Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 1.
Published in final edited form as: Adv Pharmacol. 2011;62:37–77. doi: 10.1016/B978-0-12-385952-5.00010-5

Allosteric Modulation of Metabotropic Glutamate Receptors

Douglas J Sheffler *, Karen J Gregory *,, Jerri M Rook *, P Jeffrey Conn *
PMCID: PMC3787868  NIHMSID: NIHMS513990  PMID: 21907906

Abstract

The development of receptor subtype-selective ligands by targeting allosteric sites of G protein-coupled receptors (GPCRs) has proven highly successful in recent years. One GPCR family that has greatly benefited from this approach is the metabotropic glutamate receptors (mGlus). These family C GPCRs participate in the neuromodulatory actions of glutamate throughout the CNS, where they play a number of key roles in regulating synaptic transmission and neuronal excitability. A large number of mGlu subtype-selective allosteric modulators have been identified, the majority of which are thought to bind within the transmembrane regions of the receptor. These modulators can either enhance or inhibit mGlu functional responses and, together with mGlu knockout mice, have furthered the establishment of the physiologic roles of many mGlu subtypes. Numerous pharmacological and receptor mutagenesis studies have been aimed at providing a greater mechanistic understanding of the interaction of mGlu allosteric modulators with the receptor, which have revealed evidence for common allosteric binding sites across multiple mGlu subtypes and the presence for multiple allosteric sites within a single mGlu subtype. Recent data have also revealed that mGlu allosteric modulators can display functional selectivity toward particular signal transduction cascades downstream of an individual mGlu subtype. Studies continue to validate the therapeutic utility of mGlu allosteric modulators as a potential therapeutic approach for a number of disorders including anxiety, schizophrenia, Parkinson’s disease, and Fragile X syndrome.

I. Introduction

Despite their tractability as drug targets, the majority of G protein-coupled receptor (GPCR)-based drug discovery programs have failed to yield highly selective compounds. Further, CNS disorders represent a therapeutic area with one of the highest rates of attrition in drug discovery (Kola & Landis, 2004). The traditional approach to targeting GPCRs in drug discovery has been to target the endogenous ligand (orthosteric)-binding site, to either mimic or block the actions of the endogenous neurotransmitter or hormone in a competitive manner. However, this approach has suffered from a lack of suitably subtype-selective ligands, both as tools to probe physiology and pathophysiology experimentally, and as therapeutic candidates. An alternative approach is to target allosteric sites that are topographically distinct from the orthosteric site, to either enhance (positive allosteric modulators, PAMs) or inhibit (negative allosteric modulators, NAMs) receptor activation. These allosteric modulators, offer a number of potential advantages over their orthosteric counterparts. In many cases, allosteric sites consist of regions on the receptor that show greater sequence divergence than orthosteric sites and as such have greater potential for subtype-selective ligand development. Further, in the case of an allosteric modulator that has no intrinsic activity, there is the capacity to “fine-tune” the response to the endogenous ligand, thereby retaining the spatial and temporal aspects of neurotransmission. Alternatively, allosteric modulators can also have intrinsic efficacy, activating the receptor alone (allosteric agonists) or neutral efficacy, having no effects on the receptor alone but competing with the activity of other allosteric modulators. Because the pharmacological effects of allosteric ligands are limited by their cooperativity, there is a ceiling level to their effect, which may provide greater margin of safety in the case of overdose. One disadvantage of allosteric modulators is that unlike orthosteric ligands, pure allosteric modulators with no intrinsic efficacy rely on the presence of endogenous ligand for efficacy.

Targeting allosteric sites to either enhance or inhibit receptor activation has proven to be highly successful for ligand-gated ion channels. For example, the mechanism of action of benzodiazepines is allosteric enhancement of GABAA receptor activity, which provides a safe and effective treatment for anxiety and sleep disorders (Mohler et al., 2002). Two GPCR allosteric modulators have now entered the market, demonstrating the clinical validity of this approach. The first of these modulators, Cinacalcet, is a PAM of the calcium-sensing receptor (CaSR) and was approved in 2004 for the treatment of hyperparathyroidism, a disease associated with CaSR deficiency (Lindberg et al., 2005). The second, Maraviroc, stabilizes C–C chemokine receptor type 5 (CCR5) receptor conformations that have a lower affinity for the HIV virus, allosterically inhibiting CCR5-dependent entry of HIV-1 into cells (Dorr et al., 2005) and was approved for the treatment of HIV infections in 2007. Consequently, discovery and characterization of GPCR allosteric modulators have gained significant momentum in the past two decades and represent exciting novel means of targeting therapeutically relevant GPCRs. Arguably, one of the most well-studied GPCR families with respect to allosteric modulation are the metabotropic glutamate receptors (mGlus). Indeed, the full spectrum of allosteric ligands has been discovered for these receptors.

II. Metabotropic Glutamate Receptors

The neuromodulatory actions of the major neurotransmitter glutamate within the CNS are mediated by activation of the mGlus. There are eight mGlu subtypes, and with the exception of mGlu6, which is primarily expressed in the retina, mGlus are expressed throughout the CNS. The individual subtypes show varied distribution in different brain areas, can be found both pre and postsynaptically (Fig. 1), participate in many different CNS processes, and are attractive therapeutic targets for a number of neurological and psychiatric diseases and disorders (Table I). The mGlus are family C GPCRs and the eight mGlu subtypes are generally classified into three groups based on sequence homology and pharmacology: Group I are mGlu1 and mGlu5, Group II are mGlu2 and mGlu3, and Group III are mGlu4, mGlu6, mGlu7, mGlu8. Within the same group, mGlus show ~70% sequence identity, whereas between groups, this conservation falls to ~45% (Niswender & Conn, 2010). The Group I mGlus preferentially couple to the Gq/11 family of G proteins, activating phosphoinositide hydrolysis and calcium mobilization as their major signaling mechanism. In contrast, Group II and Group III mGlus preferentially couple to Gi/o and inhibit adenylyl cyclases.

FIGURE 1.

FIGURE 1

Schematic representation of mGlus at the synapse. Group I mGlus, mGlu1 and mGlu5, are generally localized postsynaptically, while Group II mGlus (mGlu2 and mGlu3) and Group III mGlus (mGlu4, mGlu7, and mGlu8) are localized in presynaptic locations, although exceptions occur. mGlu6 is not shown in this figure as it is only found localized postsynaptically in the retina. On presynaptic terminals, Group II and III receptors often function to inhibit neurotransmitter release, whereas Group I mGlus promote release when present. Postsynaptically, Group I mGlus signal via Gq proteins to increase intracellular calcium, whereas Group II mGlus signal via Gi/o proteins to inhibit cAMP production. mGlu5 activation also can potentiate N-methyl-D-aspartate (NMDA) glutamate receptor currents. In addition to the NMDA receptor, the other ionotropic glutamate receptors, the α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) and kainite receptors, respond to glutamate with increases in intracellular sodium or calcium, promoting cell excitability. In the CNS, mGlu3 and mGlu5 have also been found to be expressed on glia.

TABLE I.

Representative Allosteric Modulators of mGlus and Their Potential Therapeutic Applications

mGlu subtype Intervention Therapeutic indication Representative modulators In vitro potency (nM) In vitro affinity (nM) Reference(s)
1 NAM Pain, anxiety, drug abuse CPCCOEt 3400–6500 4900 Ott et al. (2000), Litschig et al. (1999), Suzuki et al. (2007a)
FTIDC 6 2 Suzuki et al. (2007a, 2009)
2 Agonist/PAM Anxiety, drug abuse, schizophrenia BINA 98–347 Galici et al. (2006), Jin et al. (2010)
LY487379 270–1700 Johnson et al. (2003), Schaffhauser et al. (2003)
3 Agonist/PAM Neuroprotection None reported
4 Agonist/PAM Movement disorders, Parkinson’s disease PHCCCa 4100–6000 Maj et al. (2003), Marino et al. (2003), Niswender et al. (2008a)
VU0155041 693–798 Niswender et al. (2008a)
5 NAM Anxiety, chronic pain, depression, fragile X Syndrome, GERD, migraine, drug abuse MPEPb 2–36 3–12 Malherbe et al. (2006, 2003a), Gasparini et al. (1999), Cosford et al. (2003a, 2003b), Rodriguez et al. (2005), Porter et al. (2005)
MTEP 5–47 5–16 Malherbe et al. (2006), Cosford et al. (2003a, 2003b), Porter et al. (2005)
Fenobam 38–58 31–61 Ceccarelli et al. (2007), Porter et al. (2005)
VU0285683 24 17 Rodriguez et al. (2010)
5 PAM Cognition disorders, schizophrenia ADX47273 170–479 4300 de Paulis et al. (2006), Liu et al. (2008), Bradley et al. (2009), Rosenbrock et al. (2010)
CDPPB 27–347 1300–3800 Kinney et al. (2005), de Paulis et al. (2006), Chen et al. (2007), Bradley et al. (2009)
CPPHAc 239–810 O’Brien et al. (2004), Zhang et al. (2005), Chen et al. (2008)
VU0360172 16 Rodriguez et al. (2010)
7 NAM Depression, anxiety MMPIP 26–718 Suzuki et al. (2007b), Niswender et al. (2010)
7 Agonist/PAM Anxiety, depression, epilepsy, drug abuse AMN082 64–240 Suzuki et al. (2007b), Mitsukawa et al. (2005), Fendt et al. (2008)
8 Agonist/PAM Anxiety, drug abuse, epilepsy, pain No selective allosteric ligands reported
a

PHCCC is also a weak mGlu1 NAM (Annoura et al., 1996) and an mGlu6 agonist (Beqollari & Kammermeier, 2008).

b

MPEP is also a weak mGlu4 PAM (Mathiesen et al., 2003).

c

CPPHA is also a weak mGlu4 and mGlu8 NAM (O’Brien et al., 2004) and an mGlu1 PAM (Chen et al., 2008).

A. Structural Features of Metabotropic Glutamate Receptors

GPCRs are predicted to share a common topology consisting of seven transmembrane-spanning α-helical domains, an extracellular N terminus, and intracellular C terminus. Additional structural features of mGlus include a large extracellular N-terminal domain, termed the venus flytrap domain (VFD), that contains the endogenous ligand-binding site (Pin et al., 2003) and a cysteine-rich domain that links the VFD to the trans-membrane-spanning α-helices by a conserved disulfide bridge (Fig. 2). Crystal structures of the N-terminal domains of mGlu1, mGlu3, and mGlu7 have been solved and suggest that the VFD is made up of two lobes (Kunishima et al., 2000; Muto et al., 2007; Tsuchiya et al., 2002), forming a clam shell-shaped structure. Glutamate is thought to bind between the two lobes with the two globular domains closing into a stable conformation with glutamate inside (Bessis et al., 2000, 2002; Kunishima et al., 2000; Tsuchiya et al., 2002). Further, evidence suggests that the mGlus dimerize via interactions between their VFDs, and that activation of mGlus requires the binding of two glutamate molecules, one to each protomer (Kniazeff et al., 2011). Conformational changes induced by glutamate in the VFD are thought to be transmitted to the transmembrane spanning domains via the cysteine-rich domain to promote coupling to intracellular G proteins and activation of second messenger pathways (Liu et al., 2004; Muto et al., 2007; Rondard et al., 2006).

FIGURE 2.

FIGURE 2

Representation of the mGlu structure. mGlus possess a large N-terminal extracellular domain that contains the orthosteric binding site of the endogenous ligand glutamate, referred to as the venus flytrap domain (VFD). The extracellular VFD is connected to seven transmembrane (7TM) domains via a cysteine-rich domain. Allosteric ligands bind to sites other than the orthosteric glutamate binding site, such as within the 7TM domain. Group I mGlus couple to Gq/11 proteins, whereas the Group II and Group III mGlus couple to Gi/o proteins.

B. Localization and Functional Roles of the mGlus

1. Group I mGlus

Group I mGlus, comprising mGlu1 and mGlu5, are extensively expressed throughout the CNS in neurons, and mGlu5 is expressed in both neurons and glia. mGlu1 expression is most intense in Purkinje cells of the cerebellar cortex and the olfactory bulb with strong expression in neurons of the lateral septum, globus pallidus, ventral pallidum, most thalamic nuclei, as well as the substantia nigra. Localization of mGlu5 is greatest in corticolimbic areas responsible for controlling higher cognitive function including the striatum, hippocampus, cerebral cortex, septal nuclei as well as the thalamus, and olfactory bulb (Ferraguti & Shigemoto, 2006). While Group I mGlus are predominantly found postsynaptically, where they increase neuronal excitability, it is important to note that Group I mGlus localized presynaptically can increase or decrease neurotransmitter release (Pinheiro & Mulle, 2008) (Fig. 1). Additionally, mGlu1 and mGlu5 subtypes can have differential functional roles within a single neuronal population as is the case in CA1 pyramidal cells (Mannaioni et al., 2001). The Group I mGlus play important roles in synaptic plasticity by facilitating both long-term depression (LTD) and potentiation (LTP) of synaptic strength as well as inducing nonsynaptic conductances leading to enhanced neuronal excitability (Anwyl, 1999; Bellone et al., 2008).

Each of the mGlus has been genetically deleted in mice. The phenotypes revealed by studies conducted with these mGlu knockout (KO) animals provide insight into the potential physiological roles of individual mGlus in a wide variety of brain systems as well as their potential as targets for novel therapeutic strategies. The first mGlu subtype deleted in mice was mGlu1. While the overall gross brain morphology of these animals appears normal, they exhibit several phenotypes. As previously discussed, mGlu1 is most abundantly expressed in Purkinje cells of the cerebellar cortex. Mice lacking mGlu1 demonstrate cerebellar ataxic gait characterized by deficits in LTD in the cerebellum (Aiba et al., 1994b). Most adult Purkinje cells are individually innervated by a single climbing fiber as a consequence of developmental regulatory regression forming a strong excitatory synapse. Interestingly, mGlu1-deficient mice have abnormal regression of climbing fibers, resulting in innervations of multiple climbing fibers onto single Purkinje cells, suggesting mGlu1 is critical for guiding proper excitatory synapse formation in the cerebellum (Kano et al., 1997).

High levels of mGlu1 expression are also found within the hippocampus, suggesting a potential role of this receptor in learning and memory. Electrophysiological studies in mGlu1 KO mice reveal significantly reduced LTP in hippocampal slices. These studies also show impairment in the acquisition or retention of memory in a context-dependent fear conditioning task, suggesting deficits in hippocampal-mediated learning and memory (Aiba et al., 1994a). Recently, recordings at the CA3–CA1 synapse within the hippocampus during the acquisition of an associative learning task in mGlu1 KO mice found impaired LTP as well as the inability of the animal to learn the task (Gil-Sanz et al., 2008).

Extensive studies using mGlu5 KO mice implicate this receptor’s role in cognition, addiction, anxiety, chronic pain, and obesity. Both mGlu1 and mGlu5 KO mice demonstrate deficits in prepulse inhibition (PPI) of the acoustic startle reflex, a measure of sensorimotor gating impaired in patients with schizophrenia (Brody et al., 2003, 2004b; Gray et al., 2009), which can be reversed by antipsychotic treatment (Brody et al., 2004a). Deletion of mGlu5 also results in short-term spatial memory performance deficits in the Y-maze (Gray et al., 2009), a reduction in hippocampal CA1 LTP, and impaired performance in cognitive tasks (Lu et al., 1997). These studies, together with pharmacological studies demonstrating the involvement of mGlu5 in synaptic plasticity, including LTP and LTD (Ayala et al., 2009), support a role for mGlu5 in learning and memory (Homayoun & Moghaddam, 2010). In animal models of addiction, mGlu5 KO mice do not self-administer cocaine and do not demonstrate hyperlocomotor activity following cocaine treatment (Chiamulera et al., 2001), demonstrating that mGlu5 may play a role in addiction. Further, mGlu5 KO mice also display attenuation of stress-induced hyperthermia, an animal model of anxiety (Brodkin et al., 2002). Recently, deletion of mGlu5 was also used to demonstrate the role of mGlu5 signaling in the amygdala in the modulation of persistent pain due to peripheral inflammation (Kolber et al., 2010). Finally, mGlu5 KO mice weigh less than littermate controls, eat less when challenged with food deprivation, and are resistant to weight gain and increased insulin levels induced by a high-fat diet (Bradbury et al., 2005) indicating a potential role of mGlu5 in obesity.

2. Group II mGlus

The Group II mGlus, mGlu2 and mGlu3, are widely distributed in the CNS (Reviewed in Ferraguti & Shigemoto, 2006; Harrison et al., 2008) and in the periphery (Reviewed in Julio-Pieper et al., 2011). Generally, mGlu2 and mGlu3 are expressed presynaptically where they modulate neurotransmitter release (Fig. 1). In addition, they are also found at postsynaptic sites where they can induce hyperpolarization (Muly et al., 2007). In addition to these synaptic localizations, mGlu3 is also expressed in glia (Mudo et al., 2007; Ohishi et al., 1993; Tamaru et al., 2001). Similar to the Group I mGlus, mGlu2 and mGlu3 KO mice have been heavily utilized in order to define the individual roles of the Group II mGlus in a variety of physiologic processes. For example, mGlu2 KO mice show a loss of Group II agonist-induced reversal of PCP-induced hyperlocomotion (Spooren et al., 2000), loss of the anxiolytic effects of a Group II agonist (Linden et al., 2005), an enhanced responsiveness to cocaine (Morishima et al., 2005), alterations in synaptic transmission in a number of regions including the CA1 region of the hippocampus and the dentate gyrus (Kew et al., 2002; Yokoi et al., 1996), and loss of Group II agonist antipsychotic activity in numerous rodent models (Fell et al., 2008; Woolley et al., 2008). mGlu3 KO mice also show decreased efficacy of Group II agonists in anxiolytic models (Linden et al., 2005), an increase in basal hippocampal c-Fos expression (Linden et al., 2006), and a loss of Group II agonist-induced neuroprotection by astrocytes against NMDA excitotoxicity (Corti et al., 2007).

3. Group III mGlus

The Group III mGlus (mGlu4, mGlu6, mGlu7, and mGlu8), with the exception of mGlu6, which is localized postsynaptically on retinal ON bipolar cells (Nakajima et al., 1993), are mainly expressed presynaptically in neurons diffusely distributed throughout the CNS (Fig. 1). mGlu4 is highly expressed in the cerebellum, with lower levels of expression in the hippocampus, basal ganglia, and olfactory bulb (Lavreysen & Dautzenberg, 2008). mGlu4 expression is also found peripherally in pancreatic islet cells (Uehara et al., 2004) and taste buds along with a shorter mGlu4 splice variant (Toyono et al., 2002). Consistent with expression distribution, deleting the mGlu4 gene results in mice with impaired cerebellar synaptic plasticity and deficits in learning of complicated motor tasks (Pekhletski et al., 1996) and spatial memory performance (Gerlai et al., 1998). mGlu7 and mGlu8 are widely distributed throughout the brain. In particular, mGlu7 is localized to active zones of synapses and has an extremely low affinity for glutamate. Given this low glutamate affinity and localization, it has been proposed that mGlu7 is only activated in the presence of very high glutamate levels and thus serves to prevent overstimulation by glutamate (Kinoshita et al., 1998; Shigemoto et al., 1997). This hypothesis is further supported by the epileptic phenotype of mGlu7 KO mice (Sansig et al., 2001). Additionally, mice lacking mGlu7 exhibit deficits in memory and learning (Bushell et al., 2002; Callaerts-Vegh et al., 2006; Goddyn et al., 2008; Holscher et al., 2004, 2005; Masugi et al., 1999) and have been implicated in disorders such as anxiety and depression (Cryan et al., 2003; Stachowicz et al., 2008). mGlu8 is expressed broadly throughout the brain, albeit at lower levels than both mGlu4 and mGlu7. mGlu8 KO mice displayed increased anxiety and weight gain (Duvoisin et al., 2005; Linden et al., 2002). mGlu8 is also localized postsynaptically within the peripheral cells of the gut and pancreas and has been implicated in gastrointestinal motility and insulin secretion (Tong & Kirchgessner, 2003; Tong et al., 2002). Unlike other Group III subtypes, the mGlu6 signaling cascade has been shown to involve Go protein subunits (Dhingra et al., 2000). Mice lacking mGlu6 demonstrate deficits in ON response to light stimulation (Masu et al., 1995; Sugihara et al., 1997).

III. Pharmacological Profiles of mGlu Allosteric Modulators

A. Group I mGlus

The first mGlu allosteric modulator was identified when the selective mGlu1 antagonist CPCCOEt (Annoura et al., 1996) was determined to act via a noncompetitive mechanism (Litschig et al., 1999), which marked a major advance in supporting the rationale of targeting allosteric sites for discovery of highly subtype-selective mGlu antagonists. Several structurally distinct mGlu1 NAMs with nanomolar potencies have been published since, including Bay 36-7620 (Carroll et al., 2001), JNJ16259685 (Lavreysen et al., 2003), YM298198 (Kohara et al., 2005), and FTIDC (Suzuki et al., 2007a), which possess adequate pharmacokinetic properties for in vivo characterization. Highly selective mGlu1 PAMs have also been developed and among these compounds are Ro 67-7476 and VU71 (Hemstapat et al., 2006; Knoflach et al., 2001).

The other Group I receptor, mGlu5, has been extensively investigated, and several potent and selective NAMs are known. SIB-1757 and SIB-1893 were originally reported (Varney et al., 1999), with the subsequent structural analogs MPEP (Gasparini et al., 1999) and MTEP (Cosford et al., 2003b) providing increased potency and selectivity as well as brain penetration as compared to their predecessors. While many potent, highly selective mGlu5 NAMs have now been reported, MTEP and MPEP remain the most commonly used antagonists for in vivo characterization of this receptor. However, Rodriguez et al. (2010) recently reported discovery of VU0285683 as a potent, in vivo active, mGlu5 NAM with improved pharmacokinetic properties for systemic dosing.

In addition to mGlu5 NAMs, many mGlu5 PAMs have been identified. These include DFB, CPPHA, CDPPB, VU29, and ADX47273 (Conn et al., 2009). Although DFB and CPPHA do not have suitable potencies or solubility in physiological buffers for systemic administration and assessment of mGlu5 function in vivo, newer generation PAMs are proving highly useful in evaluating the role of mGlu5 in animal models. Most recently, VU0360172 was reported as a potent, selective mGlu5 PAM with properties that are much more favorable for in vivo studies than was the case for earlier mGlu5 PAMs (Rodriguez et al., 2010). Another pharmacological tool utilized for evaluating the physiological role of mGlu5 is the neutral ligand, 5MPEP, which binds to the MPEP site but has no effects alone. However, 5MPEP blocks the effects of both the allosteric antagonist MPEP and potentiators DFB and CDPPB (Rodriguez et al., 2005).

B. Group II mGlus

Numerous mGlu2 PAMs have been identified, the majority of which are structurally related to BINA and LY487379 (Reviewed in Rudd & McCauley, 2005). Unfortunately, there are currently no reports of selective mGlu3 PAMs. Interestingly, a single report of an mGlu2 PAM displaying mGlu3 PAM activity in a GTPγS binding assay (Govek et al., 2005) has shown that discovery of mGlu3 PAMs may be on the horizon. Compared to the mGlu2 PAMs, only a limited number of Group II NAMs, which are nonselective between mGlu2 and mGlu3 (Hemstapat et al., 2007), have been reported. Chemically, the majority of these Group II NAMs are benzo-diazepinones structurally related to MNI-137 (Hemstapat et al., 2007), with some newer compounds in this series demonstrating improved oral bioavail-ability in rodents (Woltering et al., 2010). Interestingly, LY2389575, an mGlu3 selective NAM, was recently reported (Caraci et al., 2010), providing a key tool compound that will allow further investigation of mGlu3’s physiological function.

C. Group III mGlus

Relative to Group I and Group II mGlus, the pharmacology of Group III mGlus has been the least investigated, due in large part to the difficulty in developing subtype-selective compounds. While several orthosteric compounds selective for Group III receptors have been identified, generally these compounds do not discriminate between subtypes and have relatively low potencies (Schoepp et al., 1999). However, exciting new progress has been made in developing subtype-selective Group III mGlu allosteric modulators with the development of mGlu4 and mGlu7 subtype-selective compounds. PHCCC is a pure positive allosteric modulator of mGlu4, potentiating the potency of glutamate at mGlu4 with no direct agonist activity alone (Maj et al., 2003; Marino et al., 2003). Additionally, the mGlu5 NAMs SIB-1893 and MPEP possess mGlu4 PAM activity, although at significantly lower potencies and efficacies (Mathiesen et al., 2003). Recently, two structurally distinct mGlu4 PAMs, VU0155041 and VU0080421, have been identified (Niswender et al., 2008a,b). Significant improvements in potency and selectivity for mGlu4 have been achieved with VU0155041, which is a mixed allosteric agonist/PAM. Moreover, VU0155041 exhibits increased solubility in physiological buffers, providing the opportunity to examine the role of mGlu4 in animal models of various disease states (Niswender et al., 2008a).

AMN082 has been reported as a selective mGlu7 allosteric agonist and is extensively used as an in vivo tool compound (Mitsukawa et al., 2005). Recent studies demonstrating varying activity suggest that the pharmacology of this compound is complex. Reports have shown that AMN082 does not induce calcium mobilization in a cell line coexpressing mGlu7 and a promiscuous G protein (Suzuki et al., 2007b), mGlu7-mediated activation of GIRK potassium channels in human embryonic kidney cells (Ayala et al., 2008), or activation of mGlu7 at the Schaffer collateral-CA1 synapse (Ayala et al., 2008). Therefore, AMN082-mediated activation of mGlu7 may be highly dependent upon specific signaling pathways activated and the system involved. A novel mGlu7 NAM, MMPIP, displays similar nuisances in determining its activity at mGlu7, exhibiting permissive antagonism, varying antagonist ability contingent upon the signaling pathway being investigated (Kenakin, 2005; Niswender, 2008).

IV. Quantifying Allosteric Interactions

The binding of an allosteric modulator has the potential to modulate, either in a positive or negative manner, the binding affinity and/or signaling efficacy of an orthosteric ligand. This is a consequence of changes in the conformation of the receptor when it is simultaneously bound by more than one ligand, resulting in altered “geography” of the orthosteric site and also receptor/protein interfaces. The simplest model of GPCR allosteric interactions, referred to as the allosteric ternary complex model (ATCM; Fig. 3), assumes that allosteric modulator binding to its site changes only the affinity of the orthosteric ligand and vice versa. Within this model, the interaction is governed by the concentration of each ligand, the equilibrium dissociation constants of the orthosteric and allosteric ligands (KA and KB, respectively), and the “cooperativity factor” α, a measure of the magnitude and direction of the allosteric interaction between the two conformationally linked sites (Ehlert, 1988; Stockton et al., 1983). This model has been sufficient to describe the behavior of allosteric modulators for class A GPCRs, however, there is increasing evidence of allosteric modulators that alter signaling efficacy in addition to, or independently of, any effects on orthosteric ligand binding affinity.

FIGURE 3.

FIGURE 3

Models of allosteric interactions. (A) Allosteric ternary complex model (ACTM), (B) Operational model of allosterism. In these models, the affinity (equilibrium dissociation constant) of the orthosteric ligand (A) for the receptor (R) is defined as KA, while the affinity of the allosteric modulator (B) is KB. α is the affinity cooperativity parameter, denoting the direction and magnitude of the allosteric interaction. The pharmacological effect or stimulus arising from the orthosteric agonist occupied receptor is SA, whereas that arising from the modulator occupied receptor is SB. β is the efficacy cooperativity parameter, describing the change in SA when both orthosteric and allosteric sites are occupied. τA denotes the coupling efficiency of the orthosteric ligand, Em represents the maximal system response, while n is the slope factor that links occupancy to response.

With the continued discovery and characterization of allosteric modulators for a plethora of GPCRs, it is becoming more and more evident that in addition to, or independently of, modulating affinity and/or efficacy, allosteric ligands can act as agonists in their own right (Langmead & Christopoulos, 2006). Referred to as allosteric agonists, these compounds represent additional therapeutic development options and yet another level of complexity. Further, it is conceivable that an allosteric modulator can possess more than one of these properties concurrently, for example, positive or inverse agonism along with enhancement or inhibition of orthosteric agonist affinity/efficacy, or even have opposing effects on affinity and efficacy (May et al., 2007; Schwartz & Holst, 2007). Such a phenomenon is prevalent within the ion channel field, defined as use-dependent blockade, where an antagonist is more potent at blocking the active firing channel (Winquist et al., 2005). For GPCRs, an example of this is the cannabinoid CB1 receptor allosteric modulator, Org27569, which is a positive allosteric modulator of [3H]CP 55940 binding but a NAM of CP 55940 function (Price et al., 2005). The ATCM has been extended into an allosteric “two-state” model (ATSM) to account for such allosteric effects on efficacy (Hall, 2000). Further, Parmentier et al. (2002) have suggested an alternate extension of the ATCM that accounts for the fact that class C GPCRs have very distinct ligand binding and effector coupling domains and proposes an allosteric interaction between these two domains (Parmentier et al., 2002). Indeed, mGlu allosteric modulators are generally efficacy modulators only, most likely due to the fact that the orthosteric and allosteric binding sites are in very separate domains of the receptor.

While the ATSM and the model proposed by Parmentier and colleagues describe the multitude of effects an allosteric modulator may have on ligand–receptor interactions and functional properties, due to the large number of parameters, they are not amenable to fitting experimental biological data. To this end, an operational model of allosterism (Fig. 3) was recently reported that incorporates allosteric agonism as well as the capacity to modulate both efficacy and affinity (Leach et al., 2007; May et al., 2007). In this model, two cooperativity parameters describe the cooperativity between two interacting ligands, α (affinity) and β (efficacy). For each set of ligands, these parameters should be constant regardless of the measure of GPCR function, unless there is pathway specific modulation occurring, in which case the β values will change. Clearly, allosteric interactions can be considerably complex presenting challenges with respect to detection strategies and subsequent data interpretation.

V. Structural Determinants of mGlu Allosteric Modulator Binding

Through the use of chimeric and truncated receptor constructs, all currently identified allosteric modulators of mGlus are known to bind within the transmembrane-spanning regions of the receptor (Brauner-Osborne et al., 1999; Carroll et al., 2001; Gasparini et al., 2001; Goudet et al., 2004; Knoflach et al., 2001; Litschig et al., 1999; Maj et al., 2003; Mitsukawa et al., 2005; Pagano et al., 2000). Interestingly, truncation of the N-terminal extracellular VFD, but retention of an intact transmembrane region and a functional C terminus, yields a receptor that couples to G proteins and can be positively or negatively regulated by ligands, like any class A GPCR (Goudet et al., 2004), but which no longer responds to orthosteric ligands. Importantly, allosteric modulators retain activity in cells expressing the truncated receptor, PAMs are agonists, and NAMs become inverse agonists (Chen et al., 2007, 2008; Suzuki et al., 2007a). In the absence of a crystal structure of the transmembrane spanning regions of a class C GPCR, experimental evidence suggests that this region consists of seven transmembrane-spanning α-helices (Bhave et al., 2003), despite the low sequence identity (less than 20%) between the different classes of GPCRs. This predicted common architecture between the class A and class C GPCRs has provided the basis for the use of class A crystal structures as templates for homology models of the transmembrane-spanning region of class C GPCRs. Given the lack of sequence identity between classes, homology models cannot provide structural information at atomic resolution, therefore modeling best occurs synergistically alongside experimental studies of allosteric modulators (Ballesteros & Palczewski, 2001). With the growing number of mammalian GPCR crystal structures to use as templates (Cherezov et al., 2007; Chien et al., 2010; Jaakola et al., 2008; Palczewski et al., 2000; Rasmussen et al., 2007; Rosenbaum et al., 2007; Wu et al., 2010), a number of homology models of mGlus have been published revealing possible binding modes of known allosteric modulators within the top half of the α-helical transmembrane domains (TMs) (Gregory et al., 2010; Malherbe et al., 2003a, 2003b; Miedlich et al., 2004; Ott et al., 2000; Vanejevs et al., 2008). The availability of multiple templates has sparked development of high-throughput homology modeling of GPCRs (Yarnitzky et al., 2010), which has the potential to enrich our understanding of the transmembrane region of these receptors.

A. Common Allosteric Sites on the mGlus

One of the continuing challenges faced in drug discovery in general and for mGlus in particular is establishing suitably subtype-selective ligands. For the most part, mGlu allosteric modulators have displayed better specificity than orthosteric ligands. However, there are a number of examples of allosteric modulators that interact with more than one subtype: MPEP, an mGlu5 NAM, is an mGlu4 PAM (Mathiesen et al., 2003); DFB and CPPHA (mGlu5 PAMs) are also weak mGlu4 NAMs (O’Brien et al., 2003, 2004); PHCCC, an mGlu4 PAM, is also an mGlu1 NAM (Annoura et al., 1996). This lack of selectivity across mGlus from different groups suggests similarities within the allosteric binding pockets. Initial studies seeking to identify allosteric modulator binding sites of mGlus hypothesized that it would resemble the orthosteric site of class A GPCRs, located within the top third of the TMs. Indeed, it was shown that several residues critical for retinal-rhodopsin binding corresponded to important residues for allosteric modulator binding (Malherbe et al., 2003a, 2003b). To date, mutagenesis studies have exclusively investigated the determinants of allosteric modulator binding and cooperativity at mGlu1, mGlu2, and mGlu5 (see Gregory et al., 2010 for review). Specifically, for Group I mGlus, important residues have been identified on the top half of TMs 3, 5, 6, and 7 on the inside faces of helices. Studies performing substitution of nonconserved residues from one mGlu subtype to another have provided further evidence for commonalities in the location of allosteric sites. For example, swapping residues known to be important for allosteric modulation by the selective mGlu1 compounds CPCCOEt and Ro 67-7476 onto mGlu5 result in a gain of function of these compounds at mGlu5 (Knoflach et al., 2001; Litschig et al., 1999). Similarly, exchange of residues important for binding of the mGlu5 NAM, MPEP, onto mGlu1 results in the gain of [3H]MPEP binding (Pagano et al., 2000). Interestingly, molecules from structurally distinct chemical scaffolds including both NAMs and PAMs have shown a tendency to cluster in overlapping binding sites. The mGlu5 neutral MPEP site ligand, 5MPEP, displays a competitive interaction with PAMs from the CDPPB series, which are also able to displace [3H]methoxyPEPy binding (Chen et al., 2007). Clearly, despite the availability of selective allosteric modulators for Group I mGlus, there is evidence that the location of at least one allosteric site is very similar. For mGlu2, important residues for interactions with PAMs have been identified in both TM4 and TM5, and exchange of the equivalent residues onto mGlu3 results in a gain of mGlu2 PAM activity (Hemstapat et al., 2007; Rowe et al., 2008; Schaffhauser et al., 2003). Those residues important for mGlu2 PAM function in TM5 appear to cluster near the Group I mGlu common allosteric site, however, the residues in TM4 do not. It is important to note that mutagenesis data for mGlu2 rely entirely upon functional assays, so it remains to be determined whether these residues are required for binding or for the transmission of cooperativity. Therefore, mapping of residues to TM4 does not necessarily suggest a different localization of an allosteric binding pocket on mGlu2 relative to Group I mGlus.

B. Multiple Allosteric Sites Within a mGlu Subtype

In addition to evidence of common allosteric sites utilized by both PAMs and NAMs at a single mGlu subtype, a number of studies have described allosteric ligands that do not appear to compete with common allosteric sites. The most striking example of this being CPPHA, a Group I PAM, which displaces neither [3H]methoxyPEPy at mGlu5 nor [3H]R214127 at mGlu1 (Chen et al., 2008; O’Brien et al., 2004). Moreover, 5MPEP noncompetitively inhibits CPPHA potentiation of glutamate at mGlu5, suggesting that not only does CPPHA bind to a distinct allosteric site, but also that these two allosteric sites can allosterically regulate one another (Chen et al., 2008). Preliminary site-directed mutagenesis identified a single point mutation at the top of TM1 (F585I/mGlu5; F599I/mGlu1) that abolished potentiation of orthosteric agonists by CPPHA (Chen et al., 2008). Further, residues known to perturb modulators that compete at the common Group I allosteric site (A809V/mGlu5 and V757L/mGlu1) had no effect on CPPHA potentiation. Similarly, two mGlu1 selective PAMs, VU48 and VU71, which are noncompetitive with [3H]R214127 and insensitive to mutations known to affect NAMs that bind the common Group I mGlu allosteric site, have been reported (Hemstapat et al., 2006). Collectively, these data suggest the presence of multiple distinct allosteric sites on Group I mGlus that can be targeted by PAMs. Although limited mutagenesis studies have been performed regarding mGlu2/3 NAMs, mutation of a residue important for mGlu2 PAM functional activity, N735 in TM5, does not affect the functional responses of the mGlu2/3 NAM MNI-137, implying that mGlu2/3 NAMs may occupy a separate allosteric site from the mGlu2 PAMs (Hemstapat et al., 2007). However, as noted earlier, as this study relies upon functional assays alone, it is not evident whether or not this point mutation perturbs PAM affinity or cooperativity. Therefore, an alternative conclusion is that this particular residue is important for positive cooperativity or receptor activation; if this is the case, it is not surprising that this residue would have no effect on a NAM. Although less conclusive, there is evidence in support of multiple allosteric binding pockets for mGlu4 PAMs. The presence of PHCCC, an mGlu4 PAM, does not affect the potentiation concentration–response curve of VU0155041, a structurally distinct PAM, suggesting that these two ligands are not competitive for the same allosteric site (Niswender et al., 2008a).

VI. Functional Selectivity of mGlu Allosteric Modulation

Within the GPCR field, it is becoming increasingly evident that the consequences of receptor activation are not limited to G protein-coupling alone, with the overall cellular response to GPCR activation arising from a myriad of receptor–effector interactions. Indeed, it is now well established that ligand pharmacology is dependent upon the measure of receptor activation employed, a phenomenon referred to by many names including “stimulus trafficking,” “biased agonism,” and “functional selectivity” (Galandrin et al., 2007; Kenakin, 2007; Urban et al., 2007). With multiple binding pockets to exploit and diverse chemical scaffolds being tolerated within common mGlu allosteric binding sites, it is conceivable that allosteric modulators may have the capacity to differentially modulate signaling to different effector pathways. Indeed, in rat cortical astrocytes, CPPHA potentiates signaling to phosphorylation of ERK1/2 versus inositol phosphate turnover differentially, while other PAMs such as VU-29 have similar effects on both pathways (Zhang et al., 2005). Also, MMPIP, an mGlu7 NAM, has differential effects on the receptor activation depending upon the measure of receptor activation and cellular background (Niswender et al., 2010). Further, Ro 67-4853 and other structurally related mGlu1 PAMs potentiate mGlu1-mediated calcium mobilization but act as allosteric agonists for mGlu1-mediated ERK1/2 phosphorylation and stimulation of cAMP (Sheffler & Conn, 2008). Such diversity in functional coupling of allosteric modulator-bound receptors has important implications with respect to the development of therapeutics, particularly for selection of screening assays and lead compounds. If the pathophysiology of a disease of therapeutic end-point can be attributed to the activation or inactivation of a particular pathway, then selective modulation by allosteric ligand may represent a novel, more selective means of therapeutic intervention.

VII. Therapeutic Potential of mGlu Allosteric Modulators

A. mGlu1 NAMs for Pain

Multiple studies have demonstrated the efficacy of mGlu1 NAMs in models of analgesia. For example, the mGlu1 NAM YM-298198 is analgesic in the streptozotocin-induced hyperalgesia mouse model (Kohara et al., 2005), and FTIDC displays analgesic effects in the formalin test (Satow et al., 2008). In addition, the mGlu1 NAM CPCCOEt dose-dependently reversed capsaicin-induced sensitization in spinothalamic tract cells in vivo demonstrating the potential utility of mGlu1-selective NAMs for the treatment of persistent pain associated with spinal sensitization (Neugebauer et al., 1999). Although mGlu1 NAM development has largely focused on potential in pain, mGlu1 NAMs have also demonstrated efficacy in models of anxiety (Satow et al., 2008), antipsychotic activity (Satow et al., 2008), and addiction (Xie et al., 2010).

B. mGlu5 NAMs for Anxiety, Depression, and Fragile X Syndrome

Several preclinical studies suggest a role for mGlu5 NAMs as a therapeutic approach for the treatment of anxiety. MPEP, a selective mGlu5 NAM, demonstrates anxiolytic activity in several rodent behavioral models including marble burying, elevated plus maze, fear-potentiated startle, social exploration, stress-induced hyperthermia, ultrasonic vocalizations, and the Vogel conflict test (Spooren & Gasparini, 2004). Further, Fenobam, a nonbenzodiazepine anxiolytic, was recently found to be a selective negative modulator of mGlu5 (Porter et al., 2005). Fenobam has not only been shown to be efficacious in preclinical model of anxiety (Patel et al., 1982) but also in clinical trials (Pecknold et al., 1982) establishing efficacy of mGlu5 NAMs in the treatment of anxiety. Recent studies have also linked antagonism of mGlu5 with antidepressant activity. MPEP and MTEP exhibit antidepressant activity in multiple animal behavioral models of depression, including the tail suspension test (Tatarczynska et al., 2001) and forced swim test (Li et al., 2006) in mice, as well as passive-learning in olfactory bulbectomized rats (Wieronska et al., 2002). While further testing in chronic depression models is needed, these data suggest exciting new potential in mGlu5 NAMs as novel antidepressants. Although mGlu5 NAM development has largely focused on anxiety disorders, additional studies suggest that mGlu5 NAMs have therapeutic potential in the treatment of addiction, chronic pain, migraine, Alz-heimer’s disease, gastroesophogeal reflux (GERD), as well as fragile X syndrome (FXS) (Alexander & Godwin, 2006; Goudet et al., 2009; Keywood et al., 2009; Lehmann, 2008; Slassi et al., 2005; Thathiah & De Strooper, 2011; Ure et al., 2006; Varney & Gereau, 2002).

In particular, the potential for mGlu5 NAMs for the treatment of FXS has gained momentum through recent preclinical and clinical studies. FXS is the most common inherited form of mental retardation and autism, which is caused by the transcriptional silencing of the FMR1 gene due to an expansion of a CGG repeat. The FMR1 gene product, fragile X mental retardation protein (FMRP), negatively regulates local protein synthesis in neuronal dendrites responsible for promoting LTD. In its absence, overabundance of these proteins results in reduced synaptic strength due to AMPA receptor trafficking abnormalities that lead, at least in part, to the fragile X phenotype (Bear, 2005; Dolen et al., 2010). Importantly, multiple studies have demonstrated that increased mGlu5 signaling occurs in FXS (Bear, 2005; Ronesi & Huber, 2008), a reduction in mGlu5 expression rescues FXS phenotypes (Dolen et al., 2010), and that FXS mice demonstrate diminished FXS phenotypes, such as anxiety and seizures, when treated with MPEP (Yan et al., 2005). These preclinical studies have validated the use of mGlu5 NAMs as a potential therapeutic strategy for FXS. AFQ056, a NAM of mGlu5, is currently in phase 2 clinical trials in adults with FXS and has demonstrated improvement in several patients with full FMR1 promoter methylation and no detectable FMR1 messenger RNA (Jacquemont et al., 2010).

C. mGlu5 PAMs for Schizophrenia

Positive allosteric modulation of mGlu5 has also emerged as a novel therapeutic target for the treatment of schizophrenia and cognitive disorders. Early clinical findings demonstrated that noncompetitive, use-dependent N-methyl-D-aspartate (NMDA) receptor antagonists, such as phencyclidine (PCP) and ketamine, produce a state of psychosis in humans that is not clinically distinguishable from that observed in schizophrenic patients (Luby et al., 1959). Moreover, administration of NMDA receptor function-enhancing agents, such as agonists at the glycine binding site of the NMDA receptor, results in a symptomatic improvement in schizophrenic patients (Heresco-Levy & Javitt, 2004; Heresco-Levy et al., 1999, 2005; Javitt et al., 1997). Based on these clinical studies, enhancement of glutamatergic neurotransmission via increased NMDA receptor activity has been proposed as a potential treatment for the psychotic symptoms and cognitive deficits associated with many neurological disorders.

Numerous studies suggest that mGlu5 is a closely associated signaling partner with the NMDA receptor and may play an integral role in regulating NMDA receptor function in various forebrain regions implicated in the pathology of schizophrenia (Homayoun et al., 2004; Marino & Conn, 2002). Consistently, activation of mGlu5 receptors results in the potentiation of NMDA receptor currents in hippocampal pyramidal cells, suggesting a role of mGlu5 in cognitive function (Mannaioni et al., 2001). Therefore, selective activation of mGlu5 and subsequent enhancement of NMDA receptor activity may provide a novel approach to the treatment of not only the positive symptoms but also the negative symptoms and cognitive deficits afflicting patients with schizophrenia. Multiple mGlu5 PAMs have been identified and potentiate mGlu5-mediated electrophysiological responses in midbrain and forebrain circuits, including NMDA receptor currents (Ayala et al., 2009; Chen et al., 2007; O’Brien et al., 2004). Further, the positive modulators of mGlu5, CDPPB, ADX47273, and VU0360172 demonstrate robust efficacy in models predictive of antipsychotic activity and cognition enhancement (Kinney et al., 2005; Liu et al., 2008; Rodriguez et al., 2010; Stefani & Moghaddam, 2010). These findings present exciting evidence for the use of mGlu5 potentiators in the treatment of schizophrenia. Extensive discovery efforts to develop a selective mGlu5 PAM for clinical trials are ongoing.

D. mGlu2 PAMs for Schizophrenia, Anxiety Disorders, and Drug Dependence

There is a tremendous volume of clinical and preclinical evidence that Group II mGlu activators have potential as a novel approach for the treatment of schizophrenia and anxiety disorders. Many brain regions relevant to schizophrenia and anxiety express Group II mGlus (Lindsley et al., 2006; Schoepp & Marek, 2002), and Group II agonists reduce neurotransmission in these regions (Chavez-Noriega et al., 2005; Doherty et al., 2004; Macek et al., 1996; Nicholls et al., 2006). Further, the effects of psychotomimetics on thalamocortical glutamatergic neurotransmission, a region postulated to play a role in the pathophysiology of schizophrenia (Chavez-Noriega et al., 2005; Lorrain et al., 2003; Moghaddam, 2004), are blocked by Group II mGlu agonists (Marek et al., 2000). Numerous studies have further demonstrated that Group II agonists have activity in multiple animal models of anxiolytic (Swanson et al., 2005) and antipsychotic (Marek, 2010) drug action, including reversal of PCP-induced hyperlocomotor activity, stereotyped behaviors, and working memory deficits in rats (Cartmell et al., 1999; Moghaddam & Adams, 1998). In addition, Group II agonists inhibit PCP-(Moghaddam & Adams, 1998) or ketamine-induced (Lorrain et al., 2003) glutamate release in prefrontal cortex and nucleus accumbens, two regions of the brain thought to be relevant for antipsychotic drug action. Importantly, LY2140023, the prodrug of the Group II agonist LY404039, recently demonstrated antipsychotic efficacy in a clinical trial, with schizophrenic patients showing improvements in multiple symptom clusters (Patil et al., 2007).

The efficacy of Group II agonists in anxiolytic and antipsychotic models has paved the way for evaluation of mGlu2 selective PAMs as an alternative therapeutic route. Similar to the Group II agonists, numerous mGlu2 PAMs have demonstrated efficacy in reversing PCP- or ketamine-induced hyperlocomotion in rodents (Galici et al., 2005, 2006; Govek et al., 2005; Hackler et al., 2010; Johnson et al., 2005; Pinkerton et al., 2004, 2005). mGlu2 PAMs have also been demonstrated to inhibit ketamine-induced neurotransmitter release in brain regions relevant for the antipsychotic action of Group II agonists, including norepinephrine release in the ventral hippocampus (Pinkerton et al., 2004) and histamine release in the medial prefrontal cortex (mPFC; Fell et al., 2010). Further, two distinct mGlu2 PAMs, BINA and LY487379, have been evaluated in PPI, an animal model of sensorimotor gating that is disrupted in schizophrenic patients. Although Group II agonists have no effect on PPI, LY487379 reversed amphetamine-, but not PCP-, induced disruptions of PPI (Galici et al., 2005), whereas BINA reversed PCP-induced disruptions of PPI (Galici et al., 2006). Together, these studies suggest that mGlu2 PAMs may provide a novel approach toward the sensorimotor gating deficits associated with schizophrenia. In addition to these models of antipsychotic drug action, multiple mGlu2 PAMs have demonstrated efficacy in several models of anxiolytic action, including stress-induced hyperthermia (Fell et al., 2011; Galici et al., 2006), marble burying (Fell et al., 2011), fear-potentiated startle (Johnson et al., 2003, 2005), and in the elevated plus maze (Galici et al., 2006). Together, these studies raise the possibility that selective mGlu2 PAMs might provide a novel approach to the treatment of schizophrenia and anxiety disorders.

In addition to anxiety and schizophrenia, Group II agonists have also been suggested for the treatment of drug addiction and relapse (Moussawi & Kalivas, 2010). In animal models, Group II agonists attenuate cocaine self-administration (Adewale et al., 2006; Baptista et al., 2004; Kim et al., 2005) and cocaine-seeking behavior (Baptista et al., 2004; Peters & Kalivas, 2006). In addition to their effects on cocaine, Group II agonists also inhibit heroin-seeking (Bossert et al., 2005, 2006) and alcohol-seeking (Backstrom & Hyytia, 2005) behaviors. Although these studies are promising, Group II agonists have also been demonstrated to decrease food-seeking behavior (Baptista et al., 2004; Jin et al., 2010; Peters & Kalivas, 2006), implying that Group II agonists decrease responses to both drug and natural rewards. Alternatively, recent studies with the mGlu2 PAM BINA have demonstrated attenuation of cocaine-self administration and cocaine-seeking without an effect on food responding, implying that mGlu2 PAMs may provide a better therapeutic approach to drug addiction than direct acting agonists (Dhanya et al., 2011; Jin et al., 2010).

E. mGlu3 PAMs for Neuroprotection

Numerous mGlus have been suggested as a target for neurodegenerative disorders (Nicoletti et al., 1996). In particular, a number of studies have demonstrated that Group II mGlu agonists are neuroprotective when mixed cultures of cortical neurons and astrocytes are challenged with NMDA (Bruno et al., 1997, 1998) via a glial-neuronal mechanism involving transforming growth factor β (Bruno et al., 1998). Studies with mGlu2 and mGlu3 KO mice have shown that mGlu3 expressed on the astrocytes are necessary for this effect and that, in the absence of mGlu3, the activation of neuronally expressed mGlu2 may further contribute to excitotoxicity (Corti et al., 2007). Recently, LY2389575, an mGlu3 selective NAM, was utilized in combination with mGlu3 KO mice to further demonstrate that activation of glial mGlu3 receptors was protective against amyloid β neurotoxicity (Caraci et al., 2010). Together, these studies suggest that mGlu3 selective PAMs may have efficacy as neuroprotective agents and potentially novel targets for the treatment of Alzheimer’s disease.

F. Group II NAMs for Cognitive Enhancement

Group II agonists (Spinelli et al., 2005) and muscarinic antagonists, such as scopolamine (Dunnett, 1993), induce deficits in the delayed match to position (DMTP) task in rodents, a measure of working memory. Recently, a Group II NAM has been shown to reverse Group II agonist or scopolamine-induced working memory deficits in the DMTP task (Woltering et al., 2010). Additional studies by this group demonstrated a synergistic reversal of scopolamine-induced deficits in DMTP when low doses of a Group II NAM were combined with a threshold dose of the acetylcholinesterase inhibitor donezepil (Woltering et al., 2010). Given the efficacy of donepezil and other acetylcholinesterase inhibitors in the treatment of the cognitive impairments in Alzheimer’s disease (Tsuno, 2009), Group II NAMs may have efficacy as cognitive enhancers.

G. mGlu4 PAMs for Parkinson’s Disease

mGlu4 is expressed on GABAergic fibers within the indirect pathway of motor control in the basal ganglia circuit. Activation of these receptors decreases GABA release at the striatopallidal synapse, which is overactive after the loss of dopamine neurons in Parkinson’s disease (PD) (Macinnes & Duty, 2008; Matsui & Kita, 2003; Valenti et al., 2003). Disinhibition of thalamocortical neurons is believed to alleviate Parkinsonian symptoms and is the basis for targeting mGlu4 for the treatment of PD (Conn et al., 2005). PHCCC, a selective mGlu4 PAM, potentiates mGlu4-mediated inhibition at the striatopallidal synapse (Marino et al., 2003). In addition, PHCCC, as well as VU0155041, demonstrates antiparkinsonian effects in preclinical models, such as reversal of haloperidol-induced catalepsy and reserpine-induced akinesia (Marino et al., 2003; Niswender et al., 2008a). PHCCC also exhibits neuroprotective properties and has been shown to decrease dopamine neuron degeneration in the substantia nigra in a MPTP model of PD, providing evidence of mGlu4 PAMs’ ability to potentially slow progression of PD by reducing excitotoxicity of dopamine neurons resulting from excessive excitatory stimulation (Battaglia et al., 2006; Valenti et al., 2005).

Group III mGlus demonstrate potential for the treatment of several other CNS disorders. mGlu4 KO mice are resistant to characteristics attributed to alcohol addiction (Blednov et al., 2004); injection of the mGlu7 allosteric agonist, AMN082, into the nucleus accumbens and ventral pallidum inhibits cocaine self-administration (Li et al., 2009); and mGlu8 expression is altered by cocaine and amphetamine administration (Parelkar & Wang, 2008; Zhang et al., 2009), suggesting a role for Group III receptors in addiction. Pharmacological and genetic studies implicate mGlu4 and mGlu7 in the regulation of seizures (Ngomba et al., 2008; Sansig et al., 2001; Snead et al., 2000). Additional research proposes roles of Group III mGlus in anxiety and depression (Klak et al., 2007; Palucha et al., 2007; Stachowicz et al., 2004, 2006, 2008), and neuroblastoma (Iacovelli et al., 2006).

VIII. Conclusion

Within the past 15 years, selective allosteric modulators have been developed for the vast majority of the mGlus. This advance, together with the development of mGlu KO mice, has greatly led to our better understanding of the roles of individual mGlu subtypes in a number of systems. Within this time, the mGlu allosteric modulators field has also made significant advances in the understanding of how and where these compounds interact with the receptor. These studies have suggested commonalities among mGlu allosteric sites, and that individual mGlu subtypes can have multiple alloste-ric sites. However, given the inherent complexity of allosteric interactions, there remains a great need to further investigate these mechanisms. In particular, numerous mGlu allosteric modulators have been demonstrated to display functional selectivity. Thus, allosteric modulators have the potential to engender unique receptor conformations that may have different functional consequences than regular competitive ligands. This possibility suggests that evaluation of mGlu allosteric modulators across multiple functional outputs is key to a greater understanding of how a given modulator may function in vivo. It remains to be determined whether pure modulation or both modulator and agonist properties, that is, inverse agonist/NAMs or agonist/PAMs, will be required to treat diseases with mGlu allosteric ligands. In all likelihood, each therapeutic intervention will require a different drug profile. Regardless, numerous preclinical and clinical studies continue to provide validity for utilizing mGlu allosteric ligands as a therapeutic approach, and it is likely that these studies will pave the way for additional utilization of mGlu allosteric modulators for the treatment of numerous neurological and psychiatric disorders in the coming years.

Acknowledgments

The authors would like to acknowledge funding from the National Institute of Mental Health (D. J. S., P. J. C), an American Australian Association Merck Co. Foundation fellowship (K. J. G.), and a National Institute of Mental Health Kirschstein National Research Service Award (J. M. R.).

Abbreviations

5MPEP

5-methyl-6-(phenylethynyl)-pyridine

ADX47273

S-(4-fluorophenyl)-{3-[3-(4-fluorophenyl)-[1,2,4] oxadiazol-5-yl]-piperidin-1-yl}-methanone

AMN082

N,N′-bis(diphenylmethyl)-1,2-ethanediamine

ATCM

allosteric ternary complex model

Bay 36-7620

(3aS,6aS)-hexahydro-5-methylene-6a-(2-naphthalenylmethyl)-1H-cyclopenta[c]furan-1-one

BINA

Biphenyl-indanone A

cAMP

cyclic adenosine monophosphate

CaSR

calcium-sensing receptor

CCR5

C–C chemokine receptor type 5

CDPPB

3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide

CPCCOEt

7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxylate ethyl ester

CPPHA

N-{4-chloro-2-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl) methyl]phenyl}-2-hydroxybenzamide

DFB

[(3-fluorophenyl)methylene]hydrazone-3-fluorobenzaldehyde

DMTP

delayed match to position

ERK1/2

extracellular signal-regulated kinases 1 and 2

FMRP

fragile X mental retardation protein

FTIDC

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

FXS

fragile X syndrome

GERD

gastroesophageal reflux

GPCR

G protein-coupled receptor

JNJ16259685

(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-(cis-4-methoxycyclohexyl)-methanone

KO

knockout

LTD

long-term depression

LTP

long-term potentiation

LY2389575

(3S)-1-(5-bromopyrimidin-2-yl)-N-(2,4-dichlorobenzyl) pyrrolidin-3-amine methanesulfonate hydrate

LY404039

(−)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0] hexane-4,6-dicarboxylic acid

LY487379

2,2,2-trifluoro-N-[4-(2-methoxyphenoxy) phenyl]-N-(3-pyridinylmethyl)ethanesulfonamide

mGlu

metabotropic glutamate receptor

MMPIP

6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo [4,5-c] pyridine-4(5H)-one hydrochloride

MNI-137

4-(7-bromo-4-oxo-4,5-dihydro-3H-benzo[1,4]diazepin-2-yl)-pyridine-2-carbonitrile

MPEP

2-methyl-6-(phenylethynyl)pyridine

mPFC

medial prefrontal cortex

MTEP

3-((2-methyl-1,3-thiazol-4-yl)ethynyl)pyridine

NAM

negative allosteric modulator

NMDA

N-methyl-D-aspartate

Org27569

5-chloro-3-ethyl-N-[2-[4-(1-piperidinyl)phenyl]ethyl]-1H-indole-2-carboxamide

PAM

positive allosteric modulator

PCP

phencyclidine

PD

Parkinson’s disease

PHCCC

N-Phenyl-7-(hydroxyimino)cyclopropa[b] chromen-1a-carboxamide

PPI

prepulse inhibition

Ro 67-4853

(9H-xanthene-9-carbonyl)-carbamic acid butyl ester

Ro 67-7476

(S)-2-(4-fluorophenyl)-1-(toluene-4-sulfonyl)-pyrrolidine

SIB-1757

6-methyl-2-(phenylazo)-3-pyridinol

SIB-1893

2-methyl-6-(2-phenylethenyl)pyridine

TM

transmembrane domain

VFD

venus flytrap domain

VU0155041

cis-2-{[(3,5-dichlorophenyl)amino]carbonyl} cyclohexanecarboxylic acid

VU0285683

3-fluoro-5-(3-(pyridine-2-yl)-1,2,4-oxadiazol-5-yl) benzonitrile

VU0360172

N-cyclobutyl-6-((3-fluorophenyl)ethynyl)nicotinamide

VU29

4-nitro-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide

VU48

4-nitro-N-(1-(2-bromophenyl)-3-phenyl-1H-pyrazol-5-yl) benzamide

VU71

4-nitro-N-(1,4-diphenyl-1H-pyazol-5-yl)benzamide

YM298198

6-amino-N-cyclohexyl-N,3-dimethylthiazolo[3,2-α] benzimidazole-2-carboxamide

Footnotes

Conflicts of Interest: Dr. Conn has served as a consultant over the past 3 years for: Merck and Co., Johnson and Johnson, Hoffman La Roche, GlaxoSmithKline, Lundbeck Research USA, Epix Pharmaceuticals, Invitrogen Life Technologies, Evotech Inc., Addex Pharmaceuticals, Michael J. Fox Foundation, Cephalon Inc., LEK Consulting, The Frankel Group, Prestwick Chemical Co., IMS Health, Primary Insight, Otsuka Pharmaceuticals, AstraZenca USA, NeurOP Inc., Seaside Therapeutics, Millipore Corp., Genentech, Abbott Laboratories, AMRI, Bristol Myers Squibb, and PureTech. Dr. Conn receives research support that includes salary support from Seaside Therapeutics and Johnson & Johnson. The remaining authors have no conflicts of interest to declare.

References

  1. Adewale AS, Platt DM, Spealman RD. Pharmacological stimulation of group ii metabotropic glutamate receptors reduces cocaine self-administration and cocaine-induced reinstatement of drug seeking in squirrel monkeys. The Journal of Pharmacology and Experimental Therapeutics. 2006;318(2):922–931. doi: 10.1124/jpet.106.105387. [DOI] [PubMed] [Google Scholar]
  2. Aiba A, Chen C, Herrup K, Rosenmund C, Stevens CF, Tonegawa S. Reduced hippocampal long-term potentiation and context-specific deficit in associative learning in mGluR1 mutant mice. Cell. 1994a;79(2):365–375. doi: 10.1016/0092-8674(94)90204-6. [DOI] [PubMed] [Google Scholar]
  3. Aiba A, Kano M, Chen C, Stanton ME, Fox GD, Herrup K, et al. Deficient cerebellar long-term depression and impaired motor learning in mGluR1 mutant mice. Cell. 1994b;79(2):377–388. [PubMed] [Google Scholar]
  4. Alexander GM, Godwin DW. Metabotropic glutamate receptors as a strategic target for the treatment of epilepsy. Epilepsy Research. 2006;71(1):1–22. doi: 10.1016/j.eplepsyres.2006.05.012. [DOI] [PubMed] [Google Scholar]
  5. Annoura H, Fukunaga A, Uesugi M, Tatsuoka T, Horikawa Y. A novel class of antagonists for metabotropic glutamate receptors, 7-(Hydroxyimino)cyclopropa[b]chro-men-1a-carboxylates. Bioorganic & Medicinal Chemistry Letters. 1996;6(7):763–766. [Google Scholar]
  6. Anwyl R. Metabotropic glutamate receptors: Electrophysiological properties and role in plasticity. Brain Research. Brain Research Reviews. 1999;29(1):83–120. doi: 10.1016/s0165-0173(98)00050-2. [DOI] [PubMed] [Google Scholar]
  7. Ayala JE, Chen Y, Banko JL, Sheffler DJ, Williams R, Telk AN, et al. mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology. 2009;34(9):2057–2071. doi: 10.1038/npp.2009.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Ayala JE, Niswender CM, Luo Q, Banko JL, Conn PJ. Group III mGluR regulation of synaptic transmission at the SC-CA1 synapse is developmentally regulated. Neuropharmacology. 2008;54(5):804–814. doi: 10.1016/j.neuropharm.2007.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Backstrom P, Hyytia P. 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. European Journal of Pharmacology. 2005;528(1–3):110–118. doi: 10.1016/j.ejphar.2005.10.051. [DOI] [PubMed] [Google Scholar]
  10. Ballesteros J, Palczewski K. G protein-coupled receptor drug discovery: Implications from the crystal structure of rhodopsin. Current Opinion in Drug Discovery & Development. 2001;4(5):561–574. [PMC free article] [PubMed] [Google Scholar]
  11. Baptista MA, Martin-Fardon R, Weiss F. 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. The Journal of Neuroscience. 2004;24(20):4723–4727. doi: 10.1523/JNEUROSCI.0176-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Battaglia G, Busceti CL, Molinaro G, Biagioni F, Traficante A, Nicoletti F, et al. Pharmacological activation of mGlu4 metabotropic glutamate receptors reduces nigrostriatal degeneration in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyr-idine. The Journal of Neuroscience. 2006;26(27):7222–7229. doi: 10.1523/JNEUROSCI.1595-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bear MF. Therapeutic implications of the mGluR theory of fragile X mental retardation. Genes, Brain, and Behavior. 2005;4(6):393–398. doi: 10.1111/j.1601-183X.2005.00135.x. [DOI] [PubMed] [Google Scholar]
  14. Bellone C, Luscher C, Mameli M. Mechanisms of synaptic depression triggered by metabotropic glutamate receptors. Cellular and Molecular Life Sciences. 2008;65(18):2913–2923. doi: 10.1007/s00018-008-8263-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Beqollari D, Kammermeier PJ. The mGlu(4) receptor allosteric modulator N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide acts as a direct agonist at mGlu(6) receptors. European Journal of Pharmacology. 2008;589(1–3):49–52. doi: 10.1016/j.ejphar.2008.06.054. [DOI] [PubMed] [Google Scholar]
  16. Bessis AS, Bertrand HO, Galvez T, De Colle C, Pin JP, Acher F. Three-dimensional model of the extracellular domain of the type 4a metabotropic glutamate receptor: New insights into the activation process. Protein Science. 2000;9(11):2200–2209. doi: 10.1110/ps.9.11.2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bessis AS, Rondard P, Gaven F, Brabet I, Triballeau N, Prezeau L, et al. Closure of the Venus flytrap module of mGlu8 receptor and the activation process: Insights from mutations converting antagonists into agonists. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(17):11097–11102. doi: 10.1073/pnas.162138699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bhave G, Nadin BM, Brasier DJ, Glauner KS, Shah RD, Heinemann SF, et al. Membrane topology of a metabotropic glutamate receptor. The Journal of Biological Chemistry. 2003;278(32):30294–30301. doi: 10.1074/jbc.M303258200. [DOI] [PubMed] [Google Scholar]
  19. Blednov YA, Walker D, Osterndorf-Kahanek E, Harris RA. Mice lacking metabotropic glutamate receptor 4 do not show the motor stimulatory effect of ethanol. Alcohol. 2004;34(2–3):251–259. doi: 10.1016/j.alcohol.2004.10.003. [DOI] [PubMed] [Google Scholar]
  20. Bossert JM, Busch RF, Gray SM. The novel mGluR2/3 agonist LY379268 attenuates cue-induced reinstatement of heroin seeking. Neuroreport. 2005;16(9):1013–1016. doi: 10.1097/00001756-200506210-00026. [DOI] [PubMed] [Google Scholar]
  21. Bossert JM, Gray SM, Lu L, Shaham Y. Activation of group II metabotropic glutamate receptors in the nucleus accumbens shell attenuates context-induced relapse to heroin seeking. Neuropsychopharmacology. 2006;31(10):2197–2209. doi: 10.1038/sj.npp.1300977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bradbury MJ, Campbell U, Giracello D, Chapman D, King C, Tehrani L, et al. Metabotropic glutamate receptor mGlu5 is a mediator of appetite and energy balance in rats and mice. The Journal of Pharmacology and Experimental Therapeutics. 2005;313(1):395–402. doi: 10.1124/jpet.104.076406. [DOI] [PubMed] [Google Scholar]
  23. Bradley SJ, Watson JM, Challiss RA. Effects of positive allosteric modulators on single-cell oscillatory Ca2 plus signaling initiated by the type 5 metabotropic glutamate receptor. Molecular Pharmacology. 2009;76(6):1302–1313. doi: 10.1124/mol.109.059170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Brauner-Osborne H, Jensen AA, Krogsgaard-Larsen P. Interaction of CPCCOEt with a chimeric mGlu1b and calcium sensing receptor. Neuroreport. 1999;10(18):3923–3925. doi: 10.1097/00001756-199912160-00036. [DOI] [PubMed] [Google Scholar]
  25. Brodkin J, Bradbury M, Busse C, Warren N, Bristow LJ, Varney MA. Reduced stress-induced hyperthermia in mGluR5 knockout mice. The European Journal of Neuroscience. 2002;16(11):2241–2244. doi: 10.1046/j.1460-9568.2002.02294.x. [DOI] [PubMed] [Google Scholar]
  26. Brody SA, Conquet F, Geyer MA. Disruption of prepulse inhibition in mice lacking mGluR1. The European Journal of Neuroscience. 2003;18(12):3361–3366. doi: 10.1111/j.1460-9568.2003.03073.x. [DOI] [PubMed] [Google Scholar]
  27. Brody SA, Conquet F, Geyer MA. Effect of antipsychotic treatment on the prepulse inhibition deficit of mGluR5 knockout mice. Psychopharmacology (Berl) 2004a;172(2):187–195. doi: 10.1007/s00213-003-1635-3. [DOI] [PubMed] [Google Scholar]
  28. Brody SA, Dulawa SC, Conquet F, Geyer MA. Assessment of a prepulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Molecular Psychiatry. 2004b;9(1):35–41. doi: 10.1038/sj.mp.4001404. [DOI] [PubMed] [Google Scholar]
  29. Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, Nicoletti F. Neuroprotection by glial metabotropic glutamate receptors is mediated by transforming growth factor-beta. The Journal of Neuroscience. 1998;18(23):9594–9600. doi: 10.1523/JNEUROSCI.18-23-09594.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Bruno V, Sureda FX, Storto M, Casabona G, Caruso A, Knopfel T, et al. The neuroprotective activity of group-II metabotropic glutamate receptors requires new protein synthesis and involves a glial-neuronal signaling. The Journal of Neuroscience. 1997;17(6):1891–1897. doi: 10.1523/JNEUROSCI.17-06-01891.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Bushell TJ, Sansig G, Collett VJ, van der Putten H, Collingridge GL. Altered short-term synaptic plasticity in mice lacking the metabotropic glutamate receptor mGlu7. ScientificWorldJournal. 2002;2:730–737. doi: 10.1100/tsw.2002.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Callaerts-Vegh Z, Beckers T, Ball SM, Baeyens F, Callaerts PF, Cryan JF, et al. Concomitant deficits in working memory and fear extinction are functionally dissociated from reduced anxiety in metabotropic glutamate receptor 7-deficient mice. The Journal of Neuroscience. 2006;26(24):6573–6582. doi: 10.1523/JNEUROSCI.1497-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Caraci F, Molinaro G, Battaglia G, Giuffrida ML, Riozzi B, Traficante A, et al. Targeting group-II metabotropic glutamate receptors for the treatment of psychosis associated with Alzheimer’s disease: Selective activation of mGlu2 receptors amplifies {beta}-amyloid toxicity in cultured neurons whereas dual activation of mGlu2 and mGlu3 receptors is neuroprotective. Molecular Pharmacology. 2010;79(3):618–626. doi: 10.1124/mol.110.067488. [DOI] [PubMed] [Google Scholar]
  34. Carroll FY, Stolle A, Beart PM, Voerste A, Brabet I, Mauler F, et al. BAY36-7620: A potent non-competitive mGlu1 receptor antagonist with inverse agonist activity. Molecular Pharmacology. 2001;59(5):965–973. [PMC free article] [PubMed] [Google Scholar]
  35. Cartmell J, Monn JA, Schoepp DD. The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. The Journal of Pharmacology and Experimental Therapeutics. 1999;291(1):161–170. [PubMed] [Google Scholar]
  36. Ceccarelli SM, Jaeschke G, Buettelmann B, Huwyler J, Kolczewski S, Peters JU, et al. Rational design, synthesis, and structure-activity relationship of benzoxazolones: New potent mglu5 receptor antagonists based on the fenobam structure. Bioorganic & Medicinal Chemistry Letters. 2007;17(5):1302–1306. doi: 10.1016/j.bmcl.2006.12.006. [DOI] [PubMed] [Google Scholar]
  37. Chavez-Noriega LE, Marino MJ, Schaffhauser H, Campbell UC, Conn PJ. Novel potential therapeutics for schizophrenia: Focus on the modulation of metabotropic glutamate receptor function. Current Neuropharmacology. 2005;3(1):9–34. [Google Scholar]
  38. Chen Y, Goudet C, Pin JP, Conn PJ. N-{4-Chloro-2-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phenyl}-2-hydroxybenzamide (CPPHA) acts through a novel site as a positive allosteric modulator of group 1 metabotropic glutamate receptors. Molecular Pharmacology. 2008;73(3):909–918. doi: 10.1124/mol.107.040097. [DOI] [PubMed] [Google Scholar]
  39. Chen Y, Nong Y, Goudet C, Hemstapat K, de Paulis T, Pin JP, et al. Interaction of novel positive allosteric modulators of metabotropic glutamate receptor 5 with the negative allosteric antagonist site is required for potentiation of receptor responses. Molecular Pharmacology. 2007;71(5):1389–1398. doi: 10.1124/mol.106.032425. [DOI] [PubMed] [Google Scholar]
  40. Cherezov V, Rosenbaum DM, Hanson MA, Rasmussen SG, Thian FS, Kobilka TS, et al. High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor. Science. 2007;318(5854):1258–1265. doi: 10.1126/science.1150577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Chiamulera C, Epping-Jordan MP, Zocchi A, Marcon C, Cottiny C, Tacconi S, et al. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nature Neuroscience. 2001;4(9):873–874. doi: 10.1038/nn0901-873. [DOI] [PubMed] [Google Scholar]
  42. Chien EY, Liu W, Zhao Q, Katritch V, Han GW, Hanson MA, et al. Structure of the human dopamine D3 receptor in complex with a D2/D3 selective antagonist. Science. 2010;330(6007):1091–1095. doi: 10.1126/science.1197410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Conn PJ, Battaglia G, Marino MJ, Nicoletti F. Metabotropic glutamate receptors in the basal ganglia motor circuit. Nature Reviews. Neuroscience. 2005;6(10):787–798. doi: 10.1038/nrn1763. [DOI] [PubMed] [Google Scholar]
  44. Conn PJ, Lindsley CW, Jones CK. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends in Pharmacological Sciences. 2009;30(1):25–31. doi: 10.1016/j.tips.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Corti C, Battaglia G, Molinaro G, Riozzi B, Pittaluga A, Corsi M, et al. The use of knock-out mice unravels distinct roles for mGlu2 and mGlu3 metabotropic glutamate receptors in mechanisms of neurodegeneration/neuroprotection. The Journal of Neuroscience. 2007;27(31):8297–8308. doi: 10.1523/JNEUROSCI.1889-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Cosford ND, Roppe J, Tehrani L, Schweiger EJ, Seiders TJ, Chaudary A, et al. [3H]-methoxymethyl-MTEP and [3H]-methoxy-PEPy: Potent and selective radioligands for the metabotropic glutamate subtype 5 (mGlu5) receptor. Bioorganic & Medicinal Chemistry Letters. 2003a;13(3):351–354. doi: 10.1016/s0960-894x(02)00997-6. [DOI] [PubMed] [Google Scholar]
  47. Cosford ND, Tehrani L, Roppe J, Schweiger E, Smith ND, Anderson J, et al. 3-[(2-Methyl-1,3-thiazol-4-yl)ethynyl]-pyridine: A potent and highly selective metabo-tropic glutamate subtype 5 receptor antagonist with anxiolytic activity. Journal of Medicinal Chemistry. 2003b;46(2):204–206. doi: 10.1021/jm025570j. [DOI] [PubMed] [Google Scholar]
  48. Cryan JF, Kelly PH, Neijt HC, Sansig G, Flor PJ, van Der Putten H. Antidepressant and anxiolytic-like effects in mice lacking the group III metabotropic glutamate receptor mGluR7. The European Journal of Neuroscience. 2003;17(11):2409–2417. doi: 10.1046/j.1460-9568.2003.02667.x. [DOI] [PubMed] [Google Scholar]
  49. de Paulis T, Hemstapat K, Chen Y, Zhang Y, Saleh S, Alagille D, et al. Substituent effects of N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamides on positive allosteric modulation of the metabotropic glutamate-5 receptor in rat cortical astrocytes. Journal of Medicinal Chemistry. 2006;49(11):3332–3344. doi: 10.1021/jm051252j. [DOI] [PubMed] [Google Scholar]
  50. Dhanya RP, Sidique S, Sheffler DJ, Nickols HH, Herath A, Yang L, et al. Design and synthesis of an orally active metabotropic glutamate receptor subtype-2 (mGluR2) positive allosteric modulator (PAM) that decreases cocaine self-administration in rats. Journal of Medicinal Chemistry. 2011;54(1):342–353. doi: 10.1021/jm1012165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Dhingra A, Lyubarsky A, Jiang M, Pugh EN, Jr, Birnbaumer L, Sterling P, et al. The light response of ON bipolar neurons requires G[alpha]o. The Journal of Neuroscience. 2000;20(24):9053–9058. doi: 10.1523/JNEUROSCI.20-24-09053.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Doherty JJ, Alagarsamy S, Bough KJ, Conn PJ, Dingledine R, Mott DD. Metabotropic glutamate receptors modulate feedback inhibition in a developmentally regulated manner in rat dentate gyrus. The Journal of Physiology. 2004;561(Pt 2):395–401. doi: 10.1113/jphysiol.2004.074930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Dolen G, Carpenter RL, Ocain TD, Bear MF. Mechanism-based approaches to treating fragile X. Pharmacology & Therapeutics. 2010;127(1):78–93. doi: 10.1016/j.pharmthera.2010.02.008. [DOI] [PubMed] [Google Scholar]
  54. Dorr P, Westby M, Dobbs S, Griffin P, Irvine B, Macartney M, et al. Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrobial Agents and Chemotherapy. 2005;49(11):4721–4732. doi: 10.1128/AAC.49.11.4721-4732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Dunnett SB. The role and repair of forebrain cholinergic systems in short-term memory. Studies using the delayed matching-to-position task in rats. Advances in Neurology. 1993;59:53–65. [PubMed] [Google Scholar]
  56. Duvoisin RM, Zhang C, Pfankuch TF, O’Connor H, Gayet-Primo J, Quraishi S, et al. Increased measures of anxiety and weight gain in mice lacking the group III metabotropic glutamate receptor mGluR8. The European Journal of Neuroscience. 2005;22(2):425–436. doi: 10.1111/j.1460-9568.2005.04210.x. [DOI] [PubMed] [Google Scholar]
  57. Ehlert FJ. Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. Molecular Pharmacology. 1988;33(2):187–194. [PubMed] [Google Scholar]
  58. Fell MJ, Katner JS, Johnson BG, Khilevich A, Schkeryantz JM, Perry KW, et al. Activation of metabotropic glutamate (mGlu)2 receptors suppresses histamine release in limbic brain regions following acute ketamine challenge. Neuropharmacology. 2010;58(3):632–639. doi: 10.1016/j.neuropharm.2009.11.014. [DOI] [PubMed] [Google Scholar]
  59. Fell MJ, Svensson KA, Johnson BG, Schoepp DD. Evidence for the role of metabotropic glutamate (mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist (−)-(1R,4S,5S,6S)-4-amino-2-sulfonylbi-cyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039) The Journal of Pharmacology and Experimental Therapeutics. 2008;326(1):209–217. doi: 10.1124/jpet.108.136861. [DOI] [PubMed] [Google Scholar]
  60. Fell MJ, Witkin JM, Falcone JF, Katner JS, Perry KW, Hart J, et al. 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. The Journal of Pharmacology and Experimental Therapeutics. 2011;336(1):165–177. doi: 10.1124/jpet.110.172957. [DOI] [PubMed] [Google Scholar]
  61. Fendt M, Schmid S, Thakker DR, Jacobson LH, Yamamoto R, Mitsukawa K, et al. mGluR7 facilitates extinction of aversive memories and controls amygdala plasticity. Molecular Psychiatry. 2008;13(10):970–979. doi: 10.1038/sj.mp.4002073. [DOI] [PubMed] [Google Scholar]
  62. Ferraguti F, Shigemoto R. Metabotropic glutamate receptors. Cell and Tissue Research. 2006;326(2):483–504. doi: 10.1007/s00441-006-0266-5. [DOI] [PubMed] [Google Scholar]
  63. Galandrin S, Oligny-Longpre G, Bouvier M. The evasive nature of drug efficacy: Implications for drug discovery. Trends in Pharmacological Sciences. 2007;28(8):423–430. doi: 10.1016/j.tips.2007.06.005. [DOI] [PubMed] [Google Scholar]
  64. Galici R, Echemendia NG, Rodriguez AL, Conn PJ. A selective allosteric potentiator of metabotropic glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in mouse models predictive of antipsychotic activity. The Journal of Pharmacology and Experimental Therapeutics. 2005;315(3):1181–1187. doi: 10.1124/jpet.105.091074. [DOI] [PubMed] [Google Scholar]
  65. Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, et al. Biphenyl-indanone A, a positive allosteric modulator of the metabotropic glutamate receptor subtype 2, has antipsychotic- and anxiolytic-like effects in mice. The Journal of Pharmacology and Experimental Therapeutics. 2006;318(1):173–185. doi: 10.1124/jpet.106.102046. [DOI] [PubMed] [Google Scholar]
  66. Gasparini F, Floersheim P, Flor PJ, Heinrich M, Inderbitzin W, Ott D, et al. Discovery and characterization of non-competitive antagonists of group I metabotropic glutamate receptors. Farmaco. 2001;56(1–2):95–99. doi: 10.1016/s0014-827x(01)01008-4. [DOI] [PubMed] [Google Scholar]
  67. Gasparini F, Lingenhohl K, Stoehr N, Flor PJ, Heinrich M, Vranesic I, et al. 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology. 1999;38(10):1493–1503. doi: 10.1016/s0028-3908(99)00082-9. [DOI] [PubMed] [Google Scholar]
  68. Gerlai R, Roder JC, Hampson DR. Altered spatial learning and memory in mice lacking the mGluR4 subtype of metabotropic glutamate receptor. Behavioral Neuroscience. 1998;112(3):525–532. doi: 10.1037//0735-7044.112.3.525. [DOI] [PubMed] [Google Scholar]
  69. Gil-Sanz C, Delgado-Garcia JM, Fairen A, Gruart A. Involvement of the mGluR1 receptor in hippocampal synaptic plasticity and associative learning in behaving mice. Cerebral Cortex. 2008;18(7):1653–1663. doi: 10.1093/cercor/bhm193. [DOI] [PubMed] [Google Scholar]
  70. Goddyn H, Callaerts-Vegh Z, Stroobants S, Dirikx T, Vansteenwegen D, Hermans D, et al. Deficits in acquisition and extinction of conditioned responses in mGluR7 knockout mice. Neurobiology of Learning and Memory. 2008;90(1):103–111. doi: 10.1016/j.nlm.2008.01.001. [DOI] [PubMed] [Google Scholar]
  71. Goudet C, Gaven F, Kniazeff J, Vol C, Liu J, Cohen-Gonsaud M, et al. Heptahelical domain of metabotropic glutamate receptor 5 behaves like rhodopsin-like receptors. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(1):378–383. doi: 10.1073/pnas.0304699101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Goudet C, Magnaghi V, Landry M, Nagy F, Gereau RWt, Pin JP. Metabotropic receptors for glutamate and GABA in pain. Brain Research Reviews. 2009;1(1):43–56. doi: 10.1016/j.brainresrev.2008.12.007. [DOI] [PubMed] [Google Scholar]
  73. Govek SP, Bonnefous C, Hutchinson JH, Kamenecka T, McQuiston J, Pracitto R, et al. Benzazoles as allosteric potentiators of metabotropic glutamate receptor 2 (mGluR2): Efficacy in an animal model for schizophrenia. Bioorganic & Medicinal Chemistry Letters. 2005;15(18):4068–4072. doi: 10.1016/j.bmcl.2005.06.017. [DOI] [PubMed] [Google Scholar]
  74. Gray L, van den Buuse M, Scarr E, Dean B, Hannan AJ. Clozapine reverses schizophrenia-related behaviours in the metabotropic glutamate receptor 5 knockout mouse: Association with N-methyl-D-aspartic acid receptor up-regulation. The International Journal of Neuropsychopharmacology. 2009;12(1):45–60. doi: 10.1017/S1461145708009085. [DOI] [PubMed] [Google Scholar]
  75. Gregory KJ, Dong EN, Meiler J, Conn PJ. Allosteric modulation of metabotropic glutamate receptors: Structural insights and therapeutic potential. Neuro-pharmacology. 2010;60(1):66–81. doi: 10.1016/j.neuropharm.2010.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hackler EA, Byun NE, Jones CK, Williams JM, Baheza R, Sengupta S, et al. Selective potentiation of the metabotropic glutamate receptor subtype 2 blocks phencycli-dine-induced hyperlocomotion and brain activation. Neuroscience. 2010;168(1):209–218. doi: 10.1016/j.neuroscience.2010.02.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hall DA. Modeling the functional effects of allosteric modulators at pharmacological receptors: An extension of the two-state model of receptor activation. Molecular Pharmacology. 2000;58(6):1412–1423. doi: 10.1124/mol.58.6.1412. [DOI] [PubMed] [Google Scholar]
  78. Harrison PJ, Lyon L, Sartorius LJ, Burnet PW, Lane TA. The group II metabotropic glutamate receptor 3 (mGluR3, mGlu3, GRM3): Expression, function and involvement in schizophrenia. Journal of Psychopharmacology. 2008;22(3):308–322. doi: 10.1177/0269881108089818. [DOI] [PubMed] [Google Scholar]
  79. Hemstapat K, Da Costa H, Nong Y, Brady AE, Luo Q, Niswender CM, et al. A novel family of potent negative allosteric modulators of group II metabotropic glutamate receptors. The Journal of Pharmacology and Experimental Therapeutics. 2007;322(1):254–264. doi: 10.1124/jpet.106.117093. [DOI] [PubMed] [Google Scholar]
  80. Hemstapat K, de Paulis T, Chen Y, Brady AE, Grover VK, Alagille D, et al. A novel class of positive allosteric modulators of metabotropic glutamate receptor subtype 1 interact with a site distinct from that of negative allosteric modulators. Molecular Pharmacology. 2006;70(2):616–626. doi: 10.1124/mol.105.021857. [DOI] [PubMed] [Google Scholar]
  81. Heresco-Levy U, Javitt DC. Comparative effects of glycine and D-cycloserine on persistent negative symptoms in schizophrenia: A retrospective analysis. Schizophrenia Research. 2004;66(2–3):89–96. doi: 10.1016/S0920-9964(03)00129-4. [DOI] [PubMed] [Google Scholar]
  82. Heresco-Levy U, Javitt DC, Ebstein R, Vass A, Lichtenberg P, Bar G, et al. D-serine efficacy as add-on pharmacotherapy to risperidone and olanzapine for treatment-refractory schizophrenia. Biological Psychiatry. 2005;57(6):577–585. doi: 10.1016/j.biopsych.2004.12.037. [DOI] [PubMed] [Google Scholar]
  83. Heresco-Levy U, Javitt DC, Ermilov M, Mordel C, Silipo G, Lichtenstein M. Efficacy of high-dose glycine in the treatment of enduring negative symptoms of schizophrenia. Archives of General Psychiatry. 1999;56(1):29–36. doi: 10.1001/archpsyc.56.1.29. [DOI] [PubMed] [Google Scholar]
  84. Holscher C, Schmid S, Pilz PK, Sansig G, vander Putten H, Plappert CF. Lack of the metabotropic glutamate receptor subtype 7 selectively impairs short-term working memory but not long-term memory. Behavioural Brain Research. 2004;154(2):473–481. doi: 10.1016/j.bbr.2004.03.015. [DOI] [PubMed] [Google Scholar]
  85. Holscher C, Schmid S, Pilz PK, Sansig G, van der Putten H, Plappert CF. Lack of the metabotropic glutamate receptor subtype 7 selectively modulates Theta rhythm and working memory. Learning & Memory. 2005;12(5):450–455. doi: 10.1101/lm.98305. [DOI] [PubMed] [Google Scholar]
  86. Homayoun H, Moghaddam B. Group 5 metabotropic glutamate receptors: Role in modulating cortical activity and relevance to cognition. European Journal of Pharmacology. 2010;639(1–3):33–39. doi: 10.1016/j.ejphar.2009.12.042. [DOI] [PubMed] [Google Scholar]
  87. Homayoun H, Stefani MR, Adams BW, Tamagan GD, Moghaddam B. Functional interaction between NMDA and mGlu5 receptors: Effects on working memory, instrumental learning, motor behaviors, and dopamine release. Neuropsycho-pharmacology. 2004;29(7):1259–1269. doi: 10.1038/sj.npp.1300417. [DOI] [PubMed] [Google Scholar]
  88. Iacovelli L, Arcella A, Battaglia G, Pazzaglia S, Aronica E, Spinsanti P, et al. Pharmacological activation of mGlu4 metabotropic glutamate receptors inhibits the growth of medulloblastomas. The Journal of Neuroscience. 2006;26(32):8388–8397. doi: 10.1523/JNEUROSCI.2285-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Jaakola VP, Griffith MT, Hanson MA, Cherezov V, Chien EY, Lane JR, et al. The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science. 2008;322(5905):1211–1217. doi: 10.1126/science.1164772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Jacquemont S, Curie A, des Portes V, Torrioli MG, Berry-Kravis E, Hagerman RJ, et al. Epigenetic modification of the FMR1 gene in fragile X syndrome is associated with differential response to the mGluR5 antagonist AFQ056. Science Translational Medicine. 2010;3(64):64ra61. doi: 10.1126/scitranslmed.3001708. [DOI] [PubMed] [Google Scholar]
  91. Javitt DC, Sershen H, Hashim A, Lajtha A. Reversal of phencyclidine-induced hyperactivity by glycine and the glycine uptake inhibitor glycyldodecylamide. Neuropsy-chopharmacology. 1997;17(3):202–204. doi: 10.1016/S0893-133X(97)00047-X. [DOI] [PubMed] [Google Scholar]
  92. Jin X, Semenova S, Yang L, Ardecky R, Sheffler DJ, Dahl R, et al. 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. 2010;35(10):2021–2036. doi: 10.1038/npp.2010.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Johnson MP, Baez M, Jagdmann GE, Jr, Britton TC, Large TH, Callagaro DO, et al. Discovery of allosteric potentiators for the metabotropic glutamate 2 receptor: Synthesis and subtype selectivity of N-(4-(2-methoxyphenoxy)phenyl)-N-(2,2,2- trifluoroethylsulfonyl)pyrid-3-ylmethylamine. Journal of Medicinal Chemistry. 2003;46(15):3189–3192. doi: 10.1021/jm034015u. [DOI] [PubMed] [Google Scholar]
  94. Johnson MP, Barda D, Britton TC, Emkey R, Hornback WJ, Jagdmann GE, et al. Metabotropic glutamate 2 receptor potentiators: Receptor modulation, frequency-dependent synaptic activity, and efficacy in preclinical anxiety and psychosis model(s) Psychopharmacology (Berl) 2005;179(1):271–283. doi: 10.1007/s00213-004-2099-9. [DOI] [PubMed] [Google Scholar]
  95. Julio-Pieper M, Flor PJ, Dinan TG, Cryan JF. Exciting times beyond the brain: Metabotropic glutamate receptors in peripheral and non-neural tissues. Pharmacological Reviews. 2011;63(1):35–38. doi: 10.1124/pr.110.004036. [DOI] [PubMed] [Google Scholar]
  96. Kano M, Hashimoto K, Kurihara H, Watanabe M, Inoue Y, Aiba A, et al. Persistent multiple climbing fiber innervation of cerebellar Purkinje cells in mice lacking mGluR1. Neuron. 1997;18(1):71–79. doi: 10.1016/s0896-6273(01)80047-7. [DOI] [PubMed] [Google Scholar]
  97. Kenakin T. New concepts in drug discovery: Collateral efficacy and permissive antagonism. Nature Reviews. Drug Discovery. 2005;4(11):919–927. doi: 10.1038/nrd1875. [DOI] [PubMed] [Google Scholar]
  98. Kenakin T. Functional selectivity through protean and biased agonism: Who steers the ship? Molecular Pharmacology. 2007;72(6):1393–1401. doi: 10.1124/mol.107.040352. [DOI] [PubMed] [Google Scholar]
  99. Kew JN, Pflimlin MC, Kemp JA, Mutel V. Differential regulation of synaptic transmission by mGlu2 and mGlu3 at the perforant path inputs to the dentate gyrus and CA1 revealed in mGlu2 −/− mice. Neuropharmacology. 2002;43(2):215–221. doi: 10.1016/s0028-3908(02)00084-9. [DOI] [PubMed] [Google Scholar]
  100. Keywood C, Wakefield M, Tack J. A proof-of-concept study evaluating the effect of ADX10059, a metabotropic glutamate receptor-5 negative allosteric modulator, on acid exposure and symptoms in gastro-oesophageal reflux disease. Gut. 2009;58(9):1192–1199. doi: 10.1136/gut.2008.162040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Kim JH, Austin JD, Tanabe L, Creekmore E, Vezina P. Activation of group II mGlu receptors blocks the enhanced drug taking induced by previous exposure to amphetamine. The European Journal of Neuroscience. 2005;21(1):295–300. doi: 10.1111/j.1460-9568.2004.03822.x. [DOI] [PubMed] [Google Scholar]
  102. Kinney GG, O’Brien JA, Lemaire W, Burno M, Bickel DJ, Clements MK, et al. A novel selective positive allosteric modulator of metabotropic glutamate receptor subtype 5 has in vivo activity and antipsychotic-like effects in rat behavioral models. The Journal of Pharmacology and Experimental Therapeutics. 2005;313(1):199–206. doi: 10.1124/jpet.104.079244. [DOI] [PubMed] [Google Scholar]
  103. Kinoshita A, Shigemoto R, Ohishi H, van der Putten H, Mizuno N. Immunohistochemical localization of metabotropic glutamate receptors, mGluR7a and mGluR7b, in the central nervous system of the adult rat and mouse: A light and electron microscopic study. The Journal of Comparative Neurology. 1998;393(3):332–352. [PubMed] [Google Scholar]
  104. Klak K, Palucha A, Branski P, Sowa M, Pilc A. 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. 2007;32(2):169–172. doi: 10.1007/s00726-006-0316-z. [DOI] [PubMed] [Google Scholar]
  105. Kniazeff J, Prezeau L, Rondard P, Pin JP, Goudet C. Dimers and beyond: The functional puzzles of class C GPCRs. Pharmacology & Therapeutics. 2011;130(1):9–25. doi: 10.1016/j.pharmthera.2011.01.006. [DOI] [PubMed] [Google Scholar]
  106. Knoflach F, Mutel V, Jolidon S, Kew JN, Malherbe P, Vieira E, et al. Positive allosteric modulators of metabotropic glutamate 1 receptor: Characterization, mechanism of action, and binding site. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(23):13402–13407. doi: 10.1073/pnas.231358298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Kohara A, Toya T, Tamura S, Watabiki T, Nagakura Y, Shitaka Y, et al. Radioligand binding properties and pharmacological characterization of 6-amino-N-cyclohexyl-N,3-dimethylthiazolo[3,2-a]benzimidazole-2-carboxamid e (YM-298198), a high-affinity, selective, and noncompetitive antagonist of metabotropic glutamate receptor type 1. The Journal of Pharmacology and Experimental Therapeutics. 2005;315(1):163–169. doi: 10.1124/jpet.105.087171. [DOI] [PubMed] [Google Scholar]
  108. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nature Reviews. Drug Discovery. 2004;3(8):711–715. doi: 10.1038/nrd1470. [DOI] [PubMed] [Google Scholar]
  109. Kolber BJ, Montana MC, Carrasquillo Y, Xu J, Heinemann SF, Muglia LJ, et al. Activation of metabotropic glutamate receptor 5 in the amygdala modulates pain-like behavior. The Journal of Neuroscience. 2010;30(24):8203–8213. doi: 10.1523/JNEUROSCI.1216-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, et al. Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature. 2000;407(6807):971–977. doi: 10.1038/35039564. [DOI] [PubMed] [Google Scholar]
  111. Langmead CJ, Christopoulos A. Allosteric agonists of 7TM receptors: Expanding the pharmacological toolbox. Trends in Pharmacological Sciences. 2006;27(9):475–481. doi: 10.1016/j.tips.2006.07.009. [DOI] [PubMed] [Google Scholar]
  112. Lavreysen H, Dautzenberg FM. Therapeutic potential of group III metabotropic glutamate receptors. Current Medicinal Chemistry. 2008;15(7):671–684. doi: 10.2174/092986708783885246. [DOI] [PubMed] [Google Scholar]
  113. Lavreysen H, Janssen C, Bischoff F, Langlois X, Leysen JE, Lesage AS. [3H] R214127: A novel high-affinity radioligand for the mGlu1 receptor reveals a common binding site shared by multiple allosteric antagonists. Molecular Pharmacology. 2003;63(5):1082–1093. doi: 10.1124/mol.63.5.1082. [DOI] [PubMed] [Google Scholar]
  114. Leach K, Sexton PM, Christopoulos A. Allosteric GPCR modulators: Taking advantage of permissive receptor pharmacology. Trends in Pharmacological Sciences. 2007;28(8):382–389. doi: 10.1016/j.tips.2007.06.004. [DOI] [PubMed] [Google Scholar]
  115. Lehmann A. Novel treatments of GERD: Focus on the lower esophageal sphincter. European Review for Medical and Pharmacological Sciences. 2008;12(Suppl 1):103–110. [PubMed] [Google Scholar]
  116. Li X, Li J, Peng XQ, Spiller K, Gardner EL, Xi ZX. Metabotropic glutamate receptor 7 modulates the rewarding effects of cocaine in rats: Involvement of a ventral pallidal GABAergic mechanism. Neuropsychopharmacology. 2009;34(7):1783–1796. doi: 10.1038/npp.2008.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Li X, Need AB, Baez M, Witkin JM. Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. The Journal of Pharmacology and Experimental Therapeutics. 2006;319(1):254–259. doi: 10.1124/jpet.106.103143. [DOI] [PubMed] [Google Scholar]
  118. Lindberg JS, Culleton B, Wong G, Borah MF, Clark RV, Shapiro WB, et al. Cinacalcet HCl, an oral calcimimetic agent for the treatment of secondary hyperparathy-roidism in hemodialysis and peritoneal dialysis: A randomized, double-blind, multicenter study. Journal of the American Society of Nephrology. 2005;16(3):800–807. doi: 10.1681/ASN.2004060512. [DOI] [PubMed] [Google Scholar]
  119. Linden AM, Baez M, Bergeron M, Schoepp DD. Effects of mGlu2 or mGlu3 receptor deletions on mGlu2/3 receptor agonist (LY354740)-induced brain c-Fos expression: Specific roles for mGlu2 in the amygdala and subcortical nuclei, and mGlu3 in the hippocampus. Neuropharmacology. 2006;51(2):213–228. doi: 10.1016/j.neuropharm.2006.03.014. [DOI] [PubMed] [Google Scholar]
  120. Linden AM, Johnson BG, Peters SC, Shannon HE, Tian M, Wang Y, et al. Increased anxiety-related behavior in mice deficient for metabotropic glutamate 8 (mGlu8) receptor. Neuropharmacology. 2002;43(2):251–259. doi: 10.1016/s0028-3908(02)00079-5. [DOI] [PubMed] [Google Scholar]
  121. Linden AM, Shannon H, Baez M, Yu JL, Koester A, Schoepp DD. 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) 2005;179(1):284–291. doi: 10.1007/s00213-004-2098-x. [DOI] [PubMed] [Google Scholar]
  122. Lindsley CW, Shipe WD, Wolkenberg SE, Theberge CR, Williams DL, Jr, Sur C, et al. Progress towards validating the NMDA receptor hypofunction hypothesis of schizophrenia. Current Topics in Medicinal Chemistry. 2006;6(8):771–785. doi: 10.2174/156802606777057599. [DOI] [PubMed] [Google Scholar]
  123. Litschig S, Gasparini F, Rueegg D, Stoehr N, Flor PJ, Vranesic I, et al. CPCCOEt, a noncompetitive metabotropic glutamate receptor 1 antagonist, inhibits receptor signaling without affecting glutamate binding. Molecular Pharmacology. 1999;55(3):453–461. [PubMed] [Google Scholar]
  124. Liu F, Grauer S, Kelley C, Navarra R, Graf R, Zhang G, et al. ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piper idin-1-yl}-metha-none]: A novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. The Journal of Pharmacology and Experimental Therapeutics. 2008;327(3):827–839. doi: 10.1124/jpet.108.136580. [DOI] [PubMed] [Google Scholar]
  125. Liu X, He Q, Studholme DJ, Wu Q, Liang S, Yu L. NCD3G: A novel nine-cysteine domain in family 3 GPCRs. Trends in Biochemical Sciences. 2004;29(9):458–461. doi: 10.1016/j.tibs.2004.07.009. [DOI] [PubMed] [Google Scholar]
  126. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. Effects of ketamine and N-methyl-D-aspartate on glutamate and dopamine release in the rat prefrontal cortex: Modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience. 2003;117(3):697–706. doi: 10.1016/s0306-4522(02)00652-8. [DOI] [PubMed] [Google Scholar]
  127. Lu YM, Jia Z, Janus C, Henderson JT, Gerlai R, Wojtowicz JM, et al. Mice lacking metabotropic glutamate receptor 5 show impaired learning and reduced CA1 long-term potentiation (LTP) but normal CA3 LTP. The Journal of Neuroscience. 1997;17(13):5196–5205. doi: 10.1523/JNEUROSCI.17-13-05196.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Luby ED, Cohen BD, Rosenbaum G, Gottlieb JS, Kelley R. Study of a new schizophrenomimetic drug; sernyl. Archives of Neurology and Psychiatry. 1959;81(3):363–369. doi: 10.1001/archneurpsyc.1959.02340150095011. [DOI] [PubMed] [Google Scholar]
  129. Macek TA, Winder DG, Gereau RWt, Ladd CO, Conn PJ. Differential involvement of group II and group III mGluRs as autoreceptors at lateral and medial perforant path synapses. Journal of Neurophysiology. 1996;76(6):3798–3806. doi: 10.1152/jn.1996.76.6.3798. [DOI] [PubMed] [Google Scholar]
  130. Macinnes N, Duty S. Group III metabotropic glutamate receptors act as hetero-receptors modulating evoked GABA release in the globus pallidus in vivo. European Journal of Pharmacology. 2008;580(1–2):95–99. doi: 10.1016/j.ejphar.2007.10.030. [DOI] [PubMed] [Google Scholar]
  131. Maj M, Bruno V, Dragic Z, Yamamoto R, Battaglia G, Inderbitzin W, et al. (−)-PHCCC, a positive allosteric modulator of mGluR4: Characterization, mechanism of action, and neuroprotection. Neuropharmacology. 2003;45(7):895–906. doi: 10.1016/s0028-3908(03)00271-5. [DOI] [PubMed] [Google Scholar]
  132. Malherbe P, Kratochwil N, Knoflach F, Zenner MT, Kew JN, Kratzeisen C, et al. Mutational analysis and molecular modeling of the allosteric binding site of a novel, selective, noncompetitive antagonist of the metabotropic glutamate 1 receptor. The Journal of Biological Chemistry. 2003a;278(10):8340–8347. doi: 10.1074/jbc.M211759200. [DOI] [PubMed] [Google Scholar]
  133. Malherbe P, Kratochwil N, Muhlemann A, Zenner MT, Fischer C, Stahl M, et al. Comparison of the binding pockets of two chemically unrelated allosteric antagonists of the mGlu5 receptor and identification of crucial residues involved in the inverse agonism of MPEP. Journal of Neurochemistry. 2006;98(2):601–615. doi: 10.1111/j.1471-4159.2006.03886.x. [DOI] [PubMed] [Google Scholar]
  134. Malherbe P, Kratochwil N, Zenner MT, Piussi J, Diener C, Kratzeisen C, et al. Mutational analysis and molecular modeling of the binding pocket of the metabotropic glutamate 5 receptor negative modulator 2-methyl-6-(phenylethynyl)-pyridine. Molecular Pharmacology. 2003b;64(4):823–832. doi: 10.1124/mol.64.4.823. [DOI] [PubMed] [Google Scholar]
  135. Mannaioni G, Marino MJ, Valenti O, Traynelis SF, Conn PJ. Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. The Journal of Neuroscience. 2001;21(16):5925–5934. doi: 10.1523/JNEUROSCI.21-16-05925.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Marek GJ. Metabotropic glutamate2/3 (mGlu2/3) receptors, schizophrenia and cognition. European Journal of Pharmacology. 2010;639(1–3):81–90. doi: 10.1016/j.ejphar.2010.02.058. [DOI] [PubMed] [Google Scholar]
  137. Marek GJ, Wright RA, Schoepp DD, Monn JA, Aghajanian GK. Physiological antagonism between 5-hydroxytryptamine(2A) and group II metabotropic glutamate receptors in prefrontal cortex. The Journal of Pharmacology and Experimental Therapeutics. 2000;292(1):76–87. [PubMed] [Google Scholar]
  138. Marino MJ, Conn PJ. Direct and indirect modulation of the N-methyl D-aspartate receptor. Current Drug Targets. CNS and Neurological Disorders. 2002;1(1):1–16. doi: 10.2174/1568007023339544. [DOI] [PubMed] [Google Scholar]
  139. Marino MJ, Williams DL, Jr, O’Brien JA, Valenti O, McDonald TP, Clements MK, et al. Allosteric modulation of group III metabotropic glutamate receptor 4: A potential approach to Parkinson’s disease treatment. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(23):13668–13673. doi: 10.1073/pnas.1835724100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Masu M, Iwakabe H, Tagawa Y, Miyoshi T, Yamashita M, Fukuda Y, et al. Specific deficit of the ON response in visual transmission by targeted disruption of the mGluR6 gene. Cell. 1995;80(5):757–765. doi: 10.1016/0092-8674(95)90354-2. [DOI] [PubMed] [Google Scholar]
  141. Masugi M, Yokoi M, Shigemoto R, Muguruma K, Watanabe Y, Sansig G, et al. Metabotropic glutamate receptor subtype 7 ablation causes deficit in fear response and conditioned taste aversion. The Journal of Neuroscience. 1999;19(3):955–963. doi: 10.1523/JNEUROSCI.19-03-00955.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Mathiesen JM, Svendsen N, Brauner-Osborne H, Thomsen C, Ramirez MT. Positive allosteric modulation of the human metabotropic glutamate receptor 4 (hmGluR4) by SIB-1893 and MPEP. British Journal of Pharmacology. 2003;138(6):1026–1030. doi: 10.1038/sj.bjp.0705159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Matsui T, Kita H. Activation of group III metabotropic glutamate receptors presynaptically reduces both GABAergic and glutamatergic transmission in the rat globus pallidus. Neuroscience. 2003;122(3):727–737. doi: 10.1016/j.neuroscience.2003.08.032. [DOI] [PubMed] [Google Scholar]
  144. May LT, Leach K, Sexton PM, Christopoulos A. Allosteric modulation of G protein-coupled receptors. Annual Review of Pharmacology and Toxicology. 2007;47:1–51. doi: 10.1146/annurev.pharmtox.47.120505.105159. [DOI] [PubMed] [Google Scholar]
  145. Miedlich SU, Gama L, Seuwen K, Wolf RM, Breitwieser GE. Homology modeling of the transmembrane domain of the human calcium sensing receptor and localization of an allosteric binding site. The Journal of Biological Chemistry. 2004;279(8):7254–7263. doi: 10.1074/jbc.M307191200. [DOI] [PubMed] [Google Scholar]
  146. Mitsukawa K, Yamamoto R, Ofner S, Nozulak J, Pescott O, Lukic S, et al. A selective metabotropic glutamate receptor 7 agonist: Activation of receptor signaling via an allosteric site modulates stress parameters in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(51):18712–18717. doi: 10.1073/pnas.0508063102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Moghaddam B. Targeting metabotropic glutamate receptors for treatment of the cognitive symptoms of schizophrenia. Psychopharmacology (Berl) 2004;174(1):39–44. doi: 10.1007/s00213-004-1792-z. [DOI] [PubMed] [Google Scholar]
  148. Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science. 1998;281(5381):1349–1352. doi: 10.1126/science.281.5381.1349. [DOI] [PubMed] [Google Scholar]
  149. Mohler H, Fritschy JM, Rudolph U. A new benzodiazepine pharmacology. The Journal of Pharmacology and Experimental Therapeutics. 2002;300(1):2–8. doi: 10.1124/jpet.300.1.2. [DOI] [PubMed] [Google Scholar]
  150. Morishima Y, Miyakawa T, Furuyashiki T, Tanaka Y, Mizuma H, Nakanishi S. Enhanced cocaine responsiveness and impaired motor coordination in metabo-tropic glutamate receptor subtype 2 knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(11):4170–4175. doi: 10.1073/pnas.0500914102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Moussawi K, Kalivas PW. Group II metabotropic glutamate receptors (mGlu2/3) in drug addiction. European Journal of Pharmacology. 2010;639(1–3):115–122. doi: 10.1016/j.ejphar.2010.01.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Mudo G, Trovato-Salinaro A, Caniglia G, Cheng Q, Condorelli DF. Cellular localization of mGluR3 and mGluR5 mRNAs in normal and injured rat brain. Brain Research. 2007;1149:1–13. doi: 10.1016/j.brainres.2007.02.041. [DOI] [PubMed] [Google Scholar]
  153. Muly EC, Mania I, Guo JD, Rainnie DG. Group II metabotropic glutamate receptors in anxiety circuitry: Correspondence of physiological response and subcellular distribution. The Journal of Comparative Neurology. 2007;505(6):682–700. doi: 10.1002/cne.21525. [DOI] [PubMed] [Google Scholar]
  154. Muto T, Tsuchiya D, Morikawa K, Jingami H. Structures of the extracellular regions of the group II/III metabotropic glutamate receptors. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(10):3759–3764. doi: 10.1073/pnas.0611577104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Nakajima Y, Iwakabe H, Akazawa C, Nawa H, Shigemoto R, Mizuno N, et al. Molecular characterization of a novel retinal metabotropic glutamate receptor mGluR6 with a high agonist selectivity for L-2-amino-4-phosphonobutyrate. The Journal of Biological Chemistry. 1993;268(16):11868–11873. [PubMed] [Google Scholar]
  156. Neugebauer V, Chen PS, Willis WD. Role of metabotropic glutamate receptor subtype mGluR1 in brief nociception and central sensitization of primate STT cells. Journal of Neurophysiology. 1999;82(1):272–282. doi: 10.1152/jn.1999.82.1.272. [DOI] [PubMed] [Google Scholar]
  157. Ngomba RT, Ferraguti F, Badura A, Citraro R, Santolini I, Battaglia G, et al. Positive allosteric modulation of metabotropic glutamate 4 (mGlu4) receptors enhances spontaneous and evoked absence seizures. Neuropharmacology. 2008;54(2):344–354. doi: 10.1016/j.neuropharm.2007.10.004. [DOI] [PubMed] [Google Scholar]
  158. Nicholls RE, Zhang XL, Bailey CP, Conklin BR, Kandel ER, Stanton PK. mGluR2 acts through inhibitory Galpha subunits to regulate transmission and long-term plasticity at hippocampal mossy fiber-CA3 synapses. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(16):6380–6385. doi: 10.1073/pnas.0601267103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Nicoletti F, Bruno V, Copani A, Casabona G, Knopfel T. Metabotropic glutamate receptors: A new target for the therapy of neurodegenerative disorders? Trends in Neurosciences. 1996;19(7):267–271. doi: 10.1016/S0166-2236(96)20019-0. [DOI] [PubMed] [Google Scholar]
  160. Niswender CM. Permissive antagonism induced by negative allosteric modulators of mGluR7. Neuropharmacology. 2008;55:615. (Abstract) [Google Scholar]
  161. Niswender CM, Conn PJ. Metabotropic glutamate receptors: Physiology, pharmacology, and disease. Annual Review of Pharmacology and Toxicology. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Niswender CM, Johnson KA, Miller NR, Ayala JE, Luo Q, Williams R, et al. Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Molecular Pharmacology. 2010;77(3):459–468. doi: 10.1124/mol.109.058768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Niswender CM, Johnson KA, Weaver CD, Jones CK, Xiang Z, Luo Q, et al. Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Molecular Pharmacology. 2008a;74(5):1345–1358. doi: 10.1124/mol.108.049551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Niswender CM, Lebois EP, Luo Q, Kim K, Muchalski H, Yin H, et al. Positive allosteric modulators of the metabotropic glutamate receptor subtype 4 (mGluR4): Part I. Discovery of pyrazolo[3,4-d]pyrimidines as novel mGluR4 positive allosteric modulators. Bioorganic & Medicinal Chemistry Letters. 2008b;18(20):5626–5630. doi: 10.1016/j.bmcl.2008.08.087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. O’Brien JA, Lemaire W, Chen TB, Chang RS, Jacobson MA, Ha SN, et al. A family of highly selective allosteric modulators of the metabotropic glutamate receptor subtype 5. Molecular Pharmacology. 2003;64(3):731–740. doi: 10.1124/mol.64.3.731. [DOI] [PubMed] [Google Scholar]
  166. O’Brien JA, Lemaire W, Wittmann M, Jacobson MA, Ha SN, Wisnoski DD, et al. A novel selective allosteric modulator potentiates the activity of native metabotropic glutamate receptor subtype 5 in rat forebrain. The Journal of Pharmacology and Experimental Therapeutics. 2004;309(2):568–577. doi: 10.1124/jpet.103.061747. [DOI] [PubMed] [Google Scholar]
  167. Ohishi H, Shigemoto R, Nakanishi S, Mizuno N. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: An in situ hybridization study. The Journal of Comparative Neurology. 1993;335(2):252–266. doi: 10.1002/cne.903350209. [DOI] [PubMed] [Google Scholar]
  168. Ott D, Floersheim P, Inderbitzin W, Stoehr N, Francotte E, Lecis G, et al. Chiral resolution, pharmacological characterization, and receptor docking of the noncompetitive mGlu1 receptor antagonist (+/−)-2-hydroxyimino- 1a, 2-dihydro-1H-7-oxacyclopropa[b] naphthalene-7a-carboxylic acid ethyl ester. Journal of Medicinal Chemistry. 2000;43(23):4428–4436. doi: 10.1021/jm0009944. [DOI] [PubMed] [Google Scholar]
  169. Pagano A, Ruegg D, Litschig S, Stoehr N, Stierlin C, Heinrich M, et al. The non-competitive antagonists 2-methyl-6-(phenylethynyl)pyridine and 7-hydroxyimi-nocyclopropan[b]chromen-1a-carboxylic acid ethyl ester interact with overlapping binding pockets in the transmembrane region of group I metabotropic glutamate receptors. The Journal of Biological Chemistry. 2000;275(43):33750–33758. doi: 10.1074/jbc.M006230200. [DOI] [PubMed] [Google Scholar]
  170. Palczewski K, Kumasaka T, Hori T, Behnke CA, Motoshima H, Fox BA, et al. Crystal structure of rhodopsin: A G protein-coupled receptor. Science. 2000;289(5480):739–745. doi: 10.1126/science.289.5480.739. [DOI] [PubMed] [Google Scholar]
  171. Palucha A, Klak K, Branski P, van der Putten H, Flor PJ, Pilc A. Activation of the mGlu7 receptor elicits antidepressant-like effects in mice. Psychopharmacology (Berl) 2007;194(4):555–562. doi: 10.1007/s00213-007-0856-2. [DOI] [PubMed] [Google Scholar]
  172. Parelkar NK, Wang JQ. Upregulation of metabotropic glutamate receptor 8 mRNA expression in the rat forebrain after repeated amphetamine administration. Neuroscience Letters. 2008;433(3):250–254. doi: 10.1016/j.neulet.2008.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Parmentier ML, Prezeau L, Bockaert J, Pin JP. A model for the functioning of family 3 GPCRs. Trends in Pharmacological Sciences. 2002;23(6):268–274. doi: 10.1016/s0165-6147(02)02016-3. [DOI] [PubMed] [Google Scholar]
  174. Patel JB, Martin C, Malick JB. Differential antagonism of the anticonflict effects of typical and atypical anxiolytics. European Journal of Pharmacology. 1982;86(2):295–298. doi: 10.1016/0014-2999(82)90331-4. [DOI] [PubMed] [Google Scholar]
  175. Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: A randomized Phase 2 clinical trial. Nature Medicine. 2007;13(9):1102–1107. doi: 10.1038/nm1632. [DOI] [PubMed] [Google Scholar]
  176. Pecknold JC, McClure DJ, Appeltauer L, Wrzesinski L, Allan T. Treatment of anxiety using fenobam (a nonbenzodiazepine) in a double-blind standard (diazepam) placebo-controlled study. Journal of Clinical Psychopharmacology. 1982;2(2):129–133. [PubMed] [Google Scholar]
  177. Pekhletski R, Gerlai R, Overstreet LS, Huang XP, Agopyan N, Slater NT, et al. Impaired cerebellar synaptic plasticity and motor performance in mice lacking the mGluR4 subtype of metabotropic glutamate receptor. The Journal of Neuroscience. 1996;16(20):6364–6373. doi: 10.1523/JNEUROSCI.16-20-06364.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Peters J, Kalivas PW. The group II metabotropic glutamate receptor agonist, LY379268, inhibits both cocaine- and food-seeking behavior in rats. Psychopharmacology (Berl) 2006;186(2):143–149. doi: 10.1007/s00213-006-0372-9. [DOI] [PubMed] [Google Scholar]
  179. Pin JP, Galvez T, Prezeau L. Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacology & Therapeutics. 2003;98(3):325–354. doi: 10.1016/s0163-7258(03)00038-x. [DOI] [PubMed] [Google Scholar]
  180. Pinheiro PS, Mulle C. Presynaptic glutamate receptors: Physiological functions and mechanisms of action. Nature Reviews. Neuroscience. 2008;9(6):423–436. doi: 10.1038/nrn2379. [DOI] [PubMed] [Google Scholar]
  181. Pinkerton AB, Cube RV, Hutchinson JH, James JK, Gardner MF, Rowe BA, et al. Allosteric potentiators of the metabotropic glutamate receptor 2 (mGlu2). Part 3: Identification and biological activity of indanone containing mGlu2 receptor potentiators. Bioorganic & Medicinal Chemistry Letters. 2005;15(6):1565–1571. doi: 10.1016/j.bmcl.2005.01.077. [DOI] [PubMed] [Google Scholar]
  182. Pinkerton AB, Vernier JM, Schaffhauser H, Rowe BA, Campbell UC, Rodriguez DE, et al. Phenyl-tetrazolyl acetophenones: Discovery of positive allosteric potentiatiors for the metabotropic glutamate 2 receptor. Journal of Medicinal Chemistry. 2004;47(18):4595–4599. doi: 10.1021/jm040088h. [DOI] [PubMed] [Google Scholar]
  183. Porter RH, Jaeschke G, Spooren W, Ballard TM, Buttelmann B, Kolczewski S, et al. Fenobam: A clinically validated nonbenzodiazepine anxiolytic is a potent, selective, and noncompetitive mGlu5 receptor antagonist with inverse agonist activity. The Journal of Pharmacology and Experimental Therapeutics. 2005;315(2):711–721. doi: 10.1124/jpet.105.089839. [DOI] [PubMed] [Google Scholar]
  184. Price MR, Baillie GL, Thomas A, Stevenson LA, Easson M, Goodwin R, et al. Allosteric modulation of the cannabinoid CB1 receptor. Molecular Pharmacology. 2005;68(5):1484–1495. doi: 10.1124/mol.105.016162. [DOI] [PubMed] [Google Scholar]
  185. Rasmussen SG, Choi HJ, Rosenbaum DM, Kobilka TS, Thian FS, Edwards PC, et al. Crystal structure of the human beta2 adrenergic G-protein-coupled receptor. Nature. 2007;450(7168):383–387. doi: 10.1038/nature06325. [DOI] [PubMed] [Google Scholar]
  186. Rodriguez AL, Grier MD, Jones CK, Herman EJ, Kane AS, Smith RL, et al. Discovery of novel allosteric modulators of metabotropic glutamate receptor subtype 5 reveals chemical and functional diversity and in vivo activity in rat behavioral models of anxiolytic and antipsychotic activity. Molecular Pharmacology. 2010;78(6):1105–1123. doi: 10.1124/mol.110.067207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Rodriguez AL, Nong Y, Sekaran NK, Alagille D, Tamagnan GD, Conn PJ. A close structural analog of 2-methyl-6-(phenylethynyl)-pyridine acts as a neutral allosteric site ligand on metabotropic glutamate receptor subtype 5 and blocks the effects of multiple allosteric modulators. Molecular Pharmacology. 2005;68(6):1793–1802. doi: 10.1124/mol.105.016139. [DOI] [PubMed] [Google Scholar]
  188. Rondard P, Liu J, Huang S, Malhaire F, Vol C, Pinault A, et al. Coupling of agonist binding to effector domain activation in metabotropic glutamate-like receptors. The Journal of Biological Chemistry. 2006;281(34):24653–24661. doi: 10.1074/jbc.M602277200. [DOI] [PubMed] [Google Scholar]
  189. Ronesi JA, Huber KM. Homer interactions are necessary for metabotropic glutamate receptor-induced long-term depression and translational activation. The Journal of Neuroscience. 2008;28(2):543–547. doi: 10.1523/JNEUROSCI.5019-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Rosenbaum DM, Cherezov V, Hanson MA, Rasmussen SG, Thian FS, Kobilka TS, et al. GPCR engineering yields high-resolution structural insights into beta2-adrenergic receptor function. Science. 2007;318(5854):1266–1273. doi: 10.1126/science.1150609. [DOI] [PubMed] [Google Scholar]
  191. Rosenbrock H, Kramer G, Hobson S, Koros E, Grundl M, Grauert M, et al. Functional interaction of metabotropic glutamate receptor 5 and NMDA-receptor by a metabotropic glutamate receptor 5 positive allosteric modulator. European Journal of Pharmacology. 2010;639(1–3):40–46. doi: 10.1016/j.ejphar.2010.02.057. [DOI] [PubMed] [Google Scholar]
  192. Rowe BA, Schaffhauser H, Morales S, Lubbers LS, Bonnefous C, Kamenecka TM, et al. Transposition of three amino acids transforms the human metabotropic glutamate receptor (mGluR)-3-positive allosteric modulation site to mGluR2, and additional characterization of the mGluR2-positive allosteric modulation site. The Journal of Pharmacology and Experimental Therapeutics. 2008;326(1):240–251. doi: 10.1124/jpet.108.138271. [DOI] [PubMed] [Google Scholar]
  193. Rudd MT, McCauley JA. Positive allosteric modulators of the metabotropic glutamate receptor subtype 2 (mGluR2) Current Topics in Medicinal Chemistry. 2005;5(9):869–884. doi: 10.2174/1568026054750281. [DOI] [PubMed] [Google Scholar]
  194. Sansig G, Bushell TJ, Clarke VR, Rozov A, Burnashev N, Portet C, et al. Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7. The Journal of Neuroscience. 2001;21(22):8734–8745. doi: 10.1523/JNEUROSCI.21-22-08734.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Satow A, Maehara S, Ise S, Hikichi H, Fukushima M, Suzuki G, et al. 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] The Journal of Pharmacology and Experimental Therapeutics. 2008;326(2):577–586. doi: 10.1124/jpet.108.138107. [DOI] [PubMed] [Google Scholar]
  196. Schaffhauser H, Rowe BA, Morales S, Chavez-Noriega LE, Yin R, Jachec C, et al. Pharmacological characterization and identification of amino acids involved in the positive modulation of metabotropic glutamate receptor subtype 2. Molecular Pharmacology. 2003;64(4):798–810. doi: 10.1124/mol.64.4.798. [DOI] [PubMed] [Google Scholar]
  197. Schoepp DD, Jane DE, Monn JA. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology. 1999;38(10):1431–1476. doi: 10.1016/s0028-3908(99)00092-1. [DOI] [PubMed] [Google Scholar]
  198. Schoepp DD, Marek GJ. Preclinical pharmacology of mGlu2/3 receptor agonists: Novel agents for schizophrenia? Current Drug Targets. CNS and Neurological Disorders. 2002;1(2):215–225. doi: 10.2174/1568007024606177. [DOI] [PubMed] [Google Scholar]
  199. Schwartz TW, Holst B. Allosteric enhancers, allosteric agonists and ago-allosteric modulators: Where do they bind and how do they act? Trends in Pharmacological Sciences. 2007;28(8):366–373. doi: 10.1016/j.tips.2007.06.008. [DOI] [PubMed] [Google Scholar]
  200. Sheffler DJ, Conn PJ. Allosteric potentiators of metabotropic glutamate receptor subtype 1a differentially modulate independent signaling pathways in baby hamster kidney cells. Neuropharmacology. 2008;55(4):419–427. doi: 10.1016/j.neuropharm.2008.06.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Shigemoto R, Kinoshita A, Wada E, Nomura S, Ohishi H, Takada M, et al. Differential presynaptic localization of metabotropic glutamate receptor subtypes in the rat hippocampus. The Journal of Neuroscience. 1997;17(19):7503–7522. doi: 10.1523/JNEUROSCI.17-19-07503.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Slassi A, Isaac M, Edwards L, Minidis A, Wensbo D, Mattsson J, et al. Recent advances in non-competitive mGlu5 receptor antagonists and their potential therapeutic applications. Current Topics in Medicinal Chemistry. 2005;5(9):897–911. doi: 10.2174/1568026054750236. [DOI] [PubMed] [Google Scholar]
  203. Snead OC, 3rd, Banerjee PK, Burnham M, Hampson D. Modulation of absence seizures by the GABA(A) receptor: A critical rolefor metabotropic glutamate receptor 4 (mGluR4) The Journal of Neuroscience. 2000;20(16):6218–6224. doi: 10.1523/JNEUROSCI.20-16-06218.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Spinelli S, Ballard T, Gatti-McArthur S, Richards GJ, Kapps M, Woltering T, et al. Effects of the mGluR2/3 agonist LY354740 on computerized tasks of attention and working memory in marmoset monkeys. Psychopharmacology (Berl) 2005;179(1):292–302. doi: 10.1007/s00213-004-2126-x. [DOI] [PubMed] [Google Scholar]
  205. Spooren W, Gasparini F. mGlu5 receptor antagonists: A novel class of anxiolytics? Drug News & Perspectives. 2004;17(4):251–257. doi: 10.1358/dnp.2004.17.4.829052. [DOI] [PubMed] [Google Scholar]
  206. Spooren WP, Gasparini F, van der Putten H, Koller M, Nakanishi S, Kuhn R. Lack of effect of LY314582 (a group 2 metabotropic glutamate receptor agonist) on phencyclidine-induced locomotor activity in metabotropic glutamate receptor 2 knockout mice. European Journal of Pharmacology. 2000;397(1):R1–2. doi: 10.1016/s0014-2999(00)00269-7. [DOI] [PubMed] [Google Scholar]
  207. Stachowicz K, Branski P, Klak K, van der Putten H, Cryan JF, Flor PJ, et al. Selective activation of metabotropic G-protein-coupled glutamate 7 receptor elicits anxiolytic-like effects in mice by modulating GABAergic neurotransmission. Behavioural Pharmacology. 2008;19(5–6):597–603. doi: 10.1097/FBP.0b013e32830cd839. [DOI] [PubMed] [Google Scholar]
  208. Stachowicz K, Chojnacka-Wojcik E, Klak K, Pilc A. Anxiolytic-like effects of group III mGlu receptor ligands in the hippocampus involve GABAA signaling. Pharmacological Reports. 2006;58(6):820–826. [PubMed] [Google Scholar]
  209. Stachowicz K, Klak K, Klodzinska A, Chojnacka-Wojcik E, Pilc A. Anxiolytic-like effects of PHCCC, an allosteric modulator of mGlu4 receptors, in rats. European Journal of Pharmacology. 2004;498(1–3):153–156. doi: 10.1016/j.ejphar.2004.07.001. [DOI] [PubMed] [Google Scholar]
  210. Stefani MR, Moghaddam B. Activation of type 5 metabotropic glutamate receptors attenuates deficits in cognitive flexibility induced by NMDA receptor blockade. European Journal of Pharmacology. 2010;639(1–3):26–32. doi: 10.1016/j.ejphar.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Stockton JM, Birdsall NJ, Burgen AS, Hulme EC. Modification of the binding properties of muscarinic receptors by gallamine. Molecular Pharmacology. 1983;23(3):551–557. [PubMed] [Google Scholar]
  212. Sugihara H, Inoue T, Nakanishi S, Fukuda Y. A late ON response remains in visual response of the mGluR6-deficient mouse. Neuroscience Letters. 1997;233(2–3):137–140. doi: 10.1016/s0304-3940(97)00656-3. [DOI] [PubMed] [Google Scholar]
  213. Suzuki G, Kawagoe-Takaki H, Inoue T, Kimura T, Hikichi H, Murai T, et al. Correlation of receptor occupancy of metabotropic glutamate receptor subtype 1 (mGluR1) in mouse brain with in vivo activity of allosteric mGluR1 antagonists. Journal of Pharmacological Sciences. 2009;110(3):315–325. doi: 10.1254/jphs.09011fp. [DOI] [PubMed] [Google Scholar]
  214. Suzuki G, Kimura T, Satow A, Kaneko N, Fukuda J, Hikichi H, et al. Pharmacological characterization of a new, orally active and potent allosteric metabotropic glutamate receptor 1 antagonist, 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) The Journal of Pharmacology and Experimental Therapeutics. 2007a;321(3):1144–1153. doi: 10.1124/jpet.106.116574. [DOI] [PubMed] [Google Scholar]
  215. Suzuki G, Tsukamoto N, Fushiki H, Kawagishi A, Nakamura M, Kurihara H, et al. In vitro pharmacological characterization of novel isoxazolopyridone derivatives as allosteric metabotropic glutamate receptor 7 antagonists. The Journal of Pharmacology and Experimental Therapeutics. 2007b;323(1):147–156. doi: 10.1124/jpet.107.124701. [DOI] [PubMed] [Google Scholar]
  216. Swanson CJ, Bures M, Johnson MP, Linden AM, Monn JA, Schoepp DD. Metabotropic glutamate receptors as novel targets for anxiety and stress disorders. Nature Reviews. Drug Discovery. 2005;4(2):131–144. doi: 10.1038/nrd1630. [DOI] [PubMed] [Google Scholar]
  217. Tamaru Y, Nomura S, Mizuno N, Shigemoto R. Distribution of metabotropic glutamate receptor mGluR3 in the mouse CNS: Differential location relative to pre- and postsynaptic sites. Neuroscience. 2001;106(3):481–503. doi: 10.1016/s0306-4522(01)00305-0. [DOI] [PubMed] [Google Scholar]
  218. Tatarczynska E, Klodzinska A, Chojnacka-Wojcik E, Palucha A, Gasparini F, Kuhn R, et al. Potential anxiolytic- and antidepressant-like effects of MPEP, a potent, selective and systemically active mGlu5 receptor antagonist. British Journal of Pharmacology. 2001;132(7):1423–1430. doi: 10.1038/sj.bjp.0703923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Thathiah A, De Strooper B. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nature Reviews. Neuroscience. 2011;12(2):73–87. doi: 10.1038/nrn2977. [DOI] [PubMed] [Google Scholar]
  220. Tong Q, Kirchgessner AL. Localization and function of metabotropic glutamate receptor 8 in the enteric nervous system. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2003;285(5):G992–G1003. doi: 10.1152/ajpgi.00118.2003. [DOI] [PubMed] [Google Scholar]
  221. Tong Q, Ouedraogo R, Kirchgessner AL. Localization and function of group III metabotropic glutamate receptors in rat pancreatic islets. American Journal of Physiology. Endocrinology and Metabolism. 2002;282(6):E1324–E1333. doi: 10.1152/ajpendo.00460.2001. [DOI] [PubMed] [Google Scholar]
  222. Toyono T, Seta Y, Kataoka S, Harada H, Morotomi T, Kawano S, et al. Expression of the metabotropic glutamate receptor, mGluR4a, in the taste hairs of taste buds in rat gustatory papillae. Archives of Histology and Cytology. 2002;65(1):91–96. doi: 10.1679/aohc.65.91. [DOI] [PubMed] [Google Scholar]
  223. Tsuchiya D, Kunishima N, Kamiya N, Jingami H, Morikawa K. Structural views of the ligand-binding cores of a metabotropic glutamate receptor complexed with an antagonist and both glutamate and Gd3+ Proceedings of the National Academy of Sciences of the United States of America. 2002;99(5):2660–2665. doi: 10.1073/pnas.052708599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Tsuno N. Donepezil in the treatment of patients with Alzheimer’s disease. Expert Review of Neurotherapeutics. 2009;9(5):591–598. doi: 10.1586/ern.09.23. [DOI] [PubMed] [Google Scholar]
  225. Uehara S, Muroyama A, Echigo N, Morimoto R, Otsuka M, Yatsushiro S, et al. Metabotropic glutamate receptor type 4 is involved in autoinhibitory cascade for glucagon secretion by alpha-cells of islet of Langerhans. Diabetes. 2004;53(4):998–1006. doi: 10.2337/diabetes.53.4.998. [DOI] [PubMed] [Google Scholar]
  226. Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B, Weinstein H, et al. Functional selectivity and classical concepts of quantitative pharmacology. The Journal of Pharmacology and Experimental Therapeutics. 2007;320(1):1–13. doi: 10.1124/jpet.106.104463. [DOI] [PubMed] [Google Scholar]
  227. Ure J, Baudry M, Perassolo M. Metabotropic glutamate receptors and epilepsy. Journal of the Neurological Sciences. 2006;247(1):1–9. doi: 10.1016/j.jns.2006.03.018. [DOI] [PubMed] [Google Scholar]
  228. Valenti O, Mannaioni G, Seabrook GR, Conn PJ, Marino MJ. Group III metabotropic glutamate-receptor-mediated modulation of excitatory transmission in rodent substantia nigra pars compacta dopamine neurons. The Journal of Pharmacology and Experimental Therapeutics. 2005;313(3):1296–1304. doi: 10.1124/jpet.104.080481. [DOI] [PubMed] [Google Scholar]
  229. Valenti O, Marino MJ, Wittmann M, Lis E, DiLella AG, Kinney GG, et al. Group III metabotropic glutamate receptor-mediated modulation of the striatopallidal synapse. The Journal of Neuroscience. 2003;23(18):7218–7226. doi: 10.1523/JNEUROSCI.23-18-07218.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Vanejevs M, Jatzke C, Renner S, Muller S, Hechenberger M, Bauer T, et al. Positive and negative modulation of group I metabotropic glutamate receptors. Journal of Medicinal Chemistry. 2008;51(3):634–647. doi: 10.1021/jm0611298. [DOI] [PubMed] [Google Scholar]
  231. Varney MA, Cosford ND, Jachec C, Rao SP, Sacaan A, Lin FF, et al. SIB-1757 and SIB-1893: Selective, noncompetitive antagonists of metabotropic glutamate receptor type 5. The Journal of Pharmacology and Experimental Therapeutics. 1999;290(1):170–181. [PubMed] [Google Scholar]
  232. Varney MA, Gereau RWt. Metabotropic glutamate receptor involvement in models of acute and persistent pain: Prospects for the development of novel analgesics. Current Drug Targets. CNS and Neurological Disorders. 2002;1(3):283–296. doi: 10.2174/1568007023339300. [DOI] [PubMed] [Google Scholar]
  233. Wieronska JM, Szewczyk B, Branski P, Palucha A, Pilc A. Antidepressant-like effect of MPEP, a potent, selective and systemically active mGlu5 receptor antagonist in the olfactory bulbectomized rats. Amino Acids. 2002;23(1–3):213–216. doi: 10.1007/s00726-001-0131-5. [DOI] [PubMed] [Google Scholar]
  234. Winquist RJ, Pan JQ, Gribkoff VK. Use-dependent blockade of Cav2.2 voltage-gated calcium channels for neuropathic pain. Biochemical Pharmacology. 2005;70(4):489–499. doi: 10.1016/j.bcp.2005.04.035. [DOI] [PubMed] [Google Scholar]
  235. Woltering TJ, Wichmann J, Goetschi E, Knoflach F, Ballard TM, Huwyler J, et al. Synthesis and characterization of 1,3-dihydro-benzo[b][1,4]diazepin-2-one derivatives: Part 4. In vivo active potent and selective non-competitive metabotropic glutamate receptor 2/3 antagonists. Bioorganic & Medicinal Chemistry Letters. 2010;20(23):6969–6974. doi: 10.1016/j.bmcl.2010.09.125. [DOI] [PubMed] [Google Scholar]
  236. Woolley ML, Pemberton DJ, Bate S, Corti C, Jones DN. The mGlu2 but not the mGlu3 receptor mediates the actions of the mGluR2/3 agonist, LY379268, in mouse models predictive of antipsychotic activity. Psychopharmacology (Berl) 2008;196(3):431–440. doi: 10.1007/s00213-007-0974-x. [DOI] [PubMed] [Google Scholar]
  237. Wu B, Chien EY, Mol CD, Fenalti G, Liu W, Katritch V, et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science. 2010;330(6007):1066–1071. doi: 10.1126/science.1194396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Xie X, Ramirez DR, Lasseter HC, Fuchs RA. Effects of mGluR1 antagonism in the dorsal hippocampus on drug context-induced reinstatement of cocaine-seeking behavior in rats. Psychopharmacology (Berl) 2010;208(1):1–11. doi: 10.1007/s00213-009-1700-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Yan QJ, Rammal M, Tranfaglia M, Bauchwitz RP. Suppression of two major Fragile X Syndrome mouse model phenotypes by the mGluR5 antagonist MPEP. Neuropharmacology. 2005;49(7):1053–1066. doi: 10.1016/j.neuropharm.2005.06.004. [DOI] [PubMed] [Google Scholar]
  240. Yarnitzky T, Levit A, Niv MY. Homology modeling of G-protein-coupled receptors with X-ray structures on the rise. Current Opinion in Drug Discovery & Development. 2010;13(3):317–325. [PubMed] [Google Scholar]
  241. Yokoi M, Kobayashi K, Manabe T, Takahashi T, Sakaguchi I, Katsuura G, et al. Impairment of hippocampal mossy fiber LTD in mice lacking mGluR2. Science. 1996;273(5275):645–647. doi: 10.1126/science.273.5275.645. [DOI] [PubMed] [Google Scholar]
  242. Zhang Y, Rodriguez AL, Conn PJ. Allosteric potentiators of metabotropic glutamate receptor subtype 5 have differential effects on different signaling pathways in cortical astrocytes. The Journal of Pharmacology and Experimental Therapeutics. 2005;315(3):1212–1219. doi: 10.1124/jpet.105.090308. [DOI] [PubMed] [Google Scholar]
  243. Zhang GC, Vu K, Parelkar NK, Mao LM, Stanford IM, Fibuch EE, et al. Acute administration of cocaine reduces metabotropic glutamate receptor 8 protein expression in the rat striatum in vivo. Neuroscience Letters. 2009;449(3):224–227. doi: 10.1016/j.neulet.2008.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES