Abstract
Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS) and abnormalities in the glutamatergic system underlie various CNS disorders. As metabotropic glutamate receptor 3 (mGlu3 receptor) regulates glutamatergic transmission in various brain areas, emerging literature suggests that targeting mGlu3 receptors can be a novel approach to the treatment of psychiatric and neurological disorders. For example, mGlu3 receptor negative allosteric modulators (NAMs) induce rapid antidepressant-like effects in both acute and chronic stress models. Activation of mGlu3 receptors can enhance cognition in the rodents modeling schizophrenia-like pathophysiology. The mGlu3 receptors expressed in the astrocytes induce neuroprotective effects. Although polymorphisms in GRM3 have been shown to be associated with addiction, there is not significant evidence about the efficacy of mGlu3 receptor ligands in rodent models of addiction. Collectively, drugs targeting mGlu3 receptors may provide an alternative approach to fill the unmet clinical need for safer and more efficacious therapeutics for CNS disorders.
Glutamate is the primary excitatory neurotransmitter in the mammalian central nervous system (CNS). It is a key neuromodulator that regulates synaptic plasticity and several brain functions. A growing body of evidence suggests that abnormalities in the glutamatergic neurotransmission play important role in the development of several psychiatric disorders like major depressive disorders (MDD), bipolar disorders, and schizophrenia. Excessive glutamate release can cause neuronal toxicity and has been linked to neurodegenerative disorders, like Parkinson’s disease, amyotrophic lateral sclerosis (ALS), and Huntington’s disease (Olloquequi et al., 2018). Considering the role of glutamate in the CNS, pharmacological interventions targeting the components of the glutamatergic system may provide a therapeutic benefit to patients suffering from these neurological disorders.
In the CNS, glutamate acts through two classes of receptors: ionotropic glutamate (iGlu) receptors and metabotropic glutamate(mGlu) receptors that are found throughout the nervous system (Nakanishi, 1992) (Figure-1). Ionotropic GluRs (iGluRs) are ligand-gated ion channels that mediate fast synaptic transmission. Based upon the ligand binding profile and sequence homology, iGlu receptors are subdivided into three main families: the α-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid (AMPA) receptor, kainate (KAR) receptor and the N-methyl-d-aspartate (NMDA) receptor (Hollmann and Heinemann, 1994) (Figure-1A). A functional iGlu receptor is a tetrameric complex of receptor subunit proteins that can be identical subunits (homomeric receptors) or heterogeneous (heterogeneous receptors) (Traynelis et al., 2010). Each subunit has one large extracellular (EC) amino-terminal domain, one EC ligand-binding domain, three transmembrane domains (TMD), and an intracellular carboxy-terminal domain (CTD) (Traynelis et al., 2010). AMPA receptors are tetrameric complexes composed of subunits GluA1–4, which can be assembled into functional homomeric or heteromeric complexes. All AMPA receptor subunits have a glutamate binding site (Hollmann and Heinemann, 1994). Kainate receptors are comprised of high-affinity subunits GluK1–2 and low-affinity subunits GluK5–7. Low-affinity subunits can form homo-oligomeric receptors, while high-affinity subunits GluK1 and GluK2 must combine with GluK5, GluK6, or GluK7 to form function heteromeric receptors (Herb et al., 1992). NMDA receptors are obligatory heterotetramers comprising two GluN1 with either two GluN2 (GluN2A-GluN2D) or a combination of GluN2 and GluN3 (GluN3A and GluN3B) subunits (Ulbrich and Isacoff, 2008). Glutamate binds to GluN2 subunits (Furukawa et al., 2005; Laube et al., 1997), while glycine is a ligand for GluN1 and GluN3 subunits (Kuryatov et al., 1994). Therefore, GluN1/GluN2 NMDA receptors have glutamate and glycine as obligate coagonists, whereas GluN1/GluN3 NMDA receptors’ pharmacological properties are different from conventional GluN2 containing NMDA receptors and require glycine alone for activation (Chatterton et al., 2002; Smothers and Woodward, 2007).
Figure 1:

(A) Representative structure of an ionotropic glutamate (iGlu) receptor subunit. The iGluRs consist of one extracellular (EC) amino-terminal domain and one ligand binding domain (LBD). The LBD is connected to three transmembrane domains (TMDs). The transmembrane domain is in turn connected to intracellular (IC) C-terminal tail. In iGluRs, ligands bind to the orthosteric site in the LBD and the allosteric modulators bind to the ATD or LBD and channel blockers bind within the ion-channel forming TMD. (B) Representative structure of a metabotropic glutamate (mGlu) receptor subunit. The mGlu receptors have a larger EC domain known as Venus-flytrap domain (VFD) which is connected to seven transmembrane-spanning domains via cysteine rich domain (CRD). The transmembrane domain is in turn connected to intracellular C-terminal tail. The VFD binds glutamate and other orthosteric agonists, whereas the allosteric modulators bind in the TMD. These figures were created with BioRender.com.
The mGlu receptors are class C G-protein coupled receptors (GPCRS) and exist as constitutive dimers (homodimer and heterodimer). Based on G-protein coupling, ligand binding, sequence homology, and function, they are classified into three groups. Group I includes mGlu1 and mGlu5, group II contains mGlu2 and mGlu3, and group III has mGlu4, mGlu6, mGlu7 and mGlu8 receptors (Niswender and Conn, 2010) (Figure-1B). Except for the mGlu6 receptor, which is expressed only in the retina, all others mGlu receptors are widely expressed in the CNS. The mGlu receptors have a large EC N-terminal domain known as the Venus flytrap domain (VFD) linked to the heptahelical TMD via a cysteine-rich domain (CRD; nine cysteine residues). The TMD is connected to intracellular CTD (Pin et al., 2003). The VFD contains the binding site for glutamate, divalent cations and other orthosteric ligands (Kunishima et al., 2000). Ligand binding to the VFD induces conformational changes that are propagated via CRDs to TMD- C-terminal tail and intracellular signaling partners to modulate cellular processes (Niswender and Conn, 2010). Group I mGlu receptors couple to Gq/G11 G proteins and ligand binding activates phospholipase Cβ leading to hydrolysis of phosphoinositide (PI) to diacylglycerol and inositol 1,4,5-trisphosphate (IP3). In contrast, group II and group III receptors are coupled primarily to Gi/o proteins, and activation of these receptors leads to inhibition of adenylyl cyclase. Besides canonical GPCR signaling, mGlu receptors can couple to other signaling pathways like mitogen-activated protein kinase/extracellular receptor kinase, mammalian target of rapamycin /p70 S6 kinase, and phosphatidyl inositol 3-kinase pathways to regulate various aspects of synaptic plasticity (Hou and Klann, 2004; Iacovelli et al., 2002; Li et al., 2007; Page et al., 2006).
The available mGlu receptor-selective compounds are generally allosteric modulators (compounds that modulate the influence of an agonist at a receptor), which “fine-tune” receptor activity and may have lesser side-effects than those associated with orthosteric ligands (ligands that bind to the same binding site as endogenous ligands). Along these lines, a growing body of evidence shows that modulating metabotropic glutamate receptor 3 (mGlu3 receptor) function can exhibit significant therapeutic efficacy in preclinical models of psychiatric disorders. Therefore, in this review, we have summarized clinical and preclinical evidence linking mGlu3 receptors with various psychiatric and neurological disorders (MDD, schizophrenia, addiction disorders and neurodegenerative disorders) (Table-1, 2, 3). Additionally, we have focused on the recent developments of mGlu3 receptor-selective pharmacological tools (negative allosteric modulators) and the possibility of utilizing these ligands for as novel therapeutics.
Table 1.
Summary of the antidepressant- and anxiolytic-like effects of Group II mGlu Receptor ligands
| Compound | Experimental Organisms/strains | Behavioral Tests | Dose and route of Administration | Drug treatment | Behavioral effects | References |
|---|---|---|---|---|---|---|
| MGS0039 | Rats | Forced swim test (FST) | 1, 3 mg/kg, i.p. | 24 and 1 h prior to the FST | Decreased immobility time | Chaki et al., 2004 |
| ICR mice | Tail suspension Test (TST) | 1, 3 mg/kg, i.p. | 60 min prior to the TST | Reduced immobility time | Chaki et al., 2004 | |
| CORT treated ddY mice | FST | 1.0 mg/kg, i.p. | 60 min prior to the FST | Decreased CORT-induced increase in immobility time | Ago et al., 2013 | |
| SDS exposed mice | TST and FST | 1.0 mg/kg, i.p. | 24 and 48 h prior to FST and TST, respectively | Reduced stress-induced increase in immobility time | Dong et al., 2017 | |
| SDS treated mice | Sucrose Preference Test (SPT) | 1.0 mg/kg, i.p. | 3 and 7 days prior to the test | Restored SDS-induced deficits in sucrose preference | Dong et al., 2017 | |
| TP0473292 | CSDS exposed C57BL/6 mice | TST and FST | 3.0 mg/kg; i.p. | 24 and 48 h prior to the TST and FST, respectively | Reduced immobility time | Dong et al., 2022 |
| CSDS exposed C57BL/6 mice | SPT | 3.0 mg/kg; i.p. | 3 and 7 days prior to SPT | Enhanced sucrose preference in stressed mouse | Dong et al., 2022 | |
| LY341495 | ICR mice | TST | 1, 3 mg/kg, i.p. | 60 min prior to the TST | Decreased immobility time | Chaki et al., 2004 |
| Rats | FST | 1, 3 mg/kg, i.p. | 24 and 1 h prior to the FST | Reduced immobility time | Chaki et al., 2004 | |
| C57Bl6/J mice | TST and FST | 1, 3, and 10 mg/kg; i.p. | 60 min prior to the TST | Reduced immobility time | Campo et al., 2011 | |
| Sprague-Dawley rats | SPT | 3 mg/kg, i.p. | 48 h and 10 days prior to SPT | Reversed CUS-induced impairments in sucrose preference | Dwyer at al., 2013 | |
| CORT treated ddY mice | FST | 0.3 mg/kg, i.p. | 60 min prior to the FST | Decreased CORT-induced increase in immobility time | Ago et al., 2013 | |
| Rats | FST | 1, 3 mg/kg; i.p. | 24 h and 30 min prior to the FST | Attenuated increased immobility time | Koike et al., 2013 | |
| Sprague-Dawley rats | FST | 3 mg/kg; i.p. | 24 h prior to the FST | Decreased immobility time | Koike and Chaki, 2014 | |
| C57BL/6J mice | FST | 1 mg/kg; i.p. | 30 min prior to the FST | Reduced immobility time | Fukumoto et al., 2016 | |
| CD-1 mice | TST | 0.3, 1, 3, 10 mg/kg; i.p. | 30 min prior to the TST | Decreased immobility time | Witkin et al., 2016 | |
| Swiss mice | FST | 1, 3, 10 mg/kg; i.p. | 30 min prior to the FST | Reduced immobility time | Witkin et al., 2016 | |
| Sprague-Dawley rats | FST | 0.3, 1, 3, 10 mg/kg; i.p. | 30 min prior to the FST | Decreased immobility time | Witkin et al., 2016 | |
| CD-1 mice | Inescapable shock (IES) | 3 mg/kg; i.p. | 35 min or 24 h prior to IES | Attenuated the development of escape deficits | Highland et al., 2019 | |
| RO4491533 | C57Bl6/J mice | TST | 30, 100 mg/kg; i.p. | 60 min prior to the TST | Decreased immobility time | Campo et al., 2011 |
| C57Bl6/J mice | FST | 3, 10, 30, 100 mg/kg; i.p. | 60 min prior to the FST | Decreased immobility time | Campo et al., 2011 | |
| LY3020371 | Sprague-Dawley rats | FST | 0.3, 1, 3, 10 mg/kg; i.v. | 120 min prior to the FST | Reduced immobility time | Witkin et al., 2017a |
| Swiss mice | FST | 10 mg/kg; i.p. | 30 min prior to the FST | Reduced immobility time | Witkin et al., 2017a | |
| VU650786 | CD-1 mice | TST | 3 mg/kg; i.p. | Acute | Decreased immobility time | Engers et al., 2015 |
| C57BL/6J mice | Progressive ratio test | 30 mg/kg; i.p. | Acute | Prevented motivational deficits induced by acute restraint stress | Joffe et al., 2019 | |
| C57BL/6J mice | TST and FST | 30 mg/kg; i.p. | Acute | Decreased total immobility time | Joffe et al., 2020 | |
| CVS and CORT exposed C57BL/6J mice | SPT | 30 mg/kg; i.p. | Acute | Restored CVS and CORT-induced decrease in sucrose preference. | Joffe et al., 2020 | |
| CVS and CORT exposed C57BL/6J mice | TST and FST | 30 mg/kg; i.p. | Acute | Increased CVS and CORT-induced decrease in latency to immobility | Joffe et al., 2020 | |
| VU6010572 | CD-1 mice | TST | 3 mg/kg; i.p. | Acute | Reduced immobility time | Engers et al., 2017 |
CORT- corticosterone; SDS- social defeat stress; CVS- chronic variable stress; i.p.- intraperitoneal; i.v. intravenous
Table 2.
Summary of the beneficial effects of Group II receptor ligands in behaviors relevant for schizophrenia and substance use disorders
| Compound | Experimental Organisms/strains | Behavioral Tests | Dose and route of Administration | Drug treatment | Behavioral effects | References |
|---|---|---|---|---|---|---|
| ZJ43 | Sprague-Dawley rats | Locomotor test and stereotypy | 150 mg/kg, i.p. | 20 min prior to PCP treatment | Reduced PCP-induced motor activation, stereotypic circling behavior, and head movements | Olszewski et al., 2004 |
| NIH-Swiss mice | Jumping Behavior | 200 mg/kg; i.p. | 5 min prior to MK801 treatment | Reduced MK-801-induced jumping behavior | Olszewski et al., 2008 | |
| DBA/2 mice | Stereotypy | 50, 100, 150 mg/kg, i.p. | 10 min prior to PCP treatment | Reduced PCP-induced stereotypic movements | Olszewski et al., 2008 | |
| DBA/2 mice | Resident-Intruder Assay | 150 mg/kg, i.p. | 10 min prior to PCP treatment | Reversed PCP-induced decrease in escape behavior | Olszewski et al., 2008 | |
| C57BL/6NCr mice | Novel Object recognition (NOR) | 100, 150 mg/kg, i.p. | 20 min prior to MK801 | Rescued MK-801-induced cognitive deficits in NOR | Olszewski et al. 2012b | |
| C57BL/6NCr mice | locomotor test | 100, 150 mg/kg, i.p. | 15 min prior to AMPH treatment | Lowered AMPH-induced hyperlocomotion | Olszewski et al. 2012b | |
| C57BL/6NCr, Grm2 KO and Grm3 KO mice | NOR | 150 mg/kg; i.p. | 30 min prior to the test | Reversed ethanol-induced deficits in NOR in Grm2 KO and WT mice, but not in Grm3 KO | Olszewski et al., 2017 | |
| C57BL/6NCr mice | Locomotor test | 100, 150 mg/kg, i.p. | 15 min prior to ethanol administration | Reduced ethanol-induced motor activation | Olszewski et al., 2017 | |
| C57BL/6NCr mice | Rotarod test | 150 mg/kg, i.p. | 15 min prior to ethanol administration | Reduced ethanol-induced impairments in motor coordination | Olszewski et al., 2017 | |
| 2-PMPA | Long-Evans rats | Cocaine self-administration, intracranial electrical brain-stimulation reward | 100 mg/kg, i.p. | 30 min prior to cocaine injection | Inhibited intravenous cocaine self-administration under progressive-ratio reinforcement conditions, reduced cocaine-induced brain-stimulation reward | Xi et al., 2010a |
| Long-Evans rats | Cocaine self-administration | 30–100 mg/kg, i.p. | 30 min prior to the test session | Inhibited intravenous cocaine self-administration | Xi et al., 2010b | |
| Grm3 KO, Grm2 KO and WT mice | Hyperlocomotion | 100 mg/kg; i.p. | 30 min prior to PCP treatment | Reduced PCP-induced motor activation in Grm2 KO and WT mice | Olszewski et al., 2012a | |
| C57BL/6NCr mice | Locomotor test | 100, 150 mg/kg, i.p. | 15 min prior to AMPH treatment | Reduced AMPH-induced hyperlocomotion | Olszewski et al. 2012b | |
| C57BL/6NCr mice | Hyperlocomotion | 10, 100, 150 mg/kg, i.p. | 15 min prior to PCP treatment | Reduced PCP-induced hyperlocomotion | Olszewski et al. 2012b | |
| C57BL/6NCr mice | Novel Object recognition (NOR) | 10, 100 mg/kg, i.p. | 20 min prior to MK801 treatment | Rescued MK-801-induced cognitive deficits in NOR | Olszewski et al. 2012b | |
| C57BL/6NCr mice | NOR | 100 mg/kg; i.p. | 30 min prior to the test | Reversed ethanol-induced deficits in NOR in Grm2 KO and WT mice, but not in Grm3 KO | Olszewski et al., 2017 | |
| C57BL/6NCr mice | Rotarod test | 50, 100 mg/kg, i.p. | 15 min prior to ethanol administration | Reduced ethanol-induced impairments in motor coordination | Olszewski et al., 2017 | |
| 3xTg mice | NOR | 100 mg/kg; i.p. | 31 min prior to the test | Reversed short-term memory deficits in NOR | Olszewski et al., 2017 | |
| NAAG | Long-Evans rats | Cocaine self-administration, intracranial electrical brain-stimulation reward | 300 μg/side, i.n. | 30 min prior to cocaine injection | inhibited intravenous cocaine self-administration under progressive-ratio reinforcement conditions, reduced cocaine-induced brain-stimulation reward | Xi et al., 2010a |
| VU0477950 | C57BL/6 mice | Fear Extinction | 100 mg/kg; i.p. | 30 minutes prior to the test | Impaired fear extinction learning | Walker et al. 2015 |
| LY2794193 | Sprague-Dawley Rats | Locomotor test | 10, 30 mg/kg, i.p. | 30 minutes prior to PCP treatment | Reduced PCP-induced hyperactivity | Monn et al. 2018 |
| LY379268 | C57BL/6 mice | Trace Fear Conditioning | 3 mg/kg; i.p. | 30 min prior to the test | Restored sub-chronic PCP-induced deficits in acquisition of TFC task | Dogra et al. 2021 |
| BINA | Rhesus macaques | oculomotor delayed response | 0.001 mg/kg; i.m. | 60 min prior to the test | Improvement in delayed response performance | Jin et al. 2018 |
| N-acetylcystine | Sprague Dawley-derived albino rats | Cocaine-induced reinstatement | 60 mg/kg, s.c. | 4 h prior to cocaine administration | Prevented cocaine-induced reinstatement in mGlu2/3 receptor dependent manner | Moran et al. 2005 |
| Sprague-Dawley rats | Electrophysiology | 100 mg/kg, i.p. | 2.5 h prior to electrophysiology | Restored cocaine induced impairments in long termpotentiation and long-term depression in the NAcc core. | Moussawi et al., 2009 |
AMPH- Amphetamine; i.p.- intraperitoneal; s.c. subcutaneous; i.n.- intranasal
Table 3.
Summary of the studies indicating neuroprotective effects of mGlu3 receptor activation.
| Compound | Experimental Organisms/system | Assays | Concentration | Drug treatment | Treatment effects | References |
|---|---|---|---|---|---|---|
| LY379268 | SOD1G93A mice | Rotarod test, western blotting, IHC | 0.5, 1, 5 mg/kg; s.c. | 6 μl/day (chronic activation) | Improved neurologic disability and motor coordination and increased spinal GDNF levels | Battaglia et al., 2015 |
| C57BL/6J Mice | Western blotting | 0.5, 1 mg/kg, i.p | 24 h | Increased GDNF levels in the spinal cord | Battaglia et al., 2015 | |
| C57BL/6J Mice | Western blotting/IHC | 0.5, 1, 5 mg/kg/day; s.c. | 6 μl/day/28 days | Increased GDNF levels in the cervical and thoraco-lumbar spinal cord | Battaglia et al., 2015 | |
| Grm3 and Grm2 KO (CD-1 background) | Western blotting | 1 mg/kg, i.p. | 24 h | Increased GDNF levels in the spinal cord of Grm2 KO and WT mice (not in Grm3 KO) | Battaglia et al., 2015 | |
| Cultured spinal cord astrocytes | Western blotting | 1 μM | 24 h | Increased GDNF levels in the cultured astrocytes from Grm2 KO and WT mice, but not from Grm3 KO | Battaglia et al., 2015 | |
| Mixed cultures | ICC | 1 μM | 24 h | protected motor neurons against kainate toxicity in cultures containing spinal cord astrocytes from WT or Grm2 KO mice | Battaglia et al., 2015 | |
| Cultured astrocytes | sAPPα ELISA, RT-PCR | 0.5, 1, 1, 10 μM | 3 h, 16h, 24h | Induced expression of α-secretase, PPAR-γ and enhances sAPPα release from cultured astrocytes | Durand et al., 2014 | |
| Cultured astrocytes | RT-PCR | 0.1 μM | 3 h | Induced BDNF expression | Durand et al., 2017 | |
| Neurons treated with CM derived from astrocytes treated with LY379268 | TUNEL | medium from LY379268-treated astrocytes protected hippocampal neurons from Aβ-induced cell death | Durand et al., 2017 | |||
| Astrocyte-neuronal coculture | TUNEL | 0.1 μM | 24 h | Promoted Aβ uptake | Durand et al., 2017 | |
| Primary culture Microglia | ICC | 0.1 μM | 24 h | induced phagocytic activity and Aβ uptake by microglia | Durand et al., 2017 | |
| Cultured Astrocytes | ICC | CM derived from LY379268 (0.1 μM, 3h) astrocytes | 24 h | increased Aβ uptake | Durand et al. 2019 | |
| Cultured astrocytes | Flow cytometry, SOD activity, ROS measurement | 0.1 μM | 3 treatments (48 h apart) | Decreased ROS production induced by aging in astrocytes, protected astrocytes from apoptosis induced by aging, prevented reduction in SOD activity caused by aging | Turati et al. 2020 | |
| DCG-IV | Astrocytes and neuronal cultures | TGFβ measurement assay, Microscopy | 1 μM | 10 min | Delayed increase in both intracellular and extracellular levels of TGFβ, decreased neurotoxicity | Bruno et al., 1998 |
ICC- immunocytochemistry; IHC-immunohistochemistry; TUNEL-terminal deoxynucleotidyl transferase dUTP nick end labeling; SOD-Superoxide dismutase; ROS- reactive oxygen species; TGF-β- Transforming Growth Factor-β; sAPP- Soluble amyloid precursor protein; ELISA- enzyme-linked immunosorbent assay; RT-PCR- reverse transcription polymerase chain reaction; i.p.- intraperitoneal; s.c. subcutaneous; CM- conditioned medium
Metabotropic glutamate receptor 3 (mGlu3)
Metabotropic glutamate receptor 3 (mGlu3) is a member of group II mGlu receptors. Group II metabotropic receptors are primarily presynaptic receptors, while mGlu3 receptors are also present at postsynaptic sites and glia (Jin et al., 2018; Woo et al., 2022). Because of its coupling to Gi/o G-proteins, activation of the mGlu3 receptor inhibits adenylyl cyclase activity leading to a decrease in the production of cyclic adenosine monophosphate (cAMP) and activity of cAMP-dependent protein kinases (Figure-2). Besides canonical G-protein mediated signaling, the mGlu3 receptors can signal through other non-canonical pathways and can potentiate signaling mediated by Gq- coupled receptors. For example, activation of neuronal mGlu3 receptors supports mGlu5 receptor (a Gq-coupled receptor) induced signaling and contributes to the stimulation of mGlu5 receptor-mediated PI hydrolysis in early postnatal life (Di Menna et al., 2018; Schoepp et al., 1996). This neuronal mGlu3–5 receptor interaction has potential implications for enhancing cognition and sculpturing the influence of the mGlu5 receptor on neurotoxicity (Di Menna et al., 2018; Dogra et al., 2021). Additionally, mGlu3 receptor activation in the cortical pyramidal neurons can potentiate somatic Ca2+ mobilization induced by mGlu5 receptor activation (Di Menna et al., 2018). Interestingly, mGlu3 receptor-induced long-term depression (LTD) in the prefrontal cortex (PFC) requires activation of endogenous mGlu5 receptors, but activation of mGlu3 receptors does not potentiate mGlu5-induced LTD in the hippocampus (Dogra et al., 2021) indicating selective effects of mGlu3 receptors on mGlu5 receptor-mediated signaling.
Figure 2:

Schematic depicting mGlu3 receptor-dependent functions in pyramidal neurons and astrocytes. Activation of mGlu3 receptors in the postsynaptic neurons mobilizes endocannabinoids (2-arachindonylglycerol; 2-AG) via a mGlu5 receptor-dependent signaling. Mobilization of 2-AG decreases the release of GABA from local interneurons leading to decreased inhibitory tone on pyramidal neurons and altered threshold for synaptic plasticity via N-methyl-D-aspartate (NMDA) receptors. Facilitating synaptic pathway through this mechanism enhances hippocampal-dependent cognition and can rescue cognitive deficits in rodents modeling schizophrenia-like pathophysiology. Activation of astrocytic mGlu3 receptors can promote the release of brain derived neurotrophic factor (BDNF) that has neuroprotective effects. Activation of mGlu3 receptor in the astrocytes also enhances the synthesis and release of anti-inflammatory cytokine transforming growth factor-β (TGF β) from astrocyte that protects neurons from excitotoxic insults. Astrocytic mGlu3 receptor activation also enhances glial derived neurotropic factor (GDNF) levels indicating potential role of enhancing mGlu3 receptor function in slowing neurodegeneration. AMPA- α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; GABA- γ-Aminobutyric acid; LTP- long-term potentiation; EAAT1- Excitatory amino acid transporter 1; EAAT2- Excitatory amino acid transporter 1. This figure was created with BioRender.com.
While mGlu3–5 receptor interaction has been shown to increase the extent of long-term potentiation (LTP) at SC-CA1 synapse (Figure-2), coincident activation of mGlu3 receptors with β-adrenergic receptors (βAR) in the astrocytes in the CA1 of hippocampus can counteract the effects of βAR activation on the synaptic plasticity (Walker et al., 2017). Studies employing Grm3 (the gene coding for mGlu3 receptor) knockout (KO) mice reveal that coactivation of mGlu3 with βARs induces an increase in cAMP accumulation and increased adenosine release. The adenosine in turn acts on A1 adenosine receptors present in excitatory SC terminals that can restrain the effects of βAR on synaptic plasticity at the SC-CA1 synapse (Walker et al., 2017). These findings suggest that activation of the glial mGlu3 receptor can modulate the effect of excessive βAR activation (for example, during intense stress) and may present a protective mechanism to inhibit neuronal injury. Moreover, activation of mGlu3 receptors expressed on the astrocytes increases the expression of glutamate aspartate transporters (GLAST) and glutamate transporter-1 (GLT-1), and promotes glutamate uptake from the synapse, and thereby, limiting glutamate spread across the synapse (Aronica et al., 2003). These findings point towards an exciting possibility of targeting mGlu3 receptors to regulate EC glutamate levels during pathological conditions and restricting excitotoxic brain damage. Further, it will be exciting to determine whether these mechanisms exist at different synapses in other brain areas.
The expression of mGlu3 receptors is found in several brain areas implicated in the regulation of cognition and stress-related behaviors (Jin et al., 2018; McOmish et al., 2016; Woo et al., 2022). In the rodent brain, strong mGlu3 receptor mRNA expression is observed in the cortex, basolateral amygdala (BLA), reticular thalamic nucleus, striatal matrix and nucleus accumbens (NAcc) (McOmish et al., 2016; Woo et al., 2022). The mGlu3 receptors are also expressed in the CA1 of the hippocampus, the dentate gyrus (granular layer), claustrum and superficial gray superior colliculus (Dogra et al., 2021; McOmish et al., 2016). To note, The levels of mGlu3 receptors have been reported to decrease with age, which might be related to cognitive decline seen with advanced ages (Hernandez et al., 2018), and the impact of age-related downregulation of mGlu3 receptor is worth further investigation.
In the CNS, mGlu3 receptors are activated by glutamate as well as N-acetylaspartylglutamate (NAAG) (Ghose et al., 1997; Neale, 2011; Wroblewska et al., 1997). NAAG is a peptide neurotransmitter and is co-released with glutamate. It is catabolized by glutamate carboxypeptidase II (NAAG peptidases). Therefore, compounds that block the catabolism of synaptic NAAG (NAAG peptidase inhibitors) can regulate mGlu3 receptor-dependent functions in the CNS (Neale and Olszewski, 2019; Zhong et al., 2006). These classes of compounds are increasingly in use to investigate the significance of NAAG and the role of mGlu3 receptors in various disease conditions. Besides NAAG modulators, the discovery of mGlu3 receptor-selective negative allosteric modulators (NAMs) has further advanced the field (Engers et al., 2017; Engers et al., 2015). Using these selective pharmacological tools along with the receptor-selective conditional KOs, several exciting discoveries have facilitated the mGlu3 receptor research and have advanced our understanding of how mGlu3 receptors regulate glutamatergic transmission in different brain areas during psychiatric disorders like depression and schizophrenia (Dogra et al., 2021; Joffe et al., 2019; Joffe et al., 2020; Walker et al., 2015).
Negative allosteric modulators of mGlu3 receptor have potential utility as antidepressants
Major Depressive Disorders (MDD) are one of the most widespread health concerns and a leading cause of disability worldwide. A growing body of evidence indicates that altered glutamatergic neurotransmission may contribute to the pathophysiology of MDD, and establish the glutamatergic system as an interesting target for designing novel therapeutics for MDD (Dogra and Conn, 2021; Duman et al., 2019). Specifically, rapid and sustained antidepressant effects of ketamine in patients suffering from treatment-refractory depression (TRD) (Duman, 2018) have prompted researchers to increase focus on agents regulating glutamatergic signaling in the CNS as potential novel antidepressants.
Along these lines, mGlu2/3 receptor antagonists and NAMs have been shown to induce rapid and sustained antidepressant-like responses in both acute and chronic models of depression (Table-1) (Ago et al., 2013; Campo et al., 2011; Chaki et al., 2004; Dong et al., 2022; Dong et al., 2017; Dwyer et al., 2013; Fukumoto et al., 2016; Joffe et al., 2020; Koike et al., 2013; Podkowa et al., 2015; Witkin et al., 2017a). Further, mGlu2/3 antagonists reduced the learned helplessness induced by chronic corticosterone (CORT) treatment (Ago et al., 2013; Koike et al., 2013), and reduced immobility time in the tail suspension test (TST) in CD-1 mice (Witkin et al., 2016). Moreover, pharmacological blockade of mGlu2/3 receptors using LY341495 during stress prevented the development of inescapable shock and CORT-induced escape deficits (Highland et al., 2019). It is important to note that effective dosages of conventional antidepressants like imipramine, desipramine, and fluoxetine do not show efficacy in the chronic CORT model (Ago et al., 2013; Iijima et al., 2010), while a single injection of ketamine can induce antidepressant-like effects in this model (Koike et al., 2011, 2013). Similarly, the CD-1 strain of mice shows a reduced sensitivity to acute administration of AD therapeutics like fluvoxamine, amitriptyline and imipramine in TST (van der Heyden et al., 1987). Therefore, both chronic CORT treatment and CD-1 mice can be considered preclinical models of TRD and the efficacy of mGlu2/3 receptor antagonists in these models advise the potential utility of mGlu2/3 antagonists for TRD.
Interestingly, mGlu2/3 receptor antagonists exert antidepressant-like effects comparable to ketamine in several preclinical behavioral tests and may share some neural mechanisms with ketamine to induce antidepressant effects (Dong et al., 2017; Fukumoto et al., 2016; Koike and Chaki, 2014; Witkin et al., 2017a; Witkin et al., 2016). Additionally, administration of mGlu2/3 receptor antagonist does not recapitulate ketamine-associated adverse effects like abuse liability, cognitive deficits, motor abnormalities and neurotoxicity issues (Witkin et al., 2017b) indicating that mGlu2/3 receptor antagonists possess desired safety profile that could support development of drug candidates for further studies in patients. Based on this exciting preclinical literature, TP0473292 (TS-161), a prodrug of mGlu2/3 antagonist TP0178894, has been launched into clinical trials for TRD (Watanabe et al., 2022). The results from phase-1, double-blind, placebo-controlled clinical trials in healthy objects revealed that TP0473292 is safe and well-tolerated and is a promising clinical candidate for the evaluation of efficacy in patients with depression.
Similar to the mGlu2/3 receptor antagonists, the mGlu3 receptor-selective NAMs can induce antidepressant- and anxiolytic-like effects in various preclinical models. For instance, mGlu3 receptor-selective NAM, VU650786, dose-dependently inhibited the number of marbles buried by CD-1 mice in the marble-burying test (Engers et al., 2015) indicating potential anxiolytic-like effects of mGlu3 receptor inhibition. Additionally, VU650786 reduced the immobility time in the acute model of behavioral despair (forced swim test (FST) and tail suspension test (TST)) (Engers et al., 2015; Joffe et al., 2020). Excitingly, acute treatment with VU650786 rescued anhedonia (as accessed by an increase in sucrose preference) as well as behavioral despair measured as the increase in latency to become immobile in FST and TST in chronic CORT and chronic variable stress models of depression (Joffe et al., 2020). These findings indicate that single administration of mGlu3 receptor NAMs can rapidly reverse anhedonia and behavioral despair in preclinical chronic stress models and suggest that mGlu3 receptor NAM display an efficacy profile comparable to mGlu2/3 receptor antagonists and other rapid-acting antidepressants.
The mGlu3 receptor NAM, VU650786, was further modified using a reductionist optimization approach and the resulting novel mGlu3 receptor NAM, VU6010576, also showed efficacy for reducing immobility time comparable to ketamine (30 mg/kg; i.p.) in TST in CD-1 mice (Engers et al., 2017). Recently published mechanistic studies revealed that systemic treatment with a mGlu3 receptor NAM, VU650786, activates unique pyramidal cell ensembles in the PFC (Joffe et al., 2020). It also attenuates the mGlu2/3 receptor agonist, LY379268, induced long-term depression at thalamocortical synapses. Interestingly, inhibition of thalamocortical synapses using a chemogenetic approach blocked the effects of mGlu3 receptor NAM in TST and FST indicating that mGlu3 receptor NAM may act through these synapses to induce antidepressant-like effects. Moreover, the mGlu3 receptor NAM can prevent acute restraint stress-induced impairments in plasticity at amygdalo-cortico synapses and motivational deficits (Joffe et al., 2019), which further strengthens the utility of mGlu3 receptor NAM for treating stress-related psychiatric disorders where motivational deficits are often observed. Taken together, the above-mentioned literature indicates that selective modulators of mGlu3 receptors may provide novel approaches to address mood disorders and indicate their potential utility for disorders like schizophrenia where depression a is comorbid condition.
Activation of mGlu3 receptors can rescue deficits associated with schizophrenia
Schizophrenia is a debilitating mental disorder that involves the dysfunction of multiple brain areas. A growing body of evidence reveals that changes in glutamatergic neurotransmission may contribute to the pathophysiology of schizophrenia and the agents reversing these changes could provide symptomatic relief to schizophrenia patients. Along these lines, mGlu3 receptors have emerged as a potential therapeutic target for treating schizophrenia-associated behavioral and physiological deficits (Dogra and Conn, 2022). Genome-wide association studies (GWAS) have identified genetic variants in GRM3 as potential risk factors for developing schizophrenia (Fujii et al., 2003; Jablensky et al., 2011; Mounce et al., 2014; Saini et al., 2017). Further, the risk alleles in GRM3 have been found to be associated with schizophrenia-related endophenotypes (Chang et al., 2015; Egan et al., 2004; Kinoshita et al., 2015). Such as, single nucleotide polymorphisms (SNPs) in GRM3 were associated with cognitive impairments (Chang et al., 2015; Egan et al., 2004; Jablensky et al., 2011) and lesser activation of the PFC during cognitive task performance in schizophrenia (Kinoshita et al., 2015). Also, schizophrenia risk SNP in GRM3 was associated with lower levels of N-acetylaspartate (NAA) and mRNA of glial glutamate transporter excitatory amino acid transporter 2 (EAAT2) in the PFC (Egan et al., 2004; Marenco et al., 2006). As reduced levels of NAA and EAAT2 protein have been related to impaired executive function and working memory in schizophrenia patients (Galinska et al., 2007; Spangaro et al., 2012), it is possible that the GRM3 genotype might alter glutamatergic neurotransmission in the PFC and performance in the PFC-dependent cognitive tasks, and thereby increase the risk for schizophrenia.
Interestingly, patients having GRM3 variant rs1468412 (which is associated with altered dysfunction of the glutamatergic system (Xia et al., 2012)) have been shown to have more adverse changes in the performance of working memory tasks after antipsychotic treatment (Bishop et al., 2015). Also, GRM3 polymorphisms (rs274622, rs6465084 and rs724226) can predict the improvement of negative symptoms following antipsychotic treatment (Bishop et al., 2005; Bishop et al., 2015; Fijal et al., 2009). These association studies suggest that genetic variants in GRM3 can influence the responsiveness to antipsychotic treatment and indicate the possibility of utilizing this knowledge to identify a subgroup of patients susceptible to experiencing adverse effects on cognition following antipsychotic medication. Further, polymorphisms in GRM3 have also been linked to the white matter integrity in schizophrenia patients (Mounce et al., 2014), which is considered one of the predictors of response to antipsychotics (Kraguljac et al., 2021; Reis Marques et al., 2014). These studies provide a piece of evidence that GRM3 is a susceptibility locus for schizophrenia and have persuaded researchers to probe if activating or modulating mGlu3 receptor function can be used to reverse schizophrenia-like deficits in preclinical setups.
Based on various theories proposed to explain the underlying pathology of schizophrenia, several pharmacological disease models are currently in use preclinically. For example, as the hyperdopaminergic state is the core pathophysiology of schizophrenia, initial animal models were developed using pharmacological modifications like amphetamine treatment to induce psychotic-like changes (Jones et al., 2011). Similarly, based on the clinical evidence that administration of NMDA antagonists like phencyclidine (PCP) and ketamine in health individuals induce symptoms like those seen in schizophrenia (Hu et al., 2015), NMDA antagonists are extensively used to mimic schizophrenia-like deficits. Interestingly, initial studies have shown beneficial effects of NAAG peptidase inhibitors in reducing motor abnormalities induced by NMDA antagonists dizocilpine (MK801) and PCP (Olszewski et al., 2004; Olszewski et al., 2008). PCP-induced stereotypic movements (cycling behavior, head movements) and negative symptoms were also reduced by the NAAG peptidase inhibitor, ZJ43 (Olszewski et al., 2004; Olszewski et al., 2008). Moreover, ZL43 could inhibit amphetamine-induced hyperactivity when administered before or after amphetamine treatment (Olszewski et al., 2012b). Similarly, another inhibitor, 2-PMPA, blocked PCP- and amphetamine-induced motor activation (Olszewski et al., 2012b). Also, both ZL43 and 2-PMPA blocked PCP-induced increase in the glutamate levels in the medial PFC (mPFC) and NAcc (Zuo et al., 2012), and significantly blocked MK801-induced impairments in the novel object recognition when administered before the acquisition trials (Olszewski et al., 2012b). All these studies indicate the possibility that elevation of synaptic NAAG levels may represent a novel therapeutic approach for treating all symptom domains in schizophrenia.
Interestingly, the above-mentioned effects of NAAG inhibitors were blocked by the mGlu2/3 receptor antagonist, LY341495 revealing an essential role of the mGlu2/3 receptors in mediating beneficial effects of NAAG peptidase inhibitors in rodents mimicking schizophrenia-like symptoms. Further, studies employing receptor-specific KO mice have shown that the effects of NAAG peptidase inhibitor, 2-PMPA, on PCP-induced motor activation were absent in Grm3 KO mice (Olszewski et al., 2012a) suggesting an essential role of mGlu3 receptors in rescuing schizophrenia-like symptoms induced by NMDA antagonist. These findings are supported by studies in the Grm3 KO mice revealing abnormalities in the expression of interneuron-related genes and neuronal synchrony in the PFC (Imbriglio et al., 2019) similar to those seen in schizophrenia patients (Anderson et al., 2020; Kaar et al., 2019; Kang et al., 2018; Spencer et al., 2003; Toker et al., 2018). These findings raise the possibility that insults to the mGlu3 receptors may induce cortical neuronal abnormalities and can predispose individuals to schizophrenia. Mechanistic studies conducted by Jin et al. 2017 revealed that activation of mGlu3 using endogenous agonist NAAG enhances the firing of dorsolateral PFC (DLPFC) Delay cells during a spatial working memory task performance in Rhesus macaques (Jin et al., 2018). This study stipulates that mGlu3 receptors are positioned to strengthen the high-order cognitive circuits in DLPFC and support a large body of the above-mentioned literature linking GRM3 polymorphisms with impairments in the PFC function.
Recently, the development of pharmacological probes has helped researchers to evaluate the role of mGlu3 receptors in the regulation of CNS physiology and related behaviors (Table-2). For example, subcutaneous injections of mGlu3 receptor agonist, LY2794193 has been shown to decrease PCP-induced hyperlocomotion in a dose-dependent manner (Monn et al., 2018). Using receptor-selective KOs and NAMs, it has been shown that activation of mGlu3 receptors induces LTD in the PFC (Joffe et al., 2019; Walker et al., 2015). To note, this LTD is expressed post-synaptically and is dependent upon intracellular calcium (Ca2+) mobilization induced by activation of mGlu3 receptors in layer V pyramidal neurons (Walker et al., 2015). Further mechanistic studies revealed that co-activation of another class of mGlu receptor, mGlu5, is essential for mGlu3 receptor-induced LTD in the PFC (Di Menna et al., 2018). Recently, a similar partnership between mGlu3 and mGlu5 receptors has also been observed in the CA1 region of the hippocampus. In the CA1 pyramidal neurons of the hippocampus, activation of mGlu3 receptors has been shown to induce metaplastic changes to enhance synaptic plasticity at the SC-CA1 synapse (Figure-2) (Dogra et al., 2021). This metaplasticity is dependent upon the mGlu5 and endocannabinoid receptor-mediated disinhibitory mechanisms (Dogra et al., 2021). Excitingly, activation of mGlu3 receptors has been shown to improve fear extinction (Walker et al., 2015), and promotes associative learning in trace fear conditioning tasks (Dogra et al., 2021) indicating that activation of mGlu3 receptors may rescue cognitive deficits in psychiatric disorders. Since these functions of mGlu3 receptors are dependent upon the activation of mGlu5 receptors, it will be exciting to evaluate the efficacy of mGlu3 receptor activation on other testes of cognition modulated by mGlu5 receptors (Dogra and Conn, 2022). Moreover, activation of the mGlu3 receptors has been shown to be neuroprotective (Caraci et al., 2011). Therefore, the pharmacological tools that selectively activate the mGlu3 receptor may have the potential to reduce the risk of causing neurotoxic insults while treating cognitive deficits associated with schizophrenia.
Activation of astrocytic mGlu3 receptors is neuroprotective:
Alterations in the components of glutamatergic signaling have also been reported in neurodegenerative diseases. For example, in Alzheimer’s disease (AD), decreased expression and function of astrocytic glutamate transporters have been observed (Kirvell et al., 2006; Masliah et al., 1996; Scott et al., 2011). Additionally, amyloid-β (Aβ), which is accumulated in the AD brain and plays a key role in the disease progression (Nicoll and Weller, 2003), has been shown to induce inhibitory effects on glutamate uptake by astrocytes (Matos et al., 2008; Parpura-Gill et al., 1997) that can change the availability of extrasynaptic glutamate. Considering the role of astrocytes in regulating glutamate homeostasis, significant attention has been shifted to evaluating the effects of mGlu receptor ligands on the astrocytes (Table-3). Astrocytes primarily express mGlu5 and mGlu3 receptors (Balazs et al., 1997; Condorelli et al., 1997; Nakahara et al., 1997; Schools and Kimelberg, 1999; Woo et al., 2022), whereas no expression of mGlu2 receptors and other mGlu receptors have been found on these cells.
Interestingly, activation of the mGlu3 receptors increases the release of neuroprotective soluble amyloid precursor protein from the cultured astrocytes by inducing α-secretase expression which stimulates the removal of Aβ plaques (Durand et al., 2014; Durand et al., 2019). However, this is not the only neuroprotective mechanism modulated by mGlu3 receptors in AD (Figure-2). Activation of astrocytic mGlu3 receptors can also increase brain-derived neurotropic factor (BDNF) release and induce clearance of amyloids from the EC space (Durand et al., 2017). Moreover, mGlu3 receptor stimulation in the astrocytes induces anti-oxidant effects and prevents Aβ- induced hippocampal neuronal death (Durand et al., 2017; Turati et al., 2020). Excitingly, activation of astrocytic mGlu3 receptors enhances the formation and release of anti-inflammatory cytokine, transforming growth factor-β (TGF β), which can protect neurons from excitotoxic insults (Bruno et al., 1998). These studies raise an exciting possibility of activating/potentiating mGlu3 receptor function as a therapeutic strategy to slow down Aβ-related neurodegeneration in AD. Along these lines, decreased expression of mGlu3 receptors has been reported in the hippocampal astrocytes of the PDAPP-J20 mouse model of AD (Durand et al., 2014) which might contribute to the development of AD-like pathology in these mice. Similar changes in the mGlu3 receptor expression have been reported in the aged astrocytes (Turati et al., 2020), which can be increased after treatment with group II agonist, LY379268.
Besides astrocytes, decreased Grm3 expression has also been reported in the developing microglia following inflammatory challenges (Zinni et al., 2021). Further, genetic deletion of Grm3 mimicked the pro-inflammatory phenotype observed in the inflammatory challenged microglial. Similarly, genetic loss of mGlu3 receptors has been shown to amplify ischemic brain damage in mice (Mastroiacovo et al., 2021). To note, increased infarct size as well as increased expression of proinflammatory genes (Il-1β, TNF-1α, COX-2, IL-6, and CD86) in the peri-infarct area have been reported (Mastroiacovo et al., 2021). These studies suggest that selective activation of mGlu3 receptors may be used as a therapeutic intervention in brain injuries marked by a high level of inflammation. Additionally, acute as well as chronic treatment with mGlu2/3 receptor agonist, LY379268, enhanced glial derived neurotropic factor (GDNF) and GLT-1 levels in the spinal cord of WT as well as Grm2 (gene coding for mGlu2 receptor) KO mice (Battaglia et al., 2015). The effects of LY379268 were absent in Grm3 KO mice, indicating the role of mGlu3 receptor activation in LY379268-induced enhancement of GDNF in the spinal cord. Excitingly, chronic treatment with LY379268 rescued the spinal cord motor neurons in SOD1G93A mice (a mouse model for amyotrophic lateral sclerosis, ALS) (Battaglia et al., 2015). To note, the activation of mGlu3 receptors protects cultured spinal cord motor neurons via increased GDNF production (Battaglia et al., 2015), indicating the potential role of enhancing mGlu3 receptor function in slowing neuronal degeneration in ALS patients.
mGlu3 receptor may be involved in the pathology of substance use disorders
A tremendous amount of literature suggests a crucial role of glutamatergic signaling in the development and maintenance of behavioral and plasticity changes associated with addiction. Because of the ability to regulate glutamate release in the brain areas implicated in addiction, the mGlu3 receptors are one of the molecular targets of drug-induced plasticity changes in the brain. An association between minor alleles of two GRM3 SNPs (rs274618 and rs274622) and an increased risk of heroin dependence has been revealed in a Chinese population (Jia et al., 2014). Of note, the carrier addicts of these alleles had a relatively shorter duration for the transition from the first use to dependence as compared to the homozygous for the major alleles. These studies indicate that genetic variants in GRM3 may confer an increased risk of developing heroin dependence. Interestingly, GRM3 is one of the top candidate genes that is shared between alcoholism and other psychiatric disorders (bipolar disorders, schizophrenia and stress behaviors) (Levey et al., 2014). Sequence variants in GRM3 (rs6465084 and rs148754219) are associated with alcohol dependence in male subjects (O’Brien et al., 2014; Xia et al., 2014). A mouse genome wide-microarray analysis from the PFC has revealed Grm3 as one of the hub genes in the acute ethanol responsive networks in the mice (Wolen et al., 2012). Further, a transcriptome analysis in the ethanol naive rat strains that are genetically selected to prefer or avoid ethanol has shown a differential baseline regulation of Grm3 in the PFC (Worst et al., 2005). This could be a mechanism responsible for altered neurotransmitter release following exposure to substance of abuse.
Similarly, decreased expression of mGlu3 receptor, but not mGlu2 receptor, mRNA has also been observed in the dentate gyrus of rats administered with ethanol for 2 months (Simonyi et al., 2004). Changes in the expression of Grm3 were also observed in the postmortem hippocampus of subjects exposed to chronic alcohol (Enoch et al., 2014). Given the role of the hippocampus in the processing of contextual cues that are considered important in drug reinforcement behaviors and craving, and cognitive processes (Luo et al., 2011; Volkow et al., 2004), the above mentioned findings point toward the potential role/ association of mGlu3 receptors in cognitive deficits and behavioral changes associated with chronic substance use. Along these lines, NAAG peptidase inhibitors have been shown to reverse short-term novel object recognition memory deficits induced by acute ethanol administration in WT but not Grm3 KO mice (Olszewski et al., 2017). Additional studies revealed the absence of ethanol-induced conditioned place preference in Grm3 KO mice (Lainiola et al., 2019) suggesting a role of mGlu3 receptors in the neuroplastic changes required for ethanol conditioned place preference (CPP). On the contrary, Grm3 KO mice displayed increased place preference for methamphetamine in a conditioned place preference (CPP) task and increased methamphetamine toxicity in the mPFC and the NAcc (Busceti et al., 2021). Based on that it is conceivable that potentiating mGlu3 receptors might confer a therapeutic benefit for treating methamphetamine addiction and the associated brain damage (Table-2).
Decreased levels of GRM3 have been observed in the postmortem hippocampus of cocaine addicts (Enoch et al., 2014). In addition, repeated cocaine exposure impairs mGlu2/3 receptor function in the PFC and the NAcc (Huang et al., 2007; Xi et al., 2002). And withdrawal from cocaine self-administration attenuated the expression of LTP in the NAcc core subregion following stimulation of the PFC (Moussawi et al., 2009). These deficits in plasticity were restored by treatment with N-acetylcysteine (NAC) and the effects of NAC on LTP were blocked by administration of mGlu2/3 antagonist, LY341495. These results along with the previous findings that NAC inhibits reinstatement of drug seeking via activation of mGlu2/3 receptors (Moran et al., 2005) propose an essential role of mGlu2/3 receptors in mediating cocaine-induced drug-seeking. Further, systemic administration of NAAG peptidase inhibitor, 2-PMPA, inhibited intravenous cocaine self-administration and cocaine-induced reinstatement of drug-seeking behavior (Xi et al., 2010a; Xi et al., 2010b). Similarly, microinjections of 2-PMPA or NAAG into the NAcc also inhibited cocaine-induced reinstatement and indicate that activation of mGlu3 receptors may mediate these affects. Nevertheless, further studies utilizing receptor-selective ligands, and receptor and cell-type KOs are warranted to evaluate the role of mGlu3 receptors in cocaine-induced plasticity and behavioral changes.
Concluding remarks and future directions
Emerging evidence suggests that alterations in the components of glutamatergic signaling play an important role in the pathophysiology of multiple CNS disorders (Dong et al., 2009; Javitt, 2004; Li et al., 2018; Ribeiro et al., 2017). Pharmacologically targeting of iGlu receptors is beneficial in several CNS disorders, but these manipulations produce several adverse effects including motor deficits, psychosis-like symptoms and neurotoxicity. A tremendous number of preclinical studies have shown that manipulating mGlu receptors using selective allosteric modulators has a potential for the treatment of CNS disorders (Dogra and Conn, 2021, 2022; Maksymetz et al., 2017). Among these, the mGlu3 receptors have emerged as promising agents for the treatment of several psychiatric (Dogra and Conn, 2021, 2022) and neurodegenerative disorders (Caraci et al., 2012; Durand et al., 2013) (Table-1, 2, 3). For example, mGlu3 receptor NAM induces rapid and sustained antidepressant-like effects in both acute and chronic models of depression (Joffe et al., 2020). The mGlu3 receptor selective NAMs are also efficacious in the rodent models of treatment-resistant depression. Further, mGlu3 receptor NAM can prevent motivational deficits induced by acute restraint stress (Joffe et al., 2019). In the future, it will be exciting to evaluate the effects of mGlu3 receptor NAMs on motivation in the chronic models of depression. As mGlu2/3 receptor antagonists lack side-effects associated with the NMDA receptor antagonists, the mGlu3 receptor NAMs hold great potential as next-generation safer and more efficacious antidepressants.
The progress in the mGlu3 receptor research is impeded by the lack of mGlu3 receptor-selective pharmacological ligands (agonists and positive allosteric modulators). In that scenario, human gene association studies have played an important role in defining the role of mGlu3 receptors in CNS disorders. For example, polymorphisms in GRM3 are associated with an increased risk of developing schizophrenia and cognitive impairments associated with schizophrenia (Egan et al., 2004; Fujii et al., 2003; Kinoshita et al., 2015; Saini et al., 2017). Similarly, sequence variants in the GRM3 are linked with an increased risk of developing heroin and alcohol dependence (Jia et al., 2014; O’Brien et al., 2014; Xia et al., 2014). These gene association studies have bolstered research efforts to test if modulation of mGlu3 receptor function in selective brain areas can rescue schizophrenia-like and addiction-related pathophysiological changes. Along these lines, recently, it has been reported that selective activation of neuronal mGlu3 receptors can rescue schizophrenia-like cognitive deficits in rodents (Dogra et al., 2021). Moreover, exciting evidence suggests that activation of astrocytic mGlu3 receptors can induce neuroprotective effects by promoting the release of BDNF, TGF-β and GDNF, and by modulating glutamate transporters (Figure-2) (Battaglia et al., 2015; Durand et al., 2017). Activation of mGlu3 receptors promotes cognition (Dogra et al., 2021), therefore, it will be exciting to evaluate the cognition enhancing effects of mGlu3 receptors in various models of neurodegenerative diseases. Earlier work in these models has been done using mGlu2/3 receptor agonist and/or mGlu2- and mGlu3-selective KOs. Considering the previously reported compensatory changes in these whole-body KOs (Lyon et al., 2008), testing the effects of mGlu3 receptor activation using cell-specific conditional KOs can provide important insights into the beneficial effects of mGlu3 receptors. Besides, considering the sex differences in the CNS disorders, understanding the molecular basis of the sex-differences as a biological variable can also provide useful insights to full harness the therapeutic potential of these receptors. Additionally, the findings that mGlu3 receptors can shape the influence of mGlu5 receptor activation on neurotoxic insults (Di Menna et al., 2018) might open new avenues of research in the field of neurodegenerative disorders.
In conclusion, extensive preclinical and clinical studies illuminate the potential of targeting mGlu3 receptors to develop safer and more efficacious drugs for the treatment of psychiatric and neurodegenerative disorders. Further, continued research efforts utilizing mGlu3 receptor-selective compounds will likely provide unparalleled insights into the mechanism of CNS disorders.
Highlights.
Negative allosteric modulators of mGlu3 receptors induce antidepressant-like effects in preclinical models of depression.
Agonists of mGlu2/3 receptors induce antipsychotic-like and procognitive effects in rodents.
Activation of mGlu3 receptors enhances cognition and can provide a novel approach for treating cognitive deficits in schizophrenia.
Activation of astrocytic mGlu3 receptors induces neuroprotective effects.
Acknowledgements
This work was supported by NIH grants R01MH062646 (P.J.C.) and R01NS031373 (P.J.C.).
Declaration of interests
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. and J.P. have no competing interests to declare.
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 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.
Bibliography:
- 1.Ago Y, Yano K, Araki R, Hiramatsu N, Kita Y, Kawasaki T, Onoe H, Chaki S, Nakazato A, Hashimoto H, Baba A, Takuma K, Matsuda T, 2013. Metabotropic glutamate 2/3 receptor antagonists improve behavioral and prefrontal dopaminergic alterations in the chronic corticosterone-induced depression model in mice. Neuropharmacology 65, 29–38. [DOI] [PubMed] [Google Scholar]
- 2.Anderson KM, Collins MA, Chin R, Ge T, Rosenberg MD, Holmes AJ, 2020. Transcriptional and imaging-genetic association of cortical interneurons, brain function, and schizophrenia risk. Nat Commun 11(1), 2889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Aronica E, Gorter JA, Ijlst-Keizers H, Rozemuller AJ, Yankaya B, Leenstra S, Troost D, 2003. Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J Neurosci 17(10), 2106–2118. [DOI] [PubMed] [Google Scholar]
- 4.Balazs R, Miller S, Romano C, de Vries A, Chun Y, Cotman CW, 1997. Metabotropic glutamate receptor mGluR5 in astrocytes: pharmacological properties and agonist regulation. J Neurochem 69(1), 151–163. [DOI] [PubMed] [Google Scholar]
- 5.Battaglia G, Riozzi B, Bucci D, Di Menna L, Molinaro G, Pallottino S, Nicoletti F, Bruno V, 2015. Activation of mGlu3 metabotropic glutamate receptors enhances GDNF and GLT-1 formation in the spinal cord and rescues motor neurons in the SOD-1 mouse model of amyotrophic lateral sclerosis. Neurobiol Dis 74, 126–136. [DOI] [PubMed] [Google Scholar]
- 6.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(2–3), 253–260. [DOI] [PubMed] [Google Scholar]
- 7.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(1), 145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, Nicoletti F, 1998. Neuroprotection by glial metabotropic glutamate receptors is mediated by transforming growth factor-beta. J Neurosci 18(23), 9594–9600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Busceti CL, Ginerete RP, Di Menna L, D’Errico G, Cisani F, Di Pietro P, Imbriglio T, Bruno V, Battaglia G, Fornai F, Monn JA, Pittaluga A, Nicoletti F, 2021. Behavioural and biochemical responses to methamphetamine are differentially regulated by mGlu2 and mGlu3 metabotropic glutamate receptors in male mice. Neuropharmacology 196, 108692. [DOI] [PubMed] [Google Scholar]
- 10.Campo B, Kalinichev M, Lambeng N, El Yacoubi M, Royer-Urios I, Schneider M, Legrand C, Parron D, Girard F, Bessif A, Poli S, Vaugeois JM, Le Poul E, Celanire S, 2011. Characterization of an mGluR2/3 negative allosteric modulator in rodent models of depression. J Neurogenet 25(4), 152–166. [DOI] [PubMed] [Google Scholar]
- 11.Caraci F, Battaglia G, Sortino MA, Spampinato S, Molinaro G, Copani A, Nicoletti F, Bruno V, 2012. Metabotropic glutamate receptors in neurodegeneration/neuroprotection: still a hot topic? Neurochem Int 61(4), 559–565. [DOI] [PubMed] [Google Scholar]
- 12.Caraci F, Molinaro G, Battaglia G, Giuffrida ML, Riozzi B, Traficante A, Bruno V, Cannella M, Merlo S, Wang X, Heinz BA, Nisenbaum ES, Britton TC, Drago F, Sortino MA, Copani A, Nicoletti F, 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(3), 618–626. [DOI] [PubMed] [Google Scholar]
- 13.Chaki S, Yoshikawa R, Hirota S, Shimazaki T, Maeda M, Kawashima N, Yoshimizu T, Yasuhara A, Sakagami K, Okuyama S, Nakanishi S, Nakazato A, 2004. MGS0039: a potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology 46(4), 457–467. [DOI] [PubMed] [Google Scholar]
- 14.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(3), 1043–1046. [DOI] [PubMed] [Google Scholar]
- 15.Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova M, Shin Y, Cui J, Tu S, Sevarino KA, Nakanishi N, Tong G, Lipton SA, Zhang D, 2002. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 415(6873), 793–798. [DOI] [PubMed] [Google Scholar]
- 16.Condorelli DF, Dell’Albani P, Corsaro M, Giuffrida R, Caruso A, Trovato Salinaro A, Spinella F, Nicoletti F, Albanese V, Giuffrida Stella AM, 1997. Metabotropic glutamate receptor expression in cultured rat astrocytes and human gliomas. Neurochem Res 22(9), 1127–1133. [DOI] [PubMed] [Google Scholar]
- 17.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, 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]
- 18.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]
- 19.Dogra S, Conn PJ, 2022. Metabotropic Glutamate Receptors As Emerging Targets for the Treatment of Schizophrenia. Mol Pharmacol 101(5), 275–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Dong C, Tian Z, Fujita Y, Fujita A, Hino N, Iijima M, Hashimoto K, 2022. Antidepressant-like actions of the mGlu2/3 receptor antagonist TP0178894 in the chronic social defeat stress model: Comparison with escitalopram. Pharmacol Biochem Behav 212, 173316. [DOI] [PubMed] [Google Scholar]
- 22.Dong C, Zhang JC, Yao W, Ren Q, Ma M, Yang C, Chaki S, Hashimoto K, 2017. Rapid and Sustained Antidepressant Action of the mGlu2/3 Receptor Antagonist MGS0039 in the Social Defeat Stress Model: Comparison with Ketamine. Int J Neuropsychopharmacol 20(3), 228–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dong XX, Wang Y, Qin ZH, 2009. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin 30(4), 379–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Duman RS, 2018. Ketamine and rapid-acting antidepressants: a new era in the battle against depression and suicide. F1000Res 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Duman RS, Sanacora G, Krystal JH, 2019. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 102(1), 75–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Durand D, Carniglia L, Beauquis J, Caruso C, Saravia F, Lasaga M, 2014. Astroglial mGlu3 receptors promote alpha-secretase-mediated amyloid precursor protein cleavage. Neuropharmacology 79, 180–189. [DOI] [PubMed] [Google Scholar]
- 27.Durand D, Carniglia L, Caruso C, Lasaga M, 2013. mGlu3 receptor and astrocytes: partners in neuroprotection. Neuropharmacology 66, 1–11. [DOI] [PubMed] [Google Scholar]
- 28.Durand D, Carniglia L, Turati J, Ramirez D, Saba J, Caruso C, Lasaga M, 2017. Amyloid-beta neurotoxicity and clearance are both regulated by glial group II metabotropic glutamate receptors. Neuropharmacology 123, 274–286. [DOI] [PubMed] [Google Scholar]
- 29.Durand D, Turati J, Rudi MJ, Ramirez D, Saba J, Caruso C, Carniglia L, von Bernhardi R, Lasaga M, 2019. Unraveling the beta-amyloid clearance by astrocytes: Involvement of metabotropic glutamate receptor 3, sAPPalpha, and class-A scavenger receptor. Neurochem Int 131, 104547. [DOI] [PubMed] [Google Scholar]
- 30.Dwyer JM, Lepack AE, Duman RS, 2013. mGluR2/3 blockade produces rapid and long-lasting reversal of anhedonia caused by chronic stress exposure. J Mol Psychiatry 1(1), 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Egan MF, Straub RE, Goldberg TE, Yakub I, Callicott JH, Hariri AR, Mattay VS, Bertolino A, Hyde TM, Shannon-Weickert C, Akil M, Crook J, Vakkalanka RK, Balkissoon R, Gibbs RA, Kleinman JE, Weinberger DR, 2004. Variation in GRM3 affects cognition, prefrontal glutamate, and risk for schizophrenia. Proc Natl Acad Sci U S A 101(34), 12604–12609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Engers JL, Bollinger KA, Weiner RL, Rodriguez AL, Long MF, Breiner MM, Chang S, Bollinger SR, Bubser M, Jones CK, Morrison RD, Bridges TM, Blobaum AL, Niswender CM, Conn PJ, Emmitte KA, Lindsley CW, 2017. Design and Synthesis of N-Aryl Phenoxyethoxy Pyridinones as Highly Selective and CNS Penetrant mGlu3 NAMs. ACS Med Chem Lett 8(9), 925–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Engers JL, Rodriguez AL, Konkol LC, Morrison RD, Thompson AD, Byers FW, Blobaum AL, Chang S, Venable DF, Loch MT, Niswender CM, Daniels JS, Jones CK, Conn PJ, Lindsley CW, Emmitte KA, 2015. Discovery of a Selective and CNS Penetrant Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 3 with Antidepressant and Anxiolytic Activity in Rodents. J Med Chem 58(18), 7485–7500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Enoch MA, Rosser AA, Zhou Z, Mash DC, Yuan Q, Goldman D, 2014. Expression of glutamatergic genes in healthy humans across 16 brain regions; altered expression in the hippocampus after chronic exposure to alcohol or cocaine. Genes Brain Behav 13(8), 758–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.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(5), 311–318. [DOI] [PubMed] [Google Scholar]
- 36.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(2), 71–76. [DOI] [PubMed] [Google Scholar]
- 37.Fukumoto K, Iijima M, Chaki S, 2016. The Antidepressant Effects of an mGlu2/3 Receptor Antagonist and Ketamine Require AMPA Receptor Stimulation in the mPFC and Subsequent Activation of the 5-HT Neurons in the DRN. Neuropsychopharmacology 41(4), 1046–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Furukawa H, Singh SK, Mancusso R, Gouaux E, 2005. Subunit arrangement and function in NMDA receptors. Nature 438(7065), 185–192. [DOI] [PubMed] [Google Scholar]
- 39.Galinska B, Szulc A, Tarasow E, Kubas B, Dzienis W, Siergiejczyk L, Czernikiewicz A, Walecki J, 2007. Relationship between frontal N-acetylaspartate and cognitive deficits in first-episode schizophrenia. Med Sci Monit 13 Suppl 1, 11–16. [PubMed] [Google Scholar]
- 40.Ghose S, Wroblewska B, Corsi L, Grayson DR, De Blas AL, Vicini S, Neale JH, 1997. N-acetylaspartylglutamate stimulates metabotropic glutamate receptor 3 to regulate expression of the GABA(A) alpha6 subunit in cerebellar granule cells. J Neurochem 69(6), 2326–2335. [DOI] [PubMed] [Google Scholar]
- 41.Herb A, Burnashev N, Werner P, Sakmann B, Wisden W, Seeburg PH, 1992. The KA-2 subunit of excitatory amino acid receptors shows widespread expression in brain and forms ion channels with distantly related subunits. Neuron 8(4), 775–785. [DOI] [PubMed] [Google Scholar]
- 42.Hernandez CM, McQuail JA, Schwabe MR, Burke SN, Setlow B, Bizon JL, 2018. Age-Related Declines in Prefrontal Cortical Expression of Metabotropic Glutamate Receptors that Support Working Memory. eNeuro 5(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Highland JN, Zanos P, Georgiou P, Gould TD, 2019. Group II metabotropic glutamate receptor blockade promotes stress resilience in mice. Neuropsychopharmacology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hollmann M, Heinemann S, 1994. Cloned glutamate receptors. Annu Rev Neurosci 17, 31–108. [DOI] [PubMed] [Google Scholar]
- 45.Hou L, Klann E, 2004. Activation of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway is required for metabotropic glutamate receptor-dependent long-term depression. J Neurosci 24(28), 6352–6361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.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]
- 47.Huang CC, Yang PC, Lin HJ, Hsu KS, 2007. Repeated cocaine administration impairs group II metabotropic glutamate receptor-mediated long-term depression in rat medial prefrontal cortex. J Neurosci 27(11), 2958–2968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Iacovelli L, Bruno V, Salvatore L, Melchiorri D, Gradini R, Caricasole A, Barletta E, De Blasi A, Nicoletti F, 2002. Native group-III metabotropic glutamate receptors are coupled to the mitogen-activated protein kinase/phosphatidylinositol-3-kinase pathways. J Neurochem 82(2), 216–223. [DOI] [PubMed] [Google Scholar]
- 49.Iijima M, Ito A, Kurosu S, Chaki S, 2010. Pharmacological characterization of repeated corticosterone injection-induced depression model in rats. Brain Res 1359, 75–80. [DOI] [PubMed] [Google Scholar]
- 50.Imbriglio T, Verhaeghe R, Martinello K, Pascarelli MT, Chece G, Bucci D, Notartomaso S, Quattromani M, Mascio G, Scalabri F, Simeone A, Maccari S, Del Percio C, Wieloch T, Fucile S, Babiloni C, Battaglia G, Limatola C, Nicoletti F, Cannella M, 2019. Developmental abnormalities in cortical GABAergic system in mice lacking mGlu3 metabotropic glutamate receptors. FASEB J 33(12), 14204–14220. [DOI] [PubMed] [Google Scholar]
- 51.Jablensky A, Morar B, Wiltshire S, Carter K, Dragovic M, Badcock JC, Chandler D, Peters K, Kalaydjieva L, 2011. Polymorphisms associated with normal memory variation also affect memory impairment in schizophrenia. Genes Brain Behav 10(4), 410–417. [DOI] [PubMed] [Google Scholar]
- 52.Javitt DC, 2004. Glutamate as a therapeutic target in psychiatric disorders. Mol Psychiatry 9(11), 984–997, 979. [DOI] [PubMed] [Google Scholar]
- 53.Jia W, Zhang R, Wu B, Dai ZX, Zhu YS, Li PP, Zhu F, 2014. Metabotropic glutamate receptor 3 is associated with heroin dependence but not depression or schizophrenia in a Chinese population. PLoS One 9(1), e87247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.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(3), 974–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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(6), 916–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Joffe ME, Santiago CI, Oliver KH, Maksymetz J, Harris NA, Engers JL, Lindsley CW, Winder DG, Conn PJ, 2020. mGlu2 and mGlu3 Negative Allosteric Modulators Divergently Enhance Thalamocortical Transmission and Exert Rapid Antidepressant-like Effects. Neuron 105(1), 46–59 e43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Jones CA, Watson DJ, Fone KC, 2011. Animal models of schizophrenia. Br J Pharmacol 164(4), 1162–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kaar SJ, Angelescu I, Marques TR, Howes OD, 2019. Pre-frontal parvalbumin interneurons in schizophrenia: a meta-analysis of post-mortem studies. J Neural Transm (Vienna) 126(12), 1637–1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kang SS, MacDonald AW 3rd, Chafee MV, Im CH, Bernat EM, Davenport ND, Sponheim SR, 2018. Abnormal cortical neural synchrony during working memory in schizophrenia. Clin Neurophysiol 129(1), 210–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kinoshita A, Takizawa R, Koike S, Satomura Y, Kawasaki S, Kawakubo Y, Marumo K, Tochigi M, Sasaki T, Nishimura Y, Kasai K, 2015. Effect of metabotropic glutamate receptor-3 variants on prefrontal brain activity in schizophrenia: An imaging genetics study using multi-channel near-infrared spectroscopy. Prog Neuropsychopharmacol Biol Psychiatry 62, 14–21. [DOI] [PubMed] [Google Scholar]
- 61.Kirvell SL, Esiri M, Francis PT, 2006. Down-regulation of vesicular glutamate transporters precedes cell loss and pathology in Alzheimer’s disease. J Neurochem 98(3), 939–950. [DOI] [PubMed] [Google Scholar]
- 62.Koike H, Chaki S, 2014. Requirement of AMPA receptor stimulation for the sustained antidepressant activity of ketamine and LY341495 during the forced swim test in rats. Behav Brain Res 271, 111–115. [DOI] [PubMed] [Google Scholar]
- 63.Koike H, Iijima M, Chaki S, 2011. Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression. Behav Brain Res 224(1), 107–111. [DOI] [PubMed] [Google Scholar]
- 64.Koike H, Iijima M, Chaki S, 2013. Effects of ketamine and LY341495 on the depressive-like behavior of repeated corticosterone-injected rats. Pharmacol Biochem Behav 107, 20–23. [DOI] [PubMed] [Google Scholar]
- 65.Kraguljac NV, Anthony T, Morgan CJ, Jindal RD, Burger MS, Lahti AC, 2021. White matter integrity, duration of untreated psychosis, and antipsychotic treatment response in medication-naive first-episode psychosis patients. Mol Psychiatry 26(9), 5347–5356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi S, Jingami H, Morikawa K, 2000. Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature 407(6807), 971–977. [DOI] [PubMed] [Google Scholar]
- 67.Kuryatov A, Laube B, Betz H, Kuhse J, 1994. Mutational analysis of the glycine-binding site of the NMDA receptor: structural similarity with bacterial amino acid-binding proteins. Neuron 12(6), 1291–1300. [DOI] [PubMed] [Google Scholar]
- 68.Laube B, Hirai H, Sturgess M, Betz H, Kuhse J, 1997. Molecular determinants of agonist discrimination by NMDA receptor subunits: analysis of the glutamate binding site on the NR2B subunit. Neuron 18(3), 493–503. [DOI] [PubMed] [Google Scholar]
- 69.Levey DF, Le-Niculescu H, Frank J, Ayalew M, Jain N, Kirlin B, Learman R, Winiger E, Rodd Z, Shekhar A, Schork N, Kiefer F, Wodarz N, Muller-Myhsok B, Dahmen N, Consortium G, Nothen M, Sherva R, Farrer L, Smith AH, Kranzler HR, Rietschel M, Gelernter J, Niculescu AB, 2014. Genetic risk prediction and neurobiological understanding of alcoholism. Transl Psychiatry 4, e391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li CT, Yang KC, Lin WC, 2018. Glutamatergic Dysfunction and Glutamatergic Compounds for Major Psychiatric Disorders: Evidence From Clinical Neuroimaging Studies. Front Psychiatry 9, 767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Li XM, Li CC, Yu SS, Chen JT, Sabapathy K, Ruan DY, 2007. JNK1 contributes to metabotropic glutamate receptor-dependent long-term depression and short-term synaptic plasticity in the mice area hippocampal CA1. Eur J Neurosci 25(2), 391–396. [DOI] [PubMed] [Google Scholar]
- 72.Luo AH, Tahsili-Fahadan P, Wise RA, Lupica CR, Aston-Jones G, 2011. Linking context with reward: a functional circuit from hippocampal CA3 to ventral tegmental area. Science 333(6040), 353–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lyon L, Kew JN, Corti C, Harrison PJ, Burnet PW, 2008. Altered hippocampal expression of glutamate receptors and transporters in GRM2 and GRM3 knockout mice. Synapse 62(11), 842–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Maksymetz J, Moran SP, Conn PJ, 2017. Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Mol Brain 10(1), 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Marenco S, Steele SU, Egan MF, Goldberg TE, Straub RE, Sharrief AZ, Weinberger DR, 2006. Effect of metabotropic glutamate receptor 3 genotype on N-acetylaspartate measures in the dorsolateral prefrontal cortex. Am J Psychiatry 163(4), 740–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Masliah E, Alford M, DeTeresa R, Mallory M, Hansen L, 1996. Deficient glutamate transport is associated with neurodegeneration in Alzheimer’s disease. Ann Neurol 40(5), 759–766. [DOI] [PubMed] [Google Scholar]
- 77.Mastroiacovo F, Zinni M, Mascio G, Bruno V, Battaglia G, Pansiot J, Imbriglio T, Mairesse J, Baud O, Nicoletti F, 2021. Genetic Deletion of mGlu3 Metabotropic Glutamate Receptors Amplifies Ischemic Brain Damage and Associated Neuroinflammation in Mice. Front Neurol 12, 668877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Matos M, Augusto E, Oliveira CR, Agostinho P, 2008. Amyloid-beta peptide decreases glutamate uptake in cultured astrocytes: involvement of oxidative stress and mitogen-activated protein kinase cascades. Neuroscience 156(4), 898–910. [DOI] [PubMed] [Google Scholar]
- 79.McOmish CE, Demireva EY, Gingrich JA, 2016. Developmental expression of mGlu2 and mGlu3 in the mouse brain. Gene Expr Patterns 22(2), 46–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Monn JA, Henry SS, Massey SM, Clawson DK, Chen Q, Diseroad BA, Bhardwaj RM, Atwell S, Lu F, Wang J, Russell M, Heinz BA, Wang XS, Carter JH, Getman BG, Adragni K, Broad LM, Sanger HE, Ursu D, Catlow JT, Swanson S, Johnson BG, Shaw DB, McKinzie DL, Hao J, 2018. Synthesis and Pharmacological Characterization of C4beta-Amide-Substituted 2-Aminobicyclo[3.1.0]hexane-2,6-dicarboxylates. Identification of (1 S,2 S,4 S,5 R,6 S)-2-Amino-4-[(3-methoxybenzoyl)amino]bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid (LY2794193), a Highly Potent and Selective mGlu3 Receptor Agonist. J Med Chem 61(6), 2303–2328. [DOI] [PubMed] [Google Scholar]
- 81.Moran MM, McFarland K, Melendez RI, Kalivas PW, Seamans JK, 2005. Cystine/glutamate exchange regulates metabotropic glutamate receptor presynaptic inhibition of excitatory transmission and vulnerability to cocaine seeking. J Neurosci 25(27), 6389–6393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mounce J, Luo L, Caprihan A, Liu J, Perrone-Bizzozero NI, Calhoun VD, 2014. Association of GRM3 polymorphism with white matter integrity in schizophrenia. Schizophr Res 155(1–3), 8–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, Kalivas PW, 2009. N-Acetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci 12(2), 182–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Nakahara K, Okada M, Nakanishi S, 1997. The metabotropic glutamate receptor mGluR5 induces calcium oscillations in cultured astrocytes via protein kinase C phosphorylation. J Neurochem 69(4), 1467–1475. [DOI] [PubMed] [Google Scholar]
- 85.Nakanishi S, 1992. Molecular diversity of glutamate receptors and implications for brain function. Science 258(5082), 597–603. [DOI] [PubMed] [Google Scholar]
- 86.Neale JH, 2011. N-acetylaspartylglutamate is an agonist at mGluR(3) in vivo and in vitro. J Neurochem 119(5), 891–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.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]
- 88.Nicoll JA, Weller RO, 2003. A new role for astrocytes: beta-amyloid homeostasis and degradation. Trends Mol Med 9(7), 281–282. [DOI] [PubMed] [Google Scholar]
- 89.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]
- 90.O’Brien NL, Way MJ, Kandaswamy R, Fiorentino A, Sharp SI, Quadri G, Alex J, Anjorin A, Ball D, Cherian R, Dar K, Gormez A, Guerrini I, Heydtmann M, Hillman A, Lankappa S, Lydall G, O’Kane A, Patel S, Quested D, Smith I, Thomson AD, Bass NJ, Morgan MY, Curtis D, McQuillin A, 2014. The functional GRM3 Kozak sequence variant rs148754219 affects the risk of schizophrenia and alcohol dependence as well as bipolar disorder. Psychiatr Genet 24(6), 277–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Olloquequi J, Cornejo-Cordova E, Verdaguer E, Soriano FX, Binvignat O, Auladell C, Camins A, 2018. Excitotoxicity in the pathogenesis of neurological and psychiatric disorders: Therapeutic implications. J Psychopharmacol 32(3), 265–275. [DOI] [PubMed] [Google Scholar]
- 92.Olszewski RT, Bukhari N, Zhou J, Kozikowski AP, Wroblewski JT, Shamimi-Noori S, Wroblewska B, Bzdega T, Vicini S, Barton FB, Neale JH, 2004. NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR. J Neurochem 89(4), 876–885. [DOI] [PubMed] [Google Scholar]
- 93.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(1–3), 160–161. [DOI] [PubMed] [Google Scholar]
- 94.Olszewski RT, Janczura KJ, Ball SR, Madore JC, Lavin KM, Lee JC, Lee MJ, Der EK, Hark TJ, Farago PR, Profaci CP, Bzdega T, Neale JH, 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]
- 95.Olszewski RT, Janczura KJ, Bzdega T, Der EK, Venzor F, O’Rourke B, Hark TJ, Craddock KE, Balasubramanian S, Moussa C, Neale JH, 2017. NAAG Peptidase Inhibitors Act via mGluR3: Animal Models of Memory, Alzheimer’s, and Ethanol Intoxication. Neurochem Res 42(9), 2646–2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Olszewski RT, Wegorzewska MM, Monteiro AC, Krolikowski KA, Zhou J, Kozikowski AP, Long K, Mastropaolo J, Deutsch SI, Neale JH, 2008. Phencyclidine and dizocilpine induced behaviors reduced by N-acetylaspartylglutamate peptidase inhibition via metabotropic glutamate receptors. Biol Psychiatry 63(1), 86–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Page G, Khidir FA, Pain S, Barrier L, Fauconneau B, Guillard O, Piriou A, Hugon J, 2006. Group I metabotropic glutamate receptors activate the p70S6 kinase via both mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK 1/2) signaling pathways in rat striatal and hippocampal synaptoneurosomes. Neurochem Int 49(4), 413–421. [DOI] [PubMed] [Google Scholar]
- 98.Parpura-Gill A, Beitz D, Uemura E, 1997. The inhibitory effects of beta-amyloid on glutamate and glucose uptakes by cultured astrocytes. Brain Res 754(1–2), 65–71. [DOI] [PubMed] [Google Scholar]
- 99.Pin JP, Galvez T, Prezeau L, 2003. Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacol Ther 98(3), 325–354. [DOI] [PubMed] [Google Scholar]
- 100.Podkowa K, Rzezniczek S, Marciniak M, Acher F, Pilc A, Palucha-Poniewiera A, 2015. A novel mGlu4 selective agonist LSP4–2022 increases behavioral despair in mouse models of antidepressant action. Neuropharmacology 97, 338–345. [DOI] [PubMed] [Google Scholar]
- 101.Reis Marques T, Taylor H, Chaddock C, Dell’acqua F, Handley R, Reinders AA, Mondelli V, Bonaccorso S, Diforti M, Simmons A, David AS, Murray RM, Pariante CM, Kapur S, Dazzan P, 2014. White matter integrity as a predictor of response to treatment in first episode psychosis. Brain 137(Pt 1), 172–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ribeiro FM, Vieira LB, Pires RG, Olmo RP, Ferguson SS, 2017. Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 115, 179–191. [DOI] [PubMed] [Google Scholar]
- 103.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(8), e1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schoepp DD, Salhoff CR, Wright RA, Johnson BG, Burnett JP, Mayne NG, Belagaje R, Wu S, Monn JA, 1996. The novel metabotropic glutamate receptor agonist 2R,4R-APDC potentiates stimulation of phosphoinositide hydrolysis in the rat hippocampus by 3,5-dihydroxyphenylglycine: evidence for a synergistic interaction between group 1 and group 2 receptors. Neuropharmacology 35(12), 1661–1672. [DOI] [PubMed] [Google Scholar]
- 105.Schools GP, Kimelberg HK, 1999. mGluR3 and mGluR5 are the predominant metabotropic glutamate receptor mRNAs expressed in hippocampal astrocytes acutely isolated from young rats. J Neurosci Res 58(4), 533–543. [DOI] [PubMed] [Google Scholar]
- 106.Scott HA, Gebhardt FM, Mitrovic AD, Vandenberg RJ, Dodd PR, 2011. Glutamate transporter variants reduce glutamate uptake in Alzheimer’s disease. Neurobiol Aging 32(3), 553 e551–511. [DOI] [PubMed] [Google Scholar]
- 107.Simonyi A, Christian MR, Sun AY, Sun GY, 2004. Chronic ethanol-induced subtype- and subregion-specific decrease in the mRNA expression of metabotropic glutamate receptors in rat hippocampus. Alcohol Clin Exp Res 28(9), 1419–1423. [DOI] [PubMed] [Google Scholar]
- 108.Smothers CT, Woodward JJ, 2007. Pharmacological characterization of glycine-activated currents in HEK 293 cells expressing N-methyl-D-aspartate NR1 and NR3 subunits. J Pharmacol Exp Ther 322(2), 739–748. [DOI] [PubMed] [Google Scholar]
- 109.Spangaro M, Bosia M, Zanoletti A, Bechi M, Cocchi F, Pirovano A, Lorenzi C, Bramanti P, Benedetti F, Smeraldi E, Cavallaro R, 2012. Cognitive dysfunction and glutamate reuptake: effect of EAAT2 polymorphism in schizophrenia. Neurosci Lett 522(2), 151–155. [DOI] [PubMed] [Google Scholar]
- 110.Spencer KM, Nestor PG, Niznikiewicz MA, Salisbury DF, Shenton ME, McCarley RW, 2003. Abnormal neural synchrony in schizophrenia. J Neurosci 23(19), 7407–7411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Toker L, Mancarci BO, Tripathy S, Pavlidis P, 2018. Transcriptomic Evidence for Alterations in Astrocytes and Parvalbumin Interneurons in Subjects With Bipolar Disorder and Schizophrenia. Biol Psychiatry 84(11), 787–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R, 2010. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 62(3), 405–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Turati J, Ramirez D, Carniglia L, Saba J, Caruso C, Quarleri J, Durand D, Lasaga M, 2020. Antioxidant and neuroprotective effects of mGlu3 receptor activation on astrocytes aged in vitro. Neurochem Int 140, 104837. [DOI] [PubMed] [Google Scholar]
- 114.Ulbrich MH, Isacoff EY, 2008. Rules of engagement for NMDA receptor subunits. Proc Natl Acad Sci U S A 105(37), 14163–14168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.van der Heyden JA, Molewijk E, Olivier B, 1987. Strain differences in response to drugs in the tail suspension test for antidepressant activity. Psychopharmacology (Berl) 92(1), 127–130. [DOI] [PubMed] [Google Scholar]
- 116.Volkow ND, Fowler JS, Wang GJ, 2004. The addicted human brain viewed in the light of imaging studies: brain circuits and treatment strategies. Neuropharmacology 47 Suppl 1, 3–13. [DOI] [PubMed] [Google Scholar]
- 117.Walker AG, Sheffler DJ, Lewis AS, Dickerson JW, Foster DJ, Senter RK, Moehle MS, Lv X, Stansley BJ, Xiang Z, Rook JM, Emmitte KA, Lindsley CW, Conn PJ, 2017. Co-Activation of Metabotropic Glutamate Receptor 3 and Beta-Adrenergic Receptors Modulates Cyclic-AMP and Long-Term Potentiation, and Disrupts Memory Reconsolidation. Neuropsychopharmacology 42(13), 2553–2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.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 U S A 112(4), 1196–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Watanabe M, Marcy B, Hiroki A, Watase H, Kinoshita K, Iijima M, Marumo T, Zarate CA, Chaki S, 2022. Evaluation of the Safety, Tolerability, and Pharmacokinetic Profiles of TP0473292 (TS-161), A Prodrug of a Novel Orthosteric mGlu2/3 Receptor Antagonist TP0178894, in Healthy Subjects and Its Antidepressant-Like Effects in Rodents. Int J Neuropsychopharmacol 25(2), 106–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Witkin JM, Mitchell SN, Wafford KA, Carter G, Gilmour G, Li J, Eastwood BJ, Overshiner C, Li X, Rorick-Kehn L, Rasmussen K, Anderson WH, Nikolayev A, Tolstikov VV, Kuo MS, Catlow JT, Li R, Smith SC, Mitch CH, Ornstein PL, Swanson S, Monn JA, 2017a. Comparative Effects of LY3020371, a Potent and Selective Metabotropic Glutamate (mGlu) 2/3 Receptor Antagonist, and Ketamine, a Noncompetitive N-Methyl-d-Aspartate Receptor Antagonist in Rodents: Evidence Supporting the Use of mGlu2/3 Antagonists, for the Treatment of Depression. J Pharmacol Exp Ther 361(1), 68–86. [DOI] [PubMed] [Google Scholar]
- 121.Witkin JM, Monn JA, Li J, Johnson B, McKinzie DL, Wang XS, Heinz BA, Li R, Ornstein PL, Smith SC, Mitch CH, Calligaro DO, Swanson S, Allen D, Phillips K, Gilmour G, 2017b. Preclinical predictors that the orthosteric mGlu2/3 receptor antagonist LY3020371 will not engender ketamine-associated neurotoxic, motor, cognitive, subjective, or abuse-liability-related effects. Pharmacol Biochem Behav 155, 43–55. [DOI] [PubMed] [Google Scholar]
- 122.Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, Carter G, Johnson B, Rasmussen K, Rorick-Kehn LM, 2016. The Rapidly Acting Antidepressant Ketamine and the mGlu2/3 Receptor Antagonist LY341495 Rapidly Engage Dopaminergic Mood Circuits. J Pharmacol Exp Ther 358(1), 71–82. [DOI] [PubMed] [Google Scholar]
- 123.Wolen AR, Phillips CA, Langston MA, Putman AH, Vorster PJ, Bruce NA, York TP, Williams RW, Miles MF, 2012. Genetic dissection of acute ethanol responsive gene networks in prefrontal cortex: functional and mechanistic implications. PLoS One 7(4), e33575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Woo E, Datta D, Arnsten AFT, 2022. Glutamate Metabotropic Receptor Type 3 (mGlu3) Localization in the Rat Prelimbic Medial Prefrontal Cortex. Front Neuroanat 16, 849937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Worst TJ, Tan JC, Robertson DJ, Freeman WM, Hyytia P, Kiianmaa K, Vrana KE, 2005. Transcriptome analysis of frontal cortex in alcohol-preferring and nonpreferring rats. J Neurosci Res 80(4), 529–538. [DOI] [PubMed] [Google Scholar]
- 126.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(1), 174–181. [DOI] [PubMed] [Google Scholar]
- 127.Xi ZX, Kiyatkin M, Li X, Peng XQ, Wiggins A, Spiller K, Li J, Gardner EL, 2010a. N-acetylaspartylglutamate (NAAG) inhibits intravenous cocaine self-administration and cocaine-enhanced brain-stimulation reward in rats. Neuropharmacology 58(1), 304–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xi ZX, Li X, Peng XQ, Li J, Chun L, Gardner EL, Thomas AG, Slusher BS, Ashby CR Jr., 2010b. Inhibition of NAALADase by 2-PMPA attenuates cocaine-induced relapse in rats: a NAAG-mGluR2/3-mediated mechanism. J Neurochem 112(2), 564–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Xi ZX, Ramamoorthy S, Baker DA, Shen H, Samuvel DJ, Kalivas PW, 2002. Modulation of group II metabotropic glutamate receptor signaling by chronic cocaine. J Pharmacol Exp Ther 303(2), 608–615. [DOI] [PubMed] [Google Scholar]
- 130.Xia Y, Ma D, Hu J, Tang C, Wu Z, Liu L, Xin F, 2012. Effect of metabotropic glutamate receptor 3 genotype on N-acetylaspartate levels and neurocognition in non-smoking, active alcoholics. Behav Brain Funct 8, 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Xia Y, Wu Z, Ma D, Tang C, Liu L, Xin F, Zhu D, Hu J, 2014. Association of single-nucleotide polymorphisms in a metabotropic glutamate receptor GRM3 gene subunit to alcohol-dependent male subjects. Alcohol Alcohol 49(3), 256–260. [DOI] [PubMed] [Google Scholar]
- 132.Zhong C, Zhao X, Van KC, Bzdega T, Smyth A, Zhou J, Kozikowski AP, Jiang J, O’Connor WT, Berman RF, Neale JH, Lyeth BG, 2006. NAAG peptidase inhibitor increases dialysate NAAG and reduces glutamate, aspartate and GABA levels in the dorsal hippocampus following fluid percussion injury in the rat. J Neurochem 97(4), 1015–1025. [DOI] [PubMed] [Google Scholar]
- 133.Zinni M, Mairesse J, Pansiot J, Fazio F, Iacovelli L, Antenucci N, Orlando R, Nicoletti F, Vaiman D, Baud O, 2021. mGlu3 receptor regulates microglial cell reactivity in neonatal rats. J Neuroinflammation 18(1), 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.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(26), 21773–21782. [DOI] [PMC free article] [PubMed] [Google Scholar]
