You are on page 1of 19

Acta Psychiatr Scand 2010: 122: 192–210  2010 John Wiley & Sons A/S

All rights reserved ACTA PSYCHIATRICA


DOI: 10.1111/j.1600-0447.2009.01529.x SCANDINAVICA

Review
An update on the role of glutamate in the
pathophysiology of depression
Mitchell ND, Baker GB. An update on the role of glutamate in the N. D. Mitchell, G. B. Baker
pathophysiology of depression. Department of Psychiatry, University of Alberta,
Edmonton, Canada
Objective: To review the literature on the involvement of glutamate
(Glu), including its interactions with other neurochemical systems, in
the pathophysiology of depression.
Method: A MEDLINE search using the terms glutamate, depression
and major depressive disorder, was performed.
Results: Alterations in proteins involved in glutamatergic signalling
are implicated in variations in behaviour in animal models of
depression. Drugs acting at Glu receptors appear to have
antidepressant-like effects in these models, and traditional Key words: glutamate; depression; pathophysiology;
antidepressant pharmacotherapies act on the glutamatergic system. neurobiology; affective disorders; neurotransmitters
Recent evidence from genetic studies and in vivo spectroscopy also Nicholas D. Mitchell, Department of Psychiatry, Uni-
correlate glutamatergic dysfunction with depression. Trials of N- versity of Alberta Hospital, 1E1 Walter C. Mackenzie
methyl-d-aspartate receptor antagonists in humans have provided Centre, 8440 112 Street, Edmonton, AB T6G 2B7,
mixed results. Canada.
Conclusion: A growing body of evidence indicates that the E-mail: ndm@ualberta.ca
glutamatergic system is involved in the pathophysiology of depression,
and may represent a target for intervention. Accepted for publication December 8, 2009

Summations
• While poorly understood, increasing evidence suggests that the glutamatergic system contributes to
the pathophysiology of depression.
• The role of the glutamatergic system in depression likely involves changes in signalling at multiple
receptors in various brain regions, primarily in the frontal and temporal lobes.
• Emerging evidence from therapeutic trials in humans and animal models suggests a role for drugs
targeting the glutamatergic system in the treatment of depression.

Considerations
• Most early evidence of glutamatergic dysfunction in depression arose from animal models and
postmortem studies.
• Technical limitations and the inclusion of heterogenous samples in some in vivo spectroscopy studies
lower the specificity of their results.
• Glutamatergic dysfunction is not limited to depression, and may represent a more general marker of
disequilibrium.

Recently, research into the neurobiology of depres-


Introduction
sion has focused on a number of non-monoamine
Early theories of depression focused primarily on neurotransmitters and neuromodulators (7–10),
biogenic amines (serotonin, norepinephrine and and among the most exciting avenues of investiga-
dopamine) (1–6), and most antidepressants cur- tion in this regard are those involving the excit-
rently available target monoamine neurotransmit- atory amino acid glutamate (Glu). This study will
ter reuptake mechanisms and ⁄ or receptors. provide an overview of studies in animal models

192
Glu in the pathophysiology of depression

and humans that represent increasing evidence that Glutamatergic neurotransmission occurs thro-
Glu dysfunction is involved in the neurobiology of ugh receptors (Table 1) which act as ion channels
depression. (ionotropic) and receptors which are linked to
intracellular second messenger systems (metabo-
tropic) (12, 14–16). Ionotropic Glu receptors are
Aims of the study
classified on the basis of pharmacologic affinities to
The primary purpose of this review is to summarize synthetic ligands. The N-methyl-d-aspartate
investigations on the role of the glutamatergic (NMDA), a-amino-3-hydroxy-5-methylisoxazole-
system in major depression. The secondary aims 4-propionic acid (AMPA), and kainate receptors
are to review the biochemistry of Glu and to are each composed of four subunits. Variation in
evaluate interactions with other neurochemical the expression of these various subunits gives rise to
systems, with a view to exploring proposed different response properties. Approximately 70%
mechanisms of depression. of the synapses in the mammalian brain contain
NMDA or AMPA receptors (17). These receptors
occur in particularly high density in the cerebral
Material and methods
cortex, hippocampus, amygdala, striatum and
A MEDLINE search was performed using the key septum. On a subcellular level, these receptors
words glutamate, depression and major depressive have unique responses to activation by Glu (12).
disorder for articles published up to and including Calcium influx through NMDA ion channels
June 2009. Articles were limited to those pub- activates a number of intracellular kinases and
lished in English. The references for selected phosphatases, thereby altering the characteristics of
articles were searched by hand for additional the synapse; however, neuronal death results from
relevant studies. The search was undertaken excess influx of calcium through the NMDA
between December 2008 and January 2009, and receptor occurring in the context of anoxia or
updated in June 2009. hypoglycaemia. Therefore, glutamatergic signalling
plays a role in both neuroplasticiy and excitotoxi-
city. AMPA receptors have a lower affinity for Glu
Results
than NMDA receptors, and are responsible for an
Biochemistry of glutamate initial excitatory potential when Glu is present in
the synapse. This change in membrane polarization
Glutamate is an amino acid neurotransmitter
leads to the removal of a magnesium ion from the
which cannot pass through the blood–brain barrier
channel in the NMDA receptor which must occur
(11) and must therefore be produced centrally in
before calcium transit is possible. Kainate receptors
the neurones and glia (12). Glu is primarily derived
play a role in synaptic neurotransmission, but the
from glucose and a-ketoglutarate, with a small
exact nature of this role is not yet clear. Metabo-
amount created from glutamine (Gln) (Fig. 1).
tropic Glu (mGlu) receptors are structurally differ-
Glial cells synthesize Gln, which is then trans-
ent from ionotropic receptors, consisting of seven
ported to the neurones where glutaminase converts
transmembrane domains (18). The eight currently
it to Glu. It is then packaged into secretory vesicles
identified mGlu receptors are classified in three
by the vesicle Glu transporters (VGLUT). These
groups based on the intracellular cascades that they
vesicles merge with the cellular membrane, releas-
are coupled with, sequence homology and pharma-
ing Glu into the synapse. Glutamatergic activity is
cology. The type I mGlu receptors (mGlu1 and
regulated through the removal of Glu from the
mGlu5) are present both presynaptically and post-
synapse by excitatory amino acid transporters
synaptically, but the type II (mGlu2 and mGlu3)
(EAAT). Some Glu returns to the presynaptic
and type III (mGlu4, mGlu6, mGlu7 and mGlu8)
neurones, but most is collected into astrocytes via
receptors occur on glial cells and presynaptic
this mechanism (13).
neurones where they act to regulate Glu release
(19). These receptors are thought to have multiple
Glutamate functions related to their membrane location on the
Glutaminase Decarboxylase
neurone and density in different regions of the
Glutamine Glutamate -Aminobutyric
brain, both of which vary by type.
Glutamine acid (GABA)
Synthetase Transaminase

Investigations in animal models


-Ketoglutarate
Numerous animal models have been employed to
Fig. 1. Metabolism of glutamate. investigate the pathophysiology of major depres-

193
Mitchell and Baker

Table 1. Types of glutamate receptors

Receptor Subunits Signalling mechanism Location

Ionotropic glutamate receptors


N-methyl-D-aspartate (NMDA) NR1, NR2 and NR3 Calcium channel Postsynaptic
a-Amino-3-hydroxy-5- GluR1, GluR2 and GluR3, Sodium channel (primary) calcium channel (subset) Postsynaptic
methylisoxazole-4-propionic GluR4
acid (AMPA)
Kainate GluR5, GluR6, GluR7, KA1 Sodium channel Pre- and postsynaptic
and KA2
Metabotropic glutamate receptors (mGlu)
Type I mGlu (mGlu1, mGlu5) G-protein coupled to phosphatidylinositol ⁄ calcium pathway and Pre- and postsynaptic
diacylglycerol ⁄ protein kinase-C pathway
Type II mGlu (mGlu2 and mGlu3) G-protein coupled to adenylyl cyclase Presynaptic
Type III mGlu (mGlu4, mGlu6, G-protein coupled to adenylyl cyclase and phosphodiesterase Presynaptic
mGlu7 and mGlu8)

sion (10). Perturbations in Glu levels and gluta- mGlu7 receptor show a similar decrease in depres-
matergic neurotransmission have been examined in sion-like behaviours (27, 28). One study using rats
these systems. The forced swim test is a model of bred to spontaneously demonstrate a depressive
depressive-like behaviour and a screening test for phenotype showed decreased expression of the
antidepressants (20, 21). In one study of rats mGlu2 ⁄ 3 receptor in the hippocampus compared
exposed to an adaptation of the forced swim test, with depression-resistant controls (29). Chronic
chronic forced swimming stress, decreases in Glu stress in rats, modelled through restraint, increased
levels in the prefrontal cortex and the hippocampus the expression of the KA1 subunit of the kainate
were measured with ex vivo magnetic resonance receptor in the same brain region (30). In another
spectroscopy (MRS) (22). However, in another experiment on chronic stress in rats, mGlu5 recep-
model (23), animals bred to express congenital tor expression was increased in the CA1 region of
learned helplessness showed increases in the ratio the hippocampus, but decreased in the CA3 region
of Glu ⁄ c-aminobutyric acid (GABA) in the (31). Ryan et al. (32) evaluated the interaction of
prefrontal cortex and hippocampus. These ratios genetic predisposition and environmental stress on
normalized when rats were administered either of the hippocampus of rats. At baseline, lower
two effective treatments for depression in humans, expression of the NR1 subunit of the NMDA
desipramine or electroconvulsive shock. While the receptor was present in the genetically sensitive
quality of change in Glu levels appears to differ in rats. When exposed to early maternal separation, a
these two studies, dysregulation of Glu was present form of early life stress, expression of NR1and the
in both. mGlu2 ⁄ 3 receptor increased in sensitive rats but not
Alterations in proteins associated with glutama- in genetically resistant rats. The expression of Glu
tergic neurotransmission in animals influence, and receptors and their component subunits varies in
are influenced by, depressive-like behaviours response to genetic and environmental pressures,
(Table 2). Knockout studies involve the isolation and these fluctuations are associated with depres-
and removal of a single gene product, allowing for sion in animal models.
evaluation of the effect of that protein on an Administration of glutamatergic drugs influ-
organismÕs physiology and behaviour. The vesicu- ences the behaviour of animals in models of
lar Glu transporter protein VGLUT1 is found in depression (Table 3). Antidepressant effects
the cortex and hippocampus where it plays a observed when competitive and non-competitive
primary role in loading Glu into synaptic vesicles NMDA receptor antagonists were administered in
for release, thus affecting the efficacy of glutama- a rat model of inescapable stress (33) provided
tergic neurotransmission in these regions. A line of early evidence for involvement of Glu in depres-
mice bred for reduced VGLUT1 expression had sion. The NMDA receptor antagonist ketamine
increased depressive-like behaviours in a forced reduces immobility in the forced swim test in rats
swim test (24) and when exposed to chronic mild (34). Similarly, riluzole, a molecule that inhibits the
stress (25). Antidepressant effects in forced swim release of Glu from presynaptic neurones, reverses
and tail suspension tests are observed in knockout decreases in glial metabolism, decreases in glial
mice deficient in the NR2A NMDA receptor fibrillary associated protein mRNA expression,
subunit (26), suggesting that this particular subunit and behavioural effects of chronic unpredictable
is involved in the modulation of emotional behav- stress in rats (35). The antibiotic ceftriaxone is
iours. Mice that do not express the gene for the known to increase Glu reuptake and prevents the

194
Glu in the pathophysiology of depression

development of depression-like behaviours in mice

Increased mGlu5 receptor expression in the CA1 region of the hippocampus, but decreased

Decreased expression of VGLUT1 in the frontal cortex and hippocampus of heterozygous


exposed to a tail suspension test or a forced swim

animals, associated with enhanced depressive-like behaviour on the forced swim test
Decreased depressive-like behaviours in the forced swim test and tail suspension test

Decreased depressive-like behaviours in the forced swim test and tail suspension test

in heterozygous animals. Antidepressant effects were observed with intraperitoneal


Increased NR1 and mGlu2 ⁄ 3 expression in sensitive rats with maternal separation
test (36). Compounds with activity at mGlu recep-

Decreased VGLUT1 and EAAT1 expression and increased depression-like behaviour


Increased expression in the hippocampus of animals exposed to chronic restraint
tors are also being investigated in animal models

Decreased expression of mGlu2 ⁄ 3 receptors in genetically sensitive animals


for antidepressant effects. Antagonists at type II

Lower baseline expression of NR1 subunit in genetically sensitive animals.


mGlu receptors and agonists at type III mGlu
receptors reduce depression-like behaviour (37–
39). AMN082, a specific agonist at the mGlu7
receptor, has antidepressant-like effects in wild
type rats, but has no behavioural effect on mGlu7
Results

expression in the CA3 level of the hippocampus

receptor knockout rats (40). This suggests that


there may be a specific role of the mGlu7 receptor
in depression. In one study examining sleep archi-

*Chronic mild stress in this model included: food or water deprivation, 45 cage tilt, intermittent illumination, soiled cage, paired housing, and low intensity stroboscopic illumination.
tecture, the mGlu2 ⁄ 3 receptor agonist LY354740
significantly suppressed rapid eye movement
(REM) sleep in rats and increased its latency of
onset (41). Although the clinical relevance of this
imipramine exposure

finding is not yet known, unipolar depression in


in knockout mice

in knockout mice

humans is accompanied by an increase in total


REM sleep and a decrease in REM latency. As
well, LY379268, another mGlu2 ⁄ 3 receptor agonist,
acts synergistically with fluoxetine to increase
neurogenesis in cultures of granular cells from the
transporter-1 (EAAT1)
NR1 subunit, mGlu2 ⁄ 3

rat cerebellum (42, 43). Neurogenesis, primarily in


Protein of interest

VGLUT1, excitatory
mGlu2 ⁄ 3 receptor

the hippocampus, is increasingly recognized as


mGlu5 receptor

mGlu7 receptor

NR2A subunit

amino acid

playing a role in the pathophysiology and treat-


KA1 subunit

ment of depression (44–46). Antagonists at the


VGLUT1

mGlu1 and mGlu5 receptors decrease the amount


of time mice spend immobile during forced swim
and hippocampus

and tail suspension tasks, a marker for antidepres-


Brain region
Table 2. Studies of proteins associated with glutamatergic neurotransmission in animal models of depression

sant activity, when used alone (47, 48) or in


Frontal cortex
Hippocampus

Hippocampus

Hippocampus

Hippocampus
Hippocampus
CA1 region

combination with minocycline, an antibiotic


which reduces glutamatergic neurotransmission
N ⁄A

N ⁄A

(49, 50). The potent mGlu5 receptor antagonist


2-methyl-6-(phenylethyl)-pyridine also produces
Genetic sensitivity and
Model of depression

maternal separation
Chronic mild stress*

Chronic mild stress*

antidepressant effects in a passive avoidance task


Genetic sensitivity

Chronic restraint

in rats similar to that seen with desipramine, a


tricyclic antidepressant (51). Although animal
studies provide provocative results, the effects of
TST

TST
FST

FST

FST

these same compounds on glutamatergic neuro-


transmission and depressive symptoms in humans
NR2A subunit knockout (NR2A) ⁄ ))

remain to be determined.
Adult male sprague-dawley rats
Male flinders sensitive line and

Male flinders sensitive line and


Vesicular glutamate transporter

Vesicular glutamate transporter

Traditional antidepressant therapies, which are


heterozygous (VGLUT1+ ⁄ ))

heterozygous (VGLUT1+ ⁄ ))
(mGlu7) ⁄ ))

flinders resistant line rats

flinders resistant line rats

known to affect monoamine neurotransmitter sys-


Population

Adult male Wistar rats

tems, also influence the glutamatergic system.


and wild type mice

and wild type mice

and wild type mice

and wild type mice

FST, forced swim test; TST, tail suspension test.

Using microdialysis to directly observe changes in


mGlu7 knockout

Glu in the amygdalae of rats, Reznikov et al. (52)


demonstrated increases in Glu related to acute
stress. These elevations were not affected by the
selective serotonin reuptake inhibitor (SSRI) fluo-
xetine, but were inhibited by tianeptine, an antide-
Boyce-Rustay et al. (26)

Matrisciano et al. (29)


Wieronska et al. (31)

pressant that increases serotonin reuptake in the


Tordera et al. (24)

Hunter et al. (30)


Garcia et al. (25)
Cryan et al. (27)

cortex and hippocampus, suggesting that this


Ryan et al. (32)

medication may mediate its effects through the


glutamatergic system. Tianeptine has antidepres-
Study

sant effects in animal models and in humans, and is

195
Table 3. Studies using glutamatergic agents in animal models of depression

196
Study Population Model Experimental agent Neurochemical activity Route Results

Trullas et al. (33) Male NIH-Swiss mice FST 2-Amino-7-phosphonoheptanoic Competitive NMDA receptor Intraperitoneal All three compounds reduced depressive-
TST acid (AP7) antagonist like behaviour in the FST. Only ACPC was
tested in the TST, and resulted in
decreased depressive-like behaviour
Dizolcipine Non-competitive NMDA
Mitchell and Baker

receptor antagonist
1-Aminocylopropanecarboxylic NMDA receptor partial
acid (ACPC) agonist
Wieronska et al. (51) Male wistar rats Olfactory bulbectomy 2-Methyl-6-(phenylethynyl)-pyri mGlu5 receptor antagonist Intraperitoneal Decreased depressive-like behaviour in
and passive avoid- dine (MPEP) animals following administration of MPEP
ance
Yilmaz et al. (34) Male wistar rats FST Ketamine Non-competitive NMDA Intraperitoneal Decreased depressive-like behaviour in
receptor antagonist ketamine treated animals
Chaki et al. (37) Male ICR mice FST (1R,2R,3R,5R,6R)-2-amino-3-(3,4- mGlu2 ⁄ 3 receptor antagonist Intraperitoneal Decreased depressive-like behaviour in
TST dichlorobenzyloxy)-6-fluorobicy animals administered MGS0039.
Elevated Plus Maze clo[3.1.0]hexane-2,6-dicarboxylic
acid (MGS0039)
Palucha et al. (38) Male wistar rats FST (1S,3R,4S)-1-aminocyclo-pentane- Type III mGlu receptor agonist Infusion into Administration of each compound produced
1,3,4-tricarboxylic acid (ACPT-I) hippocampus decreases in depression-like behaviour
2-Amino-4-(3-hydroxy-5meth- Type III mGlu receptor agonist
ylisoxazol-4-yl)butyric acid
(HomoAMPA)
Palucha et al. (47) Male wistar rats FST 3-[(2-Methyl-1,3-thiazol-4-yl) mGlu5 receptor antagonist Intraperitoneal Decreased depressive-like behaviour was
TST ethinyl]-pyridine (MTEP) seen on the tail suspension test but not
the FST following administration of MTEP
Belozertseva et al. (48) Male wistar rats FST (JNJ16567083 3-ethyl-2-methyl- mGlu1 receptor antagonist Intraperitoneal Both compounds reduced depressive-like
quinolin-6-yl)-(4-methoxy-cyclo- behaviour on the FST
hexyl)-methanone methanesulfo-
nate (EMQMCM)
MTEP mGlu5 receptor antagonist
Male C57BL ⁄ 6J ⁄ Han mice TST EMQMCM mGlu1 receptor antagonist All compounds reduced depression-like
behaviour in the TST
MPEP mGlu5 receptor antagonist
MTEP mGlu5 receptor antagonist
Klak et al. (39) Male wistar rats FST ())-N-phenyl-7-(hydroxamino) Positive allosteric modulator Intra-cerbroventricular ACPT-I alone reduced depression-like
cyclopropa(b)chromen-1a-carbox- of mGlu4 receptor inejctions behaviour in the FST at effective doses.
amide (PHCCC) PHCCC coadministered with non-effective
doses of ACPT-I also reduced
depression-like behaviour
ACPT-I Type III mGlu receptor agonist
Mineur et al. (36) Male C57BL ⁄ 6J mice FST Ceftriaxone Increase Glu uptake Intraperitoneal Decreased depressive-like behaviour in
TST animals treated with Ceftriaxone
Palucha et al. (40) mGlu7 knockout (mGlu7) ⁄ )) FST N,NÕ-dibenzyhydryl-ethane-1,2-de- mGlu7 receptor agonist Intraperitoneal Decreased depressive-like behaviour in
and Wild type mice TST amine dihydrochloride (AMN082) AMN082 treated wild type mice. No
behavioural effect seen in knockout mice
Table 3. Continued

Study Population Model Experimental agent Neurochemical activity Route Results

Banasr et al. (35) Male Sprague-Dawley rats Chronic unpredictable Riluzole Inhibitor of Glu release Intraperitoneal Decreased depressive-like behaviour in rats
stress treated with Riluzole. Decreases in glial
metabolism seen with chronic
unpredictable stress were reversed upon
treatment with riluzole
Matrisciano et al. (43) Cerebellar cell cultures of TST (1R,4R,5S,6R)-4-amino-2-oxabicy- mGlu2 ⁄ 3 receptor agonist N ⁄A Increased neurogenesis as a result of
flinders sensitive line rats clo[3.1.0]hexane-4,6-dicarboxylic administration of either LY379268 or
acid (LY379268) fluoxetine. A strong synergistic effect was
noted
Fluoxetine Selective serotonin reuptake
inhibitor
Molina-Hernandez et al. (49) Adult male Wistar rats FST Minocycline Reduces glutamatergic Intraperitoneal All drugs reduced depressive-like behaviour
neurotransmission similar to the effect of desipramine, and
fluoxetine
Dizolcipine Non-competitive NMDA
receptor antagonist
EMQMCM mGlu5 receptor antagonist
MTEP mGlu5 receptor antagonist
Desipramine Tricyclic antidepressant
Fluoxetine SSRI antidepressant
Molina Hernandez et al. (50) Adult male Wistar rats Minocycline Reduces glutamatergic Infusion into nucleus accum- All drugs reduced depressive-like behaviour
neurotransmission bens similar to the effect of desipramine and
fluoxetine
Dizolcipine Non-competitive NMDA
receptor antagonist
EMQMCM mGlu5 receptor antagonist
MTEP mGlu5 receptor antagonist
Desipramine TCA
Fluoxetine SSRI
Ahnaou et al. (41) mGlu2 knockout (mGlu2) ⁄ )) Sleep–wake cycle (1S,2S,5R,6S)-2-aminobicy- mGlu2 ⁄ 3 receptor agonist Subcutaneous Each compound significantly decreased
and wild type male Sprague- clo[3.1.0]hexane-2,6-dicarboxylic total REM sleep and REM latency in WT,
Dawley rats acid LY354740 but not knockout animals. When
administered together, a synergistic effect
was noted
Biphenylindanone A (BINA) Positive allosteric modulator
of mGlu2 receptor

FST, forced swim test; TST, tail suspension test; WT, wild type.

197
Glu in the pathophysiology of depression
Mitchell and Baker

known to also exert effects on the signal transduc- Chronic administration of antidepressants also
tion pathways activated by Glu receptors (53, 54). has the capacity to alter expression of proteins
In a study employing in vivo MRS in rats, involved in glutamatergic neurotransmission in
phenelzine, a monoamine oxidase inhibitor, dem- animal brains (Table 4). Intraperitoneal adminis-
onstrated an acute decrease in the rate of cycling of tration of paroxetine, an SSRI, over 3 weeks
Glu–Gln between the neurones and glia following resulted in increased expression of mGlu1,
exposure (55). This observation was made in non- mGlu2 ⁄ 3 and AMPA receptors in the hippocampus
depressed rats, and the effect on behaviour is of rats (61). Elevations in the GluR1, GluR2 and
unknown. In a study of mice bred to express GluR3 subunits of the AMPA receptor in the
reduced levels of VGLUT1, intraperitoneal imip- hippocampus occur in mice following a 30-day
ramine reversed depression-like behaviours in a administration of maprotiline, an antidepressant
model of chronic mild stress (22). Postmortem that primarily inhibits reuptake of noradrenaline
samples of rat prefrontal cortex showed decreased (62). However, a study investigating the effect of
K+-stimulated Glu outflow in rats exposed to the antidepressants imipramine and citalopram
either imipramine or phenelzine (56), which the found no changes in Glu transporter expression
authors suggest could be as a result of effects on in any cortical regions of mice when these drugs
Glu receptor expression. Indeed, a subsequent were administered for 4 weeks (63). Electroconvul-
study involving administration of imipramine for sive shock administered to rats in one study
21 days resulted in decreased inhibitory autorecep- population resulted in increases in the EAAT1
tor activity of mGlu2 and mGlu3 receptors in the which is responsible for reuptake of Glu from the
prefrontal cortex (57). In the rat hippocampus, synapse (64). None of these studies used animals
chronic adminstration of fluoxetine results in exposed to behavioural models of depression, and
increased K+-stimulated Glu release in control conclusions regarding the effects of these com-
animals, and appears to counteract a decrease pounds in depressed brains cannot be directly
occurring in rats exposed to a model of learned extrapolated from these findings.
helplessness (58). The potential antidepressant
compound LY367265, which acts as a serotonin
Investigations in humans
2A receptor agonist and inhibitor of the serotonin
transporter protein, also decreases Glu release in Evidence of dysregulation of Glu in depression in
samples of rat cerebral cortex (59). Transcranial humans has been mounting in recent years (65).
magnetic stimulation (TMS), a somatic therapy Parallel to studies in animal models, investigations
with evidence of antidepressant effect in humans, have associated depressive phenotypes in humans
administered over the frontal cortex of rats results with changes in levels of Glu and its metabolites
in increased Glu and dopamine release in the (Table 5). An early study by Kim et al. (66)
nucleus accumbens (60). The relationship of this reported elevations in plasma Glu in individuals
finding to the therapeutic effect of TMS has not with major depression compared with healthy
been determined. controls. However, this study did not differentiate

Table 4. Studies of the effects of antidepressant treatments on the glutamatergic system in animals

Study Population Antidepressant Class Route Protein of interest Results

Martinez-Turillas Male Wistar rats Paroxetine SSRI Intraperitoneal AMPA, mGlu1, and mGlu2 ⁄ 3 Increased expression in the hippocampus
et al. (61) receptors following administration for 3 weeks
Williams et al. Adult Quackenbush Imipramine TCA Intraperitoneal Glutamate aspartate No changes in glutamate transporter levels
(63) mice transporter (GLAST) and following 4-week administration of
glutamate transporter-1 (GLT-1) antidepressants
Citalopram SSRI
Ploski et al. (64) Male Sprague- Electroconculsive N ⁄ A Bilateral EAAT1 Increases in EAAT1 levels following
Dawley rats shock administration daily for 10 days
Tan et al. (62) Adult mice Maprotiline TCA Intraperitoneal GluR1, GluR2, GluR3 subunits Increased expression of AMPA subunits
of the AMPA receptor following a 30-day administration of
Maprotiline
Garcia et al. (25) Vesicular glutamate Imipramine TCA VGLUT1, excitatory VGLUT1, EAAT1 Decreased VGLUT1 and EAAT1 expression
transporter amino acid in the hippocampus and increased
heterozygous transporter-1 depression-like behaviour in heterozygous
(VGLUT1+ ⁄ )) and (EAAT1) animals. Antidepressant effects were
wild type mice observed with intraperitoneal imipramine
exposure

198
Glu in the pathophysiology of depression

Table 5. Measurements of glutamate in human depression

Study Population Primary outcome measure Results

Kim et al. (66) Neurotic depression Plasma Glu Elevated levels of plasma Glu in depressed individuals
Endogenous depression Schizophrenia
Schizoaffective disorder
Francis et al. (73) Treatment-resistant major depression Glu in neurosurgical samples No difference in Glu compared with control neurosurgical samples
undergoing psychosurgery of frontal cortex
Altamura et al. (69) Unmedicated major depression Plasma Glu No difference in plasma Glu
Maes et al. (70) Treatment-resistant major depression Plasma Glu No baseline difference in plasma Glu, treatment with antidepressants
for 5 weeks reduced levels of Glu
Mauri et al. (67) Major depression Plasma Glu Elevated plasma Glu in depressed individuals, no influence of
treatment with fluvoxamine
Levine et al. (75) Unmedicated major depression CSF Gln Increased CSF Gln in depressed individuals
Berk et al. (109) Unmedicated major depression Platelet Glu receptor response Hypersensitivity of platelet NMDA receptors in depressed individuals
Mitani et al. (68) Major depression on Plasma Glu Elevated plasma Glu in depressed individuals, severity of depression
Medication correlates with degree of increase
Frye et al. (74) Major depression bipolar I CSF Glu Decreased CSF Glu in individuals with mood disorders
Bipolar II
Hashimoto et al. Major depression-postmortem samples Glu in frontal cortex Increased Glu in the frontal cortex of patients with major depression
(72) compared with controls

between medicated and unmedicated subjects. sive Disorder. One study found increased levels of
Similarly, elevated levels of plasma and platelet Gln in the CSF associated with depression (75).
Glu have been observed in outpatients with major Magnetic resonance spectroscopy is the only
depression (67), and in one recent study these currently available imaging technique that allows
concentrations were directly proportional to the for real-time in vivo quantification of brain meta-
severity of depressive symptoms (68). Other studies bolites (76). Identification of Glu using MRS in
have not associated elevations in Glu with depres- humans is technically challenging, as the spectrum
sion (69, 70). Maes et al. (70) measured serum of Glu overlaps with a number of other neuro-
levels in patients with treatment-resistant depres- chemicals, primarily Gln. Stronger magnetic fields
sion, and reported no differences from concentra- and advanced imaging techniques allow for the
tions in healthy controls, but after 5 weeks of isolation of the Glu signal; however, early MRS
treatment with the serotonergic antidepressants studies report the combined Glu + Gln peak as
trazodone and fluoxetine, Glu levels were signifi- ÔglutamixÕ (Glx) (77). The first clinical report of
cantly decreased from baseline. A study using MRS used to examine Glx in depression was of a
electroshock therapy (ECT) showed that plasma cancer patient who had recurrent suicidal ideation
Glu in depressed patients was increased 6 h after and depressive symptoms associated with chemo-
treatment, which the authors suggested may be therapy (78). In this individual, Glx was decreased
related to the mechanism of action of ECT (71). in the cerebral white matter. Various brain regions
Measurements of Glu in the central nervous have been targeted in investigations using MRS in
system come from postmortem analysis and cere- depression (Table 6). Imaging of the anterior
brospinal fluid (CSF) collection. In a study of cingulate cortex shows decreased levels of Glx in
postmortem tissue from the human frontal cortex, depressed adults (79) and children (80, 81) which
Glu levels were elevated in subjects with a history resolves following clinical recovery (82). Interest-
of depression compared with controls (72). How- ingly, Glx appears to be elevated in the cingulate
ever, subjects were not necessarily depressed at the cortex of depressed bipolar patients (83), and this
time of death. Conversely, in neurosurgical sam- may be a biological marker distinguishing the two
ples of the frontal cortex taken from patients disorders. One conflicting study found no differ-
undergoing psychosurgery for active major depres- ences in Glx in the anterior cingulate or occipital
sion, there were no differences in Glu concentra- cortex between individuals with treatment-resistant
tions compared with tissue samples from major depression, individuals with major depres-
non-depressed controls (73). Frye et al. (74) sion without treatment-resistance, and healthy
reported decreased levels of Glu in the CSF of controls (84). A 2006 meta-analysis of published
depressed individuals from a mixed population of studies found a significant decrease in Glx in
unipolar and bipolar mood disorders. Individuals frontal structures in depressed adults (85). Glx is
with unipolar depression represented the minority reduced bilaterally in the dorsolateral prefrontal
of the cohort, and these results have not been subcortical white matter in patients with type II
replicated in a population of purely Major Depres- diabetes and depression (86) and in the left

199
Mitchell and Baker

Table 6. Magnetic resonance spectroscopy imaging studies of glutamate in human depression

Study Population Region of interest Results

Auer et al. (79) Major depression Anterior cingulate cortex Reduced Glx in individuals with MDD
Michael et al. (88) Treatment-resistant major depression Left Amygdala Reduced Glx in individuals with MDD which resolved
with successful treatment with ECT
Michael et al. (89) Major depression with melancholic Left dorsolateral prefrontal cortex Reduced Glx in individuals with MDD which resolved
features with successful treatment with ECT
Pfleiderer et al. (90) Major depression Anterior cingulate cortex Reduced Glx in individuals with MDD which resolved
with successful treatment with ECT
Rosenburg et al. (80) Paediatric major depression with Anterior cingulate cortex Reduced Glx in individuals with MDD and OCD compared
comorbid OCD with those with OCD only
Sanacora et al. (96) Major depression Occipital cortex Increased Glx in individuals with MDD
Rosenburg et al. (81) Paediatric major depression Anterior cingulate cortex Reduced Glx in individuals with MDD
Glodzik-Slobanska et al. Poststroke depression Frontal lobe (contralateral to side Increased Glx in individuals with poststroke depression
(97) of stroke)
Luborzewski et al. (91) Major depression Dorsolateral prefrontal cortex Decreased Glu in the dorsolateral prefrontal cortex of
Anterior cingulate cortex individuals with MDD which increased following
transcranial magnetic stimulation (TMS) no changes
in the Anterior cingulate cortex with TMS
Ajilore et al. (86) Major depression and type II diabetes Dorsolateral prefrontal subcortical Decreased Glx in individuals with MDD
white matter
Hasler et al. (93) Major depression Ventromedial and dorsomedial Decreased Glx in individuals with MDD
prefrontal cortex
Batra et al. (98) Premenstrual dysphoric disorder Medial prefrontal cortex No difference in Glu between individuals with PMDD
and controls
Block et al. (94) Major depression Hippocampus Decreased Gln in individuals with MDD
Milne et al. (95) First major depressive episode Hippocampus No difference in Glx between first episode patients
Recurrent major depression and controls or recurrent MDD and controls
Murck et al. (92) Major depression Dorsolateral prefrontal cortex Increases in Glx and Gln in male responders to therapeutic
sleep deprivation and increases in Gln in responders
with melancholic signs
Price et al. (84) Major depression Occipital cortex No difference in Glx in either region in depressed individuals
Treatment-resistant major depression Anterior cingulate cortex with or without treatment resistance and controls
Taylor et al. (82) Recovered major depression Anterior cingulate cortex No difference in Glx between individuals with a history
of MDD and controls

MDD, major depressive disorder; OCD, obsessive compulsive disorder.

dorsolateral prefrontal cortex (DLPFC) in paedi- certain clinical subtypes (96). Other studies exam-
atric patients with major depression (87). In ining specific sub-populations have demonstrated
patients who respond to ECT, reduced Glx levels elevated Glx in frontal lobes of patients with
in the left amygdala (88), left DLPFC (89), and poststroke depression (97), and no difference in the
anterior cingulate cortex (90) resolve following medial prefrontal cortex between controls and
treatment. Responders to TMS in one trial showed individuals with premenstrual dysphoric disorder,
lower baseline Glu concentrations in the left a catamenial variant of MDD thought to be related
DLPFC which increased in a dose-dependent to fluctuations in ovarian hormones (98). Proteins
fashion after exposure (91). Therapeutic sleep involved in glutamatergic neurotransmission and
deprivation also increases Glx and Gln in the transport have also been investigated in mood
same brain area in male responders with major disorders in humans (Table 7).
depression and in responders with melancholic Genetic variability appears to be associated
depression (92). Hasler et al. (93) have also dem- with treatment response to antidepressant medi-
onstrated decreases in Glx in the ventromedial and cations. Data from the first phase of the
dorsomedial ⁄ dorsal anterolateral prefrontal Sequenced Treatment Alternatives to Relieve
regions of depressed individuals. Newly published Depression (STAR*D) revealed that the effective-
evidence suggests that Gln is decreased in the ness of citalopram was associated with a poly-
hippocampus of depressed individuals (94), but morphism in a gene encoding the KA1 subunit of
another study found no differences in Glx between the kainate receptor (99). A separate analysis of
newly depressed patients, those with multiple the STAR*D trial showed that treatment-emer-
episodes of low mood, or matched controls (95). gent suicidal ideation with the antidepressant
One study showed increases in Glu in the occipital citalopram was associated with two single
lobe of depressed individuals, and the authors nucleotide polymorphisms in genes encoding su-
suggested that this may depend on the presence of bunits of the AMPA receptor and the kainate

200
Table 7. Studies of proteins associated with glutamatergic neurotransmission in depression in humans

Study Population Sample type Brain region Protein of interest Results

Freed et al. (112) Schizophrenia, schizoaffective Postmortem Caudate nucleus AMPA receptor No difference between diagnostic groups and controls. AMPA binding in the caudate
disorder, and affective disorder tissue was significantly higher in victims of suicide
with psychosis
Including suicide victims
Holemans et al. (103) Suicide victims with a history of Postmortem Cerebral cortex NMDA receptor No differences in receptor density in the cortex compared with controls
major depression tissue
Nowak et al. (102) Suicide victims Postmortem Frontal cortex NMDA receptor Decreased high affinity binding compared with samples from controls
tissue
Noga et al. (113) Schizophrenia including suicide Postmortem Striatum AMPA receptor Increased AMPA receptor density in victims of suicide
victims tissue
Berk et al. (109) Major depression Platelet N ⁄A NMDA receptor Increased calcium release in response to Glu that was antagonised by dizocilpine.
sample This suggests platelet NMDA receptor hypersensitivity in depression
Dwivedi et al. (114) Suicide victims with a history Postmortem Prefrontal cortex Extracellular regulated Decreases in ERK1 ⁄ 2 and MAPK activity (which are coupled to AMPA receptors)
of major depression tissue Hippocampus kinase 1 ⁄ 2 (ERK1 ⁄ 2) in the prefrontal cortex and hippocampus of depressed individuals compared with
Cerebellum Mitogen activated protein controls. No differences in the cerebellum were noted
kinase (MAPK)
McCullumsmith et al. (108) Major depression, bipolar Postmortem Striatum Excitatory amino acid Decreased EAAT4 in the striatum of depressed individuals, decreased EAAT3 in
disorder, tissue transporter (EAAT) 1–4 schizophrenia, and decreased EAAT3 and EAAT4 in bipolar disorder compared
and schizophrenia with controls
Choudary et al. (110) Major depression and bipolar Postmortem Anterior cingulate cortex Glutamate transporter Decreased expression of the genes encoding glial glutamate transporters in the
disorder including suicides tissue Dorsolateral prefrontal cortex proteins, Multiple anterior cingulate and dorsolateral prefrontal cortex in major depression. Increased
AMPA and mGlu expression of GluR1 and KA2 in the cingulate, and increased expression of GluR3,
receptor subunits GluR5, and KA2 subunits in the dorsolateral prefrontal cortex in major depression
A different pattern of changes in gene expression was noted in bipolar disorder
Karolewicz et al. (106) Major depression or dysthymia Postmortem Locus ceruleus NR1 subunit Increased NR1 subunit of the NMDA receptor in depressed individuals
tissue
Kristiansen et al. (107) Major depression, bipolar disorder, Postmortem Striatum SAP-102 Decreased mRNA for SAP-102, a protein involved in intracellular signalling and
and schizophrenia tissue membrane localization of the NMDA in all three diagnostic groups
Beneyto et al. (115) Temporal lobe epilepsy and major Neurosurgical Hippocampus Ionotropic Glu No changes in transcript expression were observed in the hippocampus. In the
depression, bipolar disorder, or sample Entorhinal cortex receptor subunits entorhinal cortex, bipolar disorder was associated with decreased GluR2, GluR3,
schizophrenia Perirhinal cortex and GluR6 mRNA expression. In the perirhinal cortex decreased expression of GluR5
occurred in all three diagnoses. Decreases in NR1 occurred in bipolar disorder,
decreased NR2A occurred in major depression, and decreased GluR1, GluR3, and
NR2B were present in both
Laje et al. (100) Outpatients with major depression Genetic N ⁄A GluR3 subunit, Treatment-emergent suicidal ideation with citalopram was associated with
sample GluR6 subunit polymorphisms in genes encoding the GluR6 subunit of the kainate receptor
and the GluR3 subunit of the AMPA receptor
Paddock et al. (99) Outpatients with major depression Genetic N ⁄A KA1 subunit Treatment response with citalopram was associated with single nucleotide
sample polymorphisms (SNP) in genes encoding the KA1 subunit of the kainate receptor
Toro et al. (105) Temporal lobe epilepsy and major Neurosurgical Hippocampus NR1 subunit Increased NR1 subunit of NMDA receptor in the hippocampus of depressed and
depression or dysthymia sample dysthymic individuals
Feyissa et al. (104) Major depression including suicide Postmortem Prefrontal cortex NR2A subunit, Decreased NR2A and NR2B NMDA receptor subunits in the prefrontal cortex of
victims sample NR2B subunit individuals with depression
Menke et al. (101) Inpatients with major depression Genetic N ⁄A GluR3 subunit, Treatment-emergent suicidal ideation associated with SNP in genes for the GluR6
sample GluR6 subunit subunit of the kainate receptor and the GluR3 subunit of the AMPA receptor

201
Glu in the pathophysiology of depression
Mitchell and Baker

receptor (100). Treatment-emergent suicidal idea- temporal lobe epilepsy and mood disorders,
tion has also been associated with different genetic changes in expression of a number of Glu receptor
polymorphisms affecting the same subunits of Glu subunits occurred in the perirhinal cortex, but did
receptors (101). NMDA receptors in the frontal not differentiate between bipolar disorder and
cortex of patients who had died by suicide exhibi- unipolar depression (115).
ted decreased high affinity binding in a postmortem Some of the most compelling evidence comes
study (102), though no difference was found in from clinical trials with glutamatergic agents (116)
receptor density in any brain region in an earlier (Table 8). Ketamine, an antagonist at the NMDA
study (103). Alterations in the expression of receptor, has rapid and prolonged antidepressant
subunits of the NMDA receptor associated with effects in individuals with treatment-resistant
depression have been observed in the prefrontal depression after a single intravenous administra-
cortex (104), the temporal lobe (105) and the locus tion in randomized trials (117, 118). Riluzole,
ceruleus (106). Kristiansen et al. (107) recently another NMDA receptor antagonist, has been used
reported decreased expression of mRNA for SAP- in clinical trials for numerous psychiatric condi-
102, a protein associated with membrane localiza- tions (119), and showed antidepressant effects as
tion of the NMDA receptor and intracellular monotherapy in one trial (120), and as an aug-
signalling, in the striatum in depression. The menting agent in another trial (121). However, a
same region of the brain exhibits decreased tran- trial of riluzole following treatment with intrave-
scription of EAAT4, which is necessary for Glu nous ketamine showed no difference in time to
reuptake (108), in depressed individuals. These relapse when compared with placebo (122). In a
changes in glutamatergic activity are not limited to study of patients with comorbid alcohol depen-
the brain. A unique study by Berk et al. (109) dence and depression, both the NMDA receptor
demonstrated elevated platelet Glu receptor sensi- antagonist memantine and the SSRI escitalopram
tivity in depressed individuals. This was attenuated had similar antidepressant effects (123).
by the NMDA receptor antagonist dizocilpine,
suggesting a specific hyperactivity of the NMDA
Models of glutamate in depression
receptors. Postmortem brain samples of individuals
with Major Depressive Disorder show decreases in Multiple observations of glutamatergic dysfunc-
the expression of genes for glial Glu transporters in tion in animal models and humans provide evi-
the anterior cingulate and DLPFC, and increases dence for a role of Glu in the pathophysiology of
in expression of certain subunits of the AMPA and depression. Zarate et al. (118) suggest that obser-
kainate receptors in these same regions (110). vations of rapid antidepressant effects following
Conflicting data on the role of the AMPA receptor administration of intravenous ketamine may lead
come from postmortem studies (111). Increased to the development of rapid acting antidepressants.
AMPA receptor density occurs in the striatum in However, with numerous MRS studies showing
victims of suicide (112, 113), but the signalling decreases in Glu related to depression (22, 78–81,
pathways activated by AMPA are decreased in a 85–94), the relationship of the antagonistic activity
study of a similar population (114). Dysregulation of ketamine at the NMDA receptor to antidepres-
in the expression of Glu receptors is not limited to sant effects seems counterintuitive. A recent study
unipolar depression. In one study examining (124) demonstrated that pretreatment with 2,3-
neurosurgical samples taken from patients with dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-

Table 8. Clinical trials of glutamatergic drugs in human depression

Study Population Intervention Results

Berman et al. (117) Treatment-resistant major depression Intravenous ketamine vs. placebo Antidepressant response with ketamine greater
than with placebo
Zarate et al. (120) Recurrent major depression Oral riluzole vs. placebo Antidepressant effect with riluzole greater with
than placebo
Zarate et al. (118) Treatment-resistant major depression Intravenous ketamine vs. placebo Antidepressant response with ketamine greater
than with placebo
Sanacora et al. (121) Treatment-resistant major depression Augmentation of antidepressants with Antidepressant effect enhanced with riluzole
oral riluzole vs. placebo greater than with placebo
Muhonen et al. (123) Alcohol dependence and major depression Oral memantine vs. escitalopram Both interventions had similar antidepressant
effects
Mathew et al. (122) Treatment-resistant major depression Intravenous ketamine + oral riluzole vs. No change in time to relapse between groups
intravenous ketamine + placebo

202
Glu in the pathophysiology of depression

2,3-dione, an AMPA receptor antagonist, blocked function (136). The HPA axis is the primary
the antidepressant action of intravenous ketamine regulator of the physiological stress reaction.
in rats. The authors suggest that increased AMPA– Over-activity of the HPA axis has long been
NMDA receptor throughput is a necessary for the associated with depression, and recent research
antidepressant effect of this medication (125). In has focused on it as a potential target of interven-
the same study, phosphorylation of the GluR1 tion (8, 137). Interactions between the HPA axis
subunit of the AMPA receptor was significantly and Glu are recognized in the literature, with early
reduced following the administration of ketamine. investigations in animals noting that direct admin-
Normally, phosphorylation of the GluR1 subunit istration of Glu into the amygdala stimulates the
plays a role in desensitization of the AMPA release of corticotropin-releasing factor (CRF)
receptor in the presence of Glu (126), reducing (138), and that administration into the periventric-
glutamatergic neurotransmission. However, phos- ular nucleus (PVN) of the hypothalamus results in
phorylation also increases membrane expression of the secretion of adrenocorticotropic hormone
the AMPA receptor (127). It has been proposed (139). Systemic exposure to Glu agonists, specifi-
that the delayed effects of traditional antidepres- cally kainate and NMDA, activates the HPA axis
sant medications are mediated through increased in rats at the level of the PVN (140), and kainate
GluR1 phosphorylation (128, 129). In a review of receptor activity in this region mediates the release
clinical trials implicating Glu in mood disorders, of CRF (141). More recent investigations suggest
Sanacora et al. (65) suggest that dysregulation of that Glu receptors play a role in the regulation of
Glu seen in depression may be a result of glial the stress response (142). Intraperitoneal admin-
dysfunction. Decreased uptake of Glu by astro- istration of 2-methyl-6-(phenylethynyl)pyridine,
cytes would increase glutamatergic neurotransmis- a mGlu5 receptor antagonist, results in an increase
sion, and the antidepressant effect of NMDA in plasma corticosterone in rats (143). Similarly,
antagonists would be a result of increased activa- increases in corticosterone levels were observed
tion of AMPA and kainate receptors. Pilc et al. following intracerebroventricular infusions of the
suggest (130) that the antidepressant effects of non-selective mGlu receptor agonist (1S,3S)-1-am-
agents acting at mGlu receptors occur as a result of inocyclopentane-1,3-dicarboxylic acid (144). How-
dampening of NMDA receptor activity and ampli- ever, agonism of mGlu2 ⁄ 3 receptors in the
fication of AMPA receptor activity. While the hypothalamus inhibits the release of CRF (145).
exact mechanisms of glutamatergic involvement in Knockout mice failing to express the gene for the
depression is not clear, the contribution of Glu to mGlu7 receptor show dysregulation at numerous
the pathophysiology of depression likely involves sites within the HPA axis including at the level of
both ionotropic and metabotropic receptors. transcription of glucocorticoid receptors and at the
The glutamatergic system communicates with level of feedback mechanisms (28). These mice also
other potential neurochemical contributors to show anxiolytic and antidepressant-like effects, and
depression. While currently available antidepres- the authors suggest that changes in the HPA axis
sants affect monoamine systems rapidly, behavio- occurring because of removal of the mGlu7 recep-
ural effects are considerably delayed. One theory tor may account for these behavioural differences.
posits that the targets of these antidepressants are Alterations in HPA axis function also influence the
considerably upstream of the neurobiological sub- glutamatergic system. Rats administered a moder-
strates that eventually produce the antidepressant ate dose of corticosterone for 21 days show
effect (131). Downstream systems are eventually increased expression of KA1 subunit mRNA in
altered over time by changes in neurotransmission the dentate gyrus (30). Proteomic analysis of
occurring upstream, and Glu has been identified as mice brains following administration of corticoste-
a potential endpoint where these changes affect the rone for 14 days revealed changes in Glu metab-
depressive phenotype (132). This suggests interplay olism in the hippocampus, hypothalamus and
of Glu and monoamine systems. Glu in the normal cerebral cortex (146). Changes in the expression
brain operates in balance with GABA, and GAB- of various proteins involved in glutamatergic
Aergic neurones synapse on glutamatergic neuro- signalling are noted in various animal models
nes to modulate their activity (15). Parallel to the which use stress to induce depression-like behav-
evidence for glutamatergic dysfunction, there is iours (30–32). The function of the interplay of Glu
also growing evidence for dysregulation of GABA and the HPA axis in depression requires further
in depression (65, 133–135). elucidation.
The hypothalamic–pituitary–adrenal (HPA) axis Brain-derived neurotrophic factor (BDNF), the
regulates various peripheral processes, including most extensively studied neurotrophin in depres-
metabolism and immunity, and influences brain sion, is implicated in intracellular mechanisms

203
Mitchell and Baker

associated with neural plasticity (9, 46, 147, 148). tion and the effects of drugs acting on Glu
Decreased levels of BDNF are observed in the receptors appear to be quite different in schizo-
hippocampi of postmortem samples of depressed phrenia and major depressive disorder (163–165).
individuals (149) and in animal models of depres- The reasons for the differences are not clear at this
sion (150). An antidepressant effect follows direct time, but further investigations into the glutama-
infusion of BDNF into the hippocampus of rat tergic systems should provide valuable information
models of depression (151), and knockout animals about the aetiology and symptomatic overlap
that do not express BDNF fail to respond to related to these two disorders. This study has
antidepressant treatment (152). Indeed, chronic focused on recent research from both studies in
administration of most antidepressant therapies both humans and animal models that have con-
results in increased expression of BDNF in humans tributed to the increasing understanding of the role
(153, 154) and increased neurogenesis in the of Glu in the development of depression.
hippocampus of animals (155). It has been sug- Findings from knockout studies show that
gested that alterations in BDNF are a necessary multiple gene products involved with glutamatergic
condition for an antidepressant effect (46). Evi- neurotransmission affect depression-like behaviour
dence suggests that glutamatergic neurotransmis- in rats and mice. Similarly, exposure to chronic
sion interacts with BDNF production and release. stress in animals appears to change the expression
Direct exposure to Glu results in release of BDNF of some Glu receptor subunits. Taken together,
from astrocytes in the basal forebrain mediated these observations suggest that glutamatergic neu-
through mGlu1 ⁄ 5 receptors (156). Administration rotransmission is involved in the genesis of these
of ketamine induces BDNF expression in the behaviours. The antidepressant-like effects of
substantia nigra of a rat model of presymptomatic glutamatergic agents, and the changes in the
ParkinsonÕs disease (157). However, one recent glutamatergic system that are seen following
study using chronic mild stress in rats showed that administration of antidepressants thought to act
effective antidepressant therapy with ketamine did on monoamine systems, also support a role for Glu
not alter expression of BDNF in the hippocampus in the treatment of depression. However, there are
(25). The relationship between Glu and BDNF is limitations to these investigations. Translating the
bidirectional, with BDNF influencing Glu release behavioural measures in animal models of depres-
and signalling. BDNF activates the phospholipase sion to emotional, cognitive and behavioural
C-gamma ⁄ Ca2+ intracellular system which pro- changes in humans is difficult.
motes Glu release in cultured mouse neurones Early studies in humans examining Glu levels
(158). This signalling is inhibited by glucocortic- were limited by available technology. As Glu does
oids, suggesting a role for chronic stress. Phos- not cross the blood–brain barrier, the use of
phorylation of subunits of Glu receptors, which in peripheral measures of Glu as a marker for central
turn affects their response characteristics, is concentrations is questionable. As well, changes in
induced by BDNF (159). The antidepressant levels of Gln in the CSF may represent changes in
effect of increased activity of AMPA receptors overall Glu throughout the central nervous system,
may be mediated through increased production of but do not account for region-specific dysregula-
BDNF (111, 160). The complex relationship tion in metabolism or neurotransmission. MRS
between Glu and BDNF in the production and allows for real-time in vivo measurement of Glu,
treatment of depression is not fully understood; and regional fluctuations of Glu observed in these
however, it provides further evidence that the role studies are the most direct evidence of changes in
of Glu in depression is interrelated with the activity glutamatergic neurotransmission associated with
of other neurochemical systems. depression. These alterations in Glu may be state
markers of depression, as they resolve with suc-
cessful treatment. The most compelling evidence
Discussion
for the role of Glu in depression in humans comes
Historically, interest in Glu dysfunction in psychi- from the rapid antidepressant effects of ketamine
atric disorders has focused primarily on schizo- (117, 118).
phrenia (161, 162), but exciting findings in recent While glutamatergic dysfunction is increasingly
years on NMDA receptor antagonists in depres- recognized in depression, a number of important
sion have led to a flurry of research on the possible questions remain to be answered. First, depression
role of Glu in the aetiology and pharmacotherapy is a clinical syndrome consisting of a variety of
of depression (10, 15, 85). In this regard, it is of cognitive, behavioural and emotional symptoms.
interest that, based on the literature to date, The function of Glu in depression may be related
generally the nature of the glutamatergic dysfunc- to some or all of these symptoms. It is also unclear

204
Glu in the pathophysiology of depression

whether certain subtypes of depression are more 5. Nestler EJ, Barrot M, Dileone RJ, Eisch A, Gold SJ,
associated with alterations in Glu. There is an early Monteggia LM. Neurobiology of depression. Neuron
2002;34:13–25.
signal that bipolar and unipolar depression may 6. Nutt DJ. The role of dopamine and norepinephrine in
differ regarding Glu levels in the cingulate mea- depression and antidepressant treatment. J Clin Psychia-
sured on MRS (83), but this needs to be confirmed try 2006;67:3–8.
in other brain regions. Second, the timing of 7. Manji HK, Drevets WC, Charney DS. The cellular neuro-
changes in central Glu in relation to the develop- biology of depression. Nat Med 2001;7:541–547.
8. Berton O, Nestler EJ. New approaches to antidepressant
ment of depressive symptoms needs to be clarified. drug discovery: beyond monoamines. Nat Rev Neurosci
It may be that Glu levels change in certain brain 2006;7:137–151.
regions prior to the development of clinical symp- 9. Sen S, Sanacora G. Major depression: emerging thera-
toms, and if so, these changes could be used to peutics. Mt Sinai J Med 2008;75:205–225.
monitor individuals at high risk of developing 10. Baker GB, Mitchell ND. Depression: clinical mechanisms.
In: Begley TP, ed. Wiley Encyclopedia of Chemical Biol-
depression, such as those discontinuing antidepres- ogy. Hoboken, NJ: John Wiley & Sons, 2009: 460–472.
sant therapy. As well, the relationship between Glu 11. Hediger MA. Glutamate transporters in kidney and brain.
and other neurotransmitter ⁄ neuromodulatory sys- Am J Physiol 1999;277:F487–F492.
tems must be further clarified. Changes in mono- 12. Szabo ST, Gould TD, Manji HK. Chapter 1. Neurotrans-
amine systems may be a cause or a result of mitters, receptors, signal transduction, and second mes-
sengers in psychiatric disorders. In: Schatzberg AF,
changes in Glu neurotransmission. One way to test Nemeroff CB, eds. The American Psychiatric Publishing
this would be to observe changes in Glu in Textbook of Psychopharmacology, 4th edn. Arilington:
individuals subjected to amino acid protocols, American Psychiatric Publishing Inc, 2009: 3–58.
which can precipitate depression through reduc- 13. Kugaya A, Sanacora G. Beyond monoamines: glutamater-
tions in central monoamines. Finally, further gic function in mood disorders. CNS Spectr 2005;10:808–
819.
research may lead to the development of glutama- 14. Schoepfer R, Monyer H, Sommer B et al. Molecular biol-
tergic antidepressants for use in clinical practice. ogy of glutamate receptors. Prog Neurobiol 1994;42:353–
Currently, trials in humans have only focused on 357.
agents acting at the NMDA receptor, however 15. Paul IA, Skolnick P. Glutamate and depression: clinical
theoretically AMPA receptor agonists or agents and preclinical studies. Ann N Y Acad Sci 2003;1003:250–
272.
acting at mGlu receptors may also provide antide- 16. McAllister AK, Usrey WM, Noctor SC, Rayport SA.
pressant benefit. Further study of the metabolism, Chapter 4. Cellular and molecular neurobiology of the
receptors, and intracellular pathways related to neuron In: Hales RE, Yudofsky SC, Gabbard GO, eds. The
Glu will continue to provide insight into the American Psychiatric Publishing Textbook of Psychiatry,
pathophysiology and treatment of depression. 5th edn. Arlington: American Psychiatric Publishing, Inc,
2008: 113–155.
17. Bekkers JM, Stevens CF. NMDA and non-NMDA
Acknowledgements receptors are co-localized at individual excitatory syn-
apses in cultured rat hippocampus. Nature 1989;341:230–
The authors gratefully acknowledge financial support from the 233.
Department of Psychiatry, University of Alberta, the Canadian 18. Palucha A, Pilc A. Metabotropic glutamate receptor
Institutes of Health Research (CIHR) and the Canada ligands as possible anxiolytic and antidepressant drugs.
Research Chairs and Canada Foundation for Innovation Pharmacol Ther 2007;115:116–147.
programs. 19. Cartmell J, Schoepp DD. Regulation of neurotransmitter
release by metabotropic glutamate receptors. J Neuro-
chem 2000;75:889–907.
Declaration of interest 20. Porsolt RD, Bertin A, Jalfre M. Behavioural despair in
mice: a primary screening test for antidepressants. Arch
The authors declare no conflict of interest. Int Pharmacodyn 1977;229:327–336.
21. Bourin M, Hascoet M, Colombel MC, Redrobe JP, Baker
GB. Differential effects of clonidine, lithium, and quinine
References in the forced swimming test in mice for antidepressant:
1. Schildkraut JJ. The catecholamine hypothesis of affective possible roles of serotonergic systems. Eur Neuropsy-
disorders: a review of supporting evidence. Am J Psy- chopharmacol 1996;6:231–236.
chiatry 1965;122:509–522. 22. Li CX, Wang Y, Gao H et al. Cerebral metabolic changes
2. Coppen A. The biochemistry of affective disorders. Br J in a depression-like rat model of chronic forced swimming
Psychiatry 1967;113:1237–1264. studied by ex vivo high resolution 1H magnetic resonance
3. Lapin IP, Oxenkrug GF. Intensification of the central spectroscopy. Neurochem Res 2008;33:2342–2349.
serotonergic processes as a possible determinant of thy- 23. Sartorius A, Mahlstedt MM, Vollmayr B, Henn FA, Ende
moleptic effect. Lancet 1969;1:132–136. G. Elevated spectroscopic glutamate ⁄ gamma-amino
4. Baker GB, Dewhurst WG. Biochemical theories of affec- butyric acid in rats bred for learned helplessness.
tive disorders. In: Dewhurst WG, Baker GB, eds. The Neuroreport 2007;18:1469–1473.
Pharmacotherapy of Affective Disorders: theory and 24. Tordera RM, Totterdell S, Wojcik SM et al. Enhanced
Practice. London: Croom Helm Ltd., 1985:1–59. anxiety, depressive-like behaviour and impaired recogni-

205
Mitchell and Baker

tion memory in mice with reduced expression of the 41. Ahnaou A, Dautzenberg FM, Geys H et al. Modulation of
vesicular glutamate transporter 1 (VGLUT1). Eur J group II metabotropic glutamate receptor (mGlu2) elicits
Neurosci 2007;25:281–290. common changes in rat and mice sleep-wake architecture.
25. Garcia LS, Comim CM, Valvassori SS et al. Ketamine Eur J Pharmacol 2009;603:62–72.
treatment reverses behavioral and physiological altera- 42. Matrisciano F, Panaccione I, Zusso M et al. Group-II
tions induced by chronic mild stress in rats. Prog Neu- metabotropic glutamate receptor ligands as adjunctive
ropsychopharmacol Biol Psychiatry 2009;33:450–455. drugs in the treatment of depression: a new strategy to
26. Boyce-Rustay JM, Holmes A. Genetic inactivation of the shorten the latency of antidepressant medication? Mol
NMDA receptor NR2A subunit has anxiolytic- and Psychiatry 2007;12:704–706.
antidepressant-like effects in mice. Neuropsychopharma- 43. Matrisciano F, Zusso M, Panaccione I et al. Synergism
cology 2006;31:2405–2414. between fluoxetine and the mGlu2 ⁄ 3 receptor agonist,
27. Cryan JF, Kelly PH, Neijt HC, Sansig G, Flor PJ, van Der LY379268, in an in vitro model for antidepressant drug-
Putten H. Antidepressant and anxiolytic-like effects in induced neurogenesis. Neuropharmacology 2008;54:428–
mice lacking the group III metabotropic glutamate 437.
receptor mGluR7. Eur J Neurosci 2003;17:2409–2417. 44. Santarelli L, Saxe M, Gross C et al. Requirement of
28. Mitsukawa K, Mombereau C, Lotscher E et al. Metabotropic hippocampal neurogenesis for the behavioural effects of
glutamate receptor subtype 7 ablation causes dysregulation antidepressants. Science 2003;301:805–809.
of the HPA axis and increases hippocampal BDNF protein 45. Sapolsky RM. Is impaired neurogenesis relevant to the
levels: implications for stress-related psychiatric disorders. affective symptoms of depression? Biol Psychiatry
Neuropsychopharmacology 2006;31:1112–1122. 2004;56:137–139.
29. Matrisicano F, Caruso A, Orlando R et al. Defective 46. Mackin P, Watson S, Ferrier N. The neurobiology of
group-II metabotropic glutamate receptors in the hippo- depression: focus on cortisol, brain-derived neurotrophic
campus of spontaneously depressed rats. Neuropharma- factor, and their interactions. Depress Mind Body
cology 2008;55:525–531. 2005;2:11–18.
30. Hunter RG, Bellani R, Bloss E, Costa A, McCarthy K, 47. Palucha A, Branski P, Szewczyk B, Wieronska JM, Klak K,
McEwen BS. Regulation of kainate receptor subunit Pilc A. Potential antidepressant-like effect of MTEP,
mRNA by stress and corticosteroids in the rat hippo- a potent and highly selective mGluR5 antagonist. Phar-
campus. PLoS ONE 2009;4:e4328. macol Biochem Behav 2005;81:901–906.
31. Wieronska JM, Branski P, Szewczyk B et al. Changes in the 48. Belozertseva IV, Kos T, Popik P, Danysz W, Bespalov AY.
expression of metabotropic glutamate receptor 5 Antidepressant-like effects of mGluR1 and mGluR5
(mGluR5) in the rat hippocampus in an animal model of antagonists in the rat forced swim and the mouse tail
depression. Pol J Pharmacol 2001;53:659–662. suspension tests. Eur Neuropsychopharmacol 2007;17:
32. Ryan B, Musazzi L, Mallei A et al. Remodelling by early- 172–179.
life stress of NMDA receptor-dependent synaptic plas- 49. Molina-Hernández M, Tellez-Alcántara NP, Pérez-Garcı́a
ticity in a gene-environment rat model of depression. Int J J, Olivera-Lopez JI, Jaramillo-Jaimes MT. Antidepressant-
Neuropsychopharmacol 2008;31:1–7. like actions of minocycline combined with several gluta-
33. Trullas R, Skolnick P. Functional antagonists at the mate antagonists. Prog Neuropsychopharmacol Biol
NMDA receptor complex exhibit antidepressant actions. Psychiatry 2008;32:380–386.
Eur J Pharmacol 1990;185:1–10. 50. Molina-Hernández M, Téllez-Alcántara NP, Pérez-Garcı́a
34. Yilmaz A, Schulz D, Aksoy A, Canbeyli R. Prolonged ef- J, Olivera-Lopez JI, Jaramillo-Jaimes MT. Desipramine or
fect of an anesthetic dose of ketamine on behavioral glutamate antagonists synergized the antidepressant-like
despair. Pharmacol Biochem Behav 2002;71:341–344. actions of intra-nucleus accumbens infusions of mino-
35. Banasr M, Chowdhury GM, Terwilliger R et al. Glial cycline in male Wistar rats. Prog Neuropsychopharmacol
pathology in an animal model of depression: reversal of Biol Psychiatry 2008;32:1660–1666.
stress-induced cellular, metabolic and behavioral deficits 51. Wieronska JM, Szewczyk B, Branski P, Palucha A, Pilc A.
by the glutamate-modulating drug riluzole. Mol Psychi- Antidepressant-like effect of MPEP, a potent, selective and
atry 2008 [Epub ahead of print]. doi:10.1038/mp.2008.106. systemically active mGlu5 receptor antagonist in the olfac-
36. Mineur YS, Picciotto MR, Sanacora G. Antidepressant- tory bulbectomized rats. Amino Acids 2002;23:213–216.
like effects of ceftriaxone in male C57BL ⁄ 6J mice. Biol 52. Reznikov LR, Grillo CA, Piroli GG, Pasumarthi RK, Rea-
Psychiatry 2007;61:250–252. gan LP, Fadel J. Acute stress-mediated increases in
37. Chaki S, Yoshikawa R, Hirota S et al. MGS0039: a potent extracellular glutamate levels in the rat amygdala: differ-
and selective group II metabotropic glutamate receptor ential effects of antidepressant treatment. Eur J Neurosci
antagonist with antidepressant-like activity. Neurophar- 2007;25:3109–3114.
macology 2004;46:457–467. 53. Svenningsson P, Bateup H, Qi H et al. Involvement of
38. Palucha A, Tatarczynska E, Branski P et al. Group III AMPA receptor phosphorylation in antidepressant
mGlu receptor agonists produce anxiolytic- and anti- actions with special reference to tianeptine. Eur J
depressant-like effects after central administration in rats. Neurosci 2007;26:3509–3517.
Neuropharmacology 2004;46:151–159. 54. Kasper S, McEwen BS. Neurobiological and clinical effects
39. Klak K, Palucha A, Branski P, Sowa M, Pilc A. Combined of the antidepressant tianeptine. CNS Drugs 2008;22:15–
administration of PHCCC, a positive allosteric modula- 26.
tor of mGlu4 receptors and ACPT-I, mGlu III receptor 55. Yang J, Shen J. In vivo evidence for reduced cortical
agonist evokes antidepressant-like effects in rats. Amino glutamate-glutamine cycling in rats treated with the
Acids 2007;32:169–172. antidepressant ⁄ antipanic drug phenelzine. Neuroscience
40. Palucha A, Klak K, Branski P, van der Putten H, Flor PJ, 2005;135:927–937.
Pilc A. Activation of the mGlu7 receptor elicits anti- 56. Michael-Titus AT, Bains S, Jeetle J, Whelpton R. Imipra-
depressant-like effects in mice. Psychopharmacology mine and phenelzine decrease glutamate overflow in the
(Berlin) 2007;194:555–562. prefrontal cortex–a possible mechanism of neuroprotec-

206
Glu in the pathophysiology of depression

tion in major depression? Neuroscience 2000;100:681– 73. Francis PT, Poynton A, Lowe SL et al. Brain amino acid
684. concentrations and Ca2+-dependent release in intractable
57. Palucha A, Branski P, Klak K, Sowa M. Chronic imipra- depression assessed antemortem. Brain Res
mine treatment reduces inhibitory properties of group II 1989;494:315–324.
mGlu receptors without affecting their density or affinity. 74. Frye MA, Tsai GE, Huggins T, Coyle JT, Post RM. Low
Pharmacol Rep 2007;59:525–530. cerebrospinal fluid glutamate and glycine in refractory
58. Ferrero AJ, Cereseto M, Reinés A et al. Chronic treatment affective disorder. Biol Psychiatry 2007;61:162–166.
with fluoxetine decreases seizure threshold in naı̈ve but 75. Levine J, Panchalingam K, Rapoport A, Gershon S, McC-
not in rats exposed to the learned helplessness paradigm: lure RJ, Pettegrew JW. Increased cerebrospinal fluid
correlation with the hippocampal glutamate release. Prog glutamine levels in depressed patients. Biol Psychiatry
Neuropsychopharmacol Biol Psychiatry 2005;29:678–686. 2000;47:586–593.
59. Wang SJ. Potential antidepressant LY 367265 presynap- 76. Jansen JFA, Backes WH, Nicolay K, Kooi ME. 1HMR
tically inhibits the release of glutamate in rat cerebral spectroscopy of the brain: absolute quantification of
cortex. Synapse 2005;55:156–163. metabolites. Radiology 2006;240:318–332.
60. Zangen A, Hyodo K. Transcranial magnetic stimulation 77. Capizzano AA, Jorge RE, Acion LC, Robinson RG. In vivo
induces increases in extracellular levels of dopamine and proton magnetic resonance spectroscopy in patients with
glutamate in the nucleus accumbens. Neuroreport mood disorders: a technically oriented review. J Magn
2002;13:2401–2405. Reson Imaging 2007;26:1378–1389.
61. Martı́nez-Turrillas R, Del Rı́o J, Frechilla D. Sequential 78. Cousins JP, Harper G. Neurobiochemical changes from
changes in BDNF mRNA expression and synaptic levels Taxol ⁄ Neupogen chemotherapy for metastatic breast
of AMPA receptor subunits in rat hippocampus after carcinoma corresponds with suicidal depression. Cancer
chronic antidepressant treatment. Neuropharmacology Lett 1996;110:163–176.
2005;49:1178–1188. 79. Auer DP, Pütz B, Kraft E, Lipinski B, Schill J, Holsboer F.
62. Tan CH, He X, Yang J, Ong WY. Changes in AMPA Reduced glutamate in the anterior cingulate cortex in
subunit expression in the mouse brain after chronic depression: an in vivo proton magnetic resonance spec-
treatment with the antidepressant maprotiline: a link be- troscopy study. Biol Psychiatry 2000;47:305–313.
tween noradrenergic and glutamatergic function? Exp 80. Rosenberg DR, Macmaster FP, Mirza Y et al. Reduced
Brain Res 2006;170:448–456. anterior cingulate glutamatergic concentrations in child-
63. Williams SM, Bryan-Lluka LJ, Pow DV. Quantitative hood OCD and major depression versus healthy controls.
analysis of immunolabeling for serotonin and for gluta- J Am Acad Child Adolesc Psychiatry 2004;43:1146–1153.
mate transporters after administration of imipramine and 81. Rosenberg DR, Macmaster FP, Mirza Y et al. Reduced
citalopram. Brain Res 2005;1042:224–232. anterior cingulate glutamate in pediatric major depres-
64. Ploski JE, Newton SS, Duman RS. Electroconvulsive sei- sion: a magnetic resonance spectroscopy study. Biol
zure-induced gene expression profile of the hippocampus Psychiatry 2005;58:700–704.
dentate gyrus granule cell layer. J Neurochem 82. Taylor MJ, Selvaraj S, Norbury R, Jezzard P, Cowen PJ.
2006;99:1122–1132. Normal glutamate but elevated myo-inositol in anterior
65. Sanacora G, Rothman DL, Mason G, Krystal JH. Clinical cingulate cortex in recovered depressed patients. J Affect
studies implementing glutamate neurotransmission in Disord 2009;119:186–189.
mood disorders. Ann N Y Acad Sci 2003;1003:292–308. 83. Frye MA, Watzl J, Banakar S et al. Increased anterior
66. Kim JS, Schmid-Burgk W, Claus D, Kornhuber HH. cingulate ⁄ medial prefrontal cortical glutamate and crea-
Increased serum glutamate in depressed patients. Arch tine in bipolar depression. Neuropsychopharmacology
Psychiatr Nervenkr 1982;232:299–304. 2007;32:2490–2499.
67. Mauri MC, Ferrera A, Boscati L et al. Plasma and platelet 84. Price RB, Shungu DC, Mao X et al. Amino acid neuro-
amino acid concentrations in patients affected by major transmitters assessed by proton magnetic resonance
depression and under fluvoxamine treatment. Neuropsy- spectroscopy: relationship to treatment resistance in ma-
chobiology 1998;37:124–129. jor depressive disorder. Biol Psychiatry 2009;65:792–800.
68. Mitani H, Shirayama Y, Yamada T, Maeda K, Ashby CR, 85. Yildiz-Yesiloglu A, Ankerst DP. Review of 1H magnetic
Kawahara R. Correlation between plasma levels of glu- resonance spectroscopy findings in major depressive dis-
tamate, alanine and serine with severity of depression. order: a meta-analysis. Pychiatry Res 2006;147:1–25.
Prog Neuropsychopharmacol Biol Psychiatry 86. Ajilore O, Haroon E, Kumaran S et al. Measurement of
2006;30:1155–1158. brain metabolites in patients with type 2 diabetes and
69. Altamura C, Maes M, Dai J, Meltzer HY. Plasma con- major depression using proton magnetic resonance spec-
centrations of excitatory amino acids, serine, glycine, troscopy. Neuropsychopharmacology 2007;32:1224–1231.
taurine, and histidine in major depression. Eur Neuro- 87. Caetano SC, Fonseca M, Olvera RL et al. Proton spec-
psychopharmacol 1995;5:71–75. troscopy study of the left dorsolateral prefrontal cortex in
70. Maes M, Verkerk R, Vandoolaeghe E, Lin A, Scharpé S. pediatric depressed patients. Neurosci Lett 2005;384:321–
Serum levels of excitatory amino acids, serine, glycine, 326.
histidine, threonine, taurine, alanine and arginine in 88. Michael N, Erfurth A, Ohrmann P, Arolt V, Heindel W,
treatment-resistant depression: modulation by treatment Pfleiderer B. Neurotrophic effects of electroconvulsive
with antidepressants and prediction of clinical respon- therapy: a proton magnetic resonance study of the left
sivity. Acta Psychiatr Scand 1998;97:302–308. amygdalar region in patients with treatment-resistant
71. Palmio J, Huuhka M, Saransaari P et al. Changes in plasma depression. Neuropsychopharmacology 2003;28:720–725.
amino acids after electroconvulsive therapy of depressed 89. Michael N, Erfurth A, Ohrmann P, Arolt V, Heindel W,
patients. Psychiatry Res 2005;137:183–190. Pfleiderer B. Metabolic changes within the left dorsolat-
72. Hashimoto K, Sawa A, Iyo M. Increased levels of glutamate eral prefrontal cortex occurring with electroconvulsive
in brains from patients with mood disorders. Biol Psy- therapy in patients with treatment resistant unipolar
chiatry 2007;62:1310–1316. depression. Psychol Med 2003;33:1277–1284.

207
Mitchell and Baker

90. Pfleiderer B, Michael N, Erfurth A et al. Effective elec- 106. Karolewicz B, Stockmeier CA, Ordway GA. Elevated levels
troconvulsive therapy reverses glutamate ⁄ glutamine def- of the NR2C subunit of the NMDA receptor in the locus
icit in the left anterior cingulum of unipolar depressed coeruleus in depression. Neuropsychopharmacology
patients. Psychiatry Res 2003;122:185–192. 2005;30:1557–1567.
91. Luborzewski A, Schubert F, Seifert F et al. Metabolic 107. Kristiansen LV, Meador-Woodruff JH. Abnormal striatal
alterations in the dorsolateral prefrontal cortex after expression of transcripts encoding NMDA interacting
treatment with high-frequency repetitive transcranial PSD proteins in schizophrenia, bipolar disorder and
magnetic stimulation in patients with unipolar major major depression. Schizophr Res 2005;78:87–93.
depression. J Psychiatr Res 2007;41:606–615. 108. McCullumsmith RE, Meador-Woodruff JH. Striatal excit-
92. Murck M, Schubert MI, Schmid D, Schussler P, Steiger A, atory amino acid transporter transcript expression in
Auer DP. The glutamatergic system and its relation to the schizophrenia, bipolar disorder, and major depressive
clinical effect of therapeutic-sleep deprivation in depres- disorder. Neuropsychopharmacology 2002;26:368–375.
sion – an MR spectroscopy study. J Psychiatr Res 109. Berk M, Plein H, Ferreira D. Platelet glutamate receptor
2009;43:175–180. supersensitivity in major depressive disorder. Clin Neu-
93. Hasler G, van der Veen JW, Tumonis T, Meyers N, Shen J, ropharmacol 2001;24:129–132.
Drevets WC. Reduced prefrontal glutamate ⁄ glutamine 110. Choudary PV, Molnar M, Evans SJ et al. Altered cortical
and gamma-aminobutyric acid levels in major depression glutamatergic and GABAergic signal transmission with
determined using proton magnetic resonance spectro- glial involvement in depression. Proc Natl Acad Sci USA
scopy. Arch Gen Psychiatry 2007;64:193–200. 2005;102:15653–15658.
94. Block W, Träber F, von Widdern O et al. Proton MR 111. Bleakman D, Alt A, Witkin JM. AMPA receptors in the
spectroscopy of the hippocampus at 3 T in patients with therapeutic management of depression. CNS Neurol
unipolar major depressive disorder: correlates and pre- Disord Drug Targets 2007;6:117–126.
dictors of treatment response. Int J Neuropsychophar- 112. Freed WJ, Dillan-Carter O, Kleinman JE. Properties of [3H]
macol 2009;12:415–422. AMPA binding in post-mortem human brain from psy-
95. Milne A, Macqueen GM, Yucel K, Soreni N, Hall GB. chotic subjects and controls: increases in caudate nucleus
Hippocampal metabolic abnormalities at first onset and associated with suicide. Exp Neurol 1993;121:48–56.
with recurrent episodes of a major depressive disorder: a 113. Noga JT, Hyde TM, Herman MM et al. Glutamate recep-
proton magnetic resonance spectroscopy study. Neuro- tors in the post-mortem striatum of schizophrenic, sui-
image 2009;47:36–41. cide, and control brains. Synapse 1997;27:168–176.
96. Sanacora G, Gueorguieva R, Epperson CN et al. Subtype- 114. Dwivedi Y, Rizavi HS, Roberts RC, Conely RC, Tamminga
specific alterations of GABA and glutamate in major CA, Pandey GN. Reduced activation and expression of
depression. Arch Gen Psychiatry 2004;61:705–713. ERK1 ⁄ 2 MAP kinase in the post-mortem brain of
97. Glodzik-Sobanska L, Slowik A, McHugh P et al. Single depressed suicide victims. J Neurochem 2001;77:916–928.
voxel proton magnetic resonance spectroscopy in post- 115. Beneyto M, Kristiansen LV, Oni-Orisan A, McCullumsmith
stroke depression. Psychiatry Res 2006;148:111–120. RE, Meador-Woodruff JH. Abnormal glutamate receptor
98. Batra NA, Seres-Mailo J, Hanstock C et al. Proton mag- expression in the medial temporal lobe in schizophrenia
netic resonance spectroscopy measurement of brain glu- and mood disorders. Neuropsychopharmacology
tamate levels in premenstrual dysphoric disorder. Biol 2007;32:1888–1902.
Psychiatry 2008;63:1178–1184. 116. Krystal JH. Ketamine and the potential role for rapid-
99. Paddock S, Laje G, Charney D et al. Association of acting antidepressant medications. Swiss Med Wkly
GRIK4 with outcome of antidepressant treatment in the 2007;137:215–216.
STAR*D cohort. Am J Psychiatry 2007;164:1136–1139. 117. Berman RM, Cappiello A, Anand A et al. Antidepressant
100. Laje G, Paddock S, Manji H et al. Genetic markers of effects of ketamine in depressed patients. Biol Psychiatry
suicidal ideation emerging during citalopram treatment of 2000;47:351–354.
major depression. Am J Psychiatry 2007;164:1530–1538. 118. Zarate CA, Singh JB, Carlson PJ et al. A randomized trial
101. Menke A, Lucae S, Kloiber S et al. Genetic markers within of an N-methyl-d-aspartate antagonist in treatment
glutamate receptors associated with antidepressant treat- resistant major depression. Arch Gen Psychiatry
ment-emergent suicidal ideation. Am J Psychiatry 2006;63:856–864.
2008;165:917–918. 119. Pittenger C, Coric V, Banasr M, Bloch M, Krystal JH,
102. Nowak G, Ordway GA, Paul IA. Alterations in the N-me- Sanacora G. Riluzole in the treatment of mood and
thyl-d-asptartate (NMDA) receptor complex in the frontal anxiety disorders. CNS Drugs 2008;22:761–786.
cortex of suicide victims. Brain Res 1995;675:157–164. 120. Zarate CA, Payne JL, Quiroz J et al. An open-label trial of
103. Holemans S, De Paermentier F, Horton RW, Crompton MR, riluzole in patients with treatment-resistant major
Katona CL, Maloteaux JM. NMDA glutamatergic depression. Am J Psychiatry 2004;161:171–174.
receptors, labelled with [3H]MK-801, in brain samples 121. Sanacora G, Kendell SF, Levin Y et al. Preliminary
from drug-free depressed suicides. Brain Res evidence of riluzole efficacy in antidepressant-treated
1993;616:138–143. patients with residual depressive symptoms. Biol Psychi-
104. Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. atry 2007;61:822–825.
Reduced levels of NR2A and NR2B subunits of NMDA 122. Mathew SJ, Murrough JW, Aan Het Rot M, Collins KA,
receptor and PSD-95 in the prefrontal cortex in major Reich DL, Charney DS. Riluzole for relapse prevention
depression. Prog Neuropsychopharmacol Biol Psychiatry following intravenous ketamine in treatment-resistant
2009;33:70–75. depression: a pilot randomized, placebo-controlled con-
105. Toro CT, Hallak JE, Dunham JS et al. The NR1 N-methyl- tinuation trial. Int J Neuropsychopharmacol 2009;17:
d-aspartate subunit and brain-derived neurotrophic factor 1–12.
in temporal lobe epilepsy hippocampus: a comparison of 123. Muhonen LH, Lönnqvist J, Juva K, Alho H. Double-blind,
patients with and without coexisting psychiatric symp- randomized comparison of memantine and escitalopram
toms. Epilepsia 2007;48:2352–2356. for the treatment of major depressive disorder comorbid

208
Glu in the pathophysiology of depression

with alcohol dependence. J Clin Psychiatry 2008;69:392– and median eminence. Psychoneuroendocrinology
399. 2009;34:1370–1379.
124. Maeng S, Zarate CA, Du J et al. Cellular mechanisms 142. Durand D, Pampillo M, Caruso C, Lasaga M. Role of
underlying the antidepressant effects of ketamine: role of metabotropic glutamate receptors in the control of neu-
alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic roendocrine function. Neuropharmacology 2008;55:577–
acid receptors. Psychiatry 2008;63:349–352. 583.
125. Maeng S, Zarate CA. The role of glutamate in mood 143. Bradbury MJ, Giracello DR, Chapman DF et al. Metabo-
disorders: results from the ketamine in major depression tropic glutamate receptor 5 antagonist-induced stimula-
study and the presumed cellular mechanism underlying its tion of hypothalamic-pituitary axis activity: interaction
antidepressant effects. Curr Psychiatry Rep 2007;9:467– with serotonergic systems. Neuropharmacology
474. 2003;44:562–572.
126. Palmer CL, Cotton L, Henley JM. The molecular phar- 144. Lang CH, Ajmal M. Metabolic, hormonal, and hemody-
macology and cell biology of alpha-amino-3-hydroxy-5- namic changes induced by metabotropic excitatory amino
methyl-4-isoxazolepropionic acid receptors. Pharmacol acid agonist (1S, 3R)-ACPD. Am J Physiol
Rev 2005;57:253–277. 1995;268:1026–1033.
127. Lee HK, Takamiya K, Han JS et al. Phosphorylation of the 145. Scaccianoce S, Matrisciano F, Del Bianco P et al. Endo-
AMPA receptor GluR1 subunit is required for synaptic genous activation of group-II metabotropic glutamate
plasticity and retention of spatial memory. Cell receptors inhibits the hypothalamic-pituitary-adrenocor-
2003;112:631–643. tical axis. Neuropharmacology 2003;44:555–561.
128. Svenningsson P, Tzavara ET, Witkin JM, Fienberg AA, 146. Skynner HA, Amos DP, Murray F et al. Proteomic analysis
Nomikos GG, Greengard P. Involvement of striatal and identifies alterations in cellular morphology and cell death
extrastriatal DARPP-32 in biochemical and behavioural pathways in mouse brain after chronic corticosterone
effects of fluoxetine (Prozac). Proc Natl Acad Sci USA treatment. Brain Res 2006;1102:12–26.
2002;99:3182–3187. 147. Altar CA, Cai N, Bliven T et al. Anterograde transport of
129. Du J, Suzuki K, Wei Y et al. The anticonvulsants lamo- brain-derived neurotrophic factor and its role in the
trigine, riluzole, and valproate differentially regulate brain. Nature 1997;389:856–860.
AMPA receptor membrane localization: relationship to 148. Martinowich K, Manji H, Lu B. New insights into BDNF
clinical effects in mood disorders. Neuropsychopharma- function in depression and anxiety. Nat Neurosci
cology 2006;32:793–802. 2007;10:1089–1093.
130. Pilc A, Chaki S, Nowak G, Witkin JM. Mood disorders: 149. Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, Tamminga
regulation by metabotropic glutamate receptors. Biochem CA, Pandey GN. Altered gene expression of brain-derived
Pharmacol 2008;75:997–1006. neurotrophic factor and receptor tyrosine kinase B in
131. Hyman SE, Nestler EJ. Initiation and adaptation: a par- postmortem brain of suicide subjects. Arch Gen Psychi-
adigm for understanding psychotropic drug action. Am J atry 2003;60:804–815.
Psychitatry 1996;153:151–162. 150. Duman RS, Monteggia LM. A neurotrophic model for
132. Skolnick P, Legutko B, Li X, Bymaster FP. Current per- stress-related mood disorders. Biol Psychiatry
spectives on the development of non-biogenic amine- 2006;59:1116–1127.
based antidepressants. Pharmacol Res 2001;43:411–423. 151. Siuciak JA, Lewis DR, Wiegand SJ, Lindsay RM. Antide-
133. Petty F. GABA and mood disorders: a brief review and pressant-like effect of brain-derived neurotrophic factor
hypothesis. J Affect Dis 1995;33:275–281. (BDNF). Pharmacol Biochem Behav 1997;56:131–137.
134. Sanacora G, Mason GF, Rothman DL et al. Reduced cor- 152. Monteggia LM, Barrot M, Powell CM et al. Essential role
tical c-aminobutyric acid levels in depressed patients of brain-derived neurotrophic factor in adult hippocam-
determined by proton magnetic resonance spectroscopy. pal function. Proc Natl Acad Sci USA 2004;101:10827–
Arch Gen Psychiatry 1999;56:1043–1047. 10832.
135. Chaki S, Okubo T, Sekiguchi Y. Non-monoamine-based 153. Shimizu E, Hashimoto K, Okamura N et al. Alterations of
approach for the treatment of depression and anxiety serum levels of brain-derived neurotrophic factor
disorders. Recent Pat CNS Drug Discov 2006;1:1–27. (BDNF) in depressed patients with or without antide-
136. Pariante CM, Lightman SL. The HPA axis in major pressants. Biol Psychiatry 2003;54:70–75.
depression: classical theories and new developments. 154. Tanis KQ, Newton SS, Duman RS. Targeting neurotrophic ⁄
Trends Neurosci 2007;31:464–468. growth factor expression and signaling for antidepressant
137. Thomson F, Craighead M. Innovative approaches for the drug development. CNS Neurol Disord Drug Targets
treatment of depression: targeting the HPA axis. Neuro- 2007;6:151–160.
chem Res 2008;33:691–707. 155. Warner-Schmidt JL, Duman RS. Hippocampal neurogen-
138. Gabr RW, Birkle DL, Azzaro AJ. Stimulation of the esis: opposing effects of stress and antidepressant treat-
amygdala by glutamate facilitates corticotropin-releasing ment. Hippocampus 2006;16:239–249.
factor release from the median eminence and activation of 156. Jean YY, Lercher LD, Dreyfus CF. Glutamate elicits
the hypothalamic-pituitary-adrenal axis in stressed rats. release of BDNF from basal forebrain astrocytes in a
Neuroendocrinology 1995;62:333–339. process dependent on metabotropic receptors and the
139. Makara GB, Stark E. Effect of intraventricular glutamate PLC pathway. Neuron Glia Biol 2008;4:35–42.
on ACTH release. Neuroendocrinology 1975;18:213–216. 157. Bustos G, Abarca J, Bustos V et al. NMDA receptors
140. Zelena D, Mergl A, Makara GB. Glutamate agonists mediate an early up-regulation of brain-derived neuro-
activate the hypothalamic-pituitary-adrenal axis through trophic factor expression in substantia nigra in a rat
hypothalamic periventricular nucleus, but not through model of presymptomatic ParkinsonÕs disease. J Neurosci
vasopressinergic neurons. Brain Res 2005;1031:185–193. Res 2009;106:647–652.
141. Evanson NK, Van Hooren DC, Herman JP. GluR5-medi- 158. Numakawa T, Kumamaru E, Adachi N, Yagasaki Y, Izumi A,
ated signalling reduces hypothalamo-pituitary-adreno- Kunugi H. Glucocorticoid receptor interaction with TrkB
cortical stress responses at the paraventricular nucleus promotes BDNF-triggered PLC-gamma signaling for

209
Mitchell and Baker

glutamate release via a glutamate transporter. Proc Natl 162. Seeman P. Glutamate and dopamine components in
Acad Sci USA 2009;106:647–652. schizophrenia. J Psychiatry Neurosci 2009;34:143–149.
159. Carvalho AL, Caldeira MV, Santos SD, Duarte CB. Role 163. Coyle JT, Tsai G, Goff D. Converging evidence of NMDA
of the brain-derived neurotrophic factor at glutamatergic receptor hypofunction in the pathophysiology of schizo-
synapses. Br J Pharmacol 2008;153:S310–S324. phrenia. Ann N Y Acad Sci 2003;1003:318–327.
160. Alt A, Nisenbaum ES, Bleakman D, Witkin JM. A role for 164. Lindsley CW, Shipe WD, Wolkenberg SE et al. Progress
AMPA receptors in mood disorders. Neuropsychophar- towards validating the NMDA receptor hypofunction
macology 2006;31:2405–2414. hypothesis of schizophrenia. Curr Top Med Chem
161. Sodhi M, Wood KH, Meador-Woodruff J. Role of 2006;6:771–785.
glutamate in schizophrenia: integrating excitatory ave- 165. Wieronska JM, Pilc A. Metabotropic glutamate receptors
nues of research. Expert Rev Neurother 2008;8:1389– in the tripartite synapse as a target for new psychotropic
1406. drugs. Neurochem Int 2009;55:85–97.

210

You might also like