Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Feb 24.
Published in final edited form as: Curr Opin Pharmacol. 2018 Feb 24;38:31–36. doi: 10.1016/j.coph.2018.02.003

The therapeutic potential of metabotropic glutamate receptor modulation for schizophrenia

Branden J Stansley 1,2, P Jeffrey Conn 1,2,*
PMCID: PMC5949078  NIHMSID: NIHMS943840  PMID: 29486374

Abstract

Accumulating evidence suggests that a dysregulation of the glutamatergic system exists in the brains of schizophrenia patients. The metabotropic glutamate (mGlu) receptors are being investigated as novel drug targets for this disease, and have shown promise in both preclinical and clinical studies. Activation of mGlu5 receptors may be efficacious for several symptom domains (positive, negative, and cognitive) and the potential for targeting mGlu5 receptors has been bolstered by recent research on mitigating toxicity profiles associated with mGlu5 activation. Additionally, genetic profiling of schizophrenia patients suggests that genes encoding for mGlu1 and mGlu3 receptors are altered, prompting preclinical studies that have demonstrated potential antipsychotic and cognitive enhancing effects of agents that activate mGlu1 and mGlu3 receptors, respectively. Development of subtype-specific drugs for the mGlu receptors, such as allosteric modulators, could provide a path forward for more efficacious and tolerable therapeutics for schizophrenia.


Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system (CNS). It mediates cellular signaling between brain cells by binding both ionotropic (i.e. N-methyl-D-aspartate (NMDA), a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate) [1] and metabotropic glutamate (mGlu) receptors [2], thus contributing to information processing through neuronal network activity and synaptic plasticity [3]. Pharmacological modulation of the glutamatergic system is therefore poised to restore neuronal signaling and provide therapeutic benefit in neurological disease states. Schizophrenia, a neuropsychiatric disorder with a prevalence of around 1% of the world population [4], is one such psychiatric disorder that is highly amenable to glutamatergic modulation [5, 6]. Schizophrenia is clinically diagnosed in the United States according the criteria set by the current edition of the Diagnostic and Statistical Manual of Mental Disorders 5 (DSM-V) [7]. Of the three main symptom clusters found in schizophrenia patients, current typical and atypical antipsychotic medications that target the monoamine receptors have shown efficacy toward the positive (e.g., auditory and visual hallucinations, delusions), but not negative (e.g., social withdrawal, flat affect) nor cognitive (e.g., impairments in executive function, working memory and cognitive flexibility) symptoms [8]. Unfortunately, the efficacy of these medications is met with significant unwanted extrapyramidal side effects and weight gain, resulting in a significant unmet clinical need for improved pharmacological therapeutics.

Ongoing basic research into the biology of glutamate receptors has yielded significant knowledge that can be leveraged to improve drug discovery efforts for schizophrenia. Although the etiology of the disease is unknown, considerable evidence suggests that NMDA receptor hypofunction may contribute to the pathophysiology of schizophrenia [9, 10]. For example, administration of the NMDA receptor antagonist phencyclidine (PCP) causes schizophrenia-like symptoms in healthy individuals [11], and the NMDA receptor antagonist ketamine can exacerbate symptoms in individuals with schizophrenia [12]. These studies suggest an important role for NMDA receptor signaling in the pathology of schizophrenia; however, modulation of NMDA receptors using orthosteric agonists produces neuronal excitotoxicity and seizures, prohibiting direct NMDA receptor targeting as a viable therapeutic option. Alternatively, targeting the mGlu receptors pharmacologically may allow for regulation of glutamatergic neurotransmission and more precise tuning of the excitatory/inhibitory balance of the brain.

The mGlu receptors are G protein-coupled protein receptors that are sub-divided into three groups (Group I: mGlu1 and mGlu5; Group II: mGlu2, mGlu3; Group III: mGlu4, mGlu6, mGlu7, mGlu8), based on sequence homology, agonist selectivity and secondary signaling cascades [13]. As pharmacology efforts have advanced, the ability to attain subtype specificity of mGlu receptors has been accomplished through development of allosteric modulators. Positive allosteric modulators (PAMs) act to potentiate endogenous glutamate signaling, while negative allosteric modulators (NAMs) reduce the receptor responsiveness to glutamate [14]. In the context of CNS disorders, drug discovery efforts have elucidated beneficial modes of action for allosteric modulators that target subtypes within each of the three groups. Hence, several mGlu receptor subtypes represent novel therapeutic targets with potential utility for treatment of schizophrenia.

Group I mGlu receptors

The group I mGlu receptors consist of mGlu1 and mGlu5, both of which are coupled to signaling proteins that stimulate phosphoinositide hydrolysis and phospholipase C [2]. For many years, mGlu5 has been considered an appealing therapeutic target due to its interaction with NMDA receptors through structural connections with scaffolding proteins such as Homer and Shank [15], as well as functionally by potentiation of NMDA receptor responses [16]. Correspondingly, positive allosteric modulation of mGlu5 has been shown to enhance long-term plasticity (both long-term potentiation (LTP) and long-term depression (LTD)) in the hippocampus, and has pro-cognitive effects in healthy rodents [17]. When mGlu5 is genetically deleted from mice, NMDA receptor-dependent plasticity and learning is impaired [18]. The mGlu5-knockout mice have also been shown to display a disruption in prepulse inhibition (PPI) [19], a model of sensory motor gating that is disrupted in schizophrenia patients, leading to the hypothesis that mGlu5 may represent a therapeutic target for schizophrenia. Preclinical research has established that mGlu5 PAMs could be efficacious for schizophrenia in assays with predictive validity [2022]. Albeit promising, the development of compounds that target the mGlu5 receptors for treatment of neurological disorders have been stymied by preclinical toxicology issues, possibly related to excessive NMDA receptor activation [23]. Mechanistic studies point to the possibility that subsets of mGlu5 PAMs can be biased away from NMDA receptor current potentiation, thus mitigating excitotoxic/seizure-inducing profiles [24]. Interestingly, compounds such as VU0409551 that do not potentiate effects of mGlu5 on NMDA receptor currents [25], retain their efficacy in rodent models, displaying antipsychotic-like and cognitive enhancing activity [26, 27]. These beneficial effects may be related to the ability of mGlu5 PAMs to modify long-term plasticity through non-NMDA receptor related mechanisms such as enhanced excitability of pyramidal neurons [28], enhancement of long-term plasticity requiring protein synthesis [29], or possibly depression of inhibitory tone through local messengers such as cannabinoids [30]. Taken together, mGlu5 represents a target that could be efficacious for positive, negative and cognitive symptoms of schizophrenia. However, this may be contingent on the potential to develop mGlu5 PAMs without adverse side-effect liability.

While mGlu5 has been heavily studied as a potential target for pharmacotherapy, mGlu1 has received less attention. However, multiple mutations in the gene encoding for human mGlu1 receptor, GRM1, have been identified in schizophrenia patients, raising the possibility that mGlu1 may be dysregulated in schizophrenia [31]. This is supported by the finding that mGlu1 mRNA expression is dysregulated in tissue of postmortem schizophrenia patients [32]. Interestingly, mutations that have been found to be associated with schizophrenia leads to a decrease in mGlu1 signaling in-vitro, and a range of highly selective mGlu1 PAMs can potentiate responses to activation of the mutant receptors [33], raising the possibility that mGlu1 PAMs could reverse deficits in mGlu1 signaling in schizophrenia patients that bear these mutations.

Preclinically, mGlu1 knockout mice have a pathological phenotype that mirrors the symptomology of schizophrenia, such as deficits in PPI [34]. Given the hyperdopaminergic state of the schizophrenic brain [35, 36], drugs that antagonize the dopamine system have been a mainstay of the therapeutic treatment. Therefore, production of new drugs that act to dampen dopaminergic efflux would be predicted to have antipsychotic efficacy. It was demonstrated that group I mGlu activation depresses dopamine release in the dorsal striatum of mice using fast-scan cyclic voltammetry ex-vivo; an effect likely attributable specifically to mGlu1 activation, as the presence of mGlu5 antagonist or using mGlu5–null mice did not affect group I mGlu agonist-evoked dopamine release [37]. Indeed, administration of a selective mGlu1 PAM, VU6004909, causes dopamine release to be significantly attenuated in the dorsal striatum after electrical stimulation of the medial forebrain bundle in rats [38]. Incongruently, mGlu1 NAMs are also beneficial in preclinical assays such as methamphetamine induced-hyperlocomotion and disruption of PPI [39]. Therefore, more research is necessary to elucidate the precise mechanisms by which mGlu1 modulation shows efficacy in these assays. These data also highlight the important fact that underlying individual pathology such as the presence of GRM1 mutations may promote the utility of mGlu1 specific drugs.

Group II mGlu receptors

The group II mGlu receptors, consisting of mGlu2 and mGlu3, couple to G-protein subunits that inhibit adenylate-cyclase activity [2]. Activation of these receptors by group II mGlu receptor orthosteric agonists has been shown to enhance the function of NMDA receptors [40], as well as regulate LTD and LTP in the prefrontal cortex and the hippocampus, respectively [4143]. Furthermore, activation of group II mGlu receptors reduces excessive activity at glutamatergic synapses in the PFC that is induced by hallucinogens such as the 5-HT2A receptor agonist DOI or administration of NMDA receptor antagonists [44, 45]. Therefore, these receptors are prime targets for the treatment of schizophrenia where NMDA receptor dysregulation and plasticity impairments are thought to be occurring in these brain areas. Indeed, seminal work testing target viability of the group II receptors for schizophrenia was conducted in rats and revealed that the mGlu2/3 agonist, LY354740, was capable of rescuing deficits in stereotypy, locomotion, spatial working memory and cortical glutamate efflux induced by the NMDA-receptor antagonist PCP [46]. This finding was strengthened by the translation of mGlu2/3 agonist efficacy in attenuating NMDA receptor-induced deficits in working memory within human subjects [47]. Clinically, the mGlu2/3 agonist LY2140023, developed by Eli Lilly and Company, entered a double blind-placebo controlled phase II clinical trial and showed safety and tolerability; without weight gain or extrapyramidal side effects seen with typical antipsychotics. Most importantly, there was also a significant improvement in positive and negative symptoms of schizophrenia compared to placebo [48]. However, the effects of LY2140023 were washed out by a large placebo effect in a follow-up phase II study [49]. Combined with a subsequent placebo controlled study where the atypical antipsychotic risperidone improved scores on the Positive and Negative Symptom Scale, the mGlu2/3 agonist LY2140023 failed to show efficacy [50]. While the reasons for clinical failure can be manifold, the importance of dose-response was exemplified by a recent non-human primate study that demonstrated mGlu2/3 agonist enhancement of working memory follows an inverted-U dose-response, where low doses improve working memory but higher doses have varied effects [51]. Nonetheless, given the mixed results from these trials and others, Eli Lilly terminated the further development of this compound as a clinical candidate. While these trials were not successful, the fact that the drug did have benefit in some patients suggests that perhaps mGlu2 or mGlu3 specific drugs could yield better results clinically.

Selective mGlu2 PAMs have been heavily pursued as drug candidates for schizophrenia given that preclinical data suggest that mGlu2 primarily mediates the antipsychotic-like effects of mGlu2/3 agonists in animal models. This is evidenced by the finding that the mGlu2/3 agonist, LY314582, fails to reduce NMDA receptor-antagonist induced hyper-locomotion in mGlu2 KO mice [52]. Several mGlu2 PAMs, including LY487379 and biphenyl-indanone A (BINA), have shown efficacy in preclinical animal models of schizophrenia [53, 54] (for in-depth review see [55]). This research paved the way for mGlu2 PAM testing in clinical trials. However, a proof of principle phase II study of AstraZeneca’s mGlu2 PAM AZD8529 did not demonstrate efficacy of the drug candidate compared to placebo, while the atypical antipsychotic risperidone demonstrated significant efficacy [56]. These results taken together illustrate the difficulty of translating preclinical research findings, and highlight the potential shortcomings in animal modeling approaches. Certainly, the underlying pathology within clinical populations of schizophrenia patients is heterogeneous and disease etiology is likely to be multifaceted; possibly making activation of a single target insufficient to provide therapeutic benefit for a wide population sample. Therefore, the ability to stratify patient population based on biomarkers or genotype may enable pharmacotherapy to be tailored and boost efficacy for individual patients.

Mutations in GRM3, the gene encoding for mGlu3, have been shown to be associated with schizophrenia and deficits in cognitive function [57, 58]. The generation of knockout mice and development of specific mGlu3 tool compounds has enabled researchers to begin to uncover the basic functions of mGlu3 in the CNS. Within the prefrontal cortex (PFC), an area of the brain important for working memory and cognition, mGlu3 functions postsynaptically to induce long-term depression (LTD) of excitatory transmission as well as induce calcium mobilization [41] and increase pyramidal cell excitability through inhibition of postsynaptic cAMP-K+ channel actions [51]. Furthermore, mGlu3 modulates plasticity within the hippocampus, a declarative memory center that is altered in schizophrenia [59]. mGlu3 is required for induction of LTD in the dentate gyrus subregion [60], and facilitates LTP through post-synaptic mechanisms in the CA1 subregion [43]. However, under some conditions activation of mGlu3 can inhibit induction of LTP. Specifically, activation of mGlu3 in hippocampal astrocytes, during corresponding beta-adrenergic receptor activation leads to an attenuation of LTP enhancement through a novel form of glial-neuronal communication [42], suggesting that mGlu3 modulates plasticity differently depending on the activity of the network.

Behaviorally, mGlu3 alterations could contribute to phenotypes associated with schizophrenia given that mGlu3 knockout mice show hyperactivity as well as deficits in reference memory and working memory tests [61], paralleling some known functions of the group I mGlu, mGlu5. Interestingly, recent preclinical evidence uncovered intriguing interactions of mGlu3 and mGlu5, where activation of mGlu3 potentiates mGlu5 signaling. Furthermore, mGlu3-dependent LTD in the PFC was shown to be dependent on mGlu5 activity [62]. Combined with the fact that mGlu3 may provide neuroprotection in response to NMDA receptor-mediated toxicity [63], a drug activating mGlu3 may reduce the risk for cytotoxicity while improving excitatory signaling. Therefore, mGlu3 represents a novel target for cognitive dysfunction in schizophrenia patients, and development of specific mGlu3 potentiators could provide valuable adjunct therapy options.

Group III mGlu receptors

The group III receptors are coupled to signaling cascades that inhibit adenylate cyclase activity, and thus primarily function as presynaptic autoreceptors. Aside from mGlu6, which is restricted to the retina, mGlu4, mGlu7, and mGlu8 are all potential targets to normalize glutamatergic tone within the CNS of schizophrenia patients [13]. Basic research in the field is in its early stages of validating these receptors as specific targets, however, group III agonists such as ACPT-I possess antipsychotic-like effects in animal models by reducing PCP-induced hyperlocomotion in rodents [64]. In addition, the selective mGlu4 PAM, ADX88178, can improve indicators of all three symptom clusters preclinically [65]. Evidence that mGlu4 could heterodimerize with mGlu2 in vivo [66] supports the possibility of targeting this mGlu2/4 complex to provide even greater subtype selectivity; although the antipsychotic efficacy of such a drug has yet to be tested experimentally. While mGlu7 and mGlu8 both play a role in cognitive functions [67, 68] and glutamatergic signaling in the hippocampus [69, 70], direct evidence for these receptors as targets for schizophrenia is lacking. In fact, current data may suggest that mGlu7 PAMs could exacerbate positive symptoms [71]. More research is certainly required using more extensive preclinical modeling before the value of these targets can be ruled out, especially as more advanced selective compounds become available.

Conclusion

The fact that the negative and cognitive symptom domains of schizophrenia are often insufficiently controlled in the clinical setting substantiates the need for new pharmacotherapies for this disorder. The mGlu receptors represent several novel targets for the treatment of schizophrenia, with receptors from each of the three mGlu sub-groups showing promise. Based on preclinical data, mGlu5 PAMs that are devoid of toxicity and seizure liability could yield effective treatment for all three symptom domains. Furthermore, the genetic alterations in the genes encoding mGlu1 and mGlu3 within the schizophrenia patient population support recent preclinical data suggesting potential antipsychotic and cognitive enhancing attributes of drugs targeting these receptors. It will be of interest to see how future investigations expound upon this data. Together, the studies outlined in this review illuminate the ongoing basic and clinical research that will undoubtedly drive discovery efforts to make safer and more efficacious mGlu targeted drugs for this disorder.

Highlights.

  • mGlu5 PAMs are efficacious for positive, negative, and cognitive symptoms in animal models

  • Clinical trials for mGlu2 PAMs have not been successful

  • Genetic studies have identified GRM1 and GRM3 as dysregulated in the schizophrenic brain

  • mGlu1 PAMs reduce dopamine release in preclinical studies and could be potentially beneficial for positive symptoms

  • mGlu3 activation appears to be pro-cognitive and neuroprotective preclinically

Acknowledgments

PJC receives funding from the National Institutes of Health (R01MH062646, R37NS031373). BJS is the recipient of a postdoctoral fellowship from the National Institute of Health (F32MH111124).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62(3):405–96. doi: 10.1124/pr.109.002451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Conn PJ, Pin JP. Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol. 1997;37:205–37. doi: 10.1146/annurev.pharmtox.37.1.205. [DOI] [PubMed] [Google Scholar]
  • 3.Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature. 1993;361(6407):31–9. doi: 10.1038/361031a0. [DOI] [PubMed] [Google Scholar]
  • 4.McGrath J, Saha S, Chant D, Welham J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol Rev. 2008;30:67–76. doi: 10.1093/epirev/mxn001. [DOI] [PubMed] [Google Scholar]
  • 5.Noetzel MJ, Jones CK, Conn PJ. Emerging approaches for treatment of schizophrenia: modulation of glutamatergic signaling. Discov Med. 2012;14(78):335–43. [PMC free article] [PubMed] [Google Scholar]
  • 6.Moghaddam B, Javitt D. From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology. 2012;37(1):4–15. doi: 10.1038/npp.2011.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tandon R, Gaebel W, Barch DM, Bustillo J, Gur RE, Heckers S, Malaspina D, Owen MJ, Schultz S, Tsuang M, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150(1):3–10. doi: 10.1016/j.schres.2013.05.028. [DOI] [PubMed] [Google Scholar]
  • 8.Li P, Snyder GL, Vanover KE. Dopamine Targeting Drugs for the Treatment of Schizophrenia: Past, Present and Future. Curr Top Med Chem. 2016;16(29):3385–3403. doi: 10.2174/1568026616666160608084834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Javitt DC. Glutamatergic theories of schizophrenia. Isr J Psychiatry Relat Sci. 2010;47(1):4–16. [PubMed] [Google Scholar]
  • 10.Coyle JT. NMDA receptor and schizophrenia: a brief history. Schizophr Bull. 2012;38(5):920–6. doi: 10.1093/schbul/sbs076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Domino E, Luby ED. Abnormal mental states induced by phencyclidine as a model of schizophrenia, Phencyclidine: Historical and Current Perspectives. NPP Books; Ann Arbor, MI: 1981. pp. 401–418. [Google Scholar]
  • 12.Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51(3):199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
  • 13.Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Conn PJ, Christopoulos A, Lindsley CW. Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat Rev Drug Discov. 2009;8(1):41–54. doi: 10.1038/nrd2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakeman P, Doan A, Aakalu VK, Lanahan AA, Sheng M, et al. Coupling of mGluR/Homer and PSD-95 complexes by the Shank family of postsynaptic density proteins. Neuron. 1999;23(3):583–92. doi: 10.1016/s0896-6273(00)80810-7. [DOI] [PubMed] [Google Scholar]
  • 16.Awad H, Hubert GW, Smith Y, Levey AI, Conn PJ. Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci. 2000;20(21):7871–9. doi: 10.1523/JNEUROSCI.20-21-07871.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ayala JE, Chen Y, Banko JL, Sheffler DJ, Williams R, Telk AN, Watson NL, Xiang Z, Zhang Y, Jones PJ, et al. mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology. 2009;34(9):2057–71. doi: 10.1038/npp.2009.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jia Z, Lu Y, Henderson J, Taverna F, Romano C, Abramow-Newerly W, Wojtowicz JM, Roder J. Selective abolition of the NMDA component of long-term potentiation in mice lacking mGluR5. Learn Mem. 1998;5(4–5):331–43. [PMC free article] [PubMed] [Google Scholar]
  • 19.Brody SA, Dulawa SC, Conquet F, Geyer MA. Assessment of a prepulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Mol Psychiatry. 2004;9(1):35–41. doi: 10.1038/sj.mp.4001404. [DOI] [PubMed] [Google Scholar]
  • 20.Kinney GG, O’Brien JA, Lemaire W, Burno M, Bickel DJ, Clements MK, Chen TB, Wisnoski DD, Lindsley CW, Tiller PR, et al. A novel selective positive allosteric modulator of metabotropic glutamate receptor subtype 5 has in vivo activity and antipsychotic-like effects in rat behavioral models. J Pharmacol Exp Ther. 2005;313(1):199–206. doi: 10.1124/jpet.104.079244. [DOI] [PubMed] [Google Scholar]
  • 21.Kinney GG, Burno M, Campbell UC, Hernandez LM, Rodriguez D, Bristow LJ, Conn PJ. Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther. 2003;306(1):116–23. doi: 10.1124/jpet.103.048702. [DOI] [PubMed] [Google Scholar]
  • 22.Gastambide F, Cotel MC, Gilmour G, O’Neill MJ, Robbins TW, Tricklebank MD. Selective remediation of reversal learning deficits in the neurodevelopmental MAM model of schizophrenia by a novel mGlu5 positive allosteric modulator. Neuropsychopharmacology. 2012;37(4):1057–66. doi: 10.1038/npp.2011.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Parmentier-Batteur S, Hutson PH, Menzel K, Uslaner JM, Mattson BA, O’Brien JA, Magliaro BC, Forest T, Stump CA, Tynebor RM, et al. Mechanism based neurotoxicity of mGlu5 positive allosteric modulators--development challenges for a promising novel antipsychotic target. Neuropharmacology. 2014;82:161–73. doi: 10.1016/j.neuropharm.2012.12.003. [DOI] [PubMed] [Google Scholar]
  • 24.Rook JM, Noetzel MJ, Pouliot WA, Bridges TM, Vinson PN, Cho HP, Zhou Y, Gogliotti RD, Manka JT, Gregory KJ, et al. Unique signaling profiles of positive allosteric modulators of metabotropic glutamate receptor subtype 5 determine differences in in vivo activity. Biol Psychiatry. 2013;73(6):501–9. doi: 10.1016/j.biopsych.2012.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25•.Rook JM, Xiang Z, Lv X, Ghoshal A, Dickerson JW, Bridges TM, Johnson KA, Foster DJ, Gregory KJ, Vinson PN, et al. Biased mGlu5-Positive Allosteric Modulators Provide In Vivo Efficacy without Potentiating mGlu5 Modulation of NMDAR Currents. Neuron. 2015;86(4):1029–40. doi: 10.1016/j.neuron.2015.03.063. A manuscript detailing the discovery that biased mGlu5 PAMs activate unique signalling pathways and exclude NMDA receptor modulation and excitotoxicity in rodents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26••.Balu DT, Li Y, Takagi S, Presti KT, Ramikie TS, Rook JM, Jones CK, Lindsley CW, Conn PJ, Bolshakov VY, et al. An mGlu5-Positive Allosteric Modulator Rescues the Neuroplasticity Deficits in a Genetic Model of NMDA Receptor Hypofunction in Schizophrenia. Neuropsychopharmacology. 2016;41(8):2052–61. doi: 10.1038/npp.2016.2. An elegant study demonstrating mGlu5 positive allosteric modulation in correcting plasticity impairments in a mouse model of schizophrenia. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ghoshal A, Moran SP, Dickerson JW, Joffe ME, Grueter BA, Xiang Z, Lindsley CW, Rook JM, Conn PJ. Role of mGlu5 receptors and inhibitory neurotransmission in M1 dependent muscarinic LTD in the prefrontal cortex: implications in schizophrenia. ACS Chem Neurosci. 2017 doi: 10.1021/acschemneuro.7b00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mannaioni G, Marino MJ, Valenti O, Traynelis SF, Conn PJ. Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. J Neurosci. 2001;21(16):5925–34. doi: 10.1523/JNEUROSCI.21-16-05925.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Huber KM, Roder JC, Bear MF. Chemical induction of mGluR5- and protein synthesis--dependent long-term depression in hippocampal area CA1. J Neurophysiol. 2001;86(1):321–5. doi: 10.1152/jn.2001.86.1.321. [DOI] [PubMed] [Google Scholar]
  • 30.Xu J, Antion MD, Nomura T, Kraniotis S, Zhu Y, Contractor A. Hippocampal metaplasticity is required for the formation of temporal associative memories. J Neurosci. 2014;34(50):16762–73. doi: 10.1523/JNEUROSCI.2869-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ayoub MA, Angelicheva D, Vile D, Chandler D, Morar B, Cavanaugh JA, Visscher PM, Jablensky A, Pfleger KD, Kalaydjieva L. Deleterious GRM1 mutations in schizophrenia. PLoS One. 2012;7(3):e32849. doi: 10.1371/journal.pone.0032849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Volk DW, Eggan SM, Lewis DA. Alterations in metabotropic glutamate receptor 1alpha and regulator of G protein signaling 4 in the prefrontal cortex in schizophrenia. Am J Psychiatry. 2010;167(12):1489–98. doi: 10.1176/appi.ajp.2010.10030318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33•.Cho HP, Garcia-Barrantes PM, Brogan JT, Hopkins CR, Niswender CM, Rodriguez AL, Venable DF, Morrison RD, Bubser M, Daniels JS, et al. Chemical modulation of mutant mGlu1 receptors derived from deleterious GRM1 mutations found in schizophrenics. ACS Chem Biol. 2014;9(10):2334–46. doi: 10.1021/cb500560h. The novel mGlu1 PAMs described here partially rescue receptor function caused by deleterious GRM1 mutations found in schizophrenic patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Brody SA, Conquet F, Geyer MA. Disruption of prepulse inhibition in mice lacking mGluR1. Eur J Neurosci. 2003;18(12):3361–6. doi: 10.1111/j.1460-9568.2003.03073.x. [DOI] [PubMed] [Google Scholar]
  • 35.Laruelle M, Abi-Dargham A, Gil R, Kegeles L, Innis R. Increased dopamine transmission in schizophrenia: relationship to illness phases. Biol Psychiatry. 1999;46(1):56–72. doi: 10.1016/s0006-3223(99)00067-0. [DOI] [PubMed] [Google Scholar]
  • 36.Howes OD, Montgomery AJ, Asselin MC, Murray RM, Valli I, Tabraham P, Bramon-Bosch E, Valmaggia L, Johns L, Broome M, et al. Elevated striatal dopamine function linked to prodromal signs of schizophrenia. Arch Gen Psychiatry. 2009;66(1):13–20. doi: 10.1001/archgenpsychiatry.2008.514. [DOI] [PubMed] [Google Scholar]
  • 37.Zhang H, Sulzer D. Glutamate spillover in the striatum depresses dopaminergic transmission by activating group I metabotropic glutamate receptors. J Neurosci. 2003;23(33):10585–92. doi: 10.1523/JNEUROSCI.23-33-10585.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yohn SECD, Foster DJ, Garcia-Barrantes PM, Joffe ME, Cheer JP, Lindsley CW, Conn PJ. Regulation of Striatal Dopamine Release by Metabotropic Glutamate Receptor Subtype 1 via an Endocannabinoid-Dependent Mechanism: Implications for the Treatment of Schizophrenia. Abstract, Society for Neuroscience. 2017 [Google Scholar]
  • 39.Satow A, Suzuki G, Maehara S, Hikichi H, Murai T, Kawagoe-Takaki H, Hata M, Ito S, Ozaki S, Kawamoto H, et al. 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-dih ydro-1H-isoindol-1-one. J Pharmacol Exp Ther. 2009;330(1):179–90. doi: 10.1124/jpet.109.151118. [DOI] [PubMed] [Google Scholar]
  • 40.Tyszkiewicz JP, Gu Z, Wang X, Cai X, Yan Z. Group II metabotropic glutamate receptors enhance NMDA receptor currents via a protein kinase C-dependent mechanism in pyramidal neurones of rat prefrontal cortex. J Physiol. 2004;554(Pt 3):765–77. doi: 10.1113/jphysiol.2003.056812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Walker AG, Wenthur CJ, Xiang Z, Rook JM, Emmitte KA, Niswender CM, Lindsley CW, Conn PJ. Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proc Natl Acad Sci U S A. 2015;112(4):1196–201. doi: 10.1073/pnas.1416196112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42••.Walker AG, Sheffler DJ, Lewis AS, Dickerson JW, Foster DJ, Senter RK, Moehle MS, Lv X, Stansley BJ, Xiang Z, et al. Co-Activation of Metabotropic Glutamate Receptor 3 and Beta-Adrenergic Receptors Modulates Cyclic-AMP, Long-Term Potentiation, and Disrupts Memory Reconsolidation. Neuropsychopharmacology. 2017 doi: 10.1038/npp.2017.136. An investigation of hippocampal mGlu3 signialling in astrocytes and its role in plasticity and cognition, providing target validation for cognitive domains. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43••.Rosenberg N, Gerber U, Ster J. Activation of Group II Metabotropic Glutamate Receptors Promotes LTP Induction at Schaffer Collateral-CA1 Pyramidal Cell Synapses by Priming NMDA Receptors. J Neurosci. 2016;36(45):11521–11531. doi: 10.1523/JNEUROSCI.1519-16.2016. This paper demonstrates an important role for mGlu3 in promoting LTP through NMDA receptor-dependent mechanisms in the hippocampus, suggesting potential cognitive enhancing effects of mGlu3 pharmacological targeting. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Homayoun H, Jackson ME, Moghaddam B. Activation of metabotropic glutamate 2/3 receptors reverses the effects of NMDA receptor hypofunction on prefrontal cortex unit activity in awake rats. J Neurophysiol. 2005;93(4):1989–2001. doi: 10.1152/jn.00875.2004. [DOI] [PubMed] [Google Scholar]
  • 45.Klodzinska A, Bijak M, Tokarski K, Pilc A. Group II mGlu receptor agonists inhibit behavioural and electrophysiological effects of DOI in mice. Pharmacol Biochem Behav. 2002;73(2):327–32. doi: 10.1016/s0091-3057(02)00845-6. [DOI] [PubMed] [Google Scholar]
  • 46.Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science. 1998;281(5381):1349–52. doi: 10.1126/science.281.5381.1349. [DOI] [PubMed] [Google Scholar]
  • 47.Krystal JH, Abi-Saab W, Perry E, D’Souza DC, Liu N, Gueorguieva R, McDougall L, Hunsberger T, Belger A, Levine L, et al. 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) 2005;179(1):303–9. doi: 10.1007/s00213-004-1982-8. [DOI] [PubMed] [Google Scholar]
  • 48.Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM, Gurovich IY, Morozova MA, et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med. 2007;13(9):1102–7. doi: 10.1038/nm1632. [DOI] [PubMed] [Google Scholar]
  • 49.Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K, Kryzhanovskaya L, Jarkova N. A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin Psychopharmacol. 2011;31(3):349–55. doi: 10.1097/JCP.0b013e318218dcd5. [DOI] [PubMed] [Google Scholar]
  • 50.Downing AM, Kinon BJ, Millen BA, Zhang L, Liu L, Morozova MA, Brenner R, Rayle TJ, Nisenbaum L, Zhao F, et al. A Double-Blind, Placebo-Controlled Comparator Study of LY2140023 monohydrate in patients with schizophrenia. BMC Psychiatry. 2014;14:351. doi: 10.1186/s12888-014-0351-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51••.Jin LE, Wang M, Yang ST, Yang Y, Galvin VC, Lightbourne TC, Ottenheimer D, Zhong Q, Stein J, Raja A, et al. mGluR2/3 mechanisms in primate dorsolateral prefrontal cortex: evidence for both presynaptic and postsynaptic actions. Mol Psychiatry. 2016 doi: 10.1038/mp.2016.129. An elegant study showing mGlu2/3 agonists demonstrate an inverted-U dose response on working memory in non-human primates, providing mechanistic insight into prefrontal cortex modulation by group II mGlu agonists. [DOI] [PMC free article] [PubMed]
  • 52.Spooren WP, Gasparini F, van der Putten H, Koller M, Nakanishi S, Kuhn R. 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. 2000;397(1):R1–2. doi: 10.1016/s0014-2999(00)00269-7. [DOI] [PubMed] [Google Scholar]
  • 53.Galici R, Echemendia NG, Rodriguez AL, Conn PJ. 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. 2005;315(3):1181–7. doi: 10.1124/jpet.105.091074. [DOI] [PubMed] [Google Scholar]
  • 54.Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, de Paulis T, Conn PJ. 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. 2006;318(1):173–85. doi: 10.1124/jpet.106.102046. [DOI] [PubMed] [Google Scholar]
  • 55.Maksymetz J, Moran SP, Conn PJ. Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Mol Brain. 2017;10(1):15. doi: 10.1186/s13041-017-0293-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Litman RE, Smith MA, Doherty JJ, Cross A, Raines S, Gertsik L, Zukin SR. AZD8529, a positive allosteric modulator at the mGluR2 receptor, does not improve symptoms in schizophrenia: A proof of principle study. Schizophr Res. 2016;172(1–3):152–7. doi: 10.1016/j.schres.2016.02.001. [DOI] [PubMed] [Google Scholar]
  • 57••.Egan MF, Straub RE, Goldberg TE, Yakub I, Callicott JH, Hariri AR, Mattay VS, Bertolino A, Hyde TM, Shannon-Weickert C, et al. Variation in GRM3 affects cognition, prefrontal glutamate, and risk for schizophrenia. Proc Natl Acad Sci U S A. 2004;101(34):12604–9. doi: 10.1073/pnas.0405077101. Clinical study in which the mGlu2 PAM, AZD8529, was shown not to have efficacy in a schizophrenic population. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58••.Saini SM, Mancuso SG, Mostaid MS, Liu C, Pantelis C, Everall IP, Bousman CA. Meta-analysis supports GWAS-implicated link between GRM3 and schizophrenia risk. Transl Psychiatry. 2017;7(8):e1196. doi: 10.1038/tp.2017.172. This paper identifies a link between GRM3 genetic variation and risk for schizophrenia through a genome-wide association study. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tamminga CA, Stan AD, Wagner AD. The hippocampal formation in schizophrenia. Am J Psychiatry. 2010;167(10):1178–93. doi: 10.1176/appi.ajp.2010.09081187. [DOI] [PubMed] [Google Scholar]
  • 60.Poschel B, Wroblewska B, Heinemann U, Manahan-Vaughan D. The metabotropic glutamate receptor mGluR3 is critically required for hippocampal long-term depression and modulates long-term potentiation in the dentate gyrus of freely moving rats. Cereb Cortex. 2005;15(9):1414–23. doi: 10.1093/cercor/bhi022. [DOI] [PubMed] [Google Scholar]
  • 61.Fujioka R, Nii T, Iwaki A, Shibata A, Ito I, Kitaichi K, Nomura M, Hattori S, Takao K, Miyakawa T, et al. Comprehensive behavioral study of mGluR3 knockout mice: implication in schizophrenia related endophenotypes. Mol Brain. 2014;7:31. doi: 10.1186/1756-6606-7-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Di Menna L, Joffe ME, Iacovelli L, Orlando R, Lindsley CW, Mairesse J, Gressens P, Cannella M, Caraci F, Copani A, Bruno V, Battaglia G, Conn PJ, Nicoletti F. Functional partnership between mGlu3 and mGlu5 metabotropic glutamate receptors in neurons. Pharmacology. 2017 doi: 10.1016/j.neuropharm.2017.10.026. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Corti C, Battaglia G, Molinaro G, Riozzi B, Pittaluga A, Corsi M, Mugnaini M, Nicoletti F, Bruno V. The use of knock-out mice unravels distinct roles for mGlu2 and mGlu3 metabotropic glutamate receptors in mechanisms of neurodegeneration/neuroprotection. J Neurosci. 2007;27(31):8297–308. doi: 10.1523/JNEUROSCI.1889-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Palucha-Poniewiera A, Klodzinska A, Stachowicz K, Tokarski K, Hess G, Schann S, Frauli M, Neuville P, Pilc A. Peripheral administration of group III mGlu receptor agonist ACPT-I exerts potential antipsychotic effects in rodents. Neuropharmacology. 2008;55(4):517–24. doi: 10.1016/j.neuropharm.2008.06.033. [DOI] [PubMed] [Google Scholar]
  • 65.Kalinichev M, Le Poul E, Bolea C, Girard F, Campo B, Fonsi M, Royer-Urios I, Browne SE, Uslaner JM, Davis MJ, et al. Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. J Pharmacol Exp Ther. 2014;350(3):495–505. doi: 10.1124/jpet.114.214437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yin S, Noetzel MJ, Johnson KA, Zamorano R, Jalan-Sakrikar N, Gregory KJ, Conn PJ, Niswender CM. Selective actions of novel allosteric modulators reveal functional heteromers of metabotropic glutamate receptors in the CNS. J Neurosci. 2014;34(1):79–94. doi: 10.1523/JNEUROSCI.1129-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Holscher C, Schmid S, Pilz PK, Sansig G, van der Putten H, Plappert CF. Lack of the metabotropic glutamate receptor subtype 7 selectively impairs short-term working memory but not long-term memory. Behav Brain Res. 2004;154(2):473–81. doi: 10.1016/j.bbr.2004.03.015. [DOI] [PubMed] [Google Scholar]
  • 68.Gerlai R, Adams B, Fitch T, Chaney S, Baez M. Performance deficits of mGluR8 knockout mice in learning tasks: the effects of null mutation and the background genotype. Neuropharmacology. 2002;43(2):235–49. doi: 10.1016/s0028-3908(02)00078-3. [DOI] [PubMed] [Google Scholar]
  • 69.Klar R, Walker AG, Ghose D, Grueter BA, Engers DW, Hopkins CR, Lindsley CW, Xiang Z, Conn PJ, Niswender CM. Activation of Metabotropic Glutamate Receptor 7 Is Required for Induction of Long-Term Potentiation at SC-CA1 Synapses in the Hippocampus. J Neurosci. 2015;35(19):7600–15. doi: 10.1523/JNEUROSCI.4543-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhai J, Tian MT, Wang Y, Yu JL, Koster A, Baez M, Nisenbaum ES. Modulation of lateral perforant path excitatory responses by metabotropic glutamate 8 (mGlu8) receptors. Neuropharmacology. 2002;43(2):223–30. doi: 10.1016/s0028-3908(02)00087-4. [DOI] [PubMed] [Google Scholar]
  • 71.Wieronska JM, Stachowicz K, Acher F, Lech T, Pilc A. Opposing efficacy of group III mGlu receptor activators, LSP1-2111 and AMN082, in animal models of positive symptoms of schizophrenia. Psychopharmacology (Berl) 2012;220(3):481–94. doi: 10.1007/s00213-011-2502-2. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES