Targeting of NMDA Receptors in New Treatments for Schizophrenia

Embed Size (px)

DESCRIPTION

Abnormalities in glutamatergic neurotransmission mediated by N-methyl-d-aspartate (NMDA) are implicated in the pathophysiology of schizophrenia, although the precise mechanisms are unknown.Read More: http://informahealthcare.com/doi/full/10.1517/14728222.2014.934225

Citation preview

  • 1. Introduction

    2. Alterations in the

    glutamine-glutamate-GABA

    cycle in schizophrenia

    3. Glycine modulatory site on the

    NMDA receptor as a

    therapeutic target

    4. Redox site of the NMDA

    receptor as a therapeutic

    target

    5. Conclusion

    6. Expert opinion

    Review

    Targeting of NMDA receptors innew treatments for schizophreniaKenji HashimotoChiba University Center for Forensic Mental Health, Division of Clinical Neuroscience, Chiba,

    Japan

    Introduction: Abnormalities in glutamatergic neurotransmission mediated by

    N-methyl-D-aspartate (NMDA) are implicated in the pathophysiology of

    schizophrenia, although the precise mechanisms are unknown.

    Areas covered: The author examines the role of the NMDA receptor in

    schizophrenia, focusing on results from preclinical and clinical studies that

    support the NMDA receptor hypothesis of schizophrenia. The author first

    reviewed papers detailing alterations in the levels of endogenous substances

    such as glutamine, glutamate, D-serine, L-serine, kynurenic acid and glutathi-

    one (GSH), all of which can affect NMDA receptor function. Next, the author

    reviewed clinical findings for glycine, D-serine, D-cycloserine, D-amino acid

    oxidase inhibitors (e.g., sodium benzoate) and glycine transporter-1 inhibitors

    (e.g., sarcosine, bitopertin), as potential therapeutic drugs. In addition, the

    author outlined how oxidative stress associated with decreased levels of the

    endogenous antioxidant GSH may play a role in the pathophysiology of

    schizophrenia. Finally, the author reviewed N-acetylcysteine (NAC), a precur-

    sor of GSH and an activator of the cystine--glutamate antiporter, as a potential

    therapeutic drug.

    Expert opinion: Given the NMDA receptor hypothesis of schizophrenia, the

    glycine modulatory site on NMDA receptors is the most attractive therapeutic

    target for this disease. In addition, both the kynurenine pathway and cystine-

    glutamate antiporter represent credible potential therapeutic targets for

    schizophrenia.

    Keywords: cystine-glutamate antiporter, D-serine, glutamate, glutathione, glycine,

    glycine transporter, kynurenic acid, N-acetylcysteine, NMDA receptor, sulforaphane

    Expert Opin. Ther. Targets (2014) 18(9):1049-1063

    1. Introduction

    Schizophrenia is a chronic, debilitating psychiatric disease, affecting ~ 1% of theworlds population. This disease shows varied and severe symptoms that generallybegin in late adolescence or early adulthood, and usually continue throughoutlife. Although the precise cause of schizophrenia is unknown, both genetic and envi-ronmental factors, such as place and time of birth, infection, prenatal events, andobstetric complications, are thought to contribute to disease pathophysiology [1].

    Glutamate (L-glutamic acid) plays an important role, as a well-established majorexcitatory neurotransmitter in the brain. The N-methyl-D-aspartate (NMDA)receptor is an ion-channel subtype of glutamate receptors and comprises differentsubunits responsible for varied biophysical and pharmacological properties (Figure 1).The NMDA receptor is composed of four subunits: two obligatory GluN1 subunitsand a combination of two GluN2 subunits (GluN2A, GluN2B, GluN2C andGluN2D) and/or GluN3 subunits (GluN3A and GluN3B) [2]. Multiple lines of evi-dence suggest that dysfunctional glutamatergic neurotransmission via the NMDAreceptor could underlie the pathophysiology of schizophrenia [3-11]. The NMDAreceptor hypothesis in schizophrenia was first proposed > 30 years ago [12]. This

    10.1517/14728222.2014.934225 2014 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 1049All rights reserved: reproduction in whole or in part not permitted

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • hypothesis evolved from the clinical findings that phencycli-dine (PCP) and its congener, ketamine, which blocks theNMDA receptor (Figure 1), induce a schizophrenia-likepsychosis with positive and negative symptoms, as well ascognitive impairment in normal healthy humans, and thatPCP exacerbates symptoms in patients with chronic schizo-phrenia [3,13]. Therefore, to date, NMDA receptor antagonismprovides the best pharmacological model of schizophrenia. Astudy using postmortem brain samples showed a decrease inthe obligatory GluN1 subunit at both mRNA and protein lev-els in the dorsolateral prefrontal cortex from schizophrenia [14],supporting the hypothesis of NMDA receptor hypofunction inschizophrenia. Further evidence comes from genetic associa-tion studies showing the influence of schizophrenia risk genes,such as D-amino acid oxidase (DAAO), G72, neuregulin 1 anddysbindin on NMDA receptor function [15,16].In this article, the author reviews recent data on the NMDA

    receptor as a new therapeutic target for schizophrenia.

    2. Alterations in the glutamine-glutamate-GABA cycle in schizophrenia

    In 1980, Kim et al. [17] reported decreased glutamate levels inthe cerebrospinal fluid (CSF) of patients with schizophreniaand proposed the glutamate hypothesis of schizophrenia.However, subsequent studies failed to replicate thesefindings [18,19]. Previously, we found an increased ratio ofglutamine to glutamate in the CSF of first episode anddrug-naive patients with schizophrenia, although individualCSF levels showed no differences between the two groups [20].In the brain, glutamine synthesis from glutamate and ammo-

    nia occurs exclusively in astrocytes. Glutamine plays major rolesin nitrogen and carbon homeostasis, in the detoxification ofammonia and as a precursor in the synthesis of glutamate inexcitatory neurons (Figure 2). Released glutamate is taken upby surrounding astrocytes via the glutamate transporter, whereit is converted to glutamine, transported back to presynapticneurons and reconverted to glutamate. Thus, the glutamine-glutamate cycle as part of glia-neuron communication playsan important role in excitatory neurotransmission [21]. In

    addition, the inhibitory amino acid, GABA, is also synthesizedfrom glutamate via glutamic acid decarboxylase (Figure 2) [21].Therefore, it is not unlikely that abnormalities in glutamine-glutamate-GABA cycling play a role in the pathophysiology ofschizophrenia [20-22]. A study using 3T 1H-magnetic resonancespectroscopy (MRS) demonstrated an increased ratio of gluta-mine to glutamate in the medial prefrontal cortex derivedfrom schizophrenic brains, although there was no difference inthe levels of individual amino acids between the patients andcontrols [23]. A more recent study using 3T 1H-MRS foundan increased ratio of glutamine to glutamate in the dorsalanterior cingulate cortex of schizophrenia brains [24]. Thesefindings support our earlier hypothesis on abnormalities inglutamine-glutamate cycling in the brains of schizophreniapatients. Amino acids, including glutamate, glutamine, glycine,L-serine and D-serine, are capable of modulating neurotransmis-sion via the NMDA receptor [25]. However, using postmortembrain samples from the Stanley Foundation, we found nochanges in these amino acids in the prefrontal cortex of patientswith schizophrenia [25].

    3. Glycine modulatory site on the NMDAreceptor as a therapeutic target

    Accepting the NMDA receptor hypofunction hypothesis forschizophrenia, increasing NMDA receptor function throughpharmacological manipulation could potentially be a newstrategy for managing schizophrenia. At present, thestrychnine-insensitive glycine modulatory site on the NMDAreceptor is the most used therapeutic target for improvingsymptoms such as positive and negative symptoms, andcognitive impairment in schizophrenia [7-10,26].

    3.1 GlycineGlycine is a well-established inhibitory neurotransmitter, viaits interaction with strychnine-sensitive glycine receptors.Glycine is also a full agonist at the strychnine-insensitiveglycine modulatory site associated with the NMDA receptor(Figure 1). Initial clinical studies with glycine were performedin the early 1990s. Glycine was found to ameliorate multipleschizophrenia symptoms (e.g., positive and negative symp-toms, cognitive impairment and depression) (Table 1) [27,28].However, glycine is extensively metabolized in the liver andshows poor distribution across the blood-brain barrier, neces-sitating large doses of glycine to effectively stimulate NMDAreceptor activation in the brain [9].

    Serine hydroxymethyltransferase 1 (Shmt1) is an enzyme inthe reversible interconversion between glycine and L-serine(Figure 2). Maekawa et al. [29] reported that levels of Shmt1mRNA in the dorsolateral prefrontal cortex of schizophreniabrains were significantly higher than in control brains, sug-gesting that Shmt1 is associated schizophrenia pathophysiol-ogy. A study of Shmt1-deficient mice proposed that thisgene could be involved in hippocampus neurogenesis andcognition [30]. Since Shmt1 impacts on NMDA receptor

    Article highlights.

    . The NMDA receptor plays an important role in thepathophysiology of schizophrenia.

    . Abnormalities in the glutamine-glutamate-GABA cycleare implicated in the pathophysiology of this disease.

    . Glycine modulatory sites on the NMDA receptor are veryattractive therapeutic targets.

    . The kynurenine pathway is also a new therapeutictarget for this disease.

    . Abnormalities in the synthesis and metabolism of theantioxidant glutathione are also implicated in thepathophysiology of this disease.

    This box summarizes key points contained in the article.

    K. Hashimoto

    1050 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • function via the glycine-L-serine-D-serine cycle (Figure 2),Shmt1 could also be a possible therapeutic target.

    3.2 D-CycloserineD-Cycloserine, a partial agonist at the NMDA receptor-associated strychnine-insensitive glycine modulatory site(Figure 1), is a less efficient NMDA receptor ligand, comparedwith endogenous full agonists, such as glycine and D-serine(Figure 1). At high doses, D-cycloserine acts as an antagonistby displacing more efficacious endogenous agonists, but atmoderate doses, D-cycloserine facilitates glutamatergic neuro-transmission via the NMDA receptor [9]. A retrospective studyand two meta-analyses show that glycine, D-serine and sarcosine(N-methylglycine) are more effective than D-cycloserine inimproving the overall psychopathology in medicated patientswith schizophrenia (Table 1) [27,28,31]. It is likely that D-cycloser-ine has a relatively narrow therapeutic window due to its partialagonist properties [9]. Recently, we reported that D-cycloserinemay act as a prodrug for D-serine, since brain levels of D-serineincreased after oral administration of D-cycloserine [32].

    3.3 DAAO inhibitorsD-Serine is synthesized by serine racemase (SRR) from L-ser-ine (Figure 2) and is a full agonist at the NMDA receptor-associated strychnine-insensitive glycine modulatory site(Figure 1) [9,33]. Studies using SRR knockout (KO) micehighlighted SRR as the major enzyme responsible for D-serineproduction in the adult brain [34-36]. Interestingly, reducedlevels of D-serine and a reduced ratio of D-serine to total serine

    have been demonstrated in both the blood and CSF ofpatients with schizophrenia [37-41]. Very recently, Lin et al.[42] reported higher plasma G72 (or DAAO activator) proteinlevels in medicated and drug-free patients with schizophrenia,suggesting that this protein could be a potential biomarker forschizophrenia. Moreover, abnormalities in genes such asDAAO and DAAO activator/G72, both of which regulateD-serine metabolism, have been reported [43-45]. These find-ings implicate aberrant synthesis and metabolism of D-serinein the pathophysiology of schizophrenia [9].

    A number of clinical studies have demonstrated a therapeu-tic benefit for D-serine (30 -- 120 mg/kg/day) in patients withschizophrenia (Table 1) [28,46-49]. Despite its clinical efficacy,orally administered D-serine is metabolized substantially byDAAO (Figure 2), diminishing its oral bioavailability. Higherdoses of D-serine raise the potential for nephrotoxicity,although no significant adverse events have as yet beenobserved at doses of up to 4 g/day [50]. We have in the pastreported that coadministration of the DAAO inhibitor poten-tiated the bioavailability of D-serine in mice, thereby enhanc-ing the efficacy of D-serine in animal models of schizophrenia[51,52]. Taken together, it is likely that coadministration ofD-serine and a DAAO inhibitor could represent a novelapproach in the treatment of schizophrenia [9,51-54].

    Recently, we reported that supplementation of D-serine inpreadolescent to adult stages may prevent the onset of psychosisin adult mice after neonatal inhibition of SRR [55], highlightingD-serine as an attractive therapeutic agent for early interventionin the onset of psychosis. A randomized, placebo-controlled

    Polyamine

    PCP receptor

    GluN2 GluN1

    Glutathione(GSH)

    GlutamateNMDA

    Ca2+, Na+

    GlycineD-SerineKynurenic acid

    PCPKetamine

    Ma2+

    Zn2+

    Figure 1. NMDA receptors in the brain. Phencyclidine (PCP) and ketamine are ion-channel blockers of the NMDA receptor.

    Glycine and D-serine are endogenous co-agonists of the glycine modulatory site on the GluN1 subunit. Kynurenic acid is an

    endogenous antagonist at glycine modulatory sites. D-Cycloserine is a partial agonist at glycine modulatory sites. Glutamate

    and NMDA are agonists at glutamate sites on the GluN2 subunit. Glutathione (GSH) and polyamine bind to the redox

    modulatory and polyamine sites of the NMDA receptor, respectively.

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1051

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • study of D-serine in subjects with schizophrenia prodrome isreported by Dr. Daniel Javitt [56]. They investigated the effectsof D-serine (16-week) on prodromal symptoms in 44 subjectsat high clinical risk for schizophrenia, based on StructuredInterview for Psychosis-Risk Syndromes/Scale of Psychosis-Risk Symptoms (SOPS) criteria. A highly significant reduction(p = 0.03, d = 0.68) was observed on SOPS negative symptoms.Furthermore, a > 20% improvement was observed in 9 out of10 subjects who completed all 16 weeks of treatment, comparedwith only 5 out of 11 placebo-treated subjects (p = 0.23). Twoplacebo-treated subjects and one D-serine-treated subjecttransitioned to psychosis during the study (Table 1) [56]. Thesedata suggested a potential for early intervention with D-serinein early stage schizophrenia, consistent with preclinicalreporting [55].

    3.4 Sodium benzoateSodium benzoate is the sodium salt of benzoic acid and isoften used as a food preservative because of its antimicrobial

    properties. Sodium benzoate is a competitive inhibitorof DAAO, which binds to the enzyme active site (Ki 16 M) [51,57]. A recent randomized, double-blind, placebo-controlled study demonstrated that benzoate (1 g/dayfor 6-week) significantly improved a variety of symptomdomains and cognition in patients with schizophrenia(Table 1) [58].

    DAAO shows very low activity in adult forebrains, withhigh activity in the cerebellum. Previously, we reported thatDAAO inhibition had no effect on D-serine levels in adultmouse forebrains [52]. Therefore, it is possible that the increasein cerebellar D-serine levels by DAAO inhibition may, in part,confer antipsychotic activity, by augmenting D-serine-medi-ated regulation of the NMDA receptor in the cerebellum [59].Nonetheless, further detailed studies on sodium benzoate inpreclinical models of schizophrenia are needed. A very recentreport shows that sodium benzoate upregulated brain-derivedneurotrophic factor (BDNF) [60]. This data imply that thetherapeutic effect of sodium benzoate is through increases in

    D-amino acid oxidaseD-Serine

    L-Serine

    3-Phosphoglyceratedehydrogenase

    3-Phosphoglycerate

    Glycine

    NH3

    NH3

    NH3

    Succinyl CoA

    Succinate

    Succinic semialdehydedehydrogenase

    Succinicsemialdehyde

    GABAtransaminase

    GABA

    Glutamic aciddecarboxylase

    Glutamatedehydrogenase

    L-GlutamateL-Glutamine

    L-Cysteine

    Glutathione synthetase

    Glutathione

    Glutaminase

    Glutamine synthetase

    -Keto-glutarate

    -Glutamylcysteine

    -Glutamylcysteine synthetase

    -Ketoglutaratedehydrogenase

    Serinehydroxymethyltransferase

    Serine racemase

    Figure 2. Amino acid synthetic and metabolic pathways. L-Glutamate, an excitatory amino acid, is synthesized from

    L-glutamine by glutaminase, and metabolized to L-glutamine by glutamine synthetase. In addition, L-glutamate is

    metabolized to GABA, an inhibitory amino acid, by glutamic acid decarboxylase. GABA is metabolized to succinic

    semialdehyde by GABA transaminase. D-Serine is synthesized from L-serine by serine racemase and is metabolized by D-amino

    acid oxidase. L-Serine is converted to glycine by serine hydroxymethyltransferase (SHMT). Glutathione is synthesized via

    g-glutamylcysteine from L-glutamate, L-cysteine and glycine.

    K. Hashimoto

    1052 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • BDNF levels, as BDNF is a known causative agent in thepathophysiology of some psychiatric diseases [61-63].

    3.5 Sarcosine (N-methylglycine)Glycine transporter-1 (GlyT-1), located on astrocytes and neu-rons, is responsible for glycine reuptake in forebrain areas, andin some regions, it may co-localize with glycinemodulatory siteson NMDA receptors [9,64]. Furthermore, GlyT-1 acts to main-tain local synaptic glycine at very low levels, suggestive of a rolein regulating glutamatergic neurotransmission via NMDAreceptors [64]. Previously, we reported that the GlyT-1 inhibitor,NFPS, improved cognitive deficits in rodents after administra-tion of NMDA receptor antagonists [65,66]. These findingsimply that increased synaptic glycine levels achieved throughinhibition of GlyT-1, lead to enhanced NMDA receptor activa-tion, in turn suggesting a potential role for GlyT-1 inhibition asa novel treatment for schizophrenia [9,64,67].

    Sarcosine (N-methylglycine) is generated by the enzymatictransfer of a methyl group from S-adenosylmethionine toglycine, and this reaction is catalyzed by the enzyme, glycineN-methyltransferase (Figure 3) [64]. Treatment with sarcosine(2 g/day) conferred beneficial effects in patients treated withantipsychotics, such as risperidone [68], but not clozapine [69].A randomized, double-blind, placebo-controlled study dem-onstrated that the sarcosine (2 g/day) group showed greaterreductions in Positive and Negative Syndrome Scale (PANSS)

    total scores than the placebo or D-serine (2 g/day) groups.This study proposes that sarcosine may offer superior resultsover D-serine, in that it benefits patients with long-term stableschizophrenia, as well as those in acute stages of the disease(Table 1) [48]. A randomized, double-blind study reportedthat sarcosine (2 g/day) alone was effective in the treatmentof acutely ill, symptomatic drug-free schizophrenics (Table 1)[70]. Additionally, a randomized, double-blind, placebo-controlled comparison study of sarcosine and D-serine dem-onstrated that sarcosine was superior to placebo in all fouroutcome measures of PANSS total, Scale for the Assessmentof Negative Symptoms, Quality of Life and Global Assess-ment of Functioning (GAF), while D-serine showed no signif-icant differences from placebo in these measures (Table 1) [71].Further large-scale, placebo-controlled, dose-finding studiesare needed to fully assess the beneficial effects of sarcosine,since the original data were based on small studies from a sin-gle group in Taiwan. Nonetheless, their findings suggest thatGlyT-1 inhibition could be a novel therapeutic target forenhancing NMDA receptor function [9,64,67].

    An epidemiological study showed an ~ 40 -- 50% lower riskfor prostate cancer in male patients with schizophrenia [72]. In2009, Sreekumar et al. [73] identified sarcosine as a biomarkerin urine, which is highly increased during prostate cancer pro-gression to metastasis. The enzyme glycine N-methyltransfer-ase is typically expressed at high levels in prostate. It would

    Table 1. Clinical trial of drugs in schizophrenics and subjects with prodromal syndrome.

    Drugs Dose and duration Clinical outcome Ref.

    Glycine Retrospective analysis andmeta-analysis

    Improved positive and negative symptoms, cognitiveimpairment and depression

    [27,28]

    D-Cycloserine Retrospective analysis andmeta-analysis

    Less effective than glycine, D-serine and sarcosine [27,28,31]

    D-Serine 30 -- 120 mg/kg/day Improved positive and negative symptoms and cognitiveimpairment

    [28,46-49]

    60 mg/kg/day and 16 weeks Improved SOPS negative symptoms score in subjects withprodromal syndrome

    [56]

    Sodium benzoate 1 g/day and 6 weeks Improved PANSS total, positive subscale, negative subscale,SANS, GAF, QOLS, CGI, HDRS and MATRICS composite score

    [58]

    Sarcosine 2 g/day and 6 weeks Improved PANSS total, negative subscale, SANS, QOL andGAF

    [68-71]

    Bitopertin 10, 30 or 60 mg/day and8 weeks (Phase IIb)

    Improved PANSS negative subscale [78]

    10, 30 mg/day and4 weeks (Phase III)

    No improvement of PANSS total in acute schizophrenia [81]

    5, 10 or 20 mg/day and24 weeks (Phase III)

    No improvement of PANSS negative subscale [79]

    N-acetylcysteine (NAC) 1 g/day and 6 weeks Improved PANSS total, negative subscale and CGI [122]2 g/day and 60 days Improved MMN, not P300, in patients with schizophrenia [123]2 g/day and 2 months Modulated EEG synchronization [125]2 g/day and 8 weeks Improved PANSS total and negative subscale [126]2 g/day and 12 weeks Improved BACS-J total score in subjects with at-risk mental

    state[128]

    BACS-J: Brief Assessment for Cognition in Schizophrenia Japanese version; CGI: Clinical global impression; GAF: Global assessment of function; HDRS: Hamilton

    depression rating scale - 17 items; MATRICS: Measurement And treatment research to improve cognition in schizophrenia; MMN: Mismatch negativity;

    PANSS: Positive and negative symptoms scale; QOL: Quality of life; QOLS: Quality of life scale; SANS: Scales for the assessment of negative symptoms - 20 items;

    SOPS: Scale of psychosis-risk symptoms.

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1053

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • therefore be of interest to study whether blood (or urine) lev-els of sarcosine are altered in male schizophrenics, since sarco-sine levels may contribute to disease pathophysiology. Takentogether, it seems that the glycine--sarcosine pathway mayinfluence the lower risk for prostate cancer in male schizo-phrenics although further detailed studies are necessary toconfirm this hypothesis [74].

    3.6 BitopertinThe selective and potent GlyT-1 inhibitor, RG1678 or bitoper-tin, was efficacious in in vivo studies using animal models ofschizophrenia, and oral administration produced a robust andsustained increase of extracellular glycine levels in ratstriatum [75,76]. A subsequent study showed increased CSFlevels of glycine in healthy volunteers after oral dosing with bito-pertin (3 -- 60 mg). Additionally, a positron emission tomogra-phy study found dose-dependent occupancy of GlyT-1 inhuman brains after oral administration of bitopertin [77].A Phase IIb multicenter, randomized, double-blind study of

    bitopertin (using 10, 30 or 60 mg/day for 8 weeks) showedthat it was effective against negative symptoms in stable, med-icated, schizophrenics (n = 323) (Table 1) [78]. Changes in thePANSS negative symptom factor score demonstrated a statisti-cally significant improvement in patient symptoms after takingbitopertin (10 and 30 mg) compared to placebo (Table 1) [78].On the other hand, bitopertin did not improve cognition inpatients, although GlyT-1 inhibitors improved cognitive

    deficits in an NMDA receptor hypofunction rodentmodel [65,66]. In January 2014, Roche reported negative resultsfrom two Phase III studies using bitopertin in schizophrenia(Table 1) [79]. Unexpectedly, the addition of bitopertin toantipsychotic therapy did not significantly reduce negativesymptoms at 24 week compared to placebo because of a highplacebo effect [79,80]. A randomized, double-blind, placebo-and active-controlled Phase II/III trial demonstrated thatbitopertin and olanzapine induced improved PANSS totalscores in patients with acute exacerbation of schizophrenia,although the changes were not statistically significant. How-ever, a greater proportion of patients receiving bitopertin(30 mg, 51.3%) or olanzapine (52.5%) compared with pla-cebo (32.9%) were ready for hospital discharge by week 4(Table 1) [81]. Further randomized, placebo-controlled studiesusing novel biomarkers to improve disease categorization andthus reduce the problem of patient heterogeneity are neededbefore the true efficacy of GlyT-1 inhibitors such as bitopertincan be ascertained [82].

    Although the reasons underlying the lack of efficacy forbitopertin in Phase III trials are currently unclear, it seems thatglycine performs a more complex role than simply binding tothe GluN1 subunit of NMDA receptors. As mentioned before,glycine is an inhibitory neurotransmitter through its interactionwith strychnine-sensitive glycine receptors [64]. One possibility isthat the increased glycine interacts with strychnine-sensitive gly-cine receptors, thereby attenuating the beneficial effects of

    NH2

    S

    O

    HO

    NH2

    SH

    OOH

    OHHO

    ONN

    H2N

    H2NN

    N

    O

    OH

    NH

    O

    OH

    NH2

    S

    OOH

    OHHO

    ONN

    H2N

    N

    N

    Glycine

    S-adenosylmethionine

    Methionine

    Sarcosine (N -methylglycine)

    S-adenosylhomocysteine

    GNMT

    Figure 3. Sarcosine synthesis pathway. Sarcosine (N-methylglycine) is generated by the enzymatic transfer of a methyl group

    from S-adenosylmethionine to glycine. This reaction is catalyzed by glycine N-methyltransferase (GNMT).

    K. Hashimoto

    1054 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • bitopertin. Another possibility may be an alteration of the gly-cine balance and activity of GlyT-1 and GlyT-2. GlyT-1 andGlyT-2 are two specific sodium/chloride-dependent high-affinity transporters, which regulate glycine synaptic levels inthe CNS. Finally, glycine is metabolized to L-serine byShmt1 (Figure 2) [29], and it is appropriate for this reaction tobe taken into consideration in this discussion.

    3.7 Kynurenic acidKynurenic acid is synthesized from the essential amino acid,L-tryptophan, via L-kynurenine, and is produced and releasedby astrocytes in the brain (Figure 4). Tryptophan 2,3-dioxyge-nase (TDO2) is an enzyme in the first step of the kynureninepathway, while kynurenine 3-monooxygenase (KMO) is arate-limiting enzyme at the branching point of this pathway(Figure 4) [83]. In addition to its well-characterized action asa competitive antagonist of glycine modulatory sites onNMDA receptors (Figure 1), kynurenic acid also acts as a non-competitive antagonist at a7 nicotinic acetylcholine receptors(nAChRs). Both of these receptors are already implicated inthe pathophysiology of schizophrenia (Figure 4) [84-86].

    Levels of kynurenic acid are increased in the CSF and brainsof patients with schizophrenia [87-90]. However, levels of3-hydroxykynurenine remained unchanged in the brain [89].Increased levels of TDO2 were reported in the anterior

    cingulate cortex of schizophrenia [91], whereas KMO geneexpression and KMO enzyme activity were significantlydecreased in postmortem brain samples from schizophrenics [92].

    Injections of L-kynurenine (100 mg/kg) increased extracel-lular levels of kynurenic acid (1500%) and induced cognitivedeficits in rats. Pretreatment with galantamine, a positivemodulator at a7 nAChRs, prevented these deficits [93].Increased kynurenic acid synthesis in the developing brainproduced long-lasting cognitive deficits in adult rats; however,these deficits could be restored to control levels by galant-amine [94]. Continuous administration of L-kynurenine dur-ing the prenatal period, but not adolescence, promotedcognitive deficits in adult rats. This implied that prenatalbut not adolescent exposure to L-kynurenine is sufficient tocause cognitive deficits in adulthood [95]. These findingssuggest that increased exposure to kynurenic acid duringdevelopment may result in cognitive deficits during adult-hood and that stimulation at a7 nAChRs could improve thesedeficits in rodents.

    These combined results highlight the possibility thatincreased levels of kynurenic acid play a causative role in thepathophysiology of schizophrenia. Considering the possibleinteraction between the NMDA receptor and a7 nAChRsin schizophrenia, it is also likely that the kynurenine pathwaycould be a new therapeutic target in this disease [83-86,96].

    L-Tryptophan

    L-Kynurenine

    Kynurenineaminotransferases

    3-Hydroxykynurenine

    Quinolinic acid

    Kynurenic acid

    7 nAch receptor

    NMDA receptor

    Serotonin5-Hydroxy

    L-Tryptophan

    KMO

    IDO TDOTPH

    N-Formylkynurenine

    Formidase

    Figure 4. L-Tryptophan in the serotonin and kynurenine pathways. The essential amino acid L-tryptophan is converted to

    5-hydroxy L-tryptophan by tryptophan hydroxylase (TPH), and this is metabolized to 5-hydroxytryptamine (5-HT: serotonin)

    by 5-hydroxytryptophan decarboxylase. The kynurenine pathway is initiated by the conversion of L-tryptophan to

    N-formylkynurenine by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). N-Formylkynurenine is

    metabolized to L-kynurenine by formidase. L-Kynurenine is metabolized to kynurenic acid by kynurenine aminotransferases.

    L-Kynurenine is also metabolized to 3-hydroxykynurenine by kynurenine 3-monooxygenase (KMO) and then to quinolinic

    acid. Kynurenic acid blocks both NMDA receptors and a7 nAChRs; both of which play a role in the pathophysiology ofschizophrenia.nAChRs: nicotinic acetylcholine receptors.

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1055

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • 4. Redox site of the NMDA receptor as atherapeutic target

    4.1 GlutathioneGlutathione (GSH; L-glutamyl-L-cysteinyl-glycine), the most

    abundant thiol present in mammalian cells, at concentrations

    of up to 12 mM is a nucleophilic scavenger and an enzyme-

    catalyzed antioxidant. It plays an important role in protecting

    the brain against reactive oxidative stress and harmful xenobi-

    otics [21,97,98]. GSH is synthesized in vivo in a two-step enzy-matic process by g-glutamylcysteine synthetase, therate-limiting enzyme that uses L-glutamate and L-cysteine as

    substrates to form dipeptide g-glutamylcysteine. This is thencombined with glycine in a reaction catalyzed by GSH

    synthetase to generate GSH (Figure 2). The balance of cellular

    synthesis and consumption of GSH are regulated by feedback

    inhibition of the g-glutamylcysteine synthetase reaction, bythe end product GSH (Figure 2) [21,97,98].GSH potentiates the NMDA receptor response to

    glutamate [99] by acting at the redox modulatory site(s)(Figure 1) [100]. Do et al. [101] reported that the CSF levels ofGSH in drug-free patients with schizophrenia were a signifi-cant 27% lower than controls. A 1H-MRS study using adouble-quantum coherence filter technique demonstratedthat the levels of GSH in the medial prefrontal cortex ofpatients were 52% lower than controls [101]. However, a sub-sequent 1H-MRS study using stimulated echo acquisitionmode (STEAM) detected no differences in GSH levels inthe anterior cingulate cortex between chronically medicatedpatients and healthy control subjects [102]. Using MEGA-PRESS (MEscher-GArwood-Point RE-Solved Spectroscopy)sequence, we found a negative correlation between GSH levelsin the prefrontal cortex and the severity of negative symptomsin patients with schizophrenia, although no differences ofGSH levels were detected in the two groups [103]. By contrast,medial temporal lobe GSH levels in first-episode patients withschizophrenia were 22% higher than the control group and nosignificant relationship was observed between GSH levels andthe PANSS total score [104]. It is clear that further studies areneeded as current MRS study data are inconsistent.Decreased plasma levels of GSH and antioxidant enzyme

    activities in both early and chronic schizophrenics have beenreported [105]. Furthermore, allelic variants of genes codingfor the key GSH synthesizing enzyme, glutathione cysteineligase modulatory (GCLM) and catalytic (GCLC) subunitsare associated with schizophrenia [106,107]. Polymorphisms oftrinucleotide repeats in the GCLC gene are associated withreduced enzyme activity and GSH levels [107]. These findingsimplicate dysfunctional regulation of GSH synthesis in thedisease pathophysiology of subjects with high-risk genotypes.It is therefore likely that abnormalities in the synthesis and

    metabolism of GSH are active in the pathophysiology ofschizophrenia, pinpointing this system as a possible newtherapeutic target [108-111].

    4.2 Effects of N-acetyl-L-cysteine in preclinical models

    of schizophreniaN-acetyl-L-cysteine (NAC) is the N-acetyl derivative of theamino acid, L-cysteine. L-cysteine is rapidly oxidized tocystine, a substrate of the cystine-glutamate antiporter, inthe pro-oxidant milieu of the brain (Figure 5). This antiportertransports cystine into cells with a one-to-one counter-transport of glutamate. Inside the cell, cystine is reduced toL-cysteine, the rate-limiting component of GSH synthesis(Figure 5) [112,113]. As NAC has the capacity to regulate bothcystine-glutamate antiporter activity and the biosynthesis ofGSH, it is a highly attractive candidate to offer therapeuticbenefit for a number of neuropsychiatric diseases [112].

    Previously, we reported that pretreatment with NAC attenu-ated behavioral abnormalities (e.g., hyperlocomotion andbehavioral sensitization) and dopaminergic neurotoxicity inrats after administration of the stimulant methamphetamine(METH) [114]. Furthermore, we reported that pretreatmentand subsequent treatment with NAC protected against dopami-nergic neurotoxicity in monkey striatum, after repeated doses ofMETH [115]. In addition, NAC reversed social withdrawalcaused by repeated dosing with the NMDA receptor antagonist,PCP, and this effect could be blocked by infusion of the cystine-glutamate antiporter inhibitor, (S)-4-carboxyphenylglycine,into the prefrontal cortex or systemic administration of thegroup II mGluR antagonist, LY341495 [116]. These findingsintimate that the effects of NAC require both cystine-glutamateantiporter and group II mGluR activation [116].

    The GCLM gene KO mice, a model for increased oxidativestress, showed a selective decrease of parvalbumin-immunoreactivity (PV-IR) interneurons in CA3 and thedentate gyrus (DG) of the ventral hippocampus, and a con-comitant reduction of b/g oscillations [117]. At the end of ado-lescence/early adulthood, PV-IR neurons show impairment asoxidative stress increases or cumulates selectively in CA3 andDG of the ventral hippocampus [117]. Interestingly, treatmentwith NAC from gestation was able to normalize most neuro-chemical alterations in GCLM KO mice to wild-type lev-els [118]. In addition, GCLM KO mice show delayedmaturation of PV interneurons, and any additional oxidativechallenge in preweaning or pubertal, but not in young adultstages, reduces the number of PV-IR interneurons. Again,these effects were preventable by NAC treatment [119].Recently, it has also been reported that treatment with NAC(60 or 120 mg/kg/day) during developmental daysP42-P70 prevented increased sensitivity to amphetamine insocial isolation-reared mice [120]. It is highly plausible thatNAC may represent a potentially useful medication to preventconversion to schizophrenia in at-risk subjects [121].

    4.3 Clinical findings for NAC in patients with

    schizophreniaNAC has been in clinical use for over 30 years as an antidote inparacetamol overdose, a mucolytic for chronic obstructive

    K. Hashimoto

    1056 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • pulmonary disease, a renal protectant in contrast-inducednephropathy and a therapeutic agent in the management ofHIV [112]. A randomized, multicenter, double-blind, placebo-controlled study showed that NAC (1 g/day for 24 weeks)improved the PANSS total score, PANSS negativesubscale score and Clinical Global Impression (CGI) scores(Table 1) [122]. Subsequent to this, Lavoie et al. [123] reportedthat treatment with NAC (2 g/day) improved impaired mis-match negativity (MMN) generation in schizophrenia patients,compared with the placebo group (Table 1). However, pretreat-ment with NAC (3 g) did not attenuate behavioral changes(such as psychotic-like symptoms, reductions in working mem-ory and sustained attention performance), impaired MMN andP300 event-related potentials in healthy subjects, after adminis-tration of ketamine (Table 1) [124]. A randomized, double-blind,placebo-controlled study demonstrated that NAC (2 g/day)modulated EEG synchronization in the brains of schizophrenics(Table 1) [125]. Additionally, a recent randomized, double-blind,placebo-controlled study demonstrated that augmentation withNAC (up to 2 g/day) promoted significantly greater improve-ment in PANSS total (p = 0.006) and negative subscale(p < 0.001) scores compared with the placebo group, indicatingthat NAC add-on therapy could be an effective augmentationstrategy for alleviating negative symptoms in schizophrenia(Table 1) [126]. These trials detected no significant differencesbetween NAC and placebo groups in the frequency of adverseeffects. A recent meta-analysis of 26 double-blind, randomizedcontrolled studies found that NAC (mean weighted effectsize = 0.45, n = 140) conferred significant beneficial effects in

    patients with schizophrenia [127]. An open study demonstratedthat NAC (2 g/day for 12 weeks) improved scores on theSchizophrenia Cognition Rating Scale (SCoRS) and Schizo-phrenia Quality of Life Scale -- Japanese version (SQLS-J), infour subjects with at-risk mental state (Table 1) [128], implyingthat NAC may prevent the onset of psychosis.

    It is likely that add-on of NAC could act as a therapeuticagent in schizophrenia, particularly since it is widely usedthroughout the world in a number of disorders [112,121,129].However, low bioavailability of NAC is an issue, necessitatingthe development of new compounds for the cysteine-glutamate antiporter or derivatives (e.g., NAC ethyl esterand NAC amide) of NAC [130,131].

    5. Conclusion

    This review provides an update on the therapeutic potential ofthe NMDA receptor in schizophrenia. The summarized dataseem to support the notion that abnormalities in variousendogenous substances able to impact on glutamatergicneurotransmission via the NMDA receptor may be implicatedin the pathophysiology of schizophrenia. Findings from anumber of preclinical and clinical studies put forward theNMDA receptor is a highly credible therapeutic target forthis disease. However, before the therapeutic potential oftargets based on the NMDA receptor hypothesis can beconfirmed, better categorization of this heterogenous diseaseis needed. This could be achieved by the use of novel diseasebiomarkers (e.g., blood levels of amino acids, oxidative

    Cystine

    Cystine-glutamateantiporter

    Cystine

    Extracellular

    Intracellular

    Glutathione (GSH)

    N-acetylcysteine(NAC)

    L-Glutamate

    L-Glutamate

    L-Cysteine

    L-Cysteine

    Figure 5. Regulation of the cystine-glutamate antiporter by N-acetylcysteine (NAC). Intracellular glutamate transports

    extracellularly via the cystine-glutamate antiporter, while extracellular cystine moves intracellularly via the same mechanism.

    NAC can increase extracellular L-cysteine levels, thereby activating L-glutamate release through the cystine-glutamate

    antiporter. In turn released L-glutamate can potentiate glutamatergic neurotransmission via ion-channel and metabotropic

    glutamate receptors. In the intracellular spaces, cystine can be reduced to L-cysteine, the rate-limiting component in the

    synthesis of the key endogenous antioxidant, glutathione (GSH). GSH is synthesized from three amino acids, L-glutamate,

    L-cysteine and glycine. Thus, administration of NAC can increase availability of L-cysteine, which in turn facilitates the

    cystine-glutamate antiporter and the production of GSH in cells. This GSH can then potentiate NMDA receptor function.

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1057

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • stress markers, inflammatory cytokines, neurophysiologicalmeasures, cognitive measures and gene analysis) [86,132-136].

    6. Expert opinion

    Schizophrenia is a chronic, debilitating psychiatric diseasethat affects ~ 1% of the worlds general population and is a lead-ing cause of disability. Despite recent advances in the psycho-pharmacological treatment of this disease, many patientsremain refractory to therapy or with residual symptoms, includ-ing negative symptoms (e.g., social withdrawal, lack of motiva-tion and reduced emotional reactivity) and cognitiveimpairment (e.g., processing speed and attention deficits, work-ing memory problems). Current antipsychotic drugs have a lim-ited impact on negative symptoms and cognitive impairment inschizophrenia. Added to this are treatment-resistant patientswho are largely resistant to current antipsychotic medications.A number of preclinical and clinical findings suggest

    involvement of the NMDA receptor in the development ofnegative symptoms and cognitive impairment, since thisreceptor mediates the release of neurotransmitters, such asdopamine, glutamate, acetylcholine and GABA. Two recentPhase III studies found that addition of the GlyT-1 inhibitor,bitopertin, to antipsychotic therapy did not significantlyreduce prominent negative symptoms at 24 weeks, comparedwith placebo [79,80]. In addition, pomaglumetad methionil, ametabotropic glutamate mGluR2/3 agonist developed onthe basis of the glutamate hypothesis of schizophrenia, failedto significantly alleviate prominent negative symptoms inmedicated patients with schizophrenia [137,138]. On August29, 2012, Eli Lilly and Co. announced their decision tostop ongoing clinical studies investigating pomaglumetadmethionil in schizophrenia, due to a failure to meet the pri-mary end point [139]. Despite a number of preclinical modelsand translational efforts, clinical studies on these two drugshave not produced positive results. Since schizophrenia is aheterogenous disease, further randomized, placebo-controlledstudies using novel biomarkers and improved diagnoses, suchas Research Domain Criteria (RDoC) [140], will be necessaryto confirm the efficacy of new drugs developed in keepingwith the glutamate hypothesis of schizophrenia.Schizophrenia is typically diagnosed between the ages

    16 and 25, indicating that onset of disease could be preventedby early intervention. Aversion of psychosis has become amajor objective in the field of clinical psychiatry. Approxi-mately one-third of subjects at high risk develop psychosiswithin 3 years, and most are diagnosed with schizophre-nia [141]. Epidemiological studies suggest that improvingnutrition and avoiding infection during pregnancy may pre-vent some cases of this illness [142]. For example, supplemen-tation with the correct amount of vitamin D in early lifecould decrease the risk of schizophrenia [141,142]. Interestingly,supplementation of omega-3 fatty acids in the form of fishoils could decrease the onset of psychosis for high-risk sub-jects, since omega-3 fatty acids are integral components of

    neuronal membranes, which also act to dampen inflammationand oxidative stress [141,143].

    Cognitive impairment is a core feature of schizophrenia, oftenpersisting even when positive symptoms (e.g., hallucinations,delusions and suspiciousness) have been treated successfully.A recent meta-analysis showed that adolescents and young adultsat high-risk for developing psychosis demonstrated cognitiveimpairment before the onset of psychosis [144]. Both preclinicaland clinical studies found that cognitive impairment could beimproved by current off-label drugs or supplements [145-149].As described above, the antioxidant NAC has the potential toprevent psychosis, although oral bioavailability of NAC ispoor. In a previous preclinical study, we reported that supple-mentation with D-serine during juvenile and preadolescentstages could prevent the onset of psychosis at the adult stage [55].A natural conclusion would be that supplementation with theendogenous, and therefore safe D-serine, could protect adoles-cents and young adults at high-risk of developing psychosis.

    Sulforaphane (SFN) is obtained from cruciferous vegetables,such as broccoli and Brussels sprouts. SFN produces potent anti-oxidant and anti-inflammatory effects via the nuclear factorE2-related factor 2 (Nrf2)/Keap1 system. Previously, we reportedthat SFN conferred antipsychotic benefits in rodent models ofschizophrenia [150,151]. Interestingly, we found that supplementa-tion with SFN during juvenile stages could prevent the onset ofcognitive impairment in the preclinical PCP model of schizo-phrenia [152]. Therefore, supplementation with SFN-rich broc-coli sprout extract during childhood and pre-adolescence mayprevent the onset of psychosis in adulthood, assisted by the factthat this extract is widely available as a supplement.

    These findings suggest that early intervention with safe sup-plements could improve cognitive impairment in adolescentsand young adults, and possibly prevent the onset of schizo-phrenia. Finally, we would like to propose a hypothesis thatnutrition in childhood and preadolescent stages could have ahigh impact on subsequent mental health at adolescent andadult stages, particularly the adequate intake of omega-3 fattyacid-rich fish and vegetables (e.g., SFN-rich broccoli sprout),and possibly prevent the onset of psychiatric diseases.

    Acknowledgments

    The author would like to thank the collaborators who workedon some papers described in this review article, and who arelisted as the co-authors of our papers in the reference list.

    Declaration of interest

    This study was supported by a Grant-in-Aid for ScientificResearch on Innovative Areas of the Ministry of Education,Culture, Sports, Science and Technology, Japan. The authorhas no other relevant affiliations or financial involvementwith any organization or entity with a financial interest in orfinancial conflict with the subject matter or materialsdiscussed in the manuscript apart from those disclosed.

    K. Hashimoto

    1058 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://informahealthcare.com/journal/ETT

  • BibliographyPapers of special note have been highlighted as

    either of interest () or of considerable interest() to readers.1. Sullivan PF. The genetics of

    schizophrenia. PLoS Med 2005;2:e212

    2. Traynelis SF, Wollmuth LP, McBain CJ,

    et al. Glutamate receptor ion channels:

    structure, regulation, and function.

    Pharmacol Rev 2010;62:405-96

    3. Javitt DC, Zukin SR. Recent advances in

    the phencyclidine model of

    schizophrenia. Am J Psychiatry

    1991;148:1301-8

    4. Coyle JT. The glutamatergic dysfunction

    hypothesis for schizophrenia.

    Harv Rev Psychiatry 1995;3:241-53

    5. Olney JW, Farber NB. Glutamate

    receptor dysfunction and schizophrenia.

    Arch Gen Psychiatry 1995;52:998-1007

    6. Krystal JH, DSouza DC, Petrakis IL,

    et al. NMDA agonists and antagonists as

    probes of glutamatergic dysfunction and

    pharmacotherapies in neuropsychiatric

    disorders. Harv Rev Psychiatry

    1999;7:125-43

    7. Hashimoto K, Okamura N, Shimizu E,

    et al. Glutamate hypothesis of

    schizophrenia and approach for possible

    therapeutic drugs. Curr Med Chem -

    CNS Agents 2004;4:147-54

    8. Hashimoto K. The NMDA receptor

    hypofunction hypothesis for

    schizophrenia and glycine modulatory

    sites on the NMDA receptors as

    potential therapeutic drugs.

    Clin Psychopharmacol Neurosci

    2006;4:3-10

    9. Hashimoto K, Malchow B, Falkai P,

    et al. Glutamate modulators as potential

    therapeutic drugs in schizophrenia and

    affective disorders. Eur Arch Psychiatry

    Clin Neurosci 2013;263:367-77

    10. Javitt DC, Schoepp D, Kalivas PW,

    et al. Translating glutamate: from

    pathophysiology to treatment.

    Sci Transl Med 2011;3:102mr2

    11. Moghaddam B, Javitt D. From

    revolution to evolution: the gluatamate

    hypothesis of schizophrenia and its

    implication for treatment.

    Neuropsychopharmacology 2012;37:4-15

    12. Coyle JT. NMDA receptor and

    schizophrenia: a brief history.

    Schizophr Bull 2012;38:920-6

    13. Krystal JH, Karper LP, Seibyl JP, et al.

    Subanesthetic effects of the noncompetitive

    NMDA antagonist, ketamine, in humans.

    Psychotomimetic, perceptual, cognitive,

    and neuroendocrine responses.

    Arch Gen Psychiatry 1994;51:199-214

    14. Weickert CS, Fung SJ, Catts VS, et al.

    Molecular evidence of N-methyl-D-

    aspartate receptor hypofunction in

    schizophrenia. Mol Psychiatry

    2013;18:1185-92

    15. Harrison PJ, Owen MJ. Genes for

    schizophrenia? Recent findings and their

    pathophysiological implications. Lancet

    2003;361:417-19

    16. Schwartz TL, Sachdeva S, Stahl SM.

    Genetic data supporting the

    NMDA glutamate receptor hypothesis

    for schizophrenia. Curr Pharm Des

    2012;18:1580-92

    17. Kim JS, Kornhuber HH, Holzmuller B,

    et al. Reduction of cerebrospinal fluid

    glutamic acid in Huntigtons chorea and

    in schizophrenic patients.

    Arch Psychiatr Nervenkr 1980;228:7-10

    18. Gattaz WF, Gattaz D, Beckmann H.

    Glutamate in schizophrenics and healthy

    controls. Arch Psychiatr Nervenkr

    1982;231:221-5

    19. Perry TL. Normal cerebrospinal fluid

    and brain glutamate levels in

    schizophrenia do not support the

    hypothesis of glutamatergic neuronal

    dysfunction. Neurosci Lett 1982;28:81-5

    20. Hashimoto K, Engberg G, Shimizu E,

    et al. Elevated glutamine/glutamate ratio

    in cerebrospinal fluid of first episode and

    drug naive schizophrenic patients.

    BMC Psychiatry 2005;5:6

    .. This is the first study showing

    abnormalities in glutamine--glutamate

    cycle in schizophrenia.

    21. Hashimoto K, Shimizu E, Iyo M.

    Dysfunction of glia-neuron

    communication in pathophysiology of

    schizophrenia. Curr Psychiatry Rev

    2005;1:151-63

    22. Hashimoto K. Abnormalities of the

    glutamine-glutamate-GABA cycle in the

    schizophrenia brain. Schizophr Res

    2014;156:281-2

    23. Shirayama Y, Obata T, Matsuzawa D,

    et al. Specific metabolites in the medial

    prefrontal cortex are associated with the

    neurocognitive deficits in schizophrenia:

    a preliminary study. Neuroimage

    2010;49:2783-90

    24. Bustillo JR, Chen H, Jones T, et al.

    Increased glutamine in patients

    undergoing long-term treatment for

    schizophrenia. A proton magnetic

    resonance spectroscopy study at 3T.

    JAMA Psychiatry 2014;71:265-72

    25. Hashimoto K, Sawa A, Iyo M. Increased

    levels of glutamate in brains from

    patients with mood disorders.

    Biol Psychiatry 2007;62:1310-16

    26. Coyle JT, Tsai G. The NMDA receptor

    glycine modulatory site: a therapeutic

    target for improving cognition and

    reducing negative symptoms in

    schizophrenia. Psychopharmacology (Berl)

    2004;174:32-8

    27. Heresco-Levy U, Javitt DC. Comparative

    effects of glycine and D-cycloserine on

    persistent negative symptoms in

    schizophrenia: a retrospective analysis.

    Schizophr Res 2004;66:89-96

    28. Tsai GE, Lin PY. Strategies to enhance

    N-methyl-D-aspartate receptor-mediated

    neurotransmission in schizophrenia.

    A critical review and meta-analysis.

    Curr Pharm Des 2010;16:522-37

    29. Maekawa M, Ohnishi T, Hashimoto K,

    et al. Analysis of strain-dependent

    prepulse inhibition points to a role for

    Shmt1 (SHMT1) in mice and in

    schizophrenia. J Neurochem

    2010;115:1374-85

    . This is the first study showing

    abnormalities in Shmt1 in

    schizophrenia.

    30. Abarinov EV, Beaudin AE, Field MS,

    et al. Disruption of shmt1 imapirs

    hippocampus neurogenesis and

    mnemonic function in mice. J Nutr

    2013;143:1028-35

    31. Tuominen HJ, Tiihonen J, Wahlbeck K.

    Glutamatergic drugs for schizophrenia:

    a systematic review and meta-analysis.

    Schizophr Res 2005;72:225-34

    32. Horio M, Mori H, Hashimoto K. Is D-

    cycloserine a prodrug for D-serine in the

    brain? Biol Psychiatry 2013;73:e33-4

    33. Wolosker H. Serine racemase and the

    serine shuttle between neurons and

    astrocytes. Biochem Biophys Acta

    2011;1814:1558-66

    34. Inoue R, Hashimoto K, Mori H.

    NMDA- and beta-amyloid1-42-induced

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1059

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://www.ncbi.nlm.nih.gov/pubmed/16033310?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16033310?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20716669?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20716669?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/1654746?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/1654746?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/1654746?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/7492260?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/7492260?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10483932?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10483932?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10483932?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10483932?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23455590?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23455590?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23455590?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21957170?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21957170?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21956446?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21956446?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21956446?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21956446?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8122957?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8122957?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8122957?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8122957?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23070074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23070074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23070074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12573388?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12573388?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12573388?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22280435?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22280435?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22280435?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6104477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6104477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6104477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6104477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6123303?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6123303?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6121307?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6121307?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6121307?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6121307?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/6121307?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24811432?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24811432?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24811432?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19850131?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19850131?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19850131?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19850131?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24402128?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24402128?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24402128?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24402128?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17574216?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17574216?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17574216?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15205876?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15205876?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15205876?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15205876?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15205876?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15061240?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15061240?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15061240?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15061240?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19909229?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19909229?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19909229?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19909229?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20977478?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20977478?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20977478?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20977478?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23700346?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23700346?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23700346?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15560967?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15560967?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22877923?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22877923?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22877923?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21224019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21224019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21224019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19118183?dopt=Abstracthttp://informahealthcare.com/journal/ETT

  • neurotoxicity is attenuated in serine

    racemase knock-out mice. J Neurosci

    2008;28:14486-91

    35. Horio M, Kohno M, Fujita Y, et al.

    Levels of D-serine in the brain and

    peripheral organs of serine racemase (Srr)

    knock-out mice. Neurochem Int

    2011;59:853-9

    36. Horio M, Kohno M, Fujita Y, et al.

    Role of serine racemase in behavioral

    sensitization in mice after repeated

    administration of methamphetamine.

    PLoS One 2012;7:e35494

    37. Hashimoto K, Fukushima T, Shimizu E,

    et al. Decreased serum levels of D-serine

    in patients with schizophrenia: evidence

    in support of the NMDA receptor

    hypofunction hypothesis of

    schizophrenia. Arch Gen Psychiatry

    2013;60:572-6

    .. This is the first study showing

    decreased serum levels of D-serine in

    patients with schizophrenia.

    38. Yamada K, Ohnishi T, Hashimoto K,

    et al. Identification of multiple serine

    racemase (SRR) mRNA isoforms and

    genetic analyses of SRR and DAO in

    schizophrenia and D-serine levels.

    Biol Psychiatry 2005;57:1493-503

    39. Hashimoto K, Engberg G, Shimizu E,

    et al. Reduced D-serine to total serine

    ratio in the cerebrospinal fluid of drug

    naive schizophrenic patients.

    Prog Neuropsychopharmacol

    Biol Psychiatry 2005;29:767-9

    . This is the first study showing a

    decreased ratio of D-serine to total

    serine in cerebrospinal fluid (CSF)

    of schizophrenia.

    40. Bendikov I, Nadri C, Amar S, et al.

    A CSF and postmortem brain study of

    D-serine metabolic parameters in

    schizophrenia. Schizophr Res

    2007;90:41-51

    41. Calcia MA, Madeira C, Alheira FV,

    et al. Plasma levels of D-serine in

    Brazillian individuals with schizophrenia.

    Schizophr Res 2012;142:83-7

    42. Lin CH, Chang HT, Chen YJ, et al.

    Distinctively higher plasma G72 protein

    levels in patients with schizophrenia than

    in the healthy individuals.

    Mol Psychiatry 2014;19:636-7

    43. Chumakov I, Blumenfeld M,

    Guerassimenko O, et al. Genetic and

    physiological data implicating the new

    human gene G72 and the gene for D-

    amino acid oxidase in schizophrenia.

    Proc Natl Acad Sci USA

    2002;99:13675-80

    .. This is the first study showing G72 as

    a susceptible gene for schizophrenia.

    44. Shinkai T, De Luca V, Hwang R, et al.

    Association analyses of the DAOA/

    G30 and D-amino-acid oxidase genes in

    schizophrenia: further evidence for a role

    in schizophrenia. Neuromolecular Med

    2007;9:169-77

    45. Verrall L, Burnet PW, Betts JF, et al.

    The neurobiology of D-amino acid

    oxidase and its involvement in

    schizophrenia. Mol Psychiatry

    2010;15:122-37

    46. Tsai G, Yang P, Chung LC, et al.

    D-serine added to antipsychotics for the

    treatment of schizophrenia.

    Biol Psychiatry 1998;44:1081-9

    47. Heresco-Levy U, Javitt DC, Ebstein R,

    et al. D-serine efficacy as add-on

    pharmacotherapy to risperidone and

    olanzapine for treatment-refractory

    schizophrenia. Biol Psychiatry

    2005;57:577-85

    48. Lane HY, Chang YC, Liu YC, et al.

    Sarcosine or D-serine add-on treatment

    for acute exacerbation of schizophrenia:

    a randomized, double-blind, placebo-

    controlled study. Arch Gen Psychiatry

    2005;62:1196-204

    49. Kantrowitz JT, Malhotra AK,

    Cornblatt B, et al. High dose D-serine in

    the treatment of schizophrenia.

    Schizophr Res 2010;121:125-30

    50. Javitt DC, Zukin SR, Heresco-levy U,

    et al. Has an angel shown the way?

    Etiological and therapeutic implications

    of the PCP/NMDA model of

    schizophrenia. Schizophr Bull

    2012;38:958-66

    51. Ferraris D, Duvall B, Ko YS, et al.

    Synthesis and biological evaluation of D-

    amino acid oxidase inhibitors.

    J Med Chem 2008;51:3357-9

    52. Hashimoto K, Fujita Y, Horio M, et al.

    Co-administration of D-amino acid

    oxidase inhibitor potentiates the efficacy

    of D-serine on prepulse inhibition deficits

    after administration of dizocilpine.

    Biol Psychiatry 2009;65:1103-6

    . This is the first study showing efficacy

    of a combination of D-serine and

    D-amino acid oxidase inhibitor in an

    animal model of schizophrenia.

    53. Ferraris DV, Tsukamoto T. Recent

    advances in the discovery of D-amino

    acid oxidase inhibitors and their

    therapeutic utility in schizophrenia.

    Curr Pharm Des 2011;17:103-11

    54. Sacchi S, Rosini E, Pollegioni L, et al.

    D-amino acid oxidase inhibitors as a novel

    class of drugs for schizophgrenia therapy.

    Curr Pharm Des 2013;19:2499-511

    55. Hagiwara H, Iyo M, Hashimoto K.

    Neonatal disruption of serine racemase

    causes schizophrenia-like behavioral

    abnormalities in adulthood: clinical rescue

    by D-serine. PLoS One 2013;8:e62438

    56. Javitt DC. Glutamatergic based early

    intervention in schizophrenia. In the

    symposium Schizophrenia: could it be

    prevented? A Satellite Meeting of the

    2014 Annual Meeting of The Society of

    Biological Psychiatry; 7 May 2014; New

    York, USA

    .. This is the first study showing early

    intervention by D-serine in subjects

    with prodromal symptoms.

    57. Hashimoto K. Food coloring, sodium

    benzoate preservative and D-serine:

    implication for behavior. In: Preedy VR,

    editor. The handbook of behavior, diet

    and nutrition. Springer Publishers,

    New York; 2011. p. 577-84

    58. Lane HY, Lin CH, Green MF, et al.

    Add-on treatment of benzoate for

    schizophrenia: a randomized, double-

    blind, placebo-controlled trial of

    D-amino acid oxidase inhibitor.

    JAMA Psychiatry 2013;70:1267-75

    .. This is the first study showing

    antipsychotic activity of sodium

    benzoate in patients with schizophrenia.

    59. Hashimoto K. Comments on The effect

    of risperidone on D-amino acid oxidase

    activity as a hypothesis for a novel

    mechanism of action in the treatment of

    schizophrenia. J Psychopharmacol

    2010;24:1133-4

    60. Jana A, Modi KK, Roy A, et al.

    Up-regulation of neurotrophic factors by

    cinnamon and its metabolite sodium

    benzoate: therapeutic implications for

    neurodegenerative disorders.

    J Neuroimmune Pharmacol

    2013;8:739-55

    61. Hashimoto K, Shimizu E, Iyo M.

    Critical role of brain-derived

    neurotrophic factor in mood disorders.

    Brain Res Brain Res Rev 2004;45:104-14

    62. Hashimoto K. BDNF variant linked to

    anxiety-related behaviors. Bioessays

    2007;29:116-19

    K. Hashimoto

    1060 Expert Opin. Ther. Targets (2014) 18(9)

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://www.ncbi.nlm.nih.gov/pubmed/19118183?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19118183?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21906644?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21906644?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21906644?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22530033?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22530033?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22530033?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475930?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475930?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475930?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475930?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475930?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15953485?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15953485?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15953485?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15953485?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15939521?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15939521?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15939521?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17156977?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17156977?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17156977?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23063707?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23063707?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23857119?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23857119?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23857119?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12364586?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12364586?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12364586?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12364586?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17627036?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17627036?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17627036?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17627036?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19786963?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19786963?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19786963?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/9836012?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/9836012?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15780844?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15780844?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15780844?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15780844?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16275807?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16275807?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16275807?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16275807?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20541910?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20541910?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22987851?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22987851?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22987851?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22987851?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/18507366?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/18507366?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19217074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19217074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19217074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19217074?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21361869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21361869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21361869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21361869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23116391?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23116391?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23630632?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23630632?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23630632?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23630632?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24089054?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24089054?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24089054?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24089054?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19939869?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475543?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475543?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475543?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23475543?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15145621?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15145621?dopt=Abstracthttp://informahealthcare.com/journal/ETT

  • 63. Hashimoto K. Sigma-1 receptor

    chaperone and brain-derived

    neurotrophic factor: emerging links

    between cardiovascular disease and

    depression. Prog Neurobiol

    2013;100:15-29

    64. Hashimoto K. Glycine transportor-1:

    a potential therapeutic target for

    schizophrenia. Curr Pharm Des

    2011;17:112-20

    65. Hashimoto K, Fujita Y, Ishima T, et al.

    Phencyclidine-induced cognitive deficits

    in mice are improved by subsequent

    subchronic administration of glycine

    transporter-1 inhibitor NFPS and

    D-serine. Eur Neuropsychopharmacol

    2008;18:414-21

    66. Karasawa J, Hashimoto K, Chaki S.

    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;186:78-83

    67. Javitt DC. Glycine transport inhibitors in

    the treatment of schizophrenia.

    Handb Exp Pharmacol 2012;213:367-99

    68. Tsai G, Lane HY, Yang P, et al. Glycine

    transporter I inhibitor, N-methylglycine

    (sarcosine), added to antipsychotics for

    the treatment of schizophrenia.

    Biol Psychiatry 2004;55:452-6

    69. Lane HY, Huang CL, Wu PL, et al.

    Glycine transporter 1 inhibitor,

    N-methylglycine (sarcosine), added to

    clozapine for the treatment of

    schizophrenia. Biol Psychiatry

    2006;60:645-9

    70. Lane HY, Liu YC, Huang CL, et al.

    Sarcosine (N-methylglycine) treatment

    for acute schizophrenia: a randomized,

    double-blind study. Biol Psychiatry

    2008;63:9-12

    71. Lane HY, Lin CH, Huang YJ, et al.

    A randomized, double-blind, placebo-

    controlled comparison study of sarcosine

    (N-methylglycine) and D-serine add-on

    treatment for schizophrenia.

    Int J Neuropsychopharmacol

    2010;13:451-60

    72. Mortensen PB. The incidence of cancer

    in schizophrenic patients. J Epidemiol

    Community Health 1989;43:43-7

    73. Sreekumar A, Poisson LM,

    Rajendiran TM, et al. Metabolomic

    profiles delineate potential role for

    sarcosine in prostate cancer progression.

    Nature 2009;457:910-14

    74. Hashimoto K. Sarcosine and decreased

    risk for prostate cancer in schizophrenia.

    Open Clin Chem J 2009;2:22-3

    75. Pinard E, Alanine A, Alberati D, et al.

    Selective GlyT1 inhibitors: discovery of [4-

    (3-fluoro-5-trifluoromethylpyridin-2- yl)

    piperazin-1-yl][5-methanesulfonyl-2-((S)-

    2,2,2-trifluoro-1-methylethoxy)phenyl]

    methanone (RG1678), a promising novel

    medicine to treat schizophrenia.

    J Med Chem 2010;53:4603-14

    76. Alberati D, Moreau JL, Lengyel J, et al.

    Glycine reuptake inhibitor RG1678:

    a pharmacologic characterization of an

    investigational agent for the treatment of

    schizophrenia. Neuropharmacology

    2012;62:1152-61

    77. Martin-Facklam M, Pizzagalli F, Zhou Y,

    et al. Glycine transporter type 1

    occupancy by bitopertin: a positron

    emission tomography study in healthy

    volunteers. Neuropsychopharmacology

    2013;38:504-12

    78. Umbricht D, Alberati D,

    Martin-Facklam M, et al. Effect of

    bitopertin, a glycine reuptake inhibitor,

    on negative symptoms of schizophrenia.

    A randomized, double-blind,

    proof-of-concept study. JAMA Psychiatry

    2014;71:637-46

    79. Roche Media Release (January 21, 2014):

    roche provides update on the first two of

    six phase III studies of bitopertin in

    schizophrenia. 2014. Available from:

    http://www.roche.com/media/

    media_releases/med-cor-2014-01-21.htm

    80. Bugarski-Kirola D. Efficacy and safety of

    adjunctive bitopertin versus placebo in

    subjects with persistent predominant

    negative symptoms of schizophrenia

    treated with antipsychotic -- Update from

    the serachlyte programme. The 4th

    Schizophrenia International Resaerch

    Society Conference; 06 April 2014;

    Florence, Italy

    81. Bugarski-Kirola D, Wang A, Abi-Saab D,

    et al. A phase II/III trial of bitopertin

    monotherapy compared with placebo in

    patients with an acute exacerbation of

    schizophrenia - results from the CandleLyte

    study. Eur Neuropsychopharmacol

    2014;27:1024-36

    82. Goff DC. Bitopertin. The good news

    and bad news. JAMA Psychiatry

    2014;71:621-2

    83. Schwarcz R, Bruno JP, Muchowski PJ,

    et al. Kynurenines in the mammalian

    brain: when physiology meets pathology.

    Nat Rev Neurosci 2012;13:465-77

    84. Hashimoto K, Koike K, Shimizu E, et al.

    alpha7 Nicotinic receptor agonists as

    potential therapeutic drugs for

    schizophrenia. Curr Med Chem

    CNS Agents 2005;5:171-84

    85. Toyohara J, Hashimoto K.

    alpha7 Nicotinic receptor agonists:

    potential therapeutic drugs for treatment

    of cognitive impairments in

    schizophrenia and Alzheimers disease.

    Open Med Chem J 2010;4:37-56

    86. Hashimoto K. Targeting of

    alpha7 nicotinic acetylcholine receptor in

    the treatment of schizophrenia and the

    use of auditory sensory gating as a

    translational biomarker. Curr Pham Des

    In press

    87. Erhardt S, Blennow K, Nordin C, et al.

    Kynurenic acid levels are elevated in the

    cerebrospinal fluid of patients with

    schziophrenia. Neurosci Lett

    2001;313:96-8

    .. This is the first study showing

    increased CSF levels of kynurenic acid

    in schizophrenia.

    88. Nilsson LK, Linderholm KR, Engberg G,

    et al. Elevated levels of kynurenic acid in

    the cerebrospinal fluid of male patients

    with schizophrenia. Schizophr Res

    2005;80:315-22

    89. Schwarcz R, Rassoulpour A, Wu HQ,

    et al. Increased cortical kynurenate

    content in schizophrenia. Biol Psychiatry

    2001;50:521-30

    90. Sathyasaikumar KV, Stachowski EK,

    Wonodi I, et al. Impaired kynurenine

    pathway metabolism in the prefrontal

    cortex of individuals with schizophrenia.

    Schizophr Bull 2011;37:1147-56

    91. Miller CL, Llenos IC, Dulay JR, et al.

    Upregulation of the initiating step of the

    kynurenine pathway in postmortem

    anterior cingulate cortex from individuals

    with schizophrenia and bipolar disorder.

    Brain Res 2006;1073-1074:25-37

    92. Wonodi I, Stine OC,

    Sathyasaikumar KV, et al.

    Downregulated kynurenine 3-

    monooxygenase gene expression and

    enzyme activity in schizophrenia and

    genetic association with schizophrenia

    endophenotypes. Arch Gen Psychiatry

    2011;68:665-74

    93. Alexander KS, Wu HQ, Schwarcz R,

    et al. Acute elevations of brain kynurenic

    Targeting of NMDA receptors in new treatments for schizophrenia

    Expert Opin. Ther. Targets (2014) 18(9) 1061

    Exp

    ert O

    pin.

    The

    r. T

    arge

    ts D

    ownl

    oade

    d fr

    om in

    form

    ahea

    lthca

    re.c

    om b

    y Y

    aser

    Yac

    oob

    on 0

    3/31

    /15

    For

    pers

    onal

    use

    onl

    y.

    http://www.ncbi.nlm.nih.gov/pubmed/23044468?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23044468?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23044468?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23044468?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23044468?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21355838?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21355838?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21355838?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17804206?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17804206?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17804206?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17804206?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17804206?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17854919?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17854919?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17854919?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17854919?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23027421?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23027421?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15023571?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15023571?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15023571?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15023571?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16780811?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16780811?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16780811?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16780811?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17659263?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17659263?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/17659263?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19887019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19887019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19887019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19887019?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/2592890?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/2592890?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19212411?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19212411?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19212411?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20491477?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22138164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22138164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22138164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22138164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23132267?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23132267?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23132267?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/23132267?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24696094?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24696094?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24696094?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24696094?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24696094?dopt=Abstracthttp://www.roche.com/media/media_releases/med-cor-2014-01-21.htmhttp://www.roche.com/media/media_releases/med-cor-2014-01-21.htmhttp://www.ncbi.nlm.nih.gov/pubmed/24735806?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24735806?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24735806?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24735806?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/24735806?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22678511?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22678511?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/15683541?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21249164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21249164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21249164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21249164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21249164?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11684348?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11684348?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11684348?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16125901?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16125901?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16125901?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11600105?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11600105?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21036897?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21036897?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21036897?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21727251?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21727251?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21727251?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21727251?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21727251?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/22038535?dopt=Abstracthttp://informahealthcare.com/journal/ETT

  • acid impair cognitive flexibility:

    normalization by the alpha7 positive

    modulator galantamine.

    Psychopharmacology (Berl)

    2012;220:627-37

    94. Alexander KS, Pocivavsek A, Wu HQ,

    et al. Early developmental elevations of

    brain kynurenic acid impair cognitive

    flexibility in adults: reversal with

    galantamine. Neuroscience

    2013;238:19-28

    95. Pocivavsek A, Thomas MA, Elmer GI,

    et al. Continuous kynurenine

    administration during the prenatal

    period, but not during adolescence,

    causes learning and memory deficits in

    adult rats. Psychopharmacology (Berl)

    2014;Epub ahead of print

    96. Muller N, Myint AM, Schwarz MJ.

    Kynurenine pathway in schizophrenia:

    pathophysiological and therapeutic

    aspects. Curr Pharm Des 2011;17:130-6

    97. Aschner M. Neuron-astrocyte

    interactions: implications for cellular

    energetics and antioxidant levels.

    Neurotoxicology 2000;21:1101-7

    98. Dringen R. Metabolism and functions of

    glutathione in brain. Prog Neurobiol

    2000;62:649-71

    99. Kohr G, Eckardt S, Luddens H, et al.

    NMDA receptor channels: subunit-

    specific potentiation by reducing agents.

    Neuron 1994;12:1031-40

    100. Sullivan JM, Traynelis SF, Chen HS,

    et al. Identification of two cysteine

    residues that are required for redox

    modulation of the NMDA subtype of

    glutamate receptor. Neuron

    1994;13:929