You are on page 1of 18

Pediatric Diabetes 2014: 15(Suppl. 20): 4764 2014 John Wiley & Sons A/S.

doi: 10.1111/pedi.12192 Published by John Wiley & Sons Ltd.


All rights reserved
Pediatric Diabetes

ISPAD Clinical Practice Consensus Guidelines 2014 Compendium

The diagnosis and management of monogenic


diabetes in children and adolescents

Rubio-Cabezas O, Hattersley AT, Njlstad PR, Australia and i School of Womens and Childrens Health,
Mlynarski W, Ellard S, White N, Chi DV, Craig ME. University of New South Wales, Sydney, Australia
The diagnosis and management of monogenic diabetes in
Key words: child genetic MODY monogenic diabetes
children and adolescents. neonatal diabetes
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764.
Corresponding author:
Maria E Craig,
Oscar Rubio-Cabezasa , Andrew T Hattersleyb , The Childrens Hospital at Westmead and Discipline of
R Njlstadc,d , Wojciech Mlynarskie , Sian
Pal Pediatrics and Child Health,
University of Sydney,
Ellardb , Neil Whitef , Dung Vu Chig and Maria E
Gray St, Kogarah,
Craigh,i Sydney, New South Wales,
a
Department of Paediatric Endocrinology, Hospital Infantil Australia.
Madrid, Spain; b Institute of
Jesus,
Universitario Nino e-mail: m.craig@unsw.edu.au
Biomedical and Clinical Sciences, University of Exeter Medical
School, Exeter, UK; c Department of Clinical Science, University Editors of the ISPAD Clinical Practice Consensus Guidelines
of Bergen, Bergen, Norway; d Department of Pediatrics, 2014 Compendium: Carlo Acerini, Carine de Beaufort, Maria
Haukeland University Hospital, Bergen, Norway; e Department Craig, David Maahs, Ragnar Hanas.
of Pediatrics, Oncology, Hematology and Diabetology, Medical
University of Lodz, Lodz, Poland; f Division of Pediatric This article is a chapter in the ISPAD Clinical Practice Consensus
Endocrinology and Metabolism, Department of Pediatrics, Guidelines 2014 Compendium. The complete set of guidelines
Washington University School of Medicine, St Louis Childrens can be found for free download at www.ispad.org. The evidence
Hospital, St. Louis, MO, USA; g Department of Pediatric grading system used in the ISPAD Guidelines is the same as that
Endocrinology, National Hospital for Pediatrics, Hanoi, used by the American Diabetes Association. See page 3 (the
Vietnam; h The Childrens Hospital at Westmead and Discipline Introduction in Pediatric Diabetes 2014; 15 (Suppl. 20): 1-3).
of Pediatrics and Child Health, University of Sydney, Sydney,

Executive summary and Recommendations neonatal diabetes mellitus (TNDM), which will
resolve but may later relapse. In addition the
diagnosis will inform other likely features, e. g.,
Monogenic diabetes is uncommon, accounting for pancreatic exocrine failure and developmental
14% of pediatric diabetes cases (B).
delay (B).
All patients diagnosed with diabetes in the first
The diagnosis of maturity-onset diabetes of the
6 months of life should have immediate molecular
young (MODY) should be suspected in cases with:
genetic testing to define their subtype of monogenic
neonatal diabetes mellitus (NDM), as type 1 diabetes
is extremely rare in this subgroup (B). In patients
A family history of diabetes in one parent and
diagnosed between 6 and 12 months of age, testing
for NDM should be limited to those without islet first degree relatives of that affected parent in
antibodies as the majority of patients in this age patients who lack the characteristics of type 1 dia-
group have type 1 diabetes (B). betes [no islet autoantibodies, low or no insulin
The molecular genetic diagnosis of NDM will give requirements 5 yr after diagnosis (stimulated
information on which patients have a potassium C-peptide >200 pmol/L)] and lack the character-
channel mutation and can be treated with high istics type 2 diabetes (marked obesity, acanthosis
dose sulfonylureas and which patients have transient nigricans).
47
Rubio-Cabezas et al.

Mild stable fasting hyperglycemia which does caused by dominantly acting heterozygous mutations
not progress. Such cases should be tested for in genes important for the development or function
glucokinase (GCK) gene mutations, which is of cells (1, 5). Over the last few years, however, a
the commonest cause of persistent, incidental number of forms of monogenic diabetes clinically and
hyperglycemia in the pediatric population (B). genetically different from MODY have been identified
(6). Some patients harbor dominant mutations arising
Specific features can suggest subtypes of MODY, de novo (i.e., not inherited from parents) so family
such as renal developmental disease or renal cysts history suggesting a monogenic condition is lacking
(HNF1B-MODY) and macrosomia and/or neonatal (79). These facts, along with a widespread lack of
hypoglycemia (HNF4A-MODY) (C). awareness, hinder clinical diagnosis so that the majority
In familial autosomal dominant symptomatic of children with genetically proven monogenic diabetes
diabetes, mutations in the hepatocyte nuclear factor are initially misdiagnosed as having type 1 (1012)
1 (HNF1A) gene (HNF1A-MODY) should be or, less commonly, type 2 diabetes (13). Although
considered as the first diagnostic possibility, while monogenic diabetes is uncommon, it accounts for
mutations in the GCK gene are the most common 14% of pediatric diabetes cases (1416).
cause in the absence of symptoms or marked
hyperglycemia (B).
Results of genetic testing should be reported and Clinical relevance of diagnosing monogenic
presented to families in a clear and unambiguous diabetes
manner, as results may have a major effect on clinical
Identification of children with monogenic diabetes
management (E).
usually improves their clinical care. Making a
Referral to a specialist in monogenic diabetes or
specific molecular diagnosis helps predict the expected
an interested clinical genetics unit is recommended
clinical course of the disease and guide the most
when predictive testing of asymptomatic individuals
appropriate management in a particular patient,
is requested (E).
including pharmacological treatment. Furthermore, it
Some forms of MODY diabetes are sensitive to sul-
has important implications for the family as it enables
fonylureas, such as HNF1A-MODY and HNF4A-
genetic counseling and frequently triggers extended
MODY (B).
genetic testing in other diabetic family members, whose
Mild fasting hyperglycemia due to GCK-MODY is
diabetes may eventually be reclassified.
not progressive during childhood; patients do not
develop complications (B) and do not respond to
low dose insulin or oral agents (C), so should not Selecting candidates for molecular testing
receive treatment.
In contrast to type 1 and type 2 diabetes, where there
is no single diagnostic test, molecular genetic testing is
Introduction both sensitive and specific for diagnosing monogenic
Monogenic diabetes results from one or more defects diabetes. Genetic testing is currently available in
in a single gene. The disease may be inherited within many countries around the world and should be
families as a dominant, recessive, or non-Mendelian strongly considered in patients with suspected mono-
trait or may present as a spontaneous case due to a de genic diabetes (see below). Appropriate informed
novo mutation. Well over 40 different genetic subtypes consent/assent must be prospectively obtained from
of monogenic diabetes have been identified to date, the patient and his/her legal guardians. Genetic
each having a typical phenotype and a specific pattern testing for some conditions is available free of charge
of inheritance. on a research basis in certain academic institu-
A familial form of mild diabetes presenting during tions (e.g., www.diabetesgenes.org, http://monogenic
adolescence or in early adulthood was first described diabetes.uchicago.edu, http://www.pediatria.umed.pl/
many years ago (1, 2). Even though diabetes presented team/en/contact, www.mody.no, and http://www.
in young patients, the disease clinically resembled euro-wabb.org/en/european-genetic-diagnostic-
elderly onset non-insulin dependent diabetes and the laboratories).
newly recognized subtype of familial diabetes became Next-generation sequencing enables the simultane-
known by the acronym MODY (3). As MODY patients ous analysis of multiple genes at a lower cost and may
passed on the disease to their offspring following an become a feasible alternative to traditional genetic
autosomal dominant pattern of inheritance, it was testing in the near future (1720). In the meantime, a
quickly suspected that it might be a monogenic disorder judicious approach to selecting candidates for molecu-
(4). MODY is by far the commonest type of monogenic lar testing is required. The simplest way of maximizing
diabetes. All currently known subtypes of MODY are the cost-effectiveness of traditional genetic testing is by
48 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

increasing its positive yield through a reasoned selec- to ensure that both clinicians and patients receive
tion of the appropriate gene(s) for analysis according to adequate and understandable information. Specific
the patients clinical, immunological, and/or biochem- recommendations describing the information that
ical phenotype (21, 22). This process may be relatively should be included in the molecular genetics laboratory
easy to undertake when clinical features directly point- report for MODY testing have been published (25).
ing to a specific syndrome are present, but results may These include the method used for mutation screening,
be very difficult to achieve when diabetes is the only whether the mutation is novel and if so, evidence for its
manifestation of the monogenic disorder. pathogenicity, and information about the likelihood of
the disease being inherited by the offspring. Referral to
a specialist unit (diabetes genetics or clinical genetics) is
When to suspect a diagnosis of type 1 diabetes
recommended when predictive testing of asymptomatic
in children may not be correct?
individuals is requested.
Features in children initially thought to have type 1
diabetes that suggest a possible diagnosis of monogenic
Specific subtypes of monogenic diabetes
diabetes are shown below. Except for age of diagnosis
and their management
less than 6 months, none of these are pathognomonic
and should be considered together rather than in The different forms of monogenic diabetes can
isolation: be classified according to the main pathogenic
mechanism into two separate groups (26): genetic
1 Diabetes presenting before 6 months of age as defects of insulin secretion and genetic defects of
type 1 diabetes is extremely rare in this age- insulin action. In children, the majority of cases
group (2, 23). result from mutations in genes causing cell loss or
2 Family history of diabetes in one parent and other dysfunction although diabetes can rarely occur from
first degree relatives of that affected parent. mutations resulting in very severe insulin resistance.
3 Absence of islet autoantibodies, especially if From a clinical perspective, clinical scenarios when a
measured at diagnosis. diagnosis of monogenic diabetes should be considered
4 Preserved -cell function, with low insulin require- include:
ments and detectable C-peptide (either in blood or
urine) over an extended partial remission phase (5 yr 1 Diabetes presenting before 6 months of age (NDM).
after diagnosis). 2 Autosomal dominant familial mild hyperglycemia
or diabetes.
3 Diabetes associated with extrapancreatic features.
When to suspect a diagnosis of type 2 diabetes
4 Monogenic insulin resistance syndromes.
in children may not be correct?
In young people, type 2 diabetes often presents around
NDM diabetes diagnosed within the first
puberty and the majority are obese. A number of
612 months of life
features that should suggest monogenic diabetes are
listed below: The clinical presentation of autoimmune type 1 dia-
betes is exceedingly rare before age 6 months (23, 27).
1 Absence of severe obesity. Even though autoantibodies against -cell antigens
2 Lack of acanthosis nigricans and/or other markers may be occasionally found in very young diabetic
of metabolic syndrome. infants (23), it is now accepted that FOXP3 mutations,
3 Ethnic background with a low prevalence of type 2 and not type 1 diabetes, will account for most of these
diabetes, e.g., European Caucasian. cases (28). Therefore, all patients diagnosed under
4 Strong family history of diabetes without obesity. 6 months should have genetic testing for monogenic
NDM. Some cases of monogenic diabetes can be diag-
nosed between 6 and 12 months (12, 29, 30) although
Interpretation of genetic findings
the vast majority of these patients have type 1 diabetes.
Despite the obvious clinical benefits derived from an Many patients with NDM are born small for
increased awareness and more widely available genetic gestational age, which reflects a prenatal deficiency
diagnostic services, care needs to be exercised in the of insulin secretion as insulin exerts potent growth-
interpretation of genetic findings (24). The way the promoting effects during intrauterine development
clinician interprets the genetic report will have a major (31). Approximately half will require lifelong treatment
effect on the further clinical management of the patient to control hyperglycemia - permanent neonatal
and his/her family. Therefore, it is crucial that the diabetes mellitus (PNDM). In the remaining cases,
results are presented in a clear and unambiguous way diabetes will remit within a few weeks or months
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 49
Rubio-Cabezas et al.

- transient neonatal diabetes mellitus (TNDM), not require any treatment by a median age of 12 wk.
although it might relapse later in life. In both One third of patients show macroglossia and, more
situations, diabetes presents more frequently as an rarely, an umbilical hernia is present. During remission,
isolated condition, but some patients show a variety of transient hyperglycemia may occur during intercurrent
associated extra-pancreatic clinical features pointing to illnesses (45). Over time, diabetes relapses in at least
a particular gene, which may help guide genetic testing 5060% of patients, usually around puberty, although
(Table 1). recurrences have been reported as young as 4 yr of age.
The genetic basis of TNDM has been mostly Relapse clinically resembles early-onset type 2 diabetes
uncovered: approximately two thirds of cases are and is characterized by a loss of the first-phase insulin
caused by abnormalities in an imprinted region on secretion. Insulin therapy is not always necessary (there
chromosome 6q24 (32, 33), with activating mutations is usually some response to oral sulfonylureas) and, if
in either of the genes encoding the two subunits of needed, insulin doses required tend to be lower than in
the ATP-sensitive potassium (KATP ) channel of the patients with type 1 diabetes.
-cell membrane (KCNJ11 or ABCC8) causing the The phases described above do not present irreme-
majority of the remaining cases (34). A minority of diably in every patient. Interestingly, some mutation
cases of TNDM is caused by mutations in other genes, carrier relatives develop type 2 diabetes or gestational
including HNF1B (35), INS (preproinsulin gene) (36), diabetes in adulthood without any evidences of having
etc. In contrast, the genetic abnormality responsible for had neonatal diabetes, suggesting that other genetic or
up to 30% of PNDM cases remains unknown, although epigenetic factors may influence the clinical expression
the commonest known cause in outbred populations alterations of chromosome 6q24 (32).
are mutations in the KATP channel or INS genes (37, The role of genetic counseling depends on the
38). If parents are related, WolcottRallison syndrome underlying molecular mechanism. Uniparental disomy
or homozygous mutations in the GCK gene are the of chromosome 6 is generally sporadic and therefore
most common etiologies (37). the risk of recurrence in siblings and offspring is low.
When paternal duplication of the 6q24 region is found,
males have a 50% chance of transmitting the mutation
Transient neonatal diabetes from imprinting
and the disease to their children. In contrast, females
anomalies on 6q24
will pass on the duplication but their children will not
Anomalies at the 6q24 locus, spanning two candidate develop the disease. In this case, TNDM may recur
genes PLAGL1 and HYMAI, are the single most in the next generation as their asymptomatic sons
common cause of neonatal diabetes and always result pass on the molecular defect to their own children.
in TNDM (39). In normal circumstances, this region is Some methylation defects (i.e., ZFP57 mutations)
maternally imprinted so that only the allele inherited show an autosomal recessive inheritance and hence
from the father is expressed. TNDM is ultimately the recurrence risk is 25% for siblings and almost
associated with overexpression of the imprinted genes negligible for the offspring of a patient.
(40), with three different molecular mechanisms
identified to date: paternal uniparental disomy of
Neonatal diabetes due to mutations in the KATP
chromosome 6 (either complete or partial; it accounts
channel genes
for 50% of sporadic TNDM cases), unbalanced
paternal duplication of 6q24 (found in most familial KATP channels are hetero-octameric complexes formed
cases), and abnormal methylation of the maternal by four pore-forming Kir6.2 subunits and four SUR1
allele (found in some sporadic cases) (41). Methylation regulatory subunits, encoded by the genes KCNJ11
defects may affect only the 6q24 locus or may arise and ABCC8, respectively (46). They regulate insulin
in the context of a generalized hypomethylation secretion by linking intracellular metabolic state to
syndrome along with other clinical features including the -cell membrane electrical activity. Any increase in
congenital heart defects, brain malformations, etc. the intracellular metabolic activity induces an increase
(42). Some cases of TNDM secondary to multiple in the ATP/ADP ratio within the pancreatic -cell
methylation defects are caused by recessively acting which makes the KATP channels close, and leads to the
mutations in ZFP57, a gene on chromosome 6p cell membrane depolarization which ultimately triggers
involved in the regulation of DNA methylation (43). insulin secretion (47). Activating mutations in KCNJ11
Patients with 6q24 abnormalities are born with or ABCC8, which prevent KATP channel closure and
severe intrauterine growth retardation and develop hence insulin secretion in response to hyperglycemia,
severe but non-ketotic hyperglycemia very early on, are the most common cause of PNDM (7, 4851) and
usually during the first week of life (41, 44). Despite the the second most common cause of TNDM (34).
severity of the initial presentation, the insulin dose can The majority of patients with mutations in KCNJ11
be tapered quickly so that the majority of patients do have PNDM rather than TNDM (90 vs. 10%). In
50 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

Table 1. Monogenic subtypes of neonatal and infancy-onset diabetes (modified from reference 37)
Gene Locus Inheritance Other clinical features References

Abnormal pancreatic development


PLAGL1/HYMAI 6q24 Variable TNDM macroglossia umbilical hernia (33)
(imprinting)
ZFP57 6p22.1 Recessive TNDM (multiple hypomethylation syn- (43)
drome) macroglossia developmental
delay umbilical defects congenital heart
disease
PDX1 13q12.1 Recessive PNDM + pancreatic agenesis (steatorrhea) (173)
PTF1A 10p12.2 Recessive PNDM + pancreatic agenesis (174)
(steatorrhea) + cerebellar
hypoplasia/aplasia + central respiratory
dysfunction
PTF1A enhancer 10p12.2 Recessive PNDM + pancreatic agenesis without CNS (89)
features
HNF1B 17q21.3 Dominant TNDM + pancreatic hypoplasia and renal (35)
cysts
RFX6 6q22.1 Recessive PNDM + intestinal atresia + gall bladder (175)
agenesis
GATA6 18q11.1-q11.2 Dominant PNDM + pancreatic agenesis + congenital (90)
heart defects + biliary abnormalities
GATA4 8p23.1 Dominant PNDM + pancreatic agenesis + congenital (176)
heart defects
GLIS3 9p24.3-p23 Recessive PNDM + congenital (177)
hypothyroidism + glaucoma + hepatic
fibrosis + renal cysts
NEUROG3 10q21.3 Recessive PNDM + enteric anendocrinosis (178)
(malabsorptive diarrhea)
NEUROD1 2q32 Recessive PNDM + cerebellar hypoplasia + visual (179)
impairment + deafness
PAX6 11p13 Recessive PNDM + microphthalmia + brain (180)
malformations
MNX1 7q36.3 Recessive PNDM + developmental delay + sacral (181) (175)
agenesis + imperforate anus
NKX2-2 20p11.22 Recessive PNDM + developmental (182)
delay + hypotonia + short
stature + deafness + constipation
Abnormal -cell function
KCNJ11 11p15.1 Spontaneous or PNDM/TNDM DEND (7)
dominant
ABCC8 11p15.1 Spontaneous, TNDM/PNDM DEND (48)
dominant or
recessive
INS 11p15.5 Recessive Isolated PNDM or TNDM (36)
GCK 7p15-p13 Recessive Isolated PNDM (83)
SLC2A2 (GLUT2) 3q26.1-q26.3 Recessive FanconiBickel syndrome: (183)
PNDM + hypergalactosemia, liver
dysfunction
SLC19A2 1q23.3 Recessive Rogers syndrome: (184)
PNDM + thiamine-responsive
megaloblastic anemia, sensorineural
deafness
Destruction of cells
INS 11p15.5 Spontaneous or Isolated PNDM (9)
dominant
EIF2AK3 2p11.2 Recessive WolcottRallison syndrome: (77)
PNDM + skeletal dysplasia + recurrent liver
dysfunction
IER3IP1 18q21.2 Recessive PNDM + microcephaly + lissencephaly + epileptic (185)
encephalopathy
FOXP3 Xp11.23-p13.3 X-linked, IPEX syndrome (autoimmune enteropathy, (186)
recessive eczema, autoimmune hypothyroidism, and
elevated IgE)
WFS1 4p16.1 Recessive PNDM*+ optic atrophy diabetes (126)
insipidus deafness

CNS, central nervous system; DEND, developmental delay, epilepsy, and neonatal diabetes syndrome; IgE, immunoglobulin; IPEX, immune
dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome; TNDM; transient neonatal diabetes mellitus.
*The mean age of diagnosis among patients with WFS1 mutations is approximately 5 yr (129).

Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 51


Rubio-Cabezas et al.

contrast, mutations in ABCC8 cause TNDM more these mutations arise de novo, there is usually no fam-
frequently (66%) (48, 52). There are no significant dif- ily history of neonatal diabetes (71) but familial cases
ferences between the two subtypes of neonatal diabetes show an autosomal dominant inheritance. Recurrence
regarding the severity of intrauterine growth retarda- risk for the offspring of an affected patient is 50%. This
tion or the age at diagnosis of diabetes (34, 53). Patients is also true for most patients with activating mutations
with KATP channel mutations typically show milder in ABCC8. However, some patients are homozygous
intrauterine growth retardation and are diagnosed or compound heterozygous for two different mutations
slightly later than patients with 6q24 abnormalities, and neonatal diabetes is recessively inherited (49). In
indicating a less severe insulin deficiency during the this case, the risk of neonatal diabetes for future siblings
last months of intrauterine development and at the time is 25% but almost inexistent for the offspring. Germline
birth. In KATP -TNDM patients, diabetes usually remits mosaicism (mutations present in the gonads but not
later and relapses earlier than in 6q24-TNDM (34). detectable in blood) has been reported in several fam-
Presenting clinical features in patients with KATP ilies (71) and hence unaffected parents of a child with
channel activating mutations suggest insulin depen- an apparently de novo mutation should be advised that
dency, with low or undetectable C-peptide levels and the recurrence risk in siblings is low but not negligible.
frequent presentation with diabetic ketoacidosis (54).
In addition to diabetes, about 20% of patients with Neonatal diabetes due to mutations in INS gene
mutations in KCNJ11 were initially found to present Heterozygous coding mutations in the INS gene
with associated neurological features (7, 54, 55) in are the second most common cause of PNDM after
keeping with the expression of KATP channels in neu- KATP channel mutations (9, 53, 72, 73). The mutation
rons and muscle cells (47, 56). The most severe defect usually results in a misfolded proinsulin molecule
included marked developmental delay and early-onset that is trapped and accumulated in the endoplasmic
epilepsy and became known as DEND (developmental reticulum, leading to endoplasmic reticulum stress and
delay, epilepsy, and neonatal diabetes) syndrome. -cell apoptosis (74).
An intermediate DEND syndrome characterized The severity of intrauterine growth retardation in
by neonatal diabetes and less severe developmental patients with heterozygous INS mutations is similar
delay without epilepsy is more common. Neurological to that of patients with KATP channel mutations.
features were considered less frequent and usually In contrast, diabetes presents at a slightly later age
milder in patients with mutations in ABCC8 (48, 49). although the ranges overlap greatly and patients do
However, a recent study suggested that mild neurode- not present with neurological features as a direct
velopmental abnormalities, including developmental consequence of the mutation (53).
coordination disorder (particularly visual-spatial The majority of heterozygous INS mutations are
dyspraxia) or attention deficits, might be found on sporadic de novo mutations. Only about 20% of
detailed testing in all patients with KATP channel probands have a positive family history of autosomal
mutations (57). dominant neonatal diabetes (53). Occasionally, INS
Approximately 90% of patients with activating mutations cause permanent diabetes after 6 months of
mutations in the KATP channel genes can be transferred age and therefore genetic testing should be considered
from insulin onto sulfonylurea tablets (58, 59). Transfer in certain situations, especially in patients with
usually improves glycemic control without increasing antibody-negative type 1 diabetes (12, 73, 75, 76).
the risk of hypoglycemia. The doses required are high In addition to heterozygous INS mutations,
when calculated on a per kg body weight basis com- homozygous or compound heterozygous mutations
pared with adults with type 2 diabetes, typically needing causing neonatal diabetes have also been described
around 0.5 mg/kg/d of glibenclamide, although doses (36). Biallelic mutations do not cause slowly progres-
as high as 2.3 mg/kg/d have been occasionally reported sive -cell destruction but result in a lack of insulin
(60, 61). Many patients have been able to progressively biosynthesis before and after birth, which explains
reduce the dose of sulfonylurea after transition while much lower birth weights and earlier presentation
maintaining excellent glycemic control (58, 62). The of diabetes in affected children. As the disease is
only side effects reported to date are transient diarrhea recessively inherited, there is a 25% recurrence risk in
and staining of the teeth (63, 64). Some brain imaging siblings but, in the absence of consanguinity, a very
studies have shown that sulfonylurea drugs may pene- low risk for the offspring of a patient.
trate bloodbrain barrier (65, 66) and very interesting
case reports suggest that sulfonylureas may partially
WolcottRallison syndrome
improve some of the neurological symptoms (6770).
Activating mutations in KCNJ11 causing neonatal Biallelic mutations in EIF2AK3 (eukaryotic transla-
diabetes are always heterozygous. As about 90% of tion initiation factor alpha 2-kinase 3) cause a rare
52 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

autosomal recessive syndrome characterized by early- with -cell autoimmunity and pancreatic islet
onset diabetes mellitus, spondyloepiphyseal dysplasia, autoantibodies. Among male infants who present with
and recurrent hepatic and/or renal dysfunction (77, 78). diabetes, immune deficiency, and/or life-threatening
EIF2AK3 encodes a protein involved in the regulation infection, mutations in FOXP3 should be considered.
of the endoplasmic reticulum stress response. Pancre- Treatment with immunosuppressive agents (sirolimus
atic development is rather normal in the absence of or steroids) is recommended (86, 87). Alternatively,
the functional protein but misfolded proteins accumu- allogeneic bone marrow transplantation with reduced-
late within the endoplasmic reticulum after birth and intensity conditioning should be considered (88).
eventually induce -cell apoptosis. Although diabetes
usually manifests during infancy, it might not present
Other causes of neonatal diabetes
until 34 yr of age. Diabetes may be the first clinical
manifestation of the syndrome and therefore this diag- The clinical features in other causes of neonatal and
nosis needs to be considered in children with PNDM infancy-onset diabetes are shown in Table 1. Pancreatic
especially if parental consanguinity is present or the scanning is unreliable in neonates and so it is best to use
patient originates from a highly inbred population (79, functional tests of exocrine pancreatic function (fecal
80). As the disease is recessively inherited, there is a 25% elastase and fecal fats) when assessing if pancreatic
recurrence risk in siblings but in the absence of consan- aplasia is present (89, 90). Apart from KATP channel
guinity, a very low risk for the offspring of a patient. neonatal diabetes, all other causes need to be treated
with subcutaneous insulin. Patients with pancreatic
aplasia/hypoplasia will also require exocrine pancreatic
Neonatal diabetes due to GCK mutations
supplements.
The enzyme glucokinase is considered the glucose
sensor of the cells, as it catalyzes the rate-limiting step
Genetic testing should be performed as soon
of glucose phosphorylation and therefore enables the
as diabetes is diagnosed in a child aged less than
cell to respond appropriately to the degree of glycemia
6 months
(81). Heterozygous mutations in the GCK gene
produce familial mild non-progressive hyperglycemia Genetic testing will allow diagnosis of a specific type of
(see below). However, complete glucokinase deficiency monogenic diabetes in over 80% of patients whose
secondary to mutations in both alleles, either diabetes is diagnosed before the age of 6 months.
homozygous or compound heterozygous, prevents As discussed above, this will influence treatment as
the cells from secreting insulin in response to well as prediction of clinical features. This means
hyperglycemia (82, 83). For this reason, patients that molecular genetic testing is now recommended
present with severe intrauterine growth retardation, at the time of diabetes diagnosis in child aged less than
are usually diagnosed with diabetes during the first 6 months. It is no longer necessary to wait to see if the
few days of life, and require exogenous insulin therapy. diabetes resolves or for other features to develop, as
Apart from diabetes, patients do not show any relevant major labs will offer comprehensive testing of all NDM
extrapancreatic features. subtypes as well as very rapid testing of subtypes that
GCK is responsible for not more than 23% of alter treatment.
cases of PNDM overall (37). This type of PNDM
is inherited in a recessive manner so the recurrence
Autosomal dominant familial mild
risk for future siblings is 25%. This diagnosis should
hyperglycemia or diabetes (MODY)
be strongly considered in probands born to parents
with asymptomatic mild hyperglycemia and therefore The different genetic subtypes of MODY differ in age
measuring fasting blood glucose in the parents of any of onset, pattern of hyperglycemia, and response to
child with neonatal diabetes, even when there is no treatment. Most of them cause isolated diabetes and
known consanguinity or family history of diabetes, is therefore may be misdiagnosed as either familial type
often recommended. Sulfonylurea treatment has been 1 or type 2 diabetes (10, 13, 91). Although the classic
tested with no clear effect (P. R. N. and A. T. H., criteria for MODY include family history of diabetes,
unpublished observations). sporadic de novo mutations in a number of causative
genes have been reported (92).
Three genes are responsible for the majority of
IPEX syndrome
MODY cases (GCK, HNF1A, and HNF4A) and will
Mutations in the FOXP3 gene are responsible for the be described in some detail below (see also Table 2).
IPEX (immune dysregulation, polyendocrinopathy, However, up to 13 different genes have been reported
enteropathy, X-linked) syndrome (84, 85). This is the to cause autosomal dominant non-insulin dependent
only well-established form of PNDM that is associated diabetes but these are so unusual they do not need to be
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 53
Rubio-Cabezas et al.

Table 2. Common subtypes of MODY and associated clinical features


Gene Locus Clinical features Treatment References
HNF4A 20q12-q13.1 Macrosomia and neonatal hypoglycemia, renal Sulfonylurea (187)
Fanconi syndrome (mutation specific)
GCK 7p15-p13 Mild asymptomatic hyperglycemia Nil/diet (188)
HNF1A 12q24.2 Renal glucosuria Sulfonylurea (189)
HNF1B 17q12 Renal developmental abnormalities, genital Insulin (190)
tract malformations

MODY, maturity-onset diabetes of the young.

tested for in children with diabetes except in a research associated with chronic microvascular or macrovascu-
setting or when there are additional phenotypes such lar complications of diabetes (100, 101) and patients do
as pancreatic exocrine dysfunction (93). not generally require any treatment (102). Of note, the
presence of a GCK mutation does not protect against
the concurrent development of polygenic type 2 dia-
Mild fasting hyperglycemia due to glucokinase betes later in life, which occurs at a similar prevalence
gene mutations (GCK-MODY, MODY2) than in the general population (103). GCK-PNDM
may manifest in GCK-MODY families since in the
The incidental finding of mild hyperglycemia
setting of consanguinity or a second de novo mutation.
(5.58 mmol/L or 100145 mg/dL) in otherwise
asymptomatic children and adolescents raises the
possibility that these patients subsequently develop
Familial diabetes due to HNF1A-MODY (MODY3)
type 1 or type 2 diabetes. In the absence of concomitant
and HNF4A-MODY (MODY1)
pancreatic autoimmunity, the risk of future type 1
diabetes is minimal (94) and a significant proportion The possibility of monogenic diabetes should be
will have a heterozygous mutation in GCK (95, 96). considered whenever a parent of a diabetic child has
In peripubertal children and adolescents, the lack of diabetes, even if they are thought to have type 1 or
obesity or other signs of insulin resistance should raise type 2 diabetes. HNF1A-MODY is the most common
concern about a diagnosis of type 2 diabetes. form of monogenic diabetes that results in familial
GCK-MODY is the commonest subtype of symptomatic diabetes, with heterozygous HNF1A
monogenic diabetes in the pediatric diabetes clinic mutations being about 10 times more frequent than
and its clinical phenotype is remarkably homogeneous heterozygous mutations in HNF4A (104). Therefore,
among patients. In contrast to other subtypes of HNF1A-MODY is the first diagnostic possibility to
monogenic diabetes, GCK-MODY patients regulate be considered in families with autosomal dominant
insulin secretion adequately but around a slightly symptomatic diabetes.
higher set point than normal subjects. As a result, In both HNF1A-MODY and HNF4A-MODY,
they show non-progressive mild hyperglycemia from glucose intolerance usually becomes evident during
birth (97). Their hemoglobin A1c (HbA1c) is mildly adolescence or early adulthood. In the early stages
elevated but usually below 7.5% (98). Despite the of the disease, fasting blood glucose may be normal
mild fasting hyperglycemia, there is usually a small but patients tend to show a large increment in blood
increment in blood glucose during an oral glucose glucose (>80 mg/dL or 5 mmol/L) after meals or at 2 h
tolerance test (<60 mg/dL or <3.5 mmol/L) (99), during an OGTT (99). Patients with HNF1A-MODY
although this should not be considered an absolute demonstrate impaired incretin effect and inappropriate
criterion because of the variability of the oral glucose glucagon responses to OGTT (105). Over time, fasting
tolerance test (OGTT). As the degree of hyperglycemia hyperglycemia and osmotic symptoms (polyuria and
is not high enough to cause osmotic symptoms, polydipsia) present but patients rarely develop ketosis
most cases are usually diagnosed incidentally when because some residual insulin secretion persists for
blood glucose is measured for any other reason. Very many years. Chronic complications of diabetes are
often, the affected parent remains undiagnosed or has frequent and their development is related to the degree
been misdiagnosed with early-onset type 2 diabetes. of metabolic control (106). The frequency of microvas-
Measuring fasting glucose in apparently unaffected cular complications (retinopathy, nephropathy, and
parents is important when considering a diagnosis of a neuropathy) is similar to that of patients with type
glucokinase mutation. 1 and type 2 diabetes. HNF1A mutations are asso-
As blood glucose does not deteriorate significantly ciated with an increased frequency of cardiovascular
over time, this subtype of monogenic diabetes is rarely disease (107).
54 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

Mutations in HNF1A show a high penetrance so that comparing a glucagon-like peptide (GLP-1) agonist
63% of mutation carriers develop diabetes before 25 yr with a sulfonylurea demonstrated lower fasting glucose
of age, 79% before age 35 and 96% before 55 yr (6). in those treated with the GLP-1 agonist (122).
The age at diagnosis of diabetes is partly determined
by the location of the mutation within the gene (108,
Genetic syndromes associated with diabetes
109). Patients with mutations affecting the terminal
exons (810) are diagnosed, on average, 8 yr later A monogenic disorder should be considered in any
than those with mutations in exons 16. On the other child with diabetes associated with multi-system
hand, exposure to maternal diabetes in utero (when extrapancreatic features (123). These syndromes may
the mutation is maternally inherited) brings forward either cause neonatal diabetes (Table 1) or present later
the age at onset of diabetes by about 12 yr (99). In in life (see below). The online Mendelian inheritance
the pediatric population, diabetes in HNF4A mutation in Man website (www.ncbi.nlm.nih.gov/omim or
carriers tend to appear at a similar age to patients with www.omim.org) can help with clinical features and
mutations in HNF1A (16). to know if the gene for a particular syndrome has been
There are some differential clinical characteristics defined and hence molecular genetic testing is available.
between patients with mutations in HNF4A and Genetic testing for some of these conditions is available
HNF1A that can help decide which gene should be on a research basis at www.euro-wabb.org (124). The
considered first in a particular family. most common syndromes usually presenting beyond
infancy are described in some detail below.
Patients with HNF1A mutations typically have a
low renal threshold for glucose reabsorption due to Diabetes insipidus, diabetes mellitus, optic atrophy,
impaired renal tubular transport of glucose and may and deafness syndrome (Wolfram syndrome)
present postprandial glycosuria before developing
The association of diabetes with progressive optic atro-
significant hyperglycemia (110).
phy below 16 yr of age is diagnostic of this autosomal
In addition to diabetes, carriers of the R76W
recessive syndrome (125). Non-autoimmune insulin-
mutation in HNF4A present with an atypical form
deficient diabetes is usually the first manifestation of
of Fanconi syndrome including hypercalciuria and
the disease and presents at a mean age of 6 yr, although
nephrocalcinosis (111).
may present anytime from early-infancy (126, 127).
About 50% of HNF4A mutation carriers are
Patients require insulin treatment from diagnosis.
macrosomic at birth and 15% have diazoxide-
Other typical clinical features, such as sensorineural
responsive neonatal hyperinsulinemic hypoglycemia
deafness, central diabetes insipidus, urinary tract
(112). In this case, hyperinsulinism typically remits
dilatations, and neurological symptoms develop later
during infancy and patients develop diabetes from
in a variable order even within the same family. Many
adolescence (113, 114). Recently, hyperinsulinemic
patients with Wolfram Syndrome (WFS) are initially
hypoglycemia has also been reported in HNF1A
diagnosed as having type 1 diabetes; subsequent loss of
mutation carriers (115) but this is very uncommon.
vision, which occurs approximately 4 yr after diabetes
diagnosis, may be misdiagnosed as diabetic retinopa-
Patients with both HNF1A- and HNF4A-MODY
thy (128, 129). Patients WFS die at a median age of
can initially be treated with diet although they will
30 yr, mainly from neurodegenerative complications.
have marked postprandial hyperglycemia with high
At least 90% of patients harbor recessively acting
carbohydrate food (99). Most patients will need
mutations in the WFS1 gene (130, 131). A second
pharmacological treatment as they show progressive
variant of the syndrome has recently been described
deterioration in glycemic control. They are extremely
in association with mutations in CISD2 (132). Patients
sensitive to sulfonylureas (116), which usually allow a
with this rare variant do not develop diabetes insipidus
better glycemic control than that achieved on insulin,
but present with additional symptoms including
especially in children and young adults (117). The initial
bleeding diathesis and peptic ulcer disease.
dose should be low (one-quarter of the normal starting
dose in adults) to avoid hypoglycemia. As long as the
Renal cysts and diabetes syndrome
patients do not have problems with hypoglycemia,
(HNF1B-MODY or MODY5)
they can be maintained on low-dose sulfonylureas
(e.g., 2040 mg gliclazide daily) for decades (118, 119). Although initially described as a rare subtype of
If there is hypoglycemia despite dose titration of a familial diabetes, it is now clear that patients with
once or twice daily sulfonylurea preparation, a slow heterozygous mutations in HNF1B rarely present
release preparation or meal time doses with a short- with isolated diabetes (133). In contrast, renal
acting agent such as nateglinide may be considered developmental disorders (especially renal cysts and
(120, 121). A recent randomized controlled trial renal dysplasia) are present in almost all patients
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 55
Rubio-Cabezas et al.

with HNF1B mutations or gene deletions (8) and mitochondrial function and may trigger episodes of
constitute the main presentation in children, even in the lactic acidosis (146).
absence of diabetes (134). Genital-tract malformations The penetrance of diabetes in mutation carriers
(particularly uterine abnormalities), hyperuricemia depends on the age considered, but is estimated to be
and gout can also occur, as well as abnormal liver above 85% at 70 yr (142). Affected males do not trans-
function tests (133). Diabetes develops later, typically mit the disease to their offspring. In contrast, females
during adolescence or early adulthood (135, 136), transmit the mutation to all their children, although
although transient neonatal diabetes has been reported some may not develop the disease (6). In addition to
in a few cases (35, 137). In addition to insulin the m.3243A>G mutation, early-onset diabetes (even
deficiency related to pancreatic hypoplasia (138), in infancy) has been reported in other less common
patients also show some degree of hepatic insulin mitochondrial disorders such as KearnsSayre
resistance (139), which explains why they do not syndrome (147) and Pearson syndrome (148).
respond adequately to sulfonylurea treatment and
require early insulin therapy (6). Moreover, mutation
carriers have lower exocrine pancreatic function with Diabetes secondary to monogenic diseases of the
reduced fecal elastase; this involves both ductal and exocrine pancreas
acinar cells (140). Therefore, the phenotype of renal
cysts and diabetes (RCAD) patients is highly variable Heterozygous mutations in CEL, which encodes
even within families sharing the same HNF1B mutation a pancreatic lipase, cause an autosomal dominant
and therefore this diagnosis should be considered not disorder of pancreatic exocrine insufficiency and
only in the diabetes clinic but also in other clinics diabetes (93). Importantly, the exocrine component
(nephrology, urology, gynecology, etc.). In patients of the syndrome is initiated already in childhood,
found to have renal cysts, imaging of the pancreas 1030 yr before diabetes develops, and can be revealed
is indicated, as the absence of the pancreatic body by lowered fecal elastase and/or pancreatic lipomatosis
and/or tail is highly indicative of HNF1B-MODY (149, 150). Other autosomal dominant monogenic
(141). Fecal elastase should also be measured, as this diseases affecting mainly the exocrine pancreas that can
is always abnormal in patients with HNF1B-MODY lead to diabetes sooner or later include cystic fibrosis
(140). Importantly, a family history of renal disease or (CFTR) (151), hereditary pancreatitis (PRSS1 and
diabetes is not essential to prompt genetic testing, as SPINK1) (152), and pancreatic agenesis/hypoplasia
spontaneous mutations and deletions of this gene are (GATA6) (90).
common (one third to two thirds of cases) (8, 134).

Monogenic insulin resistance syndromes


Mitochondrial diabetes The key features of insulin resistance syndromes are
Diabetes due to mitochondrial mutations and dele- moderate to severe acanthosis nigricans associated
tions is rarely seen in the pediatric age group as the with either severely increased insulin concentrations
vast majority of patients develop diabetes as young or increased insulin requirements (depending on
or middle-aged adults. The most common form of whether the patient has diabetes already), usually
mitochondrial diabetes is caused by the m.3243A>G in the absence of a corresponding degree of obesity.
mutation in mitochondrial DNA. Diabetes onset is Three different groups have been proposed based
usually insidious but approximately 20% of patients on the pathogenesis of the disease: primary insulin
have an acute presentation, even in diabetic ketoacido- signaling defects, insulin resistance secondary to
sis (142). Although it typically presents in adulthood, adipose tissue abnormalities, and insulin resistance as
some cases have been reported in adolescents with a a feature of complex syndromes (153). Clinical and
high degree of heteroplasmy (143, 144). Mitochondrial biochemical characterization of patients with severe
diabetes should be suspected in patients present- insulin resistance may be used to guide genetic testing,
ing with diabetes and sensorineural hearing loss as it happens with monogenic -cell diabetes (Table 3).
inherited from mothers side. Interestingly, the same However, diabetes associated with monogenic severe
m.3243A>G mutation also causes a much more severe insulin resistance is far less common than monogenic
clinical syndrome known as MELAS (myopathy, -cell failure, especially in prepubertal children as
encephalopathy, lactic acidosis, and stroke) (145). hyperglycemia is usually a late event in the natural
Patients with mitochondrial diabetes may respond history of these disorders (154). As ovarian hyperan-
initially to diet or oral hypoglycemic agents but often drogenism usually is the commonest presentation in
require insulin treatment within months or years. adolescents, there is a gender bias in the diagnosis. The
Metformin should be avoided as it interferes with most relevant disorders are briefly described below.
56 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

Table 3. Classification of syndromes of severe insulin resistance (modified from reference 154)
Insulin resistance syndrome subtype Gene (inheritance) Leptin Adiponectin Other clinical features

Primary insulin Receptor defect INSR (AR or AD) Decreased Normal or elevated No dyslipidemia
signaling defects Post receptor defects AKT2, TBC1D4 (AD) No fatty liver
Adipose tissue Monogenic obesity MC4R (AD) Increased (low in LEP) Tall stature (MC4R)
abnormalities LEP, LEPR, POMC (AR) Hypogonadism (LEP)
Others Hypoadrenalism (POMC)
Congenital generalized AGPAT2, BSCL2 (AR) Decreased Decreased Severe dyslipidemia (high
lipodystrophy Others triglycerides, low HDL
cholesterol)
Fatty liver
Partial lipodystrophy LMNA, PPARG, Variable Myopathy and cardiomyopathy
PIK3R1 (AD) (LMNA)
Others Pseudoacromegaly (PPARG)
SHORT syndrome with partial
lipodystrophy, insulin resistance
and diabetes (PIK3R1)
Complex
Alstrom ALMS1 (AR)
syndromes
BardetBiedl BBS1 to BBS18 (mostly
AR)
DNA damage repair WRN (AR)
disorders BLM (AR)
Primordial dwarfism PCNT (AR)

AD, autosomal dominant; AR, autosomal recessive; HDL, high-density lipoprotein; SHORT, short stature, hypermobility of joints, ocular depression, Riegers
anomaly, and teething delay syndrome.

Primary insulin signaling defects due to mutations levels and insulin resistance (157). Mutations in either
in the insulin receptor gene AGPAT2 or BSCL account for approximately 80%
of cases of congenital generalized lipodystrophy
Insulin receptor (INSR) gene mutations are responsible
(BerardinelliSeip syndrome) (158). This is a recessive
for a number of rare insulin resistance syndromes (155).
disorder characterized by an almost complete absence
Leptin levels are low, but adiponectin levels are normal
of subcutaneous and visceral fat with abdominal
or elevated as insulin normally inhibits adiponectin
distention due hepatic steatosis, which may evolve to
secretion. The most common form is type A insulin
hepatic fibrosis. Diabetes usually becomes apparent in
resistance syndrome, which is usually diagnosed in
early adolescence. In contrast, familial partial lipodys-
non-obese female adolescents with severe acanthosis
nigricans and hyperandrogenism (polycystic ovarian trophy is usually recognized after puberty in patients
syndrome) and may show autosomal dominant with loss of subcutaneous fat from the extremities
or autosomal recessive inheritance. Mutations in and lower trunk and progressive accumulation of
both alleles of INSR are also responsible for the subcutaneous adipose tissue in the face and around
more severe Donohue syndrome (formerly known as the neck. Visceral fat is greatly increased. In addi-
Leprechaunism) and RabsonMendenhall syndrome. tion to hyperinsulinemia, hypertriglyceridemia, and
The presenting complaint is failure to thrive, with decreased high-density lipoprotein (HDL) cholesterol,
impaired linear growth and weight gain, associated to patients also show signs of hyperandrogenism and
overgrowth of soft tissues. Postprandial hyperglycemia sometimes pseudoacromegalic growth of soft tissues.
may be severe but is usually accompanied by fasting Diabetes usually appears in late adolescence or early
hypoglycemia. adulthood. Heterozygous mutations in LMNA or
Metabolic control in patients with INSR mutations PPARG account for approximately 50% of cases
remains poor and long-term diabetes complications (157). Two recent causes of lipodystrophy and multi-
are frequent. Insulin sensitizers may be tried initially system disease are: (i) subcutaneous lipodystrophy
but most patients need extraordinarily high doses of and diabetes, deafness, mandibular hypoplasia, and
insulin, with limited effect (155). As an alternative hypogonadism in males associated with a specific
therapeutic method for young children, recombinant mutation in POLD1, a universal DNA polymerase
human insulin-like growth factor (IGF-I) has been (159) and (ii) SHORT (short stature, hypermobility
reported to improve both fasting and postprandial of joints, ocular depression, Riegers anomaly, and
glycemia although long-term effects on survival remain teething delay) syndrome with partial lipodystrophy,
unclear (156). in which IR and diabetes were caused by a hot spot
mutation in PIK3R1 encoding p85 that has a central
role in the insulin-signaling pathway (160).
Monogenic lipodystrophies
Dietary advice with a low-fat, sometimes hypocaloric
Lipodystrophies are characterized by a selective lack diet is the mainstay of treating lipodystrophies as it can
of adipose tissue, which results in decreased adipokines have a dramatic effect on metabolic derangements.
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 57
Rubio-Cabezas et al.

In partial lipodystrophy, insulin sensitizers such as testing is being used as a diagnostic tool that can
metformin and glitazones may be initially effective help define the diagnosis and treatment of children
(161) but glitazones can cause further accumulation with diabetes. As these tests are expensive, diagnostic
of fat in the face and neck (154). Patients with genetic testing should be limited to those patients who
severe congenital lipodystrophy greatly benefit from are likely to harbor a mutation on clinical grounds.
treatment with recombinant leptin (162). In partial
lipodystrophy, leptin replacement has limited value
with improvement of hypertriglyceridemia but not
Conflicts of interest
hyperglycemia (163). The authors have declared no conflicts of interest.

Ciliopathy-related insulin resistance and diabetes References



Alstrom syndrome (ALMS). This autosomal reces- 1. Fajans SS, Bell GI. MODY: history, genetics,
sive disorder shares symptoms with BardetBiedl pathophysiology, and clinical decision making.
syndrome (BBS) (see below), including progressive Diabetes Care 2011: 34: 18781884.
visual impairment related to conerod dystrophy, sen- 2. Tattersall R. Maturity-onset diabetes of the young:
sorineural hearing loss, obesity, and diabetes mellitus. a clinical history. Diabet Med 1998: 15: 1114.
3. Tattersall RB, Fajans SS. A difference between the
It can be distinguished from the latter syndrome by
inheritance of classical juvenile-onset and maturity-
the lack of polydactyly and hypogonadism and by onset type diabetes of young people. Diabetes 1975:
the absence of cognitive impairment (164). More than 24: 4453.
60% of individuals with ALMS develop cardiomy- 4. Tattersall RB. Mild familial diabetes with dominant
opathy. The syndrome is caused by mutations within inheritance. Q J Med 1974: 43: 339357.
the ALMS1 gene of unknown function (165). Patients 5. Fajans SS, Bell GI, Polonsky KS. Molecular
ALMS usually show many features of the metabolic mechanisms and clinical pathophysiology of maturity-
onset diabetes of the young. N Engl J Med 2001: 345:
syndrome including acanthosis nigricans, hyperlipi-
971980.
demia, hyperuricemia, hypertension, and slowly pro- 6. Murphy R, Ellard S, Hattersley AT. Clinical
gressive insulin-resistant diabetes (166). Lifestyle inter- implications of a molecular genetic classification
vention can initially ameliorate the metabolic abnor- of monogenic beta-cell diabetes. Nat Clin Pract
malities (167). Endocrinol Metab 2008: 4: 200213.
7. Gloyn AL, Pearson ER, Antcliff JF et al. Activating
mutations in the gene encoding the ATP-sensitive
BardetBiedl syndrome. This disorder is charac- potassium-channel subunit Kir6.2 and permanent
terized by intellectual disability, progressive visual neonatal diabetes. N Engl J Med 2004: 350: 18381849.
impairment due to conerod dystrophy, polydactyly, 8. Bellanne-Chantelot C, Clauin S, Chauveau D
obesity, diabetes mellitus, renal dysplasia, hepatic et al. Large genomic rearrangements in the hepatocyte
fibrosis, and hypogonadism. Obesity is found in nuclear factor-1beta (TCF2) gene are the most frequent
almost every patient, while diabetes affects less than cause of maturity-onset diabetes of the young type 5.
Diabetes 2005: 54: 31263132.
50% (168). While the syndrome shares some simi-
9. Sty J, Edghill EL, Flanagan SE et al. Insulin gene
larities with LawrenceMoon syndrome, these two mutations as a cause of permanent neonatal diabetes.
disorders can be distinguished by the presence of Proc Natl Acad Sci U S A 2007: 104: 1504015044.
paraplegia and the absence of polydactyly, obesity, 10. Mller AM, Dalgaard LT, Pociot F, Nerup J,
and diabetes mellitus in LawrenceMoon syndrome. Hansen T, Pedersen O. Mutations in the hepatocyte
Terms such as LawrenceMoonBardetBiedl or nuclear factor-1alpha gene in Caucasian families
LawrenceMoonBiedl syndrome should therefore be originally classified as having type I diabetes.
avoided. BBS has been linked to 18 different genetic Diabetologia 1998: 41: 15281531.
11. Lambert AP, Ellard S, Allen LI et al. Identifying
loci, referred to as BBS1 to BBS18 (169, 170). The hepatic nuclear factor 1alpha mutations in children
majority of cases are autosomal recessive (171), but and young adults with a clinical diagnosis of type 1
triallelic inheritance has been reported (172). Genetic diabetes. Diabetes Care 2003: 26: 333337.
diagnostic laboratories and detailed clinical recommen- 12. Rubio-Cabezas O, Edghill EL, Argente J,
dations for patients with ALMS and BBS are present Hattersley AT. Testing for monogenic diabetes
at http://www.euro-wabb.org. among children and adolescents with antibody-
negative clinically defined Type 1 diabetes. Diabet
Med 2009: 26: 10701074.
Conclusions 13. Awa WL, Schober E, Wiegand S et al. Reclassifi-
cation of diabetes type in pediatric patients initially
Advances in molecular genetics have led to the classified as type 2 diabetes mellitus: 15 years follow-
identification of genes associated with many clinically up using routine data from the German/Austrian DPV
identified subgroups of diabetes. Molecular genetic database. Diabetes Res Clin Pract 2011: 94: 463467.

58 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764


Monogenic diabetes in children and adolescents

14. Fendler W, Borowiec M, Baranowska-Jazwiecka 30. Mohamadi A, Clark LM, Lipkin PH, Mahone EM,
A et al. Prevalence of monogenic diabetes amongst Wodka EL, Plotnick LP. Medical and developmental
Polish children after a nationwide genetic screening impact of transition from subcutaneous insulin to oral
campaign. Diabetologia 2012: 55: 26312635. glyburide in a 15-yr-old boy with neonatal diabetes
15. Irgens HU, Molnes J, Johansson BB et al. mellitus and intermediate DEND syndrome: extending
Prevalence of monogenic diabetes in the population- the age of KCNJ11 mutation testing in neonatal DM.
based Norwegian Childhood Diabetes Registry. Pediatr Diabetes 2010: 11: 203207.
Diabetologia 2013: 56: 15121519. 31. Gicquel C, Le Bouc Y. Hormonal regulation of fetal
16. Pihoker C, Gilliam LK, Ellard S et al. Prevalence, growth. Horm Res 2006: 65 (Suppl. 3): 2833.
characteristics and clinical diagnosis of maturity onset 32. Temple IK, Gardner RJ, Mackay DJ, Barber JC,
diabetes of the young due to mutations in HNF1A, Robinson DO, Shield JP. Transient neonatal diabetes:
HNF4A, and glucokinase: results from the SEARCH widening the understanding of the etiopathogenesis of
for Diabetes in Youth. J Clin Endocrinol Metab 2013: diabetes. Diabetes 2000: 49: 13591366.
98: 40554062. 33. Gardner RJ, Mackay DJ, Mungall AJ et al. An
17. Bonnefond A, Philippe J, Durand E et al. Highly imprinted locus associated with transient neonatal
sensitive diagnosis of 43 monogenic forms of diabetes diabetes mellitus. Hum Mol Genet 2000: 9: 589596.
or obesity through one-step PCR-based enrichment 34. Flanagan SE, Patch AM, Mackay DJ et al.
in combination with next-generation sequencing. Mutations in ATP-sensitive K+ channel genes cause
Diabetes Care 2014: 37: 460467. transient neonatal diabetes and permanent diabetes
18. Ellard S, Lango Allen H, De Franco E et al. in childhood or adulthood. Diabetes 2007: 56:
Improved genetic testing for monogenic diabetes using 19301937.
targeted next-generation sequencing. Diabetologia 35. Yorifuji T, Kurokawa K, Mamada M et al. Neonatal
2013: 56: 19581963. diabetes mellitus and neonatal polycystic, dysplastic
19. Gao R, Liu Y, Gjesing AP et al. Evaluation of a target kidneys: phenotypically discordant recurrence of a
region capture sequencing platform using monogenic mutation in the hepatocyte nuclear factor-1beta gene
diabetes as a study-model. BMC Genet 2014: 15: 13. due to germline mosaicism. J Clin Endocrinol Metab
20. Johansson S, Irgens H, Chudasama KK et al. Exome 2004: 89: 29052908.
sequencing and genetic testing for MODY. PLoS One 36. Garin I, Edghill EL, Akerman I et al. Recessive
2012: 7: e38050. mutations in the INS gene result in neonatal diabetes
21. Greeley SA, John PM, Winn AN et al. The cost- through reduced insulin biosynthesis. Proc Natl Acad
effectiveness of personalized genetic medicine: the case Sci U S A 2010: 107: 31053110.
of genetic testing in neonatal diabetes. Diabetes Care 37. Rubio-Cabezas O, Ellard S. Diabetes mellitus in
2011: 34: 622627. neonates and infants: genetic heterogeneity, clinical
22. Naylor RN, John PM, Winn AN et al. Cost- approach to diagnosis, and therapeutic options. Horm
effectiveness of MODY genetic testing: translating Res Paediatr 2013: 80: 137146.
genomic advances into practical health applications. 38. Russo L, Iafusco D, Brescianini S et al. Permanent
Diabetes Care 2014: 37: 202209. diabetes during the first year of life: multiple gene
23. Iafusco D, Stazi MA, Cotichini R et al. Permanent screening in 54 patients. Diabetologia 2011: 54:
diabetes mellitus in the first year of life. Diabetologia 16931701.
2002: 45: 798804. 39. Mackay D, Bens S, Perez de Nanclares G, Siebert
24. Rubio-Cabezas O. Diagnosing monogenic diabetes: R, Temple IK. Clinical utility gene card for: transient
common misinterpretations of genetic findings. Pediatr neonatal diabetes mellitus, 6q24-related. Eur J Hum
Diabetes 2009: 10: 497499. Genet 2014: Feb 26. doi: 10.1038/ejhg.2014.27.
25. Ellard S, Bellanne-Chantelot C, Hattersley AT. 40. Ma D, Shield JP, Dean W et al. Impaired glucose
European Molecular Genetics Quality Network Mg. homeostasis in transgenic mice expressing the human
Best practice guidelines for the molecular genetic transient neonatal diabetes mellitus locus, TNDM. J
diagnosis of maturity-onset diabetes of the young. Clin Invest 2004: 114: 339348.
Diabetologia 2008: 51: 546553. 41. Temple IK, Shield JP. Transient neonatal diabetes, a
26. American Diabetes Association. Diagnosis and disorder of imprinting. J Med Genet 2002: 39: 872875.
classification of diabetes mellitus. Diabetes Care 2014: 42. Mackay DJ, Boonen SE, Clayton-Smith J et al.
37 (Suppl. 1): S81S90. A maternal hypomethylation syndrome presenting as
27. Edghill EL, Dix RJ, Flanagan SE et al. HLA transient neonatal diabetes mellitus. Hum Genet 2006:
genotyping supports a nonautoimmune etiology in 120: 262269.
patients diagnosed with diabetes under the age of 6 43. Mackay DJ, Callaway JL, Marks SM et al.
months. Diabetes 2006: 55: 18951898. Hypomethylation of multiple imprinted loci in
28. Rubio-Cabezas O, Minton JA, Caswell R et al. individuals with transient neonatal diabetes is
Clinical heterogeneity in patients with FOXP3 associated with mutations in ZFP57. Nat Genet 2008:
mutations presenting with permanent neonatal 40: 949951.
diabetes. Diabetes Care 2009: 32: 111116. 44. Docherty LE, Kabwama S, Lehmann A et al. Clinical
29. Rubio-Cabezas O, Flanagan SE, Damhuis A, presentation of 6q24 transient neonatal diabetes
Hattersley AT, Ellard S. KATP channel mutations mellitus (6q24 TNDM) and genotype-phenotype
in infants with permanent diabetes diagnosed after 6 correlation in an international cohort of patients.
months of life. Pediatr Diabetes 2012: 13: 322325. Diabetologia 2013: 56: 758762.

Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 59


Rubio-Cabezas et al.

45. Shield JP, Temple IK, Sabin M et al. An assessment diabetes due to sulfonylurea receptor 1 (SUR1)
of pancreatic endocrine function and insulin sensitivity mutations. Diabetes Care 2008: 31: 204209.
in patients with transient neonatal diabetes in 60. Sagen JV, Raeder H, Hathout E et al. Permanent
remission. Arch Dis Child Fetal Neonatal Ed 2004: neonatal diabetes due to mutations in KCNJ11
89: F341F343. encoding Kir6.2: patient characteristics and initial
46. McTaggart JS, Clark RH, Ashcroft FM. The role response to sulfonylurea therapy. Diabetes 2004: 53:
of the KATP channel in glucose homeostasis in health 27132718.
and disease: more than meets the islet. J Physiol 2010: 61. Greeley SA, Tucker SE, Naylor RN, Bell GI,
588 (Pt 17): 32013209. Philipson LH. Neonatal diabetes mellitus: a model
47. Ashcroft FM. ATP-sensitive potassium chan- for personalized medicine. Trends Endocrinol Metab
nelopathies: focus on insulin secretion. J Clin Invest 2010: 21: 464472.
2005: 115: 20472058. 62. Klupa T, Skupien J, Mirkiewicz-Sieradzka B et al.
48. Babenko AP, Polak M, Cave H et al. Activating Efficacy and safety of sulfonylurea use in permanent
mutations in the ABCC8 gene in neonatal diabetes neonatal diabetes due to KCNJ11 gene mutations: 34-
mellitus. N Engl J Med 2006: 355: 456466. month median follow-up. Diabetes Technol Ther 2010:
49. Ellard S, Flanagan SE, Girard CA et al. Permanent 12: 387391.
neonatal diabetes caused by dominant, recessive, 63. Codner E, Flanagan S, Ellard S, Garca H,
or compound heterozygous SUR1 mutations with Hattersley AT. High-dose glibenclamide can replace
opposite functional effects. Am J Hum Genet 2007: insulin therapy despite transitory diarrhea in early-
81: 375382. onset diabetes caused by a novel R201L Kir6.2
50. Flanagan SE, Edghill EL, Gloyn AL, Ellard mutation. Diabetes Care 2005: 28: 758759.
S, Hattersley AT. Mutations in KCNJ11, which 64. Kumaraguru J, Flanagan SE, Greeley SA et al.
encodes Kir6.2, are a common cause of diabetes Tooth discoloration in patients with neonatal diabetes
diagnosed in the first 6 months of life, with the after transfer onto glibenclamide: a previously
phenotype determined by genotype. Diabetologia unreported side effect. Diabetes Care 2009: 32:
2006: 49: 11901197. 14281430.
51. Vaxillaire M, Populaire C, Busiah K et al. Kir6.2 65. Mlynarski W, Tarasov AI, Gach A et al.
mutations are a common cause of permanent neonatal Sulfonylurea improves CNS function in a case of
diabetes in a large cohort of French patients. Diabetes intermediate DEND syndrome caused by a mutation
2004: 53: 27192722. in KCNJ11. Nat Clin Pract Neurol 2007: 3: 640645.
52. Proks P, Arnold AL, Bruining J et al. A 66. Fendler W, Pietrzak I, Brereton MF et al.
heterozygous activating mutation in the sulphonylurea Switching to sulphonylureas in children with iDEND
receptor SUR1 (ABCC8) causes neonatal diabetes. syndrome caused by KCNJ11 mutations results in
Hum Mol Genet 2006: 15: 17931800. improved cerebellar perfusion. Diabetes Care 2013:
53. Edghill EL, Flanagan SE, Patch AM et al. Insulin 36: 23112316.
mutation screening in 1,044 patients with diabetes: 67. Battaglia D, Lin YW, Brogna C et al. Glyburide
mutations in the INS gene are a common cause ameliorates motor coordination and glucose home-
of neonatal diabetes but a rare cause of diabetes ostasis in a child with diabetes associated with the
diagnosed in childhood or adulthood. Diabetes 2008: KCNJ11/S225T, del226-232 mutation. Pediatr Dia-
57: 10341042. betes 2012: 13: 656660.
54. Hattersley AT, Ashcroft FM. Activating mutations 68. Gurgel LC, Crispim F, Noffs MH, Belzunces E,
in Kir6.2 and neonatal diabetes: new clinical Rahal MA, Moises RS. Sulfonylrea treatment in
syndromes, new scientific insights, and new therapy. permanent neonatal diabetes due to G53D mutation
Diabetes 2005: 54: 25032513. in the KCNJ11 gene: improvement in glycemic control
55. Gloyn AL, Diatloff-Zito C, Edghill EL et al. and neurological function. Diabetes Care 2007: 30:
KCNJ11 activating mutations are associated with e108.
developmental delay, epilepsy and neonatal diabetes 69. Koster JC, Cadario F, Peruzzi C, Colombo C,
syndrome and other neurological features. Eur J Hum Nichols CG, Barbetti F. The G53D mutation in
Genet 2006: 14: 824830. Kir6.2 (KCNJ11) is associated with neonatal diabetes
56. Clark RH, McTaggart JS, Webster R et al. Muscle and motor dysfunction in adulthood that is improved
dysfunction caused by a KATP channel mutation in with sulfonylurea therapy. J Clin Endocrinol Metab
neonatal diabetes is neuronal in origin. Science 2010: 2008: 93: 10541061.
329: 458461. 70. Slingerland AS, Nuboer R, Hadders-Algra M,
57. Busiah K, Drunat S, Vaivre-Douret L et al. Hattersley AT, Bruining GJ. Improved motor
Neuropsychological dysfunction and developmental development and good long-term glycaemic control
defects associated with genetic changes in infants with with sulfonylurea treatment in a patient with the
neonatal diabetes mellitus: a prospective cohort study. syndrome of intermediate developmental delay, early-
Lancet Diabetes Endocrinol 2013: 1: 199207. onset generalised epilepsy and neonatal diabetes
58. Pearson ER, Flechtner I, Njolstad PR et al. associated with the V59M mutation in the KCNJ11
Switching from insulin to oral sulfonylureas in patients gene. Diabetologia 2006: 49: 25592563.
with diabetes due to Kir6.2 mutations. N Engl J Med 71. Edghill EL, Gloyn AL, Goriely A et al. Origin of de
2006: 355: 467477. novo KCNJ11 mutations and risk of neonatal diabetes
59. Rafiq M, Flanagan SE, Patch AM et al. Effective for subsequent siblings. J Clin Endocrinol Metab 2007:
treatment with oral sulfonylureas in patients with 92: 17731777.
60 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764
Monogenic diabetes in children and adolescents

72. Colombo C, Porzio O, Liu M et al. Seven mutations 87. Yong PL, Russo P, Sullivan KE. Use of sirolimus in
in the human insulin gene linked to permanent IPEX and IPEX-like children. J Clin Immunol 2008:
neonatal/infancy-onset diabetes mellitus. J Clin Invest 28: 581587.
2008: 118: 21482156. 88. Rao A, Kamani N, Filipovich A et al. Successful
73. Polak M, Dechaume A, Cave H et al. Heterozygous bone marrow transplantation for IPEX syndrome
missense mutations in the insulin gene are linked to after reduced-intensity conditioning. Blood 2007: 109:
permanent diabetes appearing in the neonatal period 383385.
or in early infancy: a report from the French ND 89. Weedon MN, Cebola I, Patch AM et al. Recessive
(Neonatal Diabetes) Study Group. Diabetes 2008: 57: mutations in a distal PTF1A enhancer cause isolated
11151119. pancreatic agenesis. Nat Genet 2014: 46: 6164.
74. Eizirik DL, Cardozo AK, Cnop M. The role for 90. Lango Allen H, Flanagan SE, Shaw-Smith C et al.
endoplasmic reticulum stress in diabetes mellitus. GATA6 haploinsufficiency causes pancreatic agenesis
Endocr Rev 2008: 29: 4261. in humans. Nat Genet 2012: 44: 2022.
75. Bonfanti R, Colombo C, Nocerino V et al. Insulin 91. Shields BM, Hicks S, Shepherd MH, Colclough K,
gene mutations as cause of diabetes in children negative Hattersley AT, Ellard S. Maturity-onset diabetes of
for five type 1 diabetes autoantibodies. Diabetes Care the young (MODY): how many cases are we missing?
2009: 32: 123125. Diabetologia 2010: 53: 25042508.
76. Molven A, Ringdal M, Nordbo AM et al. 92. Stanik J, Dusatkova P, Cinek O et al. De novo
Mutations in the insulin gene can cause MODY and mutations of GCK, HNF1A and HNF4A may be
autoantibody-negative type 1 diabetes. Diabetes 2008: more frequent in MODY than previously assumed.
57: 11311135. Diabetologia 2014: 57: 480484.
77. Delepine M, Nicolino M, Barrett T, Golamaully 93. Raeder H, Johansson S, Holm PI et al. Mutations
M, Lathrop GM, Julier C. EIF2AK3, encoding in the CEL VNTR cause a syndrome of diabetes and
translation initiation factor 2-alpha kinase 3, is pancreatic exocrine dysfunction. Nat Genet 2006: 38:
mutated in patients with Wolcott-Rallison syndrome. 5462.
Nat Genet 2000: 25: 406409. 94. Lorini R, Alibrandi A, Vitali L et al. Risk of type
1 diabetes development in children with incidental
78. Senee V, Vattem KM, Delepine M et al. Wolcott-
hyperglycemia: a multicenter Italian study. Diabetes
Rallison syndrome: clinical, genetic, and functional
Care 2001: 24: 12101216.
study of EIF2AK3 mutations and suggestion of genetic
95. Feigerlova E, Pruhova S, Dittertova L et al. Aetio-
heterogeneity. Diabetes 2004: 53: 18761883.
logical heterogeneity of asymptomatic hyperglycaemia
79. Habeb AM, Flanagan SE, Deeb A et al. Permanent
in children and adolescents. Eur J Pediatr 2006: 165:
neonatal diabetes: different aetiology in Arabs
446452.
compared to Europeans. Arch Dis Child 2012: 97:
96. Lorini R, Klersy C, dAnnunzio G et al. Maturity-
721723.
onset diabetes of the young in children with incidental
80. Rubio-Cabezas O, Patch AM, Minton JA et al.
hyperglycemia: a multicenter Italian study of 172
Wolcott-Rallison syndrome is the most common
families. Diabetes Care 2009: 32: 18641866.
genetic cause of permanent neonatal diabetes in 97. Prisco F, Iafusco D, Franzese A, Sulli N, Barbetti
consanguineous families. J Clin Endocrinol Metab F. MODY 2 presenting as neonatal hyperglycaemia: a
2009: 94: 41624170. need to reshape the definition of neonatal diabetes?
81. Matschinsky FM. Glucokinase, glucose homeostasis, Diabetologia 2000: 43: 13311332.
and diabetes mellitus. Curr Diab Rep 2005: 5: 171176. 98. Steele AM, Wensley KJ, Ellard S et al. Use
82. Njolstad PR, Sagen JV, Bjorkhaug L et al. of HbA1c in the identification of patients with
Permanent neonatal diabetes caused by glucokinase hyperglycaemia caused by a glucokinase mutation:
deficiency: inborn error of the glucose-insulin signaling observational case control studies. PLoS One 2013: 8:
pathway. Diabetes 2003: 52: 28542860. e65326.
83. Njlstad PR, Svik O, Cuesta-Munoz A et al. 99. Stride A, Vaxillaire M, Tuomi T et al. The genetic
Neonatal diabetes mellitus due to complete glucoki- abnormality in the beta cell determines the response to
nase deficiency. N Engl J Med 2001: 344: 15881592. an oral glucose load. Diabetologia 2002: 45: 427435.
84. Bennett CL, Christie J, Ramsdell F et al. 100. Steele AM, Shields BM, Wensley KJ, Colclough
The immune dysregulation, polyendocrinopathy, K, Ellard S, Hattersley AT. Prevalence of vascular
enteropathy, X-linked syndrome (IPEX) is caused by complications among patients with glucokinase
mutations of FOXP3. Nat Genet 2001: 27: 2021. mutations and prolonged, mild hyperglycemia. JAMA
85. Verbsky JW, Chatila TA. Immune dysregulation, 2014: 311: 279286.
polyendocrinopathy, enteropathy, X-linked (IPEX) 101. Velho G, Blanche H, Vaxillaire M et al.
and IPEX-related disorders: an evolving web of Identification of 14 new glucokinase mutations and
heritable autoimmune diseases. Curr Opin Pediatr description of the clinical profile of 42 MODY-2
2013: 25: 708714. families. Diabetologia 1997: 40: 217224.
86. Bindl L, Torgerson T, Perroni L et al. Successful 102. Stride A, Shields B, Gill-Carey O et al. Cross-
use of the new immune-suppressor sirolimus in sectional and longitudinal studies suggest pharma-
IPEX (immune dysregulation, polyendocrinopathy, cological treatment used in patients with glucokinase
enteropathy, X-linked syndrome). J Pediatr 2005: 147: mutations does not alter glycaemia. Diabetologia 2014:
256259. 57: 5456.

Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 61


Rubio-Cabezas et al.

103. Fendler W, Malachowska B, Baranowska- 117. Byrne MM, Sturis J, Menzel S et al. Altered
Jazwiecka A et al. Population-based estimates for insulin secretory responses to glucose in diabetic and
double diabetes amongst people with glucokinase nondiabetic subjects with mutations in the diabetes
monogenic diabetes, GCK-MODY. Diabet Med 2014: susceptibility gene MODY3 on chromosome 12.
31: 881883. Diabetes 1996: 45: 15031510.
104. Pearson ER, Pruhova S, Tack CJ et al. Molecular 118. Fajans SS, Brown MB. Administration of sulfony-
genetics and phenotypic characteristics of MODY lureas can increase glucose-induced insulin secretion
caused by hepatocyte nuclear factor 4alpha mutations for decades in patients with maturity-onset diabetes of
in a large European collection. Diabetologia 2005: 48: the young. Diabetes Care 1993: 16: 12541261.
878885. 119. Shepherd M, Shields B, Ellard S, Rubio-Cabezas
105. Ostoft SH, Bagger JI, Hansen T et al. Incretin effect O, Hattersley AT. A genetic diagnosis of HNF1A
and glucagon responses to oral and intravenous glucose diabetes alters treatment and improves glycaemic
in patients with maturity onset diabetes of the young - control in the majority of insulin-treated patients.
type 2 and type 3. Diabetes 2014: Mar 27. [Epub ahead Diabet Med 2009: 26: 437441.
of print]. 120. Becker M, Galler A, Raile K. Meglitinide analogues
106. Isomaa B, Henricsson M, Lehto M et al. Chronic in adolescent patients with HNF1A-MODY (MODY
diabetic complications in patients with MODY3 3). Pediatrics 2014: 133: e775e779.
diabetes. Diabetologia 1998: 41: 467473. 121. Tuomi T, Honkanen EH, Isomaa B, Sarelin L,
107. Steele AM, Shields BM, Shepherd M, Ellard S, Groop LC. Improved prandial glucose control with
Hattersley AT, Pearson ER. Increased all-cause and lower risk of hypoglycemia with nateglinide than with
cardiovascular mortality in monogenic diabetes as a glibenclamide in patients with maturity-onset diabetes
result of mutations in the HNF1A gene. Diabet Med of the young type 3. Diabetes Care 2006: 29: 189194.
2010: 27: 157161. 122. Ostoft SH, Bagger JI, Hansen T et al. Glucose-
108. Bellanne-Chantelot C, Carette C, Riveline JP lowering effects and low risk of hypoglycemia in
et al. The type and the position of HNF1A mutation patients with maturity-onset diabetes of the young
modulate age at diagnosis of diabetes in patients when treated with a GLP-1 receptor agonist: a double-
with maturity-onset diabetes of the young (MODY)-3.
blind, randomized, crossover trial. Diabetes Care 2014:
Diabetes 2008: 57: 503508.
37: 17971805.
109. Harries LW, Ellard S, Stride A, Morgan NG,
123. Schmidt F, Kapellen TM, Wiegand S et al. Diabetes
Hattersley AT. Isomers of the TCF1 gene encoding
mellitus in children and adolescents with genetic
hepatocyte nuclear factor-1 alpha show differential
syndromes. Exp Clin Endocrinol Diabetes 2012: 120:
expression in the pancreas and define the relationship
579585.
between mutation position and clinical phenotype
124. Farmer A, Ayme S, de Heredia ML et al. EURO-
in monogenic diabetes. Hum Mol Genet 2006: 15:
WABB: an EU rare diseases registry for Wolfram
22162224.
syndrome, Alstrom syndrome and Bardet-Biedl
110. Stride A, Ellard S, Clark P et al. Beta-cell
dysfunction, insulin sensitivity, and glycosuria precede syndrome. BMC Pediatr 2013: 13: 130.
diabetes in hepatocyte nuclear factor-1alpha mutation 125. Domenech E, Gomez-Zaera M, Nunes V. Wol-
carriers. Diabetes Care 2005: 28: 17511756. fram/DIDMOAD syndrome, a heterogenic and molec-
111. Hamilton AJ, Bingham C, McDonald TJ et al. The ularly complex neurodegenerative disease. Pediatr
HNF4A R76W mutation causes atypical dominant Endocrinol Rev 2006: 3: 249257.
Fanconi syndrome in addition to a beta cell phenotype. 126. Barrett TG, Bundey SE, Macleod AF. Neurode-
J Med Genet 2014: 51: 165169. generation and diabetes: UK nationwide study of
112. Pearson ER, Boj SF, Steele AM et al. Macrosomia Wolfram (DIDMOAD) syndrome. Lancet 1995: 346:
and hyperinsulinaemic hypoglycaemia in patients with 14581463.
heterozygous mutations in the HNF4A gene. PLoS 127. Marshall SL, Edidin D, Sharma V, Ogle G, Arena
Med 2007: 4: e118. VC, Orchard T. Current clinical status, glucose
113. Flanagan SE, Kapoor RR, Mali G et al. Diazoxide- control, and complication rates of children and youth
responsive hyperinsulinemic hypoglycemia caused by with type 1 diabetes in Rwanda. Pediatr Diabetes 2013:
HNF4A gene mutations. Eur J Endocrinol 2010: 162: 14: 217226.
987992. 128. de Heredia ML, Cleries R, Nunes V. Genotypic
114. Kapoor RR, Locke J, Colclough K et al. Persistent classification of patients with Wolfram syndrome:
hyperinsulinemic hypoglycemia and maturity-onset insights into the natural history of the disease and
diabetes of the young due to heterozygous HNF4A correlation with phenotype. Genet Med 2013: 15:
mutations. Diabetes 2008: 57: 16591663. 497506.
115. Stanescu DE, Hughes N, Kaplan B, Stanley CA, 129. Zmyslowska A, Borowiec M, Fichna P et al.
De Leon DD. Novel presentations of congenital Delayed recognition of Wolfram syndrome frequently
hyperinsulinism due to mutations in the MODY genes: misdiagnosed as type 1 diabetes with early chronic
HNF1A and HNF4A. J Clin Endocrinol Metab 2012: complications. Exp Clin Endocrinol Diabetes 2014:
97: E2026E2030. 122: 3538.
116. Pearson ER, Starkey BJ, Powell RJ, Gribble 130. Inoue H, Tanizawa Y, Wasson J et al. A gene
FM, Clark PM, Hattersley AT. Genetic cause of encoding a transmembrane protein is mutated in
hyperglycaemia and response to treatment in diabetes. patients with diabetes mellitus and optic atrophy
Lancet 2003: 362: 12751281. (Wolfram syndrome). Nat Genet 1998: 20: 143148.

62 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764


Monogenic diabetes in children and adolescents

131. Khanim F, Kirk J, Latif F, Barrett TG. 146. Lalau JD. Lactic acidosis induced by metformin:
WFS1/wolframin mutations, Wolfram syndrome, and incidence, management and prevention. Drug Saf 2010:
associated diseases. Hum Mutat 2001: 17: 357367. 33: 727740.
132. Amr S, Heisey C, Zhang M et al. A homozygous 147. Laloi-Michelin M, Virally M, Jardel C et al.
mutation in a novel zinc-finger protein, ERIS, is Kearns Sayre syndrome: an unusual form of
responsible for Wolfram syndrome 2. Am J Hum Genet mitochondrial diabetes. Diabetes Metab 2006: 32:
2007: 81: 673683. 182186.
133. Bingham C, Hattersley AT. Renal cysts and diabetes 148. Superti-Furga A, Schoenle E, Tuchschmid P et al.
syndrome resulting from mutations in hepatocyte Pearson bone marrow-pancreas syndrome with insulin-
nuclear factor-1beta. Nephrol Dial Transplant 2004: dependent diabetes, progressive renal tubulopathy,
19: 27032708. organic aciduria and elevated fetal haemoglobin caused
134. Ulinski T, Lescure S, Beaufils S et al. Renal by deletion and duplication of mitochondrial DNA.
phenotypes related to hepatocyte nuclear factor-1beta Eur J Pediatr 1993: 152: 4450.
(TCF2) mutations in a pediatric cohort. J Am Soc 149. Raeder H, Haldorsen IS, Ersland L et al. Pancreatic
Nephrol 2006: 17: 497503. lipomatosis is a structural marker in nondiabetic
135. Edghill EL, Bingham C, Ellard S, Hattersley AT. children with mutations in carboxyl-ester lipase.
Mutations in hepatocyte nuclear factor-1beta and their Diabetes 2007: 56: 444449.
related phenotypes. J Med Genet 2006: 43: 8490. 150. Raeder H, McAllister FE, Tjora E et al. Carboxyl-
136. Raile K, Klopocki E, Holder M et al. Expanded ester lipase maturity-onset diabetes of the young is
clinical spectrum in hepatocyte nuclear factor 1b- associated with development of pancreatic cysts and
maturity-onset diabetes of the young. J Clin upregulated MAPK signaling in secretin-stimulated
Endocrinol Metab 2009: 94: 26582664. duodenal fluid. Diabetes 2014: 63: 259269.
137. Edghill EL, Bingham C, Slingerland AS et al. 151. Moran A, Pillay K, Becker D, Acerini CL.
Hepatocyte nuclear factor-1 beta mutations cause Management of cystic fibrosis related diabetes in
neonatal diabetes and intrauterine growth retardation: children and adolescents. Pediatr Diabetes 2014: 15:
support for a critical role of HNF-1beta in human 6576.
pancreatic development. Diabet Med 2006: 23: 152. Rebours V, Boutron-Ruault MC, Schnee M et al.
13011306. The natural history of hereditary pancreatitis: a
national series. Gut 2009: 58: 97103.
138. Bellanne-Chantelot C, Chauveau D, Gautier JF
153. Semple RK, Savage DB, Cochran EK, Gorden P,
et al. Clinical spectrum associated with hepatocyte
ORahilly S. Genetic syndromes of severe insulin
nuclear factor-1beta mutations. Ann Intern Med 2004:
resistance. Endocr Rev 2011: 32: 498514.
140: 510517.
154. Parker VE, Semple RK. Genetics in endocrinology:
139. Pearson ER, Badman MK, Lockwood CR et al.
genetic forms of severe insulin resistance: what
Contrasting diabetes phenotypes associated with hep-
endocrinologists should know. Eur J Endocrinol 2013:
atocyte nuclear factor-1alpha and -1beta mutations.
169: R71R80.
Diabetes Care 2004: 27: 11021107.
155. Musso C, Cochran E, Moran SA et al. Clinical
140. Tjora E, Wathle G, Erchinger F et al. Exocrine course of genetic diseases of the insulin receptor (type
pancreatic function in hepatocyte nuclear factor A and Rabson-Mendenhall syndromes): a 30-year
1beta-maturity-onset diabetes of the young (HNF1B- prospective. Medicine (Baltimore) 2004: 83: 209222.
MODY) is only moderately reduced: compensatory 156. Regan FM, Williams RM, McDonald A et al.
hypersecretion from a hypoplastic pancreas. Diabet Treatment with recombinant human insulin-like
Med 2013: 30: 946955. growth factor (rhIGF)-I/rhIGF binding protein-3
141. Haldorsen IS, Vesterhus M, Raeder H et al. Lack of complex improves metabolic control in subjects with
pancreatic body and tail in HNF1B mutation carriers. severe insulin resistance. J Clin Endocrinol Metab
Diabet Med 2008: 25: 782787. 2010: 95: 21132122.
142. Maassen JA, T Hart LM, Van Essen E et al. 157. Garg A. Acquired and inherited lipodystrophies. N
Mitochondrial diabetes: molecular mechanisms and Engl J Med 2004: 350: 12201234.
clinical presentation. Diabetes 2004: 53 (Suppl. 1): 158. Agarwal AK, Simha V, Oral EA et al. Phenotypic
S103S109. and genetic heterogeneity in congenital generalized
143. Guillausseau PJ, Dubois-Laforgue D, Massin P lipodystrophy. J Clin Endocrinol Metab 2003: 88:
et al. Heterogeneity of diabetes phenotype in patients 48404847.
with 3243 bp mutation of mitochondrial DNA 159. Weedon MN, Ellard S, Prindle MJ et al. An in-
(maternally inherited diabetes and deafness or MIDD). frame deletion at the polymerase active site of POLD1
Diabetes Metab 2004: 30: 181186. causes a multisystem disorder with lipodystrophy. Nat
144. Laloi-Michelin M, Meas T, Ambonville C et al. The Genet 2013: 45: 947950.
clinical variability of maternally inherited diabetes and 160. Chudasama KK, Winnay J, Johansson S et al.
deafness is associated with the degree of heteroplasmy SHORT syndrome with partial lipodystrophy due to
in blood leukocytes. J Clin Endocrinol Metab 2009: impaired phosphatidylinositol 3 kinase signaling. Am
94: 30253030. J Hum Genet 2013: 93: 150157.
145. Goto Y, Nonaka I, Horai S. A mutation in the 161. Owen KR, Donohoe M, Ellard S, Hattersley AT.
tRNA(Leu)(UUR) gene associated with the MELAS Response to treatment with rosiglitazone in familial
subgroup of mitochondrial encephalomyopathies. partial lipodystrophy due to a mutation in the LMNA
Nature 1990: 348: 651653. gene. Diabet Med 2003: 20: 823827.
Pediatric Diabetes 2014: 15 (Suppl. 20): 4764 63
Rubio-Cabezas et al.

162. Beltrand J, Beregszaszi M, Chevenne D et al. 177. Senee V, Chelala C, Duchatelet S et al. Mutations
Metabolic correction induced by leptin replacement in GLIS3 are responsible for a rare syndrome
treatment in young children with Berardinelli- with neonatal diabetes mellitus and congenital
Seip congenital lipoatrophy. Pediatrics 2007: 120: hypothyroidism. Nat Genet 2006: 38: 682687.
e291e296. 178. Rubio-Cabezas O, Jensen JN, Hodgson MI et al.
163. Simha V, Subramanyam L, Szczepaniak L et al. Com- Permanent neonatal diabetes and enteric anendo
parison of efficacy and safety of leptin replacement crinosis associated with biallelic mutations in
therapy in moderately and severely hypoleptinemic NEUROG3. Diabetes 2011: 60: 13491353.
patients with familial partial lipodystrophy of the 179. Rubio-Cabezas O, Minton JA, Kantor I, Williams
Dunnigan variety. J Clin Endocrinol Metab 2012: 97: D, Ellard S, Hattersley AT. Homozygous
785792. mutations in NEUROD1 are responsible for a
164. Alstrom CH, Hallgren B, Nilsson LB, Asander novel syndrome of permanent neonatal diabetes
H. Retinal degeneration combined with obesity, and neurological abnormalities. Diabetes 2010: 59:
diabetes mellitus and neurogenous deafness: a specific 23262331.
syndrome (not hitherto described) distinct from the 180. Solomon BD, Pineda-Alvarez DE, Balog JZ et al.
Laurence-Moon-Bardet-Biedl syndrome: a clinical, Compound heterozygosity for mutations in PAX6 in a
endocrinological and genetic examination based on patient with complex brain anomaly, neonatal diabetes
a large pedigree. Acta Psychiatr Neurol Scand Suppl mellitus, and microophthalmia. Am J Med Genet A
1959: 129: 135. 2009: 149A: 25432546.
165. Hearn T, Renforth GL, Spalluto C et al. Mutation 181. Bonnefond A, Vaillant E, Philippe J et al.
of ALMS1, a large gene with a tandem repeat encoding Transcription factor gene MNX1 is a novel cause
47 amino acids, causes Alstrom syndrome. Nat Genet of permanent neonatal diabetes in a consanguineous
2002: 31: 7983. family. Diabetes Metab 2013: 39: 276280.
166. Mokashi A, Cummings EA. Presentation and course 182. Flanagan SE, De Franco E, Lango Allen H
of diabetes in children and adolescents with Alstrom et al. Analysis of transcription factors key for mouse
syndrome. Pediatr Diabetes 2011: 12 (3 Pt 2): pancreatic development establishes NKX2-2 and
270275. MNX1 mutations as causes of neonatal diabetes in
167. Paisey RB, Geberhiwot T, Waterson M et al. man. Cell Metab 2014: 19: 146154.
Modification of severe insulin resistant diabetes in 183. Yoo HW, Shin YL, Seo EJ, Kim GH. Identification of
response to lifestyle changes in Alstrom syndrome. a novel mutation in the GLUT2 gene in a patient with
Eur J Med Genet 2014: 57: 7175. Fanconi-Bickel syndrome presenting with neonatal
168. Tobin JL, Beales PL. Bardet-Biedl syndrome: beyond diabetes mellitus and galactosaemia. Eur J Pediatr
the cilium. Pediatr Nephrol 2007: 22: 926936. 2002: 161: 351353.
169. Scheidecker S, Etard C, Pierce NW et al. Exome 184. Mandel H, Berant M, Hazani A, Naveh Y.
sequencing of Bardet-Biedl syndrome patient identifies Thiamine-dependent beriberi in the thiamine-
a null mutation in the BBSome subunit BBIP1 (BBS18). responsive anemia syndrome. N Engl J Med 1984:
J Med Genet 2014: 51: 132136. 311: 836838.
170. Guo DF, Rahmouni K. Molecular basis of the 185. Abdel-Salam GM, Schaffer AE, Zaki MS et al. A
obesity associated with Bardet-Biedl syndrome. Trends homozygous IER3IP1 mutation causes microcephaly
Endocrinol Metab 2011: 22: 286293. with simplified gyral pattern, epilepsy, and permanent
171. Abu-Safieh L, Al-Anazi S, Al-Abdi L et al. In search neonatal diabetes syndrome (MEDS). Am J Med
of triallelism in Bardet-Biedl syndrome. Eur J Hum Genet A 2012: 158A: 27882796.
Genet 2012: 20: 420427. 186. Wildin RS, Ramsdell F, Peake J et al. X-linked
172. Katsanis N, Ansley SJ, Badano JL et al. Triallelic neonatal diabetes mellitus, enteropathy and
inheritance in Bardet-Biedl syndrome, a Mendelian endocrinopathy syndrome is the human equivalent of
recessive disorder. Science 2001: 293: 22562259. mouse scurfy. Nat Genet 2001: 27: 1820.
173. Stoffers DA, Zinkin NT, Stanojevic V, Clarke 187. Yamagata K, Furuta H, Oda N et al. Mutations in
WL, Habener JF. Pancreatic agenesis attributable to the hepatocyte nuclear factor-4alpha gene in maturity-
a single nucleotide deletion in the human IPF1 gene onset diabetes of the young (MODY1). Nature 1996:
coding sequence. Nat Genet 1997: 15: 106110. 384: 458460.
174. Sellick GS, Barker KT, Stolte-Dijkstra I et al. 188. Vionnet N, Stoffel M, Takeda J et al. Nonsense
Mutations in PTF1A cause pancreatic and cerebellar mutation in the glucokinase gene causes early-onset
agenesis. Nat Genet 2004: 36: 13011305. non-insulin-dependent diabetes mellitus. Nature 1992:
175. Smith SB, Qu HQ, Taleb N et al. Rfx6 directs islet 356: 721722.
formation and insulin production in mice and humans. 189. Yamagata K, Oda N, Kaisaki PJ et al. Mutations in
Nature 2010: 463: 775780. the hepatocyte nuclear factor-1alpha gene in maturity-
176. DAmato E, Giacopelli F, Giannattasio A et al. onset diabetes of the young (MODY3). Nature 1996:
Genetic investigation in an Italian child with an 384: 455458.
unusual association of atrial septal defect, attributable 190. Horikawa Y, Iwasaki N, Hara M et al. Mutation
to a new familial GATA4 gene mutation, and neonatal in hepatocyte nuclear factor-1 beta gene (TCF2)
diabetes due to pancreatic agenesis. Diabet Med 2010: associated with MODY. Nat Genet 1997: 17: 384385.
27: 11951200.

64 Pediatric Diabetes 2014: 15 (Suppl. 20): 4764

You might also like