You are on page 1of 13

The Glutamatergic Dysfunction Hypothesis

for Schizophrenia
Joseph T. Coyle, MD
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

Schizophrenia is a syndrome, undoubtedly with multiple etiologies, that variably exhibits


several features including positive and negative symptoms, cognitive deficits, onset in
young adulthood, and deterioration from the previous level of function. This review will
examine the growing evidence that dysfunction of corticolimbic glutamatergic neurotrans-
mission may contribute to or account for the manifestations of schizophrenia. Glutamater-
gic neurons represent the primary excitatory afferent and efferent systems innervating the
cortex, limbic regions, and striatum. The postsynaptic actions of glutamate are mediated
by a family of glutamate-gated ion channels that permit the influx of sodium and calcium,
thereby depolarizing (exciting) neurons. One of these receptors, the Kmethyl-D-aspartate
For personal use only.

(NMDA) receptor, is the site of action of psychotomimetics such as phencyclidine and


related anesthetics, which can reproduce in normal individuals most of the symptomatic
features of schizophrenia. An endogenous antagonist at the NMDA receptor, Kacetyl-
aspatfyl glutamate, appears to have enhanced activity in the frontal cortex and hippo-
campal formation in persons with this disorder. Glutamatergic dysfunction may be particu-
larly relevant to those forms of schizophrenia in which negative symptoms, cognitive
deficits, and deterioration are prominent features. In supporl of this inference, clinical
studies have shown that drugs that enhance NMDA-receptor function reduce negative
symptoms and cognitive deficits in persons with chronic schizophrenia who are receiving
neuroleptics. Thus, dysfunction of glutamatergic neurotransmission represents an important
organizational focus for research on the complex manifestations of schizophrenia. (HARVARD
REV PSYCHIATRY1996;3:241-53.)

Schizophrenia is one of the most persistent, disabling, toms typically commence during the second or third decade
and costly forms of mental illness. Overt psychotic symp- of life (with earlier onset in males than in females),' often
after an unremarkable premorbid development.2 The causes
of schizophrenia are probably several and interactive, since
From the Consolidated Department of Psychiatry, Harvard Medical
School, Boston, Mass. genetic factors, fetal insults, and viral infection have all
been implicated."' Although the outcome is somewhat vari-
The author is the recipient of a Senior NARSAD Award and able, the majority of affected individuals exhibit waxing and
receives research funding from NIMH and NINDS. waning of acute psychotic symptoms complicated by a pro-
gressive and enduring impairment in motivational, atten-
Original manuscript received 28 July 1995, accepted for publication
15 September 1995; revised manuscript received 22 September tional, affective, and social functions."-' Neuropsychological
1995. studies also reveal deficits in working memory, thought
organization, and language usage.'-'' These deficits corre-
Reprint requests: Joseph T. Coyle, MD, McLean Hospital, 115 Mill late with reductions in volume of corticolimbic structures of
St., Belmont, M A 021 78.
the brain including the temporal cortex, amygdala, and
Copyright 0 1996 by Harvard Medical School. Whether schizophrenia, or its subtypes, is a
progressive disorder has been a topic of debate since the
1067132291961$5.00 + 0 39/1/69598 time of Kraepelin.2 The early course of illness certainly
24 1
Harvard Rev Psychiatry
242 Coyle January/Februaty 1996

TABLE 1. Glutamate Receptors mitter in the brain and as the proximate cause of neuronal
degeneration, especially in disorders with symptomatic on-
Receptor family Ligands Subunits set in adulthood or late This review will present the
evidence that the glutamatergic dysfunction can help to
NMDA Glu, ASP, NMDA NR1, NR2A-D explain the five aspects of schizophrenia described above
KA Glu, KA GluR5-7, KA1-2 and leads to hypotheses that can be subjected to experimen-
AMPA Glu, AMPA, &A GluR1-4 tal validation. More important, if dysfunction of glutamater-
Metabotropic Glu, QA, L-APB mGluR1-6 (G- gic neurotransmission contributes to the pathophysiology of
protein) schizophrenia, it offers possibilities €or therapeutic inter-
NMDA, N-methybaspartate; KA,kainate; AMPA, a-amino-3- ventions that address not simply the positive symptoms but
hydroxy-5-methylisoxazole-4-propionic acid; Glu, glutamate; Asp, the mechanisms responsible for cognitive deficits, negative
aspartic acid; QA, quisqualate; L-APB, L-2-amino-4-phosphonobu- symptoms, and deterioration.
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

tyric acid; mGZuR, metabotropic glutamate receptor.


GLUTAMATERGIC SYSTEMS
shows progressively worsening symptoms.6s16Also, recent
studiesl'~ls have provided convincing evidence that inad- Quantitative brain morphometry, neuropsychological find-
equately treated psychosis is associated with a poorer out- ings, event-related potentials, and functional brain imaging
come, suggesting that psychosis may produce relatively have all revealed both structural and functional abnormali-
irreversible brain changes. ties in schizophrenia that affect diverse areas of the brain,
Any theory that could explain the pathophysiology of including the temporal cortex, hippocampus, anterior cingu-
schizophrenia, therefore, must take into account at least five late region, prefrontal area, amygdala, nucleus accumbens,
aspects of the disorder: positive symptoms, negative symp- striatum, and t h a l a m u ~ . ~ ' -A~ common
~'~~ feature linking
toms, cognitive deficits, onset of symptoms in young adult- the diverse brain structures affected in schizophrenia is the
For personal use only.

hood, and deterioration from the previous level of function. excitatory neuronal system that interconnects them. Over
For over 20 years the major explanatory hypothesis for the the last decade a compelling case has been made for the role
pathophysiology of schizophrenia has centered on the of glutamate (and related acidic amino acids) as the primary
mechanism of antipsychotic effects of neuroleptics in terms excitatory neurotransmitter in the central nervous sys-
of blocking D, dopamine r e ~ e p t o r s . ~ The
~ - ' ~"dopamine hy- tem.34,35Specifically, the pyramidal cells of the cerebral
pothesis" maintains that the clinical potency of neuroleptics cortex, the granule cells of the hippocampus, and the exci-
correlates impressively with affinity of these drugs for the tatory thalamocortical pathways appear to be glutamater-
D, receptors, and that stimulants, which enhance central g i ~The . ~ other
~ major neurotransmitter of the forebrain is
dopaminergic neurotransmission, can produce the positive y-aminobutyric acid (GABA), which mediates inhibitory
symptoms of schizophrenia such as delusions, hallucina- neurotransmission. Whereas the glutamatergic neurons are
tions, and thought disorder. Postmortem as well as in vivo predominantly efferent, forebrain GABAergic systems (with
imaging studie~'~-'~ have provided reasonably compelling a few important exceptions such as the striatonigral path-
evidence that dysfunction of dopaminergic neurotransmis- way) are primarily local circuit neu~ons.~' Thus, on the basis
sion is an important element of the disorder. However, of their anatomy alone, the glutamatergic neurons of the
although paradoxical dopaminergic hypofunction might con- forebrain would appear to be in a key position to play a
tribute to negative symptoms, dopaminergic dysfunction central role in the diverse pathology that characterizes
would appear to account primarily for positive symp- schizophrenia.
t o m ~ . ' ~ Furthermore,
.'~ the fact that neuroleptics may take In concert with the delineation of the functional anatomy
days to weeks to produce their full effect suggests that their of glutamatergic pathways has been the development of our
mechanism of action is indirect, requiring neuronal adapta- understanding of the receptors mediating glutamate's
tion beyond mere D, receptor blockade."' It is more difficult physiological actions (see Table 1).On the basis of pharma-
to rationalize dopaminergic dysfunction as a complete ex- cological and biophysical characteristics, three families of
planation of cognitive deficits, corticolimbic atrophy, and a glutamate-gated ion channels have been identified: N-me-
possible deteriorating course. With the advent of atypical thyhaspartate (NMDA), kainic acid (KA),and cc-amino-3-
neuroleptics, especially clozapine, other neurotransmitter hydroxy-5-methylisoxazole-4-propionicacid (AMPA) recep-
systems such as the serotonin projections acting at 5-HT, t o r ~ AMPA. ~ ~ and KA receptors mediate fast excitatory
receptors have been implicated." postsynaptic potentials, whereas NMDA receptors serve a
Recently, new insights into the pathophysiology of schizo- more modulatory function since they are recruited by depo-
phrenia have emerged from fundamental research into the l a r i z a t i ~ nThese
. ~ ~ glutamatergic-gated ion channels are the
dual role of glutamate as the major excitatory neurotrans- dominant mediators of excitatory postsynaptic potentials
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 243

and also serve as important conduits for increasing intra- AP-5


cellular calcium. In addition, a family of glutamate receptors
linked to G protein-mediated intracellular responses has Gly, DCS I Glu, NMDA
been de~cribed.~'
The NMDA receptor (see Figure l), in particular, has
been extensively characterized with regard to the complex
n
l K + L l n

modulatory sites of pharmacological imp~rt."~.""~' In ad-


dition to the recognition site for glutamate, there is a
second site to which glycine binds; occupation of the glycine
site is obligatory for glutamate-mediated channel opening.

-
D-Serine is another agonist at the glycine modulatory site,
and D-cycloserine,an antitubercular drug, serves as a partial
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

agonist, activating it at low doses and inhibiting it at high


doses.'" Other modulatory sites include a binding site for
'I I
polyamines, a binding site for zinc, and a redox-sensitive
t '
site. The NMDA-receptor channel is voltage dependent, with Cat +

magnesium occluding the channel at resting membrane Na'


potential. Finally, there is a binding site within the channel FIGURE 1. Schematic representation of the NMDA recep-
that becomes accessible, upon channel opening, to MK-801 tor. The NMDA receptor is a glutamate-activated cation
and related drugs, phencyclidine (PCP), and ketamine, channel, typically composed of two products - NR1 and
which act as noncompetitive NR2A-D. Four sites of pharmacological relevance are
Early studies of the biophysical characteristics of these shown: (1) the glutamate (Glu) recognition site at which
NMDA can also serve as an agonist and aminophosphonova-
For personal use only.

glutamate-gated ion channelss4 indicated that the NMDA


receptor family conducted Na' as well as Ca2+ currents, leric acid (AP-5)as an antagonist; (2) the glycine (G1y)-
whereas the non-NMDA receptor channels sustained pri- modulating site, which must be occupied for glutamate to be
active b-cycloserine [DCS] is a partial agonist at this site);
marily Na ' currents. The recent cloning of the genes en-
(3) the binding site for phencyclidine (PCP) and its analogue
coding the polypeptides that form the various glutamate MK-801, which act as noncompetitive antagonists at the
receptors has revealed a higher level of molecular heteroge- open channel; and (4) a cation-binding site within the
neity, pharmacological diversity, and biophysical variation."" channel, where magnesium (M$ ' ) acts as a voltage-depen-
In particular, it is now known that posttranscriptional dent antagonist.
messenger RNA (mRNA) editing transforms non-NMDA
receptors from permitting the passage of Ca2' to sustaining KACENLASPARNL GLUTAMATE
only Na' currents.
With the availability of pharmacological tools to manipu- An acidic dipeptide, N-acetylaspartyl glutamate (NAAG),
late glutamate-receptor function, the role of these receptors has been closely linked to glutamatergic neuronal systems.
in mediating critical aspects of synaptic plasticity became Found in remarkably high concentrations (0.2-2.0 mmol/L)
evident. Long-term potentiation (LTP), first characterized in brain, NAAG has an uneven regional distribution there.
in the hippocampus,'" causes a permanent increase in exci- Immunocytochemical studies with highly specific polyclonal
tatory synaptic efficacy as a consequence of a brief period of and monoclonal antibodies against NAAG48.49 reveal colocali-
high synaptic activity (for a review, see Kandel and zation of NAAG in many putative glutamatergic neuronal
Schwartz""). Glutamate activation of non-NMDA receptors systems. Postmortem studies in humans5' reveal a subpopu-
mediates the excitatory postsynaptic potentials, but Ca2+ lation of corticolimbic pyramidal neurons enriched with
influx through NMDA receptors is primarily responsible for NAAG. One well-characterized putative glutamatergic pro-
the permanent change in synaptic efficacy," which may jection that contains high concentrations of NAAG is the
involve feedback to the presynaptic glutamatergic terminals retinal ganglion cell-collicular pathway.51 Ca2+-dependent
by a diffusible second messenger such as nitric oxide. LTP is release of endogenous NAAG from the collicular terminals
now known to mediate glutamatergic synaptic plasticity in occurs as a consequence of stimulation of the optic nerve.52
many brain regions other than the hippocampus. LTP ap- NAAG is cleaved to N-acetyl aspartate (NAA)and glutamate
pears to be a critical element in the use-dependent synaptic by a specific peptidase, N-acetylated a-linked acidic dipepti-
modification involved in memory and related cognitive func- dase (NAALADase).NAALADase is a membrane-bound cell-
tions.4GGiven the fact that PCP blocks NMDA receptors, it surface peptidase with high affinity for NAAG (Km = 100
is not surprising that PCP both prevents LTP at a cellular nM). It is expressed in the brain, the kidneys, and the sexual
and disrupts cognitive functions in humans." organs, like other peptidases involved in processing neu-
Harvard Rev Psychiatry
244 Coyle January/February 1996

ropeptides, and is regulated by persistent alterations in tor antagonist MK-801,30and with the neurophysiological
excitatory n e u r o t r a n s m i ~ s i o n . ~The
~ - ~ gene
~ encoding for interactions between dopaminergic and glutamatergic neu-
NAALADase activity has been cloned.s6 rotransmission reviewed by Grace.69
The physiological actions of NAAG are complex. It is a Evidence for glutamate/dopamine interactions has been
partial agonist at NMDA receptors and thus antagonizes strengthened by studies of immediate early gene (IEG)
glutamate's activation of the NMDA r e ~ e p t o r . ~In ~ addition,
.~' expression. IEGs are regulatory factors that alter neuronal
it activates a metabotropic glutamate receptor that inhibits gene expression as a consequence of neurotransmitter-
adenylate c y c l a ~ e .Through
~~ the action of NMLADase, receptor activation. Activation of NMDA receptors is a
NAAG can also serve as a source of synaptic glutamate. potent stimulus for turning on IEG expression. Neurolep-
NAALADase activity is thus pivotal in determining whether tics, D,-receptor antagonists, cause expression of IEGs in
extracellular NAAG will remain intact, thereby inhibiting the striatum, accumbens, and corticolimbic structures, pre-
NMDA receptors, or be converted to glutamate, thereby acti- sumably by disinhibiting glutamate release, although their
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

vating the family of glutamate receptors. Consistent with role in potentializing cyclic adenosine monophosphate by
evidence of molecular heterogeneity of NMDA receptors, blocking postsynaptic D, GI effects cannot be ex~luded.~"
NAAG has been shown to selectively block hippocampal LTP The IEG response to neuroleptics is reversed by coadminis-
of recurrent inhibition, which is mediated by NMDA recep- tration of NMDA-receptor antagonist^.^^ However, the
tors on GAl3Aergic intraneurons.60 atypical neuroleptic clozapine affects IEG expression in
cortical and limbic regions but not in the s t r i a t ~ m . ~
Sys-
'
GLUTAMATE/DOPAMINE INTERACTIONS temic treatment of haloperidol-pretreated rats with D-cyclo-
serine, a partial agonist at the glycine-modulatory site on
The hypothesized dysfunction of glutamatergic neurotrans- the NMDA receptor, augments the IEG induction in the
mission in schizophrenia interdigitates with the traditional s t r i a t ~ mThus,
. ~ ~ D-cycloserinebehaves like a neuroleptic in
dopamine hypothesis of schizophrenia. Presynaptic D, re- this model, although it acts through the NMDA receptor
For personal use only.

ceptors on corticostriatal and limbic glutamatergic termi- and not the dopamine D, receptor.
nals provide a negative regulation of glutamate release.6143
Neuroleptic blockade of these presynaptic receptors may PSYCHOTOMIMETICS
thereby enhance excitatory glutamatergic input to the cau-
date and putamen and to other forebrain regions receiving Perhaps the most attractive argument implicating glutam-
dopaminergic innervation. However, neuroleptic interac- atergic dysfunction in the pathophysiology of schizophrenia
tions in the cortex may alter glutamatergic function in involves the remarkable similarities in the symptomatic
more-complex ways, as has recently been suggested by manifestations of PCP-induced psychosis and schizophre-
Fitzgerald and colleague^.^^ They showed that chronic treat- nia. These similarities were noted over 20 years ago -before
ment with haloperidol increased the amounts of the NMDA there was any inkling about the mechanism of action of PCP
receptor subunit NR1 expressed in the striatum and poste- at the NMDA r e ~ e p t o r .Javitt
~ ~ . ~and
~ Zukin3' strengthened
rior cingulate and of the AMPA receptor subunit GluRl in the inferred relationship by showing that PCP psychosis
the medial prefrontal cortex of the rat. These investigators replicates not only the positive symptoms but also the
also observed a more regionally selective effect of chronic negative symptoms and cognitive deficits of schizophrenia.
clozapine treatment, with no alterations of NR1, GluR1, or Krystal and colleagues76performed a controlled study in
GluR2 in the striatum but significant elevations of GluRl in which the anesthetic ketamine, an analogue of PCP, was
the medial prefrontal cortex and of GluR2 in the frontal administered to normal subjects to document its neuro-
cortex, parietal cortex, nucleus accumbens, and hippocam- psychiatric manifestations objectively. They noted that al-
pus. Glutamatergic neurons have reciprocal interactions though ketamine caused symptoms such as dissociation not
with dopaminergic processes in their terminal fields. observed in schizophrenia, it also precipitated positive
M ~ g h a d d a mhas
~ ~ shown, for example, that glutamate as symptoms, negative symptoms, and cognitive deficits in
well as dopamine release increases in prefrontal cortex as a normal subjects similar to those seen in individuals with
consequence of stress. Furthermore, the local pharmacologi- schizophrenia. The symptoms not associated with schizo-
cal blockade of AMPNKA receptors in prefrontal cortex phrenia may result from a diffuse inhibition of brain NMDA
reduces the stress-induced increase in dopamine release, receptors as a consequence of systemic treatment.
strongly suggesting that glutamate modulates dopamine The virtual ubiquity of NMDA receptors in the brain4'
release.66In addition, glutamate-dopamine interactions may raises the question of how the 'activity of subsets of these
occur at the level of the individual n e ~ r o n . ~These
~ . ~ ' inter- receptors might be selectively altered to yield the symptoms
actions are consistent with the behavioral studies of Carls- of schizophrenia. The molecular identification of a family
son and C a r l ~ s o nwhich
, ~ ~ showed that the locomotor effects of proteins (NR2A-D) that combine with the receptor-ion
of apomorphine, a dopamine-receptor agonist, could be channel (NR1) to form heteromeric NMDA receptors has
markedly enhanced by cotreatment with the NMDA recep- revealed a source of considerable variation in the phar-
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 245

macological characteristics of these receptors:" Consistent of sustaining glial cells but no change in neuronal cell
with this variation, Grunze and colleagues'" have found that number. Rigorous cell-counting techniques have produced
the NMDA receptors on the hippocampal GABAergic in- only limited evidence of a reduction in neuronal number
terneurons are tenfold more sensitive to the synthetic in specific brain regions in schizophrenia."" Nevertheless,
antagonist aminophosphonovaleric acid and the endogenous several have indicated reductions in neuronal
antagonist NAAG than are NMDA receptors on hippo- number in regions including the mediodorsal nucleus
campal glutamatergic neurons. of the thalamus, the nucleus accumbens, the hippocampus,
In postmortem studies on the disposition of NAAG in and the cingulate area. Others, however, have reported no
schizophrenia, Tsai and colleagues77 have found that loss of neurons in the hippocampu~"~ or the prefrontal
NAALADase, the enzyme responsible for cleaving NAAG to ~~rte~.""~"~
NAA and glutamate,"J " is reduced in activity in the frontal One neuronal system that has figured prominently in the
cortex, hippocampus, and parahippocampus in schizo- postmortem studies comprises the GABAergic interneurons
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

phrenic subjects as compared to controls. Such a reduction of the cortical and limbic regions. Benes and colleagues""
in NAALADase activity would prolong the synaptic effects found a significant reduction of the interneurons in layers 11,
of NAAG, thereby favoring NMDA-receptor inhibition, es- 111, V, and VI in the anterior cingulate area. Consistent with
pecially on the GABAergic interneurons. this decline reflecting a loss of GABAergic neurons, the activ-
The role of inhibition of NMDA-receptor function in the ity of glutamate decarboxylase and transport
symptomatic manifestations of schizophrenia is also consis- sites for ['HIGABA are reducedg6in cortex while the density
tent with the age of onset of symptoms. Children appear of postsynaptic GABA, receptors is increa~ed,~'~"" as would be
much less vulnerable than adults to the psychotomimetic expected for compensatory up-regulation of these receptors."
effects of ketamine, compatible with an age of onset in early More recently, Akbarian and colleagues9" have reported a
ad~lthood.~"."" Benes"' has described the myelinization of decrease in the mRNA encoding glutamate decarboxylase,
For personal use only.

frontal glutamatergic pathways that occurs in late adoles- although they did not find a decrease in
cence. Perhaps this late myelinization (which permits the number of apparent GABAergic neurons in the cortex. In
glutamatergic functional input) and related structural aggregate, these results point to a reduction in the functional
changes provide the substrate for the psychotomimetic ef- influence of corticolimbic GABAergic interneurons, which
fects of noncompetitive NMDA-receptor antagonists and for inhibit the activity of glutamatergic efferent neurons.
the expression of idiopathic psychosis. Neuronal degeneration in the mature nervous system
is generally associated with an astrocytic response, thereby
NEURONAL LOSS indicating an area of recent or ongoing neuronal de-
generation. Well-controlled neuropathological studies in
The issue of neuronal loss in schizophrenia has confounded schizophrenia have been rather unrevealing or contradic-
investigators since schizophrenia was first des~ribed.".""."~ tory with regard to evidence of gliosis in the structures of
There are really two critical questions: is there a loss of interest."u37.100 These negative studies have lent support to
neurons in schizophrenia, and if so, when does it take place? the position that schizophrenia does not involve neuronal
With regard to the first question, quantitative morphomet- degeneration in the mature brain. However, apoptotic
ric StudieS1%.l:~.l.~.
s4 in recent years have provided compelling neuronal death (and not necrosis) could theoretically occur
evidence of volumetric reductions in cortical, limbic, and at the onset of acute symptoms, leaving little evidence of
midbrain structures in patients suffering from schizophre- reactive gliosis. This might account for the deterioration
nia. Inasmuch as the course and symptomatic characteris- occurring in the first 2 years after the initial episode."."
tics of the schizophrenia syndrome vary ~onsiderably,'~ re- Alternatively, hypoplasia (i.e., the failure to generate the
cent studies have increasingly focused on identifying corre- neurons during brain development) could account for a
lations between structural change in specific brain regions reduction in neuronal number that would not leave the
and such symptomatic manifestations as delusions, halluci- telltale signs of gliosis. Bloom1"' has reviewed the evidence
nations, and thought disorder.'".'" An overarching feature of that schizophrenia might involve a primary developmental
the quantitative morphometric studies is that volume re- defect in the central nervous system. It remains unclear
duction and enlargement of ventricles are greater in schizo- whether these second- to third-trimester correlates of im-
phrenic patients with poor outcome, more-prominent nega- paired brain maturation include a reduction in number of
tive symptoms, neuropsychological deficits, or tardive dys- neurons generated or are associated with a disproportionate
kineSia.S.26,S5.H6 amount of early apoptosis.
Reduction of brain volume is often but not invariably
associated with a decreased number of neurons. A notable GLUTAMATE AND NEURODEGENERATION
caveat comes from studies of the consequences of enriched
environments on rat c ~ r t e x , ~ ~which
.'' demonstrate an ex- Nearly a quarter of a century ago, Olney'n2 proposed the
pansion of the neuropil with increases in the number "excitotoxic hypothesis" to explain the selective pattern of
Harvard Rev Psychiatry
246 Coyle January/February 1996

neuronal degeneration that occurs as a consequence of acute neurons in cortical and limbic regions. Pharmacological
activation of glutamate-gated ion channels. Agonists at all studies'15 have indicated that this degeneration, pro-
three subclasses of glutamate receptors cause perikaryal- voked by systemic treatment with NMDA-receptor antago-
specific neuronal degeneration, although there is consider- nists, is mediated in part by non-NMDA receptors since
able variation in the vulnerability of neurons to the three local injection of 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo-
different receptor agonists.'03 This pattern of selectivity quinoxaline prevents it. Furthermore, GABA-receptor ago-
mimics the specificity of neuronal vulnerability observed in nists (consistent with hypoactivity of GABAergic inter-
several neurodegenerative conditions including Hunting- neurons in schizophrenia) and muscarinic acetylcholine-
ton's disease, amyotrophic lateral sclerosis, spinocerebellar receptor antagonists are also protective, implicating a
degeneration, stroke, and t r a ~ m a . ~ ~These . ' ~ ~ similarities,
.'~~ permissive role of these neurotransmitters. '16 Interestingly,
along with findings from animal models, have provided the corticolimbic neuronal degeneration induced by the
compelling circumstantial evidence that pre- or postsynaptic NMDA antagonists is preceded by the expression of heat
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

dysfunction of glutamatergic neurotransmission may be the shock proteins, a marker associated with oxidative dam-
proximate cause of neuronal degeneration in such disorders. age,117and leaves minimal glial reaction, consistent with an
Pioneering research by Choi and colleagues'o6 distin- apoptotic form of death."'
guished two forms of glutamate-induced neuronal degen- Olney has developed an explanatory model for this
eration. The first results from a massive influx of N a + , counterintuitive phenomenon: the activity of corticolimbic
which causes the acute cellular edema observed by Olney GABAergic interneurons is particularly sensitive to NMDA-
after systemic monosodium glutamate treatments. The receptor antagonists. Thus, PCP reduces the glutamatergic
second is a delayed degeneration of neurons that is inde- activation of the GABAergic interneurons; the reduced
pendent of Na' but requires Ca2+.As reviewed by Coyle GABAergic tone, in consequence, disinhibits the corticolim-
and P~ttfarcken,"~oxidative damage appears to be an bic glutamatergic neurons that they innervate. The en-
For personal use only.

important mediator of the delayed degeneration resulting hanced glutamatergic efferent activity causes "excitotoxic
from transient activation of NMDA and non-NMDA neuronal death" by persistent activation of non-NMDA
receptors; evidence of this process is provided by elevated receptors on the postsynaptic neurons. Thus, interference
levels of superoxide, membrane peroxidation, and protection with NMDA receptor-driven activity of corticolimbic
by agents that scavenge oxyradicals during the period of GABAergic interneurons (or loss and/or impairment of
damage. function of these interneurons) would favor a WAMPA
Two separate mechanisms of cell death, consistent with receptor-mediated delayed neuronal degeneration in animal
the existence of acute and delayed forms of glutamate models. What further links this use-dependent neuronal
receptor-mediated neuronal degeneration, have recently at- degeneration to schizophrenic psychosis is the observation
tracted considerable attention.'Os-lloThe first, necrosis, re- that both typical and atypical neuroleptics prevent the
sults typically from an external insult, is passive in nature, neuronal
is associated with the destruction of cellular integrity, and
often elicits an inflammatory response (reactive gliosis).The CLINICAL EVIDENCE OF NEURODEGENERATION
second, apoptosis, is self-initiated, involves activation of
endonucleases, and often but not invariably requires de O'Donnell and colleagues'21 have observed a highly signifi-
novo protein synthesis. It is a mechanism involved in elimi- cant increase in P300 latency as a function of age in persons
nation of superfluous cells in development (such as occurs with schizophrenia but not in age-matched healthy controls.
with loss of trophic factors in the developing nervous sys- The P300 is an electrical wave that can be detected in the
tem) and may not elicit a persistent inflammatory cell temporal lobe 300 milliseconds after an auditory stimulus.
response. Recent evidence indicates that delayed neuronal Similar increases in P300 latency have also been observed in
degeneration resulting from transient activation of non- other neurodegenerative disorders.122~123
NMDA receptors exhibits many of the features of apoptotic NAA has been employed as a marker for neuronal integ-
neurodegeneration."' Consistent with this, apoptosis often rity in nuclear magnetic resonance (NMR) spectroscopic
involves oxidative damage as a mediator. (Bcl-2, for ex- studies. Immunocytochemical studies indicate that NAA is
ample, an oncogene that prevents immune cell apoptosis, highly localized to neurons;124its levels decline in animal
exerts some of its protective effects by preventing oxidative models of excitotoxin-inducedneuronal degenerati~nl'~ and
Thus, use-dependent glutamatergic mecha- in postmortem studies of neurodegenerative disorders.lZ6
nisms may play an important role not only in neuroplastic- Using NMR spectroscopy, Yurgelun-Todd and colleague^'^^
ity but also in determining neuronal elimination. found a significant reduction in the NAAIcreatine ratios in
In exploring the ability of noncompetitive NMDA-recep- the temporal cortex of persons with schizophrenia as com-
tor antagonists like MK-801 and PCP to block "excitotoxic" pared to suitable controls. That NAA decreases may repre-
lesions, Olney and c ~ l l e a g l l e s ' ~ discovered
~~"~ that these sent a compromise of neuronal function and not simply
"neuroprotective agents" cause a delayed degeneration of degeneration is supported by Meyerhoff and colleagues,
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 247

who used NMR spectroscopy to show that NAA is reduced in glutamatergic input,"3 it is not surprising that reduced
patients with acquired immune deficiency syndrome (AIDS) activity of superoxide dismutase in cerebrospinal fluid, in-
dementia, a condition not associated with marked neuronal creased markers for glutamatergic neurotransmission, and
loss but with substantial inflammatory changes. This dis- elevated oxidized proteins distinguish schizophrenic pa-
tinction may be important inasmuch as glutamate-mediated tients who have tardive dyskinesia from those who do not.'46
use-dependent neuronal damage in schizophrenia is prob- These findings are consistent with studies indicating that
ably subtle and slowly progressive, with neuronal compro- vitamin E, a free radical scavenger, decreases the symptoms
mise antedating apoptosis. of tardive dyskinesia.'47.'4h
Consistent with an excessive activation of non-NMDA
receptors, which may be indicative of loss of neurons ex- NMDA-RECEPTOR ACTIVATION IN SCHIZOPHRENIA
pressing these receptors, reductions in non-NMDA recep-
tors have been reported in certain cortical and limbic re- The most salient prediction from the hypothesis of NMDA-
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

gions in postmortem studies in schizophrenia. In persons receptor hypofunction as a component of schizophrenia is


with schizophrenia, a reduction in the specific binding of that agents that enhance activation of NMDA receptors
["Hlkainate to its receptors has been described in the hip- should reduce symptoms in schizophrenia. Insofar as posi-
pocampal subfields including the area dentata and CA1, 2, tive symptoms, negative symptoms, and cognitive impair-
and 4,as well as in the parahippocampal gyrus; in contrast, ments are caused by NMDA-receptor antagonists, they
NMDA-inhibitable binding of [3H]glutamate has not been should all be responsive to NMDA-receptor agonists. How-
found to be altered.'2y~'"'Concordant with these findings, a ever, NMDA-receptor agonists, as treatments, bear signif-
reduction in the amount of mRNA for the non-NMDA icant risks -convulsions and NMDA receptor-mediated
receptor gene (GluR K1) has been reported in the hippo- neuronal degeneration.'4y Therefore, researchers consider-
campal formation, most notably in CA3 but also in CA1 and ing a possible pharmacological strategy selected D-cyclo-
For personal use only.

the subiculum.13' Other researchers have reported increased serine (DCS) since it is a partial agonist at the glycine
["Hlkainate binding in the orbitofrontal cortex,'"2 medial modulatory site on the NMDA r e ~ e p t o r ~ ' .and ' ~ ~crosses
~~~'
frontal cortex, and frontal eye field'33,134 in persons with the blood-brain bar~-ier.'~".'~~ Thus, it enhances the response
schizophrenia. of the NMDA receptor to endogenous glutamate without
An aspect of schizophrenia that is consistent with the directly activating the Furthermore, as a partial
hypothesis of an ongoing neurodegenerative process is the agonist, DCS actually impairs NMDA-receptor activation at
vulnerability to tardive dyskinesia, a relatively irreversible high doses, thereby limiting neurotoxic and convulsant
movement disorder. One of the most robust risk factors for effect~.~l.l~~
tardive dyskinesia is age.1s5.1'36 Prominent negative symp- In a double-blind, placebo-controlled dose-finding
toms and evidence of organic changes in the brain are other on a cohort of patients with chronic schizophrenia who had
important predictors of increased vulnerability for tardive prominent negative symptoms and were receiving neurolep-
dyskinesia in schizophrenic patient^.'^^,'^" There is an active tics, 50 mg of DCS produced a significant reduction in
debate about the relative contribution of neuroleptic expo- negative symptoms and significantly enhanced performance
sure in the etiology of tardive dyskinesia. Reviews of case on a frontal lobe-dependent cognitive task. However, nei-
records of schizophrenic patients hospitalized before the ther 15 nor 250 mg of DCS improved symptoms, thereby
advent of n e u r o l e p t i c ~ ' ~ ~indicate
~ ' ~ " that movement disor- yielding a U-shaped dose response consistent with the com-
ders, including orofacial dyskinesias, were not uncommon. pound's partial agonist properties. In support of the lack of
Furthermore, current studies carried out in small numbers efficacy of high-dose DCS, Cascella and c o l l e a g ~ e s have
'~~
of neuroleptic-naive schizophrenic patients in developing reported that 250 mg of DCS caused a deterioration in
countries indicate that tardive dyskinesia occurs in about patients with schizophrenia. In vivo studies in rats similarly
20% and first appears approximately a decade after the demonstrated that 1 mg/kg (nearly equivalent to 50 mg in
onset of the p~ychosis.'~' an adult human-i.e., 0.7 mg/kg) enhanced the IEG re-
Brain imaging, postmortem human studies, and animal sponse to haloperidol, whereas 5 mgkg (nearly equivalent to
models strongly suggest that tardive dyskinesia involves a 250 mg in an adult human) attenuated this response.73
degeneration of striatal neurons, particularly GABAergic Thus, the U-shaped dose-response curve for DCS observed
efferent^.^^^.'^:' Although caudate volume has been reported at the receptor level and in vivo in rats was replicated in the
to increase with acute neuroleptic treatment, striatal vol- schizophrenic patients. Independently, Javitt and col-
ume is often reduced in patients with tardive d y s k i n e ~ i a . ' ~ ~l e a g u e ~ ' ~have
" demonstrated that treatment with glycine
Removal of dopaminergic innervation to the striatum, a itself also significantly reduces the negative symptoms in
functional consequence of neuroleptic treatment, causes persons with schizophrenia. Since patients in both of these
apoptotic neuronal death in the rat ~ t r i a t u m . 'Since~ ~ oxi- studies were already receiving neuroleptics, the effects of
dative damage is an important component of apoptotic glycine modulatory site agonists on positive symptoms re-
neuronal degeneration and neuroleptics increase striatal main to be determined.
Harvard Rev Psychiatry
248 Coyle January/February 1996

of interneurons, decreased glutamate decarboxylase activ-


GABA-Rc V ity," and an inferred up-regulation of GABAA receptors).
Although Akbarian and c011eagues~~ could not confirm a re-
duction in the number of GABAergic interneurons, they did
KA-Rc l_r find a significant decrease in the amount of mRNA encoding
glutamate decarboxylase,the enzyme responsible for the syn-
thesis of GABA. Inasmuch as the causes of schizophrenia are

Q
undoubtedly heterogeneous, it is not surprising that the find-
ings on the specifics of GABAergic dysfunction are not uni-
Glutamate
form. Nevertheless, reduced excitatory drive through the ac-
tion of NAAG, reduced number of GABAergic neurons, and
Y reduced ability to synthesize GABA, alone or in combination,
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

would impair the inhibitory control over corticolimbic


Neuroleptics
glutamatergic efferents. Conversely, pharmacological inter-
FIGURE 2. Schematic representation of hypothesized dys- ventions that presynaptically enhance GABAergic function
functional glutamatergic circuits in corticolimbic structures should have therapeutic effects.
in schizophrenia. NMDA receptors on local circuit GABA- An important consequence of disinhibition of corticolim-
ergic neurons are differentially sensitive to antagonism by bic glutamatergic efferents is excessive stimulation of
phencyclidine (PCP) and related drugs, as well as to endog- glutamate receptors on postsynaptic neurons. Persistent
enous N-acetylaspartyl glutamate (NAAG). The consequent activation of non-NMDA glutamate receptors can produce a
decreased local inhibition of corticolimbic glutamatergic ef-
delayed degeneration of neurons, with cytopathological fea-
ferents results in excessive activation of their postsynaptic
neurons via non-NMDA kainate (KA) receptors. Persistent tures of apoptosis- a form of neuronal death not associated
For personal use only.

activation of the KA receptors may lead to apoptotic degen- with an inflammatory glial reaction. This form of neuronal
eration of these neurons. DA, Dopamine; 5-HT serotonin. death is consistent with the reported absence of gliosis in
schizophrenic Indeed, systemic treatment with
CONCLUSIONS PCP and related noncompetitive NMDA-receptor antago-
nists causes a delayed neuronal degeneration in cortical and
The findings reviewed above can be integrated into the syn- limbic regions, with features of apoptosis.
aptic model of schizophrenia presented in Figure 2. The Whether neuronal death occurs in the mature brain in
NMDA-receptor antagonists such as PCP and ketamine re- schizophrenia- either at the time of symptomatic onset
produce the positive, negative, and cognitive symptoms of and/or as an ongoing process - remains a subject of debate.
schizophrenia in normal subjects, primarily through their Decreases in brain levels of NAA, a marker of neuronal
action at NMDA receptors on GABAergic interneurons in integrity, have been described in schizophrenia from NMR
cortical and limbic regions. The consequent hypofunction of spectroscopic studie~,"~ and an age-related prolongation of
these GABAergic neurons would increase the excitability of event-related potentials, similar to that observed in well-
projecting glutamatergic efferents from the regions. As a con- characterized neurodegenerative disorders, occurs in schizo-
sequence, the affected cortical areas would have a decreased phrenia."l Inadequate treatment early in the course of
ability to filter or restrict incoming excitatory input, resulting schizophrenia is associated with poorer long-term outcome,
in a spread of extraneous information. Regionally selective suggestive of a degenerative pro~ess.'~ Tardive dyskinesia
impairments in the inhibitory regulation of afferent and ef- might also be a manifestation of a latent neurodegenerative
ferent excitatory neurotransmission could account for such process in schizophrenia, with the threshold for manifesta-
symptoms of schizophrenia as hallucinations, delusions, and tion of symptoms lowered by neuroleptic treatment. Anti-
thought disorders. Reduced activity of NAALADase, observed oxidants reduce the symptoms of tardive dyskinesia, consis-
in the frontal cortex, hippocampus, and parahippocampus in tent with an apoptotic form of neuronal death.'47.'48How-
postmortem studies in schizophrenia, could produce an ana- ever, against this evidence for neuronal loss must be
tomically circumscribed "PCP-like" effect by favoring the considered a series of studies in which careful quantitation
maintenance of synaptic NAAG, which selectively antago- has not revealed a reduction in neuronal number.
nizes the NMDA receptors on the GABAergic interneurons. Perhaps one way of rationalizing the contradictory find-
Although this scenario is plausible and internally consis- ings in the neuropathology of schizophrenia is to recognize
tent, alternative findings have been reported. Nevertheless, the heterogeneity of the causes and the course of the
much of the postmortem research supports an operational disorder. Crow5 addressed this heterogeneity by proposing
hypofunction of the GABAergic interneurons. For example, two different forms of the disorder: type I, with little evidence
Benes and colleague^^^,^^ have developed circumstantial evi- of structural change in the brain and a predominance of
dence that there is a loss of GABAergic neurons in specific positive symptoms, and type 11, with obvious structural brain
layers of the anterior cingulate cortex (i.e., reduced numbers atrophy and prominent negative symptoms. The risk for tar-
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 249

dive dyskinesia is greater in patients with brain atrophy. phrenia revisited: the contribution of twin and high-risk stud-
Although more-recent studies do not support such a simple ies. J Nerv Ment Dis 1993;181:290-7.
dichotomy, the model proposed in this review would more 5. Crow TJ. Molecular pathology of schizophrenia: more than
likely be applicable to patients showing loss of corticolimbic one disease process. BMJ 1980;280:66-8.
6. Arndt S, Andreasen N, Flavum M, Miller D, Nopoulos P. A
volume, poor response to neuroleptic treatment, negative
longitudinal study of symptom dimensions in schizophrenia.
symptoms, and cognitive impairments. Consistent with this
Arch Gen Psychiatry 1995;52:352-60.
hypothesis, patients selected on the basis of preexisting 7. Bland RC, O m H. Fourteen-year outcome in early schizophre-
negative symptoms while being treated with neuroleptics nia. Acta Psychiatr Scand 1979;58:327-38.
exhibited a reduction in negative symptoms and improve- 8. Breier A, Schreiber JL, Dyer J, Pickar D. National Institute of
ment in cognitive function when treated with DCS, a partial Mental Health longitudinal study of chronic schizophrenia:
agonist at the glycine modulatory site on the NMDA prognosis and predictors of outcome. Arch Gen Psychiatry
receptor."" In a group of schizophrenic patients who were 1991;48:239-46.
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

receiving neuroleptics but were not selected for nega- 9. Heaton RK, Drexler M. Clinical neuropsychological findings in
tive symptoms, glycine loading also decreased negative schizophrenia and aging. In: Miller NE, Cohen GD, eds.
symptoms.'58 Schizophrenia and aging: schizophrenia, paranoia, and schizo-
phreniform disorders in later life. New York: Guilford, 1987:
Many issues remain unresolved with regard to the
145-61.
hypothesis of glutamatergic dysfunction in schizophrenia.
10. Park S, Holzman PS. Schizophrenics show spatial working
First, if the hypothesis is correct, then enhanced NMDA- memory deficits. Arch Gen Psychiatry 1992;49:975-82.
receptor function should also reduce positive symptoms. 11. Goldman-Rakic P. Prefrontal cortical dysfunction in
This might be accomplished with minimal risk of extrapy- schizophrenia: the relevance of working memory. In: Carroll
ramidal side effects. Second, the therapeutic effects of DCS B, Barrett J, eds. Psychopathology and the brain. New York:
and glycine on negative symptoms suggest that a n un- Raven, 1990:l-23.
For personal use only.

foreseen action of clozapine may be enhancement of NMDA- 12. Shenton M, Kikinis R, Jolesz F, Pollak S, LeMay M, Wible C,
receptor function. If this is the case, then DCS would et al. Abnormalities of the left temporal lobe and thought
probably not have additive effects with clozapine. To the disorder in schizophrenia: a qualitative magnetic resonance
contrary, as a partial agonist, DCS and related drugs might imaging study. N Engl J Med 1992;327:604-12.
13. Barta PE, Pearlson GD, Powers RE, Richards SS, Tune LE.
attenuate the therapeutic effects of clozapine on negative
Auditory hallucinations and smaller superior temporal gyral
symptoms. Third, clinical experience with neuroleptics and
volume in schizophrenia. Am J Psychiatry 1990;147:1457-62.
especially clozapine suggest that the full effects may not be 14. Carpenter WT Jr, Buchanan RW, Krkpatrick B, Tamminga C,
evident during the initial weeks of treatment. Given the role Wood F. Strong inference, theory testing, and the neu-
of the NMDA receptor in long-term potentiation and syn- roanatomy of schizophrenia. Arch Gen Psychiatry 1993;50:
aptic remodeling, it is possible that the therapeutic effects 825-31.
of enhancers of NMDA function might increase with time 15. Andreasen NC, Arndt S, Swayze V, Cizadlo T, Flaum M,
and might potentiate the impact of psychotherapeutic and O'Leary D, et al. Thalamic abnormalities in schizophrenia
rehabilitative interventions. Finally, most potent psycho- visualized through magnetic resonance image averaging. Sci-
tropic medications bear the risk of significant side-effects. ence 1994;266:294-8.
Since the NMDA receptor is involved in neuronal excitation 16. Saykin AJ, Shtasel DL, Gur RE, Kester DB, Mozley LH,
Stafiniak P, et al. Neuropsychological deficits in neuroleptic
and neurodegenera- tion, the risks for seizures and neuronal
naive patients with first-episode schizophrenia. Arch Gen
loss must be carefully assessed in clinical trials to enhance
Psychiatry 1994;51:124-31.
NMDA-receptor function. 17. Wyatt FLJ.Antipsychotic medication and long-term course of
The excellent secretarial assistance of Frances MacNeil and schizophrenia: therapeutic and theoretical implications. In:
helpful discussions with Drs. Benes, McCarley, Goff, Renshaw, and Shriqui CL, Nasrallah HA, eds. Contemporary issues in the
Cohen are gratefully acknowledged. treatment of schizophrenia. Washington, DC: American Psy-
chiatric Press [In press].
18. Loebel AD, Lieberman JA, Alvir JM, Mayerhoff DI, Geisler
SH, Szymanski SR. Duration of psychosis and outcome in
REFERENCES first-episode schizophrenia. Am J Psychiatry 1992;149:
1183-8.
Goldstein J. Sampling biases in studies of gender and schizo- 19. Carlsson A. Antipsychotic drugs, neurotransmitters and
phrenia. Schizophr Bull 1993;19:9-14. schizophrenia. Am J Psychiatry 1978;135:165-73.
Kraepelin E. Dementia praecox and paraphrenia. Edinburgh: 20. Snyder SH, Banerjee SP, Yamamura H. Drugs, neurotransmit-
Livingston, 1919. ters and schizophrenia. Science 1974;184:1243-53.
Kendler KS, Diehl SR. The genetics of schizophrenia: a cur- 21. Matthysse S. Antipsychotic drug actions: a clue to the neuro-
rent genetic-epidemiologic perspective. Schizophr Bull 1993; pathology of schizophrenia? Fed Proc 1973;32:200-5.
19:261-85. 22. Wong DF, Wagner HN Jr, Tune LE, Dannals RF, Pearlson GD,
Stabenah JR, Pollin W. Heredity and environment in schizo- Links JM, et al. Positron emission tomography reveals el-
Harvard Rev Psychiatry
250 Coyle January/February 1996

evated D2 dopamine receptors in drug-naive schizophrenics. ated glycine receptor site in vivo. Neuropharmacology 1991;
Science 1986;234:1558-63. 30: 1167-71.
23. Joyce JN, Lexow N, Bird E, Winokur A. Organization of 42. Lodge D, h a m JA, Church J, Davies SN, Martin 0,
dopamine D, and D, receptors in human striatum: receptor O’Shaughnessy CT, et al. Excitatory amino acids and phen-
autoradiographic studies in Huntington’s disease and schizo- cyclidine-like drugs. In: Hicks TP, Lodge D, McLennan H, eds.
phrenia. Synapse 1988;2:546-57. Excitatory amino acid transmission. New York: Alan R Liss,
24. Cross A J , Crow TJ, Owen F. ’H-Flupenthixol binding in 1987:83-90.
post-mortem brains of schizophrenics: evidence for a selective 43. Bliss TVP, Lomo T. Long-lasting potentiation of synaptic
increase in dopamine D2 receptors. Psychopharmacology transmission in the dentate area of the anaesthetized rabbit
1981;74:122-4. following stimulations of the perforant path. J Physiol 1973;
25. MacKay AV, Bird ED, Spokes EG, Rossor M, Iversen LL, 232~334-56.
Creese I, et al. Dopamine receptors and schizophrenia: drug 44. Kandel ER, Schwartz J . Principles of neural science. 3rd ed.
effect or illness? Lancet 1980;2:915-6. New York Elsevier, 1991.
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

26. Berman KF, Weinberger DR. Prefrontal dopamine and defect 45. Collingridge GL, Kehl SJ, McLennan H. Excitatory amino
symptoms in schizophrenia. In: Greden JF, Tandon R, eds. acids in synaptic transmission in the Schaffer collateral-com-
Negative schizophrenic symptoms. Washington, DC: American missural pathway of the rat hippocampus. J Physiol 1983;334:
Psychiatric Press, 199O:Bl-95. 33-46.
27. Konradi C, Kobierski L, Nguyen TV, Heckers S, Hyman SE. 46. Lynch G. Synapses, circuits and the beginnings of memory.
The CAMP-response-element-bindingprotein interacts, but fos Cambridge, Massachusetts: MIT Press, 1986.
protein does not interact, with the proenkephalin enhancer in 47. Stringer JL, Greenfield LJ, Hackett JT, Guyenet PG. Blockade
rat striatum. Proc Natl Acad Sci USA 1993;90:7005-9. of long-term potentiation by phencyclidine and u opiates in the
28. Meltzer H Y , Matsukara S, Lee JC. Classification of typical and hippocampus in vivo and in vitro. Brain Res 1983;280:127-38.
atypical antipsychotic drugs on the basis of dopamine D-1, D-2 48. Blakely RD, Coyle JT. The neurobiology of N-acetylaspartyl-
and serotonin-2 pKi values. J Pharmacol Exp Ther 1989;43: glutamate. Int Rev Neurobiol 1988;30:39-100.
587-604. 49. Tsai G, Slusher BS, Sim L, Coyle JT. Immunocytochemical
For personal use only.

29. Kornhuber J . Glutamate and schizophrenia [Letter]. Trends distribution of N-acetylaspartylglutamate in the rat forebrain
Pharmacol Sci 1990;11:357. and glutamatergic pathways. J Chem Neuroanat 1993;6:277-
30. Carlsson M, Carlsson A. Schizophrenia: a subcortical neu- 92.
rotransmitter imbalance syndrome? Schizophr Bull 1990;16: 50. Passani L, Paulsen R, Coyle JT. Vulnerability of pyramidal
425-32. neurons immunoreactive for N-acetylated dipeptide in Alz-
31. Javitt DC, Zukin SR. Recent advances in the phencyclidine heimer’s and Huntington’s diseases [Abstract]. SOCNeurosci
model of schizophrenia. Am J Psychiatry 1991;148:1301-8. Abstr 1995;21:774-7.
32. Olney JW,Sesma MA, Wozniak DF. Glutamatergic, cholin- 51. Guarda AS, Robinson MB, Ory-LavollBe L, Forloni GL,
ergic, and GABAergic systems in posterior cingulate cortex: Blakely RD, Coyle JT. Quantitation of N-acetyl-aspartyl-
interactions and possible mechanisms of limbic system dis- glutamate in microdissected rat brain nuclei and peripheral
ease. In: Vogt BA, Gabriel M, eds. Neurobiology of cingulate tissues: findings with a novel liquid phase radioimmunoassay.
cortex and limbic thalamus. Boston: Birkhauser, 1993:557-80. Mol Brain Res 1988;3:223-32.
33. Zipursky RB, Lim KO, Sullivan EV. Widespread cerebral gray 52. Tsai G, Stauch BL, Vornov JJ, Deshpande JK, Coyle JT.
matter volume deficits in schizophrenia. Arch Gen Psychiatry Selective release of N-acetylaspartylglutamate from rat optic
1992;49:195-205. nerve terminals in vivo. Brain Res 1990;518:313-6.
34. Robinson MB, Coyle JT. Glutamate and related acidic excita- 53. Robinson MB, Blakely RD, Couto R, Coyle JT. Hydrolysis of
tory neurotransmitters: from basic science to clinical applica- the brain dipeptide N-acetyl-D-aspartyl-D-glutamate:identifi-
tion. FASEB J 1987;1:446-55. cation and characterization of a novel N-acetylated-alpha-
35. Watkins JC, Olverman HJ. Agonists and antagonists for exci- linked acidic dipeptidase activity from rat brain. J Biol Chem
tatory amino acid receptors. Trends Neurosci 1987;10:265-72. 1987;262:14498-506.
36. Conti F, Fabri M, Manzoni T. Glutamate-positive corticocor- 54. Slusher BS, Robinson MB, Tsai G, Simmons ML, Richards SS,
tical neurons in the somatic sensory areas I and I1 of cats. Coyle JT. Rat brain N-acetylated alpha-linked acidic dipepti-
J Neurosci 1988;8:2948-60. dase activity: purification and immunologic characterization.
37. Roberts E. Prospects for research on schizophrenia: an hy- J Biol Chem 1990;265:21297-301.
pothesis suggesting that there is a defect in the GABA system 55. Meyerhoff JL, Robinson MB, Koller JJ, Bixler MA, Coyle JT.
in schizophrenia. Neurosci Res Prog Bull 1972;10:468-80. Kindling increases brain levels of NAAG and seizures reduce
38. Hollmann M, Heinemann S. Cloned glutamate receptors. Ann activity of a NAAG-hydrolyzing enzyme, NAALADase. In:
Rev Neurosci 1994;17:31-108. Avanzini G, Engel J Jr, Fariello R, Heinemann U, eds. Neu-
39. Schoepp DD, Conn PJ. Metabotropic glutamate receptors in rotransmitters in epilepsy. Amsterdam: Elsevier, 1992:297-
brain function and pathology. Trends Pharmacol Sci 1992;14: 305.
13-20. 56. Carter E, Feldman A, Coyle JT. Isolation of a cDNA clone for
40. Nakanishi S. Molecular diversity of glutamate receptors and an enzyme which generates glutamate from N-acetylaspartyl-
implications for brain function. Science 1992;258:597-603. glutamate [Abstract]. SOCNeurosci Abstr 1994;20:733.
41. Emmett MR, Mick SJ, Cler JA, Rao TS, Lyengar S, Wood PL. 57. Sekiguchi M, Okamoto K, Sakai Y. Low-concentration N-
Actions of o-cycloserine a t the N-methyl-o-aspartate-associ- acetylaspartylglutamate suppresses the climbing fiber re-
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 251

sponse of Purkinje cells in guinea pig cerebellar slices and the 74. Cohen BD, Rosenbaum G, Luby ED, Gottlieb JS. Comparison
responses to excitatory amino acids of Xenopus laeuis oocytes of phencyclidine hydrochloride (Sernyl) with other drugs. Arch
injected with cerebellar mRNA. Brain Res 1989:482:87-96. Gen Psychiatry 1962;6:395-401.
58. Puttfarcken P, Getz R, Coyle J . Kainic acid-induced lipid 75. Allen RM, Young SJ. Phencyclidine-induced psychosis. Am J
peroxidation: protection with butylated hydroxytoluene and Psychiatry 1978;135:1081-4.
U78517F in primary cultures of cerebellar granule cells. Brain 76. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R,
Res 1993;624:223-32. Bremner JD, et al. Subanesthetic effects of the noncompetitive
59. Wroblewska B, Wroblewski JT, Saab OH, Neale JH. N-Acetyl- NMDA antagonist, ketamine, in humans. Psychotomimetic,
aspartylglutamate inhibits forskolin-stimulated cyclic AMP perceptual, cognitive, and neuroendocrine responses. Arch
levels via a metabotropic glutamate receptor in cultured cer- Gen Psychiatry 1994;51:199-214,
ebellar granule cells. J Neurochem 1993;61:943-8. 77. Tsai G, Passani LA, Slusher BS, Carter R, Baer L, Kleinman
60. Grunze HCR, Barkai E, Hasselmo ME, McCarley RW, Rainnie JE, et al. Abnormal excitatory neurotransmitter metabolism
R, Greene RW. Selective sensitivity to NMDA antagonists of in schizophrenic brains. Arch Gen Psychiatry 1995;52:829-36.
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

inhibitory LTP compared to excitatory LTP in CA1 pyramidal 78. Slusher BS, Tsai G, Yo0 G, Coyle JT. Immunocytochemical
neurons. Neuroscience [In press]. localization of N-acetyl-aspartyl-glutamate (NAAG) hydrolyz-
61. Schwarcz R. Creese I, Coyle JT, Snyder SH. Dopamine recep- ing enzyme N-acetylated alpha-linked acidic dipeptidase
tors localised on cerebral cortical afferents to rat corpus (NAALADase).J Comp Neurol 1992;315:217-29.
striatum. Nature 1978;271:766-8. 79. Marshall BE, Longnecker DE. General anesthetics. In: Good-
62. Kerkerian L, Dusticier N, Nieoullon A. Modulatory effect of man LS, Gilman A, Rall TW, Nies AS, Taylor P, eds. The
dopamine on high-affinity glutamate uptake in the rat stria- pharmacological basis of therapeutics. New York: Pergamon,
tum. J Neurochem 1987;48:1301-6. 1990:285-310.
63. Maura G, Giardi A, Raiteri M. Release-regulating D-2 dopa- 80. Karp HN, Kaufman ND, Anand SK. Phencyclidine poisoning
mine receptors are located on striatal glutamatergic nerve in young children. J Pediatr 1980;97:1006-9.
terminals. J Pharmacol Exp Ther 1988;247:680-4. 81. Benes F. Myelination of cortico-hippocampal relays during late
For personal use only.

64. Fitzgerald LW, Deutch AY, Gasic G. Heinemann SF, Nestler adolescence: anatomical correlates to onset of schizophrenia.
EJ. Regulation of cortical and subcortical glutamate receptor Schizophr Bull 1989;15:585-94.
subunit expression by antipsychotic drugs. J Neurosci 1995; 82. Vogt 0. Proposition de fonder une organisation internationale
15:245341. pour l’etude d’anatomie pathologique de la schizophrenie et
65. Moghaddam B. Stress preferentially increases extraneuronal d’autres psychoses dites fonctionelles. In: Rosenberg P, ed.
levels of excitatory amino acids in prefrontal cortex: compari- Proceedings of the First International Congress of Neuropa-
son to hippocampus and basal ganglia. J Neurochem 1993;60: thology, Rome, 8-13 September 1952:674-7.
1650-7. 83. Corsellis JAN. Psychoses of obscure pathology. In: Blackwood
66. Jedema HP, Moghaddam B. Glutamatergic control of dopa- W, Corsellis JAN, eds. Greenfield’s neuropathology. Chicago:
mine release during stress in the rat prefrontal cortex. Edward Arnold, 1976:903-15.
J Neurochem 1994;63:785-8. 84. Weinberger DR, Torrey EF, Neophytides AN, Wyatt RJ. Struc-
67. Wang KT. Presynaptic glutamate receptors modulate dopa- tural abnormalities in the cerebral cortex of chronic schizo-
mine release from striatal synaptosomes. J Neurochem 1991; phrenic patients. Arch Gen Psychiatry 1979;36:935-9.
57:819-22. 85. Weinberger DR, Bigelow LB, Kleinman J E , Klein ST, Rosen-
68. Krebs MO, Desce JM, Kemel ML, Gauchy C, Godeheu G, blatt J E , Wyatt RJ. Cerebral ventricular enlargement in
Cheramy A, et al. Glutamatergic control of dopamine release chronic schizophrenia: an association with poor response to
in the rat striatum: evidence for presynaptic N-methyl-o- treatment. Arch Gen Psychiatry 1980;37:11-3.
aspartate receptors on dopaminergic nerve terminals. J Neu- 86. Tandon R, Greden JF. Negative symptoms of schizophrenia: a
rochem 1991;56:81-5. need for conceptual clarity. Biol Psychiatry 1991;30:321-5.
69. Grace AA. Cortical regulation of subcortical dopamine systems 87. Diamond MC, Law F, Rhodes H, Lindner B, Rosenzweig MR,
and its possible relevance to schizophrenia. J Neural Transm Krech D, et al. Increases in cortical depth and glia numbers in
Gen Sect 1993;91:111-34. rats subjected to enriched environments. J Comp Neurol
70. Dragunow M, Robertson GS, Faull RL, Robertson HA, Jansen 1966;128:117-26.
K. D, dopamine receptor antagonists induce fos and related 88. Sirevaag AM, Greenough WT. Differential rearing effects on
proteins in rat striatal neurons. Neuroscience 1990;37:287- rat visual cortex synapses 111: neuronal and glial nuclei,
94. boutons, dendrites, and capillaries. Brain Res 1987;424:320-
71. Robertson GS, Vincent SR, Fibiger HC. D, and D, dopamine 32.
receptors differentially regulate c-fos expression in striatoni- 89. Benes FM. Is there a neuroanatomic basis for schizophrenia?
gral and striatopallidal neurons. Neuroscience 1992;149:285- Neuroscientist 1995;1:112-20.
96. 90. Benes FM, Davidson J, Bird ED. Quantitative cytoarchitec-
72. Robertson GS, Matsumura H, Fibiger HC. Induction patterns tural studies of cerebral cortex of schizophrenics. Arch Gen
of fos-like immunoreactivity in the forebrain as predictors of Psychiatry 1986;43:31-5.
atypical antipsychotic activity. J Pharmacol Exp Ther 1994; 91. Bogerts B, Falkai P, Tutsch J. Cell numbers in the pallidum
271:1058-66. and hippocampus of schizophrenics. In: Shagass C. Josiassen
73. Konradi C. Effects of D-cycloserine on striatal cfos expression. RC, Bridger WH, Weiss KJ, Stoff D, Simpson GM, eds. Bio-
submitted. logical psychiatry 1985. Amsterdam: Elsevier, 1986:1178-80.
Halvard Rev Psychiatry
252 Coyle January/February 1996

92. Jeste D, Lahr JB. Hippocampal pathologic findings in schizo- bcl-2 can selectively rescue neurotrophic factor dependent
phrenia. Arch Gen Psychiatry 1989;46:1019-24. neurons from apoptosis. Cell 1993;73:295-307.
93. Pakkenberg B. Pronounced reduction of total neuron number 113. Hockenbery DM, Oltvai ZN, Yin X M , Milliman CL, Korsmeyer
in mediodorsal thalamic nucleus and nucleus accumbens in SJ. Bcl-2 functions in an antioxidant pathway to prevent
schizophrenia. Arch Gen Psychiatry 1990;47:1023-8. apoptosis. Cell 1993;75:241-4.
94. Heckers S, Heinsen H, Geiger B, Beckmann H. Hippocampal 114. Olney JW,Labruyere J , Price MT. Pathological changes in-
neuron number in schizophrenia. Arch Gen Psychiatry 1991; duced in cerebrocortical neurons by phencyclidine and related
4811002-8. drugs. Science 1989;244:1360-2.
95. Akbarian S, Bunney WE Jr, Potkin SG, Wigal SB, Hagman JO, 115. Olney JW,Farber NB. Efficacy of clozapine compared with
Sandman CA, et al. Altered distribution of nicotinamide- other antipsychotics in preventing NMDA-antagonist neuro-
adenine dinucleotide phosphate-diaphorase cells in frontal toxicity. J Clin Psychiatry 1994;55(9 suppl B):43-6.
lobe of schizophrenics implies disturbances of cortical devel- 116. Olney JW,Sesma MA, Wozniak DF. Glutamatergic, cholin-
opment. Arch Gen Psychiatry 1993;50:169-77. ergic, and GABAergic systems in posterior cingulate cortex:
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

96. Bird ED, Spokes EGS, Iversen LL. Increased dopamine con- interactions and possible mechanisms of limbic system dis-
centration in limbic areas of brain from patients dying with ease. In: Vogt BA, Gabriel M, eds. Neurobiology of cingulate
schizophrenia. Brain 1979;107:211-5. cortex and limbic thalamus. Boston: Birkhauser, 1993:557-80.
97. Benes FM, Sorensen I, Bird ED. Reduced neuronal size in 117. Sharp FR, Jasper P, Hall J, Noble L, Sagar SM. MK-801 and
posterior hippocampus of schizophrenic patients. Schizophr ketamine induce heat shock protein HSP72 in injured neurons
Bull 1991;17:597-608. in posterior cingulate and retrosplenial cortex. Ann Neurol
98. Hanada S, Mita T, Nishino N, Tanaka C. 3H-Muscimolbinding 1991;30:801-9.
sites increased in autopsied brains of chronic schizophrenics. 118. O’Callaghan JP. Biochemical analysis of glial fibrillary acidic
Life Sci 1987;12:259-66. protein as a quantitative approach to neurotoxicity assessment
99. Akbarian S, Kim JJ, Potkin SG, Hagman JO, Tafazzoli A, advantages, disadvantages and application to the assessment of
Bunney WE Jr, et al. Gene expression for glutamic acid NMDA receptor antagonist-induced neurotoxicity. Psycho-
decarboxylase is reduced without loss of neurons in prefrontal
For personal use only.

pharmacol Bull 1994;30:549-54.


cortex of schizophrenics. Arch Gen Psychiatry 1995;52:258- 119. Farber NB, Price MT, Labruyere J, Nemnich J , St Peter H,
66. Wozniak DF, et al. Antipsychotic drugs block phencyclidine
100. Falkai P, Bogerts B, Rozumek M. Limbic pathology in receptor-mediated neurotoxicity. Biol Psychiatry 1993;34:
schizophrenia: the entorhinal region - a morphometric study. 119-2 1.
Biol Psychiatry 1988;24:515-2 1. 120. Sharp FR, Butman M, Wang S, Koistinaho J , Graham SH,
101. Bloom FE. Advancing a neurodevelopmental origin for schizo- Sagar SM, et al. Haloperidol prevents induction of the hsp70
phrenia. Arch Gen Psychiatry 1993;50:224-7. heat shock gene in neurons injured by phencyclidine (PCP),
102. Olney JW.Glutamate-induced neuronal necrosis in the infant MK-801, and ketamine. J Neurosci Res 1992;33:605-16.
mouse hypothalamus. J Neuropathol Exp Neurol 1971;30:75- 121. O’Donnell BF, Faux SF, McCarley RW, Kimble MO, Salisbury
90. DF, Nestor PG, et al. Increased rate of P300 slowing with age
103. Zaczek R, Coyle JT. Excitatory amino acid analogues: in schizophrenia: electrophysiological evidence for a neurode-
neurotoxicity and seizures. Neuropharmacology 1982;21:15- generative process. Arch Gen Psychiatry 1995;52:544-9.
26. 122. Homberg V, Hefter H, Granseyer G, Strauss W, Lange H,
104. Olney JW.Excitotoxic amino acids and neuropsychiatric dis- Hennerici M. Event-related potentials in patients with Hun-
orders. Ann Rev Pharmacol Toxicol 1990;30:47-61. tington’s disease and relatives at risk in relation to detailed
105. Rothstein J, Margaret L, Dykes-Hoberg M, Kuncl R. Chronic psychometry. Electroencephalogr Clin Neurophysiol 1986;63:
inhibition of glutamate uptake produces a model of slow 552-69.
neurotoxicity. Proc Natl Acad Sci USA 1993;90:6591-5. 123. Patterson JV,Michalewski HJ, Starr A. Latency variability of
106. Choi DW, Maulucci-Gedde M, Kriegstein AR. Glutamate neu- the components of auditory event-related potentials to infre-
rotoxicity in cortical cell culture. J Neurosci 1987;7:357-68. quent stimuli in aging, Alzheimer-type dementia, and depres-
107. Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and sion. Electroencephalogr Clin Neurophysiol 1988;71:450-60.
neurodegenerative disorders. Science 1993;262:689-95. 124. Simmons ML, Frondoza CG, Coyle JT. Immunocytochemical
108. Raff M, Barres B, Burne J , Coles H, Ishizaki Y, Jacobson M. localization of N-acetyl-L-aspartate with monoclonal antibod-
Programmed cell death and the control of cell survival: lessons ies. Neuroscience 199 1;45:37-45.
from the nervous system. Science 1993;262:695-700. 125. Guimares AR, Schwartz P, Parkash MR, Berger W, Jenkens
109. Reed J . Bcl-2 and the regulation of programmed cell death. BG, Coyle JT, e t al. Quantitative in uiuo ‘H chemical shift
J Cell Biol 1994;124:1-6. imaging of excitotoxicity in rat brain. Neuroscience [In press].
110. Wyllie AH, Kerr JF, Currie AR. Cell death: the significance of 126. Tsai GC, Coyle JT. N-Acetylaspartate in neuropsychiatric
apoptosis. Int Rev Cytol 1980;68:251-306. disorders. Prog Neurobiol 1995;46:531-40.
111. Simonian NA, Getz RL, Leveque JC, Konradi C, Coyle JT. 127. Yurgelun-Todd DA, Renshaw PF, Waternaux C, Cohen BM.
Kainic acid induces apoptosis in neurons in uitro. Neuro- Proton magnetic resonance spectroscopy of the temporal lobes
science [In press]. in schizophrenics and normal controls. Schizophr Res [In
112. Allsop T, Wyatt S, Paterson H, Davies A. The proto-oncogene press].
Harvard Rev Psychiatry
Volume 3, Number 5 Coyle 253

128. Meyerhoff DJ, MacKay S , Bachman L, Poole N, Dillon WP, sistent neuroleptic-induced dyskinesia of regional changes in
Weiner MW, et al. Reduced brain N-acetylaspartate suggests brain GABA synthesis. Nature 1984;309:347-9.
neuronal loss in cognitively impaired human immunodefi- 144. Hoffman W, Casey DE. Computed tomographic evaluation
ciency virus seropositive individuals: in vivo 'H magnetic of patients with tardive dyskinesia. Schizophr Res 1991;5:
resonance spectroscopic imaging. Neurology 1993;43:509-15. 1-12.
129. Kerwin RW, Patel S, Meldrum BS, Czudek C, Reynolds GP. 145. Mitchell IJ, Lawson S, Moser B, Laidlaw SM, Cooper AJ,
Asymmetrical loss of glutamate receptor subtype in left hip- Walkinshaw G, et al. Glutamate-induced apoptosis results in a
pocampus in schizophrenia. Lancet 1988;1:583-4. loss of striatal neurons in the parkinsonian rat. Neuroscience
130. Kerwin R, Patel S , Meldrum B. Quantitative autoradiographic 1994;63:1-5.
analysis of glutamate binding sites in the hippocampal forma- 146. Tsai G, Goff D, Chang R, Flood J, Baer L, Coyle JT. Increased
tion in normal and schizophrenic brain post mortem. Neuro- glutamatergic neurotransmission and oxidative stress in tar-
science 1990;39:25-32. dive dyskinesia. Biol Psychiatry [In press].
131. Harrison PJ, McLaughlin D, Kerwin RW. Decreased hippo- 147. Adler LA, Peselow E, Rotrosen J, Duncan E. Lee M, Rosenthal
Harv Rev Psychiatry Downloaded from informahealthcare.com by University of Toronto on 11/28/14

campal expression of a glutamate receptor gene in schizo- M, et al. Vitamin E treatment of tardive dyskinesia. Am J
phrenia. Lancet 1991;337:450-2. Psychiatry 1993;150:1405-7.
132. Deakin JFW, Slater P, Simpson MDC, Gilchrist C, Skan WJ, 148. Egan MF, Hyde TM, Albers GW, Elkashef A, Alexander RC,
Royston MC, et al. Frontal cortical and left temporal Reeve A, et al. Treatment of tardive dyskinesia with vitamin
glutamatergic dysfunction in schizophrenia. J Neurochem E. Am J Psychiatry 1992;149:773-7.
198932:178 1-6. 149. Lawlor BA, Davis KL. Does modulation of glutamatergic
133. Nishikawa T, Takashima M, Toru M. Increased ['Hlkainic function represent a viable therapeutic strategy in Alzhei-
acid binding in the prefrontal cortex in schizophrenia. Neuro- mer's disease? Biol Psychiatry 1992;31:337-50.
sci Lett 1983;40:245-50. 150. Henderson G, Johnson JW, Ascher P. Competitive antagonists
134. Toru M, Watanabe S , Shibuya H, Nishikawa T, Noda K, and partial agonists at the glycine modulatory site of the
Mitsushio H, et al. Neurotransmitters, receptors and neu- mouse N-methyl-D-aspartate receptor. J Physiol (Lond) 1990;
ropeptides in post-mortem brains of chronic schizophrenic 430: 189-212.
For personal use only.

patients. Acta Psychiatr Scand 1988;78:121-37. 151. Watson GB, Bolanowski MA, Baganoff MP, Deppeler CL,
135. Kane J M , Smith JM. Tardive dyskinesia: prevalence and risk Lanthorn TH. D-Cycloserine acts as a partial agonist at the
factors, 1959 to 1979. Arch Gen Psychiatry 1982;39:473-81. glycine modulatory site of the NMDA receptor expressed in
136. Chouinard G, Annable L, Ross-Chouinard N, Mercier P. A Xenopus oocytes. Brain Res 1990;510:158-60.
5-year prospective longitudinal study of tardive dyskinesia: 152. Wlaz P, Baran H, Loscher W. Effect of the glycine/NMDA
factors predicting appearance of new cases. J Clin Psycho- receptor partial agonist, D-cycloserine, on seizure threshold
pharmacol 1988;8(4 suppl):21-6. and some pharmacodynamic effects of MK-801 in mice. Eur J
137. Waddington JL, Youssef HA. An unusual cluster of tardive Pharmacol 1994;257:217-25.
dyskinesia in schizophrenia: association with cognitive dys- 153. Iwainsky H. Mode of action, biotransformation and pharma-
function and negative symptoms. Am J Psychiatry 1986;143: cokinetics of antituberculosis drugs in animals and man. In:
1162-5. Bartmann K, ed. Handbook of experimental pharmacology.
138. Brown KW, White T. The association among negative symp- Berlin: Springer, 1988:124-33.
toms, movement disorders and frontal lobe psychological defi- 154. Thomas JW, Hood W, Monahan JB, Contreras PC,
cits in schizophrenic patients. Biol Psychiatry 1991;30:1182- O'Donohue TL. Glycine modulation of the phencyclidine bind-
90. ing site in mammalian brain. Brain Res 1988;442:396-8.
139. Fenton WS, Wyatt RJ, McGlashan TH. Spontaneous dyskine- 155. Chessell IP, Procter AW, Francis PT, Bowen DM. D-Cycloser-
sia in schizophrenia [Abstract]. Schizophr Res 1995;15:207. ine, a putative cognitive enhancer, facilitates activation of the
140. Owens DG. Involuntary movements in chronic schizophrenia: N-methyl-D-aspartate receptor-ionophore complex in Alz-
the role of illness and its treatment. In: Casey DE, ed. heimer brain. Brain Res 1991;565:345-8.
Dyskinesia research and treatment. Berlin: Springer, 1985: 156. Goff DC, Tsai G, Manoch DS, Coyle JT. A dose-finding trial of
79-87. D-cycloserine added to neuroleptic for negative symptoms in
141. Hoffman WF, Kadri N, Fenn DS, Moussaoui D, Bentounssi B, schizophrenia. Am J Psychiatry 1995;152:1213-5.
Tilane A, et al. Abnormal movements in never medicated 157. Cascella NG, Macciardi F, Cavallini R, Smeraldi E. D-Cyclo-
schizophrenics [Abstract]. Schizophr Res 1995;15:208. serine adjuvant therapy to conventional neuroleptic treatment
142. Anderson U, Haggstrom JE, Levin ED, Bondesson U, Val- in schizophrenia: an open-label study. J Neural Transm Gen
verius M, Gunne LM. Reduced glutamate decarboxylase activ- Sect 1994;95:105-1 1.
ity in the subthalamic nucleus in patients with tardive dyski- 158. Javitt DC, Zylberman I, Zukin SR, Heresco-Levy U, Linden-
nesia. Mov Disord 1989;4:37-46. mayer JP. Amelioration of negative symptoms in schizophre-
143. Gunne LM, Haggstrom JE, Sjoquist B. Association with per- nia by glycine. Am J Psychiatry 1994;151:1234-6.

You might also like