Skip to main content
Pharmacological Reviews logoLink to Pharmacological Reviews
. 2022 Jul;74(3):630–661. doi: 10.1124/pharmrev.121.000540

Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease

Deborah J Luessen , P Jeffrey Conn
Editor: Lynette Daws1
PMCID: PMC9553119  PMID: 35710132

Abstract

Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders.

Significance Statement

Allosteric modulation of metabotropic glutamate (mGlu) receptors represents a promising therapeutic strategy to normalize dysregulated cellular physiology associated with neuropsychiatric disease. This review summarizes preclinical and clinical studies using mGlu receptor allosteric modulators as experimental tools and potential therapeutic approaches for the treatment of neuropsychiatric diseases, including schizophrenia, stress, and substance use disorders.

I. Introduction

Glutamate is the primary excitatory neurotransmitter within the central nervous system (CNS). Glutamate modulates cell excitability and synaptic transmission through actions on glutamate receptors, including ionotropic glutamate and metabotropic glutamate (mGlu) receptors. Ionotropic glutamate receptors, which include amino-3-hydroxy-5-methyl-isoxazolepropionic acid, N-methyl-d-aspartate (NMDA), and kainate receptors, are ligand-gated ion channels that mediate fast excitatory synaptic transmission (Traynelis et al., 2010). mGlu receptors are members of the G-protein-coupled receptor superfamily and can be classified into three distinct groups based on their sequence homology, G-protein coupling, and ligand selectivity (Conn and Pin, 1997). The mGlu receptor subtypes are differentially expressed pre- and postsynaptically throughout the CNS and are located on both neurons and glial cells.

A. Metabotropic Glutamate Receptors: Structure and Signal Transduction

1. Structural Components of Metabotropic Glutamate Receptors

mGlu receptors feature a large extracellular N-terminal domain, coined the Venus flytrap domain (VFD), which contains the orthosteric glutamate binding site and is critical for homo- and heterodimerization of these receptors (Yin and Niswender, 2014). Extensive evidence shows that VFDs form dimers, which can exist in three main states: open-open, open-closed, and closed-closed. Antagonist binding stabilizes the open-open (inactive) conformation, whereas ligand binding induces open-closed and closed-closed conformations. Distinct residues that are associated with closure of the VFD strongly contribute to functional switching of ligands from antagonists to agonists (Bessis et al., 2002; Jingami et al., 2003; Niswender and Conn, 2010), highlighting the important role of these domains and their respective orientations for receptor activation. Importantly, a number of conserved residues interact directly with glutamate as well as divalent cations, such as calcium or magnesium, which have the ability to activate the receptor (Kubo et al., 1998; Kunishima et al., 2000; Francesconi and Duvoisin, 2004). Ligand binding results in conformational changes, originating from the VFD via cysteine-rich domains (CRDs) to the heptahelical domain (HD)–C-terminal tail. Studies using mutagenesis and crystallization have shown that the CRD, which consists of nine cysteine residues, are critical to ligand-induced signal transduction, in part via a disulfide bridge formed between a cysteine in lobe 2 of the VFD and the ninth CRD cysteine (Rondard et al., 2006; Muto et al., 2007). Additionally, the second intracellular loop of mGlu receptors regulate selectivity of G protein coupling (Pin et al., 1994; Gomeza et al., 1996) and acts as an important regulatory site for kinases, like G-protein-coupled receptor kinase 2 (Dhami et al., 2005). Importantly, allosteric modulators of mGlu receptors that bidirectionally alter glutamate activity largely bind within the HD (Niswender and Conn, 2010). The C-terminus region of mGlu receptors is important for modulating G protein coupling and undergo alternative splicing, modulatory protein-protein interactions, and regulation by phosphorylation (Niswender and Conn, 2010; Enz, 2012). In addition to interacting with G-proteins, the C-terminal domains of mGlu receptor subtypes directly interact with many proteins, including enzymes, ion channels, receptors, scaffolds, and cytoskeletal proteins (Enz, 2012).

2. Metabotropic Glutamate Receptor Signal Transduction

As detailed in Table 1, group I mGlu receptor subtypes (mGlu1/5) are widely expressed in CNS neurons (Maksymetz et al., 2017). mGlu1 and mGlu5 are primarily expressed postsynaptically but are also located at presynaptic terminals of GABA and glutamate neurons (Higley, 2014). Group I mGlu receptor subtypes canonically couple to the G protein Gq/11 alpha subunit (Gαq/11) and activate phospholipase C beta (β), which in turn results in the hydrolysis of phosphotinositides and generation of inositol 1,4,5-trisphosphate and diacyl-glycerol. This signaling pathway promotes calcium mobilization and downstream activation of protein kinase C. Additionally, group I mGlu receptor subtypes can also signal through alternative pathways including the G protein Gi/o alpha subunit (Gαi/o), Gs alpha subunit (Gαs), and other molecules independent of G proteins (Hermans and Challiss, 2001). Group I mGlu receptors activate many downstream signaling effectors, including phospholipase D, protein kinase cascades like kinase 1, cyclin-dependent protein kinase 5, mitogen-activated protein kinase (MAPK)/extracellular receptor kinase, Jun kinase, and the mammalian target of rapamycin/P70 S6 kinase (p70-S6) kinase pathways (Page et al., 2006; Li et al., 2007). These signaling cascades, such as mammalian target of rapamycin/p70 S6 kinase and MAPK/extracellular receptor kinase, are critical to synaptic plasticity mediated by group I mGlu receptors. Additionally, group I mGlu receptors represent promising therapeutic targets, based on their ability to directly couple to NMDA receptors via intracellular signaling pathways and scaffolding proteins, such as SRC Homology 3 Domain (SH3), Homer, and multiple ankyrin repeat domains protein, and guanylate kinase-associated protein–postsynaptic density-95 (Aniksztejn et al., 1991; Harvey and Collingridge, 1993; Yu et al., 1997) and their subsequent capacity to activate NMDA receptors in acute brain slices (Fitzjohn et al., 1996).

TABLE 1.

Summary of mGlu receptor subtype expression, signaling, and interacting partners

Group Receptor CNS Expression Synaptic Localization G protein Coupling Primary Signaling Pathways Interacting Partners
Group I mGlu1 Widespread in neurons Predominantly postsynaptic Primarily Gαq/11 noncanocial Gαi/0, Gαs PLCβ → IP3 + DAG hydrolysis phospholipase D MAPK/ERK mTOR/p70 S6 kinase Activates NMDA receptors activates Ca2+ channels (e.g., Cav2.1) activates CaMKIIα
mGlu5 Widespread in neurons
Group II mGlu2 Widespread in neurons Presynaptic and postsynaptic Primarily Gαi/0 Inhibition of adenylyl cyclase MAPK/ERK IP3-PI3 kinase Activation of K+ channels inhibition of voltage-gated Ca2+ channels (e.g., N- type)
mGlu3 Widespread in neurons, astrocytes
Group III mGlu4 Widespread in neurons, high in cerebellum Predominantly presynaptic Primarily Gαi/0 noncanocial Gαq/1 Inhibition of adenylyl cylcase MAPK/ERK IP3 (mGlu7) stimulation of cGMP phosphodiesterase (mGlu6) Activation of K+ channels inhibition of Ca2+ channels (P/Q subtype)
mGlu6 Retina, select neuron populations, microglia Postsynaptic in retinal cells
mGlu7 Widespread in neurons Active zone of presynaptic terminals
mGlu8 Lower and more restricted expression than mGlu4/7 Predominantly presynaptic

caMKII-alpha, Calcium/calmodulin-dependent kinase II; cGMP, cyclic guanosine monophosphate; DAG, diacyl-glycerol; G protein i/o alpha subunit; G protein Gs alpha subunit; G protein Gq alpha subunit; IP3, inositol 1,4,5-trisphosphate; MAPK/ERK, mitogen-activated protein kinase/extracellular receptor kinase; mTOR, mammalian target of rapamycin; PI3 kinase, phosphatidyl inositol 3-kinase; PLCβ, phospholipase Cβ; p70-S6 kinase, P70-S6; P/Q, P/Q-type calcium channel.

The group II mGlu receptors, mGlu2 and mGlu3, are expressed presynaptically (Niswender and Conn, 2010) on axonal preterminal regions where they can be activated by excessive synaptic or astrocytic glutamate release (Nicoletti et al., 2011; Maksymetz et al., 2017). mGlu3 is located postsynaptically as well as on astrocytes where it promotes neuroprotective effects (Nicoletti et al., 2011) and facilitates astrocytic-neuronal communication (Winder and Conn, 1996; Winder et al., 1996). Group II mGlu receptor subtypes predominantly couple to Gi/o proteins, which classically inhibit adenylyl cyclases and downstream production of 3′,5′-cAMP. Group II mGlu receptors also directly regulate ion channels, including potassium (K+) and calcium (Ca2+) channels, and other downstream signaling components via G protein beta/gamma (Gβγ) subunits (Niswender and Conn, 2010). Additionally, group II mGlu receptors can engage in a multitude of signal transduction pathways, including activation of phosphatidyl inositol 3-kinase and MAPK pathways (Muguruza et al., 2016), demonstrating the great complexity by which these receptors regulate neuronal signaling and synaptic plasticity.

The group III mGlu receptors consist of mGlu4 (Tanabe et al., 1992), mGlu6 (Nakajima et al., 1993), mGlu7 (Saugstad et al., 1994), and mGlu8 (Niswender and Conn, 2010). mGlu6 receptors are largely expressed in the retina, however, these receptors have also been shown to be expressed in the CNS, including cortical areas, superior colliculus, the accessory olfactory bulb, and axons of the corpus callosum (Vardi et al., 2011; Palazzo et al., 2020). Alternatively, other group III mGlu receptors are largely expressed within the CNS (Nakajima et al., 1993). Group III mGlu receptors canonically signal via the Gαi/o subunits of the heterotrimeric G protein complex, resulting in inhibition of adenylyl cyclase and cAMP production (Niswender and Conn, 2010). Activation of group III mGlu receptors can also regulate a variety of ion channels and inhibition of vesicular fusion via Gβγ subunits (Cartmell and Schoepp, 2000). In addition to canonical Gαi/o-mediated downstream signaling, group III mGlu receptors engage in a multitude of other signal transduction cascades. For instance, mGlu7 has been shown to selectively inhibit P/Q-type (P/Q)-type Ca2+ channels through a phospholipase C-dependent mechanism resulting in subsequent Ca2+ release from intracellular stores and diacyl-glycerol–mediated activation of protein kinase C (Perroy et al., 2000). Furthermore, activation of mGlu4 results in MAPK pathway signaling via Gβγ subunits (Iacovelli et al., 2004). Evidence suggests that the mGlu6 receptor signals through Gαo to a cyclic guanosine monophosphate-preferring phosphodiesterase (Shiells and Falk, 1990; Thoreson and Miller, 1994; Nawy, 1999).

B. Metabotropic Glutamate Receptor Regulation of Neurotransmission and Synaptic Plasticity

mGlu receptors are distributed widely throughout the CNS in a vast array of major brain regions and are localized at discrete synaptic and extrasynaptic sites in both neurons and glia. Based on their robust CNS expression and diverse signal transduction pathways, activation of mGlu receptors elicits a multitude of outcomes on synaptic transmission and contributes to many forms of synaptic plasticity (Crupi et al., 2019). These activity-dependent modifications of synaptic transmission are critical to learning and memory, for example and thus, represent important mechanisms underlying many neuropsychiatric disorders, such as schizophrenia and substance use disorders (SUDs). Here, we will briefly summarize select functional roles of mGlu receptor subtypes in neurotransmission and synaptic plasticity. In-depth discussion of the vast array of physiological roles of mGlu receptors has been presented in numerous previous reviews (Benarroch, 2008; Niswender and Conn, 2010; Mukherjee and Manahan-Vaughan, 2013; Maksymetz et al., 2017). mGlu receptors expressed presynaptically have the ability to increase or decrease neurotransmitter release at excitatory (glutamate), inhibitory (GABA), and neuromodulatory (i.e., monoamines, acetylcholine, peptides) synapses (Niswender and Conn, 2010). In most cases, mGlu receptor-mediated regulation of neurotransmitter release is mediated by mGlu receptors that are localized presynaptically; however, this can also occur via postsynaptic mGlu receptors and release of retrograde messengers, such as endocannabinoids (Yohn et al., 2020). mGlu receptor-mediated neuromodulation, in turn, has a wide array of downstream effects on neuronal activity and firing. For example, mGlu receptor-mediated inhibition of GABA transmission in the cerebellum results in a local reduction in inhibition, enhancing the efficacy of the more active fibers and accentuating the contrast between inputs with differential firing rates (Mitchell and Silver, 2000).

1. Group I: Metabotropic Glutamate1/5

Group I mGlu receptors, which include mGlu1 and mGlu5, are heterogeneously expressed throughout the brain, with high levels in regions critical to cognition, reinforcement learning, and motivation, such as the nucleus accumbens (NAc), hippocampus, medial prefrontal cortex (mPFC), and thalamus (Cleva and Olive, 2012). Further studies have revealed that the vast majority of mGlu1 and mGlu5 receptors are located postsynaptically on dendritic spines (Olive, 2009) and on axon terminals in brain regions, such as the hippocampus (Romano et al., 1995) and cerebral cortex (Muly et al., 2003; Paquet and Smith, 2003). Additionally, group I mGlu receptors in globus pallidus of nonhuman primates are found in the main body of symmetric synaptic junctions established by striatal GABA terminals as well as perisynaptic to asymmetric glutamatergic synapses (Hanson and Smith, 1999). Their activation leads to cell depolarization and increases in neuronal excitability (Niswender and Conn, 2010). Group I mGlu receptor-mediated modulation of neuronal excitability is driven by regulation of numerous ion channels, which enable fine-tuning of neuronal excitability (Conn and Pin, 1997; Anwyl, 1999; Coutinho and Knöpfel, 2002; Valenti et al., 2002).

Activation of group I mGlu receptor subtypes leads to alterations in excitability and spontaneous synaptic transmission in the mPFC and cornu ammonis (CA1) region of the hippocampus, among other brain regions (Zho et al., 2002; Yin and Niswender, 2014; Turner et al., 2018; Maksymetz et al., 2021). For example, activation of group I mGlu receptors by selective orthosteric agonists, such as (S)-3,5-dihydroxyphenylglycine (DHPG), results in direct excitatory effects on CA1 pyramidal cells, including increased cell firing and depolarization (Charpak et al., 1990; Desai and Conn, 1991; Pedarzani and Storm, 1993; Davies et al., 1995; Gereau and Conn, 1995; Mannaioni et al., 1999). These downstream effects are mediated by inhibition of K+ currents and activation of both Ca2+-dependent and independent conductance (Crépel et al., 1994; Guérineau et al., 1995). Additionally, recent studies have directly demonstrated for the first time that activation of mGlu1 increases inhibitory transmission in the mPFC by excitation of somatostatin-expressing interneurons (SST-INs) (Maksymetz et al., 2021). Extensive evidence also supports the role of mGlu1 and mGlu5 receptor subtypes in numerous forms of long-term synaptic plasticity, including long-term depression (LTD) and long-term potentiation (LTP) of transmission. At excitatory synapses in hippocampal CA1, activation of mGlu1/5 and subsequent Ca2+ mobilization has been shown to induce LTP, a modality of synaptic plasticity thought to underlie learning and memory, at numerous glutamatergic synapses (Bashir et al., 1993; Frenguelli et al., 1993; Petrozzino and Connor, 1994; Balschun et al., 1999; Chevaleyre and Castillo, 2003; Gladding et al., 2009). However, there are mixed reports regarding the role of mGlu5 in N-methyl-D-aspartate receptor (NMDAR)-dependent LTP (Fitzjohn et al., 1996, 1998; Lu et al., 1997; Francesconi and Duvoisin, 2004; Bortolotto et al., 2005; Neyman and Manahan-Vaughan, 2008). Several studies have also revealed that mGlu1 and mGlu5 subtypes regulate LTP in hippocampal SST-INs (McBain et al., 1994; Perez et al., 2001; Le Duigou and Kullmann, 2011; Pelkey et al., 2017). There is evidence for the role of group I mGlu receptors in regulation of LTP extrahippocampal brain regions. For instance, it has been reported that application of DHPG facilitated LTP of the evoked excitatory postsynaptic currents (EPSCs) in SST-INs of the prefrontal cortex (PFC) (Crowley and Joffe, 2021). Furthermore, a wealth of studies has shown that activation of group I mGlu receptors induces a LTD of synaptic transmission in rat hippocampal CA1 (Palmer et al., 1997; Chevaleyre and Castillo, 2003; Tan et al., 2003; Gladding et al., 2009) and dentate gyrus (O’Mara et al., 1995; Camodeca et al., 1999) among other brain regions (Kano and Kato, 1987; Kato, 1993; Conquet et al., 1994; Wang et al., 2015).

The development of mouse models with selective deletion of group I mGlu receptors have corroborated these findings. For instance, electrophysiological recordings from mGlu1–/– mice show that these animals display impaired hippocampal LTP, which correlates with impairments in context-specific learning and impaired LTD in the cerebellum (Aiba et al., 1994a,b; Gil-Sanz et al., 2008). A wealth of studies using constitutive knockout models further confirmed the role of mGlu5 in hippocampal LTP (Bashir et al., 1993; Lu et al., 1997). Using genetic deletion or pharmacological inhibition, it has been demonstrated that mGlu5 reduces LTP at Shaffer collateral (SC)-CA1 synapses of the hippocampus in freely moving rats and ex vivo slice preparations (Lu et al., 1997; Francesconi et al., 2004; Shalin et al., 2006). In addition, LTP induction can be primed by DHPG (Cohen et al., 1998; Raymond et al., 2000), and multiple mGlu5 positive allosteric modulators (PAMs) can induce LTP at SC-CA1 synapses (Ayala et al., 2009; Noetzel et al., 2013; Rook et al., 2015). More recently, studies using conditional knockout of Glutamate Metabotropic Receptor 5 gene (GRM5) in hippocampal CA1 pyramidal cells showed that loss of mGlu5 in this cellular population impaired LTD of inhibitory synapses compared with wild-type control mice (Xu et al., 2014), suggesting a specific role of mGlu5 in hippocampal CA1 pyramidal cells in metaplasticity by regulating inhibition. These findings are of particular importance as hippocampal LTP is known to be altered in models that recapitulate the physiologic and behavioral phenotypes associated with neuropsychriatic diseases, including schizophrenia. For example, subchronic phencyclidine (PCP) treatment in mice increases the threshold for LTP of CA1 excitatory synapses, and this effect is directly related to enhanced inhibitory input to CA1 pyramidal cells through increased activity of GABAergic neurons (Nomura et al., 2016). Therefore, the contributions of group I mGlu receptors in regulating LTP at excitatory synapses is critical for our understanding of the pathophysiology of neuropsychiatric diseases and development of novel treatments.

2. Group II: Metabotropic Glutamate2/3

The group II mGlu receptor subtypes, mGlu2 and mGlu3, are expressed throughout the CNS, notably in brain regions central to motivation, learning, and memory (Moussawi and Kalivas, 2010; Muguruza et al., 2016). In some instances, they modulate synaptic transmission and alter neuroplasticity by acting at preterminal regions away from the active zone of on glutamatergic or GABAergic synapses (Nicoletti et al., 2011). mGlu2/3 receptors located on the presynaptic membrane can be activated by substantial synaptic or astrocytic glutamate release (Muguruza et al., 2016; Maksymetz et al., 2017). Additionally, further evidence supports the existence of mGlu2 and mGlu3 expressed postsynaptically (Muguruza et al., 2016).

At many synapses, mGlu2/3 receptor activation decreases spontaneous excitatory transmission (Marek et al., 2000; Kiritoshi and Neugebauer, 2015; Bocchio et al., 2019). Studies have shown that group II mGlu receptor agonist LY 354740 decreases frequency, but not amplitude, of miniature EPSCs in the presence of tetrodotoxin, which suggests that the site of action is primarily presynaptic on the glutamatergic terminals (Han et al., 2006; Kiritoshi and Neugebauer, 2015). In the infralimbic mPFC, electrophysiology experiments in rat brain slices showed that group II mGlu receptor subtypes decrease the output of layer V pyramidal cells as the result of an inhibitory action on glutamatergic synapses (Kiritoshi and Neugebauer, 2015; Thompson and Neugebauer, 2017). More specifically, LY379268, a selective group II mGlu receptor agonist, regulates activity of pyramidal cells by modulating glutamate-driven feedforward inhibitory transmission (inhibitory postsynaptic currents) in addition to direct EPSCs. In addition to regulation of neurotransmitter release and short-term plasticity, group II mGlu receptors play important roles in long-term synaptic plasticity, which underlies many cognitive and behavioral processes disrupted in neuropsychiatric disease. Activation of either mGlu2 or mGlu3 receptor subtypes induces a robust, postsynaptic LTD of evoked synaptic responses in the PFC. Interestingly, mGlu2 and mGlu3 receptors induce LTD by divergent presynaptic and postsynaptic mechanisms, respectively (Walker et al., 2015; Joffe et al., 2019a,b). At mossy fiber pyramidal cell synapses, prolonged low-frequency stimulation results in a presynaptic form of LTD that is absent in mGlu2-deficient mice and blocked by a nonselective group II antagonist. At this synapse, the activation of mGlu2 induces LTD selectively when it is coupled to a synaptically-driven increase in presynaptic Ca2+. Additionally, mossy fiber LTP is reversible by low-frequency stimulation via the activation of group II mGlu receptors (Chen et al., 2001). Finally, exciting recent reports have demonstrated that activation of mGlu3 induces metaplastic changes, biasing stimulation of afferents to induce LTP through an mGlu5 receptor-dependent, endocannabinoid-mediated mechanism of action (Dogra et al., 2021). In this same study, targeted genetic deletion of mGlu3 from hippocampal pyramidal cells prevented the LTP-inducing effects of mGlu3 activation, revealing a novel avenue by which mGlu3 regulates long-term hippocampal synaptic plasticity.

3. Group III: Metabotropic Glutamate4/6/7/8

Group III mGlu receptors are differentially expressed in the CNS and peripheral nervous system. mGlu4 and mGlu8 subtypes are expressed in the brain but in a restricted manner (Pilc et al., 2008; Julio-Pieper et al., 2011). Although mGlu4 is primarily found in the cerebellum (Kinoshita et al., 1996b; Shigemoto et al., 1997), expression has also been reported in the cerebral cortex, striatum, olfactory bulb, pontine nuclei, lateral septum, hippocampus, thalamic nuclei, and dorsal horn (Fotuhi et al., 1994; Azkue et al., 2001). Within the CNS, mGlu8 is found presynaptically in the hippocampus, cerebellum, olfactory bulb, and cortical areas (Ferraguti and Shigemoto, 2006). Although mGlu7 is expressed widely throughout the brain, mGlu6 only exhibits limited expression in the retina (Crupi et al., 2019). mGlu7 expression has been reported in the amygdala, hypothalamus, hippocampus, thalamus, and locus coeruleus (Ngomba et al., 2011). Similar to group II mGlu receptors, group III mGlu receptor subtypes are most commonly located in or near presynaptic active zones of GABAergic and glutamatergic neuronal cells (Shigemoto et al., 1997; Ferraguti and Shigemoto, 2006). Activation of group III mGlu receptors inhibits the release of neurotransmitters, such as glutamate, GABA, and dopamine, via modulation of a variety of ion channels and Gβγ subunit-dependent inhibition of vesicular fusion (Cartmell and Schoepp, 2000).

Until recent advances in group III mGlu receptor subtype-selective pharmacological compounds, most of the research pertaining to group III mGlu receptors used the broad-spectrum agonist, L-2-amino-4-phosphonobutyric acid (L-AP4). A multitude of studies have shown that L-AP4 reduces excitatory transmission in numerous brain regions, including the hippocampus, amygdala, striatum, globus pallidus, thalamus, hypothalamus, cerebellum, NAc, and substantia nigra (Mercier and Lodge, 2014). Application of L-AP4 typically increases paired-pulse ratio (Harris and Cotman, 1983; Manzoni et al., 1997; Lorez et al., 2003), and its application has also been shown to reduce the frequency, but not the amplitude, of miniature excitatory postsynaptic events (Harris and Cotman, 1983; Gereau and Conn, 1995; Manzoni et al., 1997; Schoppa and Westbrook, 1997; Schrader and Tasker, 1997), both of which indicate the role of group III mGlu receptors as presynaptic autoreceptors in the CNS. Group III mGlu receptors are also involved in the regulation of GABA and monoamine neurotransmission (Cartmell and Schoepp, 2000; Schoepp, 2001). In addition to its ability to regulate glutamate release, L-AP4 has been found to reduce inhibitory transmission in many brain regions, including the midbrain, globus pallidus, striatum, thalamus, and hippocampus (Mercier and Lodge, 2014). Importantly, hippocampal electrophysiological studies bolstered evidence for the localization of group III mGlu receptors on both glutamate and GABA terminals, such that L-AP4 has reduced both excitatory and inhibitory transmission onto hippocampal interneurons and pyramidal cells (Semyanov and Kullmann, 2000; Kogo et al., 2004; Klar et al., 2015). The critical role of group III mGlu receptors on interneurons highlights their utility in regulating the balance of excitation and inhibition of these cells, thus regulating overall network excitability (Ferraguti and Shigemoto, 2006; Klar et al., 2015).

A multitude of studies have leveraged selective pharmacological tools and transgenic knockout lines to determine the roles of group III mGlu receptors in long-term synaptic plasticity. For instance, application of α-Cyclopropyl-4-phosphonophenylglycine (CPPG), the group III mGlu receptor antagonist, prevents LTD, but not LTP, in the CA1 region of mice (Altinbilek and Manahan-Vaughan, 2007). However, the development of selective pharmacological and genetic tools has provided additional insight to the role of Group III mGlu receptor subtypes in long-term synaptic plasticity. A recent study shows that inhibition of group III mGlu receptors elicit an NMDA receptor-dependent LTP in SC-CA2 synapses (Dasgupta et al., 2020). Further, disinhibition produced by activation of mGlu7 induces LTP at SC-CA1 synapses in the hippocampus (Klar et al., 2015). Interestingly, one study using mGlu4 knockout mice found enhanced LTP in hippocampal CA1 region but not in the PFC compared with wild-type controls (Iscru et al., 2013). Other studies have shown that mGlu receptor-dependent LTD can be induced after activation of mGlu7 (Bellone et al., 2008). Together, these demonstrate critical roles of group III mGlu receptor subtypes in regulating various forms of long-term synaptic plasticity in brain regions and circuits dysregulated in neuropsychiatric disease.

II. Targeting Metabotropic Glutamate Receptors for the Treatment of Neuropsychiatric Disease

A. Metabotropic Glutamate Receptor Allosteric Modulators

The vast diversity and distribution of mGlu receptors provides an unparalleled opportunity for selective targeting of individual mGlu receptor subtypes as novel treatment strategies for neuropsychiatric and neurologic disorders. However, longstanding efforts to develop ligands that target mGlu receptors have largely focused on agonists and antagonists that interact with the orthosteric glutamate binding sites of these receptors to mimic or block the endogenous actions of glutamate. Although this strategy has proven to be somewhat fruitful, the high conservation of orthosteric binding sites across receptor subtypes has served as a critical barrier to the development of subtype-selective orthosteric ligands. To address this issue, recent efforts have been focused on developing allosteric modulators for mGlu receptor subtypes. Allosteric modulators act by altering the receptor conformational state by binding a topographically distinct nonorthosteric site, typically found within the HD of mGlu receptors (Wu et al., 2014). Thus, allosteric modulators potentiate or attenuate the response to the endogenous orthosteric ligand (i.e., glutamate for mGlu receptors) without activating the receptor directly. Allosteric modulators can be categorized based on the direction that they modulate the response to the orthosteric agonist. For instance, allosteric modulators that increase the functional response to an orthosteric agonist are referred to as “positive allosteric modulators.” In contrast, those that attenuate the functional response to the orthosteric agonist are coined “negative allosteric modulators” (NAMs). If a compound binds to an orthosteric site without inducing effects on the response of the receptor, it is called a “neutral allosteric ligand” (Conn et al., 2009). In functional assays, such as those that measure calcium mobilization or downstream signaling (e.g., cAMP accumulation), the presence of a PAM often induces a leftward shift of the agonist concentration–response curve, whereas a NAM decreases the maximal effect of the response. In addition to this modulatory activity, a subset of allosteric modulators possesses intrinsic activity and can both potentiate agonist responses and directly activate the receptor (ago-PAMs). In sum, allosteric modulators display a number of the following pharmacological properties: 1) efficacy modulation, the signaling capacity (or “intrinsic efficacy”) of an orthosteric agent can be modified via alterations in intracellular responses; 2) affinity modulation, the conformational change induced by binding of the allosteric ligand alters the binding pocket and association/dissociation rates of orthosteric ligands; and 3) agonism/inverse agonism, receptor signaling is altered either positively (agonism) or negatively (inverse agonism) by the allosteric modulator, irrespective of the presence or absence of an orthosteric ligand.

B. Advantages of Metabotropic Glutamate Allosteric Modulators

Allosteric modulators provide several advantages as potential pharmacotherapies and experimental tools in comparison with orthosteric ligands. For example, orthosteric ligand binding sites often possess a high degree of sequence homology, which presents a challenge for the development of receptor subtype-specific ligands. In contrast, allosteric ligand binding sites are often less highly conserved than orthosteric sites, allowing for the development of highly selective allosteric modulators for receptor subtypes that, formerly, have been intractable using traditional approaches (Conn et al., 2009; Nussinov et al., 2011). Additionally, differential receptor cooperativity serves as a means of subtype selectivity. The cooperativity between orthosteric and allosteric sites is not correlated with the affinity of allosteric modulators for their binding sites, and, thus, allows some allosteric modulators to bind to more than one receptor subtype with similar affinities but elicit effects through distinct cooperativity between receptor subtypes (Boehr et al., 2009; Tsai et al., 2009).

One shortcoming of orthosteric agonists targeting mGlu receptors is the risk of receptor overactivation, which can disrupt brain circuit modulation and result in adverse side effects (Sendt et al., 2012; Rook et al., 2013). This challenge is more effectively circumvented by allosteric modulators with their ability to fine-tune active synapses versus nonphysiological activation of synapses that display low glutamatergic tone. This allows for regulation of receptor responses in brain areas where the endogenous agonist exerts its physiologic effect, reducing the risk for off target or overactivation. For instance, mGlu5 agonists can induce epileptic seizure activity (Tizzano et al., 1995) This effect is also observed with allosteric agonists but is mitigated by administration of mGlu5 PAMs that lack agonist activity (Rook et al., 2015; Gould et al., 2016). This requirement for the presence of the endogenous agonist provides spatial and temporal control of allosteric modulators and provide a sizable therapeutic edge by allowing for physiologically appropriate modulation of synaptic signaling and transmission.

In addition to serving as novel treatment strategies, these ligands are essential to driving fundamental investigation into the roles of specific signaling pathways and distinct receptors in modulating identified neural circuits and behavior under physiologic and pathologic conditions. Allosteric modulators of mGlu receptor subtypes are now being pursued as potential drug candidates for numerous neuropsychiatric diseases, including Alzheimer’s disease, Parkinson’s disease, dystonia, schizophrenia, SUDs, and other brain diseases (Conn et al., 2009; O’Brien and Conn, 2016; Foster and Conn, 2017; Maksymetz et al., 2017; Joffe and Conn, 2019; Stansley and Conn, 2019). The present review will summarize the current findings on the efficacy of mGlu receptor subtype PAMs/NAMs for the treatment of neuropsychiatric diseases, with particular focus on schizophrenia and SUDs.

III. Potential of Allosteric Modulators of Metabotropic Glutamate Receptors for Treating Neuropsychiatric Disease

A. Schizophrenia

Schizophrenia is a chronic neuropsychiatric disorder that affects approximately 1% of the world population (GBD 2019 Diseases and Injuries Collaborators, 2020). The disease is characterized by three primary clusters of symptoms: positive (auditory/visual hallucinations), negative (amotivation, anhedonia, social withdrawal), and cognitive deficits (working memory, executive function, attention). Current antipsychotic medications effectively treat the positive symptoms of the disease, such as auditory and visual hallucinations, disorganized thoughts, and delusions; however, they do not improve the negative or cognitive symptoms. Negative symptoms (e.g., flattened affect, social withdrawal) and cognitive symptoms (e.g., deficits in attention, working memory, and cognitive flexibility) are believed to be the best predictors of long-term treatment outcome (Green, 1996; Bobes et al., 2007; McEvoy, 2007). Furthermore, many patients discontinue treatment due to adverse effects, such as extrapyramidal side effects (EPS) (i.e., tardive dyskinesia, tremor, dystonia, and bradykinesia) induced by typical antipsychotics as well as a host of metabolic side effects (i.e., weight gain, hyperlipidosis, and type II diabetes) elicited most commonly by atypical antipsychotics (Lieberman et al., 2005; Meltzer, 2013; Lally and MacCabe, 2015). Therefore, development of improved therapeutic options that mitigate a broader range of symptoms of schizophrenia and are devoid of EPS is of great need.

Our current understanding of the neurochemical alterations driving the symptoms associated with schizophrenia is largely attributed to two major lines of research that are driven by the dopamine and glutamate hypotheses of schizophrenia-related dysfunction (Howes et al., 2015; McCutcheon et al., 2019). The dopamine hypothesis posits that the positive symptoms of the disease are largely driven by aberrant dopamine signaling. This notion is supported by evidence that amphetamine and other dopamine-releasing agents induce symptoms that resemble those of the positive symptoms of schizophrenia (Steeds et al., 2015; Kesby et al., 2018). Further, currently available antipsychotic medications largely target the dopamine system and act, in part, by inhibiting dopamine D2 subtype of dopamine receptors (Meltzer, 2013). In support of this model, in vivo neuroimaging studies show increased subcortical dopamine release after amphetamine challenge in individuals with schizophrenia (Laruelle et al., 1996; Breier et al., 1997; Abi-Dargham et al., 1998). However, since amphetamine exposure exacerbates only the positive symptoms of schizophrenia and dopamine-targeting antipsychotic medications only alleviate positive symptoms, dopaminergic hyperactivity alone cannot account for the negative symptoms or cognitive disturbances observed in patients with schizophrenia (Carlsson, 1988).

Another prominent line of research suggests that disruption of glutamate signaling underlies numerous symptoms of schizophrenia. This notion is derived from extensive evidence that NMDA antagonists, such as PCP, ketamine and dizocilpine (MK-801), induce symptoms closely resembling those of schizophrenia (Javitt and Zukin, 1991). In addition, administration of NMDAR antagonists exacerbates or induces controlled symptoms when administered to schizophrenia patients (Krystal et al., 1994). These findings, in addition to a wealth of preclinical evidence, support the hypothesis that NMDAR hypofunction contributes to the pathophysiology underlying schizophrenia. Thus, pharmacological agents that enhance NMDAR function represent a potential strategy that could provide therapeutic benefits to patients with schizophrenia. However, a primary obstacle is that overactivation of NMDARs using traditional orthosteric agonists induces adverse effects, such as excitotoxicity and seizures (Zeron et al., 2002; Kaufman et al., 2012; Monaghan et al., 2012; Puddifoot et al., 2012). Importantly, mGlu receptors are critical modulators of NMDAR function and regulate glutamate and GABA neurotransmission throughout the CNS (Niswender and Conn, 2010; Maksymetz et al., 2017). Therefore, pharmacological modulation of mGlu receptors holds the potential to alter NMDAR function and restore excitatory and inhibitory neurotransmission to provide therapeutic benefit in patients with schizophrenia. To this end, allosteric modulators targeting all three groups of mGlu receptors have been pursued as putative targets for novel antipsychotics (Tables 24).

TABLE 2.

Summary of preclinical efficacy of group I mGlu receptor allosteric modulators in schizophrenia-related deficits

Receptor Type Compound Positive Symptom Models Negative Symptoms Models Cognitive Models References
mGlu1 PAM VU0483605 No effect on AHL Cho et al., 2014
VU6004909 Attenuates AMPH-induced hyperlocomotion and disruptions in PPI No effect on PR Yohn et al., 2020
NAM FTIDC Attenuates MHL and PPI deficits Satow et al., 2009
CFMTI Reduced MHL and NMDAR antagonist-induced hyperlocomotion (NMDAR-HL) Ameliorated METH and ketamine-disrupted PPI Reversal of MK-801-disrupted social interaction No effect on object location memory Satow et al., 2008
mGlu5 PAM CDPPB Attenuated AMPH-induced hyperlocomotion and deficits in PPIa,b Attenuated MK-801-induced decrease in sucrose preferencec ↑Morris water maze learningd
↓MK-801 deficits in cognitive flexibilitye
↓PCP deficits in NORf
(See footnotes.)
5PAM523 Reduced AHLg,h and NMDAR-HLh ↑Contextual CF2
↑NORh
(See footnotes.)
VU0409551 Reverses MK-801-induced hyperlocomotion ↑Working memory/executive function in the DNMTP task
↑contextual CF deficits in SR−/− mice
Rook et al., 2015

AHL, amphetamine-induced hyperlocomotion; AMPH, amphetamine; CF, fear conditioning; DNMTP, delayed nonmatching to position; HL, hyperlocomotion; METH, methamphetamine; MHL, METH-induced hyperlocomotion; NOR, novel object recognition; SR, serine racemase-deficient.

aKinney et al., 2003.

bLindsley et al., 2005.

cVardigan et al., 2010.

dAyala et al., 2009.

eStefani and Moghaddam, 2010.

fHorio et al., 2013.

gParmentier-Batteur et al., 2014.

hRook et al., 2015.

TABLE 4.

Summary of preclinical efficacy of group III mGlu receptor allosteric modulators in schizophrenia-related deficits

Receptor Type Compound Positive Symptom Models Negative Symptoms Models Cognitive Models References
mGlu4 PAM ADX88178 Reduced NMDAR-HL; reduced DOI-induced head twitches Reduced immobility in FST Kalinichev et al., 2014
Lu AF21934 Reduced NMDAR-HL and AHL;a reduced DOI-induced head twitchesa Reduced MK-801-induced deficits in social interactiona Rescued MK-801-induced deficits in the delayed spatial alternation task;a reduced MK-801-induced deficits in NOR (See footnotes.)
Lu AF32615 Reduced NMDAR-HL and AHL; reduced DOI-induced head twitches Reversed MK-801-induced deficits in social interaction Reversed MK-801-induced deficits in the delayed spatial alternation task Sławińska et al., 2013
mGlu7 Ago-PAM AMN082 No effect on AHL; exacerbated NMDAR-HL;b exacerbated DOI-induced head twitchesc (See footnotes.)
NAM MMPIP ADX71743 Inhibited MK-801-induced hyperactivity and reversed deficits in PPI Reversed MK-801-induced deficits in NOR and spatial delayed alternation Cieślik et al., 2018

AHL, amphetamine-induced hyperlocomotion; HL, hyperlocomotion; NOR, novel object recognition.

aSławińska et al., 2013.

bMitsukawa et al., 2005.

cWierońska et al., 2012a.

I. Group I: Metabotropic Glutamate1/5

The mGlu1 receptor subtype shows promise as a potential therapeutic target for treating schizophrenia. Genetic studies in humans reveal an association of the human gene encoding mGlu1 (GRM1) and, specifically, loss of function single nucleotide polymorphisms in GRM1 with schizophrenia, raising the possibility that mGlu1 signaling is critical to the function of brain circuits underlying symptoms associated with this disorder (Ayalew et al., 2012; Ayoub et al., 2012; Cho et al., 2014). Numerous studies have characterized the pivotal role of mGlu1 in regulating GABA and glutamate signaling in the PFC as well as striatal dopamine dynamics. The development of mGlu1 PAMs and transgenic mouse lines have allowed for these discoveries. Potent first-generation mGlu1 PAMs were developed in the early 2000s; however, they displayed poor pharmacokinetic and metabolic profiles, limiting their use in preclinical studies (Knoflach et al., 2001; Vieira et al., 2005). More recent efforts yielded mGlu1 PAMs, such as VU 6000799, VU6000790, and VU6004909 as potent, highly selective mGlu1 PAMs with improved drug metabolism and pharmacokinetic (DMPK) properties and brain penetrance and are therefore better suited for in vivo studies (Garcia-Barrantes et al., 2015, 2016a,b,c). Recent studies have leveraged these improved mGlu1 PAMs and have yielded promising results. For instance, Yohn et al., showed that activation of mGlu1 negatively regulates striatal dopamine release through an intricate mechanism involving coactivation of muscarinic acetylcholine subtype 4 (M4) receptors and retrograde endocannabinoid signaling (Yohn et al., 2020). Additionally, a recent study has characterized the critical role of mGlu1 receptors located on SST-INs of the prelimbic PFC in regulating inhibitory output onto glutamatergic pyramidal cells, highlighting mGlu1 as a critical determinant of inhibitory/excitatory balance in the PFC (Maksymetz et al., 2021). The ability to normalize both dopamine and GABA dysfunction highlights mGlu1 as a promising target to comprehensively treat symptomology associated with schizophrenia. Importantly, mGlu1 PAMs show robust antipsychotic-like efficacy in rodent models. Specifically, the mGlu1 PAM VU6004909 reverses amphetamine-induced hyperlocomotion and deficits in prepulse inhibition (PPI) induced by amphetamine treatment (Yohn et al., 2020). Furthermore, mGu1 PAMs show promising cognitive-restoring effects in rodent models. A recent study demonstrates that mGlu1 PAM VU6004909 reverses deficits in spatial working memory induced by NMDAR antagonist, MK-801 (Maksymetz et al., 2021). In addition to the potential of mGlu1 PAMs in treating schizophrenia, mGlu1 NAMs 4-[1-(2-fluoropyridin-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide (FTIDC) and 2-cyclopropyl-5-[1-(2-fluoro-3-pyridinyl)-5-methyl-1H-1,2,3-triazol-4-yl]-2,3-dihydro-1H-isoindol-1-one (CFMTI) have displayed efficacy in animal models of antipsychotic activity (Table 2), such as decreasing NMDAR antagonist and psychostimulant-induced hyperlocomotion and deficits in PPI as well as reversing social interaction deficits elicited by MK-801, an NMDAR antagonist, in rats (Satow et al., 2008, 2009). The contrasting findings of mGlu1 PAMs and NAMs illustrate the potential complexity of mGlu1-targeting ligands and suggest that mGlu1 PAMs may primarily be effective in patients carrying GRM1 mutations. Considering these exciting findings, continued interrogation is required to determine the efficacy of mGlu1 PAMs and NAMs in nonhuman primate and clinical studies.

mGlu5 also represents an exciting target for the treatment of schizophrenia and improving cognitive function in multiple brain disorders (Foster and Conn, 2017; Nicoletti et al., 2019). Extensive research supports a bidirectional interaction between mGlu5 and NMDARs, such that activation of mGlu5 receptors facilitates NMDAR function (Doherty et al., 1997; Ugolini et al., 1999; Awad et al., 2000; Attucci et al., 2001; Mannaioni et al., 2001; Pisani et al., 2001), whereas activation of NMDARs amplifies mGlu5 receptor activity by restraining receptor desensitization (Alagarsamy et al., 2005). These observations serve as a foundation for the development of mGlu5 PAMs for the treatment of schizophrenia. A number of mGlu5 PAMs have demonstrated efficacy in rodent models used to predict antipsychotic efficacy and the treatment of cognitive disturbances (Kinney et al., 2003; Lecourtier et al., 2007; Liu et al., 2008; Conn et al., 2009; Stefani and Moghaddam, 2010; Vardigan et al., 2010; Gastambide et al., 2013; Horio et al., 2013; Nicoletti et al., 2019). However, one caveat of mGlu5 PAMs includes excitotoxicity mediated by the enhanced NMDAR activity, as high doses of mGlu5 receptor PAMs have been shown to induce seizures and neurotoxicity in rodents (Parmentier-Batteur et al., 2014; Rook et al., 2015; Conde-Ceide et al., 2016). To circumvent these adverse side effects, biased mGlu5 receptor PAMs have been developed to amplify receptor function without recruiting NMDA receptors (Rook et al., 2013, 2015). Interestingly, these biased mGlu5 PAMs have robust efficacy in animal models without potentiating NMDA receptor signaling (Rook et al., 2015; Gould et al., 2016), suggesting that efficacy of these compounds is not mediated by potentiation of NMDA receptor currents. Thus, leveraging this biased mGlu5 PAM, VU0409551, Rook et al. have shown antipsychotic-like activity and precognitive efficacy without activating NMDA receptors and without inducing the adverse effects of nonbiased mGlu5 PAMs (Rook et al., 2015).

II. Group II: Metabotropic Glutamate2/3

Based on evidence that activation of group II mGlu receptor subtypes, mGlu2 and mGlu3, produce robust antipsychotic-like effects in preclinical models (Chaki et al., 2004; Pilc et al., 2008; Conn et al., 2009; Dhanya et al., 2014; Muguruza et al., 2016), longstanding efforts have been aimed at optimizing mGlu2 and mGlu3 agonists for the treatment of schizophrenia. Despite group II mGlu receptor agonists showing efficacy in improving positive and negative symptoms in an initial phase II trial (Patil et al., 2007), larger clinical studies did not demonstrate significant efficacy of these compounds compared with placebo (Kinon et al., 2011). The development of highly selective allosteric modulators of mGlu2 and mGlu3 have allowed for delineation of the role of these receptor subtypes in the physiologic and behavioral deficits associated with schizophrenia (Table 3) with hopes of developing improved treatments for schizophrenia. Interestingly, preclinical studies have alluded to mGlu2 activation being sufficient to provide therapeutic benefit, such that the antipsychotic-like activity of group II mGlu receptor agonists are absent in mGlu2, but not mGlu3, receptor knockout mice (Spooren et al., 2000). These observations have been bolstered by the discovery of mGlu2-selective PAMs, which allowed for direct interrogation of this hypothesis. Several mGlu2 PAMs demonstrated antipsychotic-like efficacy as well as precognitive and social effects in multiple preclinical models (Galici et al., 2005, 2006; Harich et al., 2007; Nikiforuk et al., 2010; Griebel et al., 2016). For example, administration of the mGlu2 PAM, potassium 3′-([(2-cyclopentyl-6-7-dimethyl-1-oxo-2,3-dihydro-1H-inden-5-yl)oxy]methyl)biphenyl l-4-carboxylate (BINA), reduced dimethoxy-bromoamphetamine–induced head twitches and reversed MK-801-induced immobility in the forced swim test (FST) (Benneyworth et al., 2007; Kawaura et al., 2016). Additionally, (2S)-5-methyl-2-{[4-(1,1,1-trifluoro-2-methylpropan-2-yl)phenoxy]methyl}-2,3-dihydroimidazo[2,1-b][1,3]oxazole-6-carboxamid (TASP0443294), another mGlu2 PAM, showed efficacy in reducing methamphetamine and NMDAR antagonist-induced hyperlocomotion as well as reversed social memory deficits induced by MK-801 administration in mice (Hikichi et al., 2015; Lavreysen et al., 2015). Two primary mGlu2 PAMs moved forward to clinical testing; however, the mGlu2 PAM AZD 8529 did not significantly improve positive or negative symptoms in patients with schizophrenia when administered as a monotherapy (Litman et al., 2016). Alternatively, another mGlu2 PAM, JNJ-40411813/ADX71149, showed beneficial effects in patients with residual negative symptoms (Hopkins, 2013) and potential efficacy in reversing select cognitive deficits and negative symptoms after administration of ketamine in healthy volunteers (Salih et al., 2015). However, negative phase 2 schizophrenia clinical trials with JNJ-40411813/AZD8529 has tempered expectations on the utility of mGlu2 PAMs for the treatment of schizophrenia. The ongoing efficacy of this compound in large-scale clinical trials remains to be seen.

TABLE 3.

Summary of preclinical efficacy of group II mGlu receptor allosteric modulators in schizophrenia-related deficits

Receptor Type Compound Positive Symptom Models Negative Symptoms Models Cognitive Models References
mGlu2 PAM LY487379 Reduced NMDAR-HL and AHL;a attenuated AMPH but not PCP-disrupted PPIa Reduced PCP-induced deficits in social interactionb Promoted cognitive flexibility in ASSTc (See footnotes.)
BINA Reduced NMDAR-HL;d,f no effect on AHL;d reduced PCP-disrupted PPI;d reduced DOB-induced head twitchese Reduced MK-801-induced increased immobility in the FSTf (See footnotes.)
TASP0443294 Reduced MHLg and NMDAR-HLh Rescued MK-801-induced social memory deficitsg (See footnotes.)
JNJ-40411813/ADX71149 Reduced NMDAR-HL; no effect on AHL; inhibited DOM-induced head twitches Lavreysen et al., 2015
SAR 218645 No effect on NMDAR-HL or AHL; no effect on hyperactivity in DAT−/− and NR1neo −/− mice; reduced DOI-induced head twitches Reversed MK-801-induced deficits in NOR; attenuated working memory deficits in Y-maze test in NR1neo−/− mice Griebel et al., 2016
mGlu3 NAM VU0477950 Dose-dependent impairment in extinction learning Walker et al., 2015
VU0650786 Blocked the ability of mGlu2/3 agonists to restore trace fear conditioning after PCP administration Dogra et al., 2021

AHL, amphetamine-induced hyperlocomotion; AMPH, amphetamine; ASST, attentional set-shift task; DAT, dopamine transporter; DOB, dimethoxy-bromoamphetamine; HL, hyperlocomotion; METH, methamphetamine; MHL, METH-induced hyperlocomotion; NOR, novel object recognition; NR1neo, NR1 subunit reduced expression.

aGalici et al., 2005.

bHarich et al., 2007.

cNikiforuk et al., 2010.

dGalici et al., 2006.

eBenneyworth et al., 2007.

fKawaura et al., 2016.

gHikichi et al., 2015.

hLavreysen et al., 2015.

In addition to the wealth of research on mGlu2-selective PAMs, numerous studies have indicated a relationship between decreased performance PFC-dependent cognitive tasks and single nucleotide polymorphisms in the human gene encoding mGlu3 (GRM3) (Egan et al., 2004; Tan et al., 2007; Harrison et al., 2008), identifying GRM3 as a risk locus for schizophrenia in genome-wide association studies (Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2014). After the discovery of highly selective mGlu3 NAMs, studies in mice revealed that mGlu3 mediates distinct aspects of PFC synaptic plasticity and procognitive effects in rodents (Walker et al., 2015; Dogra et al., 2021). In agreement with these findings, studies in nonhuman primates have demonstrated that mGlu2/3 agonists elicit postsynaptic effects in the dorsolateral PFC that improve cognitive function (Jin et al., 2017). Together, these studies highlight mGlu3 as a novel regulator of PFC-mediated cognitive processes. Additional investigation using current and next-generation group II mGlu receptor PAMs/NAMs is essential for expanding our understanding of these receptors and their potential utility in treating schizophrenia.

III. Group III: Metabotropic Glutamate4/6/7/8

Like group II mGlu receptors, the therapeutic promise of group III mGlu receptors arose from evidence of their ability to improve the hyperglutamatergic state believed to take place in schizophrenia and modulate behavioral processes dysregulated in the disease, including cognition and motivation. For instance, mice lacking mGlu4 display impairments in spatial reversal and long-term memory (Gerlai et al., 1998), suggesting a critical role of mGlu4 in cognitive flexibility and associative learning, both of which are known to be impaired in patients with schizophrenia. Previous studies also suggest that mGlu4 activation may elicit antipsychotic-like effects in rodent models. In one study, the group III agonist (1S,3R,4S)-1-aminocyclopentane-1,3,4-tricarboxylic acid (ACPT-I) reduced PCP- and amphetamine-induced hyperlocomotion in addition to head twitching in response to 2,5-Dimethoxy-4-iodoamphetamine (DOI) (Pałucha-Poniewiera et al., 2008). Similar actions of ACPT-I administration are also observed with mGlu4-selective orthosteric agonists, LSP 1-2111 (Wierońska et al., 2012a) and LSP4-2022 (Woźniak et al., 2016). Activation of mGlu4 receptors with these compounds also improves behavioral deficits associated with negative and cognitive symptoms of schizophrenia (Wierońska et al., 2012a; Woźniak et al., 2016). More recently, mGlu4 PAMs Lu AF 21934 (Bennouar et al., 2013), Lu AF32615 (East et al., 2010), and ADX88178 (Le Poul et al., 2012) have also displayed similar outcomes in models of representing all three symptom clusters of schizophrenia (Table 4). For example, ADX88178, an mGlu4 PAM, was shown to reduce NMDAR antagonist-induced hyperlocomotion and DOI-induced head twitches while also reducing immobility time in FST (Kalinichev et al., 2014). Alternatively, the mGlu4 PAMs, Lu AF21934 and Lu AF32615, showed efficacy in restoring social interaction, reducing hyperlocomotion induced by amphetamine and NMDAR antagonists, and rescuing MK-801-induced deficits in spatial working memory and novel object recognition (Wierońska et al., 2012a; Sławińska et al., 2013a). Together, these studies highlight the potential therapeutic utility of selective mGlu4 activators for schizophrenia.

Despite limited studies focusing on the mGlu7 receptor subtype as a potential therapeutic target for schizophrenia, a polymorphism in the GRM7 gene encoding mGlu7 that decreased transcription in vitro was positively correlated with schizophrenia in a large Japanese cohort (Ohtsuki et al., 2008). These findings suggest that hypofunction of mGlu7 may contribute to the pathophysiology of schizophrenia. Because activation of mGlu7 reduces glutamatergic neurotransmission (Baskys and Malenka, 1991; Ayala et al., 2008) and acts as a heteroreceptor to modulate GABA release and the induction of LTP in brain regions, such as the hippocampus (Klar et al., 2015), it is plausible that selective activators of mGlu7 may enhance aspects of hippocampal-dependent cognitive function. However, some preclinical studies refute this hypothesis such that the mGlu7 allosteric agonist N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) exacerbates MK-801-induced hyperlocomotion (Mitsukawa et al., 2005) and DOI-induced head twitches (Wierońska et al., 2012a). Although these findings may be driven by off-target effects of AMN082 in vivo (Sukoff Rizzo et al., 2011), it has been shown that the psychotic-enhancing effects were occluded in mGlu7 knockout (KO) mice (Wierońska et al., 2012a), implying that they may be mGlu7 receptor-dependent. At this time, future studies must further confirm if the use of selective PAMs may show more promise of mGlu7 activation in schizophrenia-related models.

Although expressed at fairly low levels in the brain, the mGlu8 receptor subtype is expressed in the presynaptic active zone of mainly glutamatergic synapses (Kinoshita et al., 1996a; Shigemoto et al., 1997) where it functions to modulate neurotransmitter release and gates glutamatergic transmission into the hippocampus (Zhai et al., 2002). In line with this function, mGlu8 KO mice display deficits in hippocampal-dependent learning (Gerlai et al., 2002), suggesting that activating mGlu8 with selective ligands could treat the cognitive impairments in patients with schizophrenia. In studies determining the antipsychotic efficacy of mGlu8-targeting ligands, two studies have found that the relatively selective orthosteric mGlu8 agonist (S)-3,4-dichlorophenyl glycine ((S)-3,4-DCPG) was unable to reverse amphetamine or PCP-induced hyperactivity in Sprague-Dawley rats (Thomas et al., 2001; Robbins et al., 2007). Additionally, mGlu8 KO mice do not display significant deficits in PPI of acoustic startle; thus, it appears to be unlikely that mGlu8 is a promising target for a novel antipsychotic (Robbins et al., 2007). Despite lacking evidence for antipsychotic efficacy, continued studies are required to determine the utility of mGlu8-targeting compounds as cognitive enhancers.

B. Substance Use Disorders

SUD is a multifaceted chronically relapsing disorder characterized by excessive drug intake, repeated unsuccessful attempts to reduce or stop drug use, enhanced drug-seeking and self-administration, the emergence of drug tolerance and withdrawal, and continued drug intake despite negative consequences (Koob and Volkow, 2010). SUD represents a serious public health problem with devastating consequences to society. However, there is a lack of pharmacological agents approved to treat the disease. Drugs of abuse exert their effects by altering the signaling of numerous neurotransmitter systems and brain circuits, which serves as a challenge for developing efficacious treatments. Ongoing research highlights detrimental adaptations in GABA and glutamate neurotransmission in SUD (Tzschentke and Schmidt, 2003; Cruz et al., 2008). Drugs of abuse alter glutamate transmission through a diverse array of mechanisms. For example, cocaine increases glutamate transmission indirectly through dopamine transporter-mediated dopamine release (Ritz et al., 1987). Alternatively, it has been reported that alcohol inhibits postsynaptic NMDAR- and non-NMDAR-mediated glutamate transmission and release, possibly via inhibition of GABA interneurons (Lovinger et al., 1989, 1990; Carta et al., 2003; Hendricson et al., 2003, 2004). Notably, imbalance of glutamate and GABA systems in brain regions, such as the PFC, is associated with physiologic and behavioral aspects of SUDs, including impulsivity, reinforcement learning, and executive function. For instance, a recent clinical study using hydrogen (1H)-magnetic resonance spectroscopy revealed significantly higher glutamate levels and lower GABA levels in patients with opioid use disorder compared with healthy controls (Li et al., 2020). Additionally, this study showed that higher impulsivity and cognitive impairment were associated with lower GABA and higher glutamate levels. Furthermore, exposure to pharmacological agents that block glutamate transmission attenuate the reinforcing effects of drugs of abuse. For instance, systemic administration of NMDAR antagonists attenuates self-administration of alcohol (Shelton and Balster, 1997), cocaine (Pierce et al., 1997; Pulvirenti et al., 1997; Hyytiä et al., 1999; Blokhina et al., 2005), and nicotine (Kenny et al., 2009). Therefore, restoring GABA and glutamate balance in PFC, among other brain regions, represents a promising therapeutic strategy for treating SUD.

Numerous medications targeting GABA and/or glutamate receptors have been under longstanding investigation for the treatment of SUD, including baclofen, topiramate, and gabapentin. However, to date, there has been mixed evidence for their efficacy in clinical trials for various types of SUDs, including alcohol, nicotine, cocaine, and methamphetamine (Addolorato et al., 2012). One of the well-studied candidates, baclofen, a GABAB agonist, has yielded findings in preclinical and clinical studies suggesting its potential utility in treating alcohol use disorder. Baclofen acts presynaptically to hyperpolarize synaptic terminals, inhibits calcium influx, and prevents the release of the excitatory neurotransmitters glutamate and aspartate (Davidoff, 1985). Preclinical studies show that baclofen effectively mitigates the reinforcing properties of alcohol in addition to suppressing acquisition and maintenance of alcohol drinking behavior and relapse-like drinking in rats and mice (Cousins et al., 2002; Maccioni and Colombo, 2009). Alternatively, baclofen has also been tested as a treatment of cocaine use disorder. A human brain imaging study found that baclofen reduces the activation of limbic brain regions that occurs in response to cocaine-related cues (Brebner et al., 2002). Baclofen has undergone numerous clinical trials for the treatment of cocaine use disorder; however, these studies have yielded mixed results with the first human open-label study showing a trend toward reduced cocaine craving and self-reported cocaine consumption (Ling et al., 1998), but subsequent, larger studies have not identified statistically significant effects of baclofen on craving or cocaine intake (Shoptaw et al., 2003). In addition to the studies detailed here, baclofen and other GABA-targeting drugs, such as gabapentin, have been tested for efficacy in treating nicotine and methamphetamine use disorders. Together, these studies shed light on the potential utility of activating GABA systems for the treatment of SUDs; however, more effective compounds are required to better achieve this goal.

A wealth of studies using ligands targeting mGlu receptors has demonstrated the utility of targeting these receptors as an alternative approach to mitigate SUD-induced imbalances in glutamate and GABA signaling. For example, systemic administration of LY379268, an mGlu2/3 orthosteric agonist, decreases self-administration of cocaine (Baptista et al., 2004; Adewale et al., 2006; Xi et al., 2010), nicotine (Liechti et al., 2007), and alcohol (Bäckström and Hyytiä, 2005; Sidhpura et al., 2010). However, excessive glutamate release, a potential consequence direct activation of mGlu receptors, including mGlu5, elicits excitotoxicity via NMDAR-mediated mechanisms, limiting the utility of orthosteric mGlu-targeting compounds (Reiner and Levitz, 2018). Thus, allosteric modulators of mGlu receptors represent a novel avenue for restoring homeostasis of GABA and glutamate systems while exerting receptor subtype selects effects and circumventing EPS.

I. Group I: Metabotropic Glutamate1/5

Group I mGlu receptor subtypes, mGlu1 and mGlu5, have been extensively studied in terms of behavioral effects in animal models of SUD. Both mGlu1 and mGlu5 are robustly expressed in brain regions known to be critical to SUD pathophysiology, including the NAc, dorsal striatum, ventral midbrain and PFC (Niswender and Conn, 2010). Repeated exposure to drugs of abuse can dysregulate mGlu1 and mGlu5 expression and function. For example, chronic alcohol consumption in rodents reduces mGlu1/5 mRNA levels in various subregions of the hippocampus and increases mGlu1/5 expression in the NAc core and central nucleus of the amygdala (Simonyi et al., 2004; Obara et al., 2009). Nicotine exposure increases expression of mGlu1 mRNA in the ventral tegmental area (VTA) and amygdala (Kane et al., 2005). Further, repeated cocaine exposure disrupts mGlu1 receptor-mediated signaling in the NAc (Swanson et al., 2001). Finally, prolonged withdrawal from extended-access cocaine self-administration decreases total protein and surface expression levels of mGlu1 in the NAc compared with drug-naïve rats (Loweth et al., 2014). Alternatively, the total and surface levels of mGlu1 is unchanged in the NAc following shorter abstinence periods or during cocaine administration (Ary and Szumlinski, 2007; Loweth et al., 2014). Withdrawal time-dependent reductions in mGlu1/5 expression within ventromedial PFC have also been reported for following extended-access cocaine self-administration following extinction testing compared with drug-naïve animals (Ben-Shahar et al., 2013). Additionally, extinction of cocaine-seeking decreases the surface expression of mGlu5 receptors in the NAc (Knackstedt et al., 2010). Thus, there is extensive evidence that mGlu1 and mGlu5 receptor subtypes may be involved in drug-related behaviors and the pathophysiology of SUDs.

Notably, mGlu1 and mGlu5 have both also been implicated as regulators of drug self-administration behavior. Early studies reported that mice carrying a null mutation for the gene encoding the mGlu5 receptor lack cocaine-induced hyperlocomotion and did not acquire intravenous self-administration of cocaine (Chiamulera et al., 2001). Furthermore, genetic inactivation or pharmacological inhibition of mGlu5 receptors decreases self-administration of alcohol, cocaine, heroin, nicotine, methamphetamine and ketamine and reduces breakpoints for reinforcement for various drugs of abuse in a progressive ratio (PR) paradigm (Cleva and Olive, 2012). mGlu1 antagonist JNJ16259685 reduces alcohol self-administration and breakpoints for alcohol reinforcement under a PR schedule (Besheer et al., 2008a,b). However, other studies have shown that acquisition and fixed ratio operant responding for psychostimulants, such as cocaine and methamphetamine, is intact in mice lacking mGlu5−/−; however, deletion of mGlu5 enhanced responding on a progressive ratio schedule and impaired extinction of drug-seeking behaviors (Chesworth et al., 2013; Bird et al., 2014). These findings suggest that mGlu5 may play distinct roles in drug reinforcement and instrumental extinction learning. Furthermore, mGlu1 and mGlu5 play an important role in reinstatement of drug seeking. Numerous studies have reported that pharmacological blockade of mGlu5 attenuates the reinstatement of drug-seeking behavior induced by drug-associated cues, stress and drug priming (Bespalov et al., 2005; Bäckström and Hyytiä, 2007; Platt et al., 2008; Schroeder et al., 2008; Kumaresan et al., 2009; Martin-Fardon et al., 2009). Additionally, mGlu1 antagonism attenuates the reinstatement of nicotine and cocaine-seeking behavior elicited by drug-associated cues (Dravolina et al., 2007). A wealth of additional research supports the roles of mGlu1 and mGlu5 in various other drug-related behaviors, including conditioned place preference and interoceptive drug effects. These findings have been elegantly detailed in reviews elsewhere (Olive, 2009; Cleva and Olive, 2012; Caprioli et al., 2018; Niedzielska-Andres et al., 2021).

The extensive evidence that mGlu1 and mGlu5 play critical roles in behavioral deficits associated with SUD raises the question of whether targeting group I mGlu receptors is a promising therapeutic approach for the treatment of SUD. The development of mGlu1 and mGlu5 allosteric modulators have allowed researchers to directly test this question. Studies have investigated the efficacy of mGlu1 allosteric modulators within the context of SUD (Caprioli et al., 2018). These studies have been summarized in Table 5. Specifically, repeated administration of selective mGlu1 PAMs, SYN119 or Ro0711401, blocks incubation of cue-induced cocaine craving following extended-access cocaine self-administration and prolonged withdrawal in rats (Loweth et al., 2014). Interestingly, administration of mGlu1 PAMs during withdrawal from methamphetamine self-administration did not block incubation of methamphetamine craving. The efficacy of mGlu1 PAMs in reducing incubation of cocaine craving may be driven by mGlu1-mediated blockade of calcium permeable amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor accumulation and synaptic transmission, which is known to be a critical mechanism driving cue-induced drug craving and cocaine seeking (McCutcheon et al., 2011; Loweth et al., 2014; Ruan and Yao, 2021). However, there is evidence that mGlu1 does not regulate calcium permeable amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor accumulation during methamphetamine withdrawal (Murray et al., 2019), further supporting divergent mechanisms underlying incubation of craving for psychostimulants and selective roles of mGlu1. These studies leveraging mGlu1 PAMs provide further support that selective activation of mGlu1 is able to reduce the impact of drug-induced adaptation in mGlu1 function, which drives cocaine-seeking behavior (Halbout et al., 2014) and modulates a number of critical forms of synaptic plasticity, such as cocaine-induced plasticity in the VTA (Mameli et al., 2009) and mGlu1-LTD and synaptic potentiation in the PFC (Ruan and Yao, 2021).

TABLE 5.

Summary of preclinical efficacy of group I mGlu receptor allosteric modulators in substance use disorder models

Receptor Type Compound Drug of Abuse Behavioral Effect References
mGlu1 PAM SYN119/Ro0711401 Cocaine Blocks incubation of cue-induced cocaine craving after extended-access self-administration Loweth et al., 2014
Methamphetamine No effect on incubation of methamphetamine craving after extended-access self-administration and withdrawal Murray et al., 2019
NAM JNJ-16259685 Alcohol Decreased responding under FR/PR schedules Besheer et al., 2008a,b
Cocaine and methamphetamine Decreased self-administration under a second-order reinforcement schedule of reinforcement (FI5-FR10) Achat-Mendes et al., 2012
CPCCOEt Alcohol Decreased self-administration under FR and PR schedules of reinforcement in alcohol-preferring P-rats or C57BL/6J mice;a,b,c reduced ethanol reinforcement, consumption, and expression of ethanol CPPd 1
(See footnotes.)
EMQMCM Nicotine Inhibited cue and nicotine-induced reinstatement of nicotine-seeking behavior Dravolina et al., 2007
Morphine and cocaine Inhibited the expression of locomotor sensitization Dravolina et al., 2006; Kotlinska and Bochenski, 2007
YM298198 Cocaine Decreased cocaine-primed reinstatement of cocaine seeking Schmidt et al., 2015
mGlu5 NAM MPEP/MTEP Cocaine, nicotine, heroin, and alcohol Attenuates self-administration (intravenous or oral) Ombelet et al., 1994; Kenny et al., 2003, 2005; Paterson et al., 2003; Bäckström et al., 2004; Tessari et al., 2004; Bespalov et al., 2005; Cowen et al., 2005, 2007; Lee et al., 2005; McMillen et al., 2005; Olive et al., 2005; Hodge et al., 2006; Lominac et al., 2006; Liechti and Markou, 2007; Besheer et al., 2008b; Palmatier et al., 2008;
Cocaine Attenuated cue-induced reinstatement of cocaine seeking Bäckström and Hyytiä, 2007; Knackstedt et al., 2014; Knackstedt and Schwendt, 2016
Alcohol, cocaine, and nicotine Reduced breakpoints under PR reinforcement schedule Paterson et al., 2003; Besheer et al., 2008b
Methamphetamine Reduced incubated methamphetamine seeking; decreased self-administration under FR and PR schedules of reinforcement Gass et al., 2009; Murray et al., 2021
Partial NAM M-5MPEP/Br-5MPEPy Cocaine Decreased self-administration and attenuated the discriminative stimulus effects of cocaine Gould et al., 2016

CPP, conditioned place preference; FI, fixed interval ;FR, fixed ratio; P, preferring; SYN119, mGlu1 PAM.

aCasabona et al., 1997.

bHodge et al., 2006.

cSchroeder et al., 2005.

dLominac et al., 2006.

mGlu1 NAMs have been shown to decrease alcohol self-administration in some studies but not others. In alcohol self-administration studies in alcohol-preferring P rats, JNJ16259685, an mGlu1 NAM, decreased responding under fixed ratio and PR schedules (Besheer et al., 2008a; b), but also decreased locomotor activity and lever-pressing for sucrose, suggesting nonspecific motor effects. Similar observations were reported in additional studies looking at the effect of (−)-ethyl (7E)-7-hydroxyimino-1,7a-dihydrocyclopropa[b]chromene-1a-carboxylate (CPCCOEt), another mGlu1 NAM, on alcohol self-administration in alcohol-preferring (P)-rats or C57BL/6J mice (Casabona et al., 1997; Schroeder et al., 2005; Hodge et al., 2006). In another study, CPCCOEt reduced ethanol reinforcement, consumption, and expression of ethanol conditioned place preference while facilitating the motor-impairing effects of ethanol (Casabona et al., 1997; Lominac et al., 2006). Importantly, CPCCOEt was able to block the acute effects of ethanol on extracellular levels of dopamine and glutamate in the NAc, while potentiating the effects of acute ethanol on extracellular GABA in this region. Leveraging another mGlu1 NAM, JNJ16259685, one study reported decreased psychostimulant (cocaine and methamphetamine) administration under a second-order reinforcement schedule of reinforcement fixed interval 5-10 (FI5-FR10) (Achat-Mendes et al., 2012). In this same study, JNJ16259685 had no effect on food-reinforced responding but exerted motoric effects. The efficacy of mGlu1 NAMs in nicotine use has also been studied. The selective mGlu1 NAM EMQMCM (5 mg/kg) inhibited cue and nicotine-induced reinstatement of nicotine-seeking behavior, albeit when administered at higher doses EMQMCM reduced cue-induced reinstatement of food-seeking, indicating that high doses of this compound may have general inhibitory effects on appetitive responding (Dravolina et al., 2007). Furthermore, EMQMCM has been shown to inhibit the expression of locomotor sensitization to both morphine and cocaine (Dravolina et al., 2006; Kotlinska and Bochenski, 2007). Another study showed that intra-NAc injections of mGlu1 NAM, 6-amino-N-cyclohexyl-N,3-dimethylthiazolo[3,2-a]benzimidazole-2-carboxamide (YM298198), decreases reinstatement of cocaine seeking in rats following cocaine priming (Schmidt et al., 2015). Together, these studies highlight the potential utility of mGlu1 allosteric modulators for the treatment of various SUDs but also highlight the need for continued studies with improved formulations.

mGlu5 allosteric modulators have been of particular interest as therapeutic options for the treatment of SUDs. A summary of these studies has been included in Table 5. Due to the abundance of reports, we have focused on summarizing the effects of mGlu5 allosteric modulators on drug self-administration and reinstatement. Details on the efficacy of mGlu5 PAM/NAMs in other drug-related behaviors have been reviewed previously (Olive, 2009; Cleva and Olive, 2012; Caprioli et al., 2018). A subset of classically studied mGlu5 NAMs include the compounds 2-Methyl-6-(phenylethynyl)pyridine (MPEP) and 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine (MTEP). MPEP and MTEP have been shown to attenuate intravenous self-administration of numerous drugs of abuse, including cocaine, nicotine, methamphetamine, and heroin (Ombelet et al., 1994; Kenny et al., 2003, 2005; Paterson et al., 2003; Tessari et al., 2004; Bespalov et al., 2005; Lee et al., 2005; Liechti and Markou, 2007; Palmatier et al., 2008; Gass et al., 2009) and ethanol in a variety of rodent strains (Bäckström et al., 2004; Cowen et al., 2005; McMillen et al., 2005; Olive et al., 2005; Hodge et al., 2006; Lominac et al., 2006; Cowen et al., 2007; Besheer et al., 2008b ) without altering the reinforcing properties of natural reward or food (Paterson et al., 2003; Tessari et al., 2004; Bespalov et al., 2005; Liechti and Markou, 2007; Gass et al., 2009). These compounds have also demonstrated the ability to modulate the motivational properties of many drugs. For instance, MPEP reduces breakpoints for ethanol, cocaine, and nicotine under PR schedules of reinforcement (Paterson et al., 2003; Besheer et al., 2008b). Furthermore, administration of MTEP has been shown to reduce the reinforcing efficacy of methamphetamine as reflected by reduced breakpoints under a PR schedule off reinforcement (Gass et al., 2009). In addition to modulating the rewarding and motivational properties of drugs, MPEP and MTEP have also been shown to prevent cue and drug-induced priming of reinstatement of cocaine, nicotine or ethanol-seeking behavior (Bäckström et al., 2004; Tessari et al., 2004; Lee et al., 2005; Bäckström and Hyytiä, 2006; Iso et al., 2006; Murray et al., 2021). For instance, a recent study showed that systemic administration of MTEP reduces incubated methamphetamine seeking following self-administration and prolonged withdrawal (Murray et al., 2021), elucidating the role of mGlu5 in the incubation of methamphetamine craving and delineating distinct mechanisms from that of incubation of cocaine craving. Interestingly, cue-induced reinstatement of cocaine-seeking behavior is attenuated in response to local infusion of MPEP or MTEP into the NAc (Bäckström and Hyytiä, 2007; Knackstedt et al., 2014), while local infusion of MTEP into the dorsolateral striatum at time of context-induced relapse testing attenuated extinction learning (Knackstedt et al., 2014; Knackstedt and Schwendt, 2016). This finding was paralleled by reduced mGlu5 surface expression and LTD in brain slices of animals during prolonged abstinence from cocaine which could be reversed by bath application of the mGlu5 PAM VU-29, suggesting brain-region specific subpopulations of mGlu5 receptors may play distinct roles in regulating extinction learning and reinstatement of cocaine-seeking.

Despite the promising results of these studies, it has also been reported that MPEP and MTEP can attenuate breakpoints for food (Paterson et al., 2003), enhance the sedative properties of ethanol (Sharko and Hodge, 2008) and elicit general behavioral reduction, including decreases in inactive lever responding (Murray et al., 2021). To this end, partial mGlu5 NAMs have been developed, which feature submaximal but saturable levels of blockade and may represent an additional avenue to broaden the therapeutic window of mGlu5 NAMs. One study evaluated the efficacy of partial mGlu5 NAMs, 2-[2-(3-methoxyphenyl)ethynyl]-5-methylpyridine (M-5MPEP) and bromo-2-[2-(3-methoxyphenyl)ethynyl]-5-methylpyridine (Br-5MPEPy), in comparison with the full mGlu5 NAM MTEP in models of SUD. Gould et al., found that M-5MPEP, Br-5MPEPy, and MTEP dose-dependently decreased cocaine self-administration and attenuated the discriminative stimulus effects of cocaine, suggesting that partial mGlu5 NAM activity is sufficient to elicit therapeutic effects comparable to full mGlu5 NAMs (Gould et al., 2016). In sum, full and partial mGlu5 NAMs may represent a promising therapeutic option for the treatment of numerous SUDs; however, continued exploration of novel mGlu5 NAMs with improved therapeutic window and reduced side effects is required.

II. Group II: Metabotropic Glutamate2/3

Decades of research support a critical role of group II mGlu receptors in SUDs. mGlu2 and mGlu3 regulate neurotransmission in brain regions implicated in SUDs, including the PFC and NAc. In the PFC, mGlu2/3 receptors are tonically activated by endogenous glutamate and infusion of a selective mGlu2/3 receptor antagonist, LY341495, increases glutamate levels (Melendez et al., 2005; Xie and Steketee, 2008). Within the NAc, numerous studies suggest that endogenous glutamatergic tone on group II mGlu receptors regulating both glutamate and dopamine levels. In vivo microdialysis studies have demonstrated increased glutamate release after perfusion of selective antagonist LY143495 into the NAc and decreased extracellular glutamate levels in response to agonist (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (APDC) (Xi et al., 2002). Further, electrophysiological recordings from NAc slices reveal that mGlu2/3 receptors can act as presynaptic autoreceptors to control glutamate release. Specifically, increased paired pulse ratios and reduced miniature EPSC frequency were observed after bath application of selective agonists (S)-4-carboxy-3-hydroxyphenylglycine ((1S,3S)-ACPD) and (2S,1’S,2’S)-2-(2’-carboxycyclopropyl)glycine (L-CCG1) (Manzoni et al., 1997). Evidence also supports that mGlu2/3 receptors regulate glutamate release in VTA (Manzoni and Williams, 1999), bed nucleus of stria terminalis (Grueter and Winder, 2005), and hippocampus (Capogna, 2004), among other brain regions within the motivational circuit (Poisik et al., 2005).

The expression and function of mGlu2/3 receptor subtypes are also altered by chronic use of drugs, such as alcohol, cocaine, opioids, and nicotine. The effects of drugs of abuse on mGlu2/3 receptor function have been detailed in greater detail previously (Moussawi and Kalivas, 2010). In brief, it has been reported that ethanol-dependent rats have decreased PFC GRM2 mRNA levels and mGlu2/3 autoreceptor function in the NAc shell compared with control rats (Meinhardt et al., 2013). In addition, drinking-induced decreases in mGlu2/3 autoreceptor function have also been observed in the VTA (Ding et al., 2017). Alternatively, enhanced physiologic and behavioral sensitivity to mGlu2/3 agonists is observed in the central nucleus of the amygdala and bed nucleus of stria terminalis of ethanol-dependent rats (Kufahl et al., 2011). In addition to sensitivity to the effects of ethanol, mGlu2/3 receptors are altered by chronic exposure to other drugs of abuse. A recent study revealed that 12 days cocaine self-administration followed by 6 to 10 days of extinction training resulted in a decreased in mGlu2 expression in the NAc core of male and female rats (Logan et al., 2020). Further, the inhibitory effects of mGlu2/3 receptors on excitatory transmission in the VTA and NAc are enhanced in rodents during early withdrawal from chronic morphine (Manzoni and Williams, 1999; Martin et al., 1999). Interestingly, genetic variation in the mGlu2 gene, Grm2, has also been associated with alcohol preference and consumption in rodents, such that the presence of an abnormal stop codon preventing expression of the Grm2 results in increased alcohol preference and intake as measured by a two-bottle choice paradigm (Zhou et al., 2013; Wood et al., 2017).

Notably, mGlu2/3 receptor-dependent plasticity is impaired after exposure to drugs of abuse. For example, chronic morphine impairs mGlu2/3 receptor induced LTD at excitatory synapses in NAc medium spiny neurons (Robbe et al., 2002) and chronic cocaine exposure impairs mGlu2/3 receptor-dependent LTD in PFC pyramidal cells (Huang et al., 2007). More recently, it was shown that NAc LTP induced by high frequency stimulation of the PFC was abolished after withdrawal from self-administered cocaine via reduced mGlu2/3 receptor stimulation (Moussawi et al., 2009). Given the importance of neuroplasticity in cognition, reinforcement learning, and updating behaviors after changes in environmental contingencies (Malenka and Bear, 2004; Whitlock et al., 2006; De Roo et al., 2008) and evidence of drug-induced plasticity impairments, mGlu2/3 receptors may be a critical target underlying drug-induced deficits in synaptic plasticity. Thus, potentiating the function of mGlu2/3 may represent a promising approach to mitigate drug intake and cognitive deficits in individuals with SUD by restoring neurotransmitter homeostasis and neuroplasticity.

Early studies using the prototypical group II mGlu agonist LY379268 demonstrate decreased reinstatement of alcohol, cocaine, methamphetamine, and heroin seeking induced by cues previously associated with drug self-administration (Acri et al., 2017). Administration of LY379268 has also been shown to decrease cue- and drug priming-induced reinstatement of cocaine self-administration in nonhuman primates (Adewale et al., 2006; Justinova et al., 2016) and incubation of cocaine, methamphetamine, or nicotine self-administration in rats (Liechti et al., 2007; Crawford et al., 2013). Although less abundant than studies on group I mGlu receptor PAM/NAMs, a few studies have looked at the efficacy of mGlu2/3 allosteric modulators in behavioral models of SUD (Table 6). One study used the selective and brain penetrant mGlu2 PAM BINA in a model of intravenous cocaine self-administration and cocaine-seeking behavior in rats that had short (1 h, ShA) or long (6 h, LgA) access to cocaine. In this study, BINA decreased cocaine self-administration in both ShA and LgA rats, with no effect on food self-administration (Jin et al., 2010). Additionally, this study showed that BINA decreased cue-induced reinstatement of cocaine seeking without altering food seeking, suggesting that mGlu2 allosteric modulators may have potential as treatments for cocaine use disorder and possibly other drugs of abuse.

TABLE 6.

Summary of preclinical efficacy of group II/III mGlu receptor allosteric modulators in substance use disorder models

Group Receptor Type Compound Drug of Abuse Behavioral Effect References
II mGlu2 PAM BINA Cocaine Decreased cocaine self-administration in both short-access (1 h) and long-access (6 h) in rats; decreased cue-induced reinstatement of cocaine seeking without altering food seeking Jin et al., 2010
AZD8418/AZD8529 Nicotine Deceased nicotine self-administration and blocked cue-induced reinstatement of nicotine- and food-seeking behavior Li et al., 2016
III mGlu7 Ago-PAM AMN082 Cocaine Reduced self-administration under an FR2 schedule of reinforcement; lowered PR breakpoints in rats Li et al., 2009, 2013
Cocaine Attenuates cocaine-primed reinstatement of cocaine-seeking behavior Li et al., 2010
Alcohol and heroin Inhibited self-administration and preference Salling et al., 2008; Bahi et al., 2012

FR, fixed ratio; PR, progressive ratio

mGlu2 PAMs, AZD8418 and AZD8529, underwent preclinical and clinical evaluation for their efficacy in nicotine use disorder. Acute treatment with AZD8418 (0.37, 1.12, 3.73, 7.46, and 14.92 mg/kg) and AZD8529 (1.75, 5.83, 17.5, and 58.3 mg/kg) deceased nicotine self-administration and blocked cue-induced reinstatement of nicotine- and food-seeking behavior but did not significantly affect food-maintained responding in rats (Li et al., 2016). Chronic treatment with AZD8418 attenuated nicotine self-administration but resulted in tolerance to this effect. The inhibitory effects of chronic AZD8529 administration on nicotine self-administration persisted throughout the 14 days of treatment; however, chronic treatment with these PAMs inhibited food self-administration. The mGlu2 PAM AZD8529 has since been tested in clinical trials including a 19-week, multicenter, randomized, phase 2 clinical study comparing the efficacy of two different doses of AZD8529 (1.5 and 40 mg) in smoking cessation in female smokers. However, this trial was completed in 2017 and reported only ∼10% of either the low- or high-dose AZD8529 groups meeting the primary outcome of abstinence during the course of the 13-week study (Lassi et al., 2016).

III. Group III: Metabotropic Glutamate4/6/7/8

The group III mGlu receptor subtypes have also garnered attention as potential targets for the treatment of SUD. A multitude of studies have reported group III mGlu receptor subtypes are sensitive to the effects of drugs of abuse, including psychostimulants and alcohol. For instance, repeated amphetamine exposure increases mGlu8 mRNA levels in the dorsal striatum and NAc (Parelkar and Wang, 2008). Chronic alcohol consumption has been shown to reduce mGlu7 mRNA levels in numerous hippocampal subregions, whereas mGlu8 mRNA levels were unchanged (Simonyi et al., 2004). Additionally, expression levels of the gene encoding the mGlu7 receptor is associated with higher levels of alcohol consumption (Vadasz et al., 2007). The development of selective compounds targeting group III mGlu receptor subtypes have allowed for delineation of the distinct roles of these receptors in the physiologic and behavioral effects of drugs of abuse. For example, treatment with AMN082, a selective mGlu7 allosteric agonist, attenuates cocaine-induced decreases of ventral pallidum GABA release in both naïve rats and cocaine self-administering rats (Li et al., 2009). These findings suggest a novel role of mGlu7 receptors in regulating the effects of cocaine on NAc-ventral pallidum GABA transmission, which is one mechanism proposed to underlie the rewarding and motivational effects of cocaine. Furthermore, microinjection of L-AP4, a nonselective agonist of group III mGlu receptors, into the dorsal striatum reduced amphetamine or cocaine-induced hyperlocomotion in rats (Mao and Wang, 2000). Importantly, L-AP4 attenuated amphetamine-stimulated dopamine release in the dorsal striatum (Mao et al., 2000), suggesting that group III mGlu receptors may be involved in the acute effects of psychostimulant exposure by inhibiting dopamine release. Striatal glutamate has long been recognized to facilitate dopamine release (Mao et al., 2000). Thus, inhibition of glutamate release by group III autoreceptors may result in this inhibition of dopamine release. A study by Xi et al., showed that L-AP4 decreased extracellular glutamate levels, whereas the group III receptor antagonist (R, S)-α-methylserine-O-phosphate increased extracellular glutamate levels in the NAc of rats, respectively (Xi et al., 2003). In addition to the presynaptic roles of group III mGlu receptors in regulating drug-associated neurotransmission, mGlu7 is expressed postsynaptically on both striatopallidal and striatonigral medium spiny neurons (Kosinski et al., 1999). To this end, L-AP4 inhibits evoked synaptic responses in the NAc, in part, through a postsynaptic mechanism (Martin et al., 1997). This putative postsynaptic mechanism likely works in concert with group III mGlu receptor subtype-mediated presynaptic modulation to control synaptic responses to drugs of abuse like cocaine.

Importantly, group III mGlu receptors have been shown to be important in drug self-administration in preclinical studies in rodents. For instance, Blednov and Harris demonstrated that mGlu4 knockout mice showed normal levels of ethanol consumption but are devoid of a locomotor stimulant effect of low doses of alcohol (Blednov and Harris, 2008). Furthermore, administration of the mGlu8 agonist (S)-3,4-DCPG suppressed alcohol self-administration and cue-induced reinstatement of alcohol-seeking behavior (Bäckström and Hyytiä, 2005). The mGlu7 subtype has also been established to be critical to drug self-administration and reinstatement of drug-seeking. The development of group III mGlu selective ligands and allosteric modulators has allowed for rigorous characterization of their roles in various SUDs (Table 6). Stimulation of presynaptic mGlu7 receptors with AMN082 significantly reduced cocaine self-administration under a fixed ratio 2 (FR2) schedule of reinforcement and lowered PR breakpoints for cocaine self-administration in rats (Li et al., 2009, 2013). These effects were replicable when AMN082 was directly infused the NAc or ventral pallidum. Additionally, systemic administration of AMN082 has been shown to attenuate cocaine-primed reinstatement of cocaine-seeking behavior (Li et al., 2010). Consistent with the literature on cocaine self-administration, AMN082 administration has also been shown to significantly inhibit heroin and ethanol self-administration and preference in rodents (Salling et al., 2008; Bahi et al., 2012). Together, these findings provide evidence that the mGlu7 receptor is a promising target for the treatment of SUD. Continued studies leveraging using AMN082 or mGlu7 PAMs are required to further evaluate their efficacy novel pharmacotherapies in nonhuman primates and clinical studies.

C. Stress-Related Disorders

Stress-related disorders, including anxiety, are incredibly pervasive psychiatric conditions and represent an enormous worldwide health concern. Chronic psychosocial stressors have been implicated as some of the most common risk factors for the development of stress-related disorders, which also have high comorbidity with other neuropsychiatric diseases, including depression and SUDs (Risch et al., 2009; Duric et al., 2016). However, our understanding of the mechanisms driving the development and persistence of stress-related disorders is still unclear. Stress-related disorders have been associated with aberrant brain excitability within critical neural circuits and disruption of excitatory and inhibitory transmission has been increasingly implicated as a crucial determinant of the pathophysiology of these diseases (Nuss, 2015; Jie et al., 2018). Mood and stress-related disorders involve both bottom-up and top-down control, primarily by limbic regions of the brain. As such, exposure to various stressors is known to dysregulate transmission of both glutamatergic and GABAergic systems (Nemeroff, 2003; Popoli et al., 2011; Jie et al., 2018). For example, clinical studies using proton magnetic resonance spectroscopy to measure endogenous brain metabolites, such as glutamate, in the brain have demonstrated a positive correlation between frontal cortex glutamate levels and state anxiety levels in healthy subjects (Grachev and Apkarian, 2000). Additionally, patients with social anxiety show higher glutamate levels in brain regions, such as the anterior cingulate cortex, compared with healthy control subjects who positively correlated with the severity of their social anxiety symptoms (Phan et al., 2005). These findings, among many others, suggest that restoring the balance between glutamatergic and GABAergic transmission represents a promising therapeutic strategy for alleviating symptoms of stress-related disorders.

A wide variety of pharmacotherapeutics targeting glutamate and/or GABA systems have been under ongoing investigation for their efficacy in treating stress-related disorders, including ketamine, memantine, gabapentinoids, tiagabine, valproic acid, and topiramate (Nasir et al., 2020). For example, the NMDA antagonist ketamine, which first showed efficacy in treating symptoms of depression in 2000 (Berman et al., 2000), has shown some efficacy in the treatment of post-traumatic stress disorder and anxiety disorders (Feder et al., 2014; Liriano et al., 2019; Banov et al., 2020). In addition, benzodiazepines, which act on GABAA receptors, have historically been used for the treatment of anxiety and other stress-related disorders. However, their utility has been limited by adverse side effects and high abuse liability (Tan et al., 2011). Therefore, particular interest has been focused on the development of subtype-selective drugs that will achieve specific therapeutic benefits by balancing glutamate and GABA transmission while limiting undesirable side effects. To this end, the mGlu receptors are in prime locations within these brain regions and neural circuits to normalize excitatory/inhibitory transmission, thus modulating stress responses and serving as a promising therapeutic approach for the treatment of stress-related disorders.

I. Group I: Metabotropic Glutamate1/5

Group I mGlu receptors subtypes have been strongly implicated in the pathophysiology of stress-related disorders. For example, clinical studies leveraging positron emission tomography (PET) imaging have demonstrated a strong association between the mGlu5 receptor subtype and anxiety, obsessive compulsive disorder, and depression (Terbeck et al., 2015). Positive correlations have been reported between mGlu5 binding in regions of the amygdala, anterior cingulate cortex, and medial orbitofrontal cortex and obsessive compulsive disorder severity as assessed by the Yale-Brown Obsessive-Compulsive Scale (Akkus et al., 2014). Furthermore, mice exposed to social isolation stress exhibited selectively reduced mGlu1 levels in the PFC (Ieraci et al., 2016). Prenatal stress models have also shown robust changes in mRNA and protein levels as well as gene methylation levels of mGlu1 and mGlu5 receptor subtypes expressed in the hippocampus of offspring rats that exhibit depression-like behavior (Lin et al., 2018). Several studies leveraging genetic deletion or pharmacological manipulation of group I mGlu receptors subtypes have further substantiated the notion that these receptors may be viable targets for treating stress-related disorders (Li et al., 2006; Shin et al., 2015; Zangrandi et al., 2021). For instance, mGlu5 knockout mice or mice that received the mGlu5 NAM, MTEP, displayed detriments in stress coping mechanisms (Li et al., 2006; Shin et al., 2015; Zangrandi et al., 2021). A recent study showed that mice with conditional knockout of mGlu5 in dopamine receptor D1 neurons demonstrated divergent coping mechanisms in response to acute escapable or inescapable stress compared with littermate controls, such that mGlu5 conditional knockout mice showed enhanced active stress coping upon exposure to escapable stress task and higher levels of passive strategy in response to inescapable stress (Zangrandi et al., 2021). Numerous studies also implicate the mGlu1 receptor subtype in the mechanisms underlying stress and anxiety. One study demonstrated that administration of the mGlu1 antagonist, JNJ16259685, produced an anxiolytic phenotype in mice (Steckler et al., 2005). These findings provide strong support for the potential utility of group I mGlu receptor allosteric modulators for the treatment of stress-related disorders.

The development of mGlu1 and mGlu5 allosteric modulators have greatly advanced our understanding of the physiologic and behavioral role of these receptor subtypes in stress and anxiety disorders. For example, intrahippocampal injection of MTEP impairs fear extinction by blocking hippocampal metaplasticity mechanisms that lead to enhanced LTP (Stansley et al., 2018), suggesting a potential utility of mGlu5 PAMs for the treatment of stress-related disorders. As such, preclinical work has demonstrated efficacy of mGlu5 PAM enhancement of fear extinction (Sethna and Wang, 2014). Additionally, MTEP displays antidepressant-like activity in the tail suspension test and FST (Palucha et al., 2005, 2007; Belozertseva et al., 2007). Furthermore, the mGlu5 receptor NAM, DSR-98776, was shown to produce rapid antidepressant-like actions in rats (Kato et al., 2015). Leveraging partial mGlu5 receptor NAMs, studies have also demonstrated antidepressant- and anxiolytic-like effects without inducing sedation (Gould et al., 2016; Nickols et al., 2016). Together, these studies support the possibility that mGlu5 receptor NAMs may provide fast-acting antidepressant activity. Based on extensive preclinical evidence and clinical imaging studies, mGlu5 receptor NAMs were tested in clinical trials for their efficacy to treat depressive symptoms. However, two mGlu5 receptor NAMs, AZD2066 and Basimglurant (RG7090, RO4917523) have been tested in Phase II clinical trials and failed to show any efficacy over the placebo control (ClinicalTrials.gov Identifier: NCT01145755) (Quiroz et al., 2016). Nonetheless, higher doses significantly improved secondary outcomes (Quiroz et al., 2016). In combination with an improved tolerability profile, these findings warrant need for further investigation of mGlu5 receptor NAMs for the treatment of depressive disorders. In contrast to the wealth of literature supporting the utility of mGlu5 receptor allosteric modulators for the treatment of stress-related disorders, little is known about the utility of mGlu1 receptor subtype-targeting allosteric modulators for mitigating stress and anxiety phenotypes. However, several studies using mGlu1 receptor antagonists provide a strong foundation for future studies characterizing mGlu1 allosteric modulators in stress-related disorder. For example, mGlu1 receptor antagonists LY456236, 1-Aminoindan-1,5-dicarboxylic acid (AIDA), and JNJ-16259685 elicit anxiolytic-like effects in rodents (Tatarczyńska et al., 2001; Kłodzińska et al., 2004; Varty et al., 2005; Lima et al., 2008; Lavreysen et al., 2015). In addition to reported anxiolytic-like effects, one study showed antidepressant potential of mGlu1 receptor antagoni,sts such that administration of JNJ-16567083 decreased immobility time in the tail suspension test (Belozertseva et al., 2007). However, further studies are needed to evaluate the potential anxiolytic activity of mGlu1 receptor allosteric modulators in the context of stress physiology.

II. Group II: Metabotropic Glutamate2/3

Substantial preclinical and clinical evidence supports the role of group II mGlu receptor subtypes in the etiology and maintenance of stress-related disorders (Dogra and Conn, 2021). For example, expression changes in mGlu2 and mGlu3 receptor subtypes are observed in numerous models of anxiety and depression. Elevation in group II mGlu receptor expression in the hippocampus and PFC has been observed in the mice reared under isolated conditions (Kawasaki et al., 2011). Further, increased levels of mGlu2/3 receptors have been observed in the postmortem PFC tissue from patients with depression (Feyissa et al., 2010), providing evidence that increased mGlu2/3 receptor function may contribute to the etiology of depression. To this end, several studies have reported anxiolytic effects with mGlu2/3 agonists (Helton et al., 1998; Shekhar and Keim, 2000; Schoepp et al., 2003; Linden et al., 2005, 2018). A multitude of studies have also leveraged transgenic mouse lines featuring deletion of mGlu2 and/or mGlu3 to parse out the individual contributions of these receptor subtypes in stress-related disorders. However, these studies have yielded mixed results. One study reported that the anxiolytic efficacy of mGlu2/3 agonists is reduced or absent in single GRM2−/− and GRM3−/− mice compared with littermate controls (Linden et al., 2005). Alternatively, several studies have reported that mice lacking either mGlu2 or mGlu3 alone did not display altered anxiety phenotypes (Morishima et al., 2005; Fujioka et al., 2014; De Filippis et al., 2015). Nonetheless, because the anxiolytic efficacy of mGlu2/3 agonists has been observed across a variety of species, including humans and rodents, the lack of an anxiety phenotype in the transgenic mice may be due to species differences or compensatory changes.

As such, selective mGlu2 and mGlu3 receptor allosteric modulators have been investigated for their efficacy in treating symptomology of stress-related disorders (Table 7). mGlu3 receptor NAMs have been shown to elicit antidepressant-like and anxiolytic-like effects as measured by FST and marble-burying tests, respectively (Engers et al., 2015). Further, recent studies show that mGlu2 and mGlu3 receptor NAMs reverse passive coping behavior in the FST (Joffe et al., 2020) and mGlu3 receptor NAMs reverse motivational deficits and changes in the amygdalo-cortical plasticity induced by acute stress (Joffe et al., 2019a). Other studies using mGlu2 receptor PAMs, BINA, N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide, and LY487379, have demonstrated anxiolytic-like efficacy in multiple assays of rodent stress response and also displayed antidepressive-like efficacy (Galici et al., 2006; Fell et al., 2011; Wierońska et al., 2012b). A recent study also reported that administration of the selective mGlu3 NAM VU0650786 blocked the LY379268-induced trace fear conditioning enhancement in mice (Dogra et al., 2021). Together, these studies provide strong evidence that mGlu2 and mGlu3 allosteric modulators may also be a promising novel treatment strategy for stress-related disorders.

TABLE 7.

Summary of preclinical efficacy of group I/II mGlu receptor allosteric modulators in stress-related deficits

Group Receptor Type Compound Behavioral Effect References
I mGlu5 PAM CDPPB Facilitated the extinction of contextual fear memory; enhanced the initial fear memory formation and had no effect on memory retrieval Sethna and Wang, 2014
NAM MPEP/MTEP Antidepressant-like activity in the tail TST and FSTa,b (See footnotes.)
DSR-98776 Produces antidepressant-like actions in rats Kato et al., 2015
Partial NAM M-5MPEP/Br-5MPEPy Demonstrate antidepressant- and anxiolytic-like activity in FST, TST, SIH and marble burying tests Gould et al., 2016
VU0477573 Dose-dependent efficacy in marble-burying test Nickols et al., 2016
II mGlu2 PAM BINA Anxiolytic-like effects on SIH and EPM tests in mice Galici et al., 2006
THIIC Anxiolytic-like efficacy in SIH assay in rats and stress-induced marble-burying assay in mice Fell et al., 2011
LY487379/LY379268 Anxiolytic-like efficacy in SIH assay in mice Wierońska et al., 2012b
mGlu2 NAM VU6001966 Reverse passive coping behavior in the FST Joffe et al., 2020
mGlu3 NAM VU0650786 Antidepressant-like and anxiolytic-like effects as measured by FST and marble-burying tests Engers et al., 2015
Reverse passive coping behavior in the FST; reduce stress-induced deficits in motivation Joffe et al., 2020
Blocked LY379268-induced trace fear conditioning enhancement in mice Dogra et al., 2021

BR-5MPEPy, bromo-2-[2-(3 methoxyphenyl)ethynyl]-5-methylpyridine; M-5MPEP, 2-[2-(3 methoxyphenyl)ethynyl]-5-methylpyridine; THIIC, (trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide; TST, tail suspension test.

aLi et al., 2006.

bBelozertseva et al., 2007.

III. Group III: Metabotropic Glutamate4/6/7/8

Group III mGlu receptor subtypes have garnered attention as potential therapeutic targets for the treatment of stress-related disorders. Initial studies using nonselective group III mGlu receptor agonists have aimed to determine the role of these receptors in stress-related phenotypes. For instance, Tatarczynska et al. showed that intrahippocampal administration of the group III mGlu receptor agonist ACPT-I elicits anxiolytic- and antidepressant-like effects in mice (Tatarczyńska et al., 2001). Other studies also found anxiolytic-like effects of ACPT-I as measured by stress-induced hypothermia (SIH), elevated plus-maze (EPM) tests in mice, and the Vogel test in rats (Pałucha et al., 2004; Stachowicz et al., 2009). However, until recently, a lack of selective compounds has largely limited our current understanding of the specific contribution of each group III mGlu receptor subtype in the pathophysiology of stress-related disorders. The development of transgenic rodent models and selective pharmacological agents has allowed us to gain insights on the role of group III mGlu receptor subtypes in stress-related phenotypes. For example, mGlu7-selective antagonist 7-hydroxy-3-(4-iodophenoxy)-4H-chromen-4-one, which inhibits lateral amygdala LTP, reduces innate anxiety and freezing during acquisition of Pavlovian fear in mice (Gee et al., 2014). Additionally, mGluR7−/− mice displayed anxiolytic efficacy in a battery of behavioral tasks, including the staircase test, EPM, light-dark box, and SIH (Cryan et al., 2003). Interestingly, mGluR7−/− mice also show an increase in glucocorticoid-dependent feedback suppression of the hypothalamic–pituitary–adrenal axis and increases hippocampal brain-derived neurotrophic factor (BDNF) levels compared with wild-type littermate controls (Mitsukawa et al., 2006), further bolstering the hypothesis that mGlu7 receptors are critical in stress physiology. In support of this notion, a wealth of literature reports anxiolytic- and antidepressant-like efficacy of the mGlu7 agonist, AMN082 (Palucha et al., 2007; Palazzo et al., 2008; Pałucha-Poniewiera et al., 2010, 2014; Bradley et al., 2012; O’Connor and Cryan, 2013; Pałucha-Poniewiera and Pilc, 2013). Alternatively, mice lacking the mGlu4 receptor subtype exhibited increased anxiety in the open-field and EPM test as well as improvements in cued-fear conditioning (Davis et al., 2013). Studies using the orthosteric mGlu8 agonist, DCPG, showed that mGlu8 stimulation reduced anxiety-like behavior in open field and EPM tests while also attenuating the expression of contextual fear (Fendt et al., 2013). Together, these studies have begun to elucidate the role of each receptor subtype in the pathophysiology of stress-related disorders.

Based on this evidence, studies have focused on the potential utility of allosteric modulators of group III mGlu receptor subtypes for the treatment of stress-related disorders (Table 8). Studies have leveraged mGlu4 PAMs and shown anxiolytic- and antidepressant-like activity in rodents. For example, a study by Klak et al. reported that administration of the mGlu4-selective PAM 7-hydroxyimino-N-phenyl-1,7 adihydrocyclopropa[b]chromene-1a-carboxamide (PHCCC) in combination with a subeffective dose of (1S,3R,4S)-1-aminocyclopentane-1,3,4-tricarboxylic acid produced antidepressant-like effects in rats (Kłak et al., 2007). Intra-basolateral amygdala (BLA) administration of PHCCC was reported to elicit anticonflict effects in rats subjected to the Vogel conflict test (Stachowicz et al., 2004). Another mGlu4 PAM, ADX88178, dose-dependently reduced duration of immobility in the forced swim test and attenuated conditioned freezing in the acquisition phase of the fear conditioning test without altering freezing propensity in the expression phase of the task (Kalinichev et al., 2014). Furthermore, Sławińska et al. also showed that administration of the mGlu4 PAM, Lu AF21934, produced anxiolytic- but not antidepressant-like effects as measured by SIH, four-plate, marble-burying, and Vogel’s conflict tests (Sławińska et al., 2013b). In addition to promising evidence of the anxiolytic efficacy of mGlu4 allosteric modulators, another study reported an anxiolytic-like profile of mGlu7 NAMs. Specifically, the mGlu7 NAM, ADX71743, produced a robust anxiolytic-like phenotype as evidenced by dose-dependent reduction in the number of buried marbles and increasing open arm exploration in EPM and marble burying assays, respectively (Kalinichev et al., 2013).

TABLE 8.

Summary of preclinical efficacy of group III mGlu receptor allosteric modulators in stress-related deficits

Receptor Type Compound Behavioral Effect References
mGlu4 PAM PHCCC Antidepressant-like effects in rats;a intra-BLA administration elicits anticonflict effects in rats subjected to the Vogel conflict testb (See footnotes.)
ADX88178 Reduced duration of immobility in the FST; attenuated conditioned freezing in the acquisition phase of the fear conditioning Kalinichev et al., 2014
Lu AF21934 Anxiolytic, but not antidepressant-like, effects as measured by SIH, four-plate, marble-burying, and Vogel’s conflict tests Sławińska et al., 2013b
mGlu7 NAM ADX71743 Dose-dependent reduction in the number of buried marbles and increasing open arm exploration in EPM and marble-burying assays Kalinichev et al., 2013

aKłak et al., 2007.

bStachowicz et al., 2004.

BLA, basolateral amygdala

IV. Future Directions/Concluding Remarks

A wealth of preclinical literature over the past decade supports the potential utility of allosteric modulators of mGlu receptors as promising therapeutic options to treat multiple neuropsychiatric diseases, including schizophrenia, SUDs, and stress-related disorders, which currently have limited or no effective treatments. Thus far, investigation of mGlu receptor allosteric modulators has yielded important insights into the neuropharmacology of these diseases, and surely more discoveries are yet to be discovered. However, many outstanding questions remain that the field is primed to address and that will propel research on allosteric modulators forward. One important outstanding question involves the utility of mGlu allosteric modulators as novel cognitive enhancers. Neuropsychiatric conditions, such as schizophrenia, SUDs, and stress-related disorders, are known to produce marked deficits in cognitive behavior (Gould et al., 2012; Robinson et al., 2013; Tripathi et al., 2018; Lukasik et al., 2019). Extensive literature supports the notion that many cognitive deficits associated with neuropsychiatric diseases (working memory, attention, executive function, etc.), such as schizophrenia and SUD, are driven by aberrant glutamate and GABA signaling and associated detriments in synaptic plasticity (Logue and Gould, 2014; Guidi et al., 2015). Based on the well-established and critical role of mGlu receptor subtypes in regulating glutamate/GABA transmission and synaptic plasticity (see Metabotropic Glutamate Receptor Regulation of Neurotransmission and Synaptic Plasticity), allosteric modulators of mGlu receptors are promising targets to reverse disease-related cognitive deficits, a major unmet need of numerous neuropsychiatric diseases. For example, by leveraging mGlu5-mediated regulation of cortical and hippocampal plasticity, mGlu5 PAMs show excellent potential as cognitive enhancers. Recent work demonstrates that mGlu1 PAMs regulate spatial working memory via regulation of PFC SST-IN function in mice (Maksymetz et al., 2021). Furthermore, mGlu5 PAM VU0092273 enhances trace fear conditioning in wild-type mice but not in mGlu5-CA1-KO mice (Xiang et al., 2019). Further supporting this notion, the biased mGlu5 PAM VU0409551 demonstrated robust cognition enhancement as measured by enhancement of contextual fear conditioning acquisition and an increase in recognition memory in the novel object recognition task in rats, both commonly used rodent learning and memory assays dependent upon hippocampal function (Rook et al., 2015). In the same study, VU0409551 also enhanced working memory performance in rats, such that systemic administration increased accuracy in a delayed non-match-to-sample task.

Several studies have also provided evidence for the cognitive-enhancing abilities of group II mGlu receptor subtype allosteric modulators. For example, mGlu2 PAM SAR 218645 improved learning and memory in rodent models of schizophrenia (Griebel et al., 2016). In line with their ability to enhance hippocampal LTP, selective activation of mGlu3 has been shown to improve cognition in hippocampal dependent temporal associative tasks in rodents (Stansley and Conn, 2019). Interestingly, the functional interplay between mGlu3 and mGlu5 receptor subtypes and the involvement of mGlu3 in cortical plasticity further suggests that mGlu3 PAMs may also exert cognition-enhancing effects. Lastly, group III mGlu receptors may be a promising cognition-enhancing approach to mitigate neuropsychiatric-related deficits. This notion is further evidenced by preclinical cognition-enhancement observed with mGlu7 NAMs. Specifically, the mGlu7 NAM, ADX71743, reverses MK-801-induced deficits in novel object recognition and delayed spatial alternation in mice (Cieślik et al., 2018). Ongoing studies are required to evaluate the procognitive utility of mGlu receptor allosteric modulators for other cognitive processes and neuropsychiatric disease models.

Lastly, questions remain about pharmacological refinement of next-generation allosteric modulators of mGlu receptor subtypes. Allosteric-related factors, such as heterodimer engagement and signal bias, are essential to the translation of these compounds into the clinical setting. Expanding our understanding of allosteric modulator signal biases and interaction with heterodimer complexes, for example, will allow us to optimize treatments to restore normal physiology within the circuits underlying specific disease states. Several compounds targeting mGlu receptor subtypes display biased allosteric modulation or “functional selectivity” (Zhang et al., 2005; Sheffler and Conn, 2008; Rook et al., 2015). For example, the gadolinium ion, an allosteric modulator of the mGlu1α receptor subtype, inhibits Gαq/11-linked Ca2+ mobilization but also stimulates Gαs-mediated cAMP production when administered with glutamate (Tateyama and Kubo, 2006). Recent reports have also shown that signal bias can have crucial implications for therapeutic efficacy, as evidenced by β-arrestin-biased mGlu5 NAMs in models of Fragile X (Stoppel et al., 2017) or mGlu5 PAMs biased away from NMDAR modulation for schizophrenia (Rook et al., 2015; Gould et al., 2016). Moving forward, efforts will determine how to translate these pharmacological properties to emerging drug candidates through different systems and species to avoid context-dependent pharmacology that has the potential to hinder efficacy. Accounting for these pharmacological factors, among others, throughout model systems will likely increase the competitiveness and effectiveness of future drug candidates for the treatment of neuropsychiatric disease.

Abbreviations

AMN082

N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride

BINA

potassium 3′-([(2-cyclopentyl-6-7-dimethyl-1-o xo-2,3-dihydro-1H-inden-5-yl)oxy]methyl)biphenyl l-4-carboxylate

CA1

cornu ammonis

Ca2+

calcium

CNS

central nervous system

CRD

cysteine-rich domain

CPCCOEt

(–)-ethyl (7E)-7-hydroxyimino-1,7a-dihydrocyclopropa[b]chromene-1a-carboxylate

DHPG

(S)-3,5-dihydroxyphenylglycine

DOI

2,5-dimethoxy-4-iodoamphetamine

EPM

elevated plus maze

EPS

extrapyramidal side effects

EPSC

excitatory postsynaptic current

FST

forced swim test

Gαi/o

Gi/o alpha subunit

Gβγ

protein beta/gamma

GRM

Glutamate Metabotropic Receptor gene

HD

hepatahelical domain

KO

knockout

L-AP4

L-2-amino-4-phosphonobutyric acid

LTD

long-term depression

LTP

long-term potentiation

MAPK

mitogen-activated protein kinase

mGlu

metabotropic glutamate

MK-801

dizocilpine

MPEP

2-methyl-6-(phenylethynyl)pyridine

mPFC

medial prefrontal cortex

MTEP

3-((2-methyl-4-thiazolyl)ethynyl)pyridine

NAc

nucleus accumbens

NAM

negative allosteric modulator

NMDA

N-methyl-d-aspartate

NMDAR

N-methyl-D-aspartate

PAM

positive allosteric modulator

PCP

phencyclidine

PFC

prefrontal cortex

PHCCC

7-hydroxyimino-N-phenyl-1,7 adihydrocyclopropa[b]chromene-1a-carboxamide

PPI

prepulse inhibition

PR

progressive ratio

(S)-3

4-DCPG, (S)-3,4-dichlorophenyl glycine

SC

Shaffer collateral

SIH

stress-induced hypothermia

SST-IN

somatostatin-expressing interneurons

SUD

substance use disorder

VFD

Venus flytrap domain

VTA

ventral tegmental area

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Luessen, Conn.

Footnotes

This work was supported by National Institutes of Health National Institute of Mental Health [Grants MH119673 (PJC), MH062646 (PJC), MH073676 (PJC), and MH065215 (DJL)] and National Institutes of Health National Institute of Neurologic Disorders and Stroke [Grant NS031373 (PJC)].

P.J.C. received research support from Boehringer Ingelheim and Acadia Pharmaceuticals. P.J.C. is an inventor on multiple patents for allosteric modulators of metabotropic glutamate receptors. D.J.L. has no actual or perceived conflict of interest with the contents of this article.

References

  1. Abi-Dargham A, Gil R, Krystal J, Baldwin RM, Seibyl JP, Bowers M, van Dyck CH, Charney DS, Innis RB, Laruelle M (1998) Increased striatal dopamine transmission in schizophrenia: confirmation in a second cohort. Am J Psychiatry 155:761–767. [DOI] [PubMed] [Google Scholar]
  2. Achat-Mendes C, Platt DM, Spealman RD (2012) Antagonism of metabotropic glutamate 1 receptors attenuates behavioral effects of cocaine and methamphetamine in squirrel monkeys. J Pharmacol Exp Ther 343:214–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Acri JB, Cross AJ, Skolnick P (2017) From bench to bedside: mGluR2 positive allosteric modulators as medications to treat substance use disorders. Psychopharmacology (Berl) 234:1347–1355. [DOI] [PubMed] [Google Scholar]
  4. Addolorato G, Leggio L, Hopf FW, Diana M, Bonci A (2012) Novel therapeutic strategies for alcohol and drug addiction: focus on GABA, ion channels and transcranial magnetic stimulation. Neuropsychopharmacology 37:163–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adewale AS, Platt DM, Spealman RD (2006) Pharmacological stimulation of group ii metabotropic glutamate receptors reduces cocaine self-administration and cocaine-induced reinstatement of drug seeking in squirrel monkeys. J Pharmacol Exp Ther 318:922–931. [DOI] [PubMed] [Google Scholar]
  6. Aiba A, Chen C, Herrup K, Rosenmund C, Stevens CF, Tonegawa S (1994a) Reduced hippocampal long-term potentiation and context-specific deficit in associative learning in mGluR1 mutant mice. Cell 79:365–375. [DOI] [PubMed] [Google Scholar]
  7. Aiba A, Kano M, Chen C, Stanton ME, Fox GD, Herrup K, Zwingman TA, Tonegawa S (1994b) Deficient cerebellar long-term depression and impaired motor learning in mGluR1 mutant mice. Cell 79:377–388. [PubMed] [Google Scholar]
  8. Akkus FTerbeck SAmetamey SMRufer MTreyer VBurger CJohayem AMancilla BGSovago JBuck A, et al. (2014) Metabotropic glutamate receptor 5 binding in patients with obsessive-compulsive disorder. Int J Neuropsychopharmacol 17:1915–1922. [DOI] [PubMed] [Google Scholar]
  9. Alagarsamy S, Saugstad J, Warren L, Mansuy IM, Gereau RW IV, Conn PJ (2005) NMDA-induced potentiation of mGluR5 is mediated by activation of protein phosphatase 2B/calcineurin. Neuropharmacology 49 (Suppl 1):135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Altinbilek B, Manahan-Vaughan D (2007) Antagonism of group III metabotropic glutamate receptors results in impairment of LTD but not LTP in the hippocampal CA1 region, and prevents long-term spatial memory. Eur J Neurosci 26:1166–1172. [DOI] [PubMed] [Google Scholar]
  11. Aniksztejn L, Bregestovski P, Ben-Ari Y (1991) Selective activation of quisqualate metabotropic receptor potentiates NMDA but not AMPA responses. Eur J Pharmacol 205:327–328. [DOI] [PubMed] [Google Scholar]
  12. Anwyl R (1999) Metabotropic glutamate receptors: electrophysiological properties and role in plasticity. Brain Res Brain Res Rev 29:83–120. [DOI] [PubMed] [Google Scholar]
  13. Ary AW, Szumlinski KK (2007) Regional differences in the effects of withdrawal from repeated cocaine upon Homer and glutamate receptor expression: a two-species comparison. Brain Res 1184:295–305. [DOI] [PubMed] [Google Scholar]
  14. Attucci S, Carlà V, Mannaioni G, Moroni F (2001) Activation of type 5 metabotropic glutamate receptors enhances NMDA responses in mice cortical wedges. Br J Pharmacol 132:799–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Awad H, Hubert GW, Smith Y, Levey AI, Conn PJ (2000) Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci 20:7871–7879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Ayala JEChen YBanko JLSheffler DJWilliams RTelk ANWatson NLXiang ZZhang YJones PJ, et al. (2009) mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology 34:2057–2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Ayala JE, Niswender CM, Luo Q, Banko JL, Conn PJ (2008) Group III mGluR regulation of synaptic transmission at the SC-CA1 synapse is developmentally regulated. Neuropharmacology 54:804–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Ayalew MLe-Niculescu HLevey DFJain NChangala BPatel SDWiniger EBreier AShekhar AAmdur R, et al. (2012) Convergent functional genomics of schizophrenia: from comprehensive understanding to genetic risk prediction. Mol Psychiatry 17:887–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Ayoub MA, Angelicheva D, Vile D, Chandler D, Morar B, Cavanaugh JA, Visscher PM, Jablensky A, Pfleger KD, Kalaydjieva L (2012) Deleterious GRM1 mutations in schizophrenia. PLoS One 7:e32849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Azkue JJMurga MFernández-Capetillo OMateos JMElezgarai IBenítez ROsorio ADíez JPuente NBilbao A, et al. (2001) Immunoreactivity for the group III metabotropic glutamate receptor subtype mGluR4a in the superficial laminae of the rat spinal dorsal horn. J Comp Neurol 430:448–457. [DOI] [PubMed] [Google Scholar]
  21. Bäckström P, Bachteler D, Koch S, Hyytiä P, Spanagel R (2004) mGluR5 antagonist MPEP reduces ethanol-seeking and relapse behavior. Neuropsychopharmacology 29:921–928. [DOI] [PubMed] [Google Scholar]
  22. Bäckström P, Hyytiä P (2005) Suppression of alcohol self-administration and cue-induced reinstatement of alcohol seeking by the mGlu2/3 receptor agonist LY379268 and the mGlu8 receptor agonist (S)-3,4-DCPG. Eur J Pharmacol 528:110–118. [DOI] [PubMed] [Google Scholar]
  23. Bäckström P, Hyytiä P (2006) Ionotropic and metabotropic glutamate receptor antagonism attenuates cue-induced cocaine seeking. Neuropsychopharmacology 31:778–786. [DOI] [PubMed] [Google Scholar]
  24. Bäckström P, Hyytiä P (2007) Involvement of AMPA/kainate, NMDA, and mGlu5 receptors in the nucleus accumbens core in cue-induced reinstatement of cocaine seeking in rats. Psychopharmacology (Berl) 192:571–580. [DOI] [PubMed] [Google Scholar]
  25. Bahi A, Fizia K, Dietz M, Gasparini F, Flor PJ (2012) Pharmacological modulation of mGluR7 with AMN082 and MMPIP exerts specific influences on alcohol consumption and preference in rats. Addict Biol 17:235–247. [DOI] [PubMed] [Google Scholar]
  26. Balschun D, Manahan-Vaughan D, Wagner T, Behnisch T, Reymann KG, Wetzel W (1999) A specific role for group I mGluRs in hippocampal LTP and hippocampus-dependent spatial learning. Learn Mem 6:138–152. [PMC free article] [PubMed] [Google Scholar]
  27. Banov MD, Young JR, Dunn T, Szabo ST (2020) Efficacy and safety of ketamine in the management of anxiety and anxiety spectrum disorders: a review of the literature. CNS Spectr 25:331–342. [DOI] [PubMed] [Google Scholar]
  28. Baptista MA, Martin-Fardon R, Weiss F (2004) Preferential effects of the metabotropic glutamate 2/3 receptor agonist LY379268 on conditioned reinstatement versus primary reinforcement: comparison between cocaine and a potent conventional reinforcer. J Neurosci 24:4723–4727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bashir ZI, Bortolotto ZA, Davies CH, Berretta N, Irving AJ, Seal AJ, Henley JM, Jane DE, Watkins JC, Collingridge GL (1993) Induction of LTP in the hippocampus needs synaptic activation of glutamate metabotropic receptors. Nature 363:347–350. [DOI] [PubMed] [Google Scholar]
  30. Baskys A, Malenka RC (1991) Agonists at metabotropic glutamate receptors presynaptically inhibit EPSCs in neonatal rat hippocampus. J Physiol 444:687–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Bellone C, Lüscher C, Mameli M (2008) Mechanisms of synaptic depression triggered by metabotropic glutamate receptors. Cell Mol Life Sci 65:2913–2923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Belozertseva IV, Kos T, Popik P, Danysz W, Bespalov AY (2007) Antidepressant-like effects of mGluR1 and mGluR5 antagonists in the rat forced swim and the mouse tail suspension tests. Eur Neuropsychopharmacol 17:172–179. [DOI] [PubMed] [Google Scholar]
  33. Ben-Shahar OSacramento ADMiller BWWebb SMWroten MGSilva HECaruana ALGordon EJPloense KLDitzhazy J, et al. (2013) Deficits in ventromedial prefrontal cortex group 1 metabotropic glutamate receptor function mediate resistance to extinction during protracted withdrawal from an extensive history of cocaine self-administration. J Neurosci 33:495–506a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Benarroch EE (2008) Metabotropic glutamate receptors: synaptic modulators and therapeutic targets for neurologic disease. Neurology 70:964–968. [DOI] [PubMed] [Google Scholar]
  35. Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, Sanders-Bush E (2007) A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol Pharmacol 72:477–484. [DOI] [PubMed] [Google Scholar]
  36. Bennouar KE, Uberti MA, Melon C, Bacolod MD, Jimenez HN, Cajina M, Kerkerian-Le Goff L, Doller D, Gubellini P (2013) Synergy between L-DOPA and a novel positive allosteric modulator of metabotropic glutamate receptor 4: implications for Parkinson’s disease treatment and dyskinesia. Neuropharmacology 66:158–169. [DOI] [PubMed] [Google Scholar]
  37. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47:351–354. [DOI] [PubMed] [Google Scholar]
  38. Besheer J, Faccidomo S, Grondin JJ, Hodge CW (2008a) Effects of mGlu1-receptor blockade on ethanol self-administration in inbred alcohol-preferring rats. Alcohol 42:13–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Besheer J, Faccidomo S, Grondin JJ, Hodge CW (2008b) Regulation of motivation to self-administer ethanol by mGluR5 in alcohol-preferring (P) rats. Alcohol Clin Exp Res 32:209–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Bespalov AY, Dravolina OA, Sukhanov I, Zakharova E, Blokhina E, Zvartau E, Danysz W, van Heeke G, Markou A (2005) Metabotropic glutamate receptor (mGluR5) antagonist MPEP attenuated cue- and schedule-induced reinstatement of nicotine self-administration behavior in rats. Neuropharmacology 49 (Suppl 1):167–178. [DOI] [PubMed] [Google Scholar]
  41. Bessis AS, Rondard P, Gaven F, Brabet I, Triballeau N, Prezeau L, Acher F, Pin JP (2002) Closure of the Venus flytrap module of mGlu8 receptor and the activation process: insights from mutations converting antagonists into agonists. Proc Natl Acad Sci USA 99:11097–11102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Bird MK, Lohmann P, West B, Brown RM, Kirchhoff J, Raymond CR, Lawrence AJ (2014) The mGlu5 receptor regulates extinction of cocaine-driven behaviours. Drug Alcohol Depend 137:83–89. [DOI] [PubMed] [Google Scholar]
  43. Blednov YA, Harris RA (2008) Metabotropic glutamate receptor 5 (mGluR5) regulation of ethanol sedation, dependence and consumption: relationship to acamprosate actions. Int J Neuropsychopharmacol 11:775–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Blokhina EA, Kashkin VA, Zvartau EE, Danysz W, Bespalov AY (2005) Effects of nicotinic and NMDA receptor channel blockers on intravenous cocaine and nicotine self-administration in mice. Eur Neuropsychopharmacol 15:219–225. [DOI] [PubMed] [Google Scholar]
  45. Bobes J, Garcia-Portilla MP, Bascaran MT, Saiz PA, Bousoño M (2007) Quality of life in schizophrenic patients. Dialogues Clin Neurosci 9:215–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Bocchio MLukacs IPStacey RPlaha PApostolopoulos VLivermore LSen AAnsorge OGillies MJSomogyi P, et al. (2019) Group II metabotropic glutamate receptors mediate presynaptic inhibition of excitatory transmission in pyramidal neurons of the human cerebral cortex. Front Cell Neurosci 12:508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Boehr DD, Nussinov R, Wright PE (2009) The role of dynamic conformational ensembles in biomolecular recognition. Nat Chem Biol 5:789–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Bortolotto ZA, Collett VJ, Conquet F, Jia Z, van der Putten H, Collingridge GL (2005) The regulation of hippocampal LTP by the molecular switch, a form of metaplasticity, requires mGlu5 receptors. Neuropharmacology 49 (Suppl 1):13–25. [DOI] [PubMed] [Google Scholar]
  49. Bradley SR, Uslaner JM, Flick RB, Lee A, Groover KM, Hutson PH (2012) The mGluR7 allosteric agonist AMN082 produces antidepressant-like effects by modulating glutamatergic signaling. Pharmacol Biochem Behav 101:35–40. [DOI] [PubMed] [Google Scholar]
  50. Brebner K, Childress AR, Roberts DC (2002) A potential role for GABA(B) agonists in the treatment of psychostimulant addiction. Alcohol Alcohol 37:478–484. [DOI] [PubMed] [Google Scholar]
  51. Breier ASu TPSaunders RCarson REKolachana BSde Bartolomeis AWeinberger DRWeisenfeld NMalhotra AKEckelman WC, et al. (1997) Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA 94:2569–2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Camodeca N, Breakwell NA, Rowan MJ, Anwyl R (1999) Induction of LTD by activation of group I mGluR in the dentate gyrus in vitro. Neuropharmacology 38:1597–1606. [DOI] [PubMed] [Google Scholar]
  53. Capogna M (2004) Distinct properties of presynaptic group II and III metabotropic glutamate receptor-mediated inhibition of perforant pathway-CA1 EPSCs. Eur J Neurosci 19:2847–2858. [DOI] [PubMed] [Google Scholar]
  54. Caprioli D, Justinova Z, Venniro M, Shaham Y (2018) Effect of novel allosteric modulators of metabotropic glutamate receptors on drug self-administration and relapse: a review of preclinical studies and their clinical implications. Biol Psychiatry 84:180–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Carlsson A (1988) The current status of the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 1:179–186. [DOI] [PubMed] [Google Scholar]
  56. Carta M, Ariwodola OJ, Weiner JL, Valenzuela CF (2003) Alcohol potently inhibits the kainate receptor-dependent excitatory drive of hippocampal interneurons. Proc Natl Acad Sci USA 100:6813–6818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Cartmell J, Schoepp DD (2000) Regulation of neurotransmitter release by metabotropic glutamate receptors. J Neurochem 75:889–907. [DOI] [PubMed] [Google Scholar]
  58. Casabona G, Knöpfel T, Kuhn R, Gasparini F, Baumann P, Sortino MA, Copani A, Nicoletti F (1997) Expression and coupling to polyphosphoinositide hydrolysis of group I metabotropic glutamate receptors in early postnatal and adult rat brain. Eur J Neurosci 9:12–17. [DOI] [PubMed] [Google Scholar]
  59. Chaki SYoshikawa RHirota SShimazaki TMaeda MKawashima NYoshimizu TYasuhara ASakagami KOkuyama S, et al. (2004) MGS0039: a potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology 46:457–467. [DOI] [PubMed] [Google Scholar]
  60. Charpak S, Gähwiler BH, Do KQ, Knöpfel T (1990) Potassium conductances in hippocampal neurons blocked by excitatory amino-acid transmitters. Nature 347:765–767. [DOI] [PubMed] [Google Scholar]
  61. Chen YL, Huang CC, Hsu KS (2001) Time-dependent reversal of long-term potentiation by low-frequency stimulation at the hippocampal mossy fiber-CA3 synapses. J Neurosci 21:3705–3714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Chesworth R, Brown RM, Kim JH, Lawrence AJ (2013) The metabotropic glutamate 5 receptor modulates extinction and reinstatement of methamphetamine-seeking in mice. PLoS One 8:e68371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Chevaleyre V, Castillo PE (2003) Heterosynaptic LTD of hippocampal GABAergic synapses: a novel role of endocannabinoids in regulating excitability. Neuron 38:461–472. [DOI] [PubMed] [Google Scholar]
  64. Chiamulera C, Epping-Jordan MP, Zocchi A, Marcon C, Cottiny C, Tacconi S, Corsi M, Orzi F, Conquet F (2001) Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nat Neurosci 4:873–874. [DOI] [PubMed] [Google Scholar]
  65. Cho HPGarcia-Barrantes PMBrogan JTHopkins CRNiswender CMRodriguez ALVenable DFMorrison RDBubser MDaniels JS, et al. (2014) Chemical modulation of mutant mGlu1 receptors derived from deleterious GRM1 mutations found in schizophrenics. ACS Chem Biol 9:2334–2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Cieślik PWoźniak MKaczorowska KBrański PBurnat GChocyk ABobula BGruca PLitwa EPałucha-Poniewiera A, et al. (2018) Negative allosteric modulators of mGlu7 receptor as putative antipsychotic drugs. Front Mol Neurosci 11:316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Cleva RM, Olive MF (2012) mGlu receptors and drug addiction. Wiley Interdiscip Rev Membr Transp Signal 1:281–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Cohen AS, Raymond CR, Abraham WC (1998) Priming of long-term potentiation induced by activation of metabotropic glutamate receptors coupled to phospholipase C. Hippocampus 8:160–170. [DOI] [PubMed] [Google Scholar]
  69. Conde-Ceide SAlcázar JAlonso de Diego SALópez SMartín-Martín MLMartínez-Viturro CMPena MATong HMLavreysen HMackie C, et al. (2016) Preliminary investigation of 6,7-dihydropyrazolo[1,5-a]pyrazin-4-one derivatives as a novel series of mGlu5 receptor positive allosteric modulators with efficacy in preclinical models of schizophrenia. Bioorg Med Chem Lett 26:429–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Conn PJ, Christopoulos A, Lindsley CW (2009) Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat Rev Drug Discov 8:41–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Conn PJ, Pin JP (1997) Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol 37:205–237. [DOI] [PubMed] [Google Scholar]
  72. Conquet FBashir ZIDavies CHDaniel HFerraguti FBordi FFranz-Bacon KReggiani AMatarese VCondé F, et al. (1994) Motor deficit and impairment of synaptic plasticity in mice lacking mGluR1. Nature 372:237–243. [DOI] [PubMed] [Google Scholar]
  73. Cousins MS, Roberts DC, de Wit H (2002) GABA(B) receptor agonists for the treatment of drug addiction: a review of recent findings. Drug Alcohol Depend 65:209–220. [DOI] [PubMed] [Google Scholar]
  74. Coutinho V, Knöpfel T (2002) Metabotropic glutamate receptors: electrical and chemical signaling properties. Neuroscientist 8:551–561. [DOI] [PubMed] [Google Scholar]
  75. Cowen MS, Djouma E, Lawrence AJ (2005) The metabotropic glutamate 5 receptor antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine reduces ethanol self-administration in multiple strains of alcohol-preferring rats and regulates olfactory glutamatergic systems. J Pharmacol Exp Ther 315:590–600. [DOI] [PubMed] [Google Scholar]
  76. Cowen MS, Krstew E, Lawrence AJ (2007) Assessing appetitive and consummatory phases of ethanol self-administration in C57BL/6J mice under operant conditions: regulation by mGlu5 receptor antagonism. Psychopharmacology (Berl) 190:21–29. [DOI] [PubMed] [Google Scholar]
  77. Crawford JT, Roberts DC, Beveridge TJ (2013) The group II metabotropic glutamate receptor agonist, LY379268, decreases methamphetamine self-administration in rats. Drug Alcohol Depend 132:414–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Crépel V, Aniksztejn L, Ben-Ari Y, Hammond C (1994) Glutamate metabotropic receptors increase a Ca(2+)-activated nonspecific cationic current in CA1 hippocampal neurons. J Neurophysiol 72:1561–1569. [DOI] [PubMed] [Google Scholar]
  79. Crowley NA, Joffe ME (2021) Developing breakthrough psychiatric treatments by modulating G protein-coupled receptors on prefrontal cortex somatostatin interneurons. Neuropsychopharmacology 47:389–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Crupi R, Impellizzeri D, Cuzzocrea S (2019) Role of metabotropic glutamate receptors in neurological disorders. Front Mol Neurosci 12:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Cruz MT, Bajo M, Schweitzer P, Roberto M (2008) Shared mechanisms of alcohol and other drugs. Alcohol Res Health 31:137–147. [PMC free article] [PubMed] [Google Scholar]
  82. Cryan JF, Kelly PH, Neijt HC, Sansig G, Flor PJ, van Der Putten H (2003) Antidepressant and anxiolytic-like effects in mice lacking the group III metabotropic glutamate receptor mGluR7. Eur J Neurosci 17:2409–2417. [DOI] [PubMed] [Google Scholar]
  83. Dasgupta A, Lim YJ, Kumar K, Baby N, Pang KLK, Benoy A, Behnisch T, Sajikumar S (2020) Group III metabotropic glutamate receptors gate long-term potentiation and synaptic tagging/capture in rat hippocampal area CA2. eLife 9:e55344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Davidoff RA (1985) Antispasticity drugs: mechanisms of action. Ann Neurol 17:107–116. [DOI] [PubMed] [Google Scholar]
  85. Davies CH, Clarke VR, Jane DE, Collingridge GL (1995) Pharmacology of postsynaptic metabotropic glutamate receptors in rat hippocampal CA1 pyramidal neurones. Br J Pharmacol 116:1859–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Davis MJ, Iancu OD, Acher FC, Stewart BM, Eiwaz MA, Duvoisin RM, Raber J (2013) Role of mGluR4 in acquisition of fear learning and memory. Neuropharmacology 66:365–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. De Filippis B, Lyon L, Taylor A, Lane T, Burnet PW, Harrison PJ, Bannerman DM (2015) The role of group II metabotropic glutamate receptors in cognition and anxiety: comparative studies in GRM2(-/-), GRM3(-/-) and GRM2/3(-/-) knockout mice. Neuropharmacology 89:19–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. De Roo M, Klauser P, Garcia PM, Poglia L, Muller D (2008) Spine dynamics and synapse remodeling during LTP and memory processes. Prog Brain Res 169:199–207. [DOI] [PubMed] [Google Scholar]
  89. Desai MA, Conn PJ (1991) Excitatory effects of ACPD receptor activation in the hippocampus are mediated by direct effects on pyramidal cells and blockade of synaptic inhibition. J Neurophysiol 66:40–52. [DOI] [PubMed] [Google Scholar]
  90. Dhami GK, Babwah AV, Sterne-Marr R, Ferguson SS (2005) Phosphorylation-independent regulation of metabotropic glutamate receptor 1 signaling requires g protein-coupled receptor kinase 2 binding to the second intracellular loop. J Biol Chem 280:24420–24427. [DOI] [PubMed] [Google Scholar]
  91. Dhanya RPSheffler DJDahl RDavis MLee PSYang LNickols HHCho HPSmith LHD’Souza MS, et al. (2014) Design and synthesis of systemically active metabotropic glutamate subtype-2 and -3 (mGlu2/3) receptor positive allosteric modulators (PAMs): pharmacological characterization and assessment in a rat model of cocaine dependence. J Med Chem 57:4154–4172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Ding ZM, Ingraham CM, Hauser SR, Lasek AW, Bell RL, McBride WJ (2017) Reduced levels of mGlu2 receptors within the prelimbic cortex are not associated with elevated glutamate transmission or high alcohol drinking. Alcohol Clin Exp Res 41:1896–1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. 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]
  94. Dogra S, Stansley BJ, Xiang Z, Qian W, Gogliotti RG, Nicoletti F, Lindsley CW, Niswender CM, Joffe ME, Conn PJ (2021) Activating mGlu3 metabotropic glutamate receptors rescues schizophrenia-like cognitive deficits through metaplastic adaptations within the hippocampus. Biol Psychiatry 90:385–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Doherty AJ, Palmer MJ, Henley JM, Collingridge GL, Jane DE (1997) (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) activates mGlu5, but no mGlu1, receptors expressed in CHO cells and potentiates NMDA responses in the hippocampus. Neuropharmacology 36:265–267. [DOI] [PubMed] [Google Scholar]
  96. Dravolina OA, Danysz W, Bespalov AY (2006) Effects of group I metabotropic glutamate receptor antagonists on the behavioral sensitization to motor effects of cocaine in rats. Psychopharmacology (Berl) 187:397–404. [DOI] [PubMed] [Google Scholar]
  97. Dravolina OA, Zakharova ES, Shekunova EV, Zvartau EE, Danysz W, Bespalov AY (2007) mGlu1 receptor blockade attenuates cue- and nicotine-induced reinstatement of extinguished nicotine self-administration behavior in rats. Neuropharmacology 52:263–269. [DOI] [PubMed] [Google Scholar]
  98. Duric V, Clayton S, Leong ML, Yuan LL (2016) Comorbidity factors and brain mechanisms linking chronic stress and systemic illness. Neural Plast 2016:5460732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. East SPBamford SDietz MGEickmeier CFlegg AFerger BGemkow MJHeilker RHengerer BKotey A, et al. (2010) An orally bioavailable positive allosteric modulator of the mGlu4 receptor with efficacy in an animal model of motor dysfunction. Bioorg Med Chem Lett 20:4901–4905. [DOI] [PubMed] [Google Scholar]
  100. Egan MFStraub REGoldberg TEYakub ICallicott JHHariri ARMattay VSBertolino AHyde TMShannon-Weickert C, et al. (2004) Variation in GRM3 affects cognition, prefrontal glutamate, and risk for schizophrenia. Proc Natl Acad Sci USA 101:12604–12609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Engers JLRodriguez ALKonkol LCMorrison RDThompson ADByers FWBlobaum ALChang SVenable DFLoch MT, et al. (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 58:7485–7500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Enz R (2012) Structure of metabotropic glutamate receptor C-terminal domains in contact with interacting proteins. Front Mol Neurosci 5:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Feder AParides MKMurrough JWPerez AMMorgan JESaxena SKirkwood KAan Het Rot MLapidus KAWan LB, et al. (2014) Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry 71:681–688. [DOI] [PubMed] [Google Scholar]
  104. Fell MJWitkin JMFalcone JFKatner JSPerry KWHart JRorick-Kehn LOvershiner CDRasmussen KChaney SF, et al. (2011) N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC), a novel metabotropic glutamate 2 potentiator with potential anxiolytic/antidepressant properties: in vivo profiling suggests a link between behavioral and central nervous system neurochemical changes. J Pharmacol Exp Ther 336:165–177. [DOI] [PubMed] [Google Scholar]
  105. Fendt MImobersteg SPeterlik DChaperon FMattes CWittmann COlpe HRMosbacher JVranesic Ivan der Putten H, et al. (2013) Differential roles of mGlu(7) and mGlu(8) in amygdala-dependent behavior and physiology. Neuropharmacology 72:215–223. [DOI] [PubMed] [Google Scholar]
  106. Ferraguti F, Shigemoto R (2006) Metabotropic glutamate receptors. Cell Tissue Res 326:483–504. [DOI] [PubMed] [Google Scholar]
  107. Feyissa AM, Woolverton WL, Miguel-Hidalgo JJ, Wang Z, Kyle PB, Hasler G, Stockmeier CA, Iyo AH, Karolewicz B (2010) Elevated level of metabotropic glutamate receptor 2/3 in the prefrontal cortex in major depression. Prog Neuropsychopharmacol Biol Psychiatry 34:279–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Fitzjohn SM, Bortolotto ZA, Palmer MJ, Doherty AJ, Ornstein PL, Schoepp DD, Kingston AE, Lodge D, Collingridge GL (1998) The potent mGlu receptor antagonist LY341495 identifies roles for both cloned and novel mGlu receptors in hippocampal synaptic plasticity. Neuropharmacology 37:1445–1458. [DOI] [PubMed] [Google Scholar]
  109. Fitzjohn SM, Irving AJ, Palmer MJ, Harvey J, Lodge D, Collingridge GL (1996) Activation of group I mGluRs potentiates NMDA responses in rat hippocampal slices. Neurosci Lett 203:211–213. [DOI] [PubMed] [Google Scholar]
  110. Foster DJ, Conn PJ (2017) Allosteric modulation of GPCRs: new insights and potential utility for treatment of schizophrenia and other CNS disorders. Neuron 94:431–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Fotuhi M, Standaert DG, Testa CM, Penney JB Jr, Young AB (1994) Differential expression of metabotropic glutamate receptors in the hippocampus and entorhinal cortex of the rat. Brain Res Mol Brain Res 21:283–292. [DOI] [PubMed] [Google Scholar]
  112. Francesconi A, Duvoisin RM (2004) Divalent cations modulate the activity of metabotropic glutamate receptors. J Neurosci Res 75:472–479. [DOI] [PubMed] [Google Scholar]
  113. Francesconi W, Cammalleri M, Sanna PP (2004) The metabotropic glutamate receptor 5 is necessary for late-phase long-term potentiation in the hippocampal CA1 region. Brain Res 1022:12–18. [DOI] [PubMed] [Google Scholar]
  114. Frenguelli BG, Potier B, Slater NT, Alford S, Collingridge GL (1993) Metabotropic glutamate receptors and calcium signalling in dendrites of hippocampal CA1 neurones. Neuropharmacology 32:1229–1237. [DOI] [PubMed] [Google Scholar]
  115. Fujioka RNii TIwaki AShibata AIto IKitaichi KNomura MHattori STakao KMiyakawa T, et al. (2014) Comprehensive behavioral study of mGluR3 knockout mice: implication in schizophrenia related endophenotypes. Mol Brain 7:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Galici R, Echemendia NG, Rodriguez AL, Conn PJ (2005) A selective allosteric potentiator of metabotropic glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in mouse models predictive of antipsychotic activity. J Pharmacol Exp Ther 315:1181–1187. [DOI] [PubMed] [Google Scholar]
  117. Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, de Paulis T, Conn PJ (2006) Biphenyl-indanone A, a positive allosteric modulator of the metabotropic glutamate receptor subtype 2, has antipsychotic- and anxiolytic-like effects in mice. J Pharmacol Exp Ther 318:173–185. [DOI] [PubMed] [Google Scholar]
  118. Garcia-Barrantes PM, Cho HP, Blobaum AL, Niswender CM, Conn PJ, Lindsley CW (2015) Lead optimization of the VU0486321 series of mGlu1 PAMs. Part 1: SAR of modifications to the central aryl core. Bioorg Med Chem Lett 25:5107–5110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Garcia-Barrantes PM, Cho HP, Blobaum AL, Niswender CM, Conn PJ, Lindsley CW (2016a) Lead optimization of the VU0486321 series of mGlu1 PAMs. Part 3. Engineering plasma stability by discovery and optimization of isoindolinone analogs. Bioorg Med Chem Lett 26:1869–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Garcia-Barrantes PM, Cho HP, Metts AM, Blobaum AL, Niswender CM, Conn PJ, Lindsley CW (2016b) Lead optimization of the VU0486321 series of mGlu(1) PAMs. Part 2: SAR of alternative 3-methyl heterocycles and progress towards an in vivo tool. Bioorg Med Chem Lett 26:751–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Garcia-Barrantes PM, Cho HP, Starr TM, Blobaum AL, Niswender CM, Conn PJ, Lindsley CW (2016c) Re-exploration of the mGlu1 PAM Ro 07-11401 scaffold: discovery of analogs with improved CNS penetration despite steep SAR. Bioorg Med Chem Lett 26:2289–2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Gass JT, Osborne MP, Watson NL, Brown JL, Olive MF (2009) mGluR5 antagonism attenuates methamphetamine reinforcement and prevents reinstatement of methamphetamine-seeking behavior in rats. Neuropsychopharmacology 34:820–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Gastambide F, Gilmour G, Robbins TW, Tricklebank MD (2013) The mGlu5 positive allosteric modulator LSN2463359 differentially modulates motor, instrumental and cognitive effects of NMDA receptor antagonists in the rat. Neuropharmacology 64:240–247. [DOI] [PubMed] [Google Scholar]
  124. GBD 2019 Diseases and Injuries Collaborators (2020) Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396:1204–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Gee CEPeterlik DNeuhäuser CBouhelal RKaupmann KLaue GUschold-Schmidt NFeuerbach DZimmermann KOfner S, et al. (2014) Blocking metabotropic glutamate receptor subtype 7 (mGlu7) via the Venus flytrap domain (VFTD) inhibits amygdala plasticity, stress, and anxiety-related behavior. J Biol Chem 289:10975–10987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Gereau RW IV, Conn PJ (1995) Multiple presynaptic metabotropic glutamate receptors modulate excitatory and inhibitory synaptic transmission in hippocampal area CA1. J Neurosci 15:6879–6889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Gerlai R, Adams B, Fitch T, Chaney S, Baez M (2002) Performance deficits of mGluR8 knockout mice in learning tasks: the effects of null mutation and the background genotype. Neuropharmacology 43:235–249. [DOI] [PubMed] [Google Scholar]
  128. Gerlai R, Roder JC, Hampson DR (1998) Altered spatial learning and memory in mice lacking the mGluR4 subtype of metabotropic glutamate receptor. Behav Neurosci 112:525–532. [DOI] [PubMed] [Google Scholar]
  129. Gil-Sanz C, Delgado-García JM, Fairén A, Gruart A (2008) Involvement of the mGluR1 receptor in hippocampal synaptic plasticity and associative learning in behaving mice. Cereb Cortex 18:1653–1663. [DOI] [PubMed] [Google Scholar]
  130. Gladding CM, Fitzjohn SM, Molnár E (2009) Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacol Rev 61:395–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Gomeza J, Joly C, Kuhn R, Knöpfel T, Bockaert J, Pin JP (1996) The second intracellular loop of metabotropic glutamate receptor 1 cooperates with the other intracellular domains to control coupling to G-proteins. J Biol Chem 271:2199–2205. [DOI] [PubMed] [Google Scholar]
  132. Gould RWAmato RJBubser MJoffe MENedelcovych MTThompson ADNickols HHYuh JPZhan XFelts AS, et al. (2016) Partial mGlu5 negative allosteric modulators attenuate cocaine-mediated behaviors and lack psychotomimetic-like effects. Neuropsychopharmacology 41:1166–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Gould RW, Porrino LJ, Nader MA (2012) Nonhuman primate models of addiction and PET imaging: dopamine system dysregulation. Curr Top Behav Neurosci 11:25–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Grachev ID, Apkarian AV (2000) Chemical mapping of anxiety in the brain of healthy humans: an in vivo 1H-MRS study on the effects of sex, age, and brain region. Hum Brain Mapp 11:261–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Green MF (1996) What are the functional consequences of neurocognitive deficits in schizophrenia? Am J Psychiatry 153:321–330. [DOI] [PubMed] [Google Scholar]
  136. Griebel GPichat PBoulay DNaimoli VPotestio LFeatherstone RSahni SDefex HDesvignes CSlowinski F, et al. (2016) The mGluR2 positive allosteric modulator, SAR218645, improves memory and attention deficits in translational models of cognitive symptoms associated with schizophrenia. Sci Rep 6:35320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Grueter BA, Winder DG (2005) Group II and III metabotropic glutamate receptors suppress excitatory synaptic transmission in the dorsolateral bed nucleus of the stria terminalis. Neuropsychopharmacology 30:1302–1311. [DOI] [PubMed] [Google Scholar]
  138. Guérineau NC, Bossu JL, Gähwiler BH, Gerber U (1995) Activation of a nonselective cationic conductance by metabotropic glutamatergic and muscarinic agonists in CA3 pyramidal neurons of the rat hippocampus. J Neurosci 15:4395–4407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Guidi M, Kumar A, Foster TC (2015) Impaired attention and synaptic senescence of the prefrontal cortex involves redox regulation of NMDA receptors. J Neurosci 35:3966–3977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Halbout B, Bernardi RE, Hansson AC, Spanagel R (2014) Incubation of cocaine seeking following brief cocaine experience in mice is enhanced by mGluR1 blockade. J Neurosci 34:1781–1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Han JS, Fu Y, Bird GC, Neugebauer V (2006) Enhanced group II mGluR-mediated inhibition of pain-related synaptic plasticity in the amygdala. Mol Pain 2:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Hanson JE, Smith Y (1999) Group I metabotropic glutamate receptors at GABAergic synapses in monkeys. J Neurosci 19:6488–6496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Harich S, Gross G, Bespalov A (2007) Stimulation of the metabotropic glutamate 2/3 receptor attenuates social novelty discrimination deficits induced by neonatal phencyclidine treatment. Psychopharmacology (Berl) 192:511–519. [DOI] [PubMed] [Google Scholar]
  144. Harris EW, Cotman CW (1983) Effects of acidic amino acid antagonists on paired-pulse potentiation at the lateral perforant path. Exp Brain Res 52:455–460. [DOI] [PubMed] [Google Scholar]
  145. Harrison PJ, Lyon L, Sartorius LJ, Burnet PW, Lane TA (2008) The group II metabotropic glutamate receptor 3 (mGluR3, mGlu3, GRM3): expression, function and involvement in schizophrenia. J Psychopharmacol 22:308–322. [DOI] [PubMed] [Google Scholar]
  146. Harvey J, Collingridge GL (1993) Signal transduction pathways involved in the acute potentiation of NMDA responses by 1S,3R-ACPD in rat hippocampal slices. Br J Pharmacol 109:1085–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Helton DR, Tizzano JP, Monn JA, Schoepp DD, Kallman MJ (1998) Anxiolytic and side-effect profile of LY354740: a potent, highly selective, orally active agonist for group II metabotropic glutamate receptors. J Pharmacol Exp Ther 284:651–660. [PubMed] [Google Scholar]
  148. Hendricson AW, Sibbald JR, Morrisett RA (2004) Ethanol alters the frequency, amplitude, and decay kinetics of Sr2+-supported, asynchronous NMDAR mEPSCs in rat hippocampal slices. J Neurophysiol 91:2568–2577. [DOI] [PubMed] [Google Scholar]
  149. Hendricson AW, Thomas MP, Lippmann MJ, Morrisett RA (2003) Suppression of L-type voltage-gated calcium channel-dependent synaptic plasticity by ethanol: analysis of miniature synaptic currents and dendritic calcium transients. J Pharmacol Exp Ther 307:550–558. [DOI] [PubMed] [Google Scholar]
  150. Hermans E, Challiss RA (2001) Structural, signalling and regulatory properties of the group I metabotropic glutamate receptors: prototypic family C G-protein-coupled receptors. Biochem J 359:465–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Higley MJ (2014) Localized GABAergic inhibition of dendritic Ca(2+) signalling. Nat Rev Neurosci 15:567–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Hikichi HHiyoshi TMarumo TTomishima YKaku AIida IUrabe HTamita TYasuhara AKarasawa J, et al. (2015) Antipsychotic profiles of TASP0443294, a novel and orally active positive allosteric modulator of metabotropic glutamate 2 receptor. J Pharmacol Sci 127:352–361. [DOI] [PubMed] [Google Scholar]
  153. Hodge CW, Miles MF, Sharko AC, Stevenson RA, Hillmann JR, Lepoutre V, Besheer J, Schroeder JP (2006) The mGluR5 antagonist MPEP selectively inhibits the onset and maintenance of ethanol self-administration in C57BL/6J mice. Psychopharmacology (Berl) 183:429–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Hopkins CR (2013) Is there a path forward for mGlu(2) positive allosteric modulators for the treatment of schizophrenia? ACS Chem Neurosci 4:211–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Horio M, Fujita Y, Hashimoto K (2013) Therapeutic effects of metabotropic glutamate receptor 5 positive allosteric modulator CDPPB on phencyclidine-induced cognitive deficits in mice. Fundam Clin Pharmacol 27:483–488. [DOI] [PubMed] [Google Scholar]
  156. Howes O, McCutcheon R, Stone J (2015) Glutamate and dopamine in schizophrenia: an update for the 21st century. J Psychopharmacol 29:97–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. 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 27:2958–2968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Hyytiä P, Bäckström P, Liljequist S (1999) Site-specific NMDA receptor antagonists produce differential effects on cocaine self-administration in rats. Eur J Pharmacol 378:9–16. [DOI] [PubMed] [Google Scholar]
  159. Iacovelli L, Capobianco L, Iula M, Di Giorgi Gerevini V, Picascia A, Blahos J, Melchiorri D, Nicoletti F, De Blasi A (2004) Regulation of mGlu4 metabotropic glutamate receptor signaling by type-2 G-protein coupled receptor kinase (GRK2). Mol Pharmacol 65:1103–1110. [DOI] [PubMed] [Google Scholar]
  160. Ieraci A, Mallei A, Popoli M (2016) Social isolation stress induces anxious-depressive-like behavior and alterations of neuroplasticity-related genes in adult male mice. Neural Plast 2016:6212983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Iscru E, Goddyn H, Ahmed T, Callaerts-Vegh Z, D’Hooge R, Balschun D (2013) Improved spatial learning is associated with increased hippocampal but not prefrontal long-term potentiation in mGluR4 knockout mice. Genes Brain Behav 12:615–625. [DOI] [PubMed] [Google Scholar]
  162. Iso Y, Grajkowska E, Wroblewski JT, Davis J, Goeders NE, Johnson KM, Sanker S, Roth BL, Tueckmantel W, Kozikowski AP (2006) Synthesis and structure-activity relationships of 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine analogues as potent, noncompetitive metabotropic glutamate receptor subtype 5 antagonists; search for cocaine medications. J Med Chem 49:1080–1100. [DOI] [PubMed] [Google Scholar]
  163. Javitt DC, Zukin SR (1991) Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 148:1301–1308. [DOI] [PubMed] [Google Scholar]
  164. Jie F, Yin G, Yang W, Yang M, Gao S, Lv J, Li B (2018) Stress in regulation of GABA amygdala system and relevance to neuropsychiatric diseases. Front Neurosci 12:562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Jin LEWang MYang STYang YGalvin VCLightbourne TCOttenheimer DZhong QStein JRaja A, et al. (2017) mGluR2/3 mechanisms in primate dorsolateral prefrontal cortex: evidence for both presynaptic and postsynaptic actions. Mol Psychiatry 22:1615–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Jin X, Semenova S, Yang L, Ardecky R, Sheffler DJ, Dahl R, Conn PJ, Cosford ND, Markou A (2010) The mGluR2 positive allosteric modulator BINA decreases cocaine self-administration and cue-induced cocaine-seeking and counteracts cocaine-induced enhancement of brain reward function in rats. Neuropsychopharmacology 35:2021–2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Jingami H, Nakanishi S, Morikawa K (2003) Structure of the metabotropic glutamate receptor. Curr Opin Neurobiol 13:271–278. [DOI] [PubMed] [Google Scholar]
  168. Joffe ME, Conn PJ (2019) Antidepressant potential of metabotropic glutamate receptor mGlu2 and mGlu3 negative allosteric modulators. Neuropsychopharmacology 44:214–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Joffe ME, Santiago CI, Engers JL, Lindsley CW, Conn PJ (2019a) Metabotropic glutamate receptor subtype 3 gates acute stress-induced dysregulation of amygdalo-cortical function. Mol Psychiatry 24:916–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. 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 105:46–59.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Joffe ME, Santiago CI, Stansley BJ, Maksymetz J, Gogliotti RG, Engers JL, Nicoletti F, Lindsley CW, Conn PJ (2019b) Mechanisms underlying prelimbic prefrontal cortex mGlu3/mGlu5-dependent plasticity and reversal learning deficits following acute stress. Neuropharmacology 144:19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Julio-Pieper M, Flor PJ, Dinan TG, Cryan JF (2011) Exciting times beyond the brain: metabotropic glutamate receptors in peripheral and non-neural tissues. Pharmacol Rev 63:35–58. [DOI] [PubMed] [Google Scholar]
  173. Justinova Z, Le Foll B, Redhi GH, Markou A, Goldberg SR (2016) Differential effects of the metabotropic glutamate 2/3 receptor agonist LY379268 on nicotine versus cocaine self-administration and relapse in squirrel monkeys. Psychopharmacology (Berl) 233:1791–1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Kalinichev MLe Poul EBoléa CGirard FCampo BFonsi MRoyer-Urios IBrowne SEUslaner JMDavis MJ, et al. (2014) Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. J Pharmacol Exp Ther 350:495–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Kalinichev MRouillier MGirard FRoyer-Urios IBournique BFinn TCharvin DCampo BLe Poul EMutel V, et al. (2013) ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. J Pharmacol Exp Ther 344:624–636. [DOI] [PubMed] [Google Scholar]
  176. Kane JK, Hwang Y, Konu O, Loughlin SE, Leslie FM, Li MD (2005) Regulation of Homer and group I metabotropic glutamate receptors by nicotine. Eur J Neurosci 21:1145–1154. [DOI] [PubMed] [Google Scholar]
  177. Kano M, Kato M (1987) Quisqualate receptors are specifically involved in cerebellar synaptic plasticity. Nature 325:276–279. [DOI] [PubMed] [Google Scholar]
  178. Kato N (1993) Dependence of long-term depression on postsynaptic metabotropic glutamate receptors in visual cortex. Proc Natl Acad Sci USA 90:3650–3654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Kato T, Takata M, Kitaichi M, Kassai M, Inoue M, Ishikawa C, Hirose W, Yoshida K, Shimizu I (2015) DSR-98776, a novel selective mGlu5 receptor negative allosteric modulator with potent antidepressant and antimanic activity. Eur J Pharmacol 757:11–20. [DOI] [PubMed] [Google Scholar]
  180. Kaufman AM, Milnerwood AJ, Sepers MD, Coquinco A, She K, Wang L, Lee H, Craig AM, Cynader M, Raymond LA (2012) Opposing roles of synaptic and extrasynaptic NMDA receptor signaling in cocultured striatal and cortical neurons. J Neurosci 32:3992–4003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Kawasaki TAgo YYano KAraki RWashida YOnoe HChaki SNakazato AHashimoto HBaba A, et al. (2011) Increased binding of cortical and hippocampal group II metabotropic glutamate receptors in isolation-reared mice. Neuropharmacology 60:397–404. [DOI] [PubMed] [Google Scholar]
  182. Kawaura K, Karasawa J, Hikichi H (2016) Stimulation of the metabotropic glutamate (mGlu) 2 receptor attenuates the MK-801-induced increase in the immobility time in the forced swimming test in rats. Pharmacol Rep 68:80–84. [DOI] [PubMed] [Google Scholar]
  183. Kenny PJ, Boutrel B, Gasparini F, Koob GF, Markou A (2005) Metabotropic glutamate 5 receptor blockade may attenuate cocaine self-administration by decreasing brain reward function in rats. Psychopharmacology (Berl) 179:247–254. [DOI] [PubMed] [Google Scholar]
  184. Kenny PJ, Chartoff E, Roberto M, Carlezon WA Jr, Markou A (2009) NMDA receptors regulate nicotine-enhanced brain reward function and intravenous nicotine self-administration: role of the ventral tegmental area and central nucleus of the amygdala. Neuropsychopharmacology 34:266–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Kenny PJ, Paterson NE, Boutrel B, Semenova S, Harrison AA, Gasparini F, Koob GF, Skoubis PD, Markou A (2003) Metabotropic glutamate 5 receptor antagonist MPEP decreased nicotine and cocaine self-administration but not nicotine and cocaine-induced facilitation of brain reward function in rats. Ann N Y Acad Sci 1003:415–418. [DOI] [PubMed] [Google Scholar]
  186. Kesby JP, Eyles DW, McGrath JJ, Scott JG (2018) Dopamine, psychosis and schizophrenia: the widening gap between basic and clinical neuroscience. Transl Psychiatry 8:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Kinney GG, Burno M, Campbell UC, Hernandez LM, Rodriguez D, Bristow LJ, Conn PJ (2003) Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther 306:116–123. [DOI] [PubMed] [Google Scholar]
  188. Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K, Kryzhanovskaya L, Jarkova N; HBBI Study Group (2011) A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin Psychopharmacol 31:349–355. [DOI] [PubMed] [Google Scholar]
  189. Kinoshita A, Ohishi H, Neki A, Nomura S, Shigemoto R, Takada M, Nakanishi S, Mizuno N (1996a) Presynaptic localization of a metabotropic glutamate receptor, mGluR8, in the rhinencephalic areas: a light and electron microscope study in the rat. Neurosci Lett 207:61–64. [DOI] [PubMed] [Google Scholar]
  190. Kinoshita A, Ohishi H, Nomura S, Shigemoto R, Nakanishi S, Mizuno N (1996b) Presynaptic localization of a metabotropic glutamate receptor, mGluR4a, in the cerebellar cortex: a light and electron microscope study in the rat. Neurosci Lett 207:199–202. [DOI] [PubMed] [Google Scholar]
  191. Kiritoshi T, Neugebauer V (2015) Group II mGluRs modulate baseline and arthritis pain-related synaptic transmission in the rat medial prefrontal cortex. Neuropharmacology 95:388–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Kłak K, Pałucha A, Brański P, Sowa M, Pilc A (2007) Combined administration of PHCCC, a positive allosteric modulator of mGlu4 receptors and ACPT-I, mGlu III receptor agonist evokes antidepressant-like effects in rats. Amino Acids 32:169–172. [DOI] [PubMed] [Google Scholar]
  193. Klar R, Walker AG, Ghose D, Grueter BA, Engers DW, Hopkins CR, Lindsley CW, Xiang Z, Conn PJ, Niswender CM (2015) Activation of metabotropic glutamate receptor 7 is required for induction of long-term potentiation at SC-CA1 synapses in the hippocampus. J Neurosci 35:7600–7615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Kłodzińska A, Tatarczyńska E, Stachowicz K, Chojnacka-Wójcik E (2004) The anxiolytic-like activity of AIDA (1-aminoindan-1,5-dicarboxylic acid), an mGLu 1 receptor antagonist. J Physiol Pharmacol 55:113–126. [PubMed] [Google Scholar]
  195. Knackstedt LA, Moussawi K, Lalumiere R, Schwendt M, Klugmann M, Kalivas PW (2010) Extinction training after cocaine self-administration induces glutamatergic plasticity to inhibit cocaine seeking. J Neurosci 30:7984–7992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Knackstedt LA, Schwendt M (2016) mGlu5 receptors and relapse to cocaine-seeking: the role of receptor trafficking in postrelapse extinction learning deficits. Neural Plast 2016:9312508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Knackstedt LA, Trantham-Davidson HL, Schwendt M (2014) The role of ventral and dorsal striatum mGluR5 in relapse to cocaine-seeking and extinction learning. Addict Biol 19:87–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Knoflach F, Mutel V, Jolidon S, Kew JN, Malherbe P, Vieira E, Wichmann J, Kemp JA (2001) Positive allosteric modulators of metabotropic glutamate 1 receptor: characterization, mechanism of action, and binding site. Proc Natl Acad Sci USA 98:13402–13407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Kogo N, Dalezios Y, Capogna M, Ferraguti F, Shigemoto R, Somogyi P (2004) Depression of GABAergic input to identified hippocampal neurons by group III metabotropic glutamate receptors in the rat. Eur J Neurosci 19:2727–2740. [DOI] [PubMed] [Google Scholar]
  200. Koob GF, Volkow ND (2010) Neurocircuitry of addiction. Neuropsychopharmacology 35:217–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Kosinski CM, Risso Bradley S, Conn PJ, Levey AI, Landwehrmeyer GB, Penney JB Jr, Young AB, Standaert DG (1999) Localization of metabotropic glutamate receptor 7 mRNA and mGluR7a protein in the rat basal ganglia. J Comp Neurol 415:266–284. [PubMed] [Google Scholar]
  202. Kotlinska J, Bochenski M (2007) Comparison of the effects of mGluR1 and mGluR5 antagonists on the expression of behavioral sensitization to the locomotor effect of morphine and the morphine withdrawal jumping in mice. Eur J Pharmacol 558:113–118. [DOI] [PubMed] [Google Scholar]
  203. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr, Charney DS (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199–214. [DOI] [PubMed] [Google Scholar]
  204. Kubo Y, Miyashita T, Murata Y (1998) Structural basis for a Ca2+-sensing function of the metabotropic glutamate receptors. Science 279:1722–1725. [DOI] [PubMed] [Google Scholar]
  205. Kufahl PR, Martin-Fardon R, Weiss F (2011) Enhanced sensitivity to attenuation of conditioned reinstatement by the mGluR 2/3 agonist LY379268 and increased functional activity of mGluR 2/3 in rats with a history of ethanol dependence. Neuropsychopharmacology 36:2762–2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Kumaresan V, Yuan M, Yee J, Famous KR, Anderson SM, Schmidt HD, Pierce RC (2009) Metabotropic glutamate receptor 5 (mGluR5) antagonists attenuate cocaine priming- and cue-induced reinstatement of cocaine seeking. Behav Brain Res 202:238–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. 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 407:971–977. [DOI] [PubMed] [Google Scholar]
  208. Lally J, MacCabe JH (2015) Antipsychotic medication in schizophrenia: a review. Br Med Bull 114:169–179. [DOI] [PubMed] [Google Scholar]
  209. Laruelle MAbi-Dargham Avan Dyck CHGil RD’Souza CDErdos JMcCance ERosenblatt WFingado CZoghbi SS, et al. (1996) Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad Sci USA 93:9235–9240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Lassi G, Taylor AE, Timpson NJ, Kenny PJ, Mather RJ, Eisen T, Munafò MR (2016) The CHRNA5-A3-B4 gene cluster and smoking: from discovery to therapeutics. Trends Neurosci 39:851–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Lavreysen H, Langlois X, Donck LV, Nuñez JM, Pype S, Lütjens R, Megens A (2015) Preclinical evaluation of the antipsychotic potential of the mGlu2-positive allosteric modulator JNJ-40411813. Pharmacol Res Perspect 3:e00097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Lecourtier L, Homayoun H, Tamagnan G, Moghaddam B (2007) Positive allosteric modulation of metabotropic glutamate 5 (mGlu5) receptors reverses N-Methyl-D-aspartate antagonist-induced alteration of neuronal firing in prefrontal cortex. Biol Psychiatry 62:739–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Le Duigou C, Kullmann DM (2011) Group I mGluR agonist-evoked long-term potentiation in hippocampal oriens interneurons. J Neurosci 31:5777–5781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Lee B, Platt DM, Rowlett JK, Adewale AS, Spealman RD (2005) Attenuation of behavioral effects of cocaine by the metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)-pyridine in squirrel monkeys: comparison with dizocilpine. J Pharmacol Exp Ther 312:1232–1240. [DOI] [PubMed] [Google Scholar]
  215. Le Poul EBoléa CGirard FPoli SCharvin DCampo BBortoli JBessif ALuo BKoser AJ, et al. (2012) A potent and selective metabotropic glutamate receptor 4 positive allosteric modulator improves movement in rodent models of Parkinson’s disease. J Pharmacol Exp Ther 343:167–177. [DOI] [PubMed] [Google Scholar]
  216. Li JN, Liu XL, Li L (2020) Prefrontal GABA and glutamate levels correlate with impulsivity and cognitive function of prescription opioid addicts: A 1H-magnetic resonance spectroscopy study. Psychiatry Clin Neurosci 74:77–83. [DOI] [PubMed] [Google Scholar]
  217. Li X, D’Souza MS, Niño AM, Doherty J, Cross A, Markou A (2016) Attenuation of nicotine-taking and nicotine-seeking behavior by the mGlu2 receptor positive allosteric modulators AZD8418 and AZD8529 in rats. Psychopharmacology (Berl) 233:1801–1814. [DOI] [PubMed] [Google Scholar]
  218. Li X, Li J, Gardner EL, Xi ZX (2010) Activation of mGluR7s inhibits cocaine-induced reinstatement of drug-seeking behavior by a nucleus accumbens glutamate-mGluR2/3 mechanism in rats. J Neurochem 114:1368–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Li X, Li J, Peng XQ, Spiller K, Gardner EL, Xi ZX (2009) Metabotropic glutamate receptor 7 modulates the rewarding effects of cocaine in rats: involvement of a ventral pallidal GABAergic mechanism. Neuropsychopharmacology 34:1783–1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Li X, Need AB, Baez M, Witkin JM (2006) Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. J Pharmacol Exp Ther 319:254–259. [DOI] [PubMed] [Google Scholar]
  221. Li X, Xi ZX, Markou A (2013) Metabotropic glutamate 7 (mGlu7) receptor: a target for medication development for the treatment of cocaine dependence. Neuropharmacology 66:12–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. 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 25:391–396. [DOI] [PubMed] [Google Scholar]
  223. Lieberman JAStroup TSMcEvoy JPSwartz MSRosenheck RAPerkins DOKeefe RSEDavis SMDavis CELebowitz BD, et al. ; Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators (2005) Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 353:1209–1223. [DOI] [PubMed] [Google Scholar]
  224. Liechti ME, Lhuillier L, Kaupmann K, Markou A (2007) Metabotropic glutamate 2/3 receptors in the ventral tegmental area and the nucleus accumbens shell are involved in behaviors relating to nicotine dependence. J Neurosci 27:9077–9085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Liechti ME, Markou A (2007) Interactive effects of the mGlu5 receptor antagonist MPEP and the mGlu2/3 receptor antagonist LY341495 on nicotine self-administration and reward deficits associated with nicotine withdrawal in rats. Eur J Pharmacol 554:164–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Lima VC, Molchanov ML, Aguiar DC, Campos AC, Guimarães FS (2008) Modulation of defensive responses and anxiety-like behaviors by group I metabotropic glutamate receptors located in the dorsolateral periaqueductal gray. Prog Neuropsychopharmacol Biol Psychiatry 32:178–185. [DOI] [PubMed] [Google Scholar]
  227. Lin T, Dang S, Su Q, Zhang H, Zhang J, Zhang L, Zhang X, Lu Y, Li H, Zhu Z (2018) The impact and mechanism of methylated metabotropic glutamate receptors 1 and 5 in the hippocampus on depression-like behavior in prenatal stress offspring rats. J Clin Med 7:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Linden AM, Shannon H, Baez M, Yu JL, Koester A, Schoepp DD (2005) Anxiolytic-like activity of the mGLU2/3 receptor agonist LY354740 in the elevated plus maze test is disrupted in metabotropic glutamate receptor 2 and 3 knock-out mice. Psychopharmacology (Berl) 179:284–291. [DOI] [PubMed] [Google Scholar]
  229. Linden J, James AS, McDaniel C, Jentsch JD (2018) Dopamine D2 receptors in dopaminergic neurons modulate performance in a reversal learning task in mice. eNeuro 5:0229-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Lindsley CWWisnoski DDLeister WHO'brien JALemaire WWilliams DLBurno MSur CKinney GGPettibone DJ, et al. (2004) Discovery of positive allosteric modulators for the metabotropic glutamate receptor subtype 5 from a series of N-(1,3-diphenyl-1H- pyrazol-5-yl) benzamides that potentiate receptor function in vivo. J Med Chem 47:5825–5828. [DOI] [PubMed] [Google Scholar]
  231. Ling W, Shoptaw S, Majewska D (1998) Baclofen as a cocaine anti-craving medication: a preliminary clinical study. Neuropsychopharmacology 18:403–404. [DOI] [PubMed] [Google Scholar]
  232. Liriano F, Hatten C, Schwartz TL (2019) Ketamine as treatment for post-traumatic stress disorder: a review. Drugs Context 8:212305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Litman RE, Smith MA, Doherty JJ, Cross A, Raines S, Gertsik L, Zukin SR (2016) AZD8529, a positive allosteric modulator at the mGluR2 receptor, does not improve symptoms in schizophrenia: a proof of principle study. Schizophr Res 172:152–157. [DOI] [PubMed] [Google Scholar]
  234. Liu FGrauer SKelley CNavarra RGraf RZhang GAtkinson PJPopiolek MWantuch CKhawaja X, et al. (2008) ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. J Pharmacol Exp Ther 327:827–839. [DOI] [PubMed] [Google Scholar]
  235. Logan CN, Bechard AR, Hamor PU, Wu L, Schwendt M, Knackstedt LA (2020) Ceftriaxone and mGlu2/3 interactions in the nucleus accumbens core affect the reinstatement of cocaine-seeking in male and female rats. Psychopharmacology (Berl) 237:2007–2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Logue SF, Gould TJ (2014) The neural and genetic basis of executive function: attention, cognitive flexibility, and response inhibition. Pharmacol Biochem Behav 123:45–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Lominac KD, Kapasova Z, Hannun RA, Patterson C, Middaugh LD, Szumlinski KK (2006) Behavioral and neurochemical interactions between Group 1 mGluR antagonists and ethanol: potential insight into their anti-addictive properties. Drug Alcohol Depend 85:142–156. [DOI] [PubMed] [Google Scholar]
  238. Lorez M, Humbel U, Pflimlin MC, Kew JN (2003) Group III metabotropic glutamate receptors as autoreceptors in the cerebellar cortex. Br J Pharmacol 138:614–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Lovinger DM, White G, Weight FF (1989) Ethanol inhibits NMDA-activated ion current in hippocampal neurons. Science 243:1721–1724. [DOI] [PubMed] [Google Scholar]
  240. Lovinger DM, White G, Weight FF (1990) NMDA receptor-mediated synaptic excitation selectively inhibited by ethanol in hippocampal slice from adult rat. J Neurosci 10:1372–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Loweth JAScheyer AFMilovanovic MLaCrosse ALFlores-Barrera EWerner CTLi XFord KALe TOlive MF, et al. (2014) Synaptic depression via mGluR1 positive allosteric modulation suppresses cue-induced cocaine craving. Nat Neurosci 17:73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Lu YM, Jia Z, Janus C, Henderson JT, Gerlai R, Wojtowicz JM, Roder JC (1997) Mice lacking metabotropic glutamate receptor 5 show impaired learning and reduced CA1 long-term potentiation (LTP) but normal CA3 LTP. J Neurosci 17:5196–5205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Lukasik KM, Waris O, Soveri A, Lehtonen M, Laine M (2019) The relationship of anxiety and stress with working memory performance in a large non-depressed sample. Front Psychol 10:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Maccioni P, Colombo G (2009) Role of the GABA(B) receptor in alcohol-seeking and drinking behavior. Alcohol 43:555–558. [DOI] [PubMed] [Google Scholar]
  245. Maksymetz J, Byun NE, Luessen DJ, Li B, Barry RL, Gore JC, Niswender CM, Lindsley CW, Joffe ME, Conn PJ (2021) mGlu1 potentiation enhances prelimbic somatostatin interneuron activity to rescue schizophrenia-like physiological and cognitive deficits. Cell Rep 37:109950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Maksymetz J, Moran SP, Conn PJ (2017) Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Mol Brain 10:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Malenka RC, Bear MF (2004) LTP and LTD: an embarrassment of riches. Neuron 44:5–21. [DOI] [PubMed] [Google Scholar]
  248. Mameli M, Halbout B, Creton C, Engblom D, Parkitna JR, Spanagel R, Lüscher C (2009) Cocaine-evoked synaptic plasticity: persistence in the VTA triggers adaptations in the NAc. Nat Neurosci 12:1036–1041. [DOI] [PubMed] [Google Scholar]
  249. Mannaioni G, Attucci S, Missanelli A, Pellicciari R, Corradetti R, Moroni F (1999) Biochemical and electrophysiological studies on (S)-(+)-2-(3′-carboxybicyclo(1.1.1)pentyl)-glycine (CBPG), a novel mGlu5 receptor agonist endowed with mGlu1 receptor antagonist activity. Neuropharmacology 38:917–926. [DOI] [PubMed] [Google Scholar]
  250. Mannaioni G, Marino MJ, Valenti O, Traynelis SF, Conn PJ (2001) Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. J Neurosci 21:5925–5934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Manzoni O, Michel JM, Bockaert J (1997) Metabotropic glutamate receptors in the rat nucleus accumbens. Eur J Neurosci 9:1514–1523. [DOI] [PubMed] [Google Scholar]
  252. Manzoni OJ, Williams JT (1999) Presynaptic regulation of glutamate release in the ventral tegmental area during morphine withdrawal. J Neurosci 19:6629–6636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Mao L, Lau YS, Wang JQ (2000) Activation of group III metabotropic glutamate receptors inhibits basal and amphetamine-stimulated dopamine release in rat dorsal striatum: an in vivo microdialysis study. Eur J Pharmacol 404:289–297. [DOI] [PubMed] [Google Scholar]
  254. Mao L, Wang JQ (2000) Distinct inhibition of acute cocaine-stimulated motor activity following microinjection of a group III metabotropic glutamate receptor agonist into the dorsal striatum of rats. Pharmacol Biochem Behav 67:93–101. [DOI] [PubMed] [Google Scholar]
  255. Marek GJ, Wright RA, Schoepp DD, Monn JA, Aghajanian GK (2000) Physiological antagonism between 5-hydroxytryptamine(2A) and group II metabotropic glutamate receptors in prefrontal cortex. J Pharmacol Exp Ther 292:76–87. [PubMed] [Google Scholar]
  256. Martin G, Nie Z, Siggins GR (1997) Metabotropic glutamate receptors regulate N-methyl-D-aspartate-mediated synaptic transmission in nucleus accumbens. J Neurophysiol 78:3028–3038. [DOI] [PubMed] [Google Scholar]
  257. Martin G, Przewlocki R, Siggins GR (1999) Chronic morphine treatment selectively augments metabotropic glutamate receptor-induced inhibition of N-methyl-D-aspartate receptor-mediated neurotransmission in nucleus accumbens. J Pharmacol Exp Ther 288:30–35. [PubMed] [Google Scholar]
  258. Martin-Fardon R, Baptista MA, Dayas CV, Weiss F (2009) Dissociation of the effects of MTEP [3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]piperidine] on conditioned reinstatement and reinforcement: comparison between cocaine and a conventional reinforcer. J Pharmacol Exp Ther 329:1084–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. McBain CJ, DiChiara TJ, Kauer JA (1994) Activation of metabotropic glutamate receptors differentially affects two classes of hippocampal interneurons and potentiates excitatory synaptic transmission. J Neurosci 14:4433–4445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. McCutcheon JE, Loweth JA, Ford KA, Marinelli M, Wolf ME, Tseng KY (2011) Group I mGluR activation reverses cocaine-induced accumulation of calcium-permeable AMPA receptors in nucleus accumbens synapses via a protein kinase C-dependent mechanism. J Neurosci 31:14536–14541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. McCutcheon RA, Abi-Dargham A, Howes OD (2019) Schizophrenia, dopamine and the striatum: from biology to symptoms. Trends Neurosci 42:205–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. McEvoy JP (2007) The costs of schizophrenia. J Clin Psychiatry 68 (Suppl 14):4–7. [PubMed] [Google Scholar]
  263. McMillen BA, Crawford MS, Kulers CM, Williams HL (2005) Effects of a metabotropic, mglu5, glutamate receptor antagonist on ethanol consumption by genetic drinking rats. Alcohol Alcohol 40:494–497. [DOI] [PubMed] [Google Scholar]
  264. Meinhardt MW, Hansson AC, Perreau-Lenz S, Bauder-Wenz C, Stählin O, Heilig M, Harper C, Drescher KU, Spanagel R, Sommer WH (2013) Rescue of infralimbic mGluR2 deficit restores control over drug-seeking behavior in alcohol dependence. J Neurosci 33:2794–2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Melendez RI, Vuthiganon J, Kalivas PW (2005) Regulation of extracellular glutamate in the prefrontal cortex: focus on the cystine glutamate exchanger and group I metabotropic glutamate receptors. J Pharmacol Exp Ther 314:139–147. [DOI] [PubMed] [Google Scholar]
  266. Meltzer HY (2013) Update on typical and atypical antipsychotic drugs. Annu Rev Med 64:393–406. [DOI] [PubMed] [Google Scholar]
  267. Mercier MS, Lodge D (2014) Group III metabotropic glutamate receptors: pharmacology, physiology and therapeutic potential. Neurochem Res 39:1876–1894. [DOI] [PubMed] [Google Scholar]
  268. Mitchell SJ, Silver RA (2000) Glutamate spillover suppresses inhibition by activating presynaptic mGluRs. Nature 404:498–502. [DOI] [PubMed] [Google Scholar]
  269. Mitsukawa K, Mombereau C, Lötscher E, Uzunov DP, van der Putten H, Flor PJ, Cryan JF (2006) Metabotropic glutamate receptor subtype 7 ablation causes dysregulation of the HPA axis and increases hippocampal BDNF protein levels: implications for stress-related psychiatric disorders. Neuropsychopharmacology 31:1112–1122. [DOI] [PubMed] [Google Scholar]
  270. Mitsukawa KYamamoto ROfner SNozulak JPescott OLukic SStoehr NMombereau CKuhn RMcAllister KH, et al. (2005) A selective metabotropic glutamate receptor 7 agonist: activation of receptor signaling via an allosteric site modulates stress parameters in vivo. Proc Natl Acad Sci USA 102:18712–18717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Monaghan DT, Irvine MW, Costa BM, Fang G, Jane DE (2012) Pharmacological modulation of NMDA receptor activity and the advent of negative and positive allosteric modulators. Neurochem Int 61:581–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Morishima Y, Miyakawa T, Furuyashiki T, Tanaka Y, Mizuma H, Nakanishi S (2005) Enhanced cocaine responsiveness and impaired motor coordination in metabotropic glutamate receptor subtype 2 knockout mice. Proc Natl Acad Sci USA 102:4170–4175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Moussawi K, Kalivas PW (2010) Group II metabotropic glutamate receptors (mGlu2/3) in drug addiction. Eur J Pharmacol 639:115–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, Kalivas PW (2009) N-Acetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci 12:182–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Muguruza C, Meana JJ, Callado LF (2016) Group II metabotropic glutamate receptors as targets for novel antipsychotic drugs. Front Pharmacol 7:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Mukherjee S, Manahan-Vaughan D (2013) Role of metabotropic glutamate receptors in persistent forms of hippocampal plasticity and learning. Neuropharmacology 66:65–81. [DOI] [PubMed] [Google Scholar]
  277. Muly EC, Maddox M, Smith Y (2003) Distribution of mGluR1alpha and mGluR5 immunolabeling in primate prefrontal cortex. J Comp Neurol 467:521–535. [DOI] [PubMed] [Google Scholar]
  278. Murray CH, Christian DT, Milovanovic M, Loweth JA, Hwang EK, Caccamise AJ, Funke JR, Wolf ME (2021) mGlu5 function in the nucleus accumbens core during the incubation of methamphetamine craving. Neuropharmacology 186:108452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Murray CH, Loweth JA, Milovanovic M, Stefanik MT, Caccamise AJ, Dolubizno H, Funke JR, Foster Olive M, Wolf ME (2019) AMPA receptor and metabotropic glutamate receptor 1 adaptations in the nucleus accumbens core during incubation of methamphetamine craving. Neuropsychopharmacology 44:1534–1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Muto T, Tsuchiya D, Morikawa K, Jingami H (2007) Structures of the extracellular regions of the group II/III metabotropic glutamate receptors. Proc Natl Acad Sci USA 104:3759–3764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Nakajima Y, Iwakabe H, Akazawa C, Nawa H, Shigemoto R, Mizuno N, Nakanishi S (1993) Molecular characterization of a novel retinal metabotropic glutamate receptor mGluR6 with a high agonist selectivity for L-2-amino-4-phosphonobutyrate. J Biol Chem 268:11868–11873. [PubMed] [Google Scholar]
  282. Nasir M, Trujillo D, Levine J, Dwyer JB, Rupp ZW, Bloch MH (2020) Glutamate systems in DSM-5 anxiety disorders: their role and a review of glutamate and GABA psychopharmacology. Front Psychiatry 11:548505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Nawy S (1999) The metabotropic receptor mGluR6 may signal through G(o), but not phosphodiesterase, in retinal bipolar cells. J Neurosci 19:2938–2944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Nemeroff CB (2003) The role of GABA in the pathophysiology and treatment of anxiety disorders. Psychopharmacol Bull 37:133–146. [PubMed] [Google Scholar]
  285. Neyman S, Manahan-Vaughan D (2008) Metabotropic glutamate receptor 1 (mGluR1) and 5 (mGluR5) regulate late phases of LTP and LTD in the hippocampal CA1 region in vitro. Eur J Neurosci 27:1345–1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Ngomba RT, Santolini I, Salt TE, Ferraguti F, Battaglia G, Nicoletti F, van Luijtelaar G (2011) Metabotropic glutamate receptors in the thalamocortical network: strategic targets for the treatment of absence epilepsy. Epilepsia 52:1211–1222. [DOI] [PubMed] [Google Scholar]
  287. Nickols HHYuh JPGregory KJMorrison RDBates BSStauffer SREmmitte KABubser MPeng WNedelcovych MT, et al. (2016) VU0477573: partial negative allosteric modulator of the subtype 5 metabotropic glutamate receptor with in vivo efficacy. J Pharmacol Exp Ther 356:123–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Nicoletti F, Bockaert J, Collingridge GL, Conn PJ, Ferraguti F, Schoepp DD, Wroblewski JT, Pin JP (2011) Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology 60:1017–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Nicoletti FOrlando RDi Menna LCannella MNotartomaso SMascio GIacovelli LMatrisciano FFazio FCaraci F, et al. (2019) Targeting mGlu receptors for optimization of antipsychotic activity and disease-modifying effect in schizophrenia. Front Psychiatry 10:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Niedzielska-Andres E, Pomierny-Chamioło L, Andres M, Walczak M, Knackstedt LA, Filip M, Przegaliński E (2021) Cocaine use disorder: a look at metabotropic glutamate receptors and glutamate transporters. Pharmacol Ther 221:107797. [DOI] [PubMed] [Google Scholar]
  291. Nikiforuk A, Popik P, Drescher KU, van Gaalen M, Relo AL, Mezler M, Marek G, Schoemaker H, Gross G, Bespalov A (2010) Effects of a positive allosteric modulator of group II metabotropic glutamate receptors, LY487379, on cognitive flexibility and impulsive-like responding in rats. J Pharmacol Exp Ther 335:665–673. [DOI] [PubMed] [Google Scholar]
  292. 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]
  293. Noetzel MJ, Gregory KJ, Vinson PN, Manka JT, Stauffer SR, Lindsley CW, Niswender CM, Xiang Z, Conn PJ (2013) A novel metabotropic glutamate receptor 5 positive allosteric modulator acts at a unique site and confers stimulus bias to mGlu5 signaling. Mol Pharmacol 83:835–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Nomura T, Oyamada Y, Fernandes HB, Remmers CL, Xu J, Meltzer HY, Contractor A (2016) Subchronic phencyclidine treatment in adult mice increases GABAergic transmission and LTP threshold in the hippocampus. Neuropharmacology 100:90–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Nuss P (2015) Anxiety disorders and GABA neurotransmission: a disturbance of modulation. Neuropsychiatr Dis Treat 11:165–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Nussinov R, Tsai CJ, Csermely P (2011) Allo-network drugs: harnessing allostery in cellular networks. Trends Pharmacol Sci 32:686–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Obara I, Bell RL, Goulding SP, Reyes CM, Larson LA, Ary AW, Truitt WA, Szumlinski KK (2009) Differential effects of chronic ethanol consumption and withdrawal on homer/glutamate receptor expression in subregions of the accumbens and amygdala of P rats. Alcohol Clin Exp Res 33:1924–1934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. O’Brien DE, Conn PJ (2016) Neurobiological insights from mGlu receptor allosteric modulation. Int J Neuropsychopharmacol 19:pyv133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. O’Connor RM, Cryan JF (2013) The effects of mGlu7 receptor modulation in behavioural models sensitive to antidepressant action in two mouse strains. Behav Pharmacol 24:105–113. [DOI] [PubMed] [Google Scholar]
  300. Ohtsuki TKoga MIshiguro HHoriuchi YArai MNiizato KItokawa MInada TIwata NIritani S, et al. (2008) A polymorphism of the metabotropic glutamate receptor mGluR7 (GRM7) gene is associated with schizophrenia. Schizophr Res 101:9–16. [DOI] [PubMed] [Google Scholar]
  301. Olive MF (2009) Metabotropic glutamate receptor ligands as potential therapeutics for addiction. Curr Drug Abuse Rev 2:83–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Olive MF, McGeehan AJ, Kinder JR, McMahon T, Hodge CW, Janak PH, Messing RO (2005) The mGluR5 antagonist 6-methyl-2-(phenylethynyl)pyridine decreases ethanol consumption via a protein kinase C epsilon-dependent mechanism. Mol Pharmacol 67:349–355. [DOI] [PubMed] [Google Scholar]
  303. O’Mara SM, Rowan MJ, Anwyl R (1995) Metabotropic glutamate receptor-induced homosynaptic long-term depression and depotentiation in the dentate gyrus of the rat hippocampus in vitro. Neuropharmacology 34:983–989. [DOI] [PubMed] [Google Scholar]
  304. Ombelet W, Fourie FL, Vandeput H, Bosmans E, Cox A, Janssen M, Kruger T (1994) Teratozoospermia and in-vitro fertilization: a randomized prospective study. Hum Reprod 9:1479–1484. [DOI] [PubMed] [Google Scholar]
  305. 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 49:413–421. [DOI] [PubMed] [Google Scholar]
  306. Palazzo E, Boccella S, Marabese I, Pierretti G, Guida F, Maione S (2020) The cold case of metabotropic glutamate receptor 6: unjust detention in the retina? Curr Neuropharmacol 18:120–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Palazzo E, Fu Y, Ji G, Maione S, Neugebauer V (2008) Group III mGluR7 and mGluR8 in the amygdala differentially modulate nocifensive and affective pain behaviors. Neuropharmacology 55:537–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Palmatier MI, Liu X, Donny EC, Caggiula AR, Sved AF (2008) Metabotropic glutamate 5 receptor (mGluR5) antagonists decrease nicotine seeking, but do not affect the reinforcement enhancing effects of nicotine. Neuropsychopharmacology 33:2139–2147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Palmer MJ, Irving AJ, Seabrook GR, Jane DE, Collingridge GL (1997) The group I mGlu receptor agonist DHPG induces a novel form of LTD in the CA1 region of the hippocampus. Neuropharmacology 36:1517–1532. [DOI] [PubMed] [Google Scholar]
  310. Pałucha A, Brański P, Szewczyk B, Wierońska JM, Kłak K, Pilc A (2005) Potential antidepressant-like effect of MTEP, a potent and highly selective mGluR5 antagonist. Pharmacol Biochem Behav 81:901–906. [DOI] [PubMed] [Google Scholar]
  311. Palucha A, Klak K, Branski P, van der Putten H, Flor PJ, Pilc A (2007) Activation of the mGlu7 receptor elicits antidepressant-like effects in mice. Psychopharmacology (Berl) 194:555–562. [DOI] [PubMed] [Google Scholar]
  312. Pałucha A, Tatarczyńska E, Brański P, Szewczyk B, Wierońska JM, Kłak K, Chojnacka-Wójcik E, Nowak G, Pilc A (2004) Group III mGlu receptor agonists produce anxiolytic- and antidepressant-like effects after central administration in rats. Neuropharmacology 46:151–159. [DOI] [PubMed] [Google Scholar]
  313. Pałucha-Poniewiera A, Brański P, Lenda T, Pilc A (2010) The antidepressant-like action of metabotropic glutamate 7 receptor agonist N,N′-bis(diphenylmethyl)-1,2-ethanediamine (AMN082) is serotonin-dependent. J Pharmacol Exp Ther 334:1066–1074. [DOI] [PubMed] [Google Scholar]
  314. Pałucha-Poniewiera A, Kłodzińska A, Stachowicz K, Tokarski K, Hess G, Schann S, Frauli M, Neuville P, Pilc A (2008) Peripheral administration of group III mGlu receptor agonist ACPT-I exerts potential antipsychotic effects in rodents. Neuropharmacology 55:517–524. [DOI] [PubMed] [Google Scholar]
  315. Pałucha-Poniewiera A, Pilc A (2013) A selective mGlu7 receptor antagonist MMPIP reversed antidepressant-like effects of AMN082 in rats. Behav Brain Res 238:109–112. [DOI] [PubMed] [Google Scholar]
  316. Pałucha-Poniewiera A, Szewczyk B, Pilc A (2014) Activation of the mTOR signaling pathway in the antidepressant-like activity of the mGlu5 antagonist MTEP and the mGlu7 agonist AMN082 in the FST in rats. Neuropharmacology 82:59–68. [DOI] [PubMed] [Google Scholar]
  317. Paquet M, Smith Y (2003) Group I metabotropic glutamate receptors in the monkey striatum: subsynaptic association with glutamatergic and dopaminergic afferents. J Neurosci 23:7659–7669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Parelkar NK, Wang JQ (2008) Upregulation of metabotropic glutamate receptor 8 mRNA expression in the rat forebrain after repeated amphetamine administration. Neurosci Lett 433:250–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Parmentier-Batteur SHutson PHMenzel KUslaner JMMattson BAO’Brien JAMagliaro BCForest TStump CATynebor RM, et al. (2014) Mechanism based neurotoxicity of mGlu5 positive allosteric modulators—development challenges for a promising novel antipsychotic target. Neuropharmacology 82:161–173. [DOI] [PubMed] [Google Scholar]
  320. Paterson NE, Semenova S, Gasparini F, Markou A (2003) The mGluR5 antagonist MPEP decreased nicotine self-administration in rats and mice. Psychopharmacology (Berl) 167:257–264. [DOI] [PubMed] [Google Scholar]
  321. Patil STZhang LMartenyi FLowe SLJackson KAAndreev BVAvedisova ASBardenstein LMGurovich IYMorozova MA, et al. (2007) Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med 13:1102–1107. [DOI] [PubMed] [Google Scholar]
  322. Pedarzani P, Storm JF (1993) PKA mediates the effects of monoamine transmitters on the K+ current underlying the slow spike frequency adaptation in hippocampal neurons. Neuron 11:1023–1035. [DOI] [PubMed] [Google Scholar]
  323. Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ (2017) Hippocampal GABAergic inhibitory interneurons. Physiol Rev 97:1619–1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Perez Y, Morin F, Lacaille JC (2001) A hebbian form of long-term potentiation dependent on mGluR1a in hippocampal inhibitory interneurons. Proc Natl Acad Sci USA 98:9401–9406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Perroy J, Prezeau L, De Waard M, Shigemoto R, Bockaert J, Fagni L (2000) Selective blockade of P/Q-type calcium channels by the metabotropic glutamate receptor type 7 involves a phospholipase C pathway in neurons. J Neurosci 20:7896–7904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Petrozzino JJ, Connor JA (1994) Dendritic Ca2+ accumulations and metabotropic glutamate receptor activation associated with an N-methyl-D-aspartate receptor-independent long-term potentiation in hippocampal CA1 neurons. Hippocampus 4:546–558. [DOI] [PubMed] [Google Scholar]
  327. Phan KL, Fitzgerald DA, Cortese BM, Seraji-Bozorgzad N, Tancer ME, Moore GJ (2005) Anterior cingulate neurochemistry in social anxiety disorder: 1H-MRS at 4 Tesla. Neuroreport 16:183–186. [DOI] [PubMed] [Google Scholar]
  328. Pierce RC, Meil WM, Kalivas PW (1997) The NMDA antagonist, dizocilpine, enhances cocaine reinforcement without influencing mesoaccumbens dopamine transmission. Psychopharmacology (Berl) 133:188–195. [DOI] [PubMed] [Google Scholar]
  329. Pilc A, Chaki S, Nowak G, Witkin JM (2008) Mood disorders: regulation by metabotropic glutamate receptors. Biochem Pharmacol 75:997–1006. [DOI] [PubMed] [Google Scholar]
  330. Pin JP, Joly C, Heinemann SF, Bockaert J (1994) Domains involved in the specificity of G protein activation in phospholipase C-coupled metabotropic glutamate receptors. EMBO J 13:342–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  331. Pisani A, Gubellini P, Bonsi P, Conquet F, Picconi B, Centonze D, Bernardi G, Calabresi P (2001) Metabotropic glutamate receptor 5 mediates the potentiation of N-methyl-D-aspartate responses in medium spiny striatal neurons. Neuroscience 106:579–587. [DOI] [PubMed] [Google Scholar]
  332. Platt DM, Rowlett JK, Spealman RD (2008) Attenuation of cocaine self-administration in squirrel monkeys following repeated administration of the mGluR5 antagonist MPEP: comparison with dizocilpine. Psychopharmacology (Berl) 200:167–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Poisik O, Raju DV, Verreault M, Rodriguez A, Abeniyi OA, Conn PJ, Smith Y (2005) Metabotropic glutamate receptor 2 modulates excitatory synaptic transmission in the rat globus pallidus. Neuropharmacology 49 (Suppl 1):57–69. [DOI] [PubMed] [Google Scholar]
  334. Popoli M, Yan Z, McEwen BS, Sanacora G (2011) The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci 13:22–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Puddifoot C, Martel MA, Soriano FX, Camacho A, Vidal-Puig A, Wyllie DJ, Hardingham GE (2012) PGC-1α negatively regulates extrasynaptic NMDAR activity and excitotoxicity. J Neurosci 32:6995–7000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  336. Pulvirenti L, Balducci C, Koob GF (1997) Dextromethorphan reduces intravenous cocaine self-administration in the rat. Eur J Pharmacol 321:279–283. [DOI] [PubMed] [Google Scholar]
  337. Quiroz JA, Tamburri P, Deptula D, Banken L, Beyer U, Rabbia M, Parkar N, Fontoura P, Santarelli L (2016) Efficacy and safety of basimglurant as adjunctive therapy for major depression: a randomized clinical trial. JAMA Psychiatry 73:675–684. [DOI] [PubMed] [Google Scholar]
  338. Raymond CR, Thompson VL, Tate WP, Abraham WC (2000) Metabotropic glutamate receptors trigger homosynaptic protein synthesis to prolong long-term potentiation. J Neurosci 20:969–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Reiner A, Levitz J (2018) Glutamatergic signaling in the central nervous system: ionotropic and metabotropic receptors in concert. Neuron 98:1080–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Risch N, Herrell R, Lehner T, Liang KY, Eaves L, Hoh J, Griem A, Kovacs M, Ott J, Merikangas KR (2009) Interaction between the serotonin transporter gene (5-HTTLPR), stressful life events, and risk of depression: a meta-analysis. JAMA 301:2462–2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ (1987) Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 237:1219–1223. [DOI] [PubMed] [Google Scholar]
  342. Robbe D, Bockaert J, Manzoni OJ (2002) Metabotropic glutamate receptor 2/3-dependent long-term depression in the nucleus accumbens is blocked in morphine withdrawn mice. Eur J Neurosci 16:2231–2235. [DOI] [PubMed] [Google Scholar]
  343. Robbins MJStarr KRHoney ASoffin EMRourke CJones GAKelly FMStrum JMelarange RAHarris AJ, et al. (2007) Evaluation of the mGlu8 receptor as a putative therapeutic target in schizophrenia. Brain Res 1152:215–227. [DOI] [PubMed] [Google Scholar]
  344. Robinson OJ, Vytal K, Cornwell BR, Grillon C (2013) The impact of anxiety upon cognition: perspectives from human threat of shock studies. Front Hum Neurosci 7:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Romano C, Sesma MA, McDonald CT, O’Malley K, Van den Pol AN, Olney JW (1995) Distribution of metabotropic glutamate receptor mGluR5 immunoreactivity in rat brain. J Comp Neurol 355:455–469. [DOI] [PubMed] [Google Scholar]
  346. Rondard P, Liu J, Huang S, Malhaire F, Vol C, Pinault A, Labesse G, Pin JP (2006) Coupling of agonist binding to effector domain activation in metabotropic glutamate-like receptors. J Biol Chem 281:24653–24661. [DOI] [PubMed] [Google Scholar]
  347. Rook JMNoetzel MJPouliot WABridges TMVinson PNCho HPZhou YGogliotti RDManka JTGregory KJ, et al. (2013) Unique signaling profiles of positive allosteric modulators of metabotropic glutamate receptor subtype 5 determine differences in in vivo activity. Biol Psychiatry 73:501–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Rook JMXiang ZLv XGhoshal ADickerson JWBridges TMJohnson KAFoster DJGregory KJVinson PN, et al. (2015) Biased mGlu5-positive allosteric modulators provide In vivo efficacy without potentiating mGlu5 modulation of NMDAR currents. Neuron 86:1029–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Ruan H, Yao W-D (2021) Loss of mGluR1-LTD following cocaine exposure accumulates Ca. J Neurogenet 35:358–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Salih H, Anghelescu I, Kezic I, Sinha V, Hoeben E, Van Nueten L, De Smedt H, De Boer P (2015) Pharmacokinetic and pharmacodynamic characterisation of JNJ-40411813, a positive allosteric modulator of mGluR2, in two randomised, double-blind phase-I studies. J Psychopharmacol 29:414–425. [DOI] [PubMed] [Google Scholar]
  351. Salling MC, Faccidomo S, Hodge CW (2008) Nonselective suppression of operant ethanol and sucrose self-administration by the mGluR7 positive allosteric modulator AMN082. Pharmacol Biochem Behav 91:14–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  352. Satow AMaehara SIse SHikichi HFukushima MSuzuki GKimura TTanak TIto SKawamoto H, et al. (2008) Pharmacological effects of the metabotropic glutamate receptor 1 antagonist compared with those of the metabotropic glutamate receptor 5 antagonist and metabotropic glutamate receptor 2/3 agonist in rodents: detailed investigations with a selective allosteric metabotropic glutamate receptor 1 antagonist, FTIDC [4-[1-(2-fluoropyridine-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide]. J Pharmacol Exp Ther 326:577–586. [DOI] [PubMed] [Google Scholar]
  353. Satow ASuzuki GMaehara SHikichi HMurai TMurai TKawagoe-Takaki HHata MIto SOzaki S, et al. (2009) Unique antipsychotic activities of the selective metabotropic glutamate receptor 1 allosteric antagonist 2-cyclopropyl-5-[1-(2-fluoro-3-pyridinyl)-5-methyl-1H-1,2,3-triazol-4-yl]-2,3-dihydro-1H-isoindol-1-one. J Pharmacol Exp Ther 330:179–190. [DOI] [PubMed] [Google Scholar]
  354. Saugstad JA, Kinzie JM, Mulvihill ER, Segerson TP, Westbrook GL (1994) Cloning and expression of a new member of the L-2-amino-4-phosphonobutyric acid-sensitive class of metabotropic glutamate receptors. Mol Pharmacol 45:367–372. [PubMed] [Google Scholar]
  355. Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511:421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Schmidt HD, Kimmey BA, Arreola AC, Pierce RC (2015) Group I metabotropic glutamate receptor-mediated activation of PKC gamma in the nucleus accumbens core promotes the reinstatement of cocaine seeking. Addict Biol 20:285–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Schoepp DD (2001) Unveiling the functions of presynaptic metabotropic glutamate receptors in the central nervous system. J Pharmacol Exp Ther 299:12–20. [PubMed] [Google Scholar]
  358. Schoepp DD, Wright RA, Levine LR, Gaydos B, Potter WZ (2003) LY354740, an mGlu2/3 receptor agonist as a novel approach to treat anxiety/stress. Stress 6:189–197. [DOI] [PubMed] [Google Scholar]
  359. Schoppa NE, Westbrook GL (1997) Modulation of mEPSCs in olfactory bulb mitral cells by metabotropic glutamate receptors. J Neurophysiol 78:1468–1475. [DOI] [PubMed] [Google Scholar]
  360. Schrader LA, Tasker JG (1997) Presynaptic modulation by metabotropic glutamate receptors of excitatory and inhibitory synaptic inputs to hypothalamic magnocellular neurons. J Neurophysiol 77:527–536. [DOI] [PubMed] [Google Scholar]
  361. Schroeder JP, Overstreet DH, Hodge CW (2005) The mGluR5 antagonist MPEP decreases operant ethanol self-administration during maintenance and after repeated alcohol deprivations in alcohol-preferring (P) rats. Psychopharmacology (Berl) 179:262–270. [DOI] [PubMed] [Google Scholar]
  362. Schroeder JP, Spanos M, Stevenson JR, Besheer J, Salling M, Hodge CW (2008) Cue-induced reinstatement of alcohol-seeking behavior is associated with increased ERK1/2 phosphorylation in specific limbic brain regions: blockade by the mGluR5 antagonist MPEP. Neuropharmacology 55:546–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Semyanov A, Kullmann DM (2000) Modulation of GABAergic signaling among interneurons by metabotropic glutamate receptors. Neuron 25:663–672. [DOI] [PubMed] [Google Scholar]
  364. Sendt KV, Giaroli G, Tracy DK (2012) Beyond dopamine: glutamate as a target for future antipsychotics. ISRN Pharmacol 2012:427267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Sethna F, Wang H (2014) Pharmacological enhancement of mGluR5 facilitates contextual fear memory extinction. Learn Mem 21:647–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Shalin SC, Hernandez CM, Dougherty MK, Morrison DK, Sweatt JD (2006) Kinase suppressor of Ras1 compartmentalizes hippocampal signal transduction and subserves synaptic plasticity and memory formation. Neuron 50:765–779. [DOI] [PubMed] [Google Scholar]
  367. Sharko AC, Hodge CW (2008) Differential modulation of ethanol-induced sedation and hypnosis by metabotropic glutamate receptor antagonists in C57BL/6J mice. Alcohol Clin Exp Res 32:67–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  368. Sheffler DJ, Conn PJ (2008) Allosteric potentiators of metabotropic glutamate receptor subtype 1a differentially modulate independent signaling pathways in baby hamster kidney cells. Neuropharmacology 55:419–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Shekhar A, Keim SR (2000) LY354740, a potent group II metabotropic glutamate receptor agonist prevents lactate-induced panic-like response in panic-prone rats. Neuropharmacology 39:1139–1146. [DOI] [PubMed] [Google Scholar]
  370. Shelton KL, Balster RL (1997) Effects of gamma-aminobutyric acid agonists and N-methyl-D-aspartate antagonists on a multiple schedule of ethanol and saccharin self-administration in rats. J Pharmacol Exp Ther 280:1250–1260. [PubMed] [Google Scholar]
  371. Shiells RA, Falk G (1990) Glutamate receptors of rod bipolar cells are linked to a cyclic GMP cascade via a G-protein. Proc Biol Sci 242:91–94. [DOI] [PubMed] [Google Scholar]
  372. Shigemoto RKinoshita AWada ENomura SOhishi HTakada MFlor PJNeki AAbe TNakanishi S, et al. (1997) Differential presynaptic localization of metabotropic glutamate receptor subtypes in the rat hippocampus. J Neurosci 17:7503–7522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Shin S, Kwon O, Kang JI, Kwon S, Oh S, Choi J, Kim CH, Kim DG (2015) mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress. Nat Neurosci 18:1017–1024. [DOI] [PubMed] [Google Scholar]
  374. Shoptaw S, Yang X, Rotheram-Fuller EJ, Hsieh YC, Kintaudi PC, Charuvastra VC, Ling W (2003) Randomized placebo-controlled trial of baclofen for cocaine dependence: preliminary effects for individuals with chronic patterns of cocaine use. J Clin Psychiatry 64:1440–1448. [DOI] [PubMed] [Google Scholar]
  375. Sidhpura N, Weiss F, Martin-Fardon R (2010) Effects of the mGlu2/3 agonist LY379268 and the mGlu5 antagonist MTEP on ethanol seeking and reinforcement are differentially altered in rats with a history of ethanol dependence. Biol Psychiatry 67:804–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. 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 28:1419–1423. [DOI] [PubMed] [Google Scholar]
  377. Sławińska AWierońska JMStachowicz KMarciniak MLasoń-Tyburkiewicz MGruca PPapp MKusek MTokarski KDoller D, et al. (2013a) The antipsychotic-like effects of positive allosteric modulators of metabotropic glutamate mGlu4 receptors in rodents. Br J Pharmacol 169:1824–1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Sławińska A, Wierońska JM, Stachowicz K, Pałucha-Poniewiera A, Uberti MA, Bacolod MA, Doller D, Pilc A (2013b) Anxiolytic- but not antidepressant-like activity of Lu AF21934, a novel, selective positive allosteric modulator of the mGlu4 receptor. Neuropharmacology 66:225–235. [DOI] [PubMed] [Google Scholar]
  379. Spooren WP, Gasparini F, van der Putten H, Koller M, Nakanishi S, Kuhn R (2000) Lack of effect of LY314582 (a group 2 metabotropic glutamate receptor agonist) on phencyclidine-induced locomotor activity in metabotropic glutamate receptor 2 knockout mice. Eur J Pharmacol 397:R1–R2. [DOI] [PubMed] [Google Scholar]
  380. Stachowicz K, Kłak K, Kłodzińska A, Chojnacka-Wojcik E, Pilc A (2004) Anxiolytic-like effects of PHCCC, an allosteric modulator of mGlu4 receptors, in rats. Eur J Pharmacol 498:153–156. [DOI] [PubMed] [Google Scholar]
  381. Stachowicz K, Kłodzińska A, Palucha-Poniewiera A, Schann S, Neuville P, Pilc A (2009) The group III mGlu receptor agonist ACPT-I exerts anxiolytic-like but not antidepressant-like effects, mediated by the serotonergic and GABA-ergic systems. Neuropharmacology 57:227–234. [DOI] [PubMed] [Google Scholar]
  382. Stansley BJ, Conn PJ (2019) Neuropharmacological insight from allosteric modulation of mGlu receptors. Trends Pharmacol Sci 40:240–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Stansley BJ, Fisher NM, Gogliotti RG, Lindsley CW, Conn PJ, Niswender CM (2018) Contextual fear extinction induces hippocampal metaplasticity mediated by metabotropic glutamate receptor 5. Cereb Cortex 28:4291–4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Steckler T, Lavreysen H, Oliveira AM, Aerts N, Van Craenendonck H, Prickaerts J, Megens A, Lesage AS (2005) Effects of mGlu1 receptor blockade on anxiety-related behaviour in the rat lick suppression test. Psychopharmacology (Berl) 179:198–206. [DOI] [PubMed] [Google Scholar]
  385. Steeds H, Carhart-Harris RL, Stone JM (2015) Drug models of schizophrenia. Ther Adv Psychopharmacol 5:43–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Stefani MR, Moghaddam B (2010) Activation of type 5 metabotropic glutamate receptors attenuates deficits in cognitive flexibility induced by NMDA receptor blockade. Eur J Pharmacol 639:26–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Stoppel LJ, Auerbach BD, Senter RK, Preza AR, Lefkowitz RJ, Bear MF (2017) β-arrestin2 couples metabotropic glutamate receptor 5 to neuronal protein synthesis and is a potential target to treat fragile X. Cell Rep 18:2807–2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Sukoff Rizzo SJLeonard SKGilbert ADollings PSmith DLZhang MYDi LPlatt BJNeal SDwyer JM, et al. (2011) The metabotropic glutamate receptor 7 allosteric modulator AMN082: a monoaminergic agent in disguise? J Pharmacol Exp Ther 338:345–352. [DOI] [PubMed] [Google Scholar]
  389. Swanson CJ, Baker DA, Carson D, Worley PF, Kalivas PW (2001) Repeated cocaine administration attenuates group I metabotropic glutamate receptor-mediated glutamate release and behavioral activation: a potential role for Homer. J Neurosci 21:9043–9052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Tan HY, Chen Q, Sust S, Buckholtz JW, Meyers JD, Egan MF, Mattay VS, Meyer-Lindenberg A, Weinberger DR, Callicott JH (2007) Epistasis between catechol-O-methyltransferase and type II metabotropic glutamate receptor 3 genes on working memory brain function. Proc Natl Acad Sci USA 104:12536–12541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Tan KR, Rudolph U, Lüscher C (2011) Hooked on benzodiazepines: GABAA receptor subtypes and addiction. Trends Neurosci 34:188–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Tan Y, Hori N, Carpenter DO (2003) The mechanism of presynaptic long-term depression mediated by group I metabotropic glutamate receptors. Cell Mol Neurobiol 23:187–203. [DOI] [PubMed] [Google Scholar]
  393. Tanabe Y, Masu M, Ishii T, Shigemoto R, Nakanishi S (1992) A family of metabotropic glutamate receptors. Neuron 8:169–179. [DOI] [PubMed] [Google Scholar]
  394. Tatarczyńska E, Kłodzińska A, Kroczka B, Chojnacka-Wójcik E, Pilc A (2001) The antianxiety-like effects of antagonists of group I and agonists of group II and III metabotropic glutamate receptors after intrahippocampal administration. Psychopharmacology (Berl) 158:94–99. [DOI] [PubMed] [Google Scholar]
  395. Tateyama M, Kubo Y (2006) Dual signaling is differentially activated by different active states of the metabotropic glutamate receptor 1alpha. Proc Natl Acad Sci USA 103:1124–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Terbeck S, Akkus F, Chesterman LP, Hasler G (2015) The role of metabotropic glutamate receptor 5 in the pathogenesis of mood disorders and addiction: combining preclinical evidence with human Positron Emission Tomography (PET) studies. Front Neurosci 9:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  397. Tessari M, Pilla M, Andreoli M, Hutcheson DM, Heidbreder CA (2004) Antagonism at metabotropic glutamate 5 receptors inhibits nicotine- and cocaine-taking behaviours and prevents nicotine-triggered relapse to nicotine-seeking. Eur J Pharmacol 499:121–133. [DOI] [PubMed] [Google Scholar]
  398. Thomas NK, Wright RA, Howson PA, Kingston AE, Schoepp DD, Jane DE (2001) (S)-3,4-DCPG, a potent and selective mGlu8a receptor agonist, activates metabotropic glutamate receptors on primary afferent terminals in the neonatal rat spinal cord. Neuropharmacology 40:311–318. [DOI] [PubMed] [Google Scholar]
  399. Thompson JM, Neugebauer V (2017) Amygdala plasticity and pain. Pain Res Manag 2017:8296501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  400. Thoreson WB, Miller RF (1994) Actions of (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid (1S,3R-ACPD) in retinal ON bipolar cells indicate that it is an agonist at L-AP4 receptors. J Gen Physiol 103:1019–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Tizzano JP, Griffey KI, Schoepp DD (1995) Receptor subtypes linked to metabotropic glutamate receptor agonist-mediated limbic seizures in mice. Ann N Y Acad Sci 765:230–235, discussion 248. [DOI] [PubMed] [Google Scholar]
  402. 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 62:405–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Tripathi A, Kar SK, Shukla R (2018) Cognitive deficits in schizophrenia: understanding the biological correlates and remediation strategies. Clin Psychopharmacol Neurosci 16:7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Tsai CJ, Del Sol A, Nussinov R (2009) Protein allostery, signal transmission and dynamics: a classification scheme of allosteric mechanisms. Mol Biosyst 5:207–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Turner BD, Rook JM, Lindsley CW, Conn PJ, Grueter BA (2018) mGlu1 and mGlu5 modulate distinct excitatory inputs to the nucleus accumbens shell. Neuropsychopharmacology 43:2075–2082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  406. Tzschentke TM, Schmidt WJ (2003) Glutamatergic mechanisms in addiction. Mol Psychiatry 8:373–382. [DOI] [PubMed] [Google Scholar]
  407. Ugolini A, Corsi M, Bordi F (1999) Potentiation of NMDA and AMPA responses by the specific mGluR5 agonist CHPG in spinal cord motoneurons. Neuropharmacology 38:1569–1576. [DOI] [PubMed] [Google Scholar]
  408. Vadasz C, Saito M, Gyetvai BM, Oros M, Szakall I, Kovacs KM, Prasad VV, Toth R (2007) Glutamate receptor metabotropic 7 is cis-regulated in the mouse brain and modulates alcohol drinking. Genomics 90:690–702. [DOI] [PubMed] [Google Scholar]
  409. Valenti O, Conn PJ, Marino MJ (2002) Distinct physiological roles of the Gq-coupled metabotropic glutamate receptors Co-expressed in the same neuronal populations. J Cell Physiol 191:125–137. [DOI] [PubMed] [Google Scholar]
  410. Vardi T, Fina M, Zhang L, Dhingra A, Vardi N (2011) mGluR6 transcripts in non-neuronal tissues. J Histochem Cytochem 59:1076–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  411. Vardigan JD, Huszar SL, McNaughton CH, Hutson PH, Uslaner JM (2010) MK-801 produces a deficit in sucrose preference that is reversed by clozapine, D-serine, and the metabotropic glutamate 5 receptor positive allosteric modulator CDPPB: relevance to negative symptoms associated with schizophrenia? Pharmacol Biochem Behav 95:223–229. [DOI] [PubMed] [Google Scholar]
  412. Varty GBGrilli MForlani AFredduzzi SGrzelak MEGuthrie DHHodgson RALu SXNicolussi EPond AJ, et al. (2005) The antinociceptive and anxiolytic-like effects of the metabotropic glutamate receptor 5 (mGluR5) antagonists, MPEP and MTEP, and the mGluR1 antagonist, LY456236, in rodents: a comparison of efficacy and side-effect profiles. Psychopharmacology (Berl) 179:207–217. [DOI] [PubMed] [Google Scholar]
  413. Vieira E, Huwyler J, Jolidon S, Knoflach F, Mutel V, Wichmann J (2005) 9H-Xanthene-9-carboxylic acid [1,2,4]oxadiazol-3-yl- and (2H-tetrazol-5-yl)-amides as potent, orally available mGlu1 receptor enhancers. Bioorg Med Chem Lett 15:4628–4631. [DOI] [PubMed] [Google Scholar]
  414. Walker AG, Wenthur CJ, Xiang Z, Rook JM, Emmitte KA, Niswender CM, Lindsley CW, Conn PJ (2015) Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proc Natl Acad Sci USA 112:1196–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  415. Wang Z, Neely R, Landisman CE (2015) Activation of group I and group II metabotropic glutamate receptors causes LTD and LTP of electrical synapses in the rat thalamic reticular nucleus. J Neurosci 35:7616–7625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  416. Whitlock JR, Heynen AJ, Shuler MG, Bear MF (2006) Learning induces long-term potentiation in the hippocampus. Science 313:1093–1097. [DOI] [PubMed] [Google Scholar]
  417. Wierońska JM, Stachowicz K, Acher F, Lech T, Pilc A (2012a) Opposing efficacy of group III mGlu receptor activators, LSP1-2111 and AMN082, in animal models of positive symptoms of schizophrenia. Psychopharmacology (Berl) 220:481–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Wierońska JM, Stachowicz K, Brański P, Pałucha-Poniewiera A, Pilc A (2012b) On the mechanism of anti-hyperthermic effects of LY379268 and LY487379, group II mGlu receptors activators, in the stress-induced hyperthermia in singly housed mice. Neuropharmacology 62:322–331. [DOI] [PubMed] [Google Scholar]
  419. Winder DG, Conn PJ (1996) Roles of metabotropic glutamate receptors in glial function and glial-neuronal communication. J Neurosci Res 46:131–137. [DOI] [PubMed] [Google Scholar]
  420. Winder DG, Ritch PS, Gereau RW IV, Conn PJ (1996) Novel glial-neuronal signalling by coactivation of metabotropic glutamate and beta-adrenergic receptors in rat hippocampus. J Physiol 494:743–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Wood CMNicolas CSChoi SLRoman ENylander IFernandez-Teruel AKiianmaa KBienkowski Pde Jong TRColombo G, et al. (2017) Prevalence and influence of cys407* Grm2 mutation in Hannover-derived Wistar rats: mGlu2 receptor loss links to alcohol intake, risk taking and emotional behaviour. Neuropharmacology 115:128–138. [DOI] [PubMed] [Google Scholar]
  422. Woźniak M, Acher F, Marciniak M, Lasoń-Tyburkiewicz M, Gruca P, Papp M, Pilc A, Wierońska JM (2016) Involvement of GABAB receptor signaling in antipsychotic-like action of the novel orthosteric agonist of the mGlu4 receptor, LSP4-2022. Curr Neuropharmacol 14:413–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. Wu HWang CGregory KJHan GWCho HPXia YNiswender CMKatritch VMeiler JCherezov V, et al. (2014) Structure of a class C GPCR metabotropic glutamate receptor 1 bound to an allosteric modulator. Science 344:58–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Xi ZX, Baker DA, Shen H, Carson DS, Kalivas PW (2002) Group II metabotropic glutamate receptors modulate extracellular glutamate in the nucleus accumbens. J Pharmacol Exp Ther 300:162–171. [DOI] [PubMed] [Google Scholar]
  425. Xi ZX, Kiyatkin M, Li X, Peng XQ, Wiggins A, Spiller K, Li J, Gardner EL (2010) N-acetylaspartylglutamate (NAAG) inhibits intravenous cocaine self-administration and cocaine-enhanced brain-stimulation reward in rats. Neuropharmacology 58:304–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  426. Xi ZX, Shen H, Baker DA, Kalivas PW (2003) Inhibition of non-vesicular glutamate release by group III metabotropic glutamate receptors in the nucleus accumbens. J Neurochem 87:1204–1212. [DOI] [PubMed] [Google Scholar]
  427. Xiang Z, Lv X, Maksymetz J, Stansley BJ, Ghoshal A, Gogliotti RG, Niswender CM, Lindsley CW, Conn PJ (2019) mGlu5 positive allosteric modulators facilitate long-term potentiation via disinhibition mediated by mGlu5-endocannabinoid signaling. ACS Pharmacol Transl Sci 2:198–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Xie X, Steketee JD (2008) Repeated exposure to cocaine alters the modulation of mesocorticolimbic glutamate transmission by medial prefrontal cortex Group II metabotropic glutamate receptors. J Neurochem 107:186–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  429. Xu J, Antion MD, Nomura T, Kraniotis S, Zhu Y, Contractor A (2014) Hippocampal metaplasticity is required for the formation of temporal associative memories. J Neurosci 34:16762–16773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Yin S, Niswender CM (2014) Progress toward advanced understanding of metabotropic glutamate receptors: structure, signaling and therapeutic indications. Cell Signal 26:2284–2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  431. Yohn SEFoster DJCovey DPMoehle MSGalbraith JGarcia-Barrantes PMCho HPBubser MBlobaum ALJoffe ME, et al. (2020) Activation of the mGlu1 metabotropic glutamate receptor has antipsychotic-like effects and is required for efficacy of M4 muscarinic receptor allosteric modulators. Mol Psychiatry 25:2786–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Yu SP, Sensi SL, Canzoniero LM, Buisson A, Choi DW (1997) Membrane-delimited modulation of NMDA currents by metabotropic glutamate receptor subtypes 1/5 in cultured mouse cortical neurons. J Physiol 499:721–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Zangrandi L, Schmuckermair C, Ghareh H, Castaldi F, Heilbronn R, Zernig G, Ferraguti F, Ramos-Prats A (2021) Loss of mGluR5 in D1 receptor-expressing neurons improves stress coping. Int J Mol Sci 22:7826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Zeron MM, Hansson O, Chen N, Wellington CL, Leavitt BR, Brundin P, Hayden MR, Raymond LA (2002) Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington’s disease. Neuron 33:849–860. [DOI] [PubMed] [Google Scholar]
  435. Zhai J, Tian MT, Wang Y, Yu JL, Köster A, Baez M, Nisenbaum ES (2002) Modulation of lateral perforant path excitatory responses by metabotropic glutamate 8 (mGlu8) receptors. Neuropharmacology 43:223–230. [DOI] [PubMed] [Google Scholar]
  436. Zhang Y, Rodriguez AL, Conn PJ (2005) Allosteric potentiators of metabotropic glutamate receptor subtype 5 have differential effects on different signaling pathways in cortical astrocytes. J Pharmacol Exp Ther 315:1212–1219. [DOI] [PubMed] [Google Scholar]
  437. Zho WM, You JL, Huang CC, Hsu KS (2002) The group I metabotropic glutamate receptor agonist (S)-3,5-dihydroxyphenylglycine induces a novel form of depotentiation in the CA1 region of the hippocampus. J Neurosci 22:8838–8849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Zhou ZKarlsson CLiang TXiong WKimura MTapocik JDYuan QBarbier EFeng AFlanigan M, et al. (2013) Loss of metabotropic glutamate receptor 2 escalates alcohol consumption. Proc Natl Acad Sci USA 110:16963–16968. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Pharmacological Reviews are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES