Chapter Twelve - The NMDA Receptor and Schizophrenia: From Pathophysiology to Treatment

https://doi.org/10.1016/bs.apha.2016.01.006Get rights and content

Abstract

Schizophrenia is a severe mental illness that affects almost 1% of the population worldwide. Even though the etiology of schizophrenia is uncertain, it is believed to be a neurodevelopmental disorder that results from a combination of environmental insults and genetic vulnerabilities. Over the past 20 years, there has been a confluence of evidence from many research disciplines pointing to alterations in excitatory signaling, particularly involving hypofunction of the N-methyl-d-aspartate receptor (NMDAR), as a key contributor to the schizophrenia disease process. This review describes the structure–function relationship of the NMDAR channel and how the glycine modulatory site acts as an important regulator of its activity. In addition, this review highlights the genetic, pharmacologic, and biochemical evidence supporting the hypothesis that NMDAR hypofunction contributes to the pathophysiology of schizophrenia. Finally, this chapter highlights some of the most recent and promising pharmacological strategies that are designed to either, directly or indirectly, augment NMDAR function in an effort to treat the cognitive and negative symptoms of schizophrenia that are not helped by currently available medications.

Introduction

Schizophrenia is a chronic, disabling mental disorder that affects 0.5–1% of the population worldwide (Perala et al., 2007) that commonly presents in late adolescence and early adulthood. It is characterized clinically by positive symptoms (delusions, hallucinations, and thought disorder), negative symptoms (ie, social withdrawal, poverty of speech, and anhedonia), and cognitive deficits (ie, attention, working memory, and executive function). While the negative and positive symptoms typically present during late adolescence or early adulthood (Lewis & Lieberman, 2000), the cognitive deficits generally appear years before clinical diagnosis (Lesh, Niendam, Minzenberg, & Carter, 2011).

One leading hypothesis is that the pathophysiology of schizophrenia has a neurodevelopmental component (Lewis and Lieberman, 2000, Rapoport et al., 2012). Brain development during adolescence, the time when many schizophrenia symptoms appear or worsen, is a dynamic period marked by extensive functional and neuroanatomical changes. This period is characterized by (1) fine-tuning of excitatory, inhibitory, and monoaminergic neurotransmitter systems; (2) stabilization of synapses to increase efficiency of neural function and diminish redundancy; and (3) beginning of integration between late-maturing and early-maturing brain structures (Keshavan, Giedd, Lau, Lewis, & Paus, 2014). Therefore genetic predisposition and environmental disturbances that lead to changes or imbalances in the timing of these developmental processes could increase the risk for developing schizophrenia.

Schizophrenia is associated with various neuroanatomical and structural brain abnormalities. Diffusion tensor magnetic resonance imaging studies show that white matter alterations are present from the preclinical to the chronic stages of schizophrenia, involving the long-association white matter tracts, corticospinal tracts, interhemispheric connections, cerebello-thalamo-cortical circuit, and limbic system (Canu, Agosta, & Filippi, 2015). A number of longitudinal structural magnetic resonance imaging studies have shown reduced whole brain volume and increased lateral ventricular volume in the early stages of schizophrenia and in chronically ill patients (Fusar-Poli et al., 2013, Kempton et al., 2010, Levitt et al., 2010), with some abnormalities already present during the prodromal phase (Fusar-Poli et al., 2011). The decreased brain volume in schizophrenia is especially pronounced in cortical areas, as well as in the hippocampus (Levitt et al., 2010, Tamminga et al., 2010). The extent of progressive brain tissue decrease in patients (− 0.5%/year) has been estimated as twice that of healthy controls (− 0.2%/year) (Hulshoff Pol & Kahn, 2008). Some of these neuroanatomical alterations may be correlated with antipsychotic treatment, as the higher the cumulative exposure to antipsychotic treatment, the greater the progressive gray matter volume decreases in patients over time (Fusar-Poli et al., 2013). Studies using human postmortem brain tissue suggest that decreased neuropil, including reduced dendritic length and spine density on pyramidal neurons, might be the primary contributor to reduced cortical gray matter volumes in schizophrenia (Glausier and Lewis, 2013, Konopaske et al., 2014).

Schizophrenia is a highly heritable disorder, with the relative risk being 6–17 × greater among first-degree relatives and 40–50 × greater in the monozygotic twin of an affected sibling compared to the general population (Cardno et al., 1999). Thus, there has been a massive effort in the field of psychiatric genetics to identify genes that confer risk to developing schizophrenia. Unfortunately, the genetics are very complex with non-Mendelian inheritance. Prior to genome-wide association studies (GWAS), candidate gene studies were utilized in an attempt to discover genetic risk factors for schizophrenia. Although these studies were able to identify numerous risk genes, there was often a failure to replicate associations across studies. We now know that these early candidate gene association studies for common genetic variation had inadequate statistical power to detect the small differences seen in schizophrenia (Farrell et al., 2015). Although the largest schizophrenia GWAS to date (~ 37,000 cases and ~ 113,000 controls; exponentially larger sample sizes than previous studies) identified SNPs in ~ 600 brain-enriched genes, almost all of the previously identified candidate risk genes were not found to be associated with schizophrenia in this study (Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2014). Copy number variants (CNVs) (stretches of DNA that are either deleted or duplicated) and rare exonic variants can also lead to increased genetic risk for schizophrenia (Fromer et al., 2014, Kirov et al., 2012). As a whole, these recent large-scale studies have identified both common and rare genetic variants in N-methyl-d-aspartate receptor (NMDAR) genes, as well as components of the postsynaptic density, with increased risk for schizophrenia.

In this chapter, we describe the structure and function of the NMDAR channel, with a particular focus on how the glycine modulatory site (GMS) regulates its activity. We will then discuss how it is believed that NMDAR hypofunction contributes to the pathophysiology of schizophrenia. Finally, we will highlight some of the latest pharmacological strategies aimed at both, directly and indirectly, augmenting NMDAR function.

Section snippets

NMDA Receptor: Structure and Function

In the brain, glutamate activates metabotropic and ionotropic receptors. The latter family of receptors is comprised of 18 gene products that are further divided into three subtypes based on their agonist preference: alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) receptors, kainate receptors, and NMDARs/GluNs. The heterotetrameric NMDAR is widely distributed throughout most of the brain and is a critical postsynaptic mediator of activity-dependent synaptic plasticity. This receptor

Glycine Modulatory Site

Work done on both native NMDARs (Johnson & Ascher, 1987) and those expressed by oocytes (Kleckner & Dingledine, 1988) demonstrated the need for concomitant binding of glutamate and glycine for receptor activation. Furthermore, it was shown that d-serine or d-alanine could also act as NMDAR coagonists at the GMS (Kleckner & Dingledine, 1988). d-amino acids, including d-serine are now well-established modulators of neuronal activity in mammals (Boehning and Snyder, 2003, Wolosker et al., 2008).

NMDA Receptor Hypofunction and Schizophrenia

There is substantial pharmacologic, genetic, and biochemical evidence to support NMDAR hypofunction as a key etiological component of schizophrenia. Dissociative anesthetics such as ketamine and phencyclidine (PCP), which are NMDAR channel blockers, were known since their introduction to produce the full range of schizophrenia symptoms and cognitive deficits in healthy subjects (Javitt and Zukin, 1991, Krystal et al., 1994). In healthy volunteers, low doses of ketamine that do not cause

Augmenting NMDA Receptor Function to Treat Schizophrenia

To this day, the major pharmacological treatment strategy for schizophrenia is based on a serendipitous discovery over 50 years ago of the antipsychotic effects of chlorpromazine (Delay & Deniker, 1955), which was initially developed as an antihistamine to reduce intraoperative autonomic stress. The discovery that these antipsychotic drugs increased dopamine turnover and that their clinical potency correlated with D2 receptor affinity, led to the development of 10 “first-generation” dopamine D2

Conclusion

Data from clinical, genetic, and animal model studies strongly implicate the NMDAR as a central hub for many of the pathological brain processes that occur in schizophrenia. It has been long known that the dissociative anesthetics and NMDAR antagonists, PCP and ketamine, produce the full range of schizophrenia symptoms and cognitive deficits in healthy subjects (Javitt and Zukin, 1991, Krystal et al., 1994), while ketamine exacerbates symptoms in stabilized schizophrenia patients (Lahti et al.,

Conflict of Interest

D.T.B. has no conflicts of interest to declare.

Acknowledgments

The National Institute of Mental Health supported this work (4R00MH099252-03). I would like to personally thank Dr. Coyle for being a great mentor during my postdoctoral fellowship and continuing to be a staunch advocate of my career.

References (158)

  • V.N. Foltyn et al.

    Serine racemase modulates intracellular d-serine levels through an alpha, beta-elimination activity

    The Journal of Biological Chemistry

    (2005)
  • P. Fusar-Poli et al.

    Neuroanatomy of vulnerability to psychosis: A voxel-based meta-analysis

    Neuroscience and Biobehavioral Reviews

    (2011)
  • P. Fusar-Poli et al.

    Progressive brain changes in schizophrenia related to antipsychotic treatment? A meta-analysis of longitudinal MRI studies

    Neuroscience and Biobehavioral Reviews

    (2013)
  • C. Gao et al.

    Modulation of behavior by scaffolding proteins of the post-synaptic density

    Neurobiology of Learning and Memory

    (2013)
  • J.R. Glausier et al.

    Dendritic spine pathology in schizophrenia

    Neuroscience

    (2013)
  • G. Gonzalez-Burgos et al.

    Alterations in cortical network oscillations and parvalbumin neurons in schizophrenia

    Biological Psychiatry

    (2015)
  • M. Goto et al.

    Crystal structure of a homolog of mammalian serine racemase from Schizosaccharomyces pombe

    The Journal of Biological Chemistry

    (2009)
  • P.L. Greer et al.

    From synapse to nucleus: Calcium-dependent gene transcription in the control of synapse development and function

    Neuron

    (2008)
  • K.B. Hansen et al.

    Distinct functional and pharmacological properties of triheteromeric GluN1/GluN2A/GluN2B NMDA receptors

    Neuron

    (2014)
  • K. Hashimoto et al.

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

    Progress in Neuro-Psychopharmacology & Biological Psychiatry

    (2005)
  • D. Inta et al.

    Mice with genetically altered glutamate receptors as models of schizophrenia: A comprehensive review

    Neuroscience and Biobehavioral Reviews

    (2010)
  • J.T. Kantrowitz et al.

    d-serine for the treatment of negative symptoms in individuals at clinical high risk of schizophrenia: A pilot, double-blind, placebo-controlled, randomised parallel group mechanistic proof-of-concept trial

    Lancet. Psychiatry

    (2015)
  • K.H. Karlsgodt et al.

    Reduced dysbindin expression mediates N-methyl-d-aspartate receptor hypofunction and impaired working memory performance

    Biological Psychiatry

    (2011)
  • E. Kartvelishvily et al.

    Neuron-derived d-serine release provides a novel means to activate N-methyl-d-aspartate receptors

    The Journal of Biological Chemistry

    (2006)
  • L.S. Kegeles et al.

    Modulation of amphetamine-induced striatal dopamine release by ketamine in humans: Implications for schizophrenia

    Biological Psychiatry

    (2000)
  • M.J. Kempton et al.

    Progressive lateral ventricular enlargement in schizophrenia: A meta-analysis of longitudinal MRI studies

    Schizophrenia Research

    (2010)
  • M.S. Keshavan et al.

    Changes in the adolescent brain and the pathophysiology of psychotic disorders

    Lancet. Psychiatry

    (2014)
  • A.C. Lahti et al.

    Effects of ketamine in normal and schizophrenic volunteers

    Neuropsychopharmacology

    (2001)
  • D.A. Lewis et al.

    Catching up on schizophrenia: Natural history and neurobiology

    Neuron

    (2000)
  • J.C. Leza et al.

    Inflammation in schizophrenia: A question of balance

    Neuroscience and Biobehavioral Reviews

    (2015)
  • D.T. Balu et al.

    Akt1 deficiency in schizophrenia and impairment of hippocampal plasticity and function

    Hippocampus

    (2012)
  • D.T. Balu et al.

    Multiple risk pathways for schizophrenia converge in serine racemase knockout mice, a mouse model of NMDA receptor hypofunction

    Proceedings of the National Academy of Sciences of the United States of America

    (2013)
  • D.T. Balu et al.

    An mGlu5-positive allosteric modulator rescues the neuroplasticity deficits in a genetic model of NMDA receptor hypofunction in schizophrenia

    Neuropsychopharmacology

    (2016, Jan 7)
  • D.T. Balu et al.

    d-serine and serine racemase are localized to neurons in the adult mouse and human forebrain

    Cellular and Molecular Neurobiology

    (2014)
  • A. Banerjee et al.

    Src kinase as a mediator of convergent molecular abnormalities leading to NMDAR hypoactivity in schizophrenia

    Molecular Psychiatry

    (2015)
  • A.C. Basu et al.

    Targeted disruption of serine racemase affects glutamatergic neurotransmission and behavior

    Molecular Psychiatry

    (2009)
  • J.E. Belforte et al.

    Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes

    Nature Neuroscience

    (2009)
  • M.A. Benneyworth et al.

    Cell selective conditional null mutations of serine racemase demonstrate a predominate localization in cortical glutamatergic neurons

    Cellular and Molecular Neurobiology

    (2012)
  • R. Bergeron et al.

    NAAG, NMDA receptor and psychosis

    Current Medicinal Chemistry

    (2012)
  • R. Bergeron et al.

    Modulation of N-methyl-d-aspartate receptor function by glycine transport

    Proceedings of the National Academy of Sciences of the United States of America

    (1998)
  • H. Betz et al.

    Glycine transporters: Essential regulators of synaptic transmission

    Biochemical Society Transactions

    (2006)
  • K. Biber et al.

    Expression and signaling of group I metabotropic glutamate receptors in astrocytes and microglia

    Journal of Neurochemistry

    (1999)
  • D. Boehning et al.

    Novel neural modulators

    Annual Review of Neuroscience

    (2003)
  • 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

    The American Journal of Psychiatry

    (2007)
  • A.G. Cardno et al.

    Heritability estimates for psychotic disorders: The Maudsley twin psychosis series

    Archives of General Psychiatry

    (1999)
  • G.C. Carlson et al.

    Dysbindin-1 mutant mice implicate reduced fast-phasic inhibition as a final common disease mechanism in schizophrenia

    Proceedings of the National Academy of Sciences of the United States of America

    (2011)
  • V.S. Catts et al.

    A quantitative review of the postmortem evidence for decreased cortical N-methyl-d-aspartate receptor expression levels in schizophrenia: How can we link molecular abnormalities to mismatch negativity deficits?

    Biological Psychology

    (2015, Nov 10)
  • M.P. Caulfield et al.

    International Union of Pharmacology. XVII. Classification of muscarinic acetylcholine receptors

    Pharmacological Reviews

    (1998)
  • K.H. Choi et al.

    Adjunctive pharmacotherapy for cognitive deficits in schizophrenia: Meta-analytical investigation of efficacy

    The British Journal of Psychiatry

    (2013)
  • J.J. Corrigan et al.

    The occurrence of certain d-amino acids in insects

    Biochemistry

    (1966)
  • Cited by (203)

    View all citing articles on Scopus
    View full text