the glutamate hypothesis of schizophreniapositive schizophrenic symptoms, the glutamate hypothesis...

21
PHARMACOLOGY AND PATHOPHYSIOLOGY eNS Drugs 1997 Jon; 7 (1)' 4UJ7 1172-7047/97/IXXll-ffi47/SlO.50/O © Adis International limited. All rights reserved. The Glutamate Hypothesis of Schizophrenia Therapeutic Implications Masahiko J. Ishimaru 1 and Michio Toru 2 1 Department of Molecular and Cellular Neuroscience, Medical Research Institute of Tokyo Medical and Dental University, Tokyo, Japan 2 Department of Neuropsychiatry, Medical School of Tokyo Medical and Dental University, Tokyo, Japan Contents Summary ........................... . 1. Basis for the Glutamate Hypothesis of Schizophrenia . . . 1.1 The Dopamine Hypothesis - Validity and Limitations . 1.2 Glutamate Levels in Schizophrenic Brains and CSF 1.3 Phencyclidine-Induced Psychosis .......... . 1.4 Glutamate Receptors in Schizophrenic Brains . . . . 1.5 Interaction of Glutamatergic and Dopaminergic Systems 2. Therapeutic Implications of the Glutamate Hypothesis . 2.1 Antipsychotics and the Glutamatergic System .. 2.2 Therapeutic Target Sites on the NMDA Receptor . 2.3 Glycinergic Therapies for Schizophrenia . . . . . . 3. Some Interesting Questions . . . . . . . . . . . . . . . . 3.1 What is the Basis of Glutamatergic Hypoactivity? 3.2 Is the Glycine Site Saturated In Vivo? .. . . . . . 3.3 Is D-Serine an Endogenous Agonist for the Glycine Site? 4. Conclusion ............................ . . 47 . 48 .48 48 49 52 54 55 55 55 56 60 60 61 61 62 Summary The glutamate hypothesis of schizophrenia has been developed based on the observation that psychotic symptoms induced by phencyclidine and related agents, which are antagonists at the N-methyl-D-aspartate (NMDA) glutamate receptor, closely resemble both the positive and negative symptoms of schizo- phrenia. In contrast to the dopamine hypothesis, which explains primarily with positive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also support the glutamate hypothesis by demonstrat- ing alterations in glutamate receptors in several brain areas in patients with schizophrenia, especially remarkable increases in the receptors in frontal and parieto-temporal association fields. This increase in glutamate receptors may reflect postsynaptic up-regulation in response to a deficiency in glutamatergic neuronal activity. Thus, the glutamate hypothesis implies that schizophrenic

Upload: others

Post on 25-Aug-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

PHARMACOLOGY AND PATHOPHYSIOLOGY eNS Drugs 1997 Jon; 7 (1)' 4UJ7 1172-7047/97/IXXll-ffi47/SlO.50/O

© Adis International limited. All rights reserved.

The Glutamate Hypothesis of Schizophrenia Therapeutic Implications

Masahiko J. Ishimaru 1 and Michio Toru2

1 Department of Molecular and Cellular Neuroscience, Medical Research Institute of Tokyo Medical and Dental University, Tokyo, Japan

2 Department of Neuropsychiatry, Medical School of Tokyo Medical and Dental University, Tokyo, Japan

Contents

Summary ........................... . 1. Basis for the Glutamate Hypothesis of Schizophrenia . . .

1.1 The Dopamine Hypothesis - Validity and Limitations . 1.2 Glutamate Levels in Schizophrenic Brains and CSF 1.3 Phencyclidine-Induced Psychosis .......... . 1.4 Glutamate Receptors in Schizophrenic Brains . . . . 1.5 Interaction of Glutamatergic and Dopaminergic Systems

2. Therapeutic Implications of the Glutamate Hypothesis . 2.1 Antipsychotics and the Glutamatergic System .. 2.2 Therapeutic Target Sites on the NMDA Receptor . 2.3 Glycinergic Therapies for Schizophrenia . . . . . .

3. Some Interesting Questions . . . . . . . . . . . . . . . . 3.1 What is the Basis of Glutamatergic Hypoactivity? 3.2 Is the Glycine Site Saturated In Vivo? .. . . . . . 3.3 Is D-Serine an Endogenous Agonist for the Glycine Site?

4. Conclusion ............................ .

. 47

. 48

.48 48 49 52 54 55 55 55 56 60 60 61 61 62

Summary The glutamate hypothesis of schizophrenia has been developed based on the observation that psychotic symptoms induced by phencyclidine and related agents, which are antagonists at the N-methyl-D-aspartate (NMDA) glutamate receptor, closely resemble both the positive and negative symptoms of schizo­phrenia. In contrast to the dopamine hypothesis, which explains primarily with positive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness.

Postmortem brain assays also support the glutamate hypothesis by demonstrat­ing alterations in glutamate receptors in several brain areas in patients with schizophrenia, especially remarkable increases in the receptors in frontal and parieto-temporal association fields. This increase in glutamate receptors may reflect postsynaptic up-regulation in response to a deficiency in glutamatergic neuronal activity. Thus, the glutamate hypothesis implies that schizophrenic

Page 2: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

48 Ishimaru & Toru

symptoms might be ameliorated by augmenting glutamatergic neural transmis­sion.

Recent clinical studies have been conducted using glycine, milacemide or D-cycloserine as adjuncts to or as a replacement for antipsychotics, in an attempt to augment NMDA receptor-mediated glutamatergic functions by activating the glycine modulatory site. Glycine adjuvant treatment was moderately effective in some patients in all studies and significantly improved negative symptoms in a placebo-controlled study. Replacement of antipsychotics by milacemide, a sup­posed glycine prodrug that crosses the blood-brain barrier, did not ameliorate schizophrenic symptoms. This lack of effect may be due to the fact that milacem­ide is predominantly an inhibitor of monoamine oxidases rather than a glycine agonist. Adjuvant administration ofD-cycloserine, a partial agonist ofthe glycine site, was inconclusive, with improvement in negative symptoms occurring in one study and overall exacerbation in most patients in another study.

Well designed placebo-controlled studies using orally active full agonists of the glycine regulatory site should be pursued in the future. Also, intervention at other sites within the NMDA receptor complex should be considered.

1. Basis for the Glutamate HypotheSiS of Schizophrenia

1.1 The Dopamine Hypothesis - Validity and Limitations

The dopamine hypothesis was the first durable biological framework for understanding the aetiol­ogy and treatment of schizophrenia. This hypothe­sis was an unexpected fruit of antipsychotics, drugs that had dramatically changed clinical and research practice in the field of psychiatry. Chlorpromazine was introduced in 1952 as the first effective anti­psychotic observed by modem scientists,[I] and its development triggered a burst of discoveries of various other antipsychotics. It was subsequently shown that all these antipsychotics were dopamine receptor antagonists,[2.3] with preferential affinity for the D2 receptor.[4] In addition, amphetamines were found to induce psychotic symptoms that closely mimicked the positive symptoms of schizo­phrenia[5] by facilitating dopaminergic neural transmission. [6]

Based on these findings, the dopamine hypoth­esis of schizophrenia has been largely accepted as the basis for the positive symptoms of the illness. However, clinicians have noticed that classical antipsychotics are generally ineffective in amelio-

© Adis International Limited. All rights reserved.

rating the negative symptoms[7] that are prominent in patients with a chronic course.l8]

Antipsychotics render schizophrenia manage­able, but do not completely cure the disorder. Ex­planations other than dopaminergic hyperactivity are required for further understanding of the ill­ness, as well as for therapeutic advancement. As reviewed below, accumulating evidence indicates that the glutamate hypothesis is one such candidate that may provide a more comprehensive view of schizophrenia, into which dopaminergic hyperac­tivity may be incorporated.

1 .2 Glutamate Levels in Schizophrenic Brains and CSF

Glutamate is the major excitatory neurotrans­mitter in the mammalian brain. [9. 10] Its effects are mediated through binding to a number of recep­tors: N-methyl-D-aspartate (NMDA), kainate, a­amino-3-hydroxy-5-methyl-4-isoxazole propio­nate (AMPA) and metabotropic. Glutamate is responsible for not only the classical fast synaptic transmission through non-NMDA receptors, but a long term potentiation through the NMDA recep­tor. This latter effect underlies neuronal plastic­ity.lll]

Dysfunction in glutamatergic neural transmis­sion is assumed in various neuropsychiatric disor-

eNS Drugs 1997 Jan; 7 (1)

Page 3: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

ders.D2,13] In 1980, Kim et al.[14] observed a highly significant decrease in the level of glutamate in the CSF of patients with schizophrenia compared with controls (i.e. patients with non-specific neurologi­cal disorders or healthy volunteers). From these findings they postulated that impaired glutamat­ergic function underlies schizophrenia. Although these authors were the first to propose the gluta­mate hypothesis of schizophrenia, their original finding was not supported by the studies that fol­lowed. In subsequent studies, CSF glutamate lev­els in patients with schizophrenia were reported to be no different from,[15,16] or even higher than, those of individuals who had nonspecific neuro­logical diseases.!17]

Despite this inconsistency (which is of un­known origin), the idea of glutamatergic hypofunc­tion in schizophrenia encouraged further studies of the brains of patients with the disorder, in which various methods were employed for detecting glutamatergic abnormalities. In several studies, measurement of brain glutamate levels in postmor­tem samples from patients with schizophrenia failed to show a significant change.!15,18] On the other hand, a recent preliminary study using proton magnetic resonance spectroscopy reported that the prefrontal glutamate level measured in vivo was significantly lower in never-treated schizophrenic patients than in healthy volunteers.!19]

Nevertheless, a measurement of the total gluta­mate level in a given brain area, either postmortem or in vivo, is difficult to interpret, since only a small fraction of the total glutamate represents that which functions as a neurotransmitter. Sherman et al. assayed the synaptosomal fraction (i.e. that con­taining the pre-synaptic neurons) derived from the neocortex of patients with schizophrenia and ob­served a significant reduction in glutamate release induced by veratridine,[20] NMDA and kainate.[21] Recently, Tsai et alP2] performed a postmortem study of brains from patients who had had schizo­phrenia that focused on N-acetylaspartyl-gluta­mate (NAAG), a neuropeptide that is highly con­centrated in glutamatergic neurons.!22] NAAG is a partial antagonist of NMDA receptors and, more-

© Adis Internalional Umited. All rights reserved.

49

over, is cleaved to glutamate and N-acetylaspartate by a specific peptidase, N-acetyl-a linked acidic dipeptidase (NAALADase). In the prefrontal cor­tex and hippocampus of patients who had had chronic schizophrenia, levels of NAAG were in­creased whereas NAALADase activity and levels of glutamate and aspartate were decreased com­pared with samples from unaffected individu­als.[22] From these findings it can be postulated that a deficiency in NAALADase leads to an increase in NAAG as well as to a decrease in glutamate, both of which might impair glutamatergic neural transmission. These findings suggest that a defi­ciency in glutamatergic neural transmission occurs in the schizophrenic brain.

1,3 Phencyclidine-Induced Psychosis

Observations that phencyclidine induces psy­chosis provided strong evidence for the glutamate hypothesis of schizophrenia. Phencyclidine, like the related compound ketamine, was originally de­veloped as a dissociative anaesthetic.[23] It had to be abandoned, however, because the adverse ef­fects associated with its use, especially postopera­tive agitation and delirium that could last for hours, were too serious to be tolerated.[24]

Early reports had already pointed out that phencyclidine-induced psychotic symptoms could provide a useful model of schizophreniaps.26] The connection between phencyclidine psychosis and schizophrenia was noted when the admission rate for what appeared to be unusually long, severe and treatment-resistant initial schizophrenic psychoses suddenly tripled at a mental health centre in Wash­ington DC, US, in 1973. All the patients admitted with these symptoms turned out to have smoked 'Angel Dust' , a streetterm for phencyclidine. [27] In addition to the symptoms previously observed in healthy volunteers who were given phencyclidine such as deficits in ego boundaries and feelings of depersonalisation, derealisation and estrange­ment,[25.28.29] the patients showed the following characteristics: (i) not only positive symptoms (hallucinations and delusions), but also negative symptoms (blunt affect, poverty of speech, apathy

eNS Drugs 1997 Jan: 7 (1)

Page 4: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

50

Cell membrane

Glutamate (NMDA)

I

Competitive antagonists CPP CPPene Selfotel

Ion channel

AP-5 Noncompetitive antagonists Open channel blockers

PCP Ketamine Dizocilplne rcp CyclaZOCine

Polyamine site agonists Spermine Spermidine

Glycine site ligands Full agonist

D-Serine (endogenous agonist?)

Partial agonlsts D-Cyclosenne (agonistic) (+)-HA-966 (antagonistic)

Antagonists Kynurenate 7-CIKA

lshimaru & Toru

Fig. 1. A schematic diagram of the N-methyl-D-aspartate (NMDA) receptor ion channel complex and multiple recognition sites associated with it. Endogenous ligands for each recognition site are shown above the representation of the receptor. while exogenous ligands that are mentioned in the text are listed below. Abbreviations: AP-5 = 2-amino-phosphonovaleric acid; 7-CIKA = 7-chloro­kynurenic acid; CPP = 3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid; CPPene = 3-(2-carboxypiperazin-4-yl)-1-propenyl-1-phosphonic acid; dizocilpine (MK-801) = (+)-5-methyl-1 0, 11-dihydro-SH-dibenzo-[a,d)cycloheptan-S, 1 O-imine malate; (+ )-HA-966 = (+ )-3-amino-1-hydroxypyrrolid-2-one; PCP = phencyclidine; sellotel (CGS-197SS) = cis-4-phosphonomethyl-2-piperidine carboxylic acid; rcp = N-[1-(2thienyl)cyclohexyl)piperidine.

and attention deficit); (ii) the symptoms generally did not respond to treatment with antipsychotics; and (iii) in some individuals, the symptoms lasted for days to weeks despite abstinence from the drug. [27 ,301

Administration of phencyclidine to schizophre­nic patients evoked or worsened their symptoms for a period that lasted for days[31] to weeksJ251 Phencyclidine-intoxicated patients could be differ­entiated from those with schizophrenia only by a lack of premorbid psychopathologyJ321 Therefore, phencyclidine psychosis was considered to provide a more comprehensive model of schizophrenia than amphetamine psychosis, which mimics only the positive symptoms of schizophrenia,l331

© Adis International Limited. All rights reserved.

However, the mechanism of phencyclidine psy­chosis remained unclear for many years. The main reason for this was the diverse mechanisms of ac­tion of the drug; it interacts with various transmitter systems such as dopamine, noradrenaline (norepi­nephrine), acetylcholine, serotonin (5-hydroxy­tryptamine; 5-HT) and sigma opioid as well as with cation channels. l341 Phencyclidine-induced 'amphetamine-like' behaviours in animals, such as increased locomotor activity, stereotypy and rota­tion, were ascribed to its indirect dopamino­mimetic effectsJ351

Subsequently, Lodge and colleaguesl36,371 dem­onstrated that phencyclidine (and ketamine) blocks NMDA-induced depolarisation of spinal neurons

eNS Drugs 1997 Jan; 7 (1)

Page 5: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

while having no effect on non-NMDA mediated depolarisation.l36,37] This observation was sup­ported by a number of studies that followed, and eventually phencyclidine was shown to be an ion channel blocker, It enters the ion channel associ­ated with the NMDA receptor in its open state, is trapped at the binding site located deep within the channel, and prevents calcium ions from entering the cytoplasm (fig. 1))34] Other noncompetitive NMDA receptor antagonists, such as dizocilpine (MK-80 1), N-[ 1-(2thienyl)cyclohexyl]piperidine

51

(TCP), ketamine and cyclazocine, share these properties and the resultant anticonvulsant and neuroprotective effects.[34] All these compounds produce phencyclidine-like abnormal behaviours in animals,[38] as well as phencyclidine-like psy­chotic symptoms in humans. [39-42] Similarly, Javitt and Zukin[43] reviewed previous studies and sug­gested that, in the dose range relevant to the psychotomimetic effect, phencyclidine would act exclusively on the NMDA ion channel.[43] Phen­cyclidine-like psychotic symptoms are also ob-

Table I. Glutamate receptors in the brains of patients with schizophrenia, assessed at postmortem using radiolabelled receptor assays

Reference Year Ligand/displacer No. of patients Results (patients/controls)

Kainate (KA) Nishikawa et al.[50] 1983 [3H1KAlGlu 12110 Frontal cortex (SA 8,9+ 10+46) i; frontal cortex (SA

11+12,45+47), putamen H

Kerwin et al.[51] 1988 [3H1KAlGlu (ARG) 11/9 Left hippocampus !; right hippocampus H

Toru et al.[52] 1988 [3H1KAlGlu 14/10 Parietal cortex (SA 39) i; parietal cortex (except for SA 39), occipital cortex H

Deakin et al. [53] 1989 [3H1KAlGlu 14/14 Frontal cortex (SA 11) i; frontal cortex (SA 10), temporal cortex (SA 21 ,22,38) hippocampus, amygdala H

Kerwin et al,l54] 1990 [3H1KAlGlu (ARG) 7/8 CA3, CA4, dentate gyrus, parahippocampal gyrus, left CA 1, left CA2 !; right CA 1, right CA2 H

AMPA Kerwin et al.[54] 1990 [3H1CNQx/Glu (ARG) 7/8 CA4, left CA3 !; CA 1, CA2, right CA3, dentate gyrus,

parahippocampal gyrus H

Kurumaji et a1.155] 1992 [3H1AMPAlGlu 13110 Frontal cortex, temporal cortex, parietal cortex, occipital cortex, limbic cortex H

Freed et al.[56] 1993 [3H1AMPAlGlu 12115 Frontal cortex, caudate nucleus, nucleus accumbens H

N-Methyl-D-aspartate (NMDA)

NMDA recognition site

Kerwin et al. [54] 1990 [3H1Glu/NMDA (ARG) 7/8 Hippocampus (CA1-4, dentate gyrus, parahippocampal gyrus) H

Ion channel

Kornhuber et al. [57] 1989 [3Hldizocilpine 13112 Putamen i; frontal cortex, area entorhinalis, hippocampus H

(MK-801 )/dizocilpine

Suga et al. [58] 1990 [3Hldizocilpine/ 12110 Temporal cortex (SA 22+38+41+42+52), parietal cortex (SA dizocilpine 40,5+7+31) i; frontal cortex, temporal cortex (SA 20+21 ,36),

parietal cortex (SA 39,1+2+3+5+43), occipital cortex H

Simpson et al.[59] 1992 [3H1TCP/ketamine 13114 Frontal cortex (SA 11) i; frontal cortex (SA 10), temporal cortex (SA 38), amygdala H

Glycine binding site

Ishimaru et al.[60] 1994 [3Hlglycine/glycine 13110 Parietal cortex (SA 39,40,1+2+3+5+43), occipital cortex (SA 17,18+ 19), frontal cortex (SA 6) i; frontal cortex (except for SA 6), temporal cortex, parietal cortex (SA 5+7+31) H

Abbreviations and symbols: AMPA = a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; ARG = autoradiography; SA = Srodmann's area; CNQX = 6-cyano-7-nitroquinoxaline-2,3-dione; Glu = glutamate; TCP = N-[1-(2thienyl)cyclohexyllpiperidine; i indicates an increase; ! indicates a decrease; H indicates no change; + indicates that samples from these areas were analysed together.

© Adls International Limited, All rights reserved, eNS Drugs 1997 Jan; 7 (1)

Page 6: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

52

served with competitive NMDA receptor antago­nists, such as 3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPp),[44J 3-(2-carboxy­pi perazin -4-y 1 )-1-prop en y 1-1-phosphonic acid (CPPene),l45J and selfotel (CGS 19755).l46J

Taken together, it is concluded that blockade of NMDA receptors at any level leads to a psychotic state that closely resembles schizophrenia. Based on the finding, NMDA receptor antagonists have been employed in an attempt to reproduce the behavioural or pathological changes of schizophre­nia in animal models.[47-49)

1 .4 Glutamate Receptors in Schizophrenic Brains

Radiolabelled receptor assays and related stud­ies performed in the postmortem brains from pa­tients who had had schizophrenia have provided another line of evidence that suggests that a glutamatergic abnormality may be involved in the illness (tables I and II).

1.4. 1 Non-NMDA Receptors An increase in [3H]kainate binding in the pre­

frontal cortex of schizophrenic patients compared with unaffected individuals was reported by 2 sep­arate groups, indicating an increase in the number

Ishimaru & Toru

of these non-NMDA receptors.l50,53) [3H]Kainate binding was also increased in the angular cortex, which is a part ofthe parietal association field (see fig. 2).[52J In contrast, the number of AMPA recep­tors was not changed in the brain areas exam­ined.[55.56]

Kerwin and his colleagues investigated the hip­pocampus, and observed a significant decrease in the binding density ofkainate[51] and AMPArecep­tors,[541 as well as a decrease in GluRI[61,62] and GluR2[62] mRNA that codes for the AMPA/kainate receptor protein. Their findings are interesting in light of the hippocampal abnormality suggested in schizophrenia.165,66] However, studies from other laboratories have not supported the finding of a decrease in hippocampal kainate receptors in terms of [3H]kainate binding[53) or immunoreactivity of the AMPA/kainate receptor subtypes.l63]

1.4.2 NMDA Receptors

NMDA receptors were investigated using vari­ous ligands.

The binding of [3H]dizocilpine (a ligand that la­bels the ion channel of the NMDA receptor) was reported to increase in the putamen[571 and in 3 cor­tical areas that included the parieto-temporal asso­ciation fields[581 in brains from schizophrenic pa-

Table II. Glutamate receptor subunits in the brains of patients with schizophrenia, assessed at postmortem using gene expression and immunochemical assays

Reference

AMPAlkainate Harrison et al.[61J

Eastwood et al. [62J

Breese et al[63J

Year Method Gene probe or antibody No. of patients Results (patients/controls)

1991 ISH Oligonucleotide probe for GluR1 subunit

1995 ISH Oligonucleotide probes for GluR1 and GluR2 subunits

1995 WB Specific antibodies for GluR1, GluR2, GluR3, and GluR5-7 subunits

6/8

14/9

1219

CA3 -1.; CA1, CA4, DG, SUB H

CA3, CA4, DG, SUB (GluR1) -1.; CA3, CA4, DG, SUB, PHG (GluR2) -1.; CA1, PHG (GluR1), CA1 (GluR2) H

Cingulate cortex, hippocampus H

N-Methyl-D-aspartate Akbarian et al.164J 1996 ISH cDNA or its segments for

NR1 and NR2A-D 15/15 NR2C in prefrontal cortex (BA 10) -1.; parieto-temporal

cortex (BA 39) H; l' in NR2D relative to total NR2 in subunits prefrontal cortex (BA 10)

Abbreviations and symbols: AMPA = a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; BA = Brodmann's area; cDNA = complementary DNA; DG = dentate gyrus; ISH = in situ hybridization; PHG = parahippocampal gyrus; SUB = subiculum; WB = Western blot; -1. indicates a decrease; l' indicates an increase; H indicates no change.

© Adis Internationallirnited. All rights reserved. eNS Drugs 1997 Jan; 7 (1)

Page 7: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

Prefrontal association field

('H)Kalnate Medial Iron tal cortex (9+ 1 0+46) ;01

Eye-movement cortex (S) -'()I

Orbital cortex (11 )'511

( lH)TCP Orbital cortex (11) ""I

PH]Glyclne Premotor cortex (6) 1,5

(3) .(1).(2)

Parietal-temporal-occlpital association field

p H)Kalnate Angular cortex (39) ,

('H)Dlzocllplne Supenor panetal cortex (5+ 7+31) 163

Supramarginal cortex (40) 1b.

Supenor temporal cortex (22+3S+41 +42+52) 1631

( IH]GIYClne Angular cortex (39) i6S

Supramarginal cortex (40) 165

Somesthetlc cortex (1 +2+3+5+43) lbS

OCCipital cortex (17+1S+19) 651

(17)

53

Fig. 2. Changes in glutamate receptors in the frontal and parietal-temporal-occipital association fields and related areas that have been found in patients with schizophrenia. The ligand for each receptor is shown (see fig . 1 for relationship between ligand and receptor) . Numbers in parentheses are the Srodmann's area (SA) numbers. The upregulation of glutamate receptor subtypes was observed in the areas of cerebral cortex shaded blue. In the areas shaded grey, a particularly marked effect was seen, either more than 1 receptor subtype was reported to increase (orbital cortex SA 11, angular cortex SA 39) or an extremely large increase was observed (90 to 110% in angular cortex SA 39 and supramarginal cortex SA 40). The prefrontal association field is thought to be involved in executive tasks such as motivation, planning and socialisation. Frontal lobe dysfunction is currently suggested as a cause of the negative symptoms of schizophrenia. In contrast, the parietal-temporal-occipital association field is an area of extensive polymodal convergence, where higher sensory functions are performed. Disarrangement in such an integrative function may underlie the positive symptoms of schizophrenia, such as auditory hallucinations, reality distortion, thought disorder and cognitive deficits. Abbreviation and symbol: TCP = N-[1-(2thienyl)cyclohexyllpiperidine; + indicates that samples from these areas were analysed together.

tients (fig. 2). [3H]TCP binding, which also labels the NMDA ion channel, was increased in the pre­frontal cortex (fig. 2),[59] where an increase in [3H]kainate binding had been observed (see sec­tion l.4.1))50,53] Strychnine-insensitive [3H]gly­cine binding associated with the NMDA receptor was

© Adis International Umited. All rights reserved .

significantly increased in 6 of the 16 cortical areas examined (fig. 2).160] This increase was more pro­nounced and widespread than that of [3H]dizocilpine binding observed in the same brain series.l58]

A recent study focusing on gene expression for the NMDA receptor in prefrontal and parieto-

eNS Drugs 1997 Jan; 7 (1)

Page 8: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

54

temporal cortices reported a significant increase in the NR2D subunits relative to the whole NR2 sub­unit family in the prefrontal cortex.[64] More infor­mation from gene expression studies will be forth­coming in the near future.

An increase in glutamate receptors in the cere­bral cortex has been repeatedly observed in the schizophrenic brain (table I, fig. 2) How can this be explained given the glutamatergic hypoactivity that underlies schizophrenia? Reviewing the stud­ies performed in the same brain series, Toru et aU52] demonstrated a significant negative correla­tion between glutamate levels in several subcorti­cal structures and glutamate receptor binding in cortical areas receiving projections from these sub­cortical structures. Similar correlations were also revealed between glutamate levels and glycine binding in the parieto-temporal association field.[60] This suggests that the glutamate receptors might be up-regulated in schizophrenic brains in response to a deficiency in glutamatergic neu­rons.[52] Such a possibility is supported by the ob­servation that glutamate receptors increase in rat hippocampus following electrolytic lesions of the entorhinal cortex[67,68] and removal offimbria-for­nix,l69] both of which may result in denervation of glutamatergic fibres that project to the hippocam­pus.

Results from glutamate receptor studies also indicate that glutamatergic dysfunction may be present in 2 important association fields, prefrontal and parietal-temporal-occipital (see fig. 2). Dys­function of the frontal lobe is currently associated with negative symptoms of schizophreniaPO,71] In contrast, dysfunction in the parietal-temporal­occipital association field may underlie the patho­genesis of positive symptoms or cognitive deficits, since this brain area is involved in integrative pro­cessing of extensive sensory inputs (fig.2).l72,73] Therapeutic intervention that restores glutamat­ergic neural transmission may help in correcting the dysfunction assumed in these association fields.

© Adis International Umlted. All rights reserved.

Ishimaru & Toru

1.5 Interaction of Glutamatergic and Dopaminergic Systems

The glutamate hypothesis of schizophrenia as summarised in sections 1.2 to 1.4 is not inconsis­tent with the dopamine hypothesis of the illness (see section 1.1). Abundant interactions between dopaminergic and glutamatergic systems are known to occur in the regions of the brain that con­tain dopaminergic cell bodies[74] and nerve ter­minalsP5,76] Therefore, the glutamatergic hypo­activity that is hypothesised to be the basis of phencyclidine-induced psychosis and schizophre­nia may cause psychotic symptoms via dopamin­ergic modulation. Alternatively, dopaminergic hy­peractivity may induce secondary glutamatergic hypoactivity in broad cortical areas, which may be psychotogenic.

Interestingly, systemic administration of phen­cyclidine to rats increases dopamine turnover in mesocortical and meso limbic terminal areas, while it decreases that in the striatum.[77] This contrast favours phencyclidine-induced psychosis as a model for schizophrenia, since mesocorticall limbic dopaminergic hyperactivity has been as­sumed to underlie schizophrenic symptomatol­ogyP8,79] Furthermore, in the prefrontal cortex, a putative target of antipsychotics,[78,80] NMDA re­ceptors mediate a tonic inhibitory regulation of dopaminergic transmission)81.82] Thus, phencycli­dine may cause schizophrenia-like symptoms by disinhibiting dopaminergic neurons in the prefron­tal cortex. Up-regulation in glutamate recep­tors[50,53] and a decrease in glutamate level[22] ob­served in the prefrontal cortex of schizophrenic brains may reflect such an impairment in gluta­matergic regulation of dopaminergic activity in the area. Since prefrontal glutamatergic neurons pro­ject to the striatum and limbic forebrain areas,[83,84] dysfunction in these neurons may be causative of the subcortical dopaminergic hyperactivity sug­gested in schizophrenic brains)85-87]

However, the implications of the glutamate hypothesis extend beyond a mere modulation of the dopaminergic system. The antipsychotic­resistant symptoms observed in phencyclidine-

eNS Drugs 1997 Jan; 7 (1)

Page 9: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

induced psychosis and schizophrenia are not ex­plained by dopaminergic hyperactivity, which may occur secondary (or be primary) to glutamatergic hypoactivity.!33] Similarly, alterations in glutamate receptors have been demonstrated in various areas of the cerebral cortex, beyond those that receive dopaminergic innervation (table I, fig. 2). Thus, an interaction with the dopaminergic system may be a part of the psychotogenic mechanism of glutamatergic hypoactivity, but not all of it. This suggestion that glutamatergic dysfunction may be independent, at least in part, of dopaminergic hyperactivity may promote a better understanding of schizophrenia. The glutamate hypothesis can incorporate a dysfunction of more global neural circuits, including the dopaminergic sys­tem.l13,49,88,89] In this way, it may serve as a reason­able starting point for developing refined hypo­theses of schizophrenia, as well as for planning advanced therapeutic strategies for the illness.

2. Therapeutic Implications of the Glutamate Hypothesis

2.1 Antipsychotics and the Glutamatergic System

The glutamate hypothesis of schizophrenia im­plies that schizophrenic symptoms should be ameli­orated by administration of an agent that restores glutamatergic activity in the brain. However, typi­cal anti psychotics do not primarily have an action on the glutamatergic system, although some indi­rect effects suggested in vivo, such as a reversal of impaired glutamate releasel20,22] and upregulation of the NMDA receptor-associated ion channel,l90] may contribute to the therapeutic effect of these drugs.

On the other hand, it is tempting to postulate that atypical anti psychotics such as clozapine may affect the glutamatergic system, since clozapine is effective in ameliorating symptoms that are resistant to typical antipsychotics.l91.93] In fact, ac­cumulating evidence has shown that clozapine ant­agonises behavioural effectsl94.99] and neurotoxi­city[IOO] induced by NMDA antagonists. Repeated

© Adis International limited. All rights reserved.

55

administration of clozapine to animals increased dizocilpine-induced hyperlocomotion and, in par­allel, induced an up-regUlation of phencyclidine receptors in the frontal cortex. llOl] Regional distri­bution of specific [3H]clozapine binding was re­ported to match glutamatergic innervationl102] and clozapine increased glutamate levels in the rat me­dial prefrontal cortex.[I03]

Despite all these findings, the data are inconclu­sive with regard to the way in which clozapine interacts with NMDA receptors. For example, clozapine inhibited [3H]dizocilpine binding in some studies,l 102· 104] while it did not inhibit eH]TCP binding in another study.llOl] Current studies are largely focusing on the possible in­volvement of multiple receptor systems in the ac­tion of clozapine.lI05-107]

Thus, novel agents that augment glutamatergic neural transmission need to be developed in order to derive advanced treatments from the glutamate hypothesis.

2.2 Therapeutic Target Sites on the NMDA Receptor

In light of the finding that antagonism of the NMDA receptor induces psychotic symptoms, this receptor subtype seems to be a promising target of therapeutic attempts for schizophrenia, although alterations in kainate receptors found in postmor­tem brains suggest that non-NMDA receptors may also play some role in the illness (table I).

The NMDA receptor is a complicated molecule containing several modulatory sites (see fig. 1):

• NMDA binding site • zinc ion (Zn++) modulatory site • magnesium ion (Mg++) modulatory site and

phencyclidine receptor (both of which are loca­ted in the ionophore)

• polyamine modulatory site • strychnine-insensitive glycine binding site.

2.2.1 Glutamate/NMDA Binding Site

The recognition site for glutamate or NMDA is obviously not a useful target since agonists at this site are known to be potent neurotoxins.l12,108]

eNS Drugs 1997 Jan: 7 (1)

Page 10: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

56

2.2.2 Polyamine Modulatory Site The difficulty is that the activity of the NMDA

receptor needs to be augmented to a level that cor­rects glutamatergic hypofunction, but that avoids excitotoxicity. Polyamines, which constitute a di­verse family of endogenous molecules,uo9) may in­clude compounds that show moderate augmenta­tion of glutamatergic activity. In fact, the polyamine site agonists spermine and spermidine enhance NMDA receptor function under certain conditions. [110) Interestingly, both these molecules were reported to inhibit amphetamine-induced dopaminergic hyperactivity in the mesolimbic pathway, but not in the striatum'! II 1) Further under­standing of polyamine molecules and their action on the NMDA receptor is required.

2.2.3 Phencyclidine Receptor A compound that prevents phencyclidine from

blocking the NMDA receptor-associated ion chan­nel may be another interesting candidate for a ther­apeutic agent, especially if glutamatergic hypoactivity is proven to be caused by endogenous phencyclidine-like ligands (see section 3.1 ).[112,113) Metaphit, a phencyclidine receptor acylator, was reported to antagonise phencyclidine-induced ab­normal behaviours in the rat,11l4) but this com­pound is not suitable for clinical use because it has phencyclidine-like behavioural effects of its own.11l5) Development of safer phencyclidine an­tagonists as well as investigation of modulation at the Zn++ and Mg++ sites might be pursued in the future.

2.2.4 Glycine Binding Site At present, the most promising target for cor­

recting glutamatergic hypoactivity seems to be the glycine binding site that is associated with the NMDA receptor.

Glycine dramatically enhances the response of the NMDA receptor in a strychnine-insensitive manner.11l6) This effect has been observed in all of the available assays for NMDAreceptor-mediated functions)117) Activation of the glycine binding site is an absolute requirement for NMDA receptor function;11l8,1l9) therefore, glycine is considered to be a co-agonist at the receptor/ion channel com-

© Adis International Limited. All rights reserved.

Ishimaru & Toru

plex.!1l7) Even so, glycine may augment NMDA receptor function only moderately,(120) since it acts in a manner that increases the channel opening fre­quency in the presence of competitive NMDA ag­onists.!121) The remarkable increase in the glycine binding sites demonstrated in postmortem brain as­says (see section 1.4.2) suggests that this site may play a crucial role in the process of schizophre­nia)60)

However, there is a major problem with the ad­ministration of glycine as a therapy - it does not readily cross the blood-brain barrier, since no high­affinity transport system across the barrier exists for this hydrophilic molecule. (122) Nevertheless, it was shown that brain glycine levels could be in­creased in the rat brain following an intra-arterial injection of glycine(123) or feeding with a liquid diet containing high levels of glycine.[l24) In humans, administration of glycine was not associated with any significant adverse effects at dosages up to 3 gikg(125) or at 60g per day.(126)

Moreover, intragastric administration of glycine antagonised abnormal behaviours induced by phencyclidine in animals)127) This finding, which was supported by more recent studies in which gly­cine agonists were administered through in­tracerebroventricular injection,u28-130) promoted clinical trials involving glycinergic therapy for schizophrenia (table III).

2.3 Glycinergic Therapies for Schizophrenia

2.3. 1 Glycine The first study of glycinergic therapy for schizo­

phrenia was performed by Waziri,ll31) The impetus for this study was the finding that the activity of serine hydroxymethyltransferase (SHMT) was sig­nificantly decreased, while plasma serine levels were increased, in psychotic patients including those with schizophrenia.!151,152) Since the conver­sion of serine by SHMT is the major source of gly­cine in the brain,(153) a deficiency of this enzyme might result in a shortage of glycine. Therefore, glycine (5 to 25 g/day) was administered as an ad­juvant to 11 patients with chronic schizophrenia who had been receiving continuous antipsychotic

eNS Drugs 1997 Jan; 7 (1)

Page 11: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia 57

Table III. Summary of clinical studies that have assessed glycinergic activation in patients with schizophrenia

Reference Year Type of Dosage Study No. of Results study (g/day) duration patients psychotic symptoms extrapyramidal symptoms

(wk) (drug/placebo) rating scale effect" rating scale effect

Glycine Waziri[131] 1988 A then R 5-25 >8mo 11 NA Improved (4111) NA NS Rosse et al.[132] 1989 A 10.8 4 days-8 5M SPRS, CGI, Improved (2/5), AIMS, Improved

SANS aggravated (2/5) SARS (3/5) Costa et al.[133] 1990 A 15 5 6M SPRS Improved (2/6) NA NS Potkin et al.[134] 1992 A 15 6 9M+2F/6M+1F BPRS, CGI, Improved on CGI SARS Unchanged

SANS Javitt et al. [135] 1994 A 2-30 8 7M17M PANSS Improvement in AIMS, ERS Unchanged

negative symptoms

Milacemide Rosse et al.[136] 1990 R 1.2 4 days-4 5M BPRS, CGI, Aggravated (3/5) SARS Unchanged

SANS, WCST Rosse et al. [137] 1991 R 0.4 7 days-4 4M BPRS, CGI, Aggravated (2/4) SARS NS

SANS, WCST

Tammingaet 1992 R 1.2 6 3 (co) SPRS Unchanged NA NS a1.1138]

D-Cycloserine Cascellaet 1994 A 0.25 6 3M+4F SPRS, CGI, Aggravated (417) NA Unchanged al.[139] SANS Goff et al.[140] 1995 A 0.05-0.25 2 8M+1F BPRS, GAS, Improvement in AIMS, Unchanged

SANS, SIRP negative SARS symptoms (SANS) and reaction time (SIRP)

a Where the number of patients in the effect column does not equal the total number of patients, the remainder were unchanged.

Abbreviations: A = adjuvant; AIMS = Abnormal Involuntary Movement Scale[141]; BPRS = Brief Psychiatry Rating Scale[142]; CGI = Clinical Globallmpression[143]; co = crossover; ERS = Extrapyramidal Rating Scale[144]; F = female; GAS = Global Assessment Scale[145]; M = male; NA= not applicable (no scale used); NS = not specified; PANSS = Positive and Negative Symptom ScaleI146]; R = replacement; SANS = Scale for Assessment of Negative Symptoms[147]; SARS = Simpson-Angus Rating Scale for Extrapyramidal Symptoms[148]; SIRP = Sternberg's Item Recognition ParadigmI149]; WCST = Wisconsin Card Sorting Test11501.

medication and were socially handicapped.D3l) The treatment was successful in 4 patients who showed 'a definite salutary response', allowing the dosages of their antipsychotics to be reduced or even discontinued and the patients to become in­volved in rehabilitation.[13I) This effect lasted for several months.£13l)

These observations opened the way for further studies, although a deficiency in SHMT was not supported by other studies.£154,155) Waziri et al.D 56) later suggested that a deficiency in SHMT might lead to a more complicating metabolic abnormal­ity, in which levels of glycine as well as serine might be elevated.

© Adls International Umited. All rights reserved.

In another open label study by Rosse et al.,[132) glycine was given to 6 patients at a fixed dosage (10.8 g/day) as an adjunct to antipsychotics, and the effect was evaluated by standardised rating scales (see table III.) During the glycine adjuvant period, 2 patients showed improvement whereas 2 others worsened. Another patient showed no clin­ical response to adjuvant glycine. The remaining patient reported marked subjective improvement at the beginning of the glycine phase, but left hospital before formal ratings could be performed. In I of the 2 responders, deterioration was observed after glycine withdrawal, but a subsequent rechallenge with glycine failed to result in a persistent effect.

eNS Drugs 1997 Jan; 7 (1)

Page 12: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

58

The other responder showed no worsening after glycine discontinuation. Interestingly, a remark­able improvement was observed in extrapyramidal symptoms in 3 of the patients.[l32]

Costa et aUl33] also administered glycine (15 g/day) as an adjunctto 6 patients with schizophre­nia who had been maintained on relatively stable dosages of antipsychotics. Two patients showed re­markable improvement in Brief Psychiatric Rating Scale (BPRS) scores and none of the patients showed significant aggravation of symptoms or major adverse effects. The authors conducted an­other study involving a larger number of patients in a double-blind manner and observed a slight im­provement in symptoms, as reflected in Clinical Global Impression scores but not in the other rating scales employed.[l34] The changes in BPRS showed improvement especially in the score of hostility-suspiciousness, but this did not reach sta­tistical significance.l l34]

More recently, Javitt et aUl35] administered gly­cine to patients with schizophrenia in a double­blind, placebo-controlled study where changes were evaluated according to the Positive and Nega­tive Syndrome Scale. A significant decrease in neg­ative symptom scores was observed after the gly­cine administration period without exacerbation of positive symptoms. In contrast to the previous re­port,[l32] the patients showed no significant change in Extrapyramidal Rating Scale scores, suggesting that the improvement in negative symptoms did not result from an amelioration in antipsychotic­induced extrapyramidal symptoms.[l35]

Despite some minor inconsistencies, these stud­ies suggest that glycine administration may be a valuable adjunctive tool in the treatment of schizo­phrenia.

2.3.2 Milacemide Because the relative impermeability of the

blood-brain barrier to glycine seemed to be the ma­jor difficulty for a glycinergic activation strat­egy,[l32] 2 orally active compounds, milacemide and D-cycloserine, were assessed in clinical stud­Ies.

© Adis International Limited. All rights reseNed.

Ishimaru & Toru

Milacemide is a glycine prodrug that readily crosses the blood-brain barrier and is converted by monoamine oxidase (MAO)-B into glycinamide and glycine)l57] This drug was administered either at a high (1200 mg/day)[l36] or low (400 mg/day) dosage[l37] to patients with chronic schizophrenia. However, clinical worsening was observed in almost all the patients during the milacemide administration period in both studies, which re­sulted in withdrawal of half of them from treatment (3 of 5 and 2 of 4 patients, respectively; see table III).[l36.l37] On the other hand, Tamminga et aUl38] also administered milacemide (1200 mg/day) in a small, placebo-control crossover study and ob­served no improvement or deterioration on BPRS scores in schizophrenic patients.

It should be noted that in these studies antipsy­chotics were replaced by milacemide, while in the other studies, glycine or D-cycloserine were ad­ministered as an adjunct to antipsychotics (see ta­ble III). In replacement studies, it is particularly important to design an appropriate scheduled for the cessation and wash-out of antipsychotics that had been administered to the patients involved. Rosse et aUl36.137] employed a 4-day medication­free period before administrating milacemide, whereas in the study by Tamminga et al.,[138] there was a wash-out period of at least 4 weeks until the psychotic symptoms of the patients reached a sta­ble baseline. The difference in these wash-out pe­riods may explain the inconsistency between their results, since in the former studies cessation of an­tipsychotics might have resulted in exacerbation of schizophrenic symptoms during the period when milacemide was being administered.

The failure of milacemide to ameliorate schizo­phrenic symptoms may not necessarily argue against the validity of glycinergic intervention. This is because milacemide was shown to be an inhibitor as well as a substrate of MAO_B[l58.l59] and also an inhibitorofMAO-A,[159] both of which are the enzymes that convert dopamine to 3,4 dihydroxyphenylacetic acid (DOPAC). It is likely that inhibition of these enzymes led to an accumu­lation of dopamine,[l60] to the extent that it may

eNS Drugs 1997 Jan; 7 (1)

Page 13: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

have negated the beneficial effect of glycine. Milacemide also potentiated the anticonvulsant ef­fect of dizocilpine in such a way that the potentia­tion was not blocked by a selective antagonist of glycine sites,l l61 1 suggesting that the predominant effect of milacemide in vivo is ascribed to a mech­anism other than glycinergic activation.

2.3.3 D-Cycloserine

The long term administration of D-cycloserine in the treatment of tuberculosis is frequently asso­ciated with a high incidence of neuropsychiatric abnormalities.ll621 Interestingly, it has also been administered to psychotic patients at high dosages (1.0 to 2.0 g/day), which resulted in exacerbation of psychotic symptoms with motor excitement fol­lowed by an improved response to subsequent an­tipsychotic treatment ('symptom provocative' therapylI621). D-Cycloserine has been identified as a partial agonist at the glycine site,[163, 1641 and so has recently been utilised for the treatment of schizophrenia at moderate dosages.

However, Cascella et aLi l391 administered the drug at a dosage of 250 mg/day for 6 weeks to 7 patients and observed a deterioration in 4 of the patients, with intensified positive symptoms and emotional agitation. The authors suggested that this might be due to a biphasic action of D-cyclo­serine.[ 1391 As a partial agonist, the drug has only limited activity and could antagonise the action of an endogenous agonist at the glycine site when given at high dosages.l1 65,1661 Therefore, they sug­gested there might exist a therapeutic 'window' for cycloserine,[1391

Indeed, this window was observed in a later study - Goff et aLl l401 reported that patients given a placebo and 4 dosages of D-cycloserine (5, 15, 50 and 250 mg/day) in a consecutive schedule showed a significant reduction in Schedule for As­sessment of Negative Symptoms scores as well as in reaction time according to Sternberg's Item Rec­ognition Paradigm at 50 mg/day, while at 250 mg/day the scores were reversed to the level ob­served at 0 to 15 mg/day. This indicates that the window may occur around 50 mg/day, although the effect of drug accumulation should be taken into

© Adis International Limited. All rights reserved.

59

account. Another consideration is that the patients in these 2 studies might have differed in the sever­ity of their illness; the former study used inpatients with moderate to severe symptomatology,l'391 while the latter used outpatients who had been managed on a stable medication regimen much longer than those in the former study.ll401 This may partly explain the ineffectiveness of D-cycloserine observed in the former study.

2.3.4 Other Comments on Findings

Although most of the studies reviewed above were preliminary ones conducted with differing experimental designs, the results include some in­teresting findings (table III). Administration of glycine as an adjunct to antipsychotics was effec­tive in some patients. The effect was moderate in general, but the drug rarely aggravated symptoms. Glycine did not exacerbate extrapyramidal symp­toms and sometimes ameliorated them.

It is interesting that improvement in negative symptoms was observed in recent studies follow­ing administration of glycine[1351 or D-cycloser­ine,l 1401 Earlier studies also included several cases in which negative symptoms were ameliorated by glycine.l l3l ,132] In contrast, no quantified improve­ment in positive symptoms has been reported so far. However, it should be noted that in these stud­ies glycine and D-cycloserine were given as adju­vants to antipsychotics which had been adminis­tered for years. The positive symptoms that had responded to antipsychotic treatment might also have been ameliorated by glycine site activation. In this respect, it is interesting that, in the first study by Waziri,[131] 4 of the patients who re­sponded to glycine could be maintained on reduced doses of anti psychotics or even have the drugs dis­continued for several months. However, the reli­ability of this finding is still not clear, since Costa et aLl133] reported that the administration of gly­cine without concomitant anti psychotics had no antipsychotic effect in patients with acute schizo­phrenia. More studies are required to clarify whether glycine can successfully replace anti­psychotics in controlling positive symptoms.l 1351

eNS Drugs 1997 Jan; 7 (l)

Page 14: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

60

It also remains to be clarified whether the mod­erate effects of glycine were due to its modulatory action on dopaminergic pathways, or the result of correcting the primary deficit in glutamatergic neu­ral transmission.

Since milacemide is not an effective source of glycine and D-cycloserine is only a partial agonist at the glycine site, a full glycine site agonist that readily crosses the blood-brain barrier is required. Such an agent will provide valuable information pertaining to the validity of glycine site activation therapy as well as the glutamate hypothesis of schizophrenia. In future studies, special attention should be paid to: (i) the administration schedule of the drugs; (ii) employment of a placebo-control design; (iii) methods for evaluating the symptom­atology; and, especially, (iv) the clinical condition of patients such as sUbtypes of the illness and se­verity of the symptoms. Considering the wide va­riety of symptomatology and aetiology subsumed under a diagnosis of schizophrenia,[167] one might question the validity of evaluating drug effects in such a heterogenous population.

3. Some Interesting Questions

3.1 What is the Basis of Glutamatergic Hypoactivity?

As a model for schizophrenia, phencyclidine­psychosis allows at least 2 possible mechanisms that may cause glutamatergic hypoactivity: (i) a de­ficiency in glutamatergic neurons caused by either presynaptic or postsynaptic abnormalities; or (ii) the presence of an endogenous ligand that acts at phencyclidine receptors to block the function of NMDA receptors. Theoretically, these 2 assump­tions may lead to totally different predictions of the consequence of glycine site activation. If gluta­matergic hypoactivity is due to a deficiency in glutamatergic neurons, then exogenous glycine would be effective in activating the receptors. If, by contrast, a phencyclidine-like endogenous ligand is involved in schizophrenia, further activa­tion of NMDA receptors by exogenous glycine

© Adis Internafional limited. All rights reserved.

Ishimaru & Toru

might be ineffective, as the phencyclidine-like ligands would readily block the open channels.

At present, there is no definite answer to the question. A deficiency in glutamatergic neurons is suggested by several findings: a reduction in glu­tamate release from synaptosomes obtained from schizophrenic brains,[2o.21] a reduction in gluta­mate levels in the brains of patients with schizo­phrenia,[22] a negative correlation between gluta­mate levels and the number of its receptors, [52] and alterations in non-NMDA receptors[50-54] that are not likely to be caused by an endogen...ous NMDA antagonist.

On the other hand, the existence of a phen­cyclidine-like ligand has also been reported in an­imals,£i 12,113] suggesting that glycinergic activation may not be helpful in patients with schizophrenia. In fact, it was reported that the stereotypes induced by dizocilpine in rats were not counteracted by a glycine agonist (D-cycloserine) and were even aug­mented.[168] This led Kretschmer and Schmidt[l69] to doubt the usefulness of glycine administration to schizophrenic patients, a view that is supported by the 'negative findings' of clinical studies. Carlsson et alJ l70] raised a similar question based on their finding that D-cycloserine potentiated, while (+)­HA-966 counteracted, the locomotor stimulation induced by an NMDA antagonist (either dizocilp­ine and CPPene) coadministered with clonidine in monoamine-depleted mice. They argued, as had been suggested by another group,[I7I] that (+)-HA-966, an antagonistic partial agonist of the glycine site, might be effective in treating schizophrenia, whereas D-cycloserine might not be.£170]

However, these results should be carefully inter­preted, since the effect of D-cycloserine, a partial agonist of the glycine site, could be biphasic de­pending on the dosage.D 65,166] This is particularly important if modulation is attempted at the glycine site in vivo. When competing with an endogenous full agonist, D-cycloserine may readily act as an antagonist,[166] even though it has a relatively high intrinsic activity.£164] In fact, there are several stud­ies reporting that abnormal behaviours induced by phencyclidine or dizocilpine were successfully an-

eNS Drugs 1997 Jan: 7 (1)

Page 15: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

tagonised by glycine[l27] or by full agonists of the glycine site, such as D_serine[l28,130] and D-ala­nine)129,130] The facilitating effect of phencyclid­ine on dopamine utilisation in the medial frontal cortex was also inhibited by intracerebroventricu­lar injection of D-alanine,D72]

The precise mechanism of these effects remains unclear, An in vitro study suggested that glycine site agonists counteracted the inhibitory effect of phencyclidine on the NMDA-mediated signal transduction, but probably by some indirect mech­anism at a physiologicalleveUl73] In addition, the effect of a glycine site ligand in vivo seems to be far more complex than is predicted by the data ob­tained in vitro, since the NMDA receptor mediates either enhancement or inhibition of dopaminergic neural transmission depending on the brain area in which it is located)77,171,174]

Whatever the mechanism is, these findings sug­gest that glycine agonists may be beneficial to pa­tients with schizophrenia even if glutamatergic hypofunction is due to a phencyclidine-like endog­enous ligand.

3,2 Is the Glycine Site Saturated In Vivo?

The CSF glycine level is known to be as high as 1 ~mol/L and above in both healthy individuals and patients with schizophrenia.[16] This suggests that glycine levels in vivo might be high enough for glycine to occupy all the glycine sites)1l6] If this is the case, administration of exogenous glycine ag­onists would have no effect, a hypothesis that would dampen enthusiasm for the glycinergic ac­tivation therapy.

Early studies reported that responses mediated by NMDA receptors in brain slices are not en­hanced by glycine or D-serine, but could be blocked by glycine antagonists such as kynurenate, 7-chlorokynurenic acid and (+)-HA-966)17S] This favoured the hypothesis that the glycine site is sat­urated in vivo.

However, under in vitro conditions that closely mimicked those observed in vivo, NMDA recep­tor-mediated synaptic potentials were enhanced by glycine in neocortical slices)176] This finding

© Adis International Limited. All rights reserved.

61

was followed by studies in vivo that showed a fa­cilitation ofNMDAresponses by local application of glycine or D-serine.[l77-179] Also, in cerebellar granule cells, where the high affinity glycine trans­porter is abundant, the NMDA receptor-mediated synaptic currents were absent unless glycine was supplied in the perfused medium) I 80] These obser­vations argue that the glycine sites in vivo stay unsaturated, and provide a rationale for the admin­istration of exogenous glycine agonists for activat­ing NMDA receptors)181]

Recently, brain-specific glycine transporters were cloned in rodents and the co-localisation of these transporters with NMDA receptors was ob­served)182-184] This suggests that the glycine level in the synaptic cleft may be precisely regulated by these transporter molecules. Kynurenate, an en­dogenous tryptophan metabolite that competi­tively antagonises the glycine site,[18S-187] may also play an important regulatory role)188,189] Modifi­cation of the function of the glycine transporter, as well as that of endogenous antagonist metabolism, may provide an interesting basis for a therapeutic intervention, although more information about these regulatory mechanisms is required,

3,3 Is D-Serine an Endogenous Agonist for the Glycine Site?

D-Serine is a selective agonist at the glycine site on the NMDA receptor, and has a similar potency to glycine)1l8,190,191] Although D-serine has been employed in experimental studies, it was believed that D-enantiomers of amino acids did not natu­rally exist in mammalian tissues,[192] except in small amounts.[193,194] However, recent studies have shown that free D-serine is present in substan­tial quantities in the brain ofrat[l9S] and human)196]

The anatomical distribution of D-serine, as well as its age-related change, was highly correlated with that of the NMDA receptor,[197] Analysis of the cloned NMDA receptor expressed on Xenopus oocytes revealed a lower EDso value (dose of a drug that produces 50% of the maximal response) for D-serine than for glycine, i.e. D-serine is a more potent ligand for the receptor than glycine)198] Fur-

eNS Drugs 1997 Jan: 7 (1)

Page 16: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

62

thermore, D-serine was found to concentrate in the

synaptosomes that included the uptake system for

the molecule.l l971 Therefore, there is a good possi­

bility that D-serine is an endogenous modulator at

the glycine site associated with the NMDA recep­

tor.D 811

Considering that D-serine antagonised NMDA

antagonist-induced abnormal behaviours[128, 1301

and dopaminergic hyperactivity in the prefrontal

cortex,[ 1721 this amino acid may be considered as

another promising candidate for glycinergic acti­

vation therapy, particularly if an orally administra­

ble prodrug for D-serine is developed.

4. Conclusion

In contrast to the dopamine hypothesis, which

was conceived on the basis of the action of anti­

psychotics, the glutamate hypothesis of schizo­

phrenia has been proposed on the basis of patho­

logical findings and a therapeutic approach is

developing from it. Although glutamatergic hypo­

activity in the schizophrenic brain seems compel­

ling, it remains to be demonstrated that schizophre­

nic symptoms are successfully ameliorated by

augmenting glutamatergic neural transmission.

Initial therapeutic attempts have focused on the

glycine binding site of the NMDA receptor. The

results obtained so far are promising, but not con­

vincing. Carefully designed controlled studies us­

ing either orally active glycine agonists or agents

aimed at other targets are required. The results of

these studies may determine whether the glutamate

hypothesis survives as an authentic theory or fades

away after a temporary boom.

Acknowledgements

The authors thank D.E Wozniak (PhD), G. Brosnan­Watters (PhD) and S.c. Yancey (MEd) for reading the manu­script and for making helpful suggestions.

References 1. Deniker P. Introduction of neuroleptic chemotherapy into psy­

chiatry. In: Ayd F, Blackwell B, editors. Discoveries in bio­logical psychiatry. Philadelphia: J.B. Lippincott Company, 1970: 155-64

2. Carlsson A, Lindqvist M. Effect of chlorpromazine or haloper­idol on formation of 3-methoxytyramine and normetanephr-

© Adis International Limited. All rights reserved.

Ishimaru & Toru

ine in mouse brain. Acta Pharmacol Toxicol (Copenh) 1963; 20: 140-4

3. Snyder S. The dopamine hypothesis in schizophrenia: focus on the dopamine receptor. Am J Psychiatry 1976; 133: 197-202

4. Seeman P. Brain dopamine receptors. Pharmacol Rev 1980; 32: 229-313

5. Angrist B, Rotrosen J, Gershon S. Differential effects of am­phetamine and neuroleptics on negative vs. positive symp­toms in schizophrenia. Psychopharmacology 1980; 72: 17-9

6. Seiden L, Sabol K. Amphetamine: effects on catecholamine sys­tems and behavior. Ann Rev Pharmacol Toxicol 1993; 32: 639-77

7. Johnstone E, Crow T, Frith C, et al. Mechanism of the antipsy­chotic effect in the treatment of acute schizophrenia. Lancet 1978; I: 848-51

8. Letemendia F, Harris A. Chlorpromazine and the untreated chronic schizophrenic: a long term trial. Br J Psychiatry 1967; 113: 950-8

9. Watkins J, Evans R. Excitatory amino acid transmitters. Ann Rev Pharmacol Toxicol1981; 21: 165-204

10. Fonnum F. Glutamate: a neurotransmitter in mammalian brain. J Neurochem 1984; 42: I-II

II. Collingridge G, Lester R. Excitatory amino acid receptors in the vertebrate central nervous system. Pharmacol Rev 1989; 40: 143-210

12. Olney J. Excitatory amino acids and neuropsychiatric disorders. BioI Psychiatry 1989; 26: 505-25

13. Toru M, Kurumaji A, Ishimaru M. Excitatory amino acids: im­plications for psychiatric disorders research. Life Sci 1994; 55: 1683-99

14. Kim J, Komhuber H, Schmid-Burgk W, et al. Low cerebrospinal fluid glutamate in schizophrenic patients and a new hypothe­sis on schizophrenia. Neurosci Lett 1980; 20: 379-82

15. Perry T. Normal cerebrospinal fluid and brain glutamate levels in schizophrenia do not support the hypothesis of glutamatergic neuronal dysfunction. Neurosci Lett 1982; 28: 81-5

16. Korpi E, Kaufman C, Mamela K, et al. Cerebrospinal fluid amino acid concentrations in chronic schizophrenia. Psychi­atry Res 1987; 20: 337-45

17. Gattaz W, Gasser T, Beckmann H. Multidimensional analysis of the concentrations of 17 substances in the CSF of schizo­phrenics and controls. Bioi Psychiatry 1985; 20: 360-6

18. Korpi E, Kleinman J, Goodman S, et al. Neurotransmitter amino acids in post-mortem brains of chronic schizophrenic patients. Psychiatry Res 1987; 22: 291-301

19. Stanley J, Drost D, Williamson P, et al. In vivo proton MRS in never treated schizophrenics. In: New research abstracts. Washington, DC: American Psychiatric Association, 1992: 50

20. Sherman A, Davidson A, Baruah S, et al. Evidence of glutamatergic deficiency in schizophrenia. Neurosci Lett 1991; 121: 77-80

21. Sherman A, Hegwood T, Baruah S, et al. Deficient NMDA-me­diated glutamate release from synaptosomes of schizophre­nics. Bioi Psychiatry 1991; 30: 1191-8

22. Tsai G, Passani L, Slusher B, et al. Abnormal excitatory neuro­transmitter metabolism in schizophrenic brains. Arch Gen Psychiatry 1995; 52: 829-36

23. Greifenstein F, DeVault M, Yoshitake J, et al. A study of a 1-arylcyclohexylamine for anaesthesia. Anesth Analg 1958; 37: 283-94

24. Cohen S. Angel dust. JAMA 1977; 238: 515-6

eNS Drugs 1997 Jan; 7 (1)

Page 17: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

25. Luby E, Coben E. Rosenbaum G, et al. Study of a new schizophrenomimetic drug - Sernyl. Arch Neurol Psychiatr 1959; 81: 363-9

26. Luby E, Gottlieb J, Cohen B, et al. Model psychoses and schizo­phrenia. Am J Psychiatry 1962: 119: 61-5

27. Luisada P. The phencyclidine psychosis: phenomenology and treatment. In: Petersen R, Stillman R, editors. Phencyclidine (PCP) abuse: an appraisal. NIDA Research Monograph se­ries; vol 21. Rockville: National Institute on Drug Abuse, Division of Research, 1978: 24 I -53

28. Davies B, Beech H. The effect of I-arylcyclohexylamine (Sernyl) on twelve normal volunteers. J Ment Sci 1960; 106: 912-24

29. Bakker C, Amini F. Observations on the psychotomimetic ef­fects of Sernyl. Compr Psychiatry 196 I; 2: 269-80

30. Allen R, Young S. Phencyclidine-induced psychosis. Am J Psy­chiatry 1978; 135: 1081-4

31. Ban T, Lohrenz J, Lehmann H. Observations on the action of Sernyl - a new psychotropic drug. Can Psychiatr Assoc J 1961; 1961: 150-6

32. Erard R, Luisada P, Peele R. The PCP psychosis: prolonged intoxication or drug-precipitated functional illness? J Psyche­delic Drugs 1980; 12: 235-5 I

33. Javitt D. Negative schizophrenic symptomatology and the PCP (phencyclidine) model of schizophrenia. Hillside J Clin Psy­chiatry 1987; 9: 12-35

34. Johnson K, Jones S. Neuropharmacology of phencyclidine: ba­sic mechanism and therapeutic potential. Ann Rev Pharmacol Toxicol 1990; 30: 707-50

35. Meltzer H, Sturgeon R, Simonovic M, ct al. Phencyclidine as an indirect dopamine agonist. In: Domino E, editor. PCP (phencyclidine) - historical and current perspectives. Ann Ar­bor: NPP Books, 198 I: 207-42

36. Lodge D, Anis N, Burton N. Effects of optical isomers ofketam­ine on excitation of cat and rat spinal neurons by amino acids and acetylcholine. Neurosci Lett 1982; 29: 281-6

37. Anis N, Berry S, Burton N, et al. The dissociative anaesthetics ketamine and phencyclidine selectively reduce excitation of central mammalian neurons by N-methyl-aspartate. Br J Pharmacol 1983; 79: 565-75

38. Willetts J, Balster R, Leander J. The behavioral pharmacology of NMDA receptor antagonists. Trends Pharmacol Sci 1990; I I: 423-8

39. Troupin A, Mendius J, Cheng F, et al. MK-801. In: Meldrum B, Porter R, editors. New anticonvulsant drugs. Current prob­lems in epilepsy; vol 4. London: John Libbey & Company Ltd., 1986: 191-201

40. Fauman M, Fauman B. The psychiatric aspects of chronic phencyclidine use: a study of chronic PCP users. In: Petersen R, Stillman R, editors. Phencyclidine (PCP) abuse: an ap­praisal. NlDA Research Monograph series; vo121. Rockville: National Institute on Drug Abuse, Division of Research, 1978: 183-200

4 I. Krystal J, Karper L, Seibyl J, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Arch Gen Psychiatry 1994; 51: 199-214

42. Haertzen C. Subjective effects of narcotic antagonists cyclazo­cine and nalorphine on the addiction research center inven­tory (ARCI). Psychopharmacology (Berl) 1970; 18: 366-77

43. Javitt D, Zukin S. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 1991; 148: 130 1-8

44. Kristensen J, Svensson Jr B, Gordh T. The NMDA-receptor antagonist CPP abolishes neurogenic 'wind-up pain' after in­trathecal administration in humans. Pain 1992; 51: 249-53

!D Adis Internotionollirnited. All rights reserved.

63

45. Herrling P. D-CPPene (SDZ EAA 494), a competitive NMDA antagonist: results from animal models and first results in humans [abstract]. Neuropsychopharmacology 1994; 10 (3S): 59 IS

46. Grotta 1. Safety and tolerability of the glutamate antagonist CGS 19755 in acute stroke patients [abstract]. Strokc 1994; 25:255

47. Mansbach R, Geyer M. Effects of phencyclidine and phencyclidine biologs on sensorimotor gating in the rat. Neu­ropsychopharmacology 1989; 2: 299-308

48. Carlsson M, Carlsson A. Interactions between glutamatergic and monoaminergic systems within the basal ganglia - impli­cations for schizophrenia and Parkinson's disease. Trends Neurosci 1990; 13: 272-6

49. Olney J, Farber N. Glutamate receptor dysfunction and schizo­phrenia. Arch Gen Psychiatry 1995; 52: 998-1007

SO. Nishikawa T, Takashima M, Toru M. Increased [lHlkainic acid binding in the prefrontal cortex in schizophrenia. Neurosci Lett 1983; 40: 245-50

5 I. Kerwin R, Patel S, Meldrum B, et al. Asymmetrical loss of a glutamate receptor subtype in left hippocampus in postmor­tem schizophrenic brain. Lancet 1988; I: 583-4

52. Toru M, Watanabe S, Shibuya H, et al. Neurotransmitters, recep­tors and neuropeptides in post-mortem brains of chronic schiz­ophrenic patients. Acta Psychiatr Scand 1988; 78: 121-37

53. Deakin J, Slater P, Simpson M. Frontal cortical and left tempo­ral glutamatergic dysfunction in schizophrenia. J Neurochem 1989; 52: 1781-6

54. Kerwin P. Patel S, Meldrum B. Quantitative autoradiographic analysis of glutamate binding sites in the hippocampal forma­tion in normal and schizophrenic brain post mortem. Neurosci 1990; 39: 25-32

55. Kurumaji A, Ishimaru M, Toru M. a-[lHjAmino-3-hydroxy-5-methylisoxazole-4 propionic acid binding to human cerebral cortical membranes: minimal changes in postmortem brains of chronic schizophrenics. J Neurochem 1992; 59: 829-37

56. Freed W, Dillon-Carter 0, Kleinman J. Properties of j'H jAMPA binding in postmortem human brain from psychotic subjects and controls: increases in caudate nucleus associated with suicide. Exp Neurol 1993; 121: 48-56

57. Kornhuber J, Mack-Burkhardt F, Riederer P. [JHJMK-801 bind­ing sites in postmortem brain regions of schizophrenic pa­tients. J Neural Transm 1989; 77: 23 1-6

58. Suga I, Kobayashi T, Ogata H, et al. Increased JH-MK80 I bind­ing in the post-mortem brains of chronic schizophrenic pa­tients [abstractl. The Satellite Symposium of the 17th Congress Internationale Neuropsychopharmacologicum: 199015 Sept; Kyoto, P.28

59. Simpson M, Slater P, Royston M, et al. Alterations in phencyclidine and sigma binding sites in schizophrenic brains. Schizophr Res 1992; 6: 41-8

60. Ishimaru M, Kurumaji A, Toru M. Increases in strychnine-in­sensitive glycine binding sites in cerebral cortex of chronic schizophrenics: evidence for glutamate hypothesis. Bioi Psy­chiatry 1994; 35: 84-95

6 I. Harrison P, McLaughlin D, Kerwin R. Decreased hippocampal expression of a glutamate receptor gene in schizophrenia. Lancet 1991; 337: 450-2

62. Eastwood S, McDonald B, Burnet P, et al. Decreased expression of mRNAs encoding non-NMDA glutamate receptors GluRI and GluR2 in medial temporal lobe neurons in schizophrenia. Brain Res Mol Brain Res 1995; 29: 211-23

eNS Drugs 1997 Jan: 7 (1)

Page 18: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

64

63. Breese C, Freedman R, Leonard S. Glutamate receptor subtype expression in human postmortem brain tissue from schizo­phrenics and alcohol abusers. Brain Res 1995; 674: 82-90

64. Akbarian S, Sucher N, Bradley D, et al. Selective alterations in gene expression for NMDA receptor subunits in prefrontal cortex of schizophrenia. J Neurosci 1996; 16: 19-30

65. Bogerts B, Meertz E, Schonfeldt-Bausch R. Basal ganglia and limbic system pathology in schizophrenia. Arch Gen Psychi­atry 1985; 42: 784-91

66. Brown R, Colter N, Corsellis J, et al. Postmortem evidence of structural brain changes in schizophrenia. Arch Gen Psychia­try 1986; 43: 36-42

67. U ras J, Monaghan D, Cotman C. Plastic response of hippocam­pal excitatory amino acid receptors to differentiation and re­innervation. Neuroscience 1990; 34: 9-17

68. Uras J, Monaghan D, Cotman C. Kainate receptors in the rat hippocampus: a distribution and time course of changes in response to unilateral lesions of the entorhinal cortex. J Neu­rosci 1990; 10: 2352-62

69. Lahtinen H, Kornhonen E, Castren E, et al. Long-term alter­ations in NMDA-sensitive 1-[3H]glutamate binding in the rat hippocampus following fimbria-fornix lesioning. Exp Neurol 1993; 121: 193-9

70. Weinberger D, Berman K, Zec R. Physiological dysfunction of dorsolateral prefrontal cortex in schizophrenia: I. Regional cerebral blood flow (rCBF) evidence. Arch Gen Psychiatry 1986; 43: 114-25

71. Weinberger D. Schizophrenia and the frontal lobe. Trends Neu­rosci 1988; II: 367-70

72. Kupfermann I. Localization of higher cognitive and affective functions: the association cortices. In: Kandel E, Schwartz J, Jessell T, editors. Principles of neural science, 3rd ed. New York: Elsevier, 1991: 823-38

73. Andersen R. Inferior parietal lobule function in spatial perception and visuomotor integration. In: Mountcastle Y, editor. Hand­book of physiology. Sect. 1: The nervous system. Bethesda, MD: American Physiological Society, 1987: 483-518

74. Kalivas P. Neurotransmitter regulation of dopamine neurons in the ventral tegmental area. Brain Res Rev 1993; 18: 75-113

75. Meredith G, Pennartz C, Groenewegen H. The cellular frame­work for chemical signalling in the nucleus accumbens. Prog Brain Res 1993; 99: 3-24

76. Kotter R. Postsynaptic integration of glutamatergic and dopa­minergic signals in the striatum. Prog Neurobiol 1994; 44: 163-96

77. Deutch A. Tam S-Y, Freeman A, et al. Mesolimbic and mesocortical dopamine activation induced by phencyclidine: contrasting pattern to striatal response. Eur J Pharmacol 1987; 134: 257-64

78. Scatton B, Boireau A, Garret C, et al. Action of the palmitic ester of pipotiazine on dopamine metabolism in the nigro-striatal, meso-limbic and meso-cortical systems. Naunyn Schmiedebergs Arch PharmacoI1977; 296: 169-75

79. Kaneno S, Watanabe S, Toru M, et al. Different dopamine stim­ulation of adenylate cyclase in striatum and mesolimbic area of rats treated with chronic chlorpromazine. Brain Res 1978; 152: 396-400

80. Bannon M. Pharmacology of mesocortical dopamine neurons. Pharmacol Rev 1983; 35: 53-68

81. Tanii Y, Nishikawa T, Umino A, et al. Phencyclidine increases extracellular dopamine metabolites in rat medial frontal cor­tex as measured by ill vivo dialysis. Neurosci Lett 1990; 112: 318-23

© Adis International Limited. All rights reserved.

Ishimaru & TofU

82. Hata N, Nishikawa T, Umino A, et al. Evidence for involvement of N-methyl-D-aspartate receptor in tonic inhibitory control of dopaminergic transmission in rat medial frontal cortex. Neurosci Lett 1990; 120: 101-4

83. Beckstead R. An autoradiographic examination of corticocorti­cal and subcortical projections of the mediodorsal-projection (prefrontal) cortex in the rat. J Comp Neurol 1979; 184: 43-62

84. Cotman C, Monaghan D, Ottersen 0, et al. Anatomical organi­zation of excitatory amino acid receptors and their pathways. Trends Neurosci 1987; 10: 273-80

85. Pycock C, Kerwin R, Carter C. Effect of lesion of cortical do­pamine terminals on subcortical dopamine receptors in rats. Nature 1980; 286: 74-7

86. Glowinski J, Tassin J, Thierry A. The mesocortico-prefrontal dopaminergic neurons. Trends Neurosci 1984; 7: 415-8

87. Weinberger D. Implications of normal brain development for the pathogenesis of schizophrenia. Arch Gen Psychiatry 1987; 44: 660-9

88. Carlsson A. The current status of the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 1988; I: 179-86

89. Coyle J. The glutamatergic dysfunction hypothesis for schizo­phrenia. Harv Rev Psychiatry 1996; 3: 241-53

90. Byrd J, Bykov Y, Rothman R. Chronic haloperidol treatment up-regulates rat brain PCP receptors. Eur J Pharmacol 1987; 140: 121-2

91. Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment-re­sistant schizophrenic. Arch Gen Psychiatry 1988; 45: 789-96

92. Kane J, Honigfeld G, Singer J, et al. Clozapine in treatment-re­sistant schizophrenics. Psychopharmacol Bull 1988; 24: 62-7

93. Meltzer Y. Treatment of the neuroleptic nonresponsive schizo­phrenic patient. Schizophr Bull 1992; 18: 515-42

94. Schmidt W, Krahling H, Ruhland M. Antagonism of AP-5- and amphetamine-induced behavior by timelotem as compared with clozapine and haloperidol. Life Sci 1990; 41: 1909-14

95. Tiedtke P, BischoffC, Schmidt W. MK-SOI-induced stereotypy and its antagonism by neuroleptic drugs. J Neural Transm Gen Sect 1990; 81: 173-S2

96. Hoffman D. Typical and atypical neuroleptics antagonize MK­SOl-induced locomotion and stereotypy in rats. J Neural Transm Gen Sect 1992; 89: 1-10

97. Hoffman D, Donovan H, Cassella J. The effects of haloperidol and clozapine on the disruption of sensorimotor gating in­duced by the noncompetitive glutamate antagonist MK-801. Psychopharmacology 1993; 111: 339-44

98. Bakshi Y, Swerdlow N, Geyer M. Clozapine antagonizes phencyclidine-induced deficits in sensorimotor gating of the startle response. J Pharmacol Exp Ther 1994; 271: 787-94

99. Corbett R, Camacho F, Woods A, et al. Antipsychotic agents an­tagonize non-competitive N-methyl-D-aspartate antagonist-in­duced behaviors. Psychopharmacology 1995; 120: 67-74

100. Farber N, Foster J, Olney J. Clozapine and related analogs block NMDA antagonist neurotoxicity: implications for schizo­phrenia. Schizophr Res 1995; 15: 57-8

10 1. Lang A, Yasar E, Soosaar A, et al. The involement of sigma and phencyclidine receptors in the action of antipsychotic drugs. Pharmacol Toxicol 1992; 71: 132-8

102. Janowsky A, Berger S. Clozapine inhibits L 3H1MK-80 1 binding to the glutamate receptor ion channel complex [abstract!. Schizophr Res Abs 1989; 2: 189

103. Daly D, Moghaddam B. Actions of clozapine and haloperidol on the extracellular levels of excitatory amino acids in the prefrontal cortex and striatum of conscious rats. Neurosci Lett 1993; 152: 61-4

eNS Drugs 1997 Jan; 7 (1)

Page 19: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

104. Lidsky T. Yablonsky-Alter E. Zuck L. et al. Anti-glutamatergic effects of c1ozapine. Neurosci Lett 1993; 163: 155-8

105. Meltzer H. Matsubara S. Lee J-c. Classification of typical and atypical antipsychotic drugs on the basis of dopamine 0-1. 0-2 and serotonin pKi values. J Pharmacol Exp Ther 1989; 251: 238-46

106. Reynolds G. Developments in the drug treatment of schizophre­nia. Trends Pharmacol Sci 1992; 13: 116-21

107. Svensson T. Mathe J. Andersson J. et al. Mode of action of atypical neuroleptics in relation to the phencyclidine model of schizophrenia: role of 5-HT 2 receptor and ul-adrenorecep­tor antagonism. J Clin Psychopharmacol1995; 15 I Suppl. I: IIS-18S

108. Choi D. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1988; I: 623-34

109. Tabor C. Tabor H. Polyamines. Annu Rev Biochem 1984; 53: 749-90

110. Yoneda Y. Ogita K. Neurochemical aspects of the N-methyl-D­aspartate receptor complex. Neurosci Res 1991; 10: 1-33

III. Hirsch S. Richardson-Andrews R. Costall B. et al. The effects of some polyamines on putative behavioural indices of mesolimbic versus striatal dopaminergic function. Psycho­pharmacology 1987; 93: 101-4

112. Quiron R. Bayorh M. Zerbe R. et al. Evidence for an endoge­nous peptide ligand for the phencyclidine receptor. Pep tides 1984; 5: 967-73

113. Zukin S. Zukin R. Vale W. et al. An endogenous ligand of the brain a/PCP receptor antagonizes NMDA-induced neuro­transmitter release. Brain Res 1987; 416: 84-9

114. Contreras P. Johnson S. Freedman R. et aI. Metaphit. an acylating ligand for phencyclidine receptors: characterization of in vivo actions in the rat. J Pharmacol Exp Ther 1986; 238: 1101-7

115. Koek W. Woods J. Jacobson A. et al. Metaphit. a proposed phencyclidine receptor acylator: phencyclidine-like behav­ioral effects and evidence of absence of antagonist activity in pigeons and in rhesus monkeys. J Pharmacol Exp Ther 1986; 237: 386-92

116. Johnson J. Ascher P. Glycine potentiates the NMDA response in cultured mouse brain neurons. Nature 1987; 325: 529-31

117. Thomson A. Glycine is acoagonist at the NMDAreceptor/chan­nel complex. Prog Neurobiol 1990: 35: 53-74

118. Kleckner N. Dingledine R. Requirement for glycine in activa­tion of NMDA-receptors expressed in Xenopus oocytes. Sci­ence 1988; 241: 835-7

119. Baron B. Harrison B. Miller F. et al. Activity of 5.7-dichloro­kynurenic acid. a potent antagonist at the N-methyl-D-aspart­ate receptor-associated glycine binding site. Mol Pharmacol 1990; 38: 554-61

120. Deutsch S. Mastropaolo J. Schwartz B. et al. A 'glutamatergic hypothesis' of schizophrenia - rationale for pharmacotherapy with glycine. Clin Neuropharmacol 1989; 12: 1-13

121. Bertolino M. Vicini S. Mazzetta J. et al. Phencyclidine and glycine modulate NMDA-activated high conductance cat­ionic channels by acting at different sites. Neurosci Lett 1988; 84:351-5

122. Banos G. Daniel p. Moorhouse S. et al. The requirements ofthe brain for some amino acids. J Physiol 1975; 46: 539-48

123. Oldendorph W. Brain uptake of radio labelled amino acids. amines and hexoses after arterial injection. Am J Physiol 1971; 221: 1629-39

124. Toth E. Lajtha A. Elevation of cerebral levels of non-essential amino acids in vivo by administration of large doses. Neuro­chern Res! 981; 6: 1309-17

© Adis International Limited. All rights reserved.

65

125. Riker W. Gold H. The pharmacology of sodium hydroxyacetate with observations on the toxicity of glycine. J Am Pharm Assoc 1942; 31: 306-12

126. Daly E. Aprison M. Glycine. In: Lajtha A. editor. Handbook of neurochemistry. Vol 3. New York: Plenum Press. 1983: 467-99

127. Toth E. Lajtha A. Antagonism of phencyclidine-induced hyperac­tivity by glycine in mice. Neurochem Res 1986; II: 393-400

128. Contreras P. D-Serine antagonized phencyclidine- and MK-80 I-induced stereotyped behavior and ataxia. Neuropharma­cology 1990; 29: 291-3

129. Tanii Y. Nishikawa T. Hashimoto A. et al. Stereoselective inhi­bition by 0- and L-alanine of phencyclidine-induced locomo­tor stimulation in the rat. Brain Res 1991; 563: 281-4

130. Tanii Y. Nishikawa T. Hashimoto A. et al. Stereose1ective an­tagonism by enantiomers of alanine and serine of phencyclid­ine-induced hyperactivity. stereotypy and ataxia in the rat. J Pharmacol Exp Ther 1994; 269: 1040-8

131. Waziri R. Glycine therapy of schizophrenia. Bioi Psychiatry 1988; 23: 210-1

132. Rosse R. Theut S. Banay-Schwartz M. et al. Glycine adjuvant therapy to conventional neuroleptic treatment in schizophre­nia: an open-label. pilot study. Clin Neuropharmacol 1989; [2: 416-24

133. Costa J. Khaled E. Sramek J. et al. An open trial of glycine as an adjunct to neuroleptics in chronic treatment-refractory schizophrenics. J Clin Psychopharmacol 1990; 10: 71-2

134. Potkin S. Costa J. Roy S. et al. Glycine in the treatment of schizophrenia: theory and preliminary results. In: Meltzer H. editor. Novel antipsychotic drugs. New York: Raven Press. 1992: 179-89

135. Javitt D. Zylberman [. Zukin S. et al. Amelioration of negative symptoms in schizophrenia by glycine. Am J Psychiatry 1994; 151: 1234-6

136. Rosse R. Schwartz B. Leighton M. et al. An open-label trial of milacemide in schizophrenia: an NMDA intervention strat­egy. Clin Neuropharmacol 1990; 13: 348-54

137. Rosse R. Schwartz B. Esther Davis R. et al. An NMDA inter­vention strategy in schizophrenia with 'low-dose' milacem­ide. Clin Neuropharmacol 1991; 14: 268-72

138. Tamminga C. Cascella N. Fakouhi T. et al. Enhancement of NMDA-mediated transmission in schizophrenia - effects of milacemide. In: Meltzer H. editor. Novel antipsychotic drugs. New York: Raven Press. 1992: 171 -7

139. Cascella N. Macciardi F. Cavallini C. et al. d-Cycloserine ad­juvant therapy to conventional neuroleptic treatment in schizophrenia: an open-label study. J Neural Transm Gen Sect 1994; 95: 105-11

140. Goff D. Tsai G. Manoach D. et al. Dose-finding trial of D-cy­c10serine added to neuroleptics for negative symptoms in schizophrenia. Am J Psychiatry 1995; 152: 1213-5

141. Guy W. editor. ECDEU assessment manual for psychopharma­cology. Publication ADM 76-338. Washington. DC: US De­partment of Health. Education. and Welfare. 1976: 534-7

142. Overall J. Gorham D. The brief psychiatric rating scale. Psychol Rep 1961; 10: 799-812

143. Bonato G. Manual for the ECDU assessment battery. 2nd rev. ed. Washington. DC: US Department of Health and Human Services. 1970

144. Alpert M. Rush M. Comparison of affect in Parkinson's disease and schizophrenia. Psychopharmacol Bull 1983; 19: 118-20

145. Endicott J. Spitzer R. Fleiss J. et al. The Global Assessment Scale: a procedure for measuring overall severity of psychi­atric disturbance. Arch Gen Psychiatry 1976; 33: 766-71

eNS Drugs 1997 Jan; 7 (1)

Page 20: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

66

146. Kay S, Fiszbein A, Opler L. The Positive and Negative Syn­drome Scale (PANSS) for schizophrenia. Schizophr Bull 1987; 13: 261-76

147. Andreasen N. The scale for the Assessment of Negative Symp­toms (SANS). Iowa City, Iowa: Univ. of Iowa, 1983

148. Simpson G, Angus J. A rating scale for extrapyramidal side effects. Acta Psychiatr Scand Suppl 1970; 212: 11-9

149. Sternberg S. Memory-scanning: mental processes revealed by reaction time experiments. Am Sci 1969; 57: 421-57

150. Heaton R. A manual for the Wisconsin Card Sorting Test. Odessa, FL: Psychological Assessment Resources, 1981

lSI. Waziri R, Wilcox J, Sherman A, et al. Serine metabolism and psychosis. Psychiatry Res 1984; 12: 121-36

152. Waziri R, Mott J. A biochemical basis for psychotic symptoms in patients with brain dysfunction. Eur Arch Psychiatr Neurol Sci 1987; 236: 251-5

153. Shank R, Aprison M. The metabolism in vivo of glycine and serine in eight areas of the rat central nervous system. J Neu­rochem 1970; 17: 1461-75

154. Perry T, Hansen S. Interconversion of serine and glycine is nor­mal in psychotic patients. Psychiatry Res 1985; 15: 109-13

155. Lucca A, Cortinovis S, Lucini V. Serine and glycine metabolism in schizophrenic patients. Prog Neuropsychopharmacol BioI Psychiatry 1993; 17: 947-53

156. Waziri R, Baruah S, Sherman A. Abnormal serine-glycine me­tabolism in the brains of schizophrenics. Schizophr Res 1992; 8: 233-43

157. RobaJ. Cavalier R, Cordi A, et al. Milacemide. In: MeldrumB, Porter R, editors. New anticonvulsant drugs. London: J. Libbey & Co., 1986: 179-90

158. Janssens de Varebeke P, Pauwels G, Buyse C, et al. The novel neuropsychotropic agent milacemide is a specific enzyme-ac­tivated inhibitor of brain monoamine oxidase. Br J Neu­rochem 1989; 53: 1l09-16

159. Truong D, Diamond B, Pezzoli G, et al. Monoamine oxidase inhibitory properties of milacemide in rats. Life Sci 1989; 44: 1059-66

160. Truong D, Galloway M, Pezzoli G, et al. Milacemide increases 5-hydroxytryptamine and dopamine levels in rat brain - pos­sible mechanisms of milacemide antimyoclonic property in the p'p'-DDT-induced myoclonus. Pharmacol Biochem Behav 1989; 32: 993-1001

161. Norris D, Mastropaolo J, O'Connor D, et al. Glycinergic inter­ventions potentiate the ability of MK-80 I to raise the thresh­old voltage for tonic hindlimb extension in mice. Pharmacol Biochem Behav 1991; 43: 609-12

162. Simeon J, Fink M, Itil T, et al. d-Cycloserine therapy of psychosis by symptom provocation. Compr Psychiatry 1970; II: 80-8

163. Hood W, Compton R, Monahan J. D-cycloserine: a ligand for N-methyl-D-aspartate coupled glycine receptor has partial ag­onist properties. Neurosci Lett 1989; 98: 91-5

164. Watson G, Bolanowski M, BaganoffM, et al. D-cycloserine acts as a partial agonist at the glycine modulatory site of the NMDA receptor expressed in Xenopus oocytes. Brain Res 1990: 510: 158-60

165. Wood P, Emmett M, Rao T, et al. In vivo modulation of the N-methyl-D-aspartate receptor complex by D-serine: potenti­ation of ongoing neuronal activity as evidenced by increased cerebellar cyclic GMP. J Neurochem 1989; 53: 979-81

166. Emmett M, Mick S, Cler J, et al. Actions ofD-cycloserine atthe N-methyl-D-aspartate associated glycine receptor site in vivo. Neuropharmacology 1991; 30: 1167-71

167. Bleuler E. Dementia praecox, or the group of schizophrenias. New York: International Universities Press, 1950

© Adis International Limited. All rights reserved.

Ishimaru & Toru

168. Kretschmer B, Zadow B, Volz T, et al. The contribution of the different binding sites of the N-methyl-D-aspartate (NMDA) receptor to the expression of behavior. J Neural Transm Gen Sect 1992;87: 23-35

169. Kretschmer B, Schmidt W. Glycine agonists in the treatment of schizophrenia? Clin Neuropharmacol 1992; 15: 157-61

170. Carlsson M, Engberg G, Carlsson A. Effects of D-cycloserine and (+)-HA-966 on the locomotor stimulation induced by NMDA antagonists and clonidine in monoamine-depleted mice. J Neural Transm Gen Sect 1994; 95: 223-33

171. Bristow L, Hutson P, Thorn L, et al. The glycine/NMDA recep­tor antagonist, R-(+)-HA-966, blocks activation of the mesolimbic dopaminergic system induced by phencyclidine and dizocilpine (MK-801) in rodents. Br J Pharmacol 1993; 108: 1156-63

172. Nishikawa T, Hashimoto A, Tanii Y, et al. Disturbed neurotrans­mission via the N-methyl-D-aspartate receptor and schizo­phrenia. Development of psychiatry series. In: Moroji T, Yamamoto K, editors. The biology of schizophrenia. Amster­dam: Elsevier, 1994: 197-207

173. Wroblewski J, Fadda E, Mazzetta J, et al. Glycine and D-serine act as positive modulators of signal transduction atN-methyl­D-aspartate sensitive glutamate receptors in cultured cerebel­lar granule cells. Neuropharmacology 1989; 28: 447-52

174. Kalivas P, Duffy P, Barrow J. Regulation of the mesocorticolim­bic dopamine system by glutamic acid receptor SUbtypes. J Pharmacol Exp Ther 1989; 251: 378-87

175. Fletcher E, Lodge D. Glycine reverses antagonism of N-methyl­D-aspartate (NMDA) by l-hydroxy-3-aminopyrrolidone-2 (HA-966) but not by D-2-amino-5-phosphonovalerate (D­AP5) on rat cortical slices. Eur J Pharmacol 1988; 151: 161-2

176. Thomson A, Walker V, Flynn D. Glycine enhances NMDA-re­ceptor mediated synaptic potentials in neocortical slices. Na­ture 1989; 338: 422-4

177. Larson A, Beitz A. Glycine potentiates strychnine-induced con­vulsions: role ofNMDA receptors. J Neurosci 1988; 8: 3822-6

178. Danysz W, Wrobewski J, Brooker G, et al . Modulation of glu­tamate receptors by phencyclidine and glycine in the rat cer­ebellum: cGMP increase in vivo. Brain Res 1989; 479: 270-6

179. Salt T. Modulation of NMDA receptor-mediated responses by glycine and D-serine in the rat thalamus in vivo. Brain Res 1989; 481: 403-6

180. D'Angelo E, Rossi P, Garthwaite J. Dual-component NMDA receptor currents at a single central synapse. Nature 1990; 346: 467-70

181. Wood P. The co-agonist concept: is the NMDA-associated gly­cine receptor saturated in vivo? Life Sci 1995; 57: 301-10

182. Liu Q-R, Nelson H, Mandiyan S, et al. Cloning and expression of a glycine transporter from mouse brain. FEBS Lett 1992; 305: 110-4

183. Smith K, Borden L, Hartig P, et al. Cloning and expression of a glycine transporter reveal colocalization with NMDA recep­tors. Neuron 1992; 8: 927-35

184. Guastella J, Brecha N, Weigmann C, et al. Cloning, expression, and localization of a rat brain high-affinity glycine trans­porter. Proc Nat! Acad Sci USA 1992; 89: 7189-93

185. Watson G, Hood W, Monahan J, et al. Kynurenate antagonizes actions of N-methyl-D-aspartate through a glycine-sensitive receptor. Neurosci Res Commun 1988; 2: 169-74

186. Birch P, Grossman C, Hayes A. Kynurenic acid antagonises responses to NMDA via an action at the strychnine-insensi­tive glycine receptor. Eur J Pharmacol 1988; 154: 85-7

187. Kessler M, Terramani T, Lynch G, et al. A glycine site associated with N-methyl-D-aspartic acid receptors: characterization

eNS Drugs 1997 Jan; 7 (1)

Page 21: The Glutamate Hypothesis of Schizophreniapositive schizophrenic symptoms, the glutamate hypothesis may provide a more comprehensive view of the illness. Postmortem brain assays also

Glutamate Hypothesis of Schizophrenia

and identification of a new class of antagonists. J Neurochem 1989; 52: 1319-28

188. Huettner J. Competitive antagonism of glycine at the N-methyl­D-aspartate (NMDA) receptor. Biochem Pharmacol 1991 ; 41: 9-16

189. Leeson P, Baker R, Carling R, et al. Kynurenic acid derivatives: structure-activity relationships for excitatory amino acid an­tagonism and identification of potent and selective antago­nists at the glycine site on the N-methyl-D-aspartate receptor. J Med Chem 1991 ; 34: 1243-52

190. Snell L, Morter R, Johnson K. Structural requirements for ac­tivation of the glycine receptor that modulates the N-methyl­D-aspartate operated ion channel. Eur J Pharmacol 1988; 156: 105-10

191. Fadda E, Danysz W, Wroblewski J, et al. Glycine and D-serine increase the affinity of N-methyl-D-aspartate sensitive gluta­mate binding sites in rat brain synaptic membranes. Neuro­pharmacology 1988; 27: 1183-5

192. Corrigan J. D-Amino acids in animals. Science 1969; 164: 142-9 193. Man E, Sand house M, Burg J, et al. Accumulation ofD-aspartic

acid with age in the human brain. Science 1983; 220: 1407-8 194. Dunlop D, Neidle A, Mahale D, et al. The presence of free

D-aspartic acid in rodents and man. Biochem Biophys Res Commun 1986; 141: 27-32

67

195. Hashimoto A, Nishikawa T, Hayashi T, et al. The presence of free D-serine in rat brain. FEBS Lett 1992; 296: 33-6

196. Hashimoto A, Kumashiro S, Nishikawa T, et al. Embryonic development and postnatal changes in free D-aspartate and D-serine in the human prefrontal cortex. J Neurochem 1993; 61: 348-51

197. Hashimoto A, Nishikawa T, Oka T, et al. Endogenous D-serine in rat brain: N-methyl-D-aspartate receptor-related distribu­tion and aging. J Neurochem 1993; 60: 783-6

198. Matsui T, Sekiguchi M, Hashimoto A, e t al. Functional compar­ison of D-serine and glycine in rodents: the effect on cloned NMDA receptors and the extracellular concentration. J Neu­rochem 1995; 65: 454-8

Correspondence and reprints: Dr Masahiko J. Ishimaru, Department of Psychiatry, Washington University School of Medicine, 4940 Children's Place, Saint Louis, MO 63110, USA. E-mail: [email protected]

Fourth Eilat Conference on

New Antiepileptic Drugs Date: 6-10 September 1998

Venue: Eilat, Israel

For further information, please contact: Conference Secretariat

P.O. Box 29041 Tel Aviv 61290

ISRAEL

Tel. : +972 3 5175149/50 Fax: +972 3 5175155 e-mail: trgt@netvi ion.ncUI

© Adis Internationa l Limi ted. All righ ts reserved, e NS Drugs 1997 Jan: 7 (1)