Abstract
Accumulating evidence of glutamatergic abnormalities in the brains of schizophrenia patients has led to efforts to target various components of glutamatergic signaling as potential new approaches for schizophrenia. Exciting research suggests that metabotropic glutamate (mGlu) receptors could provide a fundamentally new approach for better symptomatic relief in patients with schizophrenia. In preclinical studies, the mGlu5 receptor positive allosteric modulators (PAMs) show efficacy in animal models relevant for all symptom domains in schizophrenia. Interestingly, biased pure mGlu5 receptor PAMs that do not potentiate coupling of mGlu5 receptors to N-methyl-D-aspartate (NMDA) receptors lack neurotoxic effects associated with mGlu5 PAMs that enhance coupling to NMDA receptors or have allosteric agonist activity. This provides a better therapeutic profile for treating schizophrenia-like symptoms. Additionally, the mGlu1 receptor PAMs modulate dopamine release in the striatum, which may contribute to their antipsychotic-like effects. Besides group I mGlu (mGlu1 and mGlu5) receptors, agonists of mGlu2/3 receptors also induce robust antipsychotic-like and procognitive effects in rodents and may be effective in treating symptoms of schizophrenia in a selective group of patients. Additionally, mGlu2/4 receptor heterodimers modulate glutamatergic neurotransmission in the prefrontal cortex at selective synapses activated in schizophrenia and therefore hold potential as novel antipsychotics. Excitingly, the mGlu3 receptor activation can enhance cognition in rodents, suggesting that mGlu3 receptor agonist/PAM could provide a novel approach for the treatment of cognitive deficits in schizophrenia. Collectively, the development of mGlu receptor-specific ligands may provide an alternative approach to meet the clinical need for safer and more efficacious therapeutics for schizophrenia.
SIGNIFICANCE STATEMENT
The currently available antipsychotic medications do not show significant efficacy for treating negative symptoms and cognitive deficits in schizophrenia. Emerging preclinical and clinical literature suggests that pharmacological targeting of metabotropic glutamate receptors could potentially provide an alternative approach for designing safer and more efficacious therapeutics for treating schizophrenia.
Introduction
Schizophrenia is a heterogeneous neuropsychiatric disorder that affects around 1% of the population worldwide (Perälä et al., 2007). The clinical features of the disease are characterized by three symptom domains: positive symptoms that include hallucinations, delusions, and thought disorders; negative symptoms including blunted emotions, anhedonia, and social withdrawal; and cognitive deficits such as impairments in attention, executive function, and working memory. The currently prescribed therapeutic agents (both typical and atypical antipsychotics) show efficacy in reducing the severity of positive symptoms but have minimal impact on negative symptoms and cognitive deficits associated with schizophrenia (Li et al., 2016a). Further, a significant portion of patients do not respond to these medications, and many patients discontinue treatment because of the class-related adverse effects, such as extrapyramidal side effects (parkinsonism, bradykinesia, dystonic reactions, tardive dyskinesia, and tremor), sedation, and metabolic side effects (weight gain, hyperlipidemia, and type II diabetes) (Li et al., 2016a). Thus, there is an unmet clinical need for designing safer therapeutic agents acting at new targets underlying the pathophysiology of schizophrenia.
Despite evidence for a central role of dysfunction of dopaminergic signaling in schizophrenia, the inability of current antipsychotics to treat schizophrenia effectively suggests that a hyperdopaminergic state does not account for all major symptoms of this disorder. Accumulating evidence suggests that dysfunction of glutamatergic signaling may contribute to the pathogenesis of schizophrenia (Coyle, 2006; Moghaddam and Javitt, 2012; Hu et al., 2015; Stahl, 2018). Pharmacological evidence for the role of glutamate in schizophrenia centers on the clinical findings that administration of the noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonists like phencyclidine (PCP) and ketamine induce/exacerbate schizophrenia-like symptoms in healthy individuals (Hu et al., 2015). Based on these clinical observations, NMDA antagonists are used preclinically to induce various schizophrenia-like behaviors, such as hyperactivity, repetitive behavior, sensorimotor gating deficits, and motivational and cognitive impairments in rodents (Goff and Coyle, 2001; Barnes et al., 2017; Lee and Zhou, 2019). Interestingly, animals with genetic knockdown of the NR1 subunit of NMDA receptors exhibit a full range of behavioral phenotypes that may be relevant for schizophrenia, including hyperlocomotion, stereotypy, heightened anxiety-like behavior, lack of cognitive flexibility, and other forms of memory impairments (Mohn et al., 1999; Belforte et al., 2010). Similarly, animals lacking NR2A or NR2B subunits of NMDA receptors show behavioral and neurophysiological changes that may reflect pathophysiology observed in schizophrenia (Ito et al., 1997; Sprengel et al., 1998; Duncan et al., 2004; Brigman et al., 2008; von Engelhardt et al., 2008; Belforte et al., 2010; Kannangara et al., 2015). Collectively, these studies have led to the hypothesis that pathologic changes in glutamatergic circuits and NMDA receptor signaling may contribute to the pathophysiology associated with schizophrenia.
Interestingly, genome-wide association studies (GWAS) and linkage studies have shown a significant association of gene encoding for the components of glutamate receptors and maintenance of glutamatergic neurotransmission with schizophrenia (Timms et al., 2013; Schizophrenia Working Group of the Psychiatric Genomics, 2014; Pocklington et al., 2015; Pers et al., 2016). These studies, along with extensive preclinical evidence mentioned above, reinforce the hypothesis that disruption of glutamatergic signaling and NMDA hypofunction may contribute to the pathophysiology of schizophrenia. Based on this, it is conceivable that pharmacological agents that reverse pathologic changes in NMDA receptor function or other aspects of glutamatergic signaling could provide symptomatic relief to patients with schizophrenia. However, direct modulation of NMDA receptors produces neuronal excitotoxicity and seizures, prohibiting direct NMDA receptor targeting as a viable therapeutic option. Therefore, it will be important to develop more optimized compounds targeting other components of the glutamatergic system and testing them in rodent models for treating schizophrenia-like deficits.
A growing body of preclinical and clinical evidence raise the exciting possibility that targeting metabotropic glutamate (mGlu) receptors may allow more subtle regulation of glutamatergic neurotransmission in key brain circuits that are relevant for schizophrenia and may provide novel drug targets for treatment of this disorder. Metabotropic glutamate receptors are G protein-coupled receptors and are classified into three groups based on amino acid sequence homology, G protein binding, pharmacological profile, and signaling. Group I mGlu receptors include mGlu1 and mGlu5 receptors, group II includes mGlu2 and mGlu3 receptors, and group III includes mGlu4, mGlu6, mGlu7, and mGlu8 receptors (Niswender and Conn, 2010; Dogra and Conn, 2021). These receptors signal via coupling with different G-proteins: Group I mGlu receptors typically couple to Gq/11 proteins and group II and III receptors are coupled to Gi/o proteins (Niswender and Conn, 2010). Their ability to modulate the glutamatergic transmission in the brain areas implicated in schizophrenia makes them exciting targets for developing improved pharmacotherapies for schizophrenia.
Genomic Variants in GRM5 and mGlu5 Receptor Hypofunction Are Associated with Schizophrenia
GRM5 (the gene encoding for mGlu5 receptor) has emerged as a promising target for the treatment of various symptoms of schizophrenia. Polymorphisms in GRM5 are associated with schizophrenia (Devon et al., 2001) and impaired cognition in patients with schizophrenia (Matosin et al., 2018). A recent study by Wang et al. (2020) has reported reduced mGlu5 receptor signaling in the postmortem dorsolateral prefrontal cortex (DLPFC) of patients with schizophrenia. The authors also observed increased serine and tyrosine phosphorylation of mGlu5 receptors in DLPFC, which may cause receptor desensitization leading to reduced mGlu5 receptor signaling observed in the patients with schizophrenia (Wang et al., 2020). Interestingly, lower mGlu5 receptor availability in the left temporal cortex was associated with higher levels of negative symptoms and worse performance in cognitive tasks in male patients with chronic schizophrenia (Régio Brambilla et al., 2020). These findings support the concept that dysregulation of mGlu5 receptor signaling/function may underly the pathophysiology of schizophrenia.
These clinical studies are complemented by studies in GRM5 knockout (KO) mice displaying sensorimotor gating deficits, spontaneous hyperactivity, and loss of NMDA receptor-mediated components of some forms of synaptic plasticity (Jia et al., 1998; Brody et al., 2004a,b; Brody and Geyer, 2004; Burrows et al., 2015). These KO mice also exhibit abnormalities in sleep and neural oscillatory processing similar to patients with schizophrenia (Aguilar et al., 2020). Further, GRM5 KO mice display impaired performance on discrimination learning and reversal learning in a trial-unique nonmatching-to-location task indicating learning and memory deficits in these mice (Zeleznikow-Johnston et al., 2018). Also, deletion of GRM5 from cortical pyramidal neurons causes increased novelty-induced locomotion, and systemic treatment with the psychostimulant methylphenidate can further enhance their locomotion (Jew et al., 2013). These findings indicate that GRM5 KO mice display deficits modeling all symptom domains of schizophrenia and reiterate the critical roles of mGlu5 receptors in schizophrenia pathophysiology. Similarly, administration of mGlu5 receptor negative allosteric modulators (NAMs) evoke behavioral deficits correlated with schizophrenia (Koros et al., 2007; Swedberg et al., 2014) and potentiate behavioral deficits induced by psychotomimetic agents (Henry et al., 2002; Campbell et al., 2004), providing evidence linking mGlu5 receptors to schizophrenia.
mGlu5 Potentiators Reduce Behavioral Disruptions That Are Relevant for Schizophrenia in Rodent Models
The mGlu5 receptor positive allosteric modulators (PAMs) can induce cognition-enhancing effects, including improvements in object recognition memory, attenuation of conditioned avoidance response, and reduced impulsivity in the five-choice serial reaction time test in rodents (Liu et al., 2008; Uslaner et al., 2009; Schlumberger et al., 2010; Huang et al., 2016; Yang et al., 2016). Also, systemic administration of the mGlu5 receptor PAMs CDPPB and ADX427273 increases hippocampus-dependent spatial learning in the Morris water maze test (Ayala et al., 2009). Similarly, another mGlu5 receptor PAM, DPFE, enhanced the acquisition of contextual fear conditioning in rats (Gregory et al., 2013). Interestingly, the mGlu5 receptor agonists/PAMs CHPG, CDPPB, ADX47273, BMS-955829, DPFE, and DFB were able to reverse the cognitive deficits and motivational impairments induced by NMDA receptor antagonists as well as hyperactivity induced by amphetamine, apomorphine, and PCP (Liu et al., 2008; Stefani and Moghaddam, 2010; Vales et al., 2010; Gastambide et al., 2012; Gregory et al., 2013; LaCrosse et al., 2015; Yang et al., 2016). These studies support the hypothesis that allosteric potentiation of the mGlu5 receptors could provide a novel pharmacotherapeutic approach for treating multiple symptom domains in schizophrenia.
Newer mGlu5 receptor-selective PAMs also show efficacy in rodent models that have traditionally been used to predict therapeutic efficacy of various drugs for schizophrenia. For example, the mGlu5 receptor-selective PAMs VU0092273 and VU0364289 dose-dependently reversed amphetamine-induced hyperactivity in rodents, a model predictive of antipsychotic activity (Rodriguez et al., 2010; Noetzel et al., 2012; Gregory et al., 2013). Similar antipsychotic-like effects were also observed with a dihydrothiazolopyridone class of mGlu5 receptor PAM (Bartolomé-Nebreda et al., 2013). Since the mGlu5 receptors are closely associated with NMDA receptors (Alagarsamy et al., 1999; Benquet et al., 2002; Collett and Collingridge, 2004; O'Riordan et al., 2018), an early hypothesis was that targeting mGlu5 receptors may exert these effects by augmenting NMDA receptor function to mitigate the NMDA receptor hypofunction and symptoms observed in schizophrenia.
Biased mGlu5 Receptor PAMs That Do Not Potentiate Coupling of mGlu5 Receptors to NMDA Receptors or Exert Allosteric Agonist Activity Do Not Induce Observable Adverse Effects
Despite the promising effects of mGlu5 receptor PAMs in the preclinical studies, the development of these compounds for the treatment of schizophrenia has been hampered by neurotoxicology issues, possibly related to intrinsic allosteric agonist activity (ago-PAMs) and excessive activation of NMDA receptors (Rook et al., 2013; Parmentier-Batteur et al., 2014). Early studies revealed that mGlu5 receptor ago-PAMs induce seizures and other adverse effects that are not as prominent with pure mGlu5 receptor PAMs that lack intrinsic allosteric agonist activity (Rook et al., 2013; Parmentier-Batteur et al., 2014). Furthermore, the known adverse effects associated with overactivation of NMDA receptors raised the possibility that mGlu5 receptor PAMs that are biased away from NMDA receptor potentiation may lack excitotoxic/seizure-inducing profiles (Rook et al., 2015). Supporting this theory, the biased mGlu5 receptor PAM VU0409551, which does not enhance mGlu5 receptor-mediated potentiation of NMDA receptor currents, induces robust antipsychotic-like (MK801-induced hyperlocomotion) and cognition-enhancing effects (novel object recognition and contextual fear conditioning) in wild-type (WT) mice (Rook et al., 2015). It also rescued deficits in contextual fear conditioning and synaptic plasticity in serine racemase KO mice, a genetic model that exhibits several behavioral abnormalities observed in schizophrenia (Balu et al., 2016). The studies with VU0409551 suggest that biased mGlu5 receptor PAMs lacking mGlu5 receptor-mediated potentiation of NMDA receptor currents retain antipsychotic-like and cognition-enhancing effects in rodent models relevant for schizophrenia. Thus, the initial hypothesis that mGlu5 receptor PAMs induce their beneficial effects by potentiating NMDA receptor currents must be reevaluated. The mechanistic studies revealed that VU0409551 could enhance cognition independent of NMDA receptor activation (Balu et al., 2016). Further, biased mGlu5 receptor PAM may increase cognition by mGlu5 receptor-mediated potentiation of excitability of hippocampal Cornu Ammonis 1 (CA1) pyramidal neurons (Mannaioni et al., 2001) or endocannabinoid-mediated depression of inhibitory neurotransmission onto pyramidal neurons (Xu et al., 2014). Collectively, the above-mentioned studies propose that a detailed understanding of the mechanisms of action of mGlu5 receptor PAMs might help to develop safer compounds with robust efficacy for treating schizophrenia.
mGlu1 Receptor PAMs May Have Potential Antipsychotic Effects
Frank and coworkers (2011) identified nonsynonymous single nucleotide polymorphisms (nsSNPs) within a functionally important cysteine-rich domain and the first transmembrane helix of the mGlu1 receptor in patients with schizophrenia. Subsequently, a study involving 605 controls and 450 patients with schizophrenia confirmed the presence of deleterious mutations in GRM1 (the gene encoding for mGlu1 receptor) in patients with schizophrenia (Ayoub et al., 2012). Also, these mutations were inheritable and were also detected in relatives with other neuropsychiatric disorders, including depression and anxiety. In vitro analysis using mGlu1 receptors bearing various schizophrenia-associated GRM1 mutations indicated altered cell surface receptor expression and reduced downstream signaling (Ayoub et al., 2012; Cho et al., 2014). Excitingly, the selective mGlu1 receptor PAMs Ro 07-11401, VU0483605, and VU0483737 were able to potentiate signaling by the mutant receptors and thereby reduce deficits in mGlu1 receptor signaling (Cho et al., 2014). Similarly, another set of mGlu1 receptor PAMs based on an N-(3-chloro-4-(1,3-dioxoisoindolin-2-yl)phenyl)-3-methylfuran-2-carboxamide scaffold also potentiated the function of mGlu1 receptors mutated for schizophrenia-associated deleterious GRM1 mutations (Garcia-Barrantes et al., 2015). These findings indicate that schizophrenia patients with specific mutations may be responsive to interventions with mGlu1 receptor PAMs and highlight the effect of the clinical heterogeneity of schizophrenia on disease prognosis.
The mGlu1 Receptor Ligands Display Antipsychotic-Like Effects in Rodents
Interestingly, recent studies revealed that the mGlu1 receptor PAM VU6004909 reduced amphetamine-induced hyperlocomotion and disruptions in prepulse inhibition (PPI) in mice (Yohn et al., 2020). These effects were absent after administration of cannabinoid type 2 (CB2) receptor antagonist, which indicates that the antipsychotic-like effects of VU6004909 are dependent on CB2 receptor activation. Further, mechanistic studies reveal that VU6004909 inhibits dopamine release in the striatum, not in the nucleus accumbens, and a crosstalk between mGlu1 and muscarinic M4 receptors has been suggested for this inhibition on dopamine release (Yohn et al., 2020). Because the mutations in GRM1 reduce mGlu1 receptor signaling and GRM1 KO mice display deficits in PPI (Brody et al., 2003), it is possible that altered receptor expression may contribute to the hyperdopaminergic state observed in schizophrenia. Therefore, mGlu1 receptor PAMs may act by correcting striatal dopamine hyperactivity leading to the antipsychotic-like activity in models mimicking schizophrenia symptomatology. Recently, it has been reported that the mGlu1 receptor PAM VU6004909 can reverse the cortical hyperactivity and cognitive deficits induced by MK801 treatment (Maksymetz et al., 2021). Future studies are needed to investigate the efficacy of mGlu1 receptor PAMs on other behavioral correlates of cognitive and negative symptoms of schizophrenia.
It is important to note that early preclinical behavioral studies also argued in favor of employing mGlu1 receptor antagonism as a pharmacotherapeutic approach to schizophrenia. For example, the mGlu1 receptor NAMs FTIDC and CFMTI blocked methamphetamine-induced hyperactivity and reduced methamphetamine-induced disruption in PPI in rodents (Satow et al., 2008, 2009). Also, CFMTI and JNJ16259685 (mGlu1 receptor antagonists) improved MK801-induced impairments in social memory (Satow et al., 2009; Hikichi et al., 2013), suggesting that mGlu1 receptor NAMs could be effective for the treatment of some impairments associated with schizophrenia. At present, the mechanistic basis for overlapping actions of mGlu1 receptor PAMs and mGlu1 receptor NAMs is not understood.
Group II (mGlu2/3) Receptor Agonists Show Efficacy for Treating Behavioral Correlates of Schizophrenia
Agonists of mGlu2/3 receptors can reverse the behavioral effects of NMDA receptor antagonists, including induction of hyperlocomotion, stereotypy, and heightened anxiety in rodents (Moghaddam and Adams, 1998; Rorick-Kehn et al., 2007; Watanabe et al., 2020). Further, the mGlu2/3 receptor agonist LY354740 reduced PCP-induced working memory deficits in mice (Moghaddam and Adams, 1998) and improved working memory impairments induced by ketamine infusion in healthy human subjects (Krystal et al., 2005). These and other studies have stimulated efforts to develop mGlu2/3 receptor agonists as novel potential antipsychotic agents. Also, mGlu2/3 receptor agonists attenuate various effects induced by the hallucinogen 2,5-Dimethoxy-4-iodoamphetamine (DOI) in rodents, including expression of immediate early gene, c-FOS, in the cortex (Zhai et al., 2003; González-Maeso et al., 2008), increased excitatory synaptic responses in the medial prefrontal cortex (mPFC) (Marek et al., 2000), and head twitch response (Gewirtz and Marek, 2000; González-Maeso et al., 2008). In addition, the mGlu2/3 receptor agonists MGS0008 and LY404039 inhibited conditioned avoidance responses in WT mice (Takamori et al., 2003; Rorick-Kehn et al., 2007). Recently, our group reported that the mGlu2/3 receptor agonist LY379268 enhances associative learning as evidenced by increased freezing in trace fear conditioning in WT mice (Dogra et al., 2021). Further, using mGlu2 and mGlu3 receptor-specific NAMs, we showed that the mGlu3 receptor is mediating cognition-enhancing effects of LY379268. Interestingly, the mGlu2/3 receptor agonist LY379268 was able to revert PCP-induced deficits in associative learning (Dogra et al., 2021). All of these studies indicate the potential utility of mGlu2/3 receptor agonists for the treatment of various schizophrenia-associated symptoms.
Clinical Trials Using mGlu2/3 Receptor Agonists Yielded Inconclusive Results
Based on the extensive preclinical literature, Eli Lilly & Co. launched LY2140023 (pomaglumetad methionil, prodrug of the active mGlu2/3 receptor agonist LY404039) into clinical trials, where it showed efficacy for improving total Positive and Negative Syndrome Scale (PANSS) scores as well as positive and negative symptoms of schizophrenia compared with placebo control in early phase II clinical trials (Patil et al., 2007; Kinon and Gómez, 2013). Excitingly, LY2140023 treatment did not induce any extrapyramidal side effects, increase in mean serum prolactin, or weight gain (Patil et al., 2007).
These encouraging proof-of-concept trials were followed by a second phase II dose-response trial in which neither LY2140023 nor olanzapine demonstrated significant efficacy compared with placebo (Kinon et al., 2011). This led to inconclusive clinical trials which were likely due to high placebo response in patients with schizophrenia. Another phase II study assessed the efficacy of LY2140023 for 24 weeks and found improvements in PANSS total score similar to the standard of care (olanzapine, risperidone, or aripiprazole) group over the initial 6 to 8 weeks of treatment, but at the endpoint lesser improvement was observed in the LY2140023 group (Adams et al., 2013). Besides, trials including LY2140023 as a monotherapy or adjunctive treatment failed to show efficacy for improving symptoms (Adams et al., 2013; Stauffer et al., 2013), which led to the cessation of the LY2140023 drug development program for schizophrenia. However, a post hoc analysis of all clinical trials showed that LY2140023 displayed therapeutic efficacy in subgroups of patients who were early in disease (≤3 years) or who were previously treated with dopamine D2 receptor drugs and were never treated with 5-hydroxy-tryptamine (5-HT)2A receptor blockers (Kinon et al., 2015). These promising results indicate the need for testing mGlu2/3 receptor agonists in patients with schizophrenia recruited based on disease duration, history of previous medication, and genetic background. Further, it will be important to tease apart the relative contributions of mGlu2 and mGlu3 receptors in mediating beneficial effects of mGlu2/3 receptor agonists.
Recently, LY2140023 was tested in phase Ib proof-of-concept studies in which high doses of LY2140023 (320 mg/day for 10 days) significantly reduced ketamine-induced Brief Psychiatric Rating Scale (BPRS) total symptoms (Kantrowitz et al., 2020). However, it did not significantly inhibit ketamine-induced changes in the pharmacological blood-oxygen-level dependent (pharmacoBOLD) signals in the dorsal anterior cingulate cortex, suggesting that the tested dose might still be too low for optimal target engagement. Besides LY2140023, a novel mGlu2/3 receptor agonist prodrug TS-134 (MGS0274 besylate) entered the clinical trials and was found to be safe and well tolerated in a double-blinded and placebo-controlled dose-response phase I clinical trial conducted in healthy subjects (Watanabe et al., 2020). Further clinical studies reported reductions in both ketamine-induced BPRS positive symptoms and pharmacoBOLD in the dorsal anterior cingulate cortex, left caudate, and nucleus accumbens after treatment with a low dose of TS-134 (20 mg/day for 6 days) (Kantrowitz et al., 2020). These results provide evidence of symptom reduction and target engagement by a mGlu2/3 receptor agonist and further suggest that dose optimization and characterization of changes in the glutamatergic neurotransmission are required to fully harness the therapeutic potential of mGlu2/3 receptor agonists. Also, it will be exciting to evaluate the therapeutic effects of TS-134 in patients with schizophrenia.
mGlu2 Receptor PAMs May Have Potential Antipsychotic Activity
In terms of subtype selectivity based on the studies performed with GRM2 (the gene encoding for mGlu2 receptor) and GRM3 (the gene encoding for mGlu3 receptor) KO mice, it has been suggested that the mGlu2 receptor, not the mGlu3 receptor, mediates the antipsychotic effects of mGlu2/3 receptor agonists in rodents (Spooren et al., 2000; Woolley et al., 2008). In the past decade, several preclinical studies have shown antipsychotic-like and anxiolytic-like effects of the mGlu2 receptor PAMs CBiPES, BINA, TASP0443294, TASP0433864, JNJ40411813/ADX71149, and JNJ-42153605 (Johnson et al., 2003, 2005; Galici et al., 2005, 2006; Govek et al., 2005; Benneyworth et al., 2007; Hiyoshi et al., 2014; Hikichi et al., 2015; Lavreysen et al., 2015). Another mGlu2 receptor PAM, SAR218645, has been shown to improve cognitive symptoms induced by an NMDA receptor antagonist, reverse working memory impairments in NR1 KO mice, and reverse amphetamine-induced disruptions in sensory processing and attention (Griebel et al., 2016). These studies point toward the SAR218645 class of PAMs as a promising candidate for the treatment of cognitive impairments in schizophrenia, especially in patients with anomalous attention and sensory gating abilities.
Based on the preclinical studies, two mGlu2 receptor PAMs, JNJ40411813 (Salih et al., 2015) and AZD8529 (Litman et al., 2016), have entered clinical trials. Both PAMs displayed safety and tolerability in healthy subjects. JNJ40411813 ameliorated deficits in attention and episodic memory in the selective population and reduced ketamine-induced negative symptoms (Salih et al., 2015). These promising trials suggest that patients with residual negative symptoms are most likely to benefit from treatment with JNJ40411813. The second compound, AZD8529, did not show any extrapyramidal motor side effects, but it failed to show any significant improvement in positive and negative symptom subscale and PANSS total score as compared with placebo (Litman et al., 2016). To note, AZD8529 was tested at only one dose selected from tolerability data obtained from the prior healthy volunteer and preclinical studies. Thus, it is possible that low systemic exposure at this dose failed to engage mGlu2 receptors or induce detectable effects. Therefore, further studies including multiple dosages of AZD8529 are warranted. Taken together, these studies reveal the potential shortcomings of preclinical research and the difficulty in translating preclinical research findings to clinics. Furthermore, considering the underlying heterogeneity of patients with schizophrenia, it may be best to test mGlu2/3 receptor agonists/mGlu2 receptor PAMs in patient populations recruited based on genotype and clinical symptoms.
Genetic Variants in GRM3 Are Associated with Schizophrenia
Emerging evidence suggests an association between genetic variations in GRM3 and risk for schizophrenia and cognitive deficits in schizophrenia (Fujii et al., 2003; Egan et al., 2004; Bishop et al., 2011, 2015; Chang et al., 2015; Saini et al., 2017). Interestingly, polymorphisms in GRM3 may predict improvement in negative symptoms in patients with schizophrenia treated with antipsychotic medications (Bishop et al., 2005, 2015; Fijal et al., 2009). Also, one GRM3 polymorphism, rs1468412, was associated with worsening of spatial working memory performance after antipsychotic treatment (Bishop et al., 2015). All of these genetic association studies suggest an important pharmacogenetic relationship between GRM3 polymorphisms and changes in cognitive and negative symptom response to antipsychotic treatment. Furthermore, this information can be used to identify patients with schizophrenia who are susceptible to adverse cognitive effects induced by antipsychotic medications. These gene polymorphisms studies are supported by findings indicating impaired working memory in GRM3 KO mice (Lainiola et al., 2014; De Filippis et al., 2015), further stipulating an essential role of the mGlu3 receptor in some forms of cognition.
mGlu3 Receptor Is a Promising Target for Enhancing Cognition in Schizophrenia
The exciting gene association studies encouraged the researchers to investigate the mechanisms by which the mGlu3 receptor regulates cognition and cognitive impairments associated with schizophrenia. Owing to the lack of receptor-specific compounds, early preclinical studies used N-acetylaspartylglutamate (NAAG) peptidase (an enzyme that inactivates the peptide transmitter) inhibitors. NAAG is a peptide neurotransmitter that activates mGlu3 receptors (Wroblewska et al., 1997) and is known to enhance cognition (Neale and Olszewski, 2019). Therefore, drugs that block the inactivation of synaptically released NAAG (NAAG peptidase inhibitors) activate mGlu3 receptors and can enhance cognition (Neale and Olszewski, 2019). Also, NAAG peptidase inhibitors blocked MK801-induced impairments in object recognition and motor activation induced by PCP and amphetamine (Olszewski et al., 2012b). These inhibitors were able to rescue PCP-induced motor activation and stereotypic behavior in WT mice (Olszewski et al., 2004) but not in GRM3 KO mice (Olszewski et al., 2012a). Interestingly, they reduced PCP-induced glutamate and dopamine release in the prefrontal cortex (PFC) and the nucleus accumbens (Zuo et al., 2012) that may account for their antipsychotic-like effects. To note, mice lacking the enzymes that synthesize NAAG in the central nervous system (CNS) have impaired object recognition memory (Becker et al., 2021), suggesting an important role of NAAG (and mGlu3 receptors) in cognition. In summary, the above-mentioned literature raises the potential therapeutic utility of targeting mGlu3 receptors for treating schizophrenia and the need for further studies aimed at understanding the mechanisms by which mGlu3 receptors may enhance cognition in schizophrenia.
Using recently developed mGlu3 specific ligands, mechanistic studies revealed that the mGlu3 receptor modulates synaptic plasticity within the PFC and hippocampus (Walker et al., 2015; Joffe et al., 2019; Dogra et al., 2021). Interestingly, mGlu3 receptors functionally interact with mGlu5 receptors in the CNS (Di Menna et al., 2018; Dogra et al., 2021) and enhance mGlu5 receptor-mediated somatic Ca2+ mobilization in the cortical pyramidal neurons. Also, activation of mGlu5 receptor is required for mGlu3 receptor-mediated long-term depression in the PFC (Di Menna et al., 2018) and extinction of fear memories (Walker et al., 2015). Further, a nonhuman primate study showed that an increase in the endogenous mGlu3 receptor agonist NAAG enhances DLPFC delay cell firing during a working memory task (Jin et al., 2018). These studies illuminate the mechanisms by which mGlu3 receptors can modulate PFC function and cognition and suggest that mGlu3 receptor PAMs have the potential to reduce the PFC-dependent cognitive impairments associated with CNS disorders like schizophrenia.
This mGlu3 receptor-induced potentiation of mGlu5 receptor function in the PFC is also observed in the hippocampus (Dogra et al., 2021). Our group discovered that activation of mGlu3 receptors in the CA1 pyramidal neurons induces metaplastic changes to induce long-term potentiation at the SC-CA1 synapse through an mGlu5 receptor-dependent, endocannabinoid-mediated disinhibition (Dogra et al., 2021). Further, the mGlu3 receptor has been shown to shape the influence of mGlu5 receptors on excitotoxic insults (Di Menna et al., 2018). Given the fact that activation of mGlu3 receptor may provide neuroprotection (Caraci et al., 2011), pharmacological agents activating mGlu3 receptors may reduce the risk for neurotoxicity while improving schizophrenia-related cognitive deficits.
mGlu4 Receptor Ligands Have the Potential To Treat Positive Symptoms of Schizophrenia
The availability of brain-penetrable receptor-selective ligands has facilitated the study of group III mGlu receptors in schizophrenia. Peripheral administration of pan-group III mGlu receptor agonist ACPT-I exerted antipsychotic-like effects (reducing MK801- and amphetamine-induced hyperactivity and DOI-induced head twitches) in rats (Pałucha-Poniewiera et al., 2008). Similar antipsychotic-like effects were also observed with mGlu4 receptor-selective agonists LSP1-2111 (Wierońska et al., 2012) and LSP4-2022 (Woźniak et al., 2016). In addition, LSP4-2022 improved negative symptoms and cognition in MK801-treated mice (Woźniak et al., 2016). Interestingly, mGlu4 receptors cooperate with other neurotransmitter receptors to induce antipsychotic-like effects in rodents (Woźniak et al., 2016, 2017). For example, the antipsychotic-like activity of the mGlu4 receptor agonist LSP4-2022 was reversed by the GABAB receptor antagonist CGP55845. Further, coadministration of subeffective doses of LSP4-2022 and GABAB receptor PAMs acted synergistically to produce antipsychotic-like effects (Woźniak et al., 2016). Similarly, the 5-HT1A receptor antagonist WAY100635 reversed antipsychotic-like actions of the mGlu4 receptor agonist LSP4-2022, whereas administration of a subeffective dose of the 5-HT1A receptor agonist with an ineffective dose of LSP4-2022 enhanced the effects of ineffective dosage of LSP4-2022 (Woźniak et al., 2017). The receptor interaction has also been extended to the M4 muscarinic receptor, where coadministration of subactive doses of LSP4-2022 and M4 receptor PAMs induced a robust antipsychotic-like effects (Cieślik et al., 2018b). Similar antipsychotic-like properties have also been reported with the mGlu4 receptor PAMs Lu AF21934 (Sławińska et al., 2013) and ADX88178 (Kalinichev et al., 2014). These studies further support the potential utility of selective mGlu4 receptor activators for treating positive symptoms of schizophrenia.
Potential Utility of mGlu2-mGlu4 Receptor Heterodimers As Novel Antipsychotics
Emerging studies suggest that mGlu4 and mGlu2 receptors can form heterodimers in native brain tissues (Yin et al., 2014). Excitingly, a recent study demonstrated that activation of mGlu2/4 receptor heterodimers inhibits DOI-induced increase in glutamatergic neurotransmission in the PFC ex vivo (Xiang et al., 2021). Further mechanistic studies revealed a synapse-specific role of these heterodimers in which these heterodimers presented activity at synapses to medial PFC (mPFC) originating from the thalamus (thalamo-mPFC) but not at the synapses to the mPFC originating from basolateral amygdala and ventral hippocampus (Xiang et al., 2021). This indicates that mGlu2/4 receptor heterodimers could selectively modulate specific functions associated with thalamo-mPFC synapses (Xiang et al., 2021). Interestingly, thalamic nuclei and the projections from the thalamus are widely known to play roles in the actions of NMDA receptor antagonists (Santana et al., 2011; Zhang et al., 2012). Moreover, the thalamocortical system is an important site of action of hallucinogenic drugs like DOI and lysergic acid diethylamide (LSD) (Scruggs et al., 2000; Marek et al., 2001; Preller et al., 2019; Inserra et al., 2021), and agents that modulate or depress transmission over this synapse may show antipsychotic-like effects. Therefore, the mGlu2/4 receptor heterodimer at the same synapse could be a novel therapeutic target for the treatment of positive symptoms associated with schizophrenia.
In line with the cellular effects of mGlu2/4 receptor activation, Lu AF21934, an mGlu4 receptor PAM with activity at mGlu2/4 receptor heterodimers, has been shown to inhibit MK801- and amphetamine-induced hyperactivity and reverse of DOI-induced head twitch in rodents (Sławińska et al., 2013; Wierońska et al., 2015). Considering the fact that the mGlu4 receptor homodimer-specific PAMs PHCCC and VU0418506 fail to potentiate L-AP4-mediated inhibition of thalamo-PFC transmission in slices (Xiang et al., 2021), it is conceivable that the previously reported antipsychotic-like effects of LuAF21934 are likely to be mediated by actions on mGlu2/4 receptor heterodimers. Nevertheless, it will be important to test the behavioral effects of homomer-selective mGlu2 and mGlu4 receptors PAMs relative to mGlu2/4 receptor modulators in rodent models of NMDA hypofunction to develop a better understanding of their role in treating positive symptoms of schizophrenia.
Deletion or Blockade of mGlu7 Receptor Impairs Cognition
Genetic variations in GRM7 (the gene encoding for mGlu7 receptor) are associated with schizophrenia (Niu et al., 2015; Li et al., 2016b; Chaumette et al., 2020). Specifically, the polymorphism rs1396409 was associated with performance intellectual quotient in a discovery cohort of 144 patients with first-episode psychosis and was further replicated in 121 ultra-high-risk patients (Chaumette et al., 2020). This polymorphism is also associated with the cognitive deficits during the onset of psychosis and highlights the impact of mGlu7 receptor signaling in cognitive impairments in early psychotic episodes. These studies are supported by pharmacological studies in which systemic administration of the mGlu7 receptor NAM MMPIP impaired cognitive performances in WT mice as depicted by reduction in the recognition index and location index in the object recognition and object location test, respectively (Hikichi et al., 2010). Also, MMPIP treatment increased the total time to complete the task in the eight-arm radial maze test and decreased social interaction in rats. These results are complemented by the behavioral profiling studies indicating deficits in various forms of cognition tasks in GRM7 KO mice. For example, GRM7 KO mice displayed deficits in fear response immediately after and 1 day after exposure to foot shock (Masugi et al., 1999) and in both contextual and cued fear learning (Goddyn et al., 2008; Fisher et al., 2020). Further, these mice showed impaired spatial working memory in the water maze test and eight-arm radial maze test (Hölscher et al., 2004; Callaerts-Vegh et al., 2006). These deficits in cognition were accompanied by disruption of social behavior in which deletion of the mGlu7 receptor impacted social preference but not sociability or social recognition (Fisher et al., 2020). As cognitive deficits and social withdrawal are core symptoms of schizophrenia, the role of the mGlu7 receptor in mediating cognition and social behaviors and the underlying circuitry are worth investigating.
mGlu7 Receptor NAMs Can Rescue Schizophrenia-Like Symptoms in Rodents
Interestingly, GRM7 KO mice exhibited a blunted effect of amphetamine on locomotor activity (Fisher et al., 2020) and DOI-induced head twitches as compared with the control mice (Wierońska et al., 2012). These effects were consistent with pharmacological studies showing beneficial effects of the mGlu7 receptor NAMs ADX71743 and MMPIP in rodent models of schizophrenia-relevant behavioral responses (Kalinichev et al., 2013; Cieślik et al., 2018a). Both compounds have been shown to decrease MK801-induced hyperlocomotion and DOI-induced head twitches in mice and rescue MK801-induced reduction in recognition index in novel object recognition tests (Cieślik et al., 2018a). Further, ADX71743 significantly reversed MK801-induced impairments in social behavior and acoustic startle response, whereas MMPIP had no effect on these behaviors. Besides these effects, ADX71743 caused a slight reduction in amphetamine-induced hyperlocomotion in mice (Kalinichev et al., 2013), and the activity of MMPIP in this behavior has not been tested so far. To note, MMPIP and ADX71743 did not exert their own effects on spontaneous locomotor activity in rodents (Hikichi et al., 2010; Cieślik et al., 2018a). Taken together, these studies suggest that mGlu7 receptor NAMs may provide a novel therapeutic approach for treating all symptom domains of schizophrenia. On the other hand, the mGlu7 receptor atypical agonist AMN082 (Mitsukawa et al., 2005) exacerbated MK801-induced hyperactivity and DOI-induced head twitches in mice (Wierońska et al., 2012). Also, the propsychotic-like effects of AMN082 were absent in GRM7 KO mice, consistent with the hypothesis that the mGlu7 receptor mediates psychosis-like effects of AMN082 in rodents. These studies suggest that activation/potentiation of the mGlu7 receptor at selective synapses may induce antipsychotic-like effects. Nevertheless, more research is certainly needed to fully evaluate the potential utility of the mGlu7 receptor ligands in models mimicking schizophrenia symptoms.
Deletion of GRM8 Does Not Induce Consistent Endophenotypes of Schizophrenia
A study by Gerlai and coworkers (2002) reported that GRM8 (the gene encoding for mGlu8 receptor) KO mice showed a reduced fear response to the electric shocks presented during the training. Further, in the context test, GRM8 KO mice showed a delayed fear response with reduced freezing relative to WT mice at the beginning of the contextual test. However, the freezing increased with time, and by the end of the test session they exhibited total freezing that was greater than that observed in the control mice (Gerlai et al., 2002). GRM8 KO mice also showed a reduction in amplitude to jump after electric shock, which suggests that the mechanisms mediating responses to aversive stimuli might be altered in these mice (Gerlai et al., 2002). To note, the GRM8 KO and the control mice used in this study were on the Institute of Cancer Research (ICR) background and experienced an impaired vision that can affect the performance of mice in behavioral tasks. Furthermore, other research groups found either a robust decrease (Fendt et al., 2010) or no impairments in the freezing response in contextual fear conditioning (Goddyn et al., 2015). Similarly, no consistent genotype-specific effects in novel object recognition tests were reported by different groups (Duvoisin et al., 2005; Fendt et al., 2010). Also, the studies involving GRM8 KO did not report consistent deficits in cognition (Gerlai et al., 2002; Goddyn et al., 2015), leading to inconclusive results about the role of the mGlu8 receptor in regulating cognitive function. Moreover, no deficits in PPI, spontaneous locomotor activity, and spatial learning in the Morris water maze test have been documented in these KOs (Duvoisin et al., 2005; Fendt et al., 2010; Goddyn et al., 2015), which may suggest that the mGlu8 receptor is not directly involved in behaviors that are relevant for schizophrenia. Pharmacological studies using mGlu8 receptor orthosteric agonist (S)-3,4-DCPG showed decrease amphetamine but not PCP-induced hyperactivity when tested at a higher dosage (80 mg/kg; i.p.) in mice (Ossowska et al., 2004). Also, it evoked extrapyramidal effects at a dose closer to the efficacious dose (100 mg/kg; i.p.). From these studies, along with KO mice reports, it is conceivable that the mGlu8 receptor is not a viable target for novel antipsychotics. Further, considering the role of mGlu8 receptors in regulating memory tasks and anxiety, mGlu8 receptor ligands could be beneficial in treating the anxiety phenotype and some cognitive deficits associated with schizophrenia.
Concluding Remarks
Emerging preclinical and clinical evidences suggest the involvement of glutamatergic neurotransmission in the pathophysiology of schizophrenia. Based on that, several NMDA receptor antagonists are currently being used to mimic schizophrenia-related behavioral deficits in preclinical studies. Considering the side effects associated with drugs targeting NMDA receptors, attention has shifted to finding novel drug targets with a safer therapeutic profile. Among these, the mGlu receptors have emerged as promising targets for the treatment of schizophrenia. The development of transgenic animals and receptor-selective pharmacological tools have advanced our understanding of the role of mGlu receptors in schizophrenia. For example, mGlu1 receptor PAMs can reverse receptor dysregulation induced by selective deleterious mutations expressed in a heterologous system and induce robust antipsychotic-like activity in rodents. Also, the mGlu5 receptor PAMs show efficacy for reducing behavioral deficits related to all three symptoms domains (positive, negative, and cognitive deficits) in schizophrenia. Interestingly, biased mGlu5 receptor PAMs that do not potentiate NMDA receptor signaling are equally efficacious as ago-PAMs but lack excitotoxicity and seizure-inducing effects. These exciting findings suggest a need for testing biased mGlu5 PAMs in various preclinical models to investigate the safety and efficacy profiles before efforts to optimize compounds suitable for clinical development. Further, based on robust antipsychotic-like effects, mGlu2/3 receptor agonists and mGlu2 receptor PAMs entered the clinical trials. These compounds failed to show efficacy for the treatment of schizophrenia but were efficacious in a selected group of patients. These trials point toward a need for testing mGlu2/3 receptor agonists in the patients recruited based on disease severity and genetic background.
Excitingly, a functional interaction between mGlu5 and mGlu3 receptors in the PFC and hippocampus has been reported and might be responsible for the cognition-enhancing effects of mGlu3 receptor potentiators. In the future, it will be crucial to investigate the role of mGlu3-mGlu5 receptor interaction in a broad range of cognitive assays regulated by mGlu5 receptors. Given the neuroprotective effects of mGlu3 receptor activation, this information will be helpful in guiding design of safer drugs for treating cognitive deficits associated with schizophrenia. Also, the mGlu2-mGlu4 receptor heterodimers with a distinct pharmacological profile may provide novel approaches to optimize desired therapeutic efficacy and safety profile. Knowledge about the role of other group III mGlu receptors is still in its infancy, and more receptor-selective compounds are needed to understand their pharmacology and physiology.
In conclusion, a broad number of preclinical and clinical studies illuminate the potential of targeting mGlu receptors to develop safe and efficacious drugs for the treatment of schizophrenia.
Abbreviations
- ago-PAM
positive allosteric modulator (PAM) with agonist activity
- CA1
Cornu Ammonis 1
- CNS
central nervous system
- DLPFC
dorsolateral prefrontal cortex
- DOI
2,5-Dimethoxy-4-iodoamphetamine
- 5-HT
5-hydroxy-tryptamine
- KO
knockout
- mGlu
metabotropic glutamate
- mPFC
medial prefrontal cortex
- NAAG
N-acetylaspartylglutamate
- NAM
negative allosteric modulator
- NMDA
N-methyl-D-aspartate
- PAM
positive allosteric modulator
- PANSS
Positive and Negative Syndrome Scale
- PCP
phencyclidine
- PFC
prefrontal cortex
- pharmacoBOLD
pharmacological blood-oxygen-level dependent
- PPI
prepulse inhibition
- WT
wild-type
Authorship Contributions
Wrote or contributed to the writing of the manuscript: Dogra, Conn.
Footnotes
This work was supported by National Institutes of Health National Institute of Mental Health [Grant R01-MH062646] (to P.J.C.) and National Institute of Neurological Disorders and Stroke [Grant R37-NS031373] (to P.J.C.).
P.J.C. receives research support from Acadia Pharmaceuticals and Boehringer Ingelheim. P.J.C. is an inventor on multiple patents for allosteric modulators of metabotropic glutamate receptors. S.D. has no competing interests to declare.
References
- Adams DH, Kinon BJ, Baygani S, Millen BA, Velona I, Kollack-Walker S, Walling DP (2013) A long-term, phase 2, multicenter, randomized, open-label, comparative safety study of pomaglumetad methionil (LY2140023 monohydrate) versus atypical antipsychotic standard of care in patients with schizophrenia. BMC Psychiatry 13:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguilar DD, Strecker RE, Basheer R, McNally JM (2020) Alterations in sleep, sleep spindle, and EEG power in mGluR5 knockout mice. J Neurophysiol 123:22–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alagarsamy S, Rouse ST, Gereau RW 4th, Heinemann SF, Smith Y, Conn PJ (1999) Activation of N-methyl-D-aspartate receptors reverses desensitization of metabotropic glutamate receptor, mGluR5, in native and recombinant systems. Ann N Y Acad Sci 868:526–530. [DOI] [PubMed] [Google Scholar]
- Ayala JEChen YBanko JLSheffler DJWilliams RTelk ANWatson NLXiang ZZhang YJones PJ, et al. (2009) mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology 34:2057–2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayoub MA, Angelicheva D, Vile D, Chandler D, Morar B, Cavanaugh JA, Visscher PM, Jablensky A, Pfleger KD, Kalaydjieva L (2012) Deleterious GRM1 mutations in schizophrenia. PLoS One 7:e32849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balu DTLi YTakagi SPresti KTRamikie TSRook JMJones CKLindsley CWConn PJBolshakov VY, et al. (2016) An mGlu5-positive allosteric modulator rescues the neuroplasticity deficits in a genetic model of NMDA receptor hypofunction in schizophrenia. Neuropsychopharmacology 41:2052–2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes SA, Der-Avakian A, Young JW (2017) Preclinical models to investigate mechanisms of negative symptoms in schizophrenia. Schizophr Bull 43:706–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartolomé-Nebreda JMConde-Ceide SDelgado FIturrino LPastor JPena MATrabanco AATresadern GWassvik CMStauffer SR, et al. (2013) Dihydrothiazolopyridone derivatives as a novel family of positive allosteric modulators of the metabotropic glutamate 5 (mGlu5) receptor. J Med Chem 56:7243–7259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Becker I, Wang-Eckhardt L, Lodder-Gadaczek J, Wang Y, Grünewald A, Eckhardt M (2021) Mice deficient in the NAAG synthetase II gene Rimkla are impaired in a novel object recognition task. J Neurochem 157:2008–2023. [DOI] [PubMed] [Google Scholar]
- Belforte JE, Zsiros V, Sklar ER, Jiang Z, Yu G, Li Y, Quinlan EM, Nakazawa K (2010) Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes. Nat Neurosci 13:76–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, Sanders-Bush E (2007) A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol Pharmacol 72:477–484. [DOI] [PubMed] [Google Scholar]
- Benquet P, Gee CE, Gerber U (2002) Two distinct signaling pathways upregulate NMDA receptor responses via two distinct metabotropic glutamate receptor subtypes. J Neurosci 22:9679–9686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bishop JR, Ellingrod VL, Moline J, Miller D (2005) Association between the polymorphic GRM3 gene and negative symptom improvement during olanzapine treatment. Schizophr Res 77:253–260. [DOI] [PubMed] [Google Scholar]
- Bishop JR, Miller DD, Ellingrod VL, Holman T (2011) Association between type-three metabotropic glutamate receptor gene (GRM3) variants and symptom presentation in treatment refractory schizophrenia. Hum Psychopharmacol 26:28–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bishop JR, Reilly JL, Harris MS, Patel SR, Kittles R, Badner JA, Prasad KM, Nimgaonkar VL, Keshavan MS, Sweeney JA (2015) Pharmacogenetic associations of the type-3 metabotropic glutamate receptor (GRM3) gene with working memory and clinical symptom response to antipsychotics in first-episode schizophrenia. Psychopharmacology (Berl) 232:145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brigman JL, Feyder M, Saksida LM, Bussey TJ, Mishina M, Holmes A (2008) Impaired discrimination learning in mice lacking the NMDA receptor NR2A subunit. Learn Mem 15:50–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brody SA, Conquet F, Geyer MA (2003) Disruption of prepulse inhibition in mice lacking mGluR1. Eur J Neurosci 18:3361–3366. [DOI] [PubMed] [Google Scholar]
- Brody SA, Conquet F, Geyer MA (2004a) Effect of antipsychotic treatment on the prepulse inhibition deficit of mGluR5 knockout mice. Psychopharmacology (Berl) 172:187–195. [DOI] [PubMed] [Google Scholar]
- Brody SA, Dulawa SC, Conquet F, Geyer MA (2004b) Assessment of a prepulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Mol Psychiatry 9:35–41. [DOI] [PubMed] [Google Scholar]
- Brody SA, Geyer MA (2004) Interactions of the mGluR5 gene with breeding and maternal factors on startle and prepulse inhibition in mice. Neurotox Res 6:79–90. [DOI] [PubMed] [Google Scholar]
- Burrows EL, McOmish CE, Buret LS, Van den Buuse M, Hannan AJ (2015) Environmental enrichment ameliorates behavioral impairments modeling schizophrenia in mice lacking metabotropic glutamate receptor 5. Neuropsychopharmacology 40:1947–1956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callaerts-Vegh Z, Beckers T, Ball SM, Baeyens F, Callaerts PF, Cryan JF, Molnar E, D’Hooge R (2006) Concomitant deficits in working memory and fear extinction are functionally dissociated from reduced anxiety in metabotropic glutamate receptor 7-deficient mice. J Neurosci 26:6573–6582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell UC, Lalwani K, Hernandez L, Kinney GG, Conn PJ, Bristow LJ (2004) The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) potentiates PCP-induced cognitive deficits in rats. Psychopharmacology (Berl) 175:310–318. [DOI] [PubMed] [Google Scholar]
- Caraci FMolinaro GBattaglia GGiuffrida MLRiozzi BTraficante ABruno VCannella MMerlo SWang X, et al. (2011) Targeting group II metabotropic glutamate (mGlu) receptors for the treatment of psychosis associated with Alzheimer’s disease: selective activation of mGlu2 receptors amplifies beta-amyloid toxicity in cultured neurons, whereas dual activation of mGlu2 and mGlu3 receptors is neuroprotective. Mol Pharmacol 79:618–626. [DOI] [PubMed] [Google Scholar]
- Chang M, Sun L, Liu X, Sun W, Ji M, Wang Z, Wang Y, You X (2015) Evaluation of relationship between GRM3 polymorphisms and cognitive function in schizophrenia of Han Chinese. Psychiatry Res 229:1043–1046. [DOI] [PubMed] [Google Scholar]
- Chaumette BSengupta SMLepage MMalla AIyer SNKebir ODion PARouleau GAKrebs M-OShah JL, et al. ; ICAAR Study Group (2020) A polymorphism in the glutamate metabotropic receptor 7 is associated with cognitive deficits in the early phases of psychosis. Schizophr Res. DOI: 10.1016/j.schres.2020.06.019. [online ahead of print]. [DOI] [PubMed] [Google Scholar]
- Cho HPGarcia-Barrantes PMBrogan JTHopkins CRNiswender CMRodriguez ALVenable DFMorrison RDBubser MDaniels JS, et al. (2014) Chemical modulation of mutant mGlu1 receptors derived from deleterious GRM1 mutations found in schizophrenics. ACS Chem Biol 9:2334–2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cieślik PWoźniak MKaczorowska KBrański PBurnat GChocyk ABobula BGruca PLitwa EPałucha-Poniewiera A, et al. (2018a) Negative allosteric modulators of mGlu7 receptor as putative antipsychotic drugs. Front Mol Neurosci 11:316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cieślik P, Woźniak M, Rook JM, Tantawy MN, Conn PJ, Acher F, Tokarski K, Kusek M, Pilc A, Wierońska JM (2018b) Mutual activation of glutamatergic mGlu4 and muscarinic M4 receptors reverses schizophrenia-related changes in rodents. Psychopharmacology (Berl) 235:2897–2913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collett VJ, Collingridge GL (2004) Interactions between NMDA receptors and mGlu5 receptors expressed in HEK293 cells. Br J Pharmacol 142:991–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coyle JT (2006) Glutamate and schizophrenia: beyond the dopamine hypothesis. Cell Mol Neurobiol 26:365–384. [DOI] [PubMed] [Google Scholar]
- De Filippis B, Lyon L, Taylor A, Lane T, Burnet PW, Harrison PJ, Bannerman DM (2015) The role of group II metabotropic glutamate receptors in cognition and anxiety: comparative studies in GRM2(-/-), GRM3(-/-) and GRM2/3(-/-) knockout mice. Neuropharmacology 89:19–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devon RS, Anderson S, Teague PW, Muir WJ, Murray V, Pelosi AJ, Blackwood DH, Porteous DJ (2001) The genomic organisation of the metabotropic glutamate receptor subtype 5 gene, and its association with schizophrenia. Mol Psychiatry 6:311–314. [DOI] [PubMed] [Google Scholar]
- Di Menna LJoffe MEIacovelli LOrlando RLindsley CWMairesse JGressèns PCannella MCaraci FCopani A, et al. (2018) Functional partnership between mGlu3 and mGlu5 metabotropic glutamate receptors in the central nervous system. Neuropharmacology 128:301–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dogra S, Conn PJ (2021) Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 196:108687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dogra S, Stansley BJ, Xiang Z, Qian W, Gogliotti RG, Nicoletti F, Lindsley CW, Niswender CM, Joffe ME, Conn PJ (2021) Activating mGlu3 metabotropic glutamate receptors rescues schizophrenia-like cognitive deficits through metaplastic adaptations within the hippocampus. Biol Psychiatry 90:385–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duncan GE, Moy SS, Perez A, Eddy DM, Zinzow WM, Lieberman JA, Snouwaert JN, Koller BH (2004) Deficits in sensorimotor gating and tests of social behavior in a genetic model of reduced NMDA receptor function. Behav Brain Res 153:507–519. [DOI] [PubMed] [Google Scholar]
- Duvoisin RM, Zhang C, Pfankuch TF, O’Connor H, Gayet-Primo J, Quraishi S, Raber J (2005) Increased measures of anxiety and weight gain in mice lacking the group III metabotropic glutamate receptor mGluR8. Eur J Neurosci 22:425–436. [DOI] [PubMed] [Google Scholar]
- Egan MFStraub REGoldberg TEYakub ICallicott JHHariri ARMattay VSBertolino AHyde TMShannon-Weickert C, et al. (2004) Variation in GRM3 affects cognition, prefrontal glutamate, and risk for schizophrenia. Proc Natl Acad Sci USA 101:12604–12609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fendt M, Bürki H, Imobersteg S, van der Putten H, McAllister K, Leslie JC, Shaw D, Hölscher C (2010) The effect of mGlu8 deficiency in animal models of psychiatric diseases. Genes Brain Behav 9:33–44. [DOI] [PubMed] [Google Scholar]
- Fijal BA, Kinon BJ, Kapur S, Stauffer VL, Conley RR, Jamal HH, Kane JM, Witte MM, Houston JP (2009) Candidate-gene association analysis of response to risperidone in African-American and white patients with schizophrenia. Pharmacogenomics J 9:311–318. [DOI] [PubMed] [Google Scholar]
- Fisher NMGould RWGogliotti RGMcDonald AJBadivuku HChennareddy SBuch ABMoore AMJenkins MTRobb WH, et al. (2020) Phenotypic profiling of mGlu7 knockout mice reveals new implications for neurodevelopmental disorders. Genes Brain Behav 19:e12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frank RAMcRae AFPocklington AJvan de Lagemaat LNNavarro PCroning MDKomiyama NHBradley SJChalliss RAArmstrong JD, et al. (2011) Clustered coding variants in the glutamate receptor complexes of individuals with schizophrenia and bipolar disorder. PLoS One 6:e19011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fujii Y, Shibata H, Kikuta R, Makino C, Tani A, Hirata N, Shibata A, Ninomiya H, Tashiro N, Fukumaki Y (2003) Positive associations of polymorphisms in the metabotropic glutamate receptor type 3 gene (GRM3) with schizophrenia. Psychiatr Genet 13:71–76. [DOI] [PubMed] [Google Scholar]
- Galici R, Echemendia NG, Rodriguez AL, Conn PJ (2005) A selective allosteric potentiator of metabotropic glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in mouse models predictive of antipsychotic activity. J Pharmacol Exp Ther 315:1181–1187. [DOI] [PubMed] [Google Scholar]
- Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, de Paulis T, Conn PJ (2006) Biphenyl-indanone A, a positive allosteric modulator of the metabotropic glutamate receptor subtype 2, has antipsychotic- and anxiolytic-like effects in mice. J Pharmacol Exp Ther 318:173–185. [DOI] [PubMed] [Google Scholar]
- Garcia-Barrantes PM, Cho HP, Niswender CM, Byers FW, Locuson CW, Blobaum AL, Xiang Z, Rook JM, Conn PJ, Lindsley CW (2015) Development of novel, CNS penetrant positive allosteric modulators for the metabotropic glutamate receptor subtype 1 (mGlu1), based on an N-(3-chloro-4-(1,3-dioxoisoindolin-2-yl)phenyl)-3-methylfuran-2-carboxamide scaffold, that potentiate wild type and mutant mGlu1 receptors found in schizophrenics. J Med Chem 58:7959–7971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gastambide F, Cotel MC, Gilmour G, O’Neill MJ, Robbins TW, Tricklebank MD (2012) Selective remediation of reversal learning deficits in the neurodevelopmental MAM model of schizophrenia by a novel mGlu5 positive allosteric modulator. Neuropsychopharmacology 37:1057–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerlai R, Adams B, Fitch T, Chaney S, Baez M (2002) Performance deficits of mGluR8 knockout mice in learning tasks: the effects of null mutation and the background genotype. Neuropharmacology 43:235–249. [DOI] [PubMed] [Google Scholar]
- Gewirtz JC, Marek GJ (2000) Behavioral evidence for interactions between a hallucinogenic drug and group II metabotropic glutamate receptors. Neuropsychopharmacology 23:569–576. [DOI] [PubMed] [Google Scholar]
- Goddyn H, Callaerts-Vegh Z, D’Hooge R (2015) Functional dissociation of group III metabotropic glutamate receptors revealed by direct comparison between the behavioral profiles of knockout mouse lines. Int J Neuropsychopharmacol 18:pyv053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goddyn H, Callaerts-Vegh Z, Stroobants S, Dirikx T, Vansteenwegen D, Hermans D, van der Putten H, D’Hooge R (2008) Deficits in acquisition and extinction of conditioned responses in mGluR7 knockout mice. Neurobiol Learn Mem 90:103–111. [DOI] [PubMed] [Google Scholar]
- Goff DC, Coyle JT (2001) The emerging role of glutamate in the pathophysiology and treatment of schizophrenia. Am J Psychiatry 158:1367–1377. [DOI] [PubMed] [Google Scholar]
- González-Maeso JAng RLYuen TChan PWeisstaub NVLópez-Giménez JFZhou MOkawa YCallado LFMilligan G, et al. (2008) Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature 452:93–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Govek SPBonnefous CHutchinson JHKamenecka TMcQuiston JPracitto RZhao LXGardner MFJames JKDaggett LP, et al. (2005) Benzazoles as allosteric potentiators of metabotropic glutamate receptor 2 (mGluR2): efficacy in an animal model for schizophrenia. Bioorg Med Chem Lett 15:4068–4072. [DOI] [PubMed] [Google Scholar]
- Gregory KJHerman EJRamsey AJHammond ASByun NEStauffer SRManka JTJadhav SBridges TMWeaver CD, et al. (2013) N-aryl piperazine metabotropic glutamate receptor 5 positive allosteric modulators possess efficacy in preclinical models of NMDA hypofunction and cognitive enhancement. J Pharmacol Exp Ther 347:438–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griebel GPichat PBoulay DNaimoli VPotestio LFeatherstone RSahni SDefex HDesvignes CSlowinski F, et al. (2016) The mGluR2 positive allosteric modulator, SAR218645, improves memory and attention deficits in translational models of cognitive symptoms associated with schizophrenia. Sci Rep 6:35320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henry SA, Lehmann-Masten V, Gasparini F, Geyer MA, Markou A (2002) The mGluR5 antagonist MPEP, but not the mGluR2/3 agonist LY314582, augments PCP effects on prepulse inhibition and locomotor activity. Neuropharmacology 43:1199–1209. [DOI] [PubMed] [Google Scholar]
- Hikichi HHiyoshi TMarumo TTomishima YKaku AIida IUrabe HTamita TYasuhara AKarasawa J, et al. (2015) Antipsychotic profiles of TASP0443294, a novel and orally active positive allosteric modulator of metabotropic glutamate 2 receptor. J Pharmacol Sci 127:352–361. [DOI] [PubMed] [Google Scholar]
- Hikichi H, Kaku A, Karasawa J, Chaki S (2013) Stimulation of metabotropic glutamate (mGlu) 2 receptor and blockade of mGlu1 receptor improve social memory impairment elicited by MK-801 in rats. J Pharmacol Sci 122:10–16. [DOI] [PubMed] [Google Scholar]
- Hikichi HMurai TOkuda SMaehara SSatow AIse SNishino MSuzuki GTakehana HHata M, et al. (2010) Effects of a novel metabotropic glutamate receptor 7 negative allosteric modulator, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one (MMPIP), on the central nervous system in rodents. Eur J Pharmacol 639:106–114. [DOI] [PubMed] [Google Scholar]
- Hiyoshi T, Marumo T, Hikichi H, Tomishima Y, Urabe H, Tamita T, Iida I, Yasuhara A, Karasawa J, Chaki S (2014) Neurophysiologic and antipsychotic profiles of TASP0433864, a novel positive allosteric modulator of metabotropic glutamate 2 receptor. J Pharmacol Exp Ther 351:642–653. [DOI] [PubMed] [Google Scholar]
- Hölscher C, Schmid S, Pilz PK, Sansig G, van der Putten H, Plappert CF (2004) Lack of the metabotropic glutamate receptor subtype 7 selectively impairs short-term working memory but not long-term memory. Behav Brain Res 154:473–481. [DOI] [PubMed] [Google Scholar]
- Hu W, MacDonald ML, Elswick DE, Sweet RA (2015) The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann N Y Acad Sci 1338:38–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang HDegnan APBalakrishnan AEaston AGulianello MHuang YMatchett MMattson GMiller RSantone KS, et al. (2016) Oxazolidinone-based allosteric modulators of mGluR5: defining molecular switches to create a pharmacological tool box. Bioorg Med Chem Lett 26:4165–4169. [DOI] [PubMed] [Google Scholar]
- Inserra A, De Gregorio D, Rezai T, Lopez-Canul MG, Comai S, Gobbi G (2021) Lysergic acid diethylamide differentially modulates the reticular thalamus, mediodorsal thalamus, and infralimbic prefrontal cortex: an in vivo electrophysiology study in male mice. J Psychopharmacol 35:469–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ito I, Futai K, Katagiri H, Watanabe M, Sakimura K, Mishina M, Sugiyama H (1997) Synapse-selective impairment of NMDA receptor functions in mice lacking NMDA receptor epsilon 1 or epsilon 2 subunit. J Physiol 500:401–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jew CP, Wu CS, Sun H, Zhu J, Huang JY, Yu D, Justice NJ, Lu HC (2013) mGluR5 ablation in cortical glutamatergic neurons increases novelty-induced locomotion. PLoS One 8:e70415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia Z, Lu Y, Henderson J, Taverna F, Romano C, Abramow-Newerly W, Wojtowicz JM, Roder J (1998) Selective abolition of the NMDA component of long-term potentiation in mice lacking mGluR5. Learn Mem 5:331–343. [PMC free article] [PubMed] [Google Scholar]
- Jin LE, Wang M, Galvin VC, Lightbourne TC, Conn PJ, Arnsten AFT, Paspalas CD (2018) mGluR2 versus mGluR3 metabotropic glutamate receptors in primate dorsolateral prefrontal cortex: postsynaptic mGluR3 strengthen working memory networks. Cereb Cortex 28:974–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffe ME, Santiago CI, Engers JL, Lindsley CW, Conn PJ (2019) Metabotropic glutamate receptor subtype 3 gates acute stress-induced dysregulation of amygdalo-cortical function. Mol Psychiatry 24:916–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson MP, Baez M, Jagdmann GE Jr, Britton TC, Large TH, Callagaro DO, Tizzano JP, Monn JA, Schoepp DD (2003) Discovery of allosteric potentiators for the metabotropic glutamate 2 receptor: synthesis and subtype selectivity of N-(4-(2-methoxyphenoxy)phenyl)-N-(2,2,2-trifluoroethylsulfonyl)pyrid-3-ylmethylamine. J Med Chem 46:3189–3192. [DOI] [PubMed] [Google Scholar]
- Johnson MP, Barda D, Britton TC, Emkey R, Hornback WJ, Jagdmann GE, McKinzie DL, Nisenbaum ES, Tizzano JP, Schoepp DD (2005) Metabotropic glutamate 2 receptor potentiators: receptor modulation, frequency-dependent synaptic activity, and efficacy in preclinical anxiety and psychosis model(s). Psychopharmacology (Berl) 179:271–283. [DOI] [PubMed] [Google Scholar]
- Kalinichev MLe Poul EBoléa CGirard FCampo BFonsi MRoyer-Urios IBrowne SEUslaner JMDavis MJ, et al. (2014) Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. J Pharmacol Exp Ther 350:495–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalinichev MRouillier MGirard FRoyer-Urios IBournique BFinn TCharvin DCampo BLe Poul EMutel V, et al. (2013) ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. J Pharmacol Exp Ther 344:624–636. [DOI] [PubMed] [Google Scholar]
- Kannangara TS, Eadie BD, Bostrom CA, Morch K, Brocardo PS, Christie BR (2015) GluN2A-/- mice lack bidirectional synaptic plasticity in the dentate gyrus and perform poorly on spatial pattern separation tasks. Cereb Cortex 25:2102–2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantrowitz JTGrinband JGoff DCLahti ACMarder SRKegeles LSGirgis RRSobeih TWall MMChoo TH, et al. (2020) Proof of mechanism and target engagement of glutamatergic drugs for the treatment of schizophrenia: RCTs of pomaglumetad and TS-134 on ketamine-induced psychotic symptoms and pharmacoBOLD in healthy volunteers. Neuropsychopharmacology 45:1842–1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinon BJ, Gómez JC (2013) Clinical development of pomaglumetad methionil: a non-dopaminergic treatment for schizophrenia. Neuropharmacology 66:82–86. [DOI] [PubMed] [Google Scholar]
- Kinon BJ, Millen BA, Zhang L, McKinzie DL (2015) Exploratory analysis for a targeted patient population responsive to the metabotropic glutamate 2/3 receptor agonist pomaglumetad methionil in schizophrenia. Biol Psychiatry 78:754–762. [DOI] [PubMed] [Google Scholar]
- Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K, Kryzhanovskaya L, Jarkova N; HBBI Study Group (2011) A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin Psychopharmacol 31:349–355. [DOI] [PubMed] [Google Scholar]
- Koros E, Rosenbrock H, Birk G, Weiss C, Sams-Dodd F (2007) The selective mGlu5 receptor antagonist MTEP, similar to NMDA receptor antagonists, induces social isolation in rats. Neuropsychopharmacology 32:562–576. [DOI] [PubMed] [Google Scholar]
- Krystal JHAbi-Saab WPerry ED’Souza DCLiu NGueorguieva RMcDougall LHunsberger TBelger ALevine L, et al. (2005) Preliminary evidence of attenuation of the disruptive effects of the NMDA glutamate receptor antagonist, ketamine, on working memory by pretreatment with the group II metabotropic glutamate receptor agonist, LY354740, in healthy human subjects. Psychopharmacology (Berl) 179:303–309. [DOI] [PubMed] [Google Scholar]
- LaCrosse AL, Burrows BT, Angulo RM, Conrad PR, Himes SM, Mathews N, Wegner SA, Taylor SB, Olive MF (2015) mGluR5 positive allosteric modulation and its effects on MK-801 induced set-shifting impairments in a rat operant delayed matching/non-matching-to-sample task. Psychopharmacology (Berl) 232:251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lainiola M, Procaccini C, Linden AM (2014) mGluR3 knockout mice show a working memory defect and an enhanced response to MK-801 in the T- and Y-maze cognitive tests. Behav Brain Res 266:94–103. [DOI] [PubMed] [Google Scholar]
- Lavreysen H, Langlois X, Donck LV, Nuñez JM, Pype S, Lütjens R, Megens A (2015) Preclinical evaluation of the antipsychotic potential of the mGlu2-positive allosteric modulator JNJ-40411813. Pharmacol Res Perspect 3:e00097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee G, Zhou Y (2019) NMDAR hypofunction animal models of schizophrenia. Front Mol Neurosci 12:185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li P, Snyder GL, Vanover KE (2016a) Dopamine targeting drugs for the treatment of schizophrenia: past, present and future. Curr Top Med Chem 16:3385–3403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li WJu KLi ZHe KChen JWang QYang BAn LFeng GSun W, et al. (2016b) Significant association of GRM7 and GRM8 genes with schizophrenia and major depressive disorder in the Han Chinese population. Eur Neuropsychopharmacol 26:136–146. [DOI] [PubMed] [Google Scholar]
- Litman RE, Smith MA, Doherty JJ, Cross A, Raines S, Gertsik L, Zukin SR (2016) AZD8529, a positive allosteric modulator at the mGluR2 receptor, does not improve symptoms in schizophrenia: a proof of principle study. Schizophr Res 172:152–157. [DOI] [PubMed] [Google Scholar]
- Liu FGrauer SKelley CNavarra RGraf RZhang GAtkinson PJPopiolek MWantuch CKhawaja X, et al. (2008) ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. J Pharmacol Exp Ther 327:827–839. [DOI] [PubMed] [Google Scholar]
- Maksymetz J, Byun NE, Luessen DJ, Li B, Barry RL, Gore JC, Niswender CM, Lindsley CW, Joffe ME, Conn PJ (2021) mGlu1 potentiation enhances prelimbic somatostatin interneuron activity to rescue schizophrenia-like physiological and cognitive deficits. Cell Rep 37:109950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mannaioni G, Marino MJ, Valenti O, Traynelis SF, Conn PJ (2001) Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. J Neurosci 21:5925–5934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marek GJ, Wright RA, Gewirtz JC, Schoepp DD (2001) A major role for thalamocortical afferents in serotonergic hallucinogen receptor function in the rat neocortex. Neuroscience 105:379–392. [DOI] [PubMed] [Google Scholar]
- Marek GJ, Wright RA, Schoepp DD, Monn JA, Aghajanian GK (2000) Physiological antagonism between 5-hydroxytryptamine(2A) and group II metabotropic glutamate receptors in prefrontal cortex. J Pharmacol Exp Ther 292:76–87. [PubMed] [Google Scholar]
- Masugi M, Yokoi M, Shigemoto R, Muguruma K, Watanabe Y, Sansig G, van der Putten H, Nakanishi S (1999) Metabotropic glutamate receptor subtype 7 ablation causes deficit in fear response and conditioned taste aversion. J Neurosci 19:955–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matosin N, Newell KA, Quidé Y, Andrews JL, Teroganova N, Green MJ, Fernandez F (2018) Effects of common GRM5 genetic variants on cognition, hippocampal volume and mGluR5 protein levels in schizophrenia. Brain Imaging Behav 12:509–517. [DOI] [PubMed] [Google Scholar]
- Mitsukawa KYamamoto ROfner SNozulak JPescott OLukic SStoehr NMombereau CKuhn RMcAllister KH, et al. (2005) A selective metabotropic glutamate receptor 7 agonist: activation of receptor signaling via an allosteric site modulates stress parameters in vivo. Proc Natl Acad Sci USA 102:18712–18717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moghaddam B, Adams BW (1998) Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science 281:1349–1352. [DOI] [PubMed] [Google Scholar]
- Moghaddam B, Javitt D (2012) From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 37:4–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohn AR, Gainetdinov RR, Caron MG, Koller BH (1999) Mice with reduced NMDA receptor expression display behaviors related to schizophrenia. Cell 98:427–436. [DOI] [PubMed] [Google Scholar]
- Neale JH, Olszewski R (2019) A role for N-acetylaspartylglutamate (NAAG) and mGluR3 in cognition. Neurobiol Learn Mem 158:9–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niswender CM, Conn PJ (2010) Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol 50:295–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niu WHuang XYu TChen SLi XWu XCao YZhang RBi YYang F, et al. (2015) Association study of GRM7 polymorphisms and schizophrenia in the Chinese Han population. Neurosci Lett 604:109–112. [DOI] [PubMed] [Google Scholar]
- Noetzel MJRook JMVinson PNCho HPDays EZhou YRodriguez ALLavreysen HStauffer SRNiswender CM, et al. (2012) Functional impact of allosteric agonist activity of selective positive allosteric modulators of metabotropic glutamate receptor subtype 5 in regulating central nervous system function. Mol Pharmacol 81:120–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Riordan KJ, Hu NW, Rowan MJ (2018) Physiological activation of mGlu5 receptors supports the ion channel function of NMDA receptors in hippocampal LTD induction in vivo. Sci Rep 8:4391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olszewski RTBukhari NZhou JKozikowski APWroblewski JTShamimi-Noori SWroblewska BBzdega TVicini SBarton FB, et al. (2004) NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR. J Neurochem 89:876–885. [DOI] [PubMed] [Google Scholar]
- Olszewski RT, Bzdega T, Neale JH (2012a) mGluR3 and not mGluR2 receptors mediate the efficacy of NAAG peptidase inhibitor in validated model of schizophrenia. Schizophr Res 136:160–161. [DOI] [PubMed] [Google Scholar]
- Olszewski RTJanczura KJBall SRMadore JCLavin KMLee JCLee MJDer EKHark TJFarago PR, et al. (2012b) NAAG peptidase inhibitors block cognitive deficit induced by MK-801 and motor activation induced by d-amphetamine in animal models of schizophrenia. Transl Psychiatry 2:e145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ossowska K, Pietraszek M, Wardas J, Wolfarth S (2004) Potential antipsychotic and extrapyramidal effects of (R,S)-3,4-dicarboxyphenylglycine [(R,S)-3,4-DCPG], a mixed AMPA antagonist/mGluR8 agonist. Pol J Pharmacol 56:295–304. [PubMed] [Google Scholar]
- Pałucha-Poniewiera A, Kłodzińska A, Stachowicz K, Tokarski K, Hess G, Schann S, Frauli M, Neuville P, Pilc A (2008) Peripheral administration of group III mGlu receptor agonist ACPT-I exerts potential antipsychotic effects in rodents. Neuropharmacology 55:517–524. [DOI] [PubMed] [Google Scholar]
- Parmentier-Batteur SHutson PHMenzel KUslaner JMMattson BAO’Brien JAMagliaro BCForest TStump CATynebor RM, et al. (2014) Mechanism based neurotoxicity of mGlu5 positive allosteric modulators--development challenges for a promising novel antipsychotic target. Neuropharmacology 82:161–173. [DOI] [PubMed] [Google Scholar]
- Patil STZhang LMartenyi FLowe SLJackson KAAndreev BVAvedisova ASBardenstein LMGurovich IYMorozova MA, et al. (2007) Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med 13:1102–1107. [DOI] [PubMed] [Google Scholar]
- Perälä JSuvisaari JSaarni SIKuoppasalmi KIsometsä EPirkola SPartonen TTuulio-Henriksson AHintikka JKieseppä T, et al. (2007) Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch Gen Psychiatry 64:19–28. [DOI] [PubMed] [Google Scholar]
- Pers TH, Timshel P, Ripke S, Lent S, Sullivan PF, O’Donovan MC, Franke L, Hirschhorn JN; Schizophrenia Working Group of the Psychiatric Genomics Consortium (2016) Comprehensive analysis of schizophrenia-associated loci highlights ion channel pathways and biologically plausible candidate causal genes. Hum Mol Genet 25:1247–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pocklington AJRees EWalters JTHan JKavanagh DHChambert KDHolmans PMoran JLMcCarroll SAKirov G, et al. (2015) Novel findings from CNVs implicate inhibitory and excitatory signaling complexes in schizophrenia. Neuron 86:1203–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preller KH, Razi A, Zeidman P, Stämpfli P, Friston KJ, Vollenweider FX (2019) Effective connectivity changes in LSD-induced altered states of consciousness in humans. Proc Natl Acad Sci USA 116:2743–2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Régio Brambilla CVeselinović TRajkumar RMauler JOrth LRuch ARamkiran SHeekeren KKawohl WWyss C, et al. (2020) mGluR5 receptor availability is associated with lower levels of negative symptoms and better cognition in male patients with chronic schizophrenia. Hum Brain Mapp 41:2762–2781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez ALGrier MDJones CKHerman EJKane ASSmith RLWilliams RZhou YMarlo JEDays EL, et al. (2010) Discovery of novel allosteric modulators of metabotropic glutamate receptor subtype 5 reveals chemical and functional diversity and in vivo activity in rat behavioral models of anxiolytic and antipsychotic activity. Mol Pharmacol 78:1105–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook JMNoetzel MJPouliot WABridges TMVinson PNCho HPZhou YGogliotti RDManka JTGregory KJ, et al. (2013) Unique signaling profiles of positive allosteric modulators of metabotropic glutamate receptor subtype 5 determine differences in in vivo activity. Biol Psychiatry 73:501–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook JMXiang ZLv XGhoshal ADickerson JWBridges TMJohnson KAFoster DJGregory KJVinson PN, et al. (2015) Biased mGlu5-positive allosteric modulators provide in vivo efficacy without potentiating mGlu5 modulation of NMDAR currents. Neuron 86:1029–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rorick-Kehn LMJohnson BGKnitowski KMSalhoff CRWitkin JMPerry KWGriffey KITizzano JPMonn JAMcKinzie DL, et al. (2007) In vivo pharmacological characterization of the structurally novel, potent, selective mGlu2/3 receptor agonist LY404039 in animal models of psychiatric disorders. Psychopharmacology (Berl) 193:121–136. [DOI] [PubMed] [Google Scholar]
- Saini SM, Mancuso SG, Mostaid MS, Liu C, Pantelis C, Everall IP, Bousman CA (2017) Meta-analysis supports GWAS-implicated link between GRM3 and schizophrenia risk. Transl Psychiatry 7:e1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salih H, Anghelescu I, Kezic I, Sinha V, Hoeben E, Van Nueten L, De Smedt H, De Boer P (2015) Pharmacokinetic and pharmacodynamic characterisation of JNJ-40411813, a positive allosteric modulator of mGluR2, in two randomised, double-blind phase-I studies. J Psychopharmacol 29:414–425. [DOI] [PubMed] [Google Scholar]
- Santana N, Troyano-Rodriguez E, Mengod G, Celada P, Artigas F (2011) Activation of thalamocortical networks by the N-methyl-D-aspartate receptor antagonist phencyclidine: reversal by clozapine. Biol Psychiatry 69:918–927. [DOI] [PubMed] [Google Scholar]
- Satow AMaehara SIse SHikichi HFukushima MSuzuki GKimura TTanak TIto SKawamoto H, et al. (2008) Pharmacological effects of the metabotropic glutamate receptor 1 antagonist compared with those of the metabotropic glutamate receptor 5 antagonist and metabotropic glutamate receptor 2/3 agonist in rodents: detailed investigations with a selective allosteric metabotropic glutamate receptor 1 antagonist, FTIDC [4-[1-(2-fluoropyridine-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide]. J Pharmacol Exp Ther 326:577–586. [DOI] [PubMed] [Google Scholar]
- Satow ASuzuki GMaehara SHikichi HMurai TMurai TKawagoe-Takaki HHata MIto SOzaki S, et al. (2009) Unique antipsychotic activities of the selective metabotropic glutamate receptor 1 allosteric antagonist 2-cyclopropyl-5-[1-(2-fluoro-3-pyridinyl)-5-methyl-1H-1,2,3-triazol-4-yl]-2,3-dihydro-1H-isoindol-1-one. J Pharmacol Exp Ther 330:179–190. [DOI] [PubMed] [Google Scholar]
- Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511:421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schlumberger C, Pietraszek M, Gravius A, Danysz W (2010) Effects of a positive allosteric modulator of mGluR5 ADX47273 on conditioned avoidance response and PCP-induced hyperlocomotion in the rat as models for schizophrenia. Pharmacol Biochem Behav 95:23–30. [DOI] [PubMed] [Google Scholar]
- Scruggs JL, Patel S, Bubser M, Deutch AY (2000) DOI-Induced activation of the cortex: dependence on 5-HT2A heteroceptors on thalamocortical glutamatergic neurons. J Neurosci 20:8846–8852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sławińska AWierońska JMStachowicz KMarciniak MLasoń-Tyburkiewicz MGruca PPapp MKusek MTokarski KDoller D, et al. (2013) The antipsychotic-like effects of positive allosteric modulators of metabotropic glutamate mGlu4 receptors in rodents. Br J Pharmacol 169:1824–1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spooren WP, Gasparini F, van der Putten H, Koller M, Nakanishi S, Kuhn R (2000) Lack of effect of LY314582 (a group 2 metabotropic glutamate receptor agonist) on phencyclidine-induced locomotor activity in metabotropic glutamate receptor 2 knockout mice. Eur J Pharmacol 397:R1–R2. [DOI] [PubMed] [Google Scholar]
- Sprengel RSuchanek BAmico CBrusa RBurnashev NRozov AHvalby OJensen VPaulsen OAndersen P, et al. (1998) Importance of the intracellular domain of NR2 subunits for NMDA receptor function in vivo. Cell 92:279–289. [DOI] [PubMed] [Google Scholar]
- Stahl SM (2018) Beyond the dopamine hypothesis of schizophrenia to three neural networks of psychosis: dopamine, serotonin, and glutamate. CNS Spectr 23:187–191. [DOI] [PubMed] [Google Scholar]
- Stauffer VL, Millen BA, Andersen S, Kinon BJ, Lagrandeur L, Lindenmayer JP, Gomez JC (2013) Pomaglumetad methionil: no significant difference as an adjunctive treatment for patients with prominent negative symptoms of schizophrenia compared to placebo. Schizophr Res 150:434–441. [DOI] [PubMed] [Google Scholar]
- Stefani MR, Moghaddam B (2010) Activation of type 5 metabotropic glutamate receptors attenuates deficits in cognitive flexibility induced by NMDA receptor blockade. Eur J Pharmacol 639:26–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swedberg MD, Ellgren M, Raboisson P (2014) mGluR5 antagonist-induced psychoactive properties: MTEP drug discrimination, a pharmacologically selective non-NMDA effect with apparent lack of reinforcing properties. J Pharmacol Exp Ther 349:155–164. [DOI] [PubMed] [Google Scholar]
- Takamori K, Hirota S, Chaki S, Tanaka M (2003) Antipsychotic action of selective group II metabotropic glutamate receptor agonist MGS0008 and MGS0028 on conditioned avoidance responses in the rat. Life Sci 73:1721–1728. [DOI] [PubMed] [Google Scholar]
- Timms AEDorschner MOWechsler JChoi KYKirkwood RGirirajan SBaker CEichler EEKorvatska ORoche KW, et al. (2013) Support for the N-methyl-D-aspartate receptor hypofunction hypothesis of schizophrenia from exome sequencing in multiplex families. JAMA Psychiatry 70:582–590. [DOI] [PubMed] [Google Scholar]
- Uslaner JM, Parmentier-Batteur S, Flick RB, Surles NO, Lam JS, McNaughton CH, Jacobson MA, Hutson PH (2009) Dose-dependent effect of CDPPB, the mGluR5 positive allosteric modulator, on recognition memory is associated with GluR1 and CREB phosphorylation in the prefrontal cortex and hippocampus. Neuropharmacology 57:531–538. [DOI] [PubMed] [Google Scholar]
- Vales K, Svoboda J, Benkovicova K, Bubenikova-Valesova V, Stuchlik A (2010) The difference in effect of mGlu2/3 and mGlu5 receptor agonists on cognitive impairment induced by MK-801. Eur J Pharmacol 639:91–98. [DOI] [PubMed] [Google Scholar]
- von Engelhardt JDoganci BJensen VHvalby ØGöngrich CTaylor ABarkus CSanderson DJRawlins JNSeeburg PH, et al. (2008) Contribution of hippocampal and extra-hippocampal NR2B-containing NMDA receptors to performance on spatial learning tasks. Neuron 60:846–860. [DOI] [PubMed] [Google Scholar]
- Walker AG, Wenthur CJ, Xiang Z, Rook JM, Emmitte KA, Niswender CM, Lindsley CW, Conn PJ (2015) Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proc Natl Acad Sci USA 112:1196–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang HYMacDonald MLBorgmann-Winter KEBanerjee ASleiman PTom AKhan ALee KCRoussos PSiegel SJ, et al. (2020) mGluR5 hypofunction is integral to glutamatergic dysregulation in schizophrenia. Mol Psychiatry 25:750–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe M, Marcy B, Kinoshita K, Fukasawa M, Hikichi H, Chaki S, Okuyama S, Gevorkyan H, Yoshida S (2020) Safety and pharmacokinetic profiles of MGS0274 besylate (TS-134), a novel metabotropic glutamate 2/3 receptor agonist prodrug, in healthy subjects. Br J Clin Pharmacol 86:2286–2301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wierońska JMSławińska AŁasoń-Tyburkiewicz MGruca PPapp MZorn SHDoller DKłeczek NNoworyta-Sokołowska KGołembiowska K, et al. (2015) The antipsychotic-like effects in rodents of the positive allosteric modulator Lu AF21934 involve 5-HT1A receptor signaling: mechanistic studies. Psychopharmacology (Berl) 232:259–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wierońska JM, Stachowicz K, Acher F, Lech T, Pilc A (2012) Opposing efficacy of group III mGlu receptor activators, LSP1-2111 and AMN082, in animal models of positive symptoms of schizophrenia. Psychopharmacology (Berl) 220:481–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woolley ML, Pemberton DJ, Bate S, Corti C, Jones DN (2008) The mGlu2 but not the mGlu3 receptor mediates the actions of the mGluR2/3 agonist, LY379268, in mouse models predictive of antipsychotic activity. Psychopharmacology (Berl) 196:431–440. [DOI] [PubMed] [Google Scholar]
- Woźniak M, Acher F, Marciniak M, Lasoń-Tyburkiewicz M, Gruca P, Papp M, Pilc A, Wierońska JM (2016) Involvement of GABAB receptor signaling in antipsychotic-like action of the novel orthosteric agonist of the mGlu4 receptor, LSP4-2022. Curr Neuropharmacol 14:413–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woźniak M, Gołembiowska K, Noworyta-Sokołowska K, Acher F, Cieślik P, Kusek M, Tokarski K, Pilc A, Wierońska JM (2017) Neurochemical and behavioral studies on the 5-HT1A-dependent antipsychotic action of the mGlu4 receptor agonist LSP4-2022. Neuropharmacology 115:149–165. [DOI] [PubMed] [Google Scholar]
- Wroblewska B, Wroblewski JT, Pshenichkin S, Surin A, Sullivan SE, Neale JH (1997) N-acetylaspartylglutamate selectively activates mGluR3 receptors in transfected cells. J Neurochem 69:174–181. [DOI] [PubMed] [Google Scholar]
- Xiang Z, Lv X, Lin X, O’Brien DE, Altman MK, Lindsley CW, Javitch JA, Niswender CM, Conn PJ (2021) Input-specific regulation of glutamatergic synaptic transmission in the medial prefrontal cortex by mGlu2/mGlu4 receptor heterodimers. Sci Signal 14:eabd2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J, Antion MD, Nomura T, Kraniotis S, Zhu Y, Contractor A (2014) Hippocampal metaplasticity is required for the formation of temporal associative memories. J Neurosci 34:16762–16773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang FSnyder LBBalakrishnan ABrown JMSivarao DVEaston AFernandes AGulianello MHanumegowda UMHuang H, et al. (2016) Discovery and preclinical evaluation of BMS-955829, a potent positive allosteric modulator of mGluR5. ACS Med Chem Lett 7:289–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin S, Noetzel MJ, Johnson KA, Zamorano R, Jalan-Sakrikar N, Gregory KJ, Conn PJ, Niswender CM (2014) Selective actions of novel allosteric modulators reveal functional heteromers of metabotropic glutamate receptors in the CNS. J Neurosci 34:79–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yohn SEFoster DJCovey DPMoehle MSGalbraith JGarcia-Barrantes PMCho HPBubser MBlobaum ALJoffe ME, et al. (2020) Activation of the mGlu1 metabotropic glutamate receptor has antipsychotic-like effects and is required for efficacy of M4 muscarinic receptor allosteric modulators. Mol Psychiatry 25:2786–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeleznikow-Johnston AM, Renoir T, Churilov L, Li S, Burrows EL, Hannan AJ (2018) Touchscreen testing reveals clinically relevant cognitive abnormalities in a mouse model of schizophrenia lacking metabotropic glutamate receptor 5. Sci Rep 8:16412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhai Y, George CA, Zhai J, Nisenbaum ES, Johnson MP, Nisenbaum LK (2003) Group II metabotropic glutamate receptor modulation of DOI-induced c-fos mRNA and excitatory responses in the cerebral cortex. Neuropsychopharmacology 28:45–52. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Yoshida T, Katz DB, Lisman JE (2012) NMDAR antagonist action in thalamus imposes δ oscillations on the hippocampus. J Neurophysiol 107:3181–3189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo D, Bzdega T, Olszewski RT, Moffett JR, Neale JH (2012) Effects of N-acetylaspartylglutamate (NAAG) peptidase inhibition on release of glutamate and dopamine in prefrontal cortex and nucleus accumbens in phencyclidine model of schizophrenia. J Biol Chem 287:21773–21782. [DOI] [PMC free article] [PubMed] [Google Scholar]