Skip to main content
PMC Canada Author Manuscripts logoLink to PMC Canada Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 17.
Published in final edited form as: Eur Neuropsychopharmacol. 2014 Aug 1;24(10):1591–1605. doi: 10.1016/j.euroneuro.2014.07.015

Glutamate-mediated excitotoxicity in schizophrenia: A review

Eric Plitman a,b, Shinichiro Nakajima a,c,d,e, Camilo de la Fuente-Sandoval f,g, Philip Gerretsen a,b,d,e, M Mallar Chakravarty h,i, Jane Kobylianskii j, Jun Ku Chung a,b, Fernando Caravaggio a,b, Yusuke Iwata a,c, Gary Remington b,e,k,l, Ariel Graff-Guerrero a,b,d,e,k,*
PMCID: PMC4470624  CAMSID: CAMS4614  PMID: 25159198

Abstract

Findings from neuroimaging studies in patients with schizophrenia suggest widespread structural changes although the mechanisms through which these changes occur are currently unknown. Glutamatergic activity appears to be increased in the early phases of schizophrenia and may contribute to these structural alterations through an excitotoxic effect. The primary aim of this review was to describe the possible role of glutamate-mediated excitotoxicity in explaining the presence of neuroanatomical changes within schizophrenia. A Medline® literature search was conducted, identifying English language studies on the topic of glutamate-mediated excitotoxicity in schizophrenia, using the terms “schizophreni*” and “glutam*” and ((“MRS” or “MRI” or “magnetic resonance”) or (“computed tomography” or “CT”)). Studies concomitantly investigating glutamatergic activity and brain structure in patients with schizophrenia were included. Results are discussed in the context of findings from preclinical studies. Seven studies were identified that met the inclusion criteria. These studies provide inconclusive support for the role of glutamate-mediated excitotoxicity in the occurrence of structural changes within schizophrenia, with the caveat that there is a paucity of human studies investigating this topic. Preclinical data suggest that an excitotoxic effect may occur as a result of a paradoxical increase in glutamatergic activity following N-methyl-D-aspartate receptor hypofunction. Based on animal literature, glutamate-mediated excitotoxicity may account for certain structural changes present in schizophrenia, but additional human studies are required to substantiate these findings. Future studies should adopt a longitudinal design and employ magnetic resonance imaging techniques to investigate whether an association between glutamatergic activity and structural changes exists in patients with schizophrenia.

Keywords: Schizophrenia, Glutamate, Glutamine, MRS, Excitotoxicity, Psychosis

1. Introduction

This section provides a comprehensive explanation of topics relevant to the study of glutamate-mediated excitotoxicty in schizophrenia, beginning with a background of the illness and its dopaminergic hypothesis. The limitations of the dopaminergic hypothesis are important in bringing forth the glutamatergic hypothesis of schizophrenia. Subsequently, N-methyl-D-aspartate (NMDA) receptor hypofunction is introduced as a model for schizophrenia, which is followed by an explanation of glutamatergic dysfunction in schizophrenia. Next, glutamate’s capacity to exert neurotoxic effects is presented. Lastly, common neuroanatomical deficits are noted. This broad introduction provides important background information for the contextualization of current research investigating glutamate-mediated excitotoxicty in schizophrenia.

1.1. Schizophrenia

Schizophrenia is a debilitating illness, present in approximately 1% of the global population and characterized by positive, negative and cognitive symptoms (Sullivan et al., 2003; Weiser et al., 2005). The primary treatment for schizophrenia is dopamine receptor antagonism with anti-psychotic medication (Frangou, 2008). The clinical effects of dopamine receptor antagonists have provided the basis for the dopamine hypothesis of schizophrenia (Creese et al., 1976; Seeman and Lee, 1975), which posits that patients with the illness have aberrant functioning of the dopaminergic system (Abi-Dargham et al., 1998; Breier et al., 1997; Hietala et al., 1995; Laruelle et al., 1996). The dopamine hypothesis is limited in that it only addresses positive symptoms (Javitt et al., 2012); antipsychotics have minimal efficacy in the treatment of negative and cognitive symptoms (George et al., 2013; Miyamoto et al., 2012). Another limitation of the dopamine hypothesis is that 20–35% of patients show partial or no response to antipsychotic treatments (Lindenmayer, 2000; Suzuki et al., 2011). In addition, this hypothesis does not appear to adequately explain the neuroanatomical changes in patients with schizophrenia (Stone et al., 2007). Thus, the dopaminergic system does not describe the illness in its entirety (Moghaddam and Javitt, 2012). The glutamatergic hypothesis provides an alternate mechanism to explain the patho-physiology of schizophrenia.

1.2. Glutamatergic hypothesis of schizophrenia

Glutamate antagonists, such as phencyclidine (PCP) and ketamine, are well known to transiently induce symptoms similar to those observed in patients with schizophrenia (Coyle et al., 2003). Glutamate antagonists are unique in that they not only produce psychotomimetic effects, but also elicit negative and cognitive symptoms (Javitt and Zukin, 1991; Vollenweider and Geyer, 2001). Such effects have been reported following the acute administration of glutamate antagonists to healthy volunteers (Adler et al., 1999; Krystal et al., 1994, 2000, 2005; Malhotra et al., 1996), while administration of these agents to patients with schizophrenia exacerbates symptoms (Lahti et al., 1995a, 1995b). The observed symptomatic effects of glutamate antagonists provide the basis for the glutamatergic hypothesis of schizophrenia (Kantrowitz and Javitt, 2012).

1.3. NMDA receptor hypofunction

Glutamate antagonists induce schizophrenia-like symptoms through modulation of the NMDA receptor. PCP and ketamine are both non-competitive antagonists that exert their physiological effects by binding to the PCP receptor, a specific hydrophobic binding site coupled to the NMDA receptor (Javitt, 2007). Through this binding, PCP and ketamine inhibit the action of glutamate at the NMDA receptor, suggesting that the pathophysiology of schizophrenia may similarly result from dysregulation of the NMDA receptor (Javitt et al., 2012). Current proponents of the glutamatergic hypothesis postulate that hypofunctional NMDA receptors located on gamma-aminobutyric acid (GABA)–ergic inhibitory interneurons disinhibit pyramidal neurons, leading to a paradoxical increase in glutamatergic activity (Moghaddam and Krystal, 2012; Nakazawa et al., 2012; Stone et al., 2007).

1.4. Glutamatergic dysfunction in schizophrenia

The role of the NMDA receptor in increasing glutamate is supported by both preclinical and human studies using NMDA receptor antagonists. Acute treatment of rodents with NMDA receptor antagonists results in increased extracellular glutamate in the striatum and prefrontal cortex (Bustos et al., 1992; Moghaddam et al., 1997), and increased glutamine (the main metabolite of glutamate) in the pre-frontal cortex (Iltis et al., 2009). Studies in healthy human participants employing proton magnetic resonance spectroscopy (1H-MRS) report increased glutamate and glutamine in the anterior cingulate after the acute administration of a sub-anaesthetic dose of ketamine (Rowland et al., 2005; Stone et al., 2012). In addition, agents that inhibit glutamate release reverse behavioural, cognitive, and cerebral blood flow changes induced by NMDA receptor antagonists in healthy human volunteers (Anand et al., 2000; Deakin et al., 2008; Doyle et al., 2013).

The aforementioned findings in rodents and healthy humans following acute treatment with NMDA receptor antagonists are comparable to 1H-MRS studies in patients with schizophrenia, which report increased glutamate levels in antipsychotic-free and naïve subjects during their first episodes of psychosis, as well as in subjects at ultra-high risk for psychosis (de la Fuente-Sandoval et al., 2011, 2013a; Kegeles et al., 2012; Kraguljac et al., 2013; Purdon et al., 2008). 1H-MRS studies have also demonstrated higher glutamine levels in antipsychotic-naïve patients with schizophrenia (Bartha et al., 1997; Theberge et al., 2002). While there is strong evidence to support increased glutamatergic activity in patients with untreated schizophrenia, it should be noted that studies investigating medicated patients with schizophrenia have reported glutamatergic marker decreases or levels similar to healthy controls (Bustillo et al., 2011; de la Fuente-Sandoval et al., 2013b; Goto et al., 2012; Kegeles et al., 2012; Ohrmann et al., 2005; Rowland et al., 2013; Theberge et al., 2003). Thus far, two studies have made direct comparisons between unmedicated and medicated patients, both showing elevated glutamate levels in the unmedicated state and normal glutamate levels in the medicated state. Using a longitudinal within-subject comparison, one study in particular administered clinically effective antipsychotic treatment (reduction of at least 30% on the total score of the Positive and Negative Syndrome Scale after 4 weeks) to antipsychotic-naïve patients with first-episode psychosis, significantly decreasing elevated baseline glutamate in the associative striatum, such that levels following treatment did not differ from controls (de la Fuente-Sandoval et al., 2013b). Notably, this study specifically included patients who responded to treatment. Another study utilized a cross-sectional approach to compare unmedicated patients, medicated patients and healthy controls, reporting increased Glx in the medial prefrontal cortex region of unmedicated patients, in comparison to controls, whereas no such difference existed between medicated patients and the control group (Kegeles et al., 2012). To further elucidate the role of treatment in changing glutamatergic activity, a recent review noted that glutamatergic levels are elevated in anti-psychotic naïve patients but are similar to those of healthy controls in medicated patients with schizophrenia, independent of stage of illness (Poels et al., 2014a). This is contrasted by a meta-analysis that demonstrated that glutamate and glutamine concentrations decrease at a faster rate with age in patients with schizophrenia, as compared to healthy controls (Marsman et al., 2013).

However, it should be noted that recent research observed higher glutamate levels in the anterior cingulate cortex of antipsychotic-treated first episode patients with unremitted psychotic symptoms and in treatment-resistant patients than in medication responders (Demjaha et al., 2014; Egerton et al., 2012). These findings suggest that an alternative underlying pathophysiology may exist in patients with treatment-resistant schizophrenia than in patients who respond well to antipsychotics – one that similarly involves the glutamatergic system, yet is not modulated by dopaminergic regulation.

1.5. Glutamate as an excitotoxic factor

Glutamate has the potential to induce neuronal dysfunction and degeneration when present in abnormally high extracellular concentrations (Lahti and Reid, 2011; Lau and Tymianski, 2010; Mehta et al., 2013). This process is referred to as excitotoxicity, a term coined by John Olney (Olney, 1969; Olney and Sharpe, 1969), who posited that excessive stimulation by glutamate has the capacity to vastly increase intracellular calcium, affecting calcium homoeostatic mechanisms and triggering a cascade of events that ultimately result in cell death (Lau and Tymianski, 2010). Though the exact mechanisms of this phenomenon are only partially known, calcium influx is highly implicated (Belousov, 2012; Choi, 1988; Hardingham and Bading, 2010). In schizophrenia, the disruption in glutamatergic signalling may result in an excitotoxic effect secondary to excess stimulation of non-NMDA glutamate receptors (i.e AMPA and Kainate), leading to the structural findings associated with the illness (Abbott and Bustillo, 2006; Deutsch et al., 2001).

1.6. Structural changes in schizophrenia

Neuroanatomical changes are often reported in patients with schizophrenia; for example, progressive loss of grey matter volume is common in both early and chronic stages of the illness (Arango et al., 2012; Hulshoff Pol and Kahn, 2008; Meyer-Lindenberg, 2011; van Haren et al., 2008). Recent meta-analyses investigating grey matter losses in schizophrenia most commonly identify volumetric reduction within superior temporal, medial temporal, superior prefrontal, medial pre-frontal and insular regions, along with the thalamus and basal ganglia (Bora et al., 2011; Chan et al., 2011; Ellison-Wright et al., 2008; Glahn et al., 2008; Honea et al., 2005; Shenton et al., 2001; Steen et al., 2006). Whole brain volume reductions, ventricular enlargement and white-matter alterations are also frequently reported (Colibazzi et al., 2013; Connor et al., 2011; Lawrie and Abukmeil, 1998; Nazeri et al., 2013; Shenton et al., 2001; Voineskos et al., 2013).

In addition, reductions in cortical thickness are common in patients with schizophrenia. Various studies have observed cortical thinning in schizophrenia, particularly within frontal, temporal, parietal and cingulate regions, though insular and occipital areas are also affected (Goldman et al., 2009; Kuperberg et al., 2003; Narr et al., 2005; Rais et al., 2010; Rimol et al., 2010; van Haren et al., 2011; White et al., 2003).

The occurrence of these neuroanatomical changes is largely unexplained. Though the changes may conceivably result from medication intake and prolonged illness progression (Moncrieff and Leo, 2010; Navari and Dazzan, 2009; Torres et al., 2013; van Haren et al., 2012; Vita et al., 2012), studies utilizing first episode schizophrenia patients have provided evidence that structural changes occur irrespective of continuous antipsychotic treatment and years of illness duration. First episode schizophrenia patients with little or no exposure to antipsychotics exhibit neuroanatomical alterations within a number of brain regions in comparison with healthy controls (Chen et al., 2014; Fornito et al., 2008; Narr et al., 2005; Ren et al., 2013; Schultz et al., 2010; Sprooten et al., 2013; Steen et al., 2006; Venkatasubramanian et al., 2008; Vita et al., 2012). Glutamate-mediated excitotoxicity may contribute to these structural changes present in patients with schizophrenia (Abbott and Bustillo, 2006; Goff and Coyle, 2001; Stone et al., 2007).

1.7. Aim of this review

The glutamatergic hypothesis offers a mechanism through which neuroanatomical changes may occur: glutamate-mediated excitotoxicity. In short, elevated glutamatergic neurotransmission, which is highly implicated in the pathology of schizophrenia, may have neurotoxic effects. The primary aim of this review was to describe the potential role of glutamate-mediated excitotoxicity as an explanatory mechanism for the neuroanatomical changes observed in patients with schizophrenia. To do so, findings from human studies were reviewed and discussed, followed by a presentation of the evidence from preclinical literature. Limitations of both human and preclinical studies were considered in drawing conclusions and providing future research directions.

2. Experimental procedures

A Medline® literature search (1946-April Week 3 2014) was performed to identify studies, reviews or case reports relevant to glutamate-mediated excitotoxicity in patients with schizophrenia. The search was conducted using the terms “schizophreni*” (Subheadings: schizophrenia, antipsychotic agents and psychotic disorders) and “glutam*” (Subheading: magnetic resonance spectroscopy) and ((“MRS” or “MRI” or “magnetic resonance”) or (“computed tomography or “CT”)). Only English language human publications were included. Reference sections of major review articles (Deutsch et al., 2001; Javitt, 2007; Marsman et al., 2013; Poels et al., 2014a; Stone et al., 2007; Tsai and Coyle, 2002) were examined for additional, relevant articles that were overlooked by the search strategy. Articles were included if they concomitantly measured markers of glutamatergic activity and brain structure using magnetic resonance imaging (MRI) or computed tomography (CT), and investigated the relationship between the two measurements. Studies utilizing participants deemed to be at risk for schizophrenia were included. The last search was conducted on April 10th 2014. Findings resulting from this search are discussed in the context of established preclinical data.

3. Results

The Medline® search yielded 622 publications. All titles and abstracts were read by two of the authors (E.P. and S.N.). Thirteen papers concurrently investigated glutamatergic activity and brain structure, and were thus selected and reviewed. Six articles were excluded because they failed to include statistics regarding the relationship between glutamatergic markers and neuroanatomical measures (Bartha et al., 1999; Duncan et al., 2013; Gruber et al., 2012; Kegeles et al., 2000; Rusch et al., 2008; Wood et al., 2010). The supplementary search through the reference sections of the specified review articles resulted in no additional articles that concurrently measured glutamatergic activity and brain structure. Thus, all remaining studies (n=7) (Aoyama et al., 2011; Klar et al., 2010; Kraguljac et al., 2013; Stone et al., 2009; Tandon et al., 2013; Theberge et al., 2007; Tsai et al., 1998) that reported on the relationship between glutamatergic activity and brain structure in patients with schizophrenia were retained. These studies are summarized in Table 1. Six of the studies utilized 1H-MRS to assess glutamatergic markers, while one measured cerebrospinal fluid (CSF) glutamate. Six of the studies used volumetric measurements to assess structure – four of which were specific to grey matter – and one study assessed structure through ventricle to brain ratio (VBR) and pre-frontal atrophy.

Table 1.

Summary of studies that met inclusion criteria (n=7).

Authors, year,
journal
n Mean age,
(SD)
Study
design
Phase of
illness
Antipsychotic
status
Glutamatergic
marker(s)a
Structural
measure(s)a
Key findings
Kraguljac et al. (2013), JAMA Psychiatry 27 SCZ; 27 HC SCZ: 32.63 (9.28); HC: 32.85 (9.39) CS Mixed Unmedicated at assessment Glx in hippocampus. Assessed by 1H-MRS Hippocampal GM volume. Assessed by MRI In SCZ:
  • Negative correlation between hippocampal Glx and GM volume in two hippocampal clusters


In HC:
  • No significant correlation between hippocampal Glx and GM volume in hippocampus

Tandon et al. (2013), Schizophr. Res. 23 HR; 24 HC HR: 15.92 (4.21); HC: 15.57 (3.99) CS At risk Unmedicated at assessment Glx in thalamus and caudate. Assessed by 1H-MRS Thalamic and caudate volume. Assessed by MRI In HR:
  • Non-significant negative correlation between thalamic Glx and thalamic volume

  • Non-significant negative correlation between caudate Glx and caudate volume

Aoyama et al. (2011), Br. J. Psychiatry 17 SCZ; 17 HC SCZ: 25 (7); HC: 29 (10) L; 80 months FES Unmedicated at initial assessment; medicated at 10- and 80-month assessments Glutamine in thalamus and anterior cingulate; tGL in thalamus and anterior cingulate. Assessed by 1H-MRS Whole brain GM volume. Assessed by MRI In SCZ:
  • Positive correlation between thalamic glutamine loss and GM volume reduction in frontal, temporal, parietal and limbic lobes

  • No significant correlation between anterior cingulate glutamine and GM volume loss

  • No significant correlation between thalamic or anterior cingulate tGL and GM volume loss

  • Study involved introduction of medication


In HC:
  • No significant correlation between loss of thalamic glutamine and GM volume reduction

Klar et al. (2010), NeuroImage 29 SCZ; 44 HC SCZ: 27.6 (6.8); HC: 30.9 (7.9) CS Mixed Medicated at assessment Glutamate in hippocampus. Assessed by 1H-MRS Hippocampal volume. Assessed by MRI In SCZ:
  • Non-significant negative correlation between hippocampal glutamate and volume in hippocampus


In HC:
  • Non-significant negative correlation between hippocampal glutamate and volume in hippocampus

Stone et al. (2009), Biol. Psychiatry 27 ARMS; 27 HC ARMS: 25 (5); HC: 25 (4) CS At risk Unmedicated at assessment Glutamate in thalamus; glutamine in anterior cingulate. Assessed by 1H-MRS Whole brain GM volume. Assessed by MRI In ARMS:
  • Positive correlation between thalamic glutamate and GM volume in left prefrontal cortex, left insula, left cingulate, left superior temporal gyrus, left temporal pole and bilaterally in cerebellum and lingual gyrus

  • Negative correlation between thalamic glutamate and GM volume in dorsal anterior cingulate extending to posterior cingulate gyrus

  • Positive correlation between anterior cingulate glutamine and GM volume in posterior cingulate gyrus

  • Negative correlation between anterior cingulate glutamine and GM volume in left cerebellum


In HC:
  • No significant correlation between thalamic glutamate and GM volume

  • Positive correlation between anterior cingulate glutamine and GM volume in right temporal cortex

  • Negative correlation between anterior cingulate glutamine and GM volume in medial frontal and orbitofrontal cortex

Thebergeetal.(2007), Br. J. Psychiatry 16 SCZ; 16 HC SCZ: 25 (8); HC: 29 (12) L; 30 months FES Unmedicated at initial assessment; medicated at 10- and 30-month assessments Glutamine in thalamus and anterior cingulate. Assessed by 1H-MRS Whole brain GM volume and 1H-MRS voxel GM volume. Assessed by MRI In SCZ:
  • Positive correlation between thalamic glutamine loss and GM volume reduction in left angular gyrus, right precuneus and left superior and inferior temporal gyrus

  • Non-significant correlations between voxel glutamine loss and voxel GM volume loss

  • Study involved introduction of medication


In HC:
  • Non-significant correlations between voxel glutamine loss and voxel GM volume loss

Tsai et al. (1998), Biol. Psychiatry 61 SCZb; 23 HC SCZ: 37.7 (7.1); HC: 35.8 (10.1) CS Chronic Unmedicated at assessment CSF glutamate VBR; prefrontal atrophy. Assessed by CT In SCZ:
  • Negative correlation between CSF glutamate and VBR

  • Non-significant negative correlation between CSF glutamate and prefrontal atrophy

ARMS: at risk mental state; CS: cross-sectional; CT: computed tomography; FES: first episode schizophrenia; Glx: Glutamate+Glutamine; GM: grey matter; HC: healthy controls;1H-MRS: proton magnetic resonance spectroscopy; HR: high-risk; L: longitudinal; MRI: magnetic resonance imaging; SCZ: schizophrenia; SD: standard deviation; tGL: Glutamate+Glutamine; VBR: ventricle–brain ratio.

a

Only measures that were utilized for the investigation of the relationship between glutamatergic markers and brain structure, and were subsequently reported upon, are included in this table.

b

Included schizoaffective patients.

Studies that met the inclusion criteria indicate that glutamate, along with its metabolite glutamine, may have a relationship with neuroanatomical measurements. One study identified a negative correlation between two clusters of grey matter volume in the hippocampus and hippocampal Glx, a combined measure of glutamate and glutamine concentrations (Kraguljac et al., 2013). By contrast, another study that measured glutamate within the hippocampus and hippocampal volume failed to find such a relationship (Klar et al., 2010).

Two other publications report on a study that employed a longitudinal design and found a relationship between thalamic glutamine and grey matter volume (Aoyama et al., 2011; Theberge et al., 2007). Theberge et al. (2007) noted an association between decreased thalamic glutamine levels and parietal and temporal grey matter volume loss over the course of 30 months, beginning with a never-treated state. Aoyama et al. (2011) extended these findings to an 80-month follow-up, and noted a positive relationship between change in both thalamic glutamine and grey matter volume within frontal, parietal, temporal and limbic regions; thalamic glutamine levels and grey matter volume both decreased over the course of 80 months. Notably, no relationships between changes in grey matter volume and anterior cingulate glutamine or tGL, a summed measure of glutamate and glutamine levels, were reported within this study. This longitudinal design included the introduction of antipsychotic medication over the duration of the study.

A study involving individuals with an at-risk mental state (ARMS) investigated the relationship between grey matter volume and glutamate in the thalamus and glutamine in the anterior cingulate (Stone et al., 2009). This study identified positive associations in participants with an ARMS between thalamic glutamate and grey matter volume in the left prefrontal cortex, insula, cingulate, superior temporal gyrus and temporal pole, and bilaterally in the cerebellum and lingual gyrus. In the same study, negative correlations were found between thalamic glutamate and grey matter volume in the dorsal anterior cingulate extending to the posterior cingulate gyrus. Anterior cingulate glutamine was positively correlated with grey matter volume in the posterior cingulate gyrus and negatively correlated with grey matter volume in the left cerebellum. In another study that included individuals at familial high risk for schizophrenia, non-significant negative correlations were observed in the at-risk group between thalamic and caudate Glx, and thalamic and caudate volumes, respectively (Tandon et al., 2013).

Lastly, a CT investigation assessing CSF glutamate and VBR in antipsychotic-free chronic schizophrenia patients reported an inverse relationship between CSF glutamate and VBR (Tsai et al., 1998). Overall, the identified studies suggest that an association between markers of glutamatergic activity and structural measures may exist, although conflicting findings are reported within available literature.

4. Discussion

The overall aim of this review was to explore the evidence in humans for the relationship between glutamate related compounds (glutamate, glutamine and Glx) and structural brain measurements in patients with schizophrenia. A review of existing literature was conducted to elucidate the role of glutamate-mediated excitotoxicity in the structural brain changes associated with schizophrenia. Unexpectedly, the search yielded only seven studies that met inclusion criteria, reflecting the paucity of literature available to effectively address this topic in humans.

4.1. Analysis of reviewed studies

Of the seven studies identified from the search, Kraguljac et al. (2013) offered the most direct evidence for a glutamate-mediated excitotoxic effect. The authors attributed structural changes observed in the hippocampus to increases in glutamatergic activity by reporting a negative correlation between two clusters of grey matter volume and Glx within the hippocampus in the schizophrenia group, whereas no such relationship existed in the healthy control group (Kraguljac et al., 2013). In this study, the patient group was unmedicated. In contrast, Klar et al. (2010), which included medicated patients, failed to find an association between hippocampal glutamate and volume; however, the study may have been underpowered to find a significant relationship (r= −0.356, p=0.074).

Interestingly, in other studies that resulted from the search, measures of thalamic glutamatergic activity were associated with volumetric loss in a number of different brain regions. Decreases in thalamic glutamine paralleled grey matter volume decreases within frontal, temporal, parietal and limbic areas (Aoyama et al., 2011; Theberge et al., 2007). Based on the regions in which volume loss occurred and the involvement of the thalamus, excitotoxic damage was considered in the explanation of these findings. Results suggest that neurodegeneration secondary to glutamate-mediated excitotoxicity may result from decreased levels of thalamic glutamatergic activity. Consistent with this notion, a decrease in thalamic glutamatergic markers was observed in both studies, which may have resulted in diminished stimulation of NMDA receptors on GABAergic interneurons in the thalamus. This hypostimulation could subsequently result in toxicity induced by paradoxically high levels of glutamate in cortical regions through the disinhibition of thalamocortical circuits that use glutamate as a neurotransmitter.

Two additional studies offer evidence that the relationship between glutamatergic markers and volumetric measures is present in the early stages of schizophrenia. In one study of participants with an ARMS, thalamic glutamate and anterior cingulate glutamine levels were associated with grey matter volume, demonstrating both positive and negative correlations depending on the brain region (Stone et al., 2009). Another study, which included individuals with familial high-risk for schizophrenia, reported that Glx in both the thalamus and the caudate was negatively associated with regional brain volume, though both correlations were not significant (Tandon et al., 2013).

Finally, one study measured CSF glutamate and VBR using CT in antipsychotic-free chronic schizophrenia patients (Tsai et al., 1998). An inverse relationship was reported between CSF glutamate and VBR, suggesting that glutamatergic cells may have degenerated over the duration of the illness as a result of toxic levels of glutamate, resulting in a lower glutamatergic measure at the time of assessment, consistent with an excitotoxic effect.

4.2. Limitations of reviewed studies

Overall, the studies listed in Table 1 have several limitations, and as such, do not individually offer sufficient evidence to make conclusive claims about the role of glutamatergic markers in the structural alterations observed in schizophrenia. The publications by Theberge et al. (2007) and Aoyama et al. (2011) were limited largely by the introduction of medication during the study. Antipsychotic treatment very likely confounded the relationship between markers of glutamatergic activity and volumetric losses; although patients were monitored throughout the study, it is expected that the introduction of medications influenced the serial neurochemical and structural measurements. This said, we acknowledge the practical challenges associated with performing a longitudinal study in medication-free patients. The studies conducted by Stone et al. (2009) and Tandon et al. (2013) were limited by the nature of the participant population. The authors included at-risk individuals, and although useful, this sample is not necessarily generalizable to patients with schizophrenia, as many at-risk participants may not progress to the illness (McGorry et al., 2009); one study reported a 35% transition rate to a psychotic disorder over a 10-year follow-up (Nelson et al., 2013). The studies by Kraguljac et al. (2013) and Klar et al. (2010) were limited by their targeted focus on glutamatergic levels and grey matter volume in the hippocampus. A common limitation shared among the aforementioned studies was the use of 1H-MRS, which quantifies the concentration of glutamate or glutamate-related compounds but cannot distinguish between intracellular and extracellular glutamate pools, therefore failing to precisely measure glutamate neurotransmission (Poels et al., 2014b). Finally, the study performed by Tsai et al. (1998) was limited by the use of VBR, as this index of brain structure lacks sensitivity to minor neuroanatomical alterations, though this measurement was necessitated by the usage of CT. Further, the same study included participants with chronic schizophrenia; in this population, ongoing medication exposure and/or illness progression very likely confounded the relationship between glutamate levels and structural irregularities. Due to the limitations present within the reviewed studies, and the general dearth of literature exploring this topic, it remains unclear whether glutamate-mediated excitotoxicity is responsible for the structural changes observed in schizophrenia.

4.3. Evidence from preclinical literature

In contrast to currently available human studies, preclinical studies offer more conclusive evidence for the role of glutamate-mediated excitotoxicity in schizophrenia. Pharmacological NMDA receptor antagonism is believed to reflect a similar state to schizophrenia, as evidenced by changes in symptomology, blood flow and cognition (Anticevic et al., 2012; Krystal et al., 2005; Lahti et al., 1995a; Nagels et al., 2011). Established and replicated preclinical studies in rodents have reported increased extracellular measures of glutamatergic activity within cortical and subcortical brain regions following NMDA receptor antagonism (Adams and Moghaddam, 1998, 2001; Bustos et al., 1992; Moghaddam et al., 1997). The administration of NMDA receptor antagonists also consistently leads to neurotoxic injury, characterized by neuronal vacuolization, neurodegeneration and the appearance of heat-shock protein in affected cells (Farber et al., 2002; Ikonomidou et al., 1999; Olney and Farber, 1995; Olney et al., 1999; Sharp et al., 1991, 1994). Moreover, preventing glutamate release in these preclinical models, using agonists of metabotropic glutamate receptors types 2 and 3, has been shown to block the neurotoxic effect of NMDA receptor antagonists (Carter et al., 2004; Moghaddam and Adams, 1998; Okamura et al., 2003; Schobel et al., 2013). A study by Schobel et al. (2013) utilized a preclinical rodent model to test the hypothesis that excess glutamate serves as a common upstream mechanism for hippocampal hypermetabolism and atrophy, after these phenomena were identified in participants who fulfilled “clinical high-risk” criteria. Acute ketamine administration led to hippocampal hypermetabolism, whereas chronic ketamine administration additionally resulted in hippocampal atrophy and parvalbumin-expressing interneuron downregulation; pre-treatment with an agonist of metabotropic glutamate receptors types 2 and 3 prevented the effects of ketamine. Thus, this study provided translational evidence to implicate increased glutamate in certain metabolic and structural abnormalities present in schizophrenia, notably concluding that excess glutamate may drive hippocampal atrophy.

Furthermore, the injury induced by NMDA receptor antagonists in preclinical models is comparable to the damage seen in schizophrenia in that both are age-dependent and affect similar locations within the brain. In schizophrenia, symptoms and certain structural changes usually appear after puberty, in late adolescence (Kessler et al., 2007; Lieberman et al., 2001; van Os and Kapur, 2009). The psychotomimetic properties of NMDA receptor antagonists are also age-dependent (Farber, 2003; Reich and Silvay, 1989). NMDA receptor antagonists begin to cause injury to rodent brain cells around the time of puberty (approximately 45 days of age), an effect that becomes more severe as rodents progress into adulthood (Auer, 1996; Farber et al., 1995). Moreover, NMDA receptor antagonism in preclinical models damages regions similar to those affected in patients with schizophrenia, including limbic and neocortical brain areas (Ellison, 1994; Hargreaves et al., 1993; Olney and Farber, 1995; Olney et al., 1989; Sharp et al., 2001). These findings offer support for the role of glutamate-mediated excitotoxicity in causing structural changes, and are consistent with the NMDA receptor hypofunction theory of schizophrenia.

The use of this preclinical model of schizophrenia also offers an opportunity to explore the mechanism by which glutamate-mediated excitotoxicity causes neuroanatomical changes. Rodent studies have suggested that the thalamus may be a primary site of NMDA receptor hypofunction. NMDA receptor hypofunction on thalamic GABAergic inter-neurons may lead to decreased GABA production and subsequent disinhibition of thalamocortical circuits, which utilize glutamate as a neurotransmitter (Sharp et al., 2001). As the thalamus has a multitude of projection neurons, NMDA receptor hypofunction on thalamic GABAergic inter-neurons provides one explanation for the increase in glutamatergic markers across multiple brain regions in schizophrenia. This is supported by studies where a GABAA agonist is injected into the thalamus, preventing NMDA receptor antagonist-induced neurotoxicity (Sharp et al., 2001; Tomitaka et al., 2000). Also, NMDA receptor antagonist administration into the anterior nucleus of the thalamus results in cortical degeneration, while injection into cortical regions has no effect (Sharp et al., 2001; Tomitaka et al., 2000). Notably, unlike systemic treatment with NMDA antagonists, cortical injection of ketamine also does not induce glutamate release (Lorrain et al., 2003).

The surge in glutamate following NMDA receptor hypofunction is believed to act on AMPA/kainate receptors, possibly resulting in calcium influx that leads to an excitotoxic effect. In support of this theory, administration of AMPA/kainate receptor antagonists and calcium channel blockers individually protects against neuronal injury by NMDA receptor antagonism (Deutsch et al., 2001; Ikonomidou et al., 1999; Sharp et al., 1995).

The ability to generalize these findings from preclinical studies to patients with schizophrenia is dependent on both the validity of the NMDA receptor hypothesis of schizophrenia and on the comparability of acute NMDA receptor deficits to the chronic NMDA receptor hypofunction presumed to exist in the illness. Thus, though findings from preclinical studies are not wholly generalizable to patients with schizophrenia, they certainly suggest that an excitotoxic effect may occur as a result of a paradoxical increase in glutamatergic activity following NMDA receptor hypofunction.

4.4. Future directions

Results from these preclinical studies provide sufficient evidence for future research to investigate the role of glutamate-mediated excitotoxicity in patients with schizophrenia. Several prior studies have assessed the relationship between functional imaging measures and glutamatergic markers (Hutcheson et al., 2012; Reid et al., 2010; Valli et al., 2011). The challenge in transferring this strategy to analysing structural imaging data relates to the fact that structural changes likely do not evolve acutely. As such, future studies should adopt a longitudinal design, serially measuring glutamatergic markers and brain structure in individuals at high risk for psychosis as they progress to their first episode of schizophrenia. In addition, the use of carbon magnetic resonance spectroscopy (13C-MRS) along with 1H-MRS would provide a complementary approach to understanding glutamatergic activity and excitotoxicity in schizophrenia. 13C-MRS has the capacity to assess neuronal and astrocyte metabolic activity, and has been previously employed to study the glutamatergic system in rodent models of schizophrenia and human participants (Eyjolfsson et al., 2011; Kondziella et al., 2006; Rothman et al., 2011). Lastly, it is for a future review to investigate the relationship between markers of glutamatergic activity and N-acetyl-aspartate measures, as the latter is believed to reflect neuronal viability (Steen et al., 2005). Further, N-acetyl-aspartate is altered in schizophrenia and has been shown to decrease following PCP administration in rodents (Brugger et al., 2011; Bustillo et al., 2012; Kraguljac et al., 2012; Natsubori et al., 2013; Reynolds et al., 2005).

4.5. Limitations of present review

This review is not without limitations. The principal aim may have been too narrow, yielding few studies specifically examining glutamate-mediated excitotoxicity in schizophrenia, thus rendering it challenging to make meaningful conclusions. Similarly, relevant articles may have been omitted from the Medline® search due to selection of the search terms.

Further, this review is biased to studies that used 1H-MRS. It may have benefited from including additional imaging techniques other than MRI and CT, such as PET and SPECT. However, the few studies that have employed these methods to investigate the glutamatergic system in schizophrenia have not addressed the relationship between structure and glutamatergic activity (Pilowsky et al., 2006; Stone et al., 2006). Moreover, MRI is currently regarded as the most sensitive, non-invasive imaging technique and does not utilize ionizing radiation. Thus, it is likely that the majority of subsequent research will be performed using this modality.

5. Conclusion

The glutamatergic hypothesis of schizophrenia provides an alternate or complementary mechanism to the dopaminergic hypothesis of schizophrenia. Excitotoxic levels of glutamate secondary to NMDA receptor hypofunction on GABAergic inhibitory interneurons may contribute to the structural abnormalities observed in schizophrenia. However, currently available literature from human studies fails to adequately address this topic.

The present review performed a Medline® search to investigate whether glutamate-mediated excitotoxicity is supported by findings from human schizophrenia studies. This search resulted in a small number of studies that met inclusion criteria: MRI or CT studies that concomitantly measured glutamatergic activity and brain structure. From the articles that met these criteria, it remains inconclusive whether glutamate-mediated excitotoxicity adequately explains the neuroanatomical changes observed in patients with schizophrenia. It is possible that publication bias exists within the literature, such that negative findings on this topic are less likely to be published.

In contrast, a subsequent discussion of preclinical studies does provide sufficient evidence of increased glutamatergic activity and associated structural alterations in response to the administration of NMDA receptor antagonists. Based on this body of literature, further studies in humans are warranted to determine the degree to which glutamate-mediated excitotoxicity could explain the structural deficits present in schizophrenia. Determining whether glutamate-mediated excitotoxicty contributes to the structural changes in schizophrenia may aid in the understanding of illness progression, along with the development of adjunctive therapies. Studies investigating the use of glutamatergic modulators to this point have provided useful, yet inconclusive evidence and as such, a better understanding of glutamatergic dysfunction within schizophrenia is required (Heresco-Levy, 2005; Javitt et al., 2012; Large et al., 2005; Weiser et al., 2012).

Future studies investigating the relationship between glutamatergic function and brain structure should employ MRI techniques and adopt a longitudinal study design, including individuals at a high risk for schizophrenia as they transition into the illness. Future research would also benefit from performing MRS studies to assess glutamatergic activity in unmedicated patients. Glutamatergic activity is commonly, reliably and non-invasively evaluated through MRS, and structure can be assessed through analyses of T1-weighted images.

Acknowledgments

We would like to thank Kristin Vesely for editing the first draft of the manuscript.

Role of funding source

The work was partially supported by a NARSAD Independent Investigator Grant from the Brain and Behavioural Research Foundation (A.G.), the Canadian Institute of Health Research MOP-114989 (A.G.), the Ontario Graduate Scholarship (E.P.) and the Early Researcher Award, Ministry of Economic Development and Innovation of Ontario (A.G., Y.I.). These sources were not involved in the collection, analysis and interpretation of data, the writing of the report or in the decision to submit the paper for publication.

Footnotes

Contributors

E.P. performed the literature search and analysis, and wrote the draft of the manuscript. S.N. also contributed to the literature search and analysis, participated in the writing of and approved the final draft of the manuscript. C.D. critically reviewed, contributed to and approved the final draft of the manuscript. P.G. was central to the formulation of the idea for the review and critically reviewed, contributed to and approved the final draft of the manuscript. M.C. contributed to the design of the review, and critically reviewed and approved the final draft of the manuscript. Ja.K. aided in the writing of the first draft of the manuscript and critically reviewed the final draft of the manuscript. Ju.K. aided in the writing of the first draft of the manuscript and critically reviewed the final draft of the manuscript. F.C. critically reviewed, contributed to and approved the final draft of manuscript. Y.I. critically reviewed, contributed to and approved the final draft of the manuscript. G.R. contributed to the design of the review, and critically reviewed and approved the final draft of the manuscript. A.G. supervised the writing of the manuscript, provided the idea for the project, and critically reviewed, contributed to and approved the first and final draft of the manuscript.

Conflict of interest

E.P. has received funding from the Ontario Graduate Scholarship. S.N. has received fellowship grant awards from the Centre for Addiction and Mental Health, Japan Society for the Promotion of Science, Canadian Institute of Health Research, and manuscript fees from Dainippon Sumitomo Pharma and Kyowa Hakko Kirin. C.D. has received grant support from Janssen (Johnson & Johnson), and has served as a consultant and/or speaker for AstraZeneca, Eli Lilly and Janssen. P.G. has received fellowship awards from the Centre for Addiction and Mental Health and the Ontario Mental Health Foundation. Y.I. has received manuscript fees from Wiley Japan. G.R. has received research support from the Canadian Diabetes Association, the Canadian Institutes of Health Research, Medicure, Neurocrine Biosciences, Novartis, Research Hospital Fund–Canada Foundation for Innovation, and the Schizophrenia Society of Ontario and has served as a consultant or speaker for Novartis, Laboratorios Farmacéuticos Rovi, Synchroneuron, and Roche. A.G. has received grant support from US National Institute of Health, Canadian Institute of Health Research, Ontario Mental Health Foundation, CONACyT, ICyTDF, NARSAD, Research Hospital Fund–Canada Foundation for Innovation, Ministry of Economic Development and Innovation of Ontario and Janssen-Cilag. He has served as a consultant for Abbott Laboratories, Gedeon Richter Plc, and Eli Lilly. M.C., Ja.K., Ju.K and F.C. have no conflicts of interest to report.

References

  1. Abbott C, Bustillo J. What have we learned from proton magnetic resonance spectroscopy about schizophrenia? A critical update Curr Opin Psychiatry. 2006;19:135–139. doi: 10.1097/01.yco.0000214337.29378.cd. [DOI] [PubMed] [Google Scholar]
  2. Abi-Dargham A, Gil R, Krystal J, Baldwin RM, Seibyl JP, Bowers M, van Dyck CH, Charney DS, Innis RB, Laruelle M. Increased striatal dopamine transmission in schizophrenia: confirmation in a second cohort. Am J Psychiatry. 1998;155:761–767. doi: 10.1176/ajp.155.6.761. [DOI] [PubMed] [Google Scholar]
  3. Adams B, Moghaddam B. Corticolimbic dopamine neuro-transmission is temporally dissociated from the cognitive and locomotor effects of phencyclidine. J Neurosci. 1998;18:5545–5554. doi: 10.1523/JNEUROSCI.18-14-05545.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Adams BW, Moghaddam B. Effect of clozapine, haloperidol, or M100907 on phencyclidine-activated glutamate efflux in the prefrontal cortex. Biol Psychiatry. 2001;50:750–757. doi: 10.1016/s0006-3223(01)01195-7. [DOI] [PubMed] [Google Scholar]
  5. Adler CM, Malhotra AK, Elman I, Goldberg T, Egan M, Pickar D, Breier A. Comparison of ketamine-induced thought disorder in healthy volunteers and thought disorder in schizophrenia. Am J Psychiatry. 1999;156:1646–1649. doi: 10.1176/ajp.156.10.1646. [DOI] [PubMed] [Google Scholar]
  6. Anand A, Charney DS, Oren DA, Berman RM, Hu XS, Cappiello A, Krystal JH. Attenuation of the neuropsychiatric effects of ketamine with lamotrigine: support for hyperglutamatergic effects of N-methyl-D-aspartate receptor antagonists. Arch Gen Psychiatry. 2000;57:270–276. doi: 10.1001/archpsyc.57.3.270. [DOI] [PubMed] [Google Scholar]
  7. Anticevic A, Gancsos M, Murray JD, Repovs G, Driesen NR, Ennis DJ, Niciu MJ, Morgan PT, Surti TS, Bloch MH, Ramani R, Smith MA, Wang XJ, Krystal JH, Corlett PR. NMDA receptor function in large-scale anticorrelated neural systems with implications for cognition and schizophrenia. Proc Natl Acad Sci USA. 2012;109:16720–16725. doi: 10.1073/pnas.1208494109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Aoyama N, Theberge J, Drost DJ, Manchanda R, Northcott S, Neufeld RW, Menon RS, Rajakumar N, Pavlosky WF, Densmore M, Schaefer B, Williamson PC. Grey matter and social functioning correlates of glutamatergic metabolite loss in schizophrenia. Br J Psychiatry. 2011;198:448–456. doi: 10.1192/bjp.bp.110.079608. [DOI] [PubMed] [Google Scholar]
  9. Arango C, Rapado-Castro M, Reig S, Castro-Fornieles J, Gonzalez-Pinto A, Otero S, Baeza I, Moreno C, Graell M, Janssen J, Parellada M, Moreno D, Bargallo N, Desco M. Progressive brain changes in children and adolescents with first-episode psychosis. Arch Gen Psychiatry. 2012;69:16–26. doi: 10.1001/archgenpsychiatry.2011.150. [DOI] [PubMed] [Google Scholar]
  10. Auer RN. Effect of age and sex on N-methyl-D-aspartate antagonist-induced neuronal necrosis in rats. Stroke. 1996;27:743–746. doi: 10.1161/01.str.27.4.743. [DOI] [PubMed] [Google Scholar]
  11. Bartha R, al-Semaan YM, Williamson PC, Drost DJ, Malla AK, Carr TJ, Densmore M, Canaran G, Neufeld RW. A short echo proton magnetic resonance spectroscopy study of the left mesial-temporal lobe in first-onset schizophrenic patients. Biol Psychiatry. 1999;45:1403–1411. doi: 10.1016/s0006-3223(99)00007-4. [DOI] [PubMed] [Google Scholar]
  12. Bartha R, Williamson PC, Drost DJ, Malla A, Carr TJ, Cortese L, Canaran G, Rylett RJ, Neufeld RW. Measurement of glutamate and glutamine in the medial pre-frontal cortex of never-treated schizophrenic patients and healthy controls by proton magnetic resonance spectroscopy. Arch Gen Psychiatry. 1997;54:959–965. doi: 10.1001/archpsyc.1997.01830220085012. [DOI] [PubMed] [Google Scholar]
  13. Belousov AB. Novel model for the mechanisms of glutamate-dependent excitotoxicity: role of neuronal gap junctions. Brain Res. 2012;1487:123–130. doi: 10.1016/j.brainres.2012.05.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bora E, Fornito A, Radua J, Walterfang M, Seal M, Wood SJ, Yucel M, Velakoulis D, Pantelis C. Neuroanatomical abnormalities in schizophrenia: a multimodal voxelwise meta-analysis and meta-regression analysis. Schizophr Res. 2011;127:46–57. doi: 10.1016/j.schres.2010.12.020. [DOI] [PubMed] [Google Scholar]
  15. Breier A, Su TP, Saunders R, Carson RE, Kolachana BS, de Bartolomeis A, Weinberger DR, Weisenfeld N, Malhotra AK, Eckelman WC, Pickar D. Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA. 1997;94:2569–2574. doi: 10.1073/pnas.94.6.2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Brugger S, Davis JM, Leucht S, Stone JM. Proton magnetic resonance spectroscopy and illness stage in schizophrenia—a systematic review and meta-analysis. Biol Psychiatry. 2011;69:495–503. doi: 10.1016/j.biopsych.2010.10.004. [DOI] [PubMed] [Google Scholar]
  17. Bustillo J, Galloway MP, Ghoddoussi F, Bolognani F, Perrone-Bizzozero N. Medial-frontal cortex hypometabolism in chronic phencyclidine exposed rats assessed by high resolution magic angle spin 11.7 T proton magnetic resonance spectroscopy. Neurochem Int. 2012;61:128–131. doi: 10.1016/j.neuint.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bustillo JR, Chen H, Gasparovic C, Mullins P, Caprihan A, Qualls C, Apfeldorf W, Lauriello J, Posse S. Glutamate as a marker of cognitive function in schizophrenia: a proton spectroscopic imaging study at 4 T. Biol Psychiatry. 2011;69:19–27. doi: 10.1016/j.biopsych.2010.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bustos G, Abarca J, Forray MI, Gysling K, Bradberry CW, Roth RH. Regulation of excitatory amino acid release by N-methyl-D-aspartate receptors in rat striatum: in vivo micro-dialysis studies. Brain Res. 1992;585:105–115. doi: 10.1016/0006-8993(92)91195-k. [DOI] [PubMed] [Google Scholar]
  20. Carter K, Dickerson J, Schoepp DD, Reilly M, Herring N, Williams J, Sallee FR, Sharp JW, Sharp FR. The mGlu2/3 receptor agonist LY379268 injected into cortex or thalamus decreases neuronal injury in retrosplenial cortex produced by NMDA receptor antagonist MK-801: possible implications for psychosis. Neuropharmacology. 2004;47:1135–1145. doi: 10.1016/j.neuropharm.2004.08.018. [DOI] [PubMed] [Google Scholar]
  21. Chan RC, Di X, McAlonan GM, Gong QY. Brain anatomical abnormalities in high-risk individuals, first-episode, and chronic schizophrenia: an activation likelihood estimation meta-analysis of illness progression. Schizophr Bull. 2011;37:177–188. doi: 10.1093/schbul/sbp073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen Z, Deng W, Gong Q, Huang C, Jiang L, Li M, He Z, Wang Q, Ma X, Wang Y, Chua SE, McAlonan GM, Sham PC, Collier DA, McGuire P, Li T. Extensive brain structural network abnormality in first-episode treatment-naive patients with schizophrenia: morphometrical and covariation study. Psychol Med. 2014;44:2489–2501. doi: 10.1017/S003329171300319X. [DOI] [PubMed] [Google Scholar]
  23. Choi DW. Glutamate neurotoxicity and diseases of the nervous system. Neuron. 1988;1:623–634. doi: 10.1016/0896-6273(88)90162-6. [DOI] [PubMed] [Google Scholar]
  24. Colibazzi T, Wexler BE, Bansal R, Hao X, Liu J, Sanchez-Pena J, Corcoran C, Lieberman JA, Peterson BS. Anatomical abnormalities in gray and white matter of the cortical surface in persons with schizophrenia. PloS One. 2013;8:e55783. doi: 10.1371/journal.pone.0055783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Connor CM, Crawford BC, Akbarian S. White matter neuron alterations in schizophrenia and related disorders. Int J Dev Neurosci. 2011;29:325–334. doi: 10.1016/j.ijdevneu.2010.07.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Coyle JT, Tsai G, Goff D. Converging evidence of NMDA receptor hypofunction in the pathophysiology of schizophrenia. Ann N Y Acad Sci. 2003;1003:318–327. doi: 10.1196/annals.1300.020. [DOI] [PubMed] [Google Scholar]
  27. Creese I, Burt DR, Snyder SH. Science. Vol. 192. New York, N.Y: 1976. Dopamine receptor binding predicts clinical and pharmacological potencies of antischizophrenic drugs; pp. 481–483. [DOI] [PubMed] [Google Scholar]
  28. Deakin JF, Lees J, McKie S, Hallak JE, Williams SR, Dursun SM. Glutamate and the neural basis of the subjective effects of ketamine: a pharmaco-magnetic resonance imaging study. Arch Gen Psychiatry. 2008;65:154–164. doi: 10.1001/archgenpsychiatry.2007.37. [DOI] [PubMed] [Google Scholar]
  29. Demjaha A, Egerton A, Murray RM, Kapur S, Howes OD, Stone JM, McGuire PK. Antipsychotic treatment resistance in schizophrenia associated with elevated glutamate levels but normal dopamine function. Biol Psychiatry. 2014;75:e11–e13. doi: 10.1016/j.biopsych.2013.06.011. [DOI] [PubMed] [Google Scholar]
  30. Deutsch SI, Rosse RB, Schwartz BL, Mastropaolo J. A revised excitotoxic hypothesis of schizophrenia: therapeutic implications. Clin Neuropharmacol. 2001;24:43–49. doi: 10.1097/00002826-200101000-00008. [DOI] [PubMed] [Google Scholar]
  31. de la Fuente-Sandoval C, Leon-Ortiz P, Azcarraga M, Favila R, Stephano S, Graff-Guerrero A. Striatal glutamate and the conversion to psychosis: a prospective 1H-MRS imaging study. Int J Neuropsychopharmacol. 2013a;16:471–475. doi: 10.1017/S1461145712000314. [DOI] [PubMed] [Google Scholar]
  32. de la Fuente-Sandoval C, Leon-Ortiz P, Azcarraga M, Stephano S, Favila R, Diaz-Galvis L, Alvarado-Alanis P, Ramirez-Bermudez J, Graff-Guerrero A. Glutamate levels in the associative striatum before and after 4 weeks of antipsychotic treatment in first-episode psychosis: a longitudinal proton magnetic resonance spectroscopy study. JAMA Psychiatry. 2013;70:1057–1066. doi: 10.1001/jamapsychiatry.2013.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. de la Fuente-Sandoval C, Leon-Ortiz P, Favila R, Stephano S, Mamo D, Ramirez-Bermudez J, Graff-Guerrero A. Higher levels of glutamate in the associative-striatum of subjects with prodromal symptoms of schizophrenia and patients with first-episode psychosis. Neuropsychopharmacology. 2011;36:1781–1791. doi: 10.1038/npp.2011.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Doyle OM, De Simoni S, Schwarz AJ, Brittain C, O’Daly OG, Williams SC, Mehta MA. Quantifying the attenuation of the ketamine pharmacological magnetic resonance imaging response in humans: a validation using antipsychotic and glutamatergic agents. J Pharmacol Exp Ther. 2013;345:151–160. doi: 10.1124/jpet.112.201665. [DOI] [PubMed] [Google Scholar]
  35. Duncan NW, Wiebking C, Tiret B, Marjanska M, Hayes DJ, Lyttleton O, Doyon J, Northoff G. Glutamate concentration in the medial prefrontal cortex predicts resting-state cortical-subcortical functional connectivity in humans. PloS One. 2013;8:e60312. doi: 10.1371/journal.pone.0060312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Egerton A, Brugger S, Raffin M, Barker GJ, Lythgoe DJ, McGuire PK, Stone JM. Anterior cingulate glutamate levels related to clinical status following treatment in first-episode schizophrenia. Neuropsychopharmacology. 2012;37:2515–2521. doi: 10.1038/npp.2012.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Ellison G. Competitive and non-competitive NMDA antagonists induce similar limbic degeneration. Neuroreport. 1994;5:2688–2692. doi: 10.1097/00001756-199412000-00070. [DOI] [PubMed] [Google Scholar]
  38. Ellison-Wright I, Glahn DC, Laird AR, Thelen SM, Bullmore E. The anatomy of first-episode and chronic schizophrenia: an anatomical likelihood estimation meta-analysis. Am J Psychiatry. 2008;165:1015–1023. doi: 10.1176/appi.ajp.2008.07101562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Eyjolfsson EM, Nilsen LH, Kondziella D, Brenner E, Haberg A, Sonnewald U. Altered 13C glucose metabolism in the cortico-striato-thalamo-cortical loop in the MK-801 rat model of schizophrenia. J Cereb Blood Flow Metab. 2011;31:976–985. doi: 10.1038/jcbfm.2010.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Farber NB. The NMDA receptor hypofunction model of psychosis. Ann N Y Acad Sci. 2003;1003:119–130. doi: 10.1196/annals.1300.008. [DOI] [PubMed] [Google Scholar]
  41. Farber NB, Kim SH, Dikranian K, Jiang XP, Heinkel C. Receptor mechanisms and circuitry underlying NMDA antagonist neurotoxicity. Mol Psychiatry. 2002;7:32–43. doi: 10.1038/sj.mp.4000912. [DOI] [PubMed] [Google Scholar]
  42. Farber NB, Wozniak DF, Price MT, Labruyere J, Huss St J, Peter H, Olney JW. Age-specific neurotoxicity in the rat associated with NMDA receptor blockade: potential relevance to schizophrenia? Biol Psychiatry. 1995;38:788–796. doi: 10.1016/0006-3223(95)00046-1. [DOI] [PubMed] [Google Scholar]
  43. Fornito A, Yucel M, Wood SJ, Adamson C, Velakoulis D, Saling MM, McGorry PD, Pantelis C. Surface-based morphometry of the anterior cingulate cortex in first episode schizophrenia. Hum Brain Mapp. 2008;29:478–489. doi: 10.1002/hbm.20412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Frangou S. Schizophrenia. Medicine. 2008;36:405–409. [Google Scholar]
  45. George M, Amrutheshwar R, Rajkumar RP, Kattimani S, Dkhar SA. Newer antipsychotics and upcoming molecules for schizophrenia. Eur J Clin Pharmacol. 2013;69:1497–1509. doi: 10.1007/s00228-013-1498-4. [DOI] [PubMed] [Google Scholar]
  46. Glahn DC, Laird AR, Ellison-Wright I, Thelen SM, Robinson JL, Lancaster JL, Bullmore E, Fox PT. Meta-analysis of gray matter anomalies in schizophrenia: application of anatomic likelihood estimation and network analysis. Biol Psychiatry. 2008;64:774–781. doi: 10.1016/j.biopsych.2008.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Goff DC, Coyle JT. The emerging role of glutamate in the pathophysiology and treatment of schizophrenia. Am J Psychiatry. 2001;158:1367–1377. doi: 10.1176/appi.ajp.158.9.1367. [DOI] [PubMed] [Google Scholar]
  48. Goldman AL, Pezawas L, Mattay VS, Fischl B, Verchinski BA, Chen Q, Weinberger DR, Meyer-Lindenberg A. Widespread reductions of cortical thickness in schizophrenia and spectrum disorders and evidence of heritability. Arch Gen Psychiatry. 2009;66:467–477. doi: 10.1001/archgenpsychiatry.2009.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Goto N, Yoshimura R, Kakeda S, Nishimura J, Moriya J, Hayashi K, Katsuki A, Hori H, Umene-Nakano W, Ikenouchi-Sugita A, Korogi Y, Nakamura J. Six-month treatment with atypical antipsychotic drugs decreased frontal-lobe levels of glutamate plus glutamine in early-stage first-episode schizophrenia. Neuropsychiatr Dis Treat. 2012;8:119–122. doi: 10.2147/NDT.S25582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gruber O, Hasan A, Scherk H, Wobrock T, Schneider-Axmann T, Ekawardhani S, Schmitt A, Backens M, Reith W, Meyer J, Falkai P. Association of the brain-derived neurotrophic factor val66met polymorphism with magnetic resonance spectroscopic markers in the human hippocampus: in vivo evidence for effects on the glutamate system. Eur Arch Psychiatry Clin Neurosci. 2012;262:23–31. doi: 10.1007/s00406-011-0214-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–696. doi: 10.1038/nrn2911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hargreaves RJ, Rigby M, Smith D, Hill RG, Iversen LL. Competitive as well as uncompetitive N-methyl-D-aspartate receptor antagonists affect cortical neuronal morphology and cerebral glucose metabolism. Neurochem Res. 1993;18:1263–1269. doi: 10.1007/BF00975046. [DOI] [PubMed] [Google Scholar]
  53. Heresco-Levy U. Glutamatergic neurotransmission modulators as emerging new drugs for schizophrenia. Expert Opin Emerg Drugs. 2005;10:827–844. doi: 10.1517/14728214.10.4.827. [DOI] [PubMed] [Google Scholar]
  54. Hietala J, Syvalahti E, Vuorio K, Rakkolainen V, Bergman J, Haaparanta M, Solin O, Kuoppamaki M, Kirvela O, Ruotsalainen U, et al. Presynaptic dopamine function in striatum of neuroleptic-naive schizophrenic patients. Lancet. 1995;346:1130–1131. doi: 10.1016/s0140-6736(95)91801-9. [DOI] [PubMed] [Google Scholar]
  55. Honea R, Crow TJ, Passingham D, Mackay CE. Regional deficits in brain volume in schizophrenia: a meta-analysis of voxel-based morphometry studies. Am J Psychiatry. 2005;162:2233–2245. doi: 10.1176/appi.ajp.162.12.2233. [DOI] [PubMed] [Google Scholar]
  56. Hulshoff Pol HE, Kahn RS. What happens after the first episode? A review of progressive brain changes in chronically ill patients with schizophrenia. Schizophr Bull. 2008;34:354–366. doi: 10.1093/schbul/sbm168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hutcheson NL, Reid MA, White DM, Kraguljac NV, Avsar KB, Bolding MS, Knowlton RC, den Hollander JA, Lahti AC. Multimodal analysis of the hippocampus in schizophrenia using proton magnetic resonance spectroscopy and functional magnetic resonance imaging. Schizophr Res. 2012;140:136–142. doi: 10.1016/j.schres.2012.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, Dikranian K, Tenkova TI, Stefovska V, Turski L, Olney JW. Science. Vol. 283. New York, N.Y: 1999. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain; pp. 70–74. [DOI] [PubMed] [Google Scholar]
  59. Iltis I, Koski DM, Eberly LE, Nelson CD, Deelchand DK, Valette J, Ugurbil K, Lim KO, Henry PG. Neuro-chemical changes in the rat prefrontal cortex following acute phencyclidine treatment: an in vivo localized (1)H MRS study. NMR Biomed. 2009;22:737–744. doi: 10.1002/nbm.1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Javitt DC. Glutamate and schizophrenia: phencyclidine, N-methyl-D-aspartate receptors, and dopamine-glutamate interactions. Int Rev Neurobiol. 2007;78:69–108. doi: 10.1016/S0074-7742(06)78003-5. [DOI] [PubMed] [Google Scholar]
  61. Javitt DC, Zukin SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry. 1991;148:1301–1308. doi: 10.1176/ajp.148.10.1301. [DOI] [PubMed] [Google Scholar]
  62. Javitt DC, Zukin SR, Heresco-Levy U, Umbricht D. Has an angel shown the way? Etiological and therapeutic implications of the PCP/NMDA model of schizophrenia. Schizophr Bull. 2012;38:958–966. doi: 10.1093/schbul/sbs069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kantrowitz J, Javitt DC. Glutamatergic transmission in schizophrenia: from basic research to clinical practice. Curr Opin Psychiatry. 2012;25:96–102. doi: 10.1097/YCO.0b013e32835035b2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kegeles LS, Mao X, Stanford AD, Girgis R, Ojeil N, Xu X, Gil R, Slifstein M, Abi-Dargham A, Lisanby SH, Shungu DC. Elevated prefrontal cortex gamma-aminobutyric acid and glutamate-glutamine levels in schizophrenia measured in vivo with proton magnetic resonance spectroscopy. Arch Gen Psychiatry. 2012;69:449–459. doi: 10.1001/archgenpsychiatry.2011.1519. [DOI] [PubMed] [Google Scholar]
  65. Kegeles LS, Shungu DC, Anjilvel S, Chan S, Ellis SP, Xanthopoulos E, Malaspina D, Gorman JM, Mann JJ, Laruelle M, Kaufmann CA. Hippocampal pathology in schizophrenia: magnetic resonance imaging and spectroscopy studies. Psychiatry Res. 2000;98:163–175. doi: 10.1016/s0925-4927(00)00044-5. [DOI] [PubMed] [Google Scholar]
  66. Kessler RC, Amminger GP, Aguilar-Gaxiola S, Alonso J, Lee S, Ustun TB. Age of onset of mental disorders: a review of recent literature. Curr Opin Psychiatry. 2007;20:359–364. doi: 10.1097/YCO.0b013e32816ebc8c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Klar AA, Ballmaier M, Leopold K, Hake I, Schaefer M, Bruhl R, Schubert F, Gallinat J. Interaction of hippocampal volume and N-acetylaspartate concentration deficits in schizophrenia: a combined MRI and 1H-MRS study. NeuroImage. 2010;53:51–57. doi: 10.1016/j.neuroimage.2010.06.006. [DOI] [PubMed] [Google Scholar]
  68. Kondziella D, Brenner E, Eyjolfsson EM, Markinhuhta KR, Carlsson ML, Sonnewald U. Glial-neuronal interactions are impaired in the schizophrenia model of repeated MK801 exposure. Neuropsychopharmacology. 2006;31:1880–1887. doi: 10.1038/sj.npp.1300993. [DOI] [PubMed] [Google Scholar]
  69. Kraguljac NV, Reid MA, White DM, den Hollander J, Lahti AC. Regional decoupling of N-acetyl-aspartate and glutamate in schizophrenia. Neuropsychopharmacology. 2012;37:2635–2642. doi: 10.1038/npp.2012.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kraguljac NV, White DM, Reid MA, Lahti AC. Increased hippocampal glutamate and volumetric deficits in unmedicated patients with schizophrenia. JAMA Psychiatry. 2013;70:1294–1302. doi: 10.1001/jamapsychiatry.2013.2437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Krystal JH, Bennett A, Abi-Saab D, Belger A, Karper LP, D’Souza DC, Lipschitz D, Abi-Dargham A, Charney DS. Dissociation of ketamine effects on rule acquisition and rule implementation: possible relevance to NMDA receptor contributions to executive cognitive functions. Biol Psychiatry. 2000;47:137–143. doi: 10.1016/s0006-3223(99)00097-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
  73. Krystal JH, Perry EB, Jr, Gueorguieva R, Belger A, Madonick SH, Abi-Dargham A, Cooper TB, Macdougall L, Abi-Saab W, D’Souza DC. Comparative and interactive human psychopharmacologic effects of ketamine and amphetamine: implications for glutamatergic and dopaminergic model psychoses and cognitive function. Arch Gen Psychiatry. 2005;62:985–994. doi: 10.1001/archpsyc.62.9.985. [DOI] [PubMed] [Google Scholar]
  74. Kuperberg GR, Broome MR, McGuire PK, David AS, Eddy M, Ozawa F, Goff D, West WC, Williams SC, van der Kouwe AJ, Salat DH, Dale AM, Fischl B. Regionally localized thinning of the cerebral cortex in schizophrenia. Arch Gen Psychiatry. 2003;60:878–888. doi: 10.1001/archpsyc.60.9.878. [DOI] [PubMed] [Google Scholar]
  75. Lahti AC, Holcomb HH, Medoff DR, Tamminga CA. Ketamine activates psychosis and alters limbic blood flow in schizophrenia. Neuroreport. 1995a;6:869–872. doi: 10.1097/00001756-199504190-00011. [DOI] [PubMed] [Google Scholar]
  76. Lahti AC, Koffel B, LaPorte D, Tamminga CA. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology. 1995b;13:9–19. doi: 10.1016/0893-133X(94)00131-I. [DOI] [PubMed] [Google Scholar]
  77. Lahti AC, Reid MA. Is there evidence for neurotoxicity in the prodromal and early stages of schizophrenia? Neuropsychopharmacology. 2011;36:1779–1780. doi: 10.1038/npp.2011.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Large CH, Webster EL, Goff DC. The potential role of lamotrigine in schizophrenia. Psychopharmacology. 2005;181:415–436. doi: 10.1007/s00213-005-0020-9. [DOI] [PubMed] [Google Scholar]
  79. Laruelle M, Abi-Dargham A, Van Dyck CH, Gil R, D’Souza CD, Erdos J, McCance E, Fingado C, Baldwin RM, Krystal JH, Charney DS, Innis RB, Seibyl JP, Rosenblatt W, Zoghbi SS. Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad Sci USA. 1996;93:9235. doi: 10.1073/pnas.93.17.9235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010;460:525–542. doi: 10.1007/s00424-010-0809-1. [DOI] [PubMed] [Google Scholar]
  81. Lawrie SM, Abukmeil SS. Brain abnormality in schizophrenia. A systematic and quantitative review of volumetric magnetic resonance imaging studies. Br J Psychiatry. 1998;172:110–120. doi: 10.1192/bjp.172.2.110. [DOI] [PubMed] [Google Scholar]
  82. Lieberman JA, Perkins D, Belger A, Chakos M, Jarskog F, Boteva K, Gilmore J. The early stages of schizophrenia: speculations on pathogenesis, pathophysiology, and therapeutic approaches. Biol Psychiatry. 2001;50:884–897. doi: 10.1016/s0006-3223(01)01303-8. [DOI] [PubMed] [Google Scholar]
  83. Lindenmayer JP. Treatment refractory schizophrenia. Psychiatr Q. 2000;71:373–384. doi: 10.1023/a:1004640408501. [DOI] [PubMed] [Google Scholar]
  84. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. Effects of ketamine and N-methyl-D-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience. 2003;117:697–706. doi: 10.1016/s0306-4522(02)00652-8. [DOI] [PubMed] [Google Scholar]
  85. Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, Breier A. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14:301–307. doi: 10.1016/0893-133X(95)00137-3. [DOI] [PubMed] [Google Scholar]
  86. Marsman A, van den Heuvel MP, Klomp DW, Kahn RS, Luijten PR, Hulshoff Pol HE. Glutamate in schizophrenia: a focused review and meta-analysis of (1)H-MRS studies. Schizophr Bull. 2013;39:120–129. doi: 10.1093/schbul/sbr069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. McGorry PD, Nelson B, Amminger GP, Bechdolf A, Francey SM, Berger G, Riecher-Rossler A, Klosterkotter J, Ruhrmann S, Schultze-Lutter F, Nordentoft M, Hickie I, McGuire P, Berk M, Chen EY, Keshavan MS, Yung AR. Intervention in individuals at ultra-high risk for psychosis: a review and future directions. J Clin Psychiatry. 2009;70:1206–1212. doi: 10.4088/JCP.08r04472. [DOI] [PubMed] [Google Scholar]
  88. Mehta A, Prabhakar M, Kumar P, Deshmukh R, Sharma PL. Excitotoxicity: bridge to various triggers in neurodegenerative disorders. Eur J Pharmacol. 2013;698:6–18. doi: 10.1016/j.ejphar.2012.10.032. [DOI] [PubMed] [Google Scholar]
  89. Meyer-Lindenberg A. Neuroimaging and the question of neurodegeneration in schizophrenia. Prog Neurobiol. 2011;95:514–516. doi: 10.1016/j.pneurobio.2011.07.007. [DOI] [PubMed] [Google Scholar]
  90. Miyamoto S, Miyake N, Jarskog LF, Fleischhacker WW, Lieberman JA. Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry. 2012;17:1206–1227. doi: 10.1038/mp.2012.47. [DOI] [PubMed] [Google Scholar]
  91. Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–2927. doi: 10.1523/JNEUROSCI.17-08-02921.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Moghaddam B, Adams BW. Science. Vol. 281. New York, N.Y: 1998. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats; pp. 1349–1352. [DOI] [PubMed] [Google Scholar]
  93. Moghaddam B, Javitt D. From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology. 2012;37:4–15. doi: 10.1038/npp.2011.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Moghaddam B, Krystal JH. Capturing the angel in “angel dust”: twenty years of translational neuroscience studies of NMDA receptor antagonists in animals and humans. Schizophr Bull. 2012;38:942–949. doi: 10.1093/schbul/sbs075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Moncrieff J, Leo J. A systematic review of the effects of antipsychotic drugs on brain volume. Psychol Med. 2010;40:1409–1422. doi: 10.1017/S0033291709992297. [DOI] [PubMed] [Google Scholar]
  96. Nagels A, Kirner-Veselinovic A, Krach S, Kircher T. Neural correlates of S-ketamine induced psychosis during overt continuous verbal fluency. NeuroImage. 2011;54:1307–1314. doi: 10.1016/j.neuroimage.2010.08.021. [DOI] [PubMed] [Google Scholar]
  97. Nakazawa K, Zsiros V, Jiang Z, Nakao K, Kolata S, Zhang S, Belforte JE. GABAergic interneuron origin of schizophrenia pathophysiology. Neuropharmacology. 2012;62:1574–1583. doi: 10.1016/j.neuropharm.2011.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Narr KL, Toga AW, Szeszko P, Thompson PM, Woods RP, Robinson D, Sevy S, Wang Y, Schrock K, Bilder RM. Cortical thinning in cingulate and occipital cortices in first episode schizophrenia. Biol Psychiatry. 2005;58:32–40. doi: 10.1016/j.biopsych.2005.03.043. [DOI] [PubMed] [Google Scholar]
  99. Natsubori T, Inoue H, Abe O, Takano Y, Iwashiro N, Aoki Y, Koike S, Yahata N, Katsura M, Gonoi W, Sasaki H, Takao H, Kasai K, Yamasue H. Reduced Frontal glutamate+glutamine and N-acetylaspartate levels in patients with chronic schizophrenia but not in those at clinical high risk for psychosis or with first-episode schizophrenia. Schizophr Bull. 2013 doi: 10.1093/schbul/sbt124. http://dx.doi.org/10.1093/schbul/sbt124. [DOI] [PMC free article] [PubMed]
  100. Navari S, Dazzan P. Do antipsychotic drugs affect brain structure? A systematic and critical review of MRI findings. Psychol Med. 2009;39:1763–1777. doi: 10.1017/S0033291709005315. [DOI] [PubMed] [Google Scholar]
  101. Nazeri A, Chakravarty MM, Felsky D, Lobaugh NJ, Rajji TK, Mulsant BH, Voineskos AN. Alterations of superficial white matter in schizophrenia and relationship to cognitive performance. Neuropsychopharmacology. 2013;38:1954–1962. doi: 10.1038/npp.2013.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Nelson B, Yuen HP, Wood SJ, Lin A, Spiliotacopoulos D, Bruxner A, Broussard C, Simmons M, Foley DL, Brewer WJ, Francey SM, Amminger GP, Thompson A, McGorry PD, Yung AR. Long-term follow-up of a group at ultra high risk (“prodromal”) for psychosis: the PACE 400 study. JAMA Psychiatry. 2013;70:793–802. doi: 10.1001/jamapsychiatry.2013.1270. [DOI] [PubMed] [Google Scholar]
  103. Ohrmann P, Siegmund A, Suslow T, Spitzberg K, Kersting A, Arolt V, Heindel W, Pfleiderer B. Evidence for glutamatergic neuronal dysfunction in the prefrontal cortex in chronic but not in first-episode patients with schizophrenia: a proton magnetic resonance spectroscopy study. Schizophr Res. 2005;73:153–157. doi: 10.1016/j.schres.2004.08.021. [DOI] [PubMed] [Google Scholar]
  104. Okamura N, Hashimoto K, Shimizu E, Koike K, Ohgake S, Koizumi H, Kumakiri C, Komatsu N, Iyo M. Protective effect of LY379268, a selective group II metabotropic glutamate receptor agonist, on dizocilpine-induced neuropathological changes in rat retrosplenial cortex. Brain Res. 2003;992:114–119. doi: 10.1016/j.brainres.2003.08.045. [DOI] [PubMed] [Google Scholar]
  105. Olney JW. Science. Vol. 164. New York, N.Y: 1969. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate; pp. 719–721. [DOI] [PubMed] [Google Scholar]
  106. Olney JW, Farber NB. Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry. 1995;52:998–1007. doi: 10.1001/archpsyc.1995.03950240016004. [DOI] [PubMed] [Google Scholar]
  107. Olney JW, Labruyere J, Price MT. Science. Vol. 244. New York, N.Y: 1989. Pathological changes induced in cerebrocortical neurons by phencyclidine and related drugs; pp. 1360–1362. [DOI] [PubMed] [Google Scholar]
  108. Olney JW, Newcomer JW, Farber NB. NMDA receptor hypofunction model of schizophrenia. J Psychiatr Res. 1999;33:523–533. doi: 10.1016/s0022-3956(99)00029-1. [DOI] [PubMed] [Google Scholar]
  109. Olney JW, Sharpe LG. Science. Vol. 166. New York, N.Y: 1969. Brain lesions in an infant rhesus monkey treated with monsodium glutamate; pp. 386–388. [Google Scholar]
  110. Pilowsky LS, Bressan RA, Stone JM, Erlandsson K, Mulligan RS, Krystal JH, Ell PJ. First in vivo evidence of an NMDA receptor deficit in medication-free schizophrenic patients. Mol Psychiatry. 2006;11:118–119. doi: 10.1038/sj.mp.4001751. [DOI] [PubMed] [Google Scholar]
  111. Poels EM, Kegeles LS, Kantrowitz JT, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR. Glutamatergic abnormalities in schizophrenia: a review of proton MRS findings. Schizophr Res. 2014a;152:325–332. doi: 10.1016/j.schres.2013.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Poels EM, Kegeles LS, Kantrowitz JT, Slifstein M, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR. Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry. 2014b;19:20–29. doi: 10.1038/mp.2013.136. [DOI] [PubMed] [Google Scholar]
  113. Purdon SE, Valiakalayil A, Hanstock CC, Seres P, Tibbo P. Elevated 3 T proton MRS glutamate levels associated with poor Continuous Performance Test (CPT-0X) scores and genetic risk for schizophrenia. Schizophr Res. 2008;99:218–224. doi: 10.1016/j.schres.2007.11.028. [DOI] [PubMed] [Google Scholar]
  114. Rais M, van Haren NE, Cahn W, Schnack HG, Lepage C, Collins L, Evans AC, Hulshoff Pol HE, Kahn RS. Cannabis use and progressive cortical thickness loss in areas rich in CB1 receptors during the first five years of schizophrenia. Eur Neuropsychopharmacol. 2010;20:855–865. doi: 10.1016/j.euroneuro.2010.08.008. [DOI] [PubMed] [Google Scholar]
  115. Reich DL, Silvay G. Ketamine: an update on the first twenty-five years of clinical experience. Can J Anaesth. 1989;36:186–197. doi: 10.1007/BF03011442. [DOI] [PubMed] [Google Scholar]
  116. Reid MA, Stoeckel LE, White DM, Avsar KB, Bolding MS, Akella NS, Knowlton RC, den Hollander JA, Lahti AC. Assessments of function and biochemistry of the anterior cingulate cortex in schizophrenia. Biol Psychiatry. 2010;68:625–633. doi: 10.1016/j.biopsych.2010.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Ren W, Lui S, Deng W, Li F, Li M, Huang X, Wang Y, Li T, Sweeney JA, Gong Q. Anatomical and functional brain abnormalities in drug-naive first-episode schizophrenia. Am J Psychiatry. 2013;170:1308–1316. doi: 10.1176/appi.ajp.2013.12091148. [DOI] [PubMed] [Google Scholar]
  118. Reynolds LM, Cochran SM, Morris BJ, Pratt JA, Reynolds GP. Chronic phencyclidine administration induces schizophrenia-like changes in N-acetylaspartate and N-acetylaspartylglutamate in rat brain. Schizophr Res. 2005;73:147–152. doi: 10.1016/j.schres.2004.02.003. [DOI] [PubMed] [Google Scholar]
  119. Rimol LM, Hartberg CB, Nesvag R, Fennema-Notestine C, Hagler DJ, Jr, Pung CJ, Jennings RG, Haukvik UK, Lange E, Nakstad PH, Melle I, Andreassen OA, Dale AM, Agartz I. Cortical thickness and subcortical volumes in schizophrenia and bipolar disorder. Biol Psychiatry. 2010;68:41–50. doi: 10.1016/j.biopsych.2010.03.036. [DOI] [PubMed] [Google Scholar]
  120. Rothman DL, De Feyter HM, de Graaf RA, Mason GF, Behar KL. 13C MRS studies of neuroenergetics and neurotransmitter cycling in humans. NMR Biomed. 2011;24:943–957. doi: 10.1002/nbm.1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Rowland LM, Bustillo JR, Mullins PG, Jung RE, Lenroot R, Landgraf E, Barrow R, Yeo R, Lauriello J, Brooks WM. Effects of ketamine on anterior cingulate glutamate metabolism in healthy humans: a 4-T proton MRS study. Am J Psychiatry. 2005;162:394–396. doi: 10.1176/appi.ajp.162.2.394. [DOI] [PubMed] [Google Scholar]
  122. Rowland LM, Kontson K, West J, Edden RA, Zhu H, Wijtenburg SA, Holcomb HH, Barker PB. In vivo measurements of glutamate, GABA, and NAAG in schizophrenia. Schizophr Bull. 2013;39:1096–1104. doi: 10.1093/schbul/sbs092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Rusch N, Tebartz van Elst L, Valerius G, Buchert M, Thiel T, Ebert D, Hennig J, Olbrich HM. Neurochemical and structural correlates of executive dysfunction in schizophrenia. Schizophr Res. 2008;99:155–163. doi: 10.1016/j.schres.2007.05.024. [DOI] [PubMed] [Google Scholar]
  124. Schobel SA, Chaudhury NH, Khan UA, Paniagua B, Styner MA, Asllani I, Inbar BP, Corcoran CM, Lieberman JA, Moore H, Small SA. Imaging patients with psychosis and a mouse model establishes a spreading pattern of hippocampal dysfunction and implicates glutamate as a driver. Neuron. 2013;78:81–93. doi: 10.1016/j.neuron.2013.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Schultz CC, Koch K, Wagner G, Roebel M, Schachtzabel C, Gaser C, Nenadic I, Reichenbach JR, Sauer H, Schlosser RG. Reduced cortical thickness in first episode schizophrenia. Schizophr Res. 2010;116:204–209. doi: 10.1016/j.schres.2009.11.001. [DOI] [PubMed] [Google Scholar]
  126. Seeman P, Lee T. Science. Vol. 188. New York, N.Y: 1975. Antipsychotic drugs: direct correlation between clinical potency and presynaptic action on dopamine neurons; pp. 1217–1219. [DOI] [PubMed] [Google Scholar]
  127. Sharp FR, Butman M, Koistinaho J, Aardalen K, Nakki R, Massa SM, Swanson RA, Sagar SM. Phencyclidine induction of the hsp 70 stress gene in injured pyramidal neurons is mediated via multiple receptors and voltage gated calcium channels. Neuroscience. 1994;62:1079–1092. doi: 10.1016/0306-4522(94)90345-x. [DOI] [PubMed] [Google Scholar]
  128. Sharp FR, Jasper P, Hall J, Noble L, Sagar SM. MK-801 and ketamine induce heat shock protein HSP72 in injured neurons in posterior cingulate and retrosplenial cortex. Ann Neurol. 1991;30:801–809. doi: 10.1002/ana.410300609. [DOI] [PubMed] [Google Scholar]
  129. Sharp FR, Tomitaka M, Bernaudin M, Tomitaka S. Psychosis: pathological activation of limbic thalamocortical circuits by psychomimetics and schizophrenia? Trends Neurosci. 2001;24:330–334. doi: 10.1016/s0166-2236(00)01817-8. [DOI] [PubMed] [Google Scholar]
  130. Sharp JW, Petersen DL, Langford MT. DNQX inhibits phencyclidine (PCP) and ketamine induction of the hsp70 heat shock gene in the rat cingulate and retrosplenial cortex. Brain Res. 1995;687:114–124. doi: 10.1016/0006-8993(95)00477-8. [DOI] [PubMed] [Google Scholar]
  131. Shenton ME, Dickey CC, Frumin M, McCarley RW. A review of MRI findings in schizophrenia. Schizophr Res. 2001;49:1–52. doi: 10.1016/s0920-9964(01)00163-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Sprooten E, Papmeyer M, Smyth AM, Vincenz D, Honold S, Conlon GA, Moorhead TW, Job D, Whalley HC, Hall J, McIntosh AM, Owens DC, Johnstone EC, Lawrie SM. Cortical thickness in first-episode schizophrenia patients and individuals at high familial risk: a cross-sectional comparison. Schizophr Res. 2013;151:259–264. doi: 10.1016/j.schres.2013.09.024. [DOI] [PubMed] [Google Scholar]
  133. Steen RG, Hamer RM, Lieberman JA. Measurement of brain metabolites by 1H magnetic resonance spectroscopy in patients with schizophrenia: a systematic review and meta-analysis. Neuropsychopharmacology. 2005;30:1949–1962. doi: 10.1038/sj.npp.1300850. [DOI] [PubMed] [Google Scholar]
  134. Steen RG, Mull C, McClure R, Hamer RM, Lieberman JA. Brain volume in first-episode schizophrenia: systematic review and meta-analysis of magnetic resonance imaging studies. Br J Psychiatry. 2006;188:510–518. doi: 10.1192/bjp.188.6.510. [DOI] [PubMed] [Google Scholar]
  135. Stone JM, Day F, Tsagaraki H, Valli I, McLean MA, Lythgoe DJ, O’Gorman RL, Barker GJ, McGuire PK. Glutamate dysfunction in people with prodromal symptoms of psychosis: relationship to gray matter volume. Biol Psychiatry. 2009;66:533–539. doi: 10.1016/j.biopsych.2009.05.006. [DOI] [PubMed] [Google Scholar]
  136. Stone JM, Dietrich C, Edden R, Mehta MA, De Simoni S, Reed LJ, Krystal JH, Nutt D, Barker GJ. Ketamine effects on brain GABA and glutamate levels with 1H-MRS: relationship to ketamine-induced psychopathology. Mol Psychiatry. 2012;17:664–665. doi: 10.1038/mp.2011.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Stone JM, Erlandsson K, Arstad E, Bressan RA, Squassante L, Teneggi V, Ell PJ, Pilowsky LS. Ketamine displaces the novel NMDA receptor SPET probe [(1 2 3)I]CNS-1261 in humans in vivo. Nucl Med Biol. 2006;33:239–243. doi: 10.1016/j.nucmedbio.2005.12.001. [DOI] [PubMed] [Google Scholar]
  138. Stone JM, Morrison PD, Pilowsky LS. Glutamate and dopamine dysregulation in schizophrenia—a synthesis and selective review. J Psychopharmacol. 2007;21:440–452. doi: 10.1177/0269881106073126. [DOI] [PubMed] [Google Scholar]
  139. Sullivan PF, Kendler KS, Neale MC. Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch Gen Psychiatry. 2003;60:1187–1192. doi: 10.1001/archpsyc.60.12.1187. [DOI] [PubMed] [Google Scholar]
  140. Suzuki T, Remington G, Mulsant BH, Rajji TK, Uchida H, Graff-Guerrero A, Mamo DC. Treatment resistant schizophrenia and response to antipsychotics: a review. Schizophr Res. 2011;133:54–62. doi: 10.1016/j.schres.2011.09.016. [DOI] [PubMed] [Google Scholar]
  141. Tandon N, Bolo NR, Sanghavi K, Mathew IT, Francis AN, Stanley JA, Keshavan MS. Brain metabolite alterations in young adults at familial high risk for schizophrenia using proton magnetic resonance spectroscopy. Schizophr Res. 2013;148:59–66. doi: 10.1016/j.schres.2013.05.024. [DOI] [PubMed] [Google Scholar]
  142. Theberge J, Al-Semaan Y, Williamson PC, Menon RS, Neufeld RW, Rajakumar N, Schaefer B, Densmore M, Drost DJ. Glutamate and glutamine in the anterior cingulate and thalamus of medicated patients with chronic schizophrenia and healthy comparison subjects measured with 4.0-T proton MRS. Am J Psychiatry. 2003;160:2231–2233. doi: 10.1176/appi.ajp.160.12.2231. [DOI] [PubMed] [Google Scholar]
  143. Theberge J, Bartha R, Drost DJ, Menon RS, Malla A, Takhar J, Neufeld RW, Rogers J, Pavlosky W, Schaefer B, Densmore M, Al-Semaan Y, Williamson PC. Glutamate and glutamine measured with 4.0 T proton MRS in never-treated patients with schizophrenia and healthy volunteers. Am J Psychiatry. 2002;159:1944–1946. doi: 10.1176/appi.ajp.159.11.1944. [DOI] [PubMed] [Google Scholar]
  144. Theberge J, Williamson KE, Aoyama N, Drost DJ, Manchanda R, Malla AK, Northcott S, Menon RS, Neufeld RW, Rajakumar N, Pavlosky W, Densmore M, Schaefer B, Williamson PC. Longitudinal grey-matter and glutamatergic losses in first-episode schizophrenia. Br J Psychiatry. 2007;191:325–334. doi: 10.1192/bjp.bp.106.033670. [DOI] [PubMed] [Google Scholar]
  145. Tomitaka S, Tomitaka M, Tolliver BK, Sharp FR. Bilateral blockade of NMDA receptors in anterior thalamus by dizocilpine (MK-801) injures pyramidal neurons in rat retrosplenial cortex. Eur J Neurosci. 2000;12:1420–1430. doi: 10.1046/j.1460-9568.2000.00018.x. [DOI] [PubMed] [Google Scholar]
  146. Torres US, Portela-Oliveira E, Borgwardt S, Busatto GF. Structural brain changes associated with antipsychotic treatment in schizophrenia as revealed by voxel-based morphometric MRI: an activation likelihood estimation meta-analysis. BMC Psychiatry. 2013;13:342. doi: 10.1186/1471-244X-13-342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Tsai G, Coyle JT. Glutamatergic mechanisms in schizophrenia. Annu Rev Pharmacol Toxicol. 2002;42:165–179. doi: 10.1146/annurev.pharmtox.42.082701.160735. [DOI] [PubMed] [Google Scholar]
  148. Tsai G, van Kammen DP, Chen S, Kelley ME, Grier A, Coyle JT. Glutamatergic neurotransmission involves structural and clinical deficits of schizophrenia. Biol Psychiatry. 1998;44:667–674. doi: 10.1016/s0006-3223(98)00151-6. [DOI] [PubMed] [Google Scholar]
  149. Valli I, Stone J, Mechelli A, Bhattacharyya S, Raffin M, Allen P, Fusar-Poli P, Lythgoe D, O’Gorman R, Seal M, McGuire P. Altered medial temporal activation related to local glutamate levels in subjects with prodromal signs of psychosis. Biol Psychiatry. 2011;69:97–99. doi: 10.1016/j.biopsych.2010.08.033. [DOI] [PubMed] [Google Scholar]
  150. Venkatasubramanian G, Jayakumar PN, Gangadhar BN, Keshavan MS. Automated MRI parcellation study of regional volume and thickness of prefrontal cortex (PFC) in antipsychotic-naive schizophrenia. Acta Psychiatr Scand. 2008;117:420–431. doi: 10.1111/j.1600-0447.2008.01198.x. [DOI] [PubMed] [Google Scholar]
  151. Vita A, De Peri L, Deste G, Sacchetti E. Progressive loss of cortical gray matter in schizophrenia: a meta-analysis and meta-regression of longitudinal MRI studies. Transl Psychiatry. 2012;2:e190. doi: 10.1038/tp.2012.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Voineskos AN, Foussias G, Lerch J, Felsky D, Remington G, Rajji TK, Lobaugh N, Pollock BG, Mulsant BH. Neuroimaging evidence for the deficit subtype of schizophrenia. JAMA Psychiatry. 2013;70:472–480. doi: 10.1001/jamapsychiatry.2013.786. [DOI] [PubMed] [Google Scholar]
  153. Vollenweider FX, Geyer MA. A systems model of altered consciousness: integrating natural and drug-induced psychoses. Brain Res Bull. 2001;56:495–507. doi: 10.1016/s0361-9230(01)00646-3. [DOI] [PubMed] [Google Scholar]
  154. Weiser M, Davidson M, Noy S. Comments on risk for schizophrenia. Schizophr Res. 2005;79:15–21. doi: 10.1016/j.schres.2005.05.005. [DOI] [PubMed] [Google Scholar]
  155. Weiser M, Heresco-Levy U, Davidson M, Javitt DC, Werbeloff N, Gershon AA, Abramovich Y, Amital D, Doron A, Konas S, Levkovitz Y, Liba D, Teitelbaum A, Mashiach M, Zimmerman Y. A multicenter, add-on randomized controlled trial of low-dose D-serine for negative and cognitive symptoms of schizophrenia. J Clin Psychiatry. 2012;73:e728–e734. doi: 10.4088/JCP.11m07031. [DOI] [PubMed] [Google Scholar]
  156. White T, Andreasen NC, Nopoulos P, Magnotta V. Gyrification abnormalities in childhood- and adolescent-onset schizophrenia. Biol Psychiatry. 2003;54:418–426. doi: 10.1016/s0006-3223(03)00065-9. [DOI] [PubMed] [Google Scholar]
  157. Wood SJ, Kennedy D, Phillips LJ, Seal ML, Yucel M, Nelson B, Yung AR, Jackson G, McGorry PD, Velakoulis D, Pantelis C. Hippocampal pathology in individuals at ultra-high risk for psychosis: a multi-modal magnetic resonance study. NeuroImage. 2010;52:62–68. doi: 10.1016/j.neuroimage.2010.04.012. [DOI] [PubMed] [Google Scholar]
  158. van Haren NE, Cahn W, Hulshoff Pol HE, Kahn RS. The course of brain abnormalities in schizophrenia: can we slow the progression? J Psychopharmacol. 2012;26:8–14. doi: 10.1177/0269881111408964. [DOI] [PubMed] [Google Scholar]
  159. van Haren NE, Hulshoff Pol HE, Schnack HG, Cahn W, Brans R, Carati I, Rais M, Kahn RS. Progressive brain volume loss in schizophrenia over the course of the illness: evidence of maturational abnormalities in early adulthood. Biol Psychiatry. 2008;63:106–113. doi: 10.1016/j.biopsych.2007.01.004. [DOI] [PubMed] [Google Scholar]
  160. van Haren NE, Schnack HG, Cahn W, van den Heuvel MP, Lepage C, Collins L, Evans AC, Hulshoff Pol HE, Kahn RS. Changes in cortical thickness during the course of illness in schizophrenia. Arch Gen Psychiatry. 2011;68:871–880. doi: 10.1001/archgenpsychiatry.2011.88. [DOI] [PubMed] [Google Scholar]
  161. van Os J, Kapur S. Schizophrenia. Lancet. 2009;374:635–645. doi: 10.1016/S0140-6736(09)60995-8. [DOI] [PubMed] [Google Scholar]

RESOURCES