Review
Glutamate signaling in the pathophysiology and therapy of schizophrenia

https://doi.org/10.1016/j.pbb.2011.03.023Get rights and content

Abstract

Glutamatergic neurotransmission, particularly through the N-methyl-d-aspartate (NMDA) receptor, has drawn attention for its role in the pathophysiology of schizophrenia. This paper reviews the neurodevelopmental origin and genetic susceptibility of schizophrenia relevant to NMDA neurotransmission, and discusses the relationship between NMDA hypofunction and different domains of symptom in schizophrenia as well as putative treatment modality for the disorder. A series of clinical trials and a meta-analysis which compared currently available NMDA-enhancing agents suggests that glycine, d-serine, and sarcosine are more efficacious than d-cycloserine in improving the overall psychopathology of schizophrenia without side effect or safety concern. In addition, enhancing glutamatergic neurotransmission via activating the AMPA receptor, metabotropic glutamate receptor or inhibition of d-amino acid oxidase (DAO) is also reviewed. More studies are needed to determine the NMDA vulnerability in schizophrenia and to confirm the long-term efficacy, functional outcome, and safety of these NMDA-enhancing agents in schizophrenic patients, particularly those with refractory negative and cognitive symptoms, or serious adverse effects while taking the existing antipsychotic agents.

Research highlights

► NMDA neurotransmission has drawn attention for its role in schizophrenia. ► NMDA function involves in developmental and genetic susceptibility of schizophrenia. ► NMDA hypofunction accounts for different domains of symptom in schizophrenia. ► Enhancing NMDA function via the coagonist or glycine transporter is new treatment.

Introduction

There are plenty of studies which have suggested that dysregulation of dopaminergic (Davis et al., 1991, Toda and Abi-Dargham, 2007), γ-aminobutyric acid (GABA) (Benes and Berretta, 2001, Lewis et al., 2005), glutamatergic (Goff and Coyle, 2001, Moghaddam, 2003) neurotransmission and their interactions (Carlsson et al., 2001) are involved in the pathophysiology of schizophrenia. Among these, the hypofunction of N-methyl-d-aspartate (NMDA) glutamatergic neurotransmission has gained much attention since two decades ago (Olney and Farber, 1995, Javitt, 2008).

Conventional antipsychotics, which block D2 dopamine receptors (Farde et al., 1986), exert effects mainly on positive symptoms. Second-generation antipsychotics (SGAs) have been suggested to be superior to conventional agents in terms of efficacy for positive symptoms, negative symptoms and cognitive deficits but the therapeutic gain is modest (Green et al., 1997, Lane and Chang, 1999, Leucht et al., 2003, Livingston, 1994). Overall, there is a considerable percentage of patients resistant or only partially responsive to available antipsychotic medications (Lieberman et al., 2005). Life threatening side-effect profiles of SGAs, particularly the metabolic syndrome, limit the clinical use of these agents (Lu et al., 2004, Newcomer, 2007, Simon et al., 2009). Moreover, most schizophrenic patients still suffer from lifelong illness and deteriorating function (Hwu et al., 2002, Malla and Payne, 2005, Tsuang et al., 2000). Hence, there is a great need to develop new therapies that will provide better long-term efficacy, functional improvement and safety profiles for schizophrenic patients.

Glutamate is the most abundant amino acid neurotransmitter in the mammalian brain. There are two types of glutamate receptors: metabotropic and ionotropic receptors. More evidence regarding the involvement of glutamatergic system in schizophrenia focuses on the ionotropic receptors which are subdivided to 3 subtypes: NMDA, quisqualate/α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainate (Lodge, 2009). Relief of the depolarization blockade of NMDA receptor requires the activation of non-NMDA receptor. Studies on long-term potentiation (LTP) indicate that NMDA receptors interact with AMPA receptors (Yu et al., 2008). However, the NMDA receptor is the best studied and most relevant subtype of glutamate receptors to understand the pathophysiology of schizophrenia. The NMDA receptor has been demonstrated to play an important role in neurocognition and neurotoxicity (Lipson and Rosenberg, 1994, Kalia et al., 2008). The psychosis due to the blockade of the NMDA receptor is similar to the clinical manifestation of schizophrenia. Up to date, NMDA synapse remains to be the only therapeutic target that is confirmed to have clinical efficacy. Thus, we will focus on the glutamatergic neurotransmission via NMDA receptor in the following review.

In addition to the agonists and coagonists, the NMDA receptor can be regulated by a variety of molecules, including polyamines, proton, zinc, magnesium, phencyclidine (PCP) and ketamine. These sophisticated regulatory mechanisms suggest that NMDA receptor can adapt to endogenous and exogenous signals to maintain and facilitate a variety of vital brain functions including cognition, memory, neurodevelopment, synaptic plasticity and psychosis (Bliss and Collingridge, 1993).

The NMDA receptor is composed of multiple subunits including NR1 and one of either the NR2 (NR2 A–D) or NR3 (NR3 A–B) to form heteromeric receptor-channels with different pharmacologic and biophysical characteristics (Laurie and Seeburg, 1994) (Fig. 1). The NMDA receptor possesses a number of unique characteristics. For example, it has binding sites not only for glutamate or aspartate, but also a separate coagonist site for the endogenous ligands, d-serine, d-alanine, and glycine. Occupancy of the coagonist site can increase the frequency of opening of the channels activated by NMDA agonists, facilitating excitatory transmission in the brain (Johnson and Ascher, 1987). In fact, the binding of both glycine (or d-serine, d-alanine (Chessell et al., 1991)) and glutamate is required to open the NMDAR channel ionophore (Mayer et al., 1989, Nong et al., 2003, Thomson et al., 1989) (Fig. 1).

Since d-alanine, which presents only in the pituitary, is less likely to play physiological role in the neocortex, most studies focused on the binding of d-serine and glycine on the d-serine/glycine site of NMDAR. Endogenous d-serine/glycine site agonists also play a role in neuromodulation. Binding to the d-serine/glycine site enhances the affinity and efficacy of the glutamate neurotransmission (Fadda et al., 1988), increases the duration and frequency of the open channel state (Vyklický et al., 1990), and promotes turnover of the NMDAR (Nong et al., 2003). Distribution of d-serine parallels to that of NR1, and d-Serine binds more tightly to the NMDAR than glycine (Furukawa and Gouaux, 2003).

The NMDAR d-serine/glycine site on the NR1 subunit is not fully saturated at synapses in brain regions such as the prefrontal cortex, neocortex, hippocampus, thalamus and brainstem slices, suggesting that agonists of the d-serine/glycine site are capable of regulating NMDAR-mediated neurotransmission (Labrie and Roder, 2010). Glycine is abundant throughout the brain and serves as a major inhibitory neurotransmitter in the hindbrain. Synaptic concentrations of glycine are primarily derived from astroglial cells, and its clearance is mediated by glycine transporter 1 (GlyT-1) (Kinney et al., 2003, Lim et al., 2004). d-Serine has been found to be more potent than glycine in targeting the d-serine/glycine site of most NMDARs (Boehning and Snyder, 2003).

Treatment with DAO results in depletion of d-serine which has been shown to attenuate NMDAR activity in cerebellar and hippocampal slices, hippocampal cell cultures, and retina preparations (Mothet et al., 2000, Gustafson et al., 2007). Supporting the physiological role, DAO inhibitor can facilitate the effects of d-serine on prepulse inhibition (PPI) (Hashimoto et al., 2009). d-Serine levels were reduced in the cerebrospinal fluid of drug naїve patients with schizophrenia (Hashimoto et al., 2005, Bendikov et al., 2007). Diminished d-serine along with elevation in l-serine also suggests a dysfunction of serine racemase (SRR) activity (Hashimoto et al., 2003). Changes in SRR protein expression have been found in the postmortem brains of schizophrenic individuals (Bendikov et al., 2007, Steffek et al., 2006, Verrall et al., 2007). Similarly, glycine levels have been found to be reduced in drug-free schizophrenic individuals and inversely correlate with the severity of negative symptoms (Sumiyoshi et al., 2004, Neeman et al., 2005). However, high-dose glycine impairs the prepulse inhibition measure of sensorimotor gating in humans, which does not support the glycine treatment for cognition (O'Neill et al., 2010). Due to glycine's complex metabolism of both excitatory and inhibitory signaling, d-serine is likely a better choice than glycine when considering applying full agonist for treatment.

Other regulators involved in the metabolism of d-serine such as d-amino acid oxidase (DAO) (Verrall et al., 2007, Bendikov et al., 2007), protein-interacting with kinase C (PICK1) (Beneyto and Meador-Woodruff, 2006) and alanine–serine–cysteine transporter 1 (Asc-1) (Burnet et al., 2008) were also found to be related to the d-serine levels. Depletion of d-serine was found to be associated with NMDAR-mediated neurological functions and NMDAR-induced neurotoxicity, as well as NMDAR-dependent LTP in many brain regions especially the hippocampus (Gustafson et al., 2007, Shleper et al., 2005, Mothet et al., 2006). d-Serine supplement can entirely reverse the effects of decreased NMDAR-mediated neurotransmission (Mothet et al., 2000, Mothet et al., 2006, Yang et al., 2003, Panatier et al., 2006, Gustafson et al., 2007), moreover, enhance NMDAR signaling demonstrated both in vitro (Chen et al., 2003, Martina et al., 2003, Yang et al., 2003, Chapman et al., 2003) and in mice which lack DAO activity (Wake et al., 2001), Acs-1 (Xie et al., 2005) or have diminished GAD67 expression (Reynolds et al., 2004, Torrey et al., 2005). In human genetic studies, significant associations between DAO (Ohnuma et al., 2009) and G72 (DAOA)/G30 (Shinkai et al., 2007) gene polymorphisms and schizophrenia were also observed in case–control association analyses.

mGluRs are relevant to schizophrenia as they have a role in modulating NMDAR-mediated neurotransmission. mGluRs are guanine nucleotide binding proteins [G] protein coupled receptors. When mGluRs are activated by glutamate, they release GDP, which subsequently alters activities of enzymes, ion channels, and vesicle transport. There are 8 subtypes of mGluRs, classified into 3 groups based on signaling pathways, pharmacology properties, and similarities in DNA sequences. Group 1 mGluRs use Gq proteins, and when activated by glutamate, signaling cascade involves phospholipase C which cleaves phosphatidylinositol-4-5 bisphosphate into diacylglycerol and inositol 1, 4, 5-triphosphate results in calcium release. Group 2 and Group 3 mGluRs interactions with G1/o species and signaling proteins include adenyl cyclase, which creates cyclic adenosine monophosphate (cAMP). mGluRs have been found at presynaptic glutamate terminals and GABA interneurons (Herron et al., 1986). Group 1 mGluRs can be found near synaptic dendritic spines, mGluR5 is located in the cortex, and hippocampal mGluR2 and mGluR3 are located both pre and postsynaptically on glutamatergic and GABAergic neurons. mGluR3 is also found in glia. One of the most striking effects of mGluR activation in a number of areas in the brain is promoting NMDAR-mediated neurotransmission (Conn et al., 2008). Specifically, Group 1 mGluRs increase presynaptic glutamate release, while Group 2 mGluRs decrease presynaptic glutamate release.

There exists a positive feedback mechanism between mGluR5 and NMDAR in which activation of mGluR5 potentiates NMDAR generated currents, and NMDAR mediated depolarization results in activation of a serine/threonine protein phosphatase that dephosphorylates mGluR5, which reverses desensitization of mGluR5, thereby allowing mGluR5 to be depolarized again (Alagarsamy et al., 1999). mGluR5 is located in the hippocampus and cortex, especially postsynaptic in CA 1 pyramidal cells. mGluR5 is also found in GABA neurons. Animal models of GluR5 knockouts exhibit decreased EPSP in the hippocampus and deficient NMDA dependent LTP and learning, indicating that mGluR5 is needed to maintain normal NMDAR-mediated neurotransmission. In addition, mGluR5 antagonists increase the effects of NMDAR antagonists. Group 2 mGluRs decrease glutamate release from presynaptic terminals, and likely function as autoregulation to protect neurons from excitotoxicity.

Group 2 mGluRs agonists block the effects of psychotomimetic agents on glutamatergic neurotransmission (Conn et al., 2008). Interestingly, however, activating mGluR2s has a paradoxical effect on dopamine release. Van Berckel et al. (2006) studied a mGluR2 agonist, LY354740 effect in four baboons. Amphetamine administration increased DA synaptic concentration as evidenced by decreased D2 receptor binding by [11C]raclopride, detected by PET scanning. In baboons treated by LY354740 there were even less unoccupied D2 receptors, indicating that stimulation of Group 2 mGluRs, thereby decreasing glutamatergic neurotransmission, increased in amphetamine induced release of dopamine in non human primates. However, the relationship between glutamate and dopamine is complex, as midbrain DA neurons are both activated by glutamate directly and inhibited indirectly by glutamate's effects on GABAergic interneurons. In terms of potential drug development, research has focused on Group 1, which includes mGluR1 and mGluR5, and Group 2, which includes mGluR2 and mGluR3.

Section snippets

Neurodevelopmental origin and genetic vulnerability of schizophrenia

Since NMDA receptor is essential and ubiquitous, it is highly regulated. Only when genetic insult(s) reaches a threshold of low NMDA function and impair the neurodevelopment, the nervous system will become vulnerable to schizophrenia. Findings from genetic studies suggest that the accumulation of multiple variations of candidate genes, rather than any single gene, may serve as a general model for the pathogenesis of schizophrenia (Girirajan and Eichler, 2010). Several susceptible factors have

Evidence of NMDA hypofunction in schizophrenia

Much evidence suggests that hypofunction of NMDA receptor-mediated neurotransmission is a critical deficit in schizophrenia (Bachus and Kleinman, 1996, Coyle, 1996, Javitt, 2004, Olney and Farber, 1995). The involvement of the NMDA system in schizophrenia is further evidenced by the effects of the NMDA-receptor antagonists, PCP and the dissociative anesthetic, ketamine, both of which induce psychiatric and physiological changes resembling schizophrenia more closely than the symptoms induced by

Evidence of NMDA hypofunction in animal genetic models

Several genes associated with glutamatergic NMDA synapses show relevance to schizophrenia (Hui et al., 2009). Some of these genes were studied in animal genetic models to demonstrate their implication in the pathophysiology of schizophrenia.

Mice which express only 5%–10% of the NR1 subunit of the NMDA receptor exhibit schizophrenia-like behaviors, such as abnormalities in motor activity, sensory processing, stereotypy, sexual, and social behavior (Duncan et al., 2004, Mohn et al., 1999,

NMDA enhancing treatments

Although the NMDA antagonism can account well for the phenomenology of schizophrenia, a critical challenge is whether the NMDA hypothesis can be applied to develop new therapeutic approaches for schizophrenia (Buchanan et al., 2007, Coyle et al., 2002, Javitt, 2004). There have been several NMDA-enhancing agents that can be classified by 3 different mechanisms of action: 1) full agonists of the coagonist site: glycine, d-serine and d-alanine; 2) partial agonist of the coagonist site: d

Effects on different symptom domains of schizophrenia: negative symptoms, cognitive deficits and quality of life

Negative symptoms are often refractory to antipsychotic treatment with the exception of clozapine. Although some studies suggest that newer SGAs targeting both dopamine D2 and serotonin 5HT2 receptors (Kapur et al., 1999) are superior to conventional agents for treating negative symptoms but their effects are not unequivocal (Green et al., 1997, Lane and Chang, 1999, Leucht et al., 2003, Livingston, 1994). The NMDA-enhancing agents add-on therapy has been demonstrated to be beneficial on the

Clinical efficacy of NMDA enhancing agents

A recent meta-analysis of all the double-blind, placebo-controlled studies in patients with schizophrenia examined the efficacy, the dose–response, the effects of concomitant antipsychotics, and side effects of all the NMDA-enhancing agents (Tsai and Lin, 2010). The result of 800 subjects from 26 studies showed that the NMDA-enhancing agents are significantly effective in most schizophrenic symptom domains with the effect size in the order of depressive (0.40), negative (0.38), cognitive

Perspective

While the results of the trials with NMDA-enhancing agents are encouraging, enhancement of glutamatergic neurotransmission via pathways other than NMDA receptors for the treatment of schizophrenia is drawing more attentions. Here we discuss two examples which suggest the potential therapeutic effects from non-NMDA targets.

Summary

This review has discussed the role of glutamatergic signaling, particularly the molecules of the NMDA synapse, and plays in the pathophysiology of schizophrenia from views of neurodevelopment, pathological finding, genetic vulnerability, animal models, pharmacology and clinical trials. Quite a few candidate genes have been identified to be involved in the neurodevelopment and glutamate-associated signaling relevant to schizophrenia; of them, DAO and G72 (DAOA) are directly involved in NMDA

Acknowledgments

The authors extend their sincere thanks to National Science Council, Taiwan (NSC-97-2314-B-039-006-MY3, NSC-98-2627-B-039-001, and NSC 99-3114-B-182A-003), National Health Research Institutes, Taiwan (NHRI-EX-100-9904NI), Department of Health, Taiwan (DOH99-TD-I-111-TM001), Taiwan Department of Health Clinical Trial and Research Center of Excellence (DOH100-TD-B-111-004), and China Medical University Hospital, Taiwan (DMR-98-093 and DMR-98-095) for funding this research.

References (223)

  • I.P. Chessell et al.

    d-Cycloserine, a putative cognitive enhancer, facilitates activation of the N-methyl-d-aspartate receptor-ionophore complex in Alzheimer brain

    Brain Res

    (1991)
  • A. Corvin et al.

    d-Amino acid oxidase (DAO) genotype and mood symptomatology in schizophrenia

    Neurosci Lett

    (2007)
  • S.D. Detera-Wadleigh et al.

    G72/G30 in schizophrenia and bipolar disorder: review and meta-analysis

    Biol Psychiatry

    (2006)
  • G.E. Duncan et al.

    Deficits in sensorimotor gating and tests of social behavior in a genetic model of reduced NMDA receptor function

    Behav Brain Res

    (2004)
  • E. Fadda 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)
  • K. Fukui et al.

    Molecular cloning and chromosomal localization of a human gene encoding d-amino-acid oxidase

    J Biol Chem

    (1992)
  • D.C. Goff et al.

    Once-weekly d-cycloserine effects on negative symptoms and cognition in schizophrenia: an exploratory study

    Schizophr Res

    (2008)
  • T.P. Green et al.

    Disposition of sodium benzoate in newborn infants with hyperammonemia

    J Pediatr

    (1983)
  • K. Hashimoto et al.

    Reduced d-serine to total serine ratio in the cerebrospinal fluid of drug naive schizophrenic patients

    Prog Neuropsychopharmacol Biol Psychiatry

    (2005)
  • K. Hashimoto et al.

    Co-administration of a d-amino acid oxidase inhibitor potentiates the efficacy of d-serine in attenuating prepulse inhibition deficits after administration of dizocilpine

    Biol Psychiatry

    (2009)
  • A. Hayashi-Takagi et al.

    Disturbed synaptic connectivity in schizophrenia: convergence of genetic risk factors during neurodevelopment

    Brain Res Bull

    (2010)
  • U. Heresco-Levy et al.

    High-dose glycine added to olanzapine and risperidone for the treatment of schizophrenia

    Biol Psychiatry

    (2004)
  • U. Heresco-Levy et al.

    d-Serine efficacy as add-on pharmacotherapy to risperidone and olanzapine for treatment-refractory schizophrenia

    Biol Psychiatry

    (2005)
  • J.F. Howes et al.

    Talampanel

    Neurotherapeutics

    (2007)
  • H.G. Hwu et al.

    Symptom patterns and subgrouping of schizophrenic patients: significance of negative symptoms assessed on admission

    Schizophr Res

    (2002)
  • H. Jaaro-Peled et al.

    Neurodevelopmental mechanisms of schizophrenia: understanding disturbed postnatal brain maturation through neuregulin-1-ErbB4 and DISC1

    Trends Neurosci

    (2009)
  • D.C. Javitt

    Glycine transport inhibitors and the treatment of schizophrenia

    Biol Psychiatry

    (2008)
  • D.C. Javitt et al.

    Bennett Research Award. Reversal of phencyclidine-induced effects by glycine and glycine transport inhibitors

    Biol Psychiatry

    (1999)
  • L.V. Kalia et al.

    NMDA receptors in clinical neurology: excitatory times ahead

    Lancet Neurol

    (2008)
  • J.T. Kantrowitz et al.

    High dose d-serine in the treatment of schizophrenia

    Schizophr Res

    (2010)
  • J. Karasawa et al.

    d-Serine and a glycine transporter inhibitor improve MK-801-induced cognitive deficits in a novel object recognition test in rats

    Behav Brain Res

    (2008)
  • R.M. Karlsson et al.

    Loss of glial glutamate and aspartate transporter (excitatory amino acid transporter 1) causes locomotor hyperactivity and exaggerated responses to psychotomimetics: rescue by haloperidol and metabotropic glutamate 2/3 agonist

    Biol Psychiatry

    (2008)
  • C.M. Adler et al.

    Comparison of ketamine-induced thought disorder in healthy volunteers and thought disorder in schizophrenia

    Am J Psychiatry

    (1999)
  • S. Alagarsamy et al.

    Activation of NMDA receptors reverses desensitization of mGluR5 in native and recombinant systems

    Nat Neurosci

    (1999)
  • A.C. Altamura et al.

    Factors affecting outcome in schizophrenia and their relevance for psychopharmacological treatment

    Int Clin Psychopharmacol

    (2007)
  • S.E. Bachus et al.

    The neuropathology of schizophrenia

    J Clin Psychiatry

    (1996)
  • T.M. Ballard et al.

    Severe impairment of NMDA receptor function in mice carrying targeted point mutations in the glycine binding site results in drug-resistant nonhabituating hyperactivity

    J Neurosci

    (2002)
  • G.R. Bartlett

    The inhibition of d-amino acid oxidase by benzoic acid and various monosubstituted benzoic acid derivatives

    J Am Chem Soc

    (1948)
  • A.C. Basu et al.

    Targeted disruption of serine racemase affects glutamatergic neurotransmission and behavior

    Mol Psychiatry

    (2009)
  • M.L. Batshaw et al.

    Evidence of lack of toxicity of sodium phenylacetate and sodium benzoate in treating urea cycle enzymopathies

    J Inherit Metab Dis

    (1981)
  • M.L. Batshaw et al.

    Treatment of urea cycle disorders

    Enzyme

    (1987)
  • M. Beneyto et al.

    Lamina-specific abnormalities of AMPA receptor trafficking and signaling molecule transcripts in the prefrontal cortex in schizophrenia

    Synapse

    (2006)
  • T.V. Bliss et al.

    A synaptic model of memory: long-term potentiation in the hippocampus

    Nature

    (1993)
  • D. Boehning et al.

    Novel neural modulators

    Annu Rev Neurosci

    (2003)
  • J.P. Bourgeois et al.

    Changes of synaptic density in the primary visual cortex of the macaque monkey from fetal to adult stage

    J Neurosci

    (1993)
  • A. Breier

    Cognitive deficit in schizophrenia and its neurochemical basis

    Br J Psychiatry

    (1999)
  • R.W. Buchanan et al.

    The Cognitive and Negative Symptoms in Schizophrenia Trial (CONSIST): the efficacy of glutamatergic agents for negative symptoms and cognitive impairments

    Am J Psychiatry

    (2007)
  • M. Cannon et al.

    Obstetric complications and schizophrenia: historical and meta-analytic review

    Am J Psychiatry

    (2002)
  • A.G. Cardno et al.

    Heritability estimates for psychotic disorders: the Maudsley twin psychosis series

    Arch Gen Psychiatry

    (1999)
  • A. Carlsson et al.

    Interactions between monoamines, glutamate, and GABA in schizophrenia: new evidence

    Annu Rev Pharmacol Toxicol

    (2001)
  • Cited by (130)

    • The neurobiology of schizophrenia

      2022, Neurobiology of Brain Disorders: Biological Basis of Neurological and Psychiatric Disorders, Second Edition
    View all citing articles on Scopus
    View full text