietary protein and fat are present in the diet.
377

Chapter 38: Keto-Adaptation in Health and Fitness 377

Glucose flux

Fat Oxidation Capacity

Insulin Chronic Disease

Body Composition
Triglycerides Longevity

Oxidative Stress

Fat Utilization Quality of Life


Inflammation

Ketones Physical Resilience


Membrane Integrity

Satiety

Days Weeks/Months Long-term


Time

FIGURE 38.1 Theoretical accumulation of benefit during sustained nutritional ketosis leading to keto-adaptation.

The metabolic pathways responsible for synthesis, have documented acute and short-term responses
transport, uptake, and oxidation of ketones are all of ketogenic diets, the precise temporal pattern of
up-regulated. Within days, blood levels of ketones responses to nutritional ketosis ultimately leading
increase from less than 0.1  mmol/L to 0.5 up to to the keto-adapted phenotype requires additional
3–5  mmol/L. This range is referred to as nutri- work (Figure 38.1).
tional ketosis, and is associated with a positive
effect on a broad range of cardiometabolic risk K E T O - A D A P TAT I O N A N D
factors. It is noteworthy that ketoacidosis does not H E A LT H I M P L I C AT I O N S
occur until ketones are above 10 mmol/L, a level Ketogenic diets have been shown to improve
only reached in individuals with insulin insuf- numerous health outcomes, especially those asso-
ficiency (i.e., poorly controlled type-1 diabetics). ciated with insulin resistance, the underlying com-
Even in a starvation-induced ketogenic state with mon feature in metabolic syndrome (MetS), type
extreme exercise, an individual with a functioning 2 diabetes, cardiovascular disease (CVD), obesity,
pancreas will not experience the increased acidity polycystic ovarian syndrome (PCOS), and many
associated with ketoacidosis. other chronic health conditions.
The near-exclusive reliance on lipid-based fuel
is a fundamental characteristic of keto-adaptation, Ketogenic Diets Improve Insulin
but a new perspective of nutritional ketosis Resistance, Metabolic Syndrome,
has emerged following the identification of β- and Type 2 Diabetes
hydroxybutyrate (βOHB) as a potent epigenetic and A primary application of ketogenic diets in adults
signaling molecule (Newman and Verdin, 2014a). is the management of insulin-resistant conditions.
We now appreciate that keto-adaptation involves Insulin resistance is the primary feature underlying
cellular adaptations that can phenotypically mani- type 2 diabetes, but it also exists across a contin-
fest in different ways between people, probably uum in the general population. It is defined by an
because of genetic and epigenetic effects. The dura- impaired functioning of the insulin response at the
tion of this chapter focuses on mechanisms associ- cellular level, which given the pleiotropic effects of
ated with keto-adaptation as it pertains to health insulin can manifest in many different signs and
outcomes, and explores the emerging potential symptoms. Most commonly, insulin resistance
of well-formulated ketogenic diets to extend the is viewed from the perspective of impaired skel-
physical and cognitive performance of athletes etal muscle glucose uptake and increased hepatic
and military personnel. Although many studies glucose output, manifesting in hyperglycemia.
378

378 section IV: Ketone-Based Metabolism

Thus, individuals with insulin resistance have a spectrum improvements across a range of risk fac-
fundamental problem metabolizing dietary car- tors relative to a traditional low-fat diet in patients
bohydrate and maintaining a normal glucose level. with metabolic syndrome, in effect putting the dis-
Fasting blood glucose values greater than 100 mg/ order into remission (Volek et al., 2009b). A large
dL are indicative of impaired glucose tolerance, number of studies have shown that the features of
and values greater than 7.0 mmol/L (126 mg/dL) metabolic syndrome improve, often in dramatic
on two occasions indicate type 2 diabetes. Since fashion, with adequate carbohydrate restriction
type 2 diabetes is discussed in a separate chapter, (Volek and Feinman, 2005; Volek et al., 2008).
we focus our discussion on the impact of ketogenic Insulin resistance is one of the primary predic-
diets on milder forms of insulin resistance that tors of nonalcoholic fatty liver disease (NAFLD),
manifest in the cluster of disturbances that encom- which is characterized by increased liver fat,
pass metabolic syndrome. inflammation, and resultant hypertriglyceride-
Metabolic syndrome is best described as pre- mia, all of which increase CVD risk. Although
diabetes and provides an early sign that the body the pathophysiology and mechanisms of NAFLD
is mismanaging dietary carbohydrate. One aspect are yet to be fully elucidated, there is a clear link
of this mismanagement is that a greater proportion to increased carbohydrate consumption, de novo
of incoming carbohydrate is converted to fat in the lipogenesis, and increased adiposity. Insulin resis-
liver (Petersen et  al., 2007). This greater conver- tance in the peripheral tissues inhibits the cellu-
sion of dietary carbohydrate into fat (i.e., de novo lar uptake of glucose, thus a shunting of dietary
lipogenesis), much of it entering the circulation carbohydrate toward the liver occurs. Glucose
as saturated fat, is a metabolic abnormality that flux into hepatic tissue is not insulin dependent,
causes a significant amount of collateral damage in thus an increased rate of glycogen synthesis or de
the body. For all these reasons, insulin resistance is novo lipogenesis, during the postprandial state, is
a problem that effectively manifests itself as carbo- experienced. Hepatic insulin resistance is associ-
hydrate intolerance. Like other food intolerances, ated with increased concentration of malonyl-
the most logical and effective approach to man- CoA; inhibition of carnitine palmitoyl transferase
aging carbohydrate intolerance is to restrict the 1 (CPT-1), a transporter for fatty acids into the
offending nutrient. When dietary carbohydrate is mitochondria; inhibition of beta-oxidation; and
restricted to a level below which it is not signifi- increased accumulation of triglycerides, cho-
cantly converted to fat (a threshold that varies from lesterol esters, and very low density lipoprotein
person to person), signs and symptoms of insulin (Browning et  al., 2011; Zivkovic et  al., 2007).
resistance improve or often disappear completely. Since keto-adaptation results in significantly
Based on our research, a ketogenic diet can resolve decreased de novo lipogenesis and accelerated
all the signs and symptoms of metabolic syndrome beta-oxidation, as well as decreased inflammation
(Volek and Feinman, 2005; Volek et al., 2008). and serum triglycerides, it would seem obvious to
The main features of metabolic syndrome are also have beneficial effects on NAFLD. However,
abdominal obesity; high blood levels of glucose, only a few studies have examined effects on liver
insulin, and triglycerides; low HDL-cholesterol; fat. Early during keto-adaptation, approximately
and high blood pressure. Other features not typi- 2 weeks, there is significantly decreased hepatic
cally included in the diagnosis of metabolic syn- triglyceride content, as compared to a higher car-
drome include elevated constitutive inflammation, bohydrate diet (Browning et al., 2011). Six months
vascular dysfunction, and disturbances in fatty acid of a ketogenic diet has been demonstrated to
composition. It is commonly stated that obesity is improve steatosis, necroinflammatory grade, and
the cause of metabolic syndrome, but this perspec- centrilobular fibrosis, all of which are histological
tive fails to acknowledge the cause of obesity, which markers of NAFLD (Tendler et al., 2007).
for many involves the overconsumption of dietary
carbohydrate relative to a person’s tolerance. Thus, Weight Loss and Body
the primary driver of metabolic syndrome is over- Composition Responses
consumption of sugars and starches relative to the Dozens of studies have examined weight-loss
level a person can metabolize without resorting to responses to low-carbohydrate and low-fat diets
increased de novo lipogenesis and other manifes- over the last 15  years. Although there is great
tations of carbohydrate intolerance (e.g., increased variability in the comparison of diets of any dura-
oxidative stress). Although the molecular details of tion, it is noteworthy that low-carbohydrate diets
insulin resistance have not been fully elucidated, do at least as well and usually better than low-fat
a well-formulated ketogenic diet results in broad diets according to recent meta-analyses (Ajala
379

Chapter 38: Keto-Adaptation in Health and Fitness 379

et  al., 2013; Bueno et  al., 2013). In fact a recent nitrogen excretion (Sherwin et al., 1975) and leu-
meta-analysis, which used Bayesian hierarchi- cine oxidation (Nair et  al., 1988)  and increases
cal modeling to provide an estimate of the likeli- muscle protein synthesis (Nair et  al., 1988). As
hood of achieving a desired degree of weight loss, such, nutritional ketosis may be an important
reported that the probability of greater weight loss determinant of lean mass preservation during pro-
associated with a low-carbohydrate diet was >99% longed periods of energy deficit induced by caloric
(Sackner-Bernstein et  al., 2015). It is noteworthy restriction and/or high energy expenditure.
that the degree of insulin resistance has an effect Preservation of lean mass can further be sup-
on expected weight loss. In a post hoc analysis of ported through resistance training. In fact, our
the A to Z study, it was reported that individuals research has shown 12 weeks of resistance training
who were insulin sensitive had similar degrees of to result in increases in lean body mass, along with
weight loss on a low- and high-carbohydrate diet greater fat loss, during a ketogenic diet versus a
after 1  year, but the those with higher levels of low-fat diet (Quann et al., 2007; Volek et al., 2010).
insulin resistance lost significantly more weight on Similar results have been observed in response to a
a low-carbohydrate diet (McClain et al., 2013). ketogenic diet versus a typical Western diet (Rauch
Several weight-loss studies have also mea- et al., 2014).
sured changes in fat and lean body mass. Similar While the metabolic adaptations that occur
to changes in total body mass, low-carbohydrate during carbohydrate restriction clearly have an
diets have been shown to result in a greater loss of important role in weight loss, satiety is an impor-
fat mass (Krieger et al., 2006). Outcomes for lean tant factor as well. In weight-loss trials comparing
mass, however, are less clear due to variations in low-carbohydrate to low-fat diets, energy deficit
diet and limitations in diet formulation and assess- for the low-carbohydrate diet is often achieved
ment methods. Over 30  years ago Phinney et  al. without intentional restriction, indicating greater
(1983) reported that nitrogen balance was positive satiety. Furthermore, very low calorie diets, which
after the first week of a eucaloric ketogenic diet. are often ketogenic due to the inherently low
An important factor not always appreciated in level of carbohydrates and protein, are known
studies of ketogenic diets is the need to adjust elec- to reduce appetite. This is supported by a recent
trolyte intake to offset the natriuretic effect of car- meta-analysis in which both ketogenic and very-
bohydrate restriction. Unfortunately, the majority low calorie diets were considered (Gibson et  al.,
of studies conducted since then have not consid- 2015). The similarities in satiety between these two
ered this natriuretic effect. However, our research diets, despite the considerable differences in pro-
has shown it is possible not only to preserve lean tein, fat, and energy content, suggests that ketosis
mass during weight loss with a ketogenic diet but may suppress appetite. This is supported by stud-
also to increase it (Volek et al., 2002). An impor- ies showing a eucaloric or ad libitum ketogenic
tant consideration is the water retention associ- diet to be more satiating than nonketogenic diets
ated with high-carbohydrate diets and glycogen with higher protein content (Johnstone et al., 2008;
storage (Olsson and Saltin, 1970), and hence the Veldhorst et al., 2010).
water loss that accompanies glycogen loss during
carbohydrate restriction. Readings from dual- Improved Lipoprotein Profile
energy X-ray absorptiometry DXA (Going et  al., Although higher probability of weight loss is a
1993) and biological impedance (Koulmann et al., major benefit of a ketogenic diet, there are also pro-
2000), which are two commonly used methods of found changes in lipoprotein metabolism (Volek
assessing body composition, fluctuate in accor- et  al., 2008). The reduced conversion of carbohy-
dance with body water content. Furthermore, drate to fat in the liver (lipogenesis) plus the low
these methods generally include total body water insulin state enabling accelerated fat oxidation are
as a component of lean mass. Therefore, the water major factors contributing to the improvements
loss that typically occurs during the initiation of in processing of lipoproteins commonly observed.
carbohydrate restriction can give a false indication The most consistent response is a sharp decrease
of functional muscle loss. in plasma triglycerides, most dramatically in those
As previously described, nutritional ketosis with preexisting hypertriglyceridemia. A ketogenic
is characterized by an elevation in blood ketones diet also demonstrates a striking reduction in the
and, depending on the extent of carbohydrate postprandial lipemic response to a high-fat meal
restriction, is not always synonymous with a low- (Volek et  al., 2000; Volek et  al., 2009b) and has
carbohydrate diet. Infusion of βOHB, the primary positive effects on postabsorptive and postpran-
ketone elevated during nutrition ketosis, reduces dial vascular function (Volek et al., 2009a). Almost
380

380 section IV: Ketone-Based Metabolism

as consistent as their triglyceride-lowering effects, of the alternative paradigm that deemphasizes


ketogenic diets increase HDL-C. The increase in carbohydrate. For example, there are few placebo-
HDL-C may not occur as quickly as the decrease controlled randomized control trials, few studies
in triglycerides, but there is some evidence that it of very low carbohydrate intake, and even fewer
develops more slowly over time (Shai et al., 2008). studies that allowed adequate time to adapt to a
The response in LDL-C concentration to a keto- low-carbohydrate diet. Carbohydrate loading has
genic diet is highly variable, with many people not shown improved performance in all studies,
showing a decrease and many an increase, with a despite significantly increased muscle glycogen
subset (~15%) showing a marked rise. While there levels (Burke et  al., 2000). The most obvious fact
is variability in the LDL-C concentration response, that has been downplayed in prior research has
there are more uniform patterns in qualitative fea- been the profound effect of carbohydrate intake
tures of the LDL particle. Dietary carbohydrate on inhibition of fat utilization, and the perspective
restriction consistently decreases small, dense LDL that fat is a preferred fuel.
particles, which are more atherogenic due to their
longer residence time in the circulation and greater Fat as a Premium Fuel
propensity for oxidation, whereas low-fat/high- Conventional wisdom has been that carbohydrate
carbohydrate diets have the opposite effect. This is the preferred source of fuel for exercise, but this
inverse relationship between dietary carbohydrate perspective is rapidly changing considering the
content and LDL particle size is evident over a wide many benefits associated with emphasizing fatty
range of carbohydrate intakes (Krauss, 2005), and acids and ketones as primary fuels, and relegat-
it can have quite dramatic (i.e., positive) effects in ing blood sugar and glycogen to secondary status.
response to a ketogenic diet (Volek et al., 2009b). Ten hypothetical reasons a high capacity to utilize
lipid-based fuels might be beneficial for athletes
K E T O - A D A P TAT I O N are listed here:
IN SPORT AND FITNES S
Beyond the clinical applications of keto-adaptation 1. Fat is stored in adipose tissue in greater
for promoting weight loss and managing insulin quantities than carbohydrates by at least an
resistant conditions, there is a growing interest in order of magnitude (>20,000 kcal in even the
ketogenic diets by athletes and active individu- thinnest athlete vs. 2,000 kcal as glycogen).
als interested in better health, performance, and A key element contributing to deteriorating
recovery. cognitive and physical performance during
physically demanding exercise is reduced
Brief History carbohydrate availability coupled with an
More than 70 years ago (Christensen and Hansen, inability to effectively utilize the thousands
1939) described that consumption of a high-car- of calories stored as fat. Paradoxically,
bohydrate diet, compared to a high-fat diet, for 1 deteriorating performance associated with
week prior to a submaximal event was associated glycogen depletion occurs in the presence of
with enhanced performance. Nearly 3 decades an abundance of fuel stored as body fat that
later, Bergstrom and Hultman (1966, 1967) dis- the athlete cannot access efficiently, unless
covered that muscle glycogen depletion was asso- they have previously keto-adapted for
ciated with fatigue, and that a high-carbohydrate several weeks.
diet maintained muscle glycogen and perfor- 2. Fat is a more efficient source of fuel than
mance. This set the stage for the next 40+ years, carbohydrates. A molecule of palmitic
during which time the supremacy of carbohydrate acid has over twice as many carbons as a
has become deeply embedded in the minds of most molecule of glucose, and over twice the
scientists, coaches, and athletes (Hargreaves et al. energy per unit weight (i.e., 9 vs. 4 kcal/g).
2004). There has been a strong confirmation bias Fat is also more efficient, since it is not
to support high-carbohydrate diets, reinforced by stored along with a lot of extra water
a burgeoning sports beverage industry profiting weight like glycogen. Finally, ketones have
from this paradigm. been shown to have greater efficiency
It needs to be acknowledged that a lot of great in providing cellular energy and work
work has been done by many researchers detail- output (i.e., more work per unit oxygen
ing how carbohydrate affects exercise metab- consumed) (Sato et al., 1995).
olism and performance, but there has been a 3. Relying on body fat for energy significantly
surprising lack of recognition, or willful neglect, decreases the need to fuel during exercise.
381

Chapter 38: Keto-Adaptation in Health and Fitness 381

This has physiologic benefits as it avoids cause of aging and inflammation, and the
the need to digest and absorb nutrients at reduction by βOHB of these processes was
a time when the majority of blood flow is remarkable.
diverted to active skeletal muscles. It also 9. One of the most common perceived
alleviates the gastrointestinal problems benefits following keto-adaptation
associated with fueling during exercise that is enhanced recovery from exercise.
are common among endurance athletes. This is frequently demonstrated by
4. When oxidized inside the mitochondria, an expedited return to training and
ketones generate less reactive oxygen competition following physically
species relative to glucose or other fuels, demanding events and long training
in essence making them a cleaner burning sessions. This area of research has
fuel (Maalouf et al., 2007; Veech, 2004). not been investigated thoroughly, but
5. Deriving greater amounts of energy from we speculate that less oxidative stress
fat at higher exercise intensities translates and the anti-inflammatory effects of
into less reliance on limited glycogen a ketogenic diet may contribute to the
stores, glycolysis, and generation of perceived improvement in recovery by
hydrogen ions. many athletes. Carbohydrate ingestion
6. Making fat the primary fuel often results and acute hyperglycemia activate a
in greater ease of fat loss and improved host of inflammatory and free radical-
body composition, which translates generating pathways (Aljada et al.,
into improved efficiency and power-to- 2006; Dhindsa et al., 2004). Some of
weight ratio. Hypothetically this would these include: stimulation of NAPDH
be advantageous for endurance as well as oxidase, superoxide generation by
many strength/power athletes. leukocytes, TNF-α production, and
7. When ketone concentrations increase activation of NF-κB which regulates the
to the 1–5 mmol/L range, the brain will transcriptional activity of over 100 pro-
begin to preferentially oxidize ketones inflammatory genes. Consistent with the
(Cahill et al., 1970; Owen et al., 1967). In notion that carbohydrate can aggravate
a non-keto-adapted athlete, continuous inflammatory balance, many studies
exercise often results in profound mental have reported that low carbohydrate
confusion (i.e., “hitting the wall” or diets decrease markers of constitutive
“bonking”). A brain adapted to extracting inflammation in athletes (Phinney
the majority of its energy requirements et al., 1983) and non-athletes (Youm
from ketones is resistant to this energy et al., 2015).
crisis, since ketones are derived from 10. A low carbohydrate intake enhances the
fatty acids and actually increase in effects of exercise on several positive cel-
concentration as exercise duration lular adaptations associated with health
increases. A keto-adapted brain is able to (Hawley and Burke, 2010). For example,
tolerate extraordinarily low levels of blood in the post-exercise period consumption
sugar without any signs or symptoms of even small amounts of carbohydrate
hypoglycemia (Cahill and Aoki, 1980; rapidly decreases the release of fatty acids
Drenick et al., 1972). from fat stores and oxidation of fat in the
8. Independent of its role as a vital fuel muscle (Long et al., 2008). In some ath-
source for the brain and other organs/ letes, a surge in insulin may be followed
tissues when carbohydrates are limited, by a low blood sugar eliciting a stress
βOHB has recently been shown to response characterized by a counter-reg-
directly signal cellular processes, acting ulatory hormonal response that can man-
more like a hormone (Newman and ifest as carbohydrate cravings, lethargy,
Verdin, 2014a, 2014b). Specifically, it poor physical/mental performance and
was demonstrated that at blood levels suboptimal recovery. Carbohydrate con-
characteristic of nutritional ketosis, βOHB sumption during recovery has also been
acts as a class I histone deactylase inhibitor, shown to diminish the beneficial effects of
switching on specific genes that protect exercise on insulin sensitivity and other
cells from free radical damage (Shimazu cardio-metabolic risk markers (Holtz
et al., 2013). Oxidative stress is a root et al., 2008; Stephens and Braun, 2008).
382

382 section IV: Ketone-Based Metabolism

T H E K E TO - A DA P T E D highest values reported in the literature (with the


AT H L E T E notable exception of the cyclists in Phinney et al.,
Although a critical mass of experimental data on 1983) are 50% lower (i.e., ~1.0 g/min). Fuel use
ketogenic diets and ketone physiology has been during the 3-hour run was also dramatically dif-
generated in the last decade, demonstrating safety ferent. The high-carbohydrate group used an even
and therapeutic efficacy in managing a range of mix of fuel, whereas nearly 90% of energy expend-
clinical conditions, there has been little effort iture was derived from fat in the low-carb athletes.
focused on studying how ketogenic diets affect This translated into a two-fold greater use of fat
performance and recovery. The two notable stud- (~1.2 vs 0.6 g/min) during exercise. Figure 38.2
ies that shed light on the metabolic phenotype shows how this enhanced reliance on fat oxida-
of the keto-adapted athlete come from Phinney tion could enable endurance athletes to compete
in 1983 (Phinney et al., 1983) and Volek in 2016 without need for exogenous fuel. Serum glycerol
(Volek et al., 2016). concentrations, a marker of adipose tissue lipoly-
Phinney first demonstrated in a small (n=5) but sis (fat breakdown), were two-fold higher during
highly controlled metabolic ward study that 4 weeks exercise and recovery in low-carbohydrate run-
of adaptation to a ketogenic diet in high-level cyclists ners, further supporting the dramatically acceler-
resulted in profound metabolic adaptations and res- ated rates of fat metabolism in the keto-adapted
toration of endurance performance following an runners (Figure 38.2).
initial decline. These cyclists showed a remarkable Despite the extraordinary transformation
near exclusive reliance on fat as the sole fuel at 64% to extract the majority of energy from fat dur-
maximal oxygen consumption, while significantly ing submaximal exercise in the low-carbohydrate
decreasing glycogen breakdown and glucose oxida- athletes, they showed a surprisingly high level
tion. For more than 3 decades these findings have of muscle glycogen at rest, glycogen breakdown
been largely ignored but never refuted. during exercise, and synthesis of glycogen dur-
We designed the FASTER (Fat Adapted ing recovery. This result was different from that
Substrate Use in Trained Elite Runners) study observed in keto-adapted cyclist after 4 weeks,
to validate and extend Phinney’s work by docu- suggesting that complete adaptations in glycogen
menting the keto-adapted phenotype in elite ath- metabolism may occur over a longer period than
letes after long-term keto-adaptation (Volek et al. 1  month. Although speculative, we hypothesize
2016). The primary goal was to characterize the there is a need to provide a source of oxaloacetate
metabolic differences between high-caliber ultra- for optimal Krebs cycle functioning in the keto-
endurance runners habitually consuming a very adapted state, and breakdown of glycogen, without
low-carbohydrate ketogenic diet versus a more terminal oxidation, provides a source of carbons
traditional high-carbohydrate diet. Ten low-carb for that purpose.
and 10 high- carb high-caliber ultra-runners were
recruited and studied. They were carefully matched S U M M A RY
on age, body mass, maximum aerobic capacity, The long-standing dogma in sports nutrition
and competition times; the main difference was has been that athletes need to consume a high-
their habitual diet. The high-carbohydrate group carbohydrate diet in order to excel physically and
consumed over half their intake as carbohydrate, cognitively. This approach may work for some
whereas the low-carbohydrate athletes consumed athletes, but for many it is associated with less
~10% of their energy intake from carbohydrate than desirable health and performance outcomes.
for an average of 20  months. Athletes performed Ketogenic diets represent an alternative approach
a maximal oxygen consumption test during which with a rapidly growing level of scientific and obser-
peak fat oxidation was determined, and on a sepa- vational support. Although it is clear that keto-
rate day they ran on a treadmill at 64% of maximal genic diets are an effective weight-loss intervention
oxygen consumption for 3 hours. often leading to greater fat loss than low-fat diets,
The most remarkable finding was an extraor- it remains to be determined whether the benefit is
dinary ability to oxidize fat in low-carbohydrate exclusively a result of carbohydrate restriction or
athletes, which was on average two-fold higher whether nutritional ketosis provides added benefit.
(1.5 vs. 0.7 g/min, respectively). Even the low- Given the new perspective of βOHB as more than
est “fat burner” in the low-carbohydrate group a metabolite, there are now more reasons to spec-
exceeded the highest “fat burner” in the high- ulate why a well-formulated ketogenic diet would
carbohydrate group. These rates of fat oxidation be associated with more robust health and perfor-
are even more remarkable considering that the mance effects. In regard to metabolic syndrome,
383

Chapter 38: Keto-Adaptation in Health and Fitness 383

120
Phinney et al., 1983
110

100

Fat oxidation @ 64% VO2 max (g/h)


90 Volek et al., 2016

80

70

60

50

40
Phinney et al., 1983
30
Volek et al., 2016
20
High-Carb (non- Keto-Adapted
keto-adapted) Athletes
Athletes

FIGURE 38.2 Rate of fat oxidation during submaximal exercise in non-keto-adapted (left) vs. keto-adapted (right)
endurance athletes. The dotted line represents the estimated rate of fat oxidation required to run the marathon leg of an
Ironman triathlon in 2 hr 40 min with minimal glycogen contribution and no exogenous fuel (Volek et al., 2015). Circles
= individual data points; Xs = mean responses.

ketogenic diets are more likely than low-fat diets approaches to the management of type 2 diabetes.
to effect global improvement in the diverse signs Am J Clin Nutr 97, 505–516.
and symptoms of this complex disease highly asso- Aljada, A., Friedman, J., Ghanim, H., Mohanty, P.,
ciated with type 2 diabetes. Moreover, treating any Hofmeyer, D., Chaudhuri, A., and Dandona, P.
of the individual metabolic syndrome markers (2006). Glucose ingestion induces an increase in
intranuclear nuclear factor kappaB, a fall in cel-
with carbohydrate restriction holds the promise to
lular inhibitor kappaB, and an increase in tumor
benefit the others. Therefore, a ketogenic diet is the
necrosis factor alpha messenger RNA by mononu-
preferred primary intervention. clear cells in healthy human subjects. Metabolism
In regard to physical activity, ketogenic diets 55, 1177–1185.
dramatically transform energy metabolism during Bergstrom, J., and Hultman, E. (1966). Muscle glyco-
exercise and may also facilitate recovery through gen synthesis after exercise:  an enhancing factor
modulation of inflammation, free radical gen- localized to the muscle cells in man. Nature 210,
eration, and antioxidant defense mechanisms. 309–310.
However, as with weight loss, it is not clear whether Bergstrom, J., and Hultman, E. (1967). A study of the
nutritional ketosis provides added benefit beyond glycogen metabolism during exercise in man.
carbohydrate restriction without ketosis. There are Scand J Clin Lab Invest 19, 218–228.
many health benefits associated with nutritional Bonadonna, R.C., Groop, L.C., Zych, K., Shank, M.,
ketosis that are relevant for athletes. In regard to and DeFronzo, R.A. (1990). Dose-dependent effect
exercise performance, field testing indicates a high of insulin on plasma free fatty acid turnover and
oxidation in humans. Am J Physiol 259, E736–750.
level of success among many endurance athletes,
Browning, J.D., Baker, J.A., Rogers, T., Davis, J.,
but there has been very little laboratory testing Satapati, S., and Burgess, S.C. (2011). Short-
focused on performance outcomes. Hopefully term weight loss and hepatic triglyceride reduc-
that will change as we dawn on a new era of sports tion:  evidence of a metabolic advantage with
nutrition that looks beyond the high-carbohydrate dietary carbohydrate restriction. Am J Clin Nutr
paradigm to alternative approaches that empha- 93, 1048–1052.
size a fat-based metabolism. Bueno, N.B., de Melo, I.S., de Oliveira, S.L., and da
Rocha Ataide, T. (2013). Very-low-carbohydrate
REFERENCES ketogenic diet v. low-fat diet for long-term weight
Ajala, O., English, P., and Pinkney, J. (2013). Systematic loss: a meta-analysis of randomised controlled tri-
review and meta-analysis of different dietary als. Br J Nutr 110, 1178–1187.
384

384 section IV: Ketone-Based Metabolism

Burke, L.M., Hawley, J.A., Schabort, E.J., St Clair Koulmann, N., Jimenez, C., Regal, D., Bolliet, P.,
Gibson, A., Mujika, I., and Noakes, T.D. (2000). Launay, J.C., Savourey, G., and Melin, B. (2000).
Carbohydrate loading failed to improve 100-km Use of bioelectrical impedance analysis to estimate
cycling performance in a placebo- controlled trial. body fluid compartments after acute variations of
J Appl Physiol (1985) 88, 1284–1290. the body hydration level. Med Sci Sports Exerc 32,
Cahill, G., Jr., Felig, P., Owen, O., and Wahren, J. 857–864.
(1970). Metabolic adaptation to prolonged starva- Krauss, R.M. (2005). Dietary and genetic probes of
tion in man. Nord Med 83, 89. atherogenic dyslipidemia. Arterioscler Thromb
Cahill, G.F., Jr., and Owen, O.E. (1968). Starvation and Vasc Biol 25, 2265–2272.
survival. Trans Am Clin Climatol Assoc 79, 13–20. Krieger, J.W., Sitren, H.S., Daniels, M.J., and
Christensen, E.H., and Hansen, O. (1939). Langkamp-Henken, B. (2006). Effects of varia-
Untersuchungen iber die verbrennungsvorgange tion in protein and carbohydrate intake on body
bei langdavernder, schwerer Muskelarbeit. Skand mass and composition during energy restriction: a
Arch Physiol 81. meta-regression 1. Am J Clin Nutr 83, 260–274.
Dhindsa, S., Tripathy, D., Mohanty, P., Ghanim, H., Long, W., 3rd, Wells, K., Englert, V., Schmidt, S.,
Syed, T., Aljada, A., and Dandona, P. (2004). Hickey, M.S., and Melby, C.L. (2008). Does prior
Differential effects of glucose and alcohol on reac- acute exercise affect postexercise substrate oxida-
tive oxygen species generation and intranuclear tion in response to a high carbohydrate meal? Nutr
nuclear factor-kappaB in mononuclear cells. Metab (Lond) 5, 2.
Metabolism 53, 330–334. Maalouf, M., Sullivan, P.G., Davis, L., Kim, D.Y., and
Drenick, E.J., Alvarez, L.C., Tamasi, G.C., and Rho, J.M. (2007). Ketones inhibit mitochondrial
Brickman, A.S. (1972). Resistance to symptom- production of reactive oxygen species production
atic insulin reactions after fasting. J Clin Invest 51, following glutamate excitotoxicity by increasing
2757–2762. NADH oxidation. Neuroscience 145, 256–264.
Forsythe, C.E., Phinney, S.D., Fernandez, M.L., Quann, McClain, A.D., Otten, J.J., Hekler, E.B., and Gardner,
E.E., Wood, R.J., Bibus, D.M., Kraemer, W.J., C.D. (2013). Adherence to a low-fat vs. low-
Feinman, R.D., and Volek, J.S. (2008). Comparison carbohydrate diet differs by insulin resistance sta-
of low fat and low carbohydrate diets on circulat- tus. Diabetes Obes Metab 15, 87–90.
ing fatty acid composition and markers of inflam- Menke, A., Casagrande, S., Geiss, L., and Cowie, C.C.
mation. Lipids 43, 65–77. (2015). Prevalence of and trends in diabetes among
Gibson, A.A., Seimon, R.V., Lee, C.M., Ayre, J., adults in the United States, 1988–2012. JAMA 314,
Franklin, J., Markovic, T.P., Caterson, I.D., and 1021–1029.
Sainsbury, A. (2015). Do ketogenic diets really Nair, K.S., Welle, S.L., Halliday, D., and Campbell, R.G.
suppress appetite? A systematic review and meta- (1988). Effect of beta-hydroxybutyrate on whole-
analysis. Obes Rev 16, 64–76. body leucine kinetics and fractional mixed skeletal
Going, S.B., Massett, M.P., Hall, M.C., Bare, L.A., Root, muscle protein synthesis in humans. J Clin Invest
P.A., Williams, D.P., and Lohman, T.G. (1993). 82, 198–205.
Detection of small changes in body composition Newman, J.C., and Verdin, E. (2014a). Beta-
by dual-energy x-ray absorptiometry. Am J Clin hydroxybutyrate:  much more than a metabolite.
Nutr 57, 845–850. Diabetes Res Clin Pract 106, 173–181.
Hargreaves M, Hawley JA, Jeukendrup A. Pre-exercise Newman, J.C., and Verdin, E. (2014b). Ketone bodies
carbohydrate and fat ingestion: effects on metabo- as signaling metabolites. Trends Endocrinol Metab
lism and performance. J Sports Sci. 2004 25, 42–52.
Hawley, J.A., and Burke, L.M. (2010). Carbohydrate Olsson, K.E., and Saltin, B. (1970). Variation in total
availability and training adaptation: effects on cell body water with muscle glycogen changes in man.
metabolism. Exerc Sport Sci Rev 38, 152–160. Acta Physiol Scand 80, 11–18.
Holtz, K.A., Stephens, B.R., Sharoff, C.G., Chipkin, Owen, O.E., Morgan, A.P., Kemp, H.G., Sullivan,
S.R., and Braun, B. (2008). The effect of carbo- J.M., Herrera, M.G., and Cahill, G.F., Jr. (1967).
hydrate availability following exercise on whole- Brain metabolism during fasting. J Clin Invest 46,
body insulin action. Appl Physiol Nutr Metab 33, 1589–1595.
946–956. Petersen, K.F., Dufour, S., Savage, D.B., Bilz, S.,
Jensen, M.D., Caruso, M., Heiling, V., and Miles, J.M. Solomon, G., Yonemitsu, S., Cline, G.W., Befroy,
(1989). Insulin regulation of lipolysis in nondia- D., Zemany, L., Kahn, B.B., et al. (2007). The role
betic and IDDM subjects. Diabetes 38, 1595–1601. of skeletal muscle insulin resistance in the patho-
Johnstone, A.M., Horgan, G.W., Murison, S.D., genesis of the metabolic syndrome. Proc Natl Acad
Bremner, D.M., and Lobley, G.E. (2008). Effects of Sci USA 104, 12587–12594.
a high-protein ketogenic diet on hunger, appetite, Phinney, S.D., Bistrian, B.R., Wolfe, R.R., and
and weight loss in obese men feeding ad libitum. Blackburn, G.L. (1983). The human metabolic
Am J Clin Nutr 87, 44–55. response to chronic ketosis without caloric
385

Chapter 38: Keto-Adaptation in Health and Fitness 385

restriction:  physical and biochemical adaptation. but not energy expenditure in normal-weight
Metabolism 32, 757–768. human subjects fed in energy balance. Br J Nutr
Quann, E.E., Ballard, K.D., Puglisi, M.J., Forsythe, C.E., 104, 1395–1405.
Volk, B.M., Silvestre, R., and Volek, J.S. (2007). Volek, J.S., Gomez, A.L., and Kraemer, W.J. (2000).
Carbohydrate restriction and resistance training Fasting lipoprotein and postprandial triacylglyc-
have additive effects on body composition during erol responses to a low-carbohydrate diet supple-
weight loss in men. J Am Diet Assoc 107, A14. mented with n-3 fatty acids. J Am Coll Nutr 19,
Rauch, J.T., Silva, J.E., Lowery, R.P., McCleary, S.A., 383–391.
Shields, K.A., Ormes, J.A., and Wilson, J.M. Volek, J.S., Sharman, M.J., Love, D.M., Avery, N.G.,
(2014). The effects of ketogenic dieting on skeletal Gomez, A.L., Scheett, T.P., and Kraemer, W.J.
muscle and fat mass. J Int Soc Sports Nutr 11, 1. (2002). Body composition and hormonal responses
Sackner-Bernstein, J., Kanter, D., and Kaul, S. (2015). to a carbohydrate-restricted diet. Metabolism 51,
Dietary intervention for overweight and obese 864–870.
adults: comparison of low-carbohydrate and low- Volek, J.S., and Feinman, R.D. (2005). Carbohydrate
fat diets: a meta-analysis. PLoS One 10, e0139817. restriction improves the features of metabolic syn-
Sato, K., Kashiwaya, Y., Keon, C.A., Tsuchiya, N., King, drome: metabolic syndrome may be defined by the
M.T., Radda, G.K., Chance, B., Clarke, K., and response to carbohydrate restriction. Nutr Metab
Veech, R.L. (1995). Insulin, ketone bodies, and (Lond) 2, 31.
mitochondrial energy transduction. FASEB J 9, Volek, J.S., Fernandez, M.L., Feinman, R.D., and
651–658. Phinney, S.D. (2008). Dietary carbohydrate
Seshadri, P., Iqbal, N., Stern, L., Williams, M., Chicano, restriction induces a unique metabolic state posi-
K.L., Daily, D.A., McGrory, J., Gracely, E.J., Rader, tively affecting atherogenic dyslipidemia, fatty acid
D.J., and Samaha, F.F. (2004). A randomized study partitioning, and metabolic syndrome. Prog Lipid
comparing the effects of a low-carbohydrate diet Res 47, 307–318.
and a conventional diet on lipoprotein subfrac- Volek, J.S., Ballard, K.D., Silvestre, R., Judelson, D.A.,
tions and C-reactive protein levels in patients with Quann, E.E., Forsythe, C.E., Fernandez, M.L.,
severe obesity. Am J Med 117, 398–405. and Kraemer, W.J. (2009a). Effects of dietary car-
Shai, I., Schwarzfuchs, D., Henkin, Y., Shahar, D.R., bohydrate restriction versus low-fat diet on flow-
Witkow, S., Greenberg, I., Golan, R., Fraser, D., mediated dilation. Metabolism 58, 1769–1777.
Bolotin, A., Vardi, H., et  al. (2008). Weight loss Volek, J.S., Phinney, S.D., Forsythe, C.E., Quann,
with a low-carbohydrate, Mediterranean, or low- E.E., Wood, R.J., Puglisi, M.J., Kraemer, W.J.,
fat diet. N Engl J Med 359, 229–241. Bibus, D.M., Fernandez, M.L., and Feinman, R.D.
Sherwin, R.S., Hendler, R.G., and Felig, P. (1975). Effect (2009b). Carbohydrate restriction has a more
of ketone infusions on amino acid and nitrogen favorable impact on the metabolic syndrome than
metabolism in man. J Clin Invest 55, 1382–1390. a low fat diet. Lipids 44, 297–309.
Shimazu, T., Hirschey, M.D., Newman, J., He, W., Volek, J.S., Quann, E.E., and Forsythe, C.E. (2010).
Shirakawa, K., Le Moan, N., Grueter, C.A., Lim, Low-carbohydrate diets promote a more favor-
H., Saunders, L.R., Stevens, R.D., et  al. (2013). able body composition than low-fat diets. Strength
Suppression of oxidative stress by beta- hydroxy- Cond Journal 32, 6.
butyrate, an endogenous histone deacetylase Volek, J.S., Freidenreich, D.J., Saenz, C., Kunces, L.J.,
inhibitor. Science 339, 211–214. Creighton, B.C., Bartley, J.M., Davitt, P.M., Munoz,
Stephens, B.R., and Braun, B. (2008). Impact of nutri- C.X., Anderson, J.M., Maresh, C.M., Lee, E.C.,
ent intake timing on the metabolic response to Schuenke, M.D., Aerni, G., Kraemer, W.J., Phinney,
exercise. Nutr Rev 66, 473–476. S.D. (2016). Metabolic characteristics of keto-
Tendler, D., Lin, S., Yancy, W.S., Jr., Mavropoulos, J., adapted ultra-endurance runners. Metabolism.
Sylvestre, P., Rockey, D.C., and Westman, E.C. Mar;65(3):100–10.
(2007). The effect of a low-carbohydrate, ketogenic Volek, J.S., Noakes, T., and Phinney, S.D. (2015).
diet on nonalcoholic fatty liver disease:  a pilot Rethinking fat as a fuel for endurance exercise. Eur
study. Dig Dis Sci 52, 589–593. J Sport Sci 15, 13–20.
Veech, R.L. (2004). The therapeutic implications of Youm, Y.H., Nguyen, K.Y., Grant, R.W., Goldberg, E.L.,
ketone bodies:  the effects of ketone bodies in Bodogai, M., Kim, D., D’Agostino, D., Planavsky,
pathological conditions:  ketosis, ketogenic diet, N., Lupfer, C., Kanneganti, T.D., et al. (2015). The
redox states, insulin resistance, and mitochondrial ketone metabolite beta-hydroxybutyrate blocks
metabolism. Prostaglandins Leukot Essent Fatty NLRP3 inflammasome-mediated inflammatory
Acids 70, 309–319. disease. Nat Med 21, 263–269.
Veldhorst, M.A., Westerterp, K.R., van Vught, A.J., and Zivkovic, A.M., German, J.B., and Sanyal, A.J. (2007).
Westerterp-Plantenga, M.S. (2010). Presence or Comparative review of diets for the metabolic syn-
absence of carbohydrates and the proportion of fat drome:  implications for nonalcoholic fatty liver
in a high-protein diet affect appetite suppression disease. Am J Clin Nutr 86, 285–300.
386

39
Advancing the Awareness and Application
of Ketogenic Therapies Globally
The Charlie Foundation and Matthew’s Friends

BETH ZUPEC-KANIA, RD, CD, JIM ABRAHAMS,


EMMA WILLIAMS, MBE, AND SUSAN A. MASINO, PHD

OV E RV I E W therapies including neurological and neurodegen-


The Charlie Foundation was born out of the erative disorders and certain cancers.
desire to spare children from the unnecessary
suffering that Charlie Abrahams endured before GENESIS OF THE CHARLIE
he achieved seizure freedom with the ketogenic F O U N D AT I O N
diet. Jim and Nancy Abrahams shared their story Charlie Abrahams, the youngest in a family of
in 1994 on Dateline and through a 1999 movie three children, had no apparent health problems
called First Do No Harm. Across the Atlantic in until his first birthday in March 1993, when he
the United Kingdom, Matthew Williams suf- experienced his first seizure. This was followed by
fered for 6  years with a devastating seizure dis- a series of tumultuous events that changed his life
order before becoming seizure-free within 2 as well as the lives of his siblings, Joseph and Jamie;
weeks of starting the diet. Based on this experi- his parents, Jim and Nancy; and ultimately thou-
ence, Matthew’s Friends was founded in 2004 by sands (possibly millions) of others. His father Jim
Emma Williams, Matthew’s mother, with a simi- described those months as follows:
lar mission as the Charlie Foundation. Despite
these dramatic testimonials and dedicated foun- After thousands of epileptic seizures, an
dations, the diet remained underutilized. Several incredible array of drugs, dozens of blood
key breakthroughs came in 2008:  The Charlie draws, eight hospitalizations, a mountain of
Foundation had commissioned medical profes- EEGs, MRIs, CST scans and PET scans, one
sionals with ketogenic experience to collaborate fruitless brain surgery, five pediatric neurolo-
on guidelines for prescribing the diet. This cul- gists in three cities, two homeopaths, one faith
minated in a publication in Epilepsia, a presti- healer, and countless prayers, Charlie’s seizures
gious international medical journal. During the were unchecked, his development “delayed”
same year, a Class I study was published in Lancet and he had a prognosis of continued seizures
Neurology confirming the diet’s efficacy as a treat- and “progressive retardation.”
ment for epilepsy; more positive Class  I  studies
followed. Use of ketogenic diet therapy spread Charlie lived that period in his bed, on a blan-
rapidly worldwide, and with increased use came ket, or in an infant seat, where he was safely
a broader understanding of its benefits for other padded during the violent seizures that caused
disorders. Less restrictive versions of the diet were him to thrash uncontrollably. The adverse effects
developed to meet the needs of older children and of the antiseizure medications were nearly as
adults. In 2012 the Charlie Foundation also began destructive as the seizures themselves. Between
educating all people with epilepsy to eliminate seizures, he was groggy, chronically constipated,
sugar, reduce refined carbohydrates, and choose a and in the words of his father, seemingly “drunk.”
predominantly whole foods diet. In addition, both Charlie also underwent a high-risk brain sur-
foundations have expanded efforts to address gery to attempt to remove the area of the brain
other conditions that can benefit from ketogenic suspected to be the source of his seizures. He
387

Chapter 39: Awareness and Application of Ketogenic Therapies Globally 387

experienced zero improvement, and the seizures bills, and a drugged, poorly functioning child in
continued. a rapid decline. Adoption of the ketogenic diet was
Exhausted by the failure of the medical com- the common denominator, and, for some, their
munity to provide an effective therapy and reel- lifesaver.
ing from the severe disruption inflicted on the One letter came from a family who had to
rest of the family, Jim began looking for another mortgage their farm to pay their son’s medical
option. He spent hours in the medical library of bills. The child was admitted to a hospital for treat-
UCLA reading through literature on epilepsy, and ment of uncontrollable seizures and placed under
it was there that he came across an epilepsy book heavy sedation. The family requested a trial of the
containing a chapter on ketogenic diet as a treat- ketogenic diet, but the treating physicians refused
ment for epilepsy. He subsequently discovered a to consider this. Instead, they wanted to add more
1992 study from Johns Hopkins University that medications and had recommended surgery. With
had been published in Epilepsia documenting 29% the help of a compassionate nurse, the family made
seizure freedom with an additional 30% significant the difficult decision to leave without medical
improvement in 58 consecutive cases of children authorization. They flew directly to Johns Hopkins
who were placed on a ketogenic diet (Kinsman Hospital. Several days later, this child, like Charlie,
et al., 1992). Notably, these children were as com- became seizure-free.
promised by their disease as Charlie. Jim brought Jim was compelled to use his professional skills
this book and the study to Charlie’s pediatric neu- as a movie producer and his connections in the
rologist. Although he was aware of the diet, he industry to find ways to help more families learn
immediately dismissed it as a treatment that “he about the diet. He wrote, directed, and produced
had never seen work.” Despite this discourage- a dramatization of the devastating impact epilepsy
ment, Jim and Nancy made the decision to take can have, and the potential for the diet to stop sei-
Charlie to Johns Hopkins Hospital in Baltimore, zures when nothing else has helped. In 1997 this
Maryland. (At the time, Johns Hopkins was one story aired in a made-for-TV film called First Do
of the few hospitals in the United States that had No Harm. The film starred Meryl Streep (who
provided the ketogenic diet continuously since its received a Humanitas Award for her performance),
discovery in 1921.) Dr. John Freeman and the die- and the movie became a conduit for spreading the
titian Millicent Kelly formed the hospital team that word about the ketogenic diet worldwide. Twenty-
implemented Charlie’s ketogenic diet. two years later, the Charlie Foundation continues
Within 3 days of starting this treatment, Charlie to hear from people who learned about the diet
was seizure-free. Over the course of the following through this movie. Despite the diet’s over-90-year
month he was weaned off of the four failed antisei- history, many physicians first learn about the diet
zure medications. Free from the sedating effects of from the families of their patients.
antiseizure medication, Charlie could finally hold The Abrahams founded the Charlie Foundation
his head up and once again interact with his family. to Cure Pediatric Epilepsy in 1994. Funding was
Although the diet required careful measurement provided for public awareness and research in ani-
and specific foods, Jim described this as a “walk mal models. Videos were developed for families
on the beach” when compared with their previous and medical professionals, and the Johns Hopkins
circumstances. This abrupt and almost incompre- team received financial support for a book, The
hensible reversal left the Abrahams with a diverse Ketogenic Diet: A Treatment for Epilepsy (now in its
range of emotions; exhilaration and gratitude over 6th edition). Over the last two decades, the Charlie
Charlie’s sudden recovery was mixed with anger Foundation has organized educational confer-
over the 9 months of unnecessary suffering that he ences, maintained a website, and trained medical
had endured. teams at hospitals worldwide.
Jim and Nancy appeared on NBC-TVs
Dateline in 1994 to recap those months. They soon G E N E S I S O F   M AT T H E W ’ S
found their mailbox overflowing with letters from FRIENDS
other families, thanking them for bringing the Matthew Williams was born in 1995. He suffers
diet out into the light of day. Each letter described with a catastrophic form of epilepsy called Dravet
a similar journey to the one the Abrahams expe- syndrome, and his seizures started when he was
rienced with Charlie. The names and ages of the 9  months old. Like Charlie, his seizures were
kids were different, but the stories were the same. severe, frequent, and uncontrolled by medication.
Uncontrolled seizures, multiple medications, Emma, his mother, asked if Matthew could try the
trips to the emergency room, mounting medical ketogenic diet when he was 2  years old; she was
388

388 section IV: Ketone-Based Metabolism

told the diet “didn’t work.” As none of the medi- shocked and angry at the lack of information
cations helped, she continued to ask periodically and the underutilization of the diet—even when
about the ketogenic diet and was given a variety of Emma had asked about it years earlier! She was
responses; she was told the diet was “too hard” and determined to raise awareness and access to the
caused “terrible side effects.” Like the Abrahams, ketogenic diet.
Emma battled on, trying innumerable medica-
tions that did not help Matthew’s seizures and TEN YEARS
themselves caused devastating side effects. Neither O F   A C C E L E R AT I N G A N D
family was told that it has long been known that C O M P L E M E N TA RY E F F O R T S
once the first medication fails the chance of sei- The past 10 years have been a period of enormous
zure control with another medication or an added growth and revitalization for the field of metabolic
medication is lower, and even lower with succes- therapy and for both foundations. Beth Zupec-
sively added drugs (down to 2.7% seizure control Kania, a registered dietitian and nutritionist,
at the third round; Mohanraj and Brodie, 2006). joined the Charlie Foundation in 2006 to develop
There is even evidence that polypharmacy can resources and trainings for ketogenic therapies.
aggravate seizures (Perucca et  al., 1998; Shorvon She designed an online ketogenic diet calculator
and Reynolds, 1979). Matthew continued to suffer program (KetoDietCalculator.org), which allows
from thousands of seizures, and his situation did nutritionists to efficiently create diets including
not improve on what Emma termed the “merry- meal plans, special recipes, and infant and feeding
go-round” of medications. Emma was told that if tube formulas. A  Help Line within the program
Matthew lived to age 12 he would likely be con- supports clinicians by answering daily questions.
fined to a residential home. KetoDietCalculator is provided without cost to
When Matthew was 7, at a routine appoint- licensed nutritionists, who can choose to extend
ment Emma insisted on trying the diet—the only access to their patients or clients. It receives regular
other option was new combinations of drugs that updates and additions and has been used by over
had already been proven ineffective and shown to 50,000 people worldwide. Figure 39.1 displays a
cause terrible side effects in Matthew: screenshot of a typical meal (4:1 ratio of fat to non-
fat grams) designed using the KetoDietCalculator.
“The side effects of the medications were awful, In 2007, the Charlie Foundation estimated
he was on a considerable amount of medica- that fewer than 15 of the 200 children’s hospitals
tion and his quality of life was so poor already in the United States had a ketogenic diet therapy
that it really could not have got any worse for program. Frustrated by this lack of progress, the
any of us.” Charlie Foundation commissioned a group of phy-
sicians and dietitians to collaborate on methods of
Fortunately there was a new option. Six years providing ketogenic diets. Their report, published
after Emma first asked about the ketogenic diet, in 2008 in Epilepsia was titled “Optimal Clinical
Matthew and 144 other children with severe epi- Management of Children Receiving the Ketogenic
lepsy were enrolled in a clinical research trial of Diet:  Recommendations of the International
the ketogenic diet spearheaded by Dr.  Helen Ketogenic Diet Study Group” (Kossoff et al., 2009).
Cross at Great Ormond Street Hospital (GOSH), This consensus statement became the cornerstone
London, England (Neal et  al., 2008). Within 2 for developing new programs. The report identi-
weeks of starting the diet Matthew’s seizures had fied which patients were most likely to benefit,
reduced by 90% and within 8 months he was off all outlined methods of starting the diet, and pro-
medication. His behavior improved dramatically. vided monitoring guidelines. The group summa-
Sadly for Matthew, however, the damage had been rized its conclusions as follows:
done. Years of thousands of seizures had caused
terrible brain damage, his family had broken apart, The ketogenic diet (KD) should be strongly
and Emma was now a single mum to Matthew and considered in a child who has failed two to
his younger sister, Alice. three anticonvulsant therapies, regardless of
Inspired by Matthew, and hoping she could age or gender, and particularly in those with
spare other families from what they had endured, symptomatic generalized epilepsies. It can
and particularly prevent others from lifelong dis- be considered the treatment of choice for
abilities, Emma started Matthew’s Friends in two distinct disorders of brain metabolism,
2004—initially running the charity out of her Glut-1 deficiency syndrome and Pyruvate
kitchen. She, like Jim and Nancy Abrahams, was Dehydrogenase Deficiency Disorder. In the
389

Chapter 39: Awareness and Application of Ketogenic Therapies Globally 389

FIGURE 39.1 Screen shot of a 4:1 ketogenic meal designed in Ketodietcalculator.org.

particular epilepsy syndromes of Dravet syn- Association of Epilepsy Centers) to centers that
drome, infantile spasms, myoclonic-astatic provide medically approved epilepsy treatments.
epilepsy, tuberous sclerosis complex, the KD
could be offered earlier. A S P E C T RU M
O F   K E TO G E N I C D I E T
In the same year as this collaborative report, a THERAPIES
randomized-controlled study of children receiv- Expanding use of the ketogenic diet for epilepsy
ing the classic and medium chain triglyceride- paved the way to variations in the diet, particularly
supplemented ketogenic diets was published in for adults and for older children who find it diffi-
Lancet Neurology (Neal et  al., 2008). This is the cult to adhere to a restrictive diet. Dr. Eric Kossoff
clinical trial in which Matthew Williams partici- at Johns Hopkins developed a modified version of
pated. Children between the ages of 2 and 16 with the ketogenic diet known as the modified Atkins
refractory seizures continued their current treat- diet (Kossoff et al., 2003). At about the same time,
ment and were randomized to a ketogenic diet Heidi Pfeifer, a Massachusetts General Hospital
or continued standard of care for 3  months. The dietitian, developed the low glycemic index treat-
results were clear:  children treated with the diet ment (LGIT; Pfeiffer and Thiele, 2005). These two
were significantly improved compared with base- newer diets are similar to the classic ketogenic diet
line and with the control group; some had over a in their high-fat, low-carbohydrate content, but
90% reduction in their seizures or were seizure- are different in that they could be initiated outside
free. Seizure frequency in children who continued of the hospital and do not require careful weigh-
to receive the standard of care worsened compared ing of each food. The modified Atkins diet was
with baseline, and none had over a 90% reduction shown to be effective in a Class I study (Sharma
or were seizure-free. This Class I study is consid- et al., 2013). Clinical reviews began to report the
ered the highest level of conclusive evidence and efficacy of the modified Atkins and LGIT diets as
was the affirmation that doctors and advocates being “nearly as effective” as the classic ketogenic
had been waiting for. It had been nearly 90 years diet (Coppola et al., 2011; Karimzadeh et al., 2014;
since the ketogenic diet was developed and finally Thibert et al., 2012). These two diets are often eas-
a comparison of the diet as treatment for difficult- ier for older children and adults to manage and
to-control epilepsy against multiple antiseizure are sometimes used as predicates to the classic
medications proved that the diet was superior in ketogenic diet.
effectiveness. Recently, another Class I study found For now, the classic ketogenic diet remains the
similar results (Lambrechts et al., 2016). most effective diet therapy based on current sci-
The combination of the Epilepsia and Lancet entific evidence. However, less strict versions may
Neurology publications gave further credence to be more realistic for certain individuals, especially
the diet, and in 2010 led to the requirement for older children and adults. These metabolic diet
inclusion of ketogenic diet therapies in Level 3 treatments are referred to collectively as “ketogenic
and 4 Epilepsy Centers in the United States. This therapies.” The Charlie Foundation published a
distinction is granted by the NAEC (National chart (Table 39.1) that outlines the differences
390
TABLE 39.1 COMPARISON OF DIET THERAPIES FOR EPILEPSY

QUESTIONS Ketogenic Therapies MCT Oil Low Glycemic Index Modified Atkins
Treatment

Is medical supervision required Yes Yes Yes Yes


Is diet high in fat? Yes Yes Yes Yes
Is diet low in carbohydrate? Yes Yes Yes Yes
What is the ratio of fat to carbo- 4:1, 3:1, 2:1, 1:1 Approximately 1:1 Approximately 1:1 Approximately 2:1
hydrate & protein?
How much carbohydrate is 8gm carb on a 4:1 40-50gm 40-60gm 10gm adolescents or 15gm adults for
allowed on a 1000 Calorie 16gm carb on a 3:1 1 month
diet? 30gm carb on a 2:1 20gm afterwards
40-60gm carb on a 1:1
How are foods measured? Weighed Weighed or measured Measured or estimated Estimated
Are meal plans used? Yes Yes Yes Optional
Where is the diet started? Hospital Hospital Home Home
Are calories controlled? Yes Yes Yes No
Are vitamin and mineral supple- Yes Yes Yes Yes
ments required?
Are liquids (fluids) restricted? No No No No
Is a pre-diet laboratory evalua- Yes Yes Yes Yes
tion required?
Can there be side-effects? Yes Yes Yes Yes
What is the overall difference in This is an individualized and An individualized and This is individualized but This diet focuses on limiting the amount
design of these diets? structured diet that pro- structured diet contain- less structured diet than of carbohydrate while encouraging
vides specific meal plans. ing Medium Chain the ketogenic diet. It uses fat. Carbohydrate may be consumed
Foods are weighed and Triglycerides (MCT) which exchange lists for planning at any time during the day as long as
meals should be consumed are highly ketogenic. This meal and emphasizes com- it is within limits and should be con-
in their entirety for best allows more carbohydrate plex carbohydrates. The sumed with fat. Suggested meal plans
results. The ratio of this diet and protein than the clas- balance of low glycemic are used as a guide. Protein is not
can be adjusted to effect sic ketogenic diet. A 2008 carbohydrates in combi- limited but too much is discouraged
better seizure-control and study showed that both nation with fat result in
also liberalized for better diets are equal in eliminat- steady glucose levels. It is
tolerance. This diet is also ing seizures. A source of not intended to promote
considered a low glycemic essential fatty acids must be ketosis.
therapy and results in included with this diet.
steady glucose levels.
The Charlie Foundation for Ketogenic Therapies © 2015 www.charliefoundation.org
391

FIGURE 39.2 Charlie Foundation logo.

FIGURE 39.3 Charlie thanking Meryl Streep for her role in First Do No Harm.
392

392 section IV: Ketone-Based Metabolism

FIGURE 39.4 Charlie reading to his preschool students in Los Angeles.

between the therapies to assist families and health high-carbohydrate diet (Volek et al., 2008). Due in
professionals in selecting the best option. part to the appetite-suppressing effect of ketosis,
Both the Charlie Foundation and Matthew’s fewer calories are consumed—resulting in weight
Friends offer a wealth of recipes and expertise, and adipose tissue loss along with lowered blood
including demonstration videos. Their tireless glucose, triglycerides, and saturated fatty acids.
dedication to developing and sharing recipes has People inquiring about ketogenic therapy
helped make the diet enormously more palatable for brain cancer have become the second-largest
and accessible to patients and their families. group (next to epilepsy sufferers and their fami-
lies) requesting information from both organiza-
O T H E R C O N D I T I O N S T H AT tions. These individuals often experience seizures;
M AY B E N E F I T therefore the role that the ketogenic diet offers as
A study published in 2008 in Lipids laid the ground- an antiseizure, anti-inflammatory, and antitumor
work for an entirely new application of ketogenic therapy offers multifaceted benefits (Seyfried and
diet therapy. Metabolic syndrome is a diagnosis Mukherjee, 2005; Veech, 2004). Several studies
that includes three or more of the following abnor- are currently in progress through the National
malities; abdominal obesity, elevated blood pres- Institutes of Health (NIH) investigating ketogenic
sure, elevated fasting blood glucose, and elevated diet effects on cancer. Recently Matthews Friends
lipids or triglycerides. This syndrome is currently has partnered with the Astro Brain Tumour Fund
an epidemic in many areas of the world, includ- to help adults and children with brain cancer.
ing the United States and the United Kingdom. Additional applications of ketogenic diet ther-
Improvements in metabolic syndrome were signif- apies for other conditions have grown exponen-
icantly better in a group that followed a modified tially in recent years. Benefits have been reported
ketogenic diet versus those who followed a low-fat, in a multitude of neurological disorders and

FIGURE 39.5 Matthew’s Friends logo


393

Chapter 39: Awareness and Application of Ketogenic Therapies Globally 393

therapies facilitates selecting the best diet for indi-


viduals taking into account their condition, diag-
nosis, age, and ability to comply. Matthew’s Friends
developed a support system to help families prior
to and during the diet to enhance the likelihood of
success. Adjusting the therapy during the course of
treatment to optimize effectiveness is often neces-
sary. Discontinuing the diet is usually the goal in
childhood epilepsy: two to three years is the typi-
cal course of treatment. However, in some cases of
epilepsy and other neurological disorders it may
be necessary to continue with a modified version
of the diet indefinitely. These discussions are fre-
quently addressed at the Charlie Foundation and
have become the topics of collaborative journal
articles and professional guidelines spearheaded
by nutritionists.
News of ketogenic diet therapies spreads
quickly through social media and the popu-
lar press. Thousands of copies of the Charlie
Foundation’s Parent’s Guide to the Ketogenic Diet
have been distributed in English and Spanish.
Consultant chef Dawn Martenz and nutrition-
ist Beth Zupec-Kania have collaborated on a new
cookbook, Keto Cookbook II (Demos Publications).
Cooking videos of the most popular keto recipes
have been added to the charliefoundation.org
FIGURE 39.6 Emma and Matthew Williams website, and posts of creative snacks and meals
are added to the Facebook page regularly. In addi-
animal models of disorders including autism, cer- tion, an App is under development for the modi-
tain mitochondrial diseases, diabetes, migraine, fied ketogenic diet. Similarly, Matthew’s Friends is
Prader-Willi syndrome, neurodegenerative dis- dedicated to developing and publishing delicious
eases including Alzheimer’s and Parkinson’s, trau- keto-friendly meals.
matic brain injury, and stroke. New research may Both the Charlie Foundation and Matthew’s
expand the mission of the Charlie Foundation Friends have been continuously supportive of
and Matthew’s Friends—or may bring new part- research and have partnered in funding global
ners supporting metabolic therapies for diverse symposia on ketogenic diets. They stay abreast of
applications. new research related to ketogenic diet therapies.
In response to the increasing demands for A recent study in the journal Obesity (2015) showed
adult resources, the Charlie Foundation pro- dramatic improvements in the health of obese chil-
duced a guide in 2014 titled “Modified Ketogenic dren in just 10 days after eliminating sugar in their
Therapy for Neurologic and Other Conditions.” diets (Lustig et al., 2016). The metabolic impact of
Intended for use under medical supervision, it a sugar-free diet is one element of ketogenic diets
describes portion sizes for protein, fat, and car- that can be undertaken by anyone without the
bohydrate and advice for optimal nutrition. The need for medical supervision. Eliminating sugar
Charlie Foundation has shared resources with can be a difficult task for most people and requires
oncology medical professionals and expanded its strong motivation. A  2007 study published in
website information to include this new popula- Neuroscience and Behavioral Reviews showed that
tion of users. A new link on the charliefoundation. rats that were fed sugar intermittently had behav-
org landing page has been added to ketogenic diet ior and neurochemical changes that resemble the
studies conducted through the NIH for cancer, effects of substance abuse (Avena et  al., 2008).
epilepsy, and other disorders. Eliminating refined (processed) foods is a second
Medical supervision for ketogenic therapies step that can be taken to improve one’s diet.
and choosing the appropriate diet therapy for each The Charlie Foundation has translated these
individual is an evolving focus. The spectrum of and other similar research findings into pragmatic
394

394 section IV: Ketone-Based Metabolism

guidelines. A  new publication, Does What I  Eat


Affect My Epilepsy? outlines steps that can be taken BOX 39.1
to eliminate sugar and refined foods and to con- CHARLIE FOUNDATION
sume a mostly whole foods diet. This has been EDUCATION RESOURCES
distributed widely (in English and Spanish) for all
people with epilepsy regardless of their interest in
ketogenic diet therapy. Parent’s Guide to the Ketogenic Diet
For conditions that may benefit from keto- Modified Ketogenic Diet Therapy:  1:1 and
genic therapies, the Foundation designed a Pre- 2:1 Prescriptions
ketogenic Diet. This document is distributed (free)
Does What I Eat Affect My Epilepsy?
to health professionals to provide to their patients
or clients who are potential candidates for keto- Preketogenic Diet; Low-Carb, Gluten-Free,
genic diet therapy. Similar to the classic ketogenic High-Fat
diet, it eliminates gluten and sugar; however, it is KetoDietCalculator Guide for the Nutritionist
not intended to induce ketosis. Instead, it can be and the User
described as a whole foods Mediterranean-style
diet that prepares the user for the transition to Professional’s Guide to the Ketogenic Diet
ketogenic therapy and also aids in determining Frequently Asked Questions about
which diet in the spectrum is best suited for their Ketogenic Diets
needs and preferences. In addition, healthcare
Comparison of Ketogenic Diet Therapies
professionals can use the Pre-ketogenic Diet as a
screening tool to identify which individuals are Ketogenic Diet Primer for Health Professionals
able to adhere to the lifestyle changes required for
ketogenic diets.

T H E C H A R L I E F O U N D AT I O N professionals with the intent of advancing research


A N D M AT T H E W ’ S F R I E N D S — and clinical use of ketogenic therapies. Plans for
L O O K I N G TO   T H E   F U T U R E future symposia are in-the-making through the
In 2013 the Charlie Foundation renamed itself the year 2022. As new clinical and scientific research
Charlie Foundation for Ketogenic Therapies to continues to emerge, the Foundation will respond
better define its mission for the future: advocacy, with further resources to promote safe and effec-
awareness, and education. An expansion of edu- tive use of ketogenic therapies and make it less
cation efforts includes adult-focused media and daunting for the user to manage.
training. Development of new resources in both Matthew’s Friends grew rapidly from Emma’s
English and Spanish remains a goal. A list of cur- kitchen table, and in 2011 opened its own clinic.
rent publications is shown in Box 39.1. The charity has continued to grow rapidly, and
Throughout the 22  years of its existence, the recently expanded into New Zealand and Canada.
Charlie Foundation has continued to receive daily In 2016 it embarked on a new program called
e-mails and letters from families and health pro- KetoCollege, to offer training for teams around the
fessionals requesting assistance. Jim Abrahams globe, adding to and complementing the efforts
has responded to the majority of these requests, of the Charlie Foundation to train ketogenic diet
triaging some of them to support staff. Although teams at their home institutions around the world.
Charlie has been seizure- and drug-free and off of Now seizure-free young adults—no longer on
the diet since 1999, Charlie’s family continues to a special diet—Charlie Abrahams and Matthew
represent ketogenic therapies in their Los Angeles Williams are two clear examples that even cata-
community and on a national level through part- strophic epilepsy can be cured by a ketogenic
nerships with other nonprofits and with supportive diet. A  question frequently asked about Charlie,
commercial organizations. Over 160 medical cen- Matthew, and others who have become seizure-
ters have been trained in the spectrum of ketogenic free on a ketogenic therapy is “How is it possible
therapies in the United States and beyond, includ- that they can come off of the diet and remain
ing Canada, Portugal, Austria, Jamaica, Slovenia, seizure-free?” At the present time, there is no clear
Kuwait, Saudi Arabia, and the Republic of Georgia. answer to this question. Emerging genetic research
Beginning in 2008, the Charlie Foundation has may soon clarify whether early application of keto-
also sponsored and organized global symposia genic therapy may either ameliorate the disease
bringing together leading scientists and medical or even cure it. The field of nutritional genomics
395

Chapter 39: Awareness and Application of Ketogenic Therapies Globally 395

looks at the effect of nutritional changes on genes the ketogenic diet in refractory childhood epi-
and suggests that maladaptive epigenetic changes lepsy. Acta Neuro Scand. doi: 10.1111/ane.12592
can be altered by diet. Targeted diet therapies are Lustig, R.H., Mulligan, K., Noworolski, S.M., Tai, V.W.,
already being prescribed along with genetic testing Wen, M.J., Erkin-Cakmak, A., Gugliucci, A., and
for certain metabolic disorders. Schwarz, J.M. (2016). Isocaloric fructose restric-
tion and metabolic improvement in children with
Implementation of a restrictive diet is easiest
obesity and metabolic syndrome. Obesity (Silver
in infants and children (given their reliance on the
Spring) 24, 453–460.
parents) and becomes more challenging as children Mohanraj, R., and Brodie, M.J. (2006). Diagnosing
gain independence. Early initiation of ketogenic refractory epilepsy:  response to sequential treat-
therapies is better tolerated in young children— ment schedules. Eur J Neurol 13, 277–282.
which may result in improved compliance and Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
outcomes. Nevertheless the diet does work in Edwards, N., Fitzsimmons, G., Whitney, A., and
adults, and has ever-increasing applications. Cross, J.H. (2008). The ketogenic diet for the treat-
ment of childhood epilepsy:  a randomised con-
REFERENCES trolled trial. Lancet Neurol 7, 500–506.
Avena, N.M., Rada, P., and Hoebel, B.G. (2008). Perucca, E., Gram, L., Avanzini, G., Dulac, O. (1998)
Evidence for sugar addiction: behavioral and neu- Antiepileptic drugs as a cause of worsening sei-
rochemical effects of intermittent, excessive sugar zures. Epilepsia 39, 5–17
intake. Neurosci Biobehav Rev 32, 20–39. Pfeiffer, H.H., and Thiele, E.A. (2005). Low-glycemic-
Coppola, G., D’Aniello, A., Messana, T., Di Pasquale, index treatment:  a liberalized ketogenic diet for
F., della Corte, R., Pascotto, A., and Verrotti, A. treatment of intractable epilepsy. Neurology 65,
(2011). Low glycemic index diet in children and 1810–1812.
young adults with refractory epilepsy: first Italian Seyfried, T.N., and Mukherjee, P. (2005). Targeting
experience. Seizure 20, 526–528. energy metabolism in brain cancer:  review and
Karimzadeh, P., Sedighi, M., Beheshti, M., Azargashb, hypothesis. Nutr Metab 2, 30.
E., Ghofrani, M., and Abdollahe-Gorgi, F. (2014). Sharma, S., Sankhyan, N., Gulati, S., and Agarwala, A.
Low glycemic index treatment in pediatric refrac- (2013). Use of the modified Atkins diet for treat-
tory epilepsy: the first Middle East report. Seizure ment of refractory childhood epilepsy: a random-
23, 570–572. ized controlled trial. Epilepsia 54, 481–486.
Kinsman, S.L., Vining, E.P., Quaskey, S.A., Mellits, D., Shorvon, S.D., and Reynolds, E.H. (1979) Reduction
and Freeman, J.M. (1992). Efficacy of the keto- in polypharmacy for epilepsy. Br Med J 2:1023–5.
genic diet for intractable seizure disorders: review Thibert, R.L., Pfeifer, H.H., Larson, A.M., Raby,
of 58 cases. Epilepsia 33, 1132–1136. A.R., Reynolds, A.A., Morgan, A.K., and Thiele,
Kossoff, E.H., Krauss, G.L., McGrogan, J.R., and E.A. (2012). Low glycemic index treatment for
Freeman, J.M. (2003). Efficacy of the Atkins diet seizures in Angelman syndrome. Epilepsia 53,
as therapy for intractable epilepsy. Neurology 61, 1498–1502.
1789–1791. Veech, R.L. (2004). The therapeutic implications of
Kossoff, E.H., Zupec-Kania, B.A., Åmark, P.E., ketone bodies:  the effects of ketone bodies in
Ballaban-Gil, K.R., Bergqvist, A.G.C., Blackford, pathological conditions:  ketosis, ketogenic diet,
R., Buchhalter, J.R., Caraballo, R.H., Cross, redox states, insulin resistance, and mitochondrial
J.H., Dahlin, M.G., et  al. (2009). Optimal clini- metabolism. Prostaglandins Leukot Essent Fatty
cal management of children receiving the keto- Acids 70, 309–319.
genic diet: recommendations of the International Volek, J.S., Fernandez, M.L., Feinman, R.D., and
Ketogenic Diet Study Group. Epilepsia 50, Phinney, S.D. (2008). Dietary carbohydrate
304–317. restriction induces a unique metabolic state posi-
Lambrechts, D.A.J.E., de Kinderen, R.J.A., Vles, J.S.H., tively affecting atherogenic dyslipidemia, fatty acid
de Louw, A.J.A., Aldenkamp, A.P., and Majoie, partitioning, and metabolic syndrome. Prog Lipid
H.J.M. (2016). A randomized controlled trial of Res 47, 307–318.
396
397

INDEX

Page references followed by an italics f indicate figures, t indicate tables, and b indicate boxes.

Abbott, N. J., 289, 292–294 adverse effects. see side effects


Abdallah, D. M., 174–175 aerobic fermentation, 80
Abrahams, Charlie, 386 aerobic glycolysis, 80
Abrahams, Jim, 3 aging
AC-1202, 319–320 cell senescence, 257
acetoacetate. see also ketone bodies neuroprotection, ketosis, 221–222, 221f
acetone from, 113 reactive oxygen species, 256–257
administration, 124 telomere shortening, 256–257
Alzheimer’s, 122f aging and neurodegeneration, 216–222
brain energy source, 113 cataplerosis–anaplerosis balance, 218
brain uptake, 296f glucose and ketone metabolism, 113–127 (see also
hippocampal slices, 187–190, 189t Alzheimer’s disease)
inflammation, 150–151 glucose metabolism, oxidative stress, 217–218, 218f
ketogenic diet, 90, 136 HIF1α stabilization, 219–221
Krebs cycle, 243f ketone bodies, 216–217
mitochondrial metabolism, 315f ketosis, 217
NADP system, 263f ketosis neuroprotection, clinical relevance,
neuroprotection, 135 218–219, 219f
prolyl-hydroxylase inhibition, 220 Ahn, Y., 107
redox potential, 262t D-alanine, seizure onset protection, 348
synthesis and metabolism, 177, 187, 228, 244, Allen, B. G., 92, 367
247–248, 248f, 296f Allen, Frederick Madison, 364, 365
VGLUTs, 281 Alpers disease, status epilepticus, 61
acetone “alternative” ketogenic diets, 5–13. see also specific types
from acetoacetate, 113 choice, 11–12
ketogenesis, 119f effectiveness, vs. classical ketogenic diet, 10–12, 10t
acidosis, 67 initiation and follow-up, 12–13
actin, 293 low glycemic index treatment, 6f, 9–10, 9b
Adabi Mohazab, R., 174 medium chain triglyceride, 5–7, 6b, 6f
adenosine modified Atkins diet, 6f, 7–9, 8b, 8t
on inflammation, 151f, 152–153 Alzheimer, Alois, 242
pain, 201 Alzheimer’s disease, 113–127, 241–249
retaliatory metabolite, 210 anaplerosis, 124–125
adenosine homeostasis arteriovenous difference, brain, 114–115, 115t
seizures, 209–210 brain energy status, neuroprotection, and
temporal lobe epilepsy, 211–213, 212f mitochondrial function, 125–126
adenosine kinase, epilepsy overexpression, 209–210 early brain development and evolution, 126
adherence, 19 environmental factors, 241–241–242
Adk gene, epilepsy, 210–211 genetics, 241
adult polyglucosan body disease, triheptanoin, 341–342 glucose, cerebral use, 243–244
398

398 Index

Alzheimer’s disease, (cont.) astrocytes


glucose, presymptomatic hypometabolism, 116–118, BAD-altered, metabolic changes, 272
116t, 117f blood-brain barrier, 290f, 292–293
glucose, uptake and metabolism, 113–114 athlete, keto-adapted, 382, 383f
hyperketonemia with cognitive deficit, 123–124, 125t autism spectrum disorder, 101–107
hypometabolism, 216–217 animal models, 105–107
incidence, 241 definition, 101
ketone bodies, available forms, 247–249, 248f, 249f effects, 105
ketone kinetics and transport, 118–120, 120f, 120t epidemiology, 101
ketone regional uptake, early disease, 121–122, etiology, 101
121f, 122f future research, 107
ketones, brain fuel, 113, 118, 119f ketogenic diet, 103–105
ketone supplementation, 318–320, 319f mitochondrial and metabolic dysfunction, 102–103,
medium chain triglycerides, dose-response 104t–105t
ketogenesis, 6f, 122–123, 123f pathophysiology, 101–102
medium chain triglycerides, safety, 126
nutritional and metabolic factors, 241–242 Babayan, Vigen K., 311
pathophysiology, 242–244, 243f BAD, 271
perspectives, 126–127, 127t on KATP channels, 271, 275
PET-FDG protocol, 114 BAD channels, metabolic seizure resistance,
predisposing factors, 241–242 271–274, 277f
prevention, 113 glucose and ketone body metabolism, 271–272, 282
risk factors, 113 KATP channels, 275–277, 276f
treatment, 244–247, 245f–246f metabolic function alteration, 272, 273f, 277f
amyloid, 244 neurons and astrocyte metabolic changes, 272
ketone body metabolism, 244–247, 245f–246f vs. other BCL-2 family proteins, 273
triheptanoin, 341 Bansal, S., 29
amino acids, for neurological disorders, 346–349 Banting, William, 363
seizures and epilepsy Barborka, C. J., 16–17, 23
D-amino acids, 347–349, 349b basement membranes, blood-brain barrier, 290f, 292
L-amino acids, 346–347, 347b Bastible, C., 16–17
traumatic brain injury, 349 BCL-2-associated agonist of cell death (BAD), 271. see
AMPA receptor agonists, medium chain fatty acids, also BAD channels, metabolic seizure resistance
331–332, 331t behavioral effects, 231
amyloid precursor protein (APP), 242, 243 Bellisario, V., 236
amyotrophic lateral sclerosis (ALS), 157–158 Bergqvist, A. G. C., 69
triheptanoin, 341 beta-oxidation metabolic pathway, 248, 249f
anaplerosis, 124–125, 218 β-hydroxybutyrate (BHB), 307. see also ketone bodies
propionyl-CoA carboxylation pathway, aging brain and Alzheimer’s disease, 113, 115t,
336–337, 338f 121–122, 121f
TCA cycling, 336, 337f, 338f aging brain and Alzheimer’s disease, kinetics,
Angelman syndrome, 52–53 120, 120t
low glycemic index treatment, 9–10, 9b antioxidant function, 138
anorexigenic food intake, 229, 229f brain energy source, 115t, 121–122, 121f
antiepileptogenesis. see epileptogenesis actions brain uptake, 296f
anti-excitotoxic effects, 137 cancer, 81–82, 83
antioxidants. see also specific types on cellular processes, 381
mechanisms of action, 257–258, 258f hippocampal slices, 187–190, 189t
neuroprotective mechanisms, 137–138 inflammation, 150–151, 151f
apolipoprotein E (ApoE), 241 KATP channels, 157
apolipoprotein E-4 (ApoE-4), 241, 242 ketogenesis, 119f
L-arginine ketogenic diet, 90, 281
seizure onset protection, 347 liver synthesis, 156
traumatic brain injury, 349 medium chain triglycerides, 123f
ascorbate, 258, 261, 262t metabolism and sum reactions, 243, 243f, 246f–248f,
ascorbic acid, 258f, 259 247–248
astrocyte-neuron lactate shuttle, 282–285, 283f–285f mineral salts supplementation, with medium chain
antiepileptic effects, 283–286, 284f, 285f triglycerides, 311–312
electrical regulation, 284, 285f neuroprotection, 124, 124f, 125t, 134
399

Index 399

nutritional ketosis, 377, 379 brain development, ketones, 126


Parkinson disease, 158 brain energy
on reactive oxygen species, 83 acetoacetate, 113
rodent model, 233f, 235–236 Alzheimer’s disease, 126
synthesis and metabolism, 187, 228, 296f β-hydroxybutyrate, 115t, 121–122, 121f
D-β-hydroxybutyrate bioenergetics, 77
administration, 124 ketones, 113, 118, 119f, 380–381
ionizing radiation protection, 263, 264f mitochondria, 125–126
ketogenesis, 119f requirements, 113
ketogenic diets, 136 brain energy metabolism
mitochondrial metabolism, 315f PET-FDG protocol, 114
neuroprotection, 135 triheptanoin, 339–340
on redox states, cellular, 261–263, 262f, 263f brain evolution, ketones, 126
on transcription, 261t, 264 brain injury, traumatic. see traumatic brain injury
Blackburn, Elizabeth, 256–257 brain ketones, 113, 118, 119f
Blomqvist, G., 121, 121f, 122 Bucher, T., 258
blood-brain barrier, 289–299 Buckley, J. A., 105
caveoline-based vesicle trafficking, 291f, 293 1,3-butanediol supplementation, 311
cellular associations, 289–292
astrocytes, 290f, 291–292 Cahill, G. F., 81, 258, 307, 317, 376
basement membranes, 290f, 292 calorie restriction
endothelial cells, 289–292, 290f, 291f classic ketogenic diet, 29
pericytes, 290f, 292 on inflammation, 151–152, 151f
endothelial cells, 289, 290f pain, 197–198
functions on tumor growth, 81–82
nutrient transport to brain, 291f, 294–295, 296f Canavan disease, triheptanoin, 341
transit regulation, 290f, 291f, 294 cancer, 79–83. see also brain cancer
glucose transporters, 294–295, 296f, 297f aspartate, 80–81
ketogenic diet, 295–299 calorie restriction, 81–82
cerebral ketone uptake regulation, 295–297, 297f cell metabolism, 80–81
ketone bodies on, 298–299 genetics and somatic mutation theory, 79
neuroinflammation, 297–298 as genetic vs. mitochondrial
neuroprotection, 298 metabolic disease, 79–80
molecules glucose, 80–81
leukocyte adhesion molecules, 293 Glucose/Ketone Index calculator, 82–83
tight junctions, 290f, 292–293 glutamine, 81
plasmalemma vesicle-associated hypoxia-inducible factor 1, 89–90
protein number, 293 ketogenic diet, 82–83
structure and functions, 289, 290f ketone supplementation, 320–321
transporters, 290f, 291f, 293, 296f mitochondrial dysfunction, 80
body composition, keto-adaptation, 378–379 nuclear factor-kappa B, 90
Bough, K. J., 192 PI3K/AKT signaling pathway, 89
brain radiation therapy, 92
ketones, 113 reactive oxygen species, 90
metabolic rate, glucose, 115 Caraballo, R. H., 44–47
PPARγ, 170–172, 172f carbohydrate
brain, aging, glucose and ketone metabolism, 113–127. depletion, 376
see also Alzheimer’s disease stored, 376
brain cancer, 392 carbohydrate, dietary
brain cancer, malignant, 88–94 decreasing, 376
glioblastoma multiforme, 88 glycemic effect, 366f, 367
ketogenic diet, humans, 93–94 carbohydrate loading, 380
ketogenic diet, with standard carbohydrate restriction, dietary
therapies, 90–91 diabetes mellitus, controlled studies, 367, 368t
metabolic remodeling, 88 diabetes mellitus, randomized controlled trials,
preclinical evidence, 91 367, 369t
standard therapies, 91–93, 92f obesity, randomized controlled
tumor metabolism and Warburg trials, 363, 363t
effect, 88–90, 89f carboxy-terminal binding protein (CtBP), 356
400

400 Index

cardiac arrest and resuscitation diabetes mellitus type 2, 362–371


ketosis neuroprotection, 218–219, 219f carbohydrate, glycemic effect, 366f, 367
survival after, 217–218, 218f carbohydrate restriction, randomized controlled trials,
cardiac hypertrophy, triheptanoin, 342 367, 369t
cardiac side effects, 69–70 dietary recommendations, evolution, 365
cardiomyopathy, 69–70 dietary recommendations, post-insulin era, 364
Castellano, C. A., 121, 121f, 122f keto-adaptation, 378
cataplerosis, 124–125, 218 ketogenic diet
caveoline-based vesicle trafficking, 291f, 293 metabolic ward study, 367
cell senescence, 257 nonrandomized studies, 370, 370t
cerebral metabolic rate, glucose, 115 origins and use, 363–364
Champ, C. E., 94 pilot study, 367–370
Chang, P., 7, 328–332, 329f, 330t, 331t remission case report, 370, 371f
Charlie Foundation, 3, 27, 386–392, 391f, 392f ketone supplementation, 321
achievements, 388–389, 389f low carbohydrate diets, controlled studies, 367, 368t
education resources, 393–394, 394b nutrition therapy, low glycemic dietary patterns,
future, 394–395 365–367
genesis, 386–387 pathophysiology, 362
history, 386 prevalence, 362
ketogenic diet therapy spectrum, 389–392, 390t progression and morbidity, 362
research support, 393–394 dietary therapy, adults, 16–24. see also specific types
childhood absence epilepsy (CAE), 56–57 demand, 17–19
Chuang, Y. C., 173–174 children transitioning to adult epilepsy
claudins, 292 providers, 17–18
clinical trials, 77 refractory epilepsy, 18
cognition, 22 super-refractory status epilepticus, 18–19
cognitive impairment, mild effects, adverse
ketogenic treatments, 127, 127t gastrointestinal, 22–23
medium chain triglycerides, 123f, 124, 125t lipids, 23
cognitive impairment, severe. see menstrual cycle, 23
Alzheimer’s disease other, 23
constipation, 67–68 effects, beneficial, 22t
convulsive status epilepticus, 61 cognition and mood, 22
Coppola, G., 317 weight loss, 22
cortical spreading depression, 160 history, 16–17
Couch, S. C., 70 results
counseling, prediet, 27 efficacy, 19–22, 20f, 21t
Crabtree effect, 80 feasibility, tolerability, and adherence, 19
Cross, Helen, 388 discontinuation, ketogenic diet, 70–71
prevalence, 66, 67t
D’Agostino, D. P., 91, 310–313, 317 DNA methylation levels, epilepsy, 211–213, 212f
Darlington, C. D., 79 Does What I Eat Affect Epilepsy?, 394
Davidson, T. E., 231 Doose syndrome, 35, 46–47
Deanna Protocol, 158 Dravet syndrome, 45–46
decanoic acid, 331. see also Drenick, E. J., 114, 118, 121f
medium chain fatty acids Dressler, A., 43, 45–46
AMPA receptors and seizure control, 331 drug resistant epilepsy, 40
blood-brain barrier penetration, 328 duration, treatment, 31
mitochondrial proliferation, 332 dyslipidemia, 229, 230t
seizure control, 330, 332
2-deoxyglucose, 353–359 education, prediet, 27
anticonvulsant actions, 353–355 effectiveness, timing, 29–30
antiepileptic actions, 355–356 efficacy, 19–22, 20f, 21t
chemical structure, 354f electrolyte imbalances, 69
glucose metabolism and glycolytic Ellenbroek, J. H., 229, 229f
pathway, 353, 354f El-Rashidy, O. F., 30
depression, 159 endocrine effects, 227–230, 228t, 229f, 230f, 230t
diabetes mellitus, Alzheimer’s disease and vascular endothelial cells, blood-brain barrier, 289–292, 290f, 291f
dementia, 242 energy, brain. see brain energy
401

Index 401

energy regulation fitness, keto-adaptation, 380–381


dysregulation, 77 food refusal, 70
long-term, 228–229 Freeman, J. M., 3, 40, 387
short-term, 228 Freemantle, E., 123, 124f
epigenetics, epilepsy, 210–211 free radicals. see also reactive nitrogen species (RNS);
epilepsy. see also specific topics reactive oxygen species (ROS)
adenosine kinase overexpression, 209–210 Alzheimer’s disease, 243–244
Adk gene, 210–211 redox state, 261, 262t
2-deoxyglucose for, 355–356 removal, in vivo, 263–264
DNA methylation levels, 211–213, 212f fructose-1,6-diphosphate (FDP), 357
energy metabolism and triheptanoin, 337–339, 338f
epigenetics, 210–211 Gahring, L. C., 170–171
fasting, 209 gene theory of cancer, 79
kainic acid status epilepticus (KASE) model, 210, 211 Geyelin, H. R., 16
ketogenic diet mechanisms, 281–282 Gibson, C. L., 298
ketone supplementation, 317 Giménez-Cassina, A., 272–274
PPARγ, 173–174 glioblastoma multiforme, 88. see also brain cancer,
epilepsy indications, established, 40–47 malignant
Doose syndrome, 35, 46–47 glioma, malignant, 88. see also brain cancer, malignant
Dravet syndrome, 45–46 glucagon, 229–230
infantile spasms, 42–44 glucose, 336
Lennox Gastaut syndrome, 44–45 blood levels, ketogenic diet, 228, 228t
refractory nonsurgical epilepsy, 40–42 chemical structure, 354f
epileptic encephalopathies, 44. see also specific types decreases, 281–282
epileptogenesis actions, 209–213 pain, 200–201
adenosine homeostasis and seizures, 209–210 glucose, brain
definition, 355 blood-brain barrier, 294–295, 296f, 297f
DNA methylation levels, 211–213, 212f cerebral metabolic rate, 115
epigenetics, 210–211 requirements, 113
ketogenic diet, 137, 165, 198 (see also ketogenic diet; glucose, brain, Alzheimer’s
seizure control) hypometabolism, presymptomatic, 116–118, 116t, 117f
ketogenic diet and adenosine homeostasis, uptake vs. metabolism, 118
211–213, 212f glucose-insulin axis, 376
erythropoietin, neuroprotection, 220 Glucose/Ketone Index Calculator (GKIC), 82–83
Eun, S. H., 43–44 glucose metabolism
evaluation, pre-treatment, 66–67, 67t, 71t BAD on, 271–272
Evangeliou, A., 105 2-deoxyglucose, 353, 354f
excitotoxic effects, 137 oxidative stress, 217–218, 218f
glucose suppression, 313–314
family-centered, team-based approach, 26 glucose transporters (GLUTs), 294–295, 296f, 297f
fasting GLUT 1 (glucose transporter type 1), 293, 297f
classic ketogenic diet, 28–29 GLUT 1 (glucose transporter type 1) deficiency, 18,
epilepsy, 209 35–37, 308
hyperketonemia, 307 clinical presentation and phenotype, 35–36, 36f
neuroprotection, 134 diagnosis, 35
pain, 196–197 ketogenic diet efficacy, 20
spinal cord injury, 134 ketogenic diet therapies, 36–37
fats ketone supplementation, 317–318
dietary, as fuel, 380–381 (see also brain energy) metabolic concept, 35, 36f
stored, 376 open questions, 37
fatty acid metabolism, beta-oxidation metabolic pathway, triheptanoin, 341
248, 249f glutamate, 349
Feinman, R. D., 378 D-glutamate, seizure onset protection, 348
fever, on inflammation, 147–148 glutathione
fever-induced refractory epileptic encephalopathy in mechanisms of action, 259, 261, 262t
school-age children (FIRES), 147 on reactive oxygen species, 264
status epilepticus, 60, 61 glycemic dietary patterns, diabetes mellitus, 365–367
fibroblast growth factor 21, 147–148 glycemic index (GI), 9, 365
First Do No Harm, 3, 41, 386, 391f glycemic load, 366
402

402 Index

glycine, seizure onset protection, 346 implementation, 26–31


glycolysis, 336, 354f classic ketogenic diet
glycolytic flux, neuroprotection, 157 caloric restriction, 29
glycolytic pathway, 2-deoxyglucose, 353, 354f fasting, 28–29
Go Lower Low Carbohydrate Diet, 367–370 liquid/formula vs. food, 29
Gottstein, U., 114 education and counseling, prediet, 27
GPR40, 199, 200 effectiveness, timing of determination, 29–30
GPR41, 201 family-centered, team-based approach, 26
GPR120, 201 initiation, in-patient vs. out-patient, 27–28
Greenwood, C. E., 231 ketogenic diet team, composition, 26–27
growth, 70 ratios, importance, 30–31
Gudsnuk, K., 233f, 235–236, 235f, 235t response predictor, 31
Guelpa, G., 16 seizure freedom, 30
treatment duration, 31
Hadera, M. G., 340 infantile spasms, 42–44
Harmon, Denon, 256 infections, 68
Hayflick, L., 256, 257 inflammasome, 138–139
HDAC inhibitor, 321 NOD-like receptor P3, 298
ionizing radiation protection, 264 inflammation, 147–153
hepatitis, 69 animal models, neurological disease, 148–150
Herbert, M. R., 105 multiple sclerosis, 148–150
HIF1α pain, 148
activation, redox state, 216 Parkinson’s disease, 150
ketones, 219 studies, 148, 149t
neuroprotection clinical evidence, 153
ketosis, 221–222, 221f fever, 147–148
stabilization, 219–221 fibroblast growth factor 21, 147–148
oxygen homeostasis, hypoxia, 220 FIRES, 147
regulation, 221 future indications, 153
hippocampal slice, 186–193 ketone supplementation, 314–316
for ketogenic diet and epilepsy studies, 187 mechanisms, 150–153, 151f
ketogenic diet–fed animal slices, 188f, 189t, 191 adenosine modulation, 151f, 152–153
ketone body direct application, caloric restriction, 151–152, 151f
187–190, 188f, 189t ketone bodies, 150–151
temporal lobe epilepsy model, 186–187 mitochondrial membrane potential, 153
whole-cell patch clamp for glucose and ATP control, polyunsaturated fatty acids, 151f, 152
188f, 189t, 190–191, 191f reactive oxygen species, 151f, 153
Hippocrates, 16, 28 uncoupling proteins, 153
Hong, A. M., 43 neuroinflammation, 297–298
Hong, S., 175 neuroprotection, 138–140
hormonal mechanisms, 227 pain, 147, 148, 196 (see also pain)
Hoyer, S., 114–115, 122 initiation, in-patient vs. out-patient, 27–28
Huntington’s disease, triheptanoin, 342 Inoue, T., 283–286, 284f, 285f
Huttenlocher, P. R., 5, 328 insulin
hydrogen peroxide, 264 blood levels, ketogenic diet, 228, 228t
synthesis, monoamine oxidase, 254–255, 255f glucose-insulin axis, 376
3-hydroxybutyrate ketone monoester (KME), hyperinsulinemia, 313–314
318–320, 319f insulin resistance
3-hydroxybutyrate methyl ester (HBME), 318 brain, Alzheimer’s disease, 243
hyperglycemia, 313–314 keto-adaptation, 377–378
hyperinsulinemia, 313–314 ketone supplementation, 321
hyperketonemia nonalcohoic fatty liver disease, 378
benefits, 307 weight loss, keto-adaptation, 379
with cognitive deficit, clinical insulin sensitivity
studies, 123–124, 125t brain ketone use, 118
hyperlipidemia, 69 ketone supplementation, 313–314
hypoglycemia, 68 International Consensus Statement for Ketogenic Diet
hypometabolism, Alzheimer’s disease, 216–217 (ICSKD), 26
hypoxia-inducible factor 1 (HIF-1), 89–90 intrauterine growth restriction (IUGR), 234
403

Index 403

ionizing radiation other applications, 392–393


HDAC inhibitor protection, 264 research support, 393–394
ketone ester effects, post-exposure, 266–267, 267f ketone-based metabolism, 282
ketosis protection, 263 ketone bodies and ketones,271. see also acetoacetate; β-
in vitro studies, 264–266, 265f, 266f hydroxybutyrate (BHB); specific types
L-isoleucine, seizure onset protection, 346 alternative energy substrates, 216–217
Alzheimer’s disease, 244–247, 245f–246f
JAMs, 292–293 available forms, 247–249, 248f, 249f
Jaworski, D. M., 80–81 on blood-brain barrier, 298–299
Jeong, E. A., 176, 177 blood levels, ketogenic diet, 228, 228t
Joensson, E. A., 370 brain, 113, 118, 119f
Joslin, Elliott P., 364, 365, 367 development and evolution, 126
Juge, N., 188–190 fuel, 113, 118, 119f, 380–381
junctional adhesion molecules (JAMs), 292–293 uptake, 295–297, 297f
juvenile myoclonic epilepsy (JME), 18, 57–58 uptake, regional, early Alzheimer’s, 121–122,
121f, 122f
kainic acid status epilepticus (KASE) model, 210, 211 breakdown, 28
Kang, H., 69 exogenous, 307
Kashiwaya, Y., 314, 316, 318, 321, 322 vs. glucose, 217
KATP channels, BAD on, 271 as “good medicine,” 81
KATP channels, metabolic seizure resistance, increased, 281
274–277, 277f on inflammation, 138–140, 150–151
BAD disruption on, 275 kinetics and transport, 118–120, 120f, 120t
metabolism and excitability link, 274–275 metabolism, BAD on, 271–272
seizure protection, BAD-elicited, 275–277, 276f neuroinflammation, 297–298
Kelly, Millicent, 387 neuroprotection, 126, 156–157, 298
Kessler, S. K., 30–31 on pain, 200
keto-adaptation, 312, 376–383 on radiation effects, post-exposure, 266–267, 267f
athlete, 382, 383f on tumors, 91
effects, 376–377 ketone ester supplementation, 312
fundamentals, 376–377 ketone salts supplementation, 311–312
health implications, 377–380 ketone supplementation, 310–322
insulin resistance, metabolic syndrome, diabetes applications, 317–322
type 2, 377–378 Alzheimer’s disease, 318–320, 319f
lipoprotein profile, 379–380 cancer, 320–321
weight loss and body composition responses, epilepsy/seizure disorders, 317
378–379 GLUT1 deficiency syndrome, 317–318
history, 376 insulin resistance/type 2 diabetes mellitus, 321
nutritional ketosis, 377, 377f weight loss, 321–322
sport and fitness, 380–381 development and testing, 310–312
starvation ketosis, 377 1,3-butanediol, 311
Keto Cookbook II, 393 formulations, 310
KetoDietCalculator, 388 ketone esters, 312
ketogenesis, 118, 119f ketone salts, 311–312
ketogenic diet. see also specific types and topics medium chain triglycerides, 310–311
history and early research, 3–4, 26 βHB mineral salts and medium chain triglycerides,
ketosis, 217 311–312
mechanisms of action, 90–91 (see also specific types) therapeutic ketosis from, 310
pediatrics indications, 4 therapeutic mechanisms, 313–316
spectrum, 389–392, 390t glucose suppression and insulin sensitivity
types, 3–4 enhancement, 313–314
use, 26 on inflammation, 314–316
ketogenic diet, advancing awareness, 386–395 metabolic efficiency, enhanced, 314, 315f
brain cancer, 392 mitochondrial health and function, 316
Charlie Foundation, 3, 27, 386–392, 391f, 392f (see oxidative stress, inhibition, 316
also Charlie Foundation) ketosis, nutritional (therapeutic), 217, 307, 310, 379.
Matthew’s Friends, 386, 388–389, 392f (see also see also ketone supplementation
Matthew’s Friends) benefits, 377, 377f
metabolic syndrome, 392 β-hydroxybutyrate, 377, 379
404

404 Index

ketosis, starvation, 377 Maurois, P., 174


kidney stones, 68 McDonald, T. S., 339
Kilaru, S., 47 medium chain fatty acids, 329f
Kim, D. Y., 190 AMPA receptor agonists, 331–332, 331t
Kimura, R., 188 seizure control, 328–330, 329f, 329t
kindling, 355 medium chain triglyceride (MCT) ketogenic diet,
Klein, P., 19 5–7, 6b, 6f
Klement, R. J., 94 history, 328
Klingenberg, M., 258 initiation and follow-up, 12–13
Kossoff, E. H., 7, 8b, 20, 30–31, 57, 389 mechanisms, 328–333
Krebs, H. A, 258–261 AMPA receptor agonism, 331–332, 331t
Kreb’s cycle, 244 implications, 332–333
Kverneland, M., 58 seizure control, 328–330, 329f, 329t
seizure control, ketones, 332
laboratory, 165–166 molecular mechanism, 328–333
lactate dehydrogenase (LDH), 281–286 medium chain triglycerides (MCTs), 5
astrocyte-neuron lactate shuttle, 282–283, 283f coconut oil and palm kernel oil, 123
epilepsy ketogenesis, dose-response, 6f, 122–123, 123f
mechanisms, 281–282 products, 123–124, 125t
metabolic target, 283–286, 284f, 285f safety, 126
future directions, 286 supplementation, 310–311
lactate fermentation, 80 supplementation, with βHB
Lambrechts, D. A., 20 mineral salts, 311–312
Lardy, Henry, 314 MELAS, status epilepticus, 61
Laux, L., 45 menstrual cycle, 23
Lemmon, M. E., 44 metabolic acidosis, 67
Lennox Gastaut syndrome, 44–45 metabolic diseases
D-leucine, 347–348 autism spectrum disorders, 102–103, 104t–105t
L-leucine, 346–347 cancer, 79–80
leukocyte adhesion molecules, metabolic potential energy, 376
blood-brain barrier, 293 metabolic remodeling, 88
lipids, 23 metabolic syndrome
lipogenesis, 379 keto-adaptation, 378
lipoprotein profile, keto-adaptation, 379–380 ketogenic therapy, 392
liquid/formula vs. food, classic ketogenic diet, 29 plasma markers, ketogenic diet, 229, 230t
Liu, Christiana, 5 metabolism. see also specific types
Lomax, M. E., 265–266 ketogenic diet mechanisms, 227
long chain fatty acid oxidation disorders, ketone supplementation, 314, 315f
triheptanoin, 341 metabolism, mitochondrial
long-chain triglycerides, 5 acetoacetate, 315f
low glycemic index treatment (LGIT), 6f, 9–10, 9b D-β-hydroxybutyrate, 315f
development, 389 D-methionine, seizure onset protection, 348
initiation and follow-up, 12–13 middle cerebral artery occlusion, triheptanoin, 341
Luna-Medina, R., 175 migraine, 159–160
Lying-Tunell, U., 114, 115, 115t, 121f, 122 mild cognitive impairment
ketogenic treatments, 127, 127t
Magee, B. A., 320 medium chain triglycerides, 123f, 124, 125t
Magrath, G., 9 mitochondrial dysfunction
Mantis, J. G., 106 autism spectrum disorders, 102–103
Marie, A., 16 brain energy status and neuroprotection, 125–126
Marin-Valencia, I., 339 cancer, 79–80
Martinez, C. C., 17 cytopathies, 27
Matthew’s Friends, 27, 392f, 393f function, 102
achievements, 388–389 ketone supplementation, 316
future, 394–395 membrane potential, inflammation, 153
genesis, 387–388 nervous system, 102
ketogenic diet therapy spectrum, mitochondrial metabolism
389–392, 390t acetoacetate, 315f
research support, 393–394 D-β-hydroxybutyrate, 315f
405

Index 405

modified Atkins diet (MAD), 6f, 7–9, 8b, 8t neuroprotection, mechanisms, 216, 221–222, 221f
development, 389 anaplerotic, 136
effectiveness, vs. classical ketogenic diet, 10–11, 10t anti-excitotoxic, 137
efficacy, 19–22, 20f, 21t antioxidant, 137–138
initiation and follow-up, 12–13 inflammation, 138–140
mood, 22 non-anaplerotic, 137
mood disorders, 159 neurovascular unit, 289, 290f
Moorman, M. A., 256, 257 Newport, M. T., 318, 320
Moreno, S., 170 NF E2-related factor 2(Nrf2) transcription factor, 138,
multiple sclerosis, 148–150 172, 221, 266
myoclonic astatic epilepsy (MAE), 35, 46–47 Nielsen, J. V., 370
Nizamuddin, J., 69
Nabbout, R., 45, 60, 61, 63 NLRP3 inflammasome, 138–139, 148, 149t, 150, 151f,
NADPH, 264 153, 197, 298, 315–316
NADP-linked enzymes, liver activity, 259, 259t NOD-like receptor P3 (NLRP3) inflammasome, 138–
NADP system, 261–263, 262f, 263f 139, 148, 149t, 150, 151f, 153, 197, 298, 315–316
D-β-hydroxybutyrate on, 264 nonalcohoic fatty liver disease, insulin resistance, 378
Neal, E. G., 3, 4, 10, 10t nonconvulsive status epilepticus, 61–62
Nebeling, L. C., 83, 93 nonsurgical epilepsy, refractory, 40–42
Nei, M., 20 Nordli, D. R., 43
neurodegeneration and aging, 216–222. see also aging NORSE, status epilepticus, 60, 61
and neurodegeneration Nrf2, 138, 172, 221, 266
neuroinflammation, 297–298 nuclear factor-kappa B, 90
neurological disorders, 156–160. see also Numis, A. L., 43–44
specific types nutritional ketosis. see ketosis, nutritional
amino acid treatment, 346–349 (see also amino acids, Nylen, K., 106
for neurological disorders)
amyotrophic lateral sclerosis, 157–158 obesity, 362–371
glycolytic flux, 157 carbohydrate restrictions, randomized controlled
ketone bodies, 156–157 trials, 363, 363t
migraine, 159–160 diabetes mellitus type 2, 362 (see also diabetes mellitus
mood disorders, 159 type 2)
neuroprotection, 156–157 ketogenic diet
Parkinson disease, 158–159 origins and use, 363
neuron restrictive silencing factor (NRSF), 356 pilot study, 367–370
neurons, BAD-altered, metabolic changes, 272 prevalence, 362
neuropathic pain, 196. see also pain occludin, 292–293
neuroprotection, 156–157, 298, 376 Ogawa, M., 115, 121f, 122
acetoacetate, 136 Oguni, H., 46
aging, 218–219, 219f Olovnik, A. M., 256
aging, ketosis, 221–222, 221f oncogenic paradox, 80
antioxidants, 137–138 orexigenic food intake, 229, 229f
β-hydroxybutyrate, 124, 124f, 125t, 134 osteopenia, 70
D-β-hydroxybutyrate, 134 overweight, prevalence, 362
cardiac arrest and resuscitation, 218–219, 219f Owen, O. E., 114, 118, 121f, 376
erythropoietin, 220 oxidative phosphorylation, 80
fasting, 134 oxidative stress
glycolytic flux, 157 on glucose metabolism, 217–218, 218f
HIF1α, 219–222, 221f ketone supplementation, 316
inflammation, 138–140 oxygen free radicals. see also reactive oxygen
ketone bodies, 156–157, 298 species (ROS)
ketones, 126 redox state, 261, 262t
neurological disorders, 156–157
Parkinson’s disease, 158–159 pain, 196–201
polyunsaturated fatty acids, 156–157 adenosine transmission, 201
PPARγ, post-SE and during SRS, 171, 172f calorie restriction, 197–198
spinal cord injury, drug candidates, 133–134 economic cost, 196
stroke, 218–219, 219f fasting, 196–197
vascular endothelial growth factor, 220–221 glucose, 200–201
406

406 Index

pain, (cont.) pregnancy


GPR40, 199, 200 establishment and maintenance, 233–234
GPR41, 201 before and during gestation, 233f, 234–236, 235f,
GPR120, 201 235t, 236f
inflammation, 147, 148, 196 Pre-ketogenic diet, 394
inflammatory, 196 presenilin 1/2 genes, 241
ketogenic diet, 198–199, 198f, 199f Prins, M. L., 298–299
ketone bodies, 200 D-proline, seizure onset protection, 348
neuropathic, 196 prolyl-hydroxylase (PHD), 220, 221, 221f
polyunsaturated fatty acids, 200 propionyl-CoA carboxylation pathway, anaplerosis,
quality of life, 196 336–337, 338f
severe, treatment, 196 puberty, ketogenic diet long-term effects, 231–234, 232f,
pancreatitis, 69 232t, 233f
Paoli, A., 228–229, 228t, 229f, 236–237 pyruvate decarboxylase deficiency, triheptanoin, 341
Parent’s Guide to the Ketogenic Diet, 393 pyruvate dehydrogenase
Parkinson’s disease deficiency, 50
on inflammation, animal models, 150 inhibition, Alzheimer’s disease, 242, 243
neuroprotection, 158–159
Pasteur effect, 80 QT interval prolongation, 69
Patel, S. P., 136
pentose phosphate shunt (PPP), 354f, 357 radiation, ionizing
pentylenetetrazole (PTZ), 317, 346 HDAC inhibitor protection, 264
pericytes, blood-brain barrier, 290f, 292 ketone ester effects, post-exposure, 266–267, 267f
peroxisome proliferator-activated receptors (PPARs) ketosis protection, 263, 264f
historical overview, 167 in vitro studies, 264–266, 265f, 266f
isoforms and domains, 168, 169f radiation therapy, cancer, 92
polyunsaturated fatty acids, 200 Raju, K. N., 30
structure and functional diversity, 167–168 Rasmussen syndrome, status epilepticus, 61
peroxisome proliferator-activated receptor γ (PPARγ), 7, ratios, importance, 30–31
137, 151, 151f, 152, 157, 167–179 reactive nitrogen species (RNS), 92, 254–268
activation, 168 damage caused by, 255–256, 255f, 256f
agonists, 174 origins, reactions, and target molecules, 254–255,
neuroprotection post-SE and during SRS, 175 255f, 256t
pretreatment, acute seizure models, 174 reactive oxygen species (ROS), 254–268
on seizure thresholds and kindling models, 174–175 in aging, 256–257
on SRS and spatial learning, 175 Alzheimer’s disease, 243–244
brain, 170–172, 172f D-β-hydroxybutyrate
epilepsy, 173–174 on cellular redox states, 261–263, 262f, 263f
functional consequences, 177–178 on transcription, 261t, 264
gene regulation and activity, 169–170, 169f cancer, 90
ketogenic diet cellular redox states vs. standard redox potentials,
activation, evidence, 175–176 258–261, 259f, 259t, 261t, 262t
activation, potential mechanisms, 176–177 damage, 255–256, 255f, 256f
regulation, 173–177 enzymes destroying, 264
mechanisms of action, 178–179, 179f on inflammation, 151f, 153
neuroprotection, homeostatic, 171, 172f mechanisms of action, 257–258, 258f
splice variants and activity, 168–169, 169f origins, reactions, and target molecules, 254–255,
structure and domains, 168, 169f 255f, 256t
Pfeifer, Heidi, 389 redox state of oxygen free radicals and other reactants,
Phinney, S. D., 379, 382 261, 262t
PI3K/AKT signaling pathway, 89 removal, in vivo, 263–264
Pires, M. E., 43 removal, NADPH generation, 264
plasmalemma vesicle-associated protein number, 293 redox potentials, standard, vs. cellular redox states,
polycystic ovary syndrome, 117 258–261, 259f, 259t, 261t, 262t
polyunsaturated fatty acids (PUFAs) redox states, cellular
on inflammation, 151f, 152 D-β-hydroxybutyrate on, 261–263, 262f, 263f
neuroprotection, 156–157 oxygen free radicals and other reactants, 261, 262t
pain, 200 vs. standard redox potentials, 258–261, 259f, 259t,
peroxisome proliferator-activated receptors, 200 261t, 262t
407

Index 407

refractory epilepsy, 18 growth, 70


refractory nonsurgical epilepsy, 40–42 hepatitis, 69
renal calculi, 68 hyperlipidemia, 69
reproduction, 227–237 hypoglycemia, 68
behavioral, 231 infections, 68
endocrine, 227–230, 228t, 229f, 230f, 230t kidney stones, 68
high-fat/ketogenic diets, 227 osteopenia, 70
long-term effects, 233–234 pancreatitis, 69
pregnancy vitamin deficiencies, 68–69
establishment and maintenance, 233–234 vomiting, 67
before and during gestation, 233f, 234–236, 235f, Simard-Tremblay, S., 47
235t, 236f Slc2a1, 293
puberty, 231–233, 232f, 232t, 233f social screening, 66–67
reproduction, 233–234 somatic mutation theory, 79
response predictor, 31 Spilioti, M., 105
resting energy expenditure (REE), 29 spinal cord injury, 133–141
Rett syndrome, 50–51, 106 epidemiology, 133
triheptanoin, 342 inflammation, 138–140
Richardson, R., 364 intermittent fasting, 134
Rouach, N., 282–283 ketogenic diet, mechanisms
Ruskin, D. N., 106 anaplerotic, 136
anti-excitotoxic, 137
Sada, N., 283–286, 284f, 285f antioxidant, 137–138
S-adenosylhomocysteine (SAH) hydrolase, 212, 212f non-anaplerotic, 137
S-adenosylmethionine (SAM) dependent ketogenic diet, outcome, 134–135
transmethylation pathway, 211–212, 212f ketogenic diet, translation to
Samoilova, M., 188 clinical setting, 140–141
Sansum, W. D., 364 management, acute, 133
Sarruf, D. A., 170 neuroprotective drug candidates, 133–134
Scheck, A. C., 83, 91–92, 92f rehabilitation, 133
Schmid, W., 267 spontaneous recurrent seizures, PPARγ
Schoeler, N. E., 41 agonists on, 175
Schrödinger, Erwin, 257 sport, keto-adaptation, 380–381
SCN1A gene mutations, 45 Stafstrom, C. E., 192
screening, pre-treatment, 66–67, 67t, 71t status epilepticus (SE), 60–63
medical, 66 cause, 60
social, 66–67 challenges, 61t, 62–63
seizure. see also epilepsy convulsive, 61
fasting, 209 duration, 60
freedom from, 30 efficacy, ketogenic diet, 60, 61t
ketone supplementation, 317 implementation, 62, 62f
seizure control. see also epileptogenesis actions incidence, 60
ketones, 332 nonconvulsive, 61–62
medium chain fatty acids, 328–330, 329f, 329t super-refractory, 60
senescence, cell, 257 treatment, first-line, 60
D-serine, seizure onset protection, 348 types, 60
L-serine stroke
seizure onset protection, 347 neuroprotection, 218–219, 219f
traumatic brain injury, 349 triheptanoin, 341
severe myoclonic epilepsy of infancy (SMEI), 45–46 Sturge-Weber syndrome, 53
Seyfried, T. N., 80 succinic semialdehyde dehydrogenase (SSADH)
Sharma, S., 3, 10, 10t deficiency, 105–106
Shimazu, T., 81 sugar, dietary, children, 393
side effects, 66–71 superoxide dismutase (MnSOD), 138, 264
acidosis, 67 oxidative stress, 172f
cardiac, 69–70 PPARγ deficiency on, 172
constipation, 67–68 reactive oxygen species, 254–255, 255f, 256t, 264
electrolyte imbalances, 69 super-refractory status epilepticus (SRSE), 18–19, 60
food refusal, 70 Sussman, D., 234–235
408

408 Index

Tanner, G. R., 190 neurological conditions, 341–342


Taub, K. S., 30 pyruvate decarboxylase deficiency, 341
TCA cycle, 336, 337f, 338f tuberous sclerosis complex, 51–52
team-based approach tumor metabolism, 88–90, 89f
family-centered, 26
team composition, 26–27 uncoupling proteins, on inflammation, 153
telomere shortening, 256–257 unlimited protein diet. see modified Atkins diet (MAD)
temporal lobe epilepsy
hippocampus model, 186–187 vascular endothelial growth factor (VEGF),
ketogenic diet and adenosine homeostasis, neuroprotection, 220–221
211–213, 212f Veech, R. L., 81, 258–260, 259t, 307, 314, 318
Thakur, K. T., 19 Verdin, ERic, 316
therapeutic ketosis, 310. see also vesicular glutamate transporters (VGLUTs), 190,
ketone supplementation 193, 286
Thio, L. L., 188 acetoacetate on, 281
thioredoxin, 259, 259f, 261 functions, 281
tight junctions, blood-brain ketogenic diet, 282
barrier, 290f, 292–293 Viggiano, A., 317
tocopherol, 258, 258f Villeneuve, N., 61
transcription, D-β-hydroxybutyrate on, 261t, 264 Vining, E., 70
transmethylation pathway, 211, 212f vitamin deficiencies, 68–69
transporters. see also specific types Volek, J. S., 378, 379–380, 382, 383f
blood-brain barrier, 290f, 291f, 293, 296f vomiting, 67
traumatic brain injury, 133–141, 298
amino acid treatment, 349 Warburg, Otto, 88
on inflammation, 138–140 Warburg effect, 80, 82, 88–90, 89f
ketogenic diet, translation to Warburg’s theory, 79–80
clinical setting, 140–141 weight loss, 22
ketogenic diet effects, 135–136 keto-adaptation, 378–379
ketogenic diet mechanisms ketone supplementation, 321–322
anaplerotic, 136 Westman, E. C., 367–370, 370t
anti-excitotoxic, 137 Wilder, R. M., 16, 26, 165
antioxidant, 137–138 Williams, Matthew, 386, 387–388, 393f
non-anaplerotic, 137 Williams, S., 70
triglycerides, 5 Williams-Karnesky, R. L., 210, 211–212
triheptanoin, 317–318, 336–342 Willis, S., 340
anaplerosis, 336–337, 337f, 338f Winocur, G., 231
anticonvulsant effects, 338f, 340 Wirrell, E. C., 28
brain energy metabolism, 339–340 Worden, L. T., 71
cardiac hypertrophy, 341
epilepsy, energy metabolism, Yudkin, John, 363
337–339, 338f
GLUT1 deficiency, 341 zona occludin proteins, 293
long chain fatty acid oxidation disorders, 341 Zuccoli, G., 93
muscle disorders, 342 Zupec-Kania, Beth, 388, 393

You might also like