Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor’s role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels.
SIGNIFICANCE STATEMENT
Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7’s potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Introduction
Structure and Function of mGlu7
Of the eight known metabotropic glutamate (mGlu) receptors, mGlu7 is the most widely distributed in the brain, as well as the most evolutionarily conserved (Flor et al., 1997; Niswender and Conn, 2010). mGlu7 is a class C G-protein coupled receptor and a member of the group III metabotropic glutamate receptors, along with mGlu4, mGlu6, and mGlu8 (Niswender and Conn, 2010). This group of mGlu receptors canonically signals through the Gi/o G-protein pathway to attenuate adenylate cyclase activity and regulate ion channel activity (Fig. 1) (Houamed et al., 1991; Tanabe et al., 1992; Okamoto et al., 1994; Niswender and Conn, 2010; Iacovelli et al., 2014). mGlu7 is localized predominantly presynaptically within the brain and is activated by high μM to mM concentrations of the primary excitatory neurotransmitter of the central nervous system (CNS), glutamate (Okamoto et al., 1994). This localization is further consolidated to the active zone of the presynaptic terminal (Somogyi et al., 2003), where the protein organizes in dense clusters partially dependent on interactions with the transsynaptic adhesion proteins Extracellular leucine-rich repeat and fibronectin type III domain containing 1 and 2 (ELFN1/2), as well as intracellular scaffolding protein interacting with C kinase 1 (PICK1) (El Far et al., 2000; El Far and Betz, 2002; Perroy et al., 2002; Bertaso et al., 2008; Niswender and Conn, 2010; Tomioka et al., 2014; Stachniak et al., 2019; Park et al., 2020). Additionally, the localization and downstream signal transduction of mGlu7 is dependent on a number of other cytosolic proteins, such as calmodulin (Nakajima et al., 1999; O’Connor et al., 1999; El Far and Betz, 2002) and Munc 13-1 (Martín et al., 2010). mGlu7 exhibits widespread expression in the CNS but exhibits particularly high expression within the amygdala, hippocampus, and hypothalamus (Kinoshita et al., 1998; Fisher et al., 2018). The receptor is also detected at low levels in peripheral tissues in the reproductive system, endocrine tissues, and smooth muscle (Uhlén et al., 2015; Alam et al., 2020; Sjöstedt et al., 2020).
Fig. 1.
Cryo-electron microscopy image of mGlu7 homodimer with accompanying downstream molecular pathways associated with receptor activation in response to high synaptic levels of glutamate. Structure PDB ID: 7EPC from Du et al. (2021). Structures of human mGlu2 and mGlu7 homo- and heterodimers (Du et al., 2021). Figure created with BioRender.com. CaMKII, calmodulin-dependent kinase II; CREB, cAMP response element binding protein; ERK, extracellular-regulated kinase; GIRK, G-protein coupled inwardly rectifying potassium channel; JNK, c-Jun N-terminal kinase; MEK, mitogen-activated protein kinase kinase; MEKK, mitogen-activated protein kinase kinase kinase; mGlu7, metabotropic glutamate receptor 7; MKK4, mitogen-activated protein kinase kinase 4; MKK7, mitogen-activated protein kinase kinase 7; PICK1, protein interacting with C kinase 1; PKCα, protein kinase C alpha.
At the level of protein structure, mGlu7 is embedded within the cell membrane by a 7-transmembrane domain and flanked by an N-terminal glutamate-binding domain, also known as the Venus flytrap domain, a cysteine-rich domain, and a C-terminal cytoplasmic domain (Pin et al., 2003; Ferraguti and Shigemoto, 2006; Du et al., 2021). The transmembrane domain and C-terminal domain regions are responsible for associating with heterotrimeric G-proteins for signal transduction, while the N-terminal Venus flytrap domain and cysteine-rich domain are responsible for ligand binding and also play roles in receptor dimerization (Niswender and Conn, 2010; Kniazeff et al., 2011; Levitz et al., 2016; Koehl et al., 2019; Fisher et al., 2021; Habrian et al., 2023). This dimerization of mGlu7, as well as all mGlu receptors, is essential for proper signaling and necessary for trafficking from the endoplasmic reticulum to the plasma membrane (Conn and Pin, 1997; Kunishima et al., 2000; Jingami et al., 2003; Pin et al., 2003; Niswender and Conn, 2010). The mGlu7 receptor monomer is also capable of heterodimerization with members of the group II or group III mGlu receptor family (Fig. 2) (Doumazane et al., 2011; Levitz et al., 2016; Du et al., 2021); heterodimerization has distinct effects on receptor pharmacology and can further alter the characteristics of intracellular signaling downstream of mGlu activation (Doumazane et al., 2011; Kammermeier, 2012; Yin and Niswender, 2014; Liu et al., 2017; Moreno Delgado et al., 2017; Habrian et al., 2019; Lee et al., 2020, 2023; Thibado et al., 2021; Xiang et al., 2021; Lin et al., 2022; Wang et al., 2023).
Fig. 2.
Currently known mGlu heterodimers containing mGlu7 and the functional/pharmacological effects of heterodimerization in comparison with mGlu7/7 homodimer activity. mGlu2/7 and mGlu3/7 heterodimers have shown pharmacological changes in ligand binding, as well as functional changes in response to ligands compared with mGlu7 homodimers (Du et al., 2021; McCullock and Kammermeier, 2021; Habrian et al., 2023). Changes in mGlu7 allosteric modulator function have also been reported at the Schaffer collateral–Cornu Ammonis 1 synapse region of the hippocampus that are consistent with the expression of an mGlu7/8 heterodimer (Lin et al., 2022). Figure created with BioRender.com
While downstream signaling increases in complexity in the context of mGlu7 heterodimers, canonical mGlu7 signaling is primarily mediated by activity of Gi/o G-proteins (Fig. 1) (Okamoto et al., 1994). Both Gα and βγ subunits play distinct roles in mGlu7-mediated synaptic release control, which work together to modulate neurotransmitter release. For example, Gα activation leads to adenylate cyclase inhibition, reduction in cAMP levels, and reduced protein kinase A (PKA) activity (Millán et al., 2002; Pelkey et al., 2008). While it is known that cAMP levels and PKA activation are important for synaptic release control (Trudeau et al., 1996; Kaneko and Takahashi, 2004; Nagy et al., 2004; Cho et al., 2015), the function of these effectors in modulating neurotransmitter release via mGlu7 activation is not fully understood. cAMP reduction contributes to decreases in presynaptic calcium ion (Ca2+) flux, along with essential functions of the βγ subunits in inhibiting presynaptic Ca2+ channels and activating protein kinase C (PKC) (Perroy et al., 2000; Martín et al., 2007, 2010, 2011). This inhibition of Ca2+ flux is also dependent on mGlu7’s interaction with PICK1 (Perroy et al., 2002; Bertaso et al., 2008). While it has been shown that this reduction in Ca2+ flux is capable of reducing synapsin-1 phosphorylation, modulating synaptic release (Hay et al., 2000), it is also hypothesized that mGlu7 activation reduces phosphorylation of synapsin-1 through a cAMP/PKA-dependent mechanism, which is also predicted to reduce neurotransmitter release (Fig. 1) (Hay et al., 2000; Patzke et al., 2019, 2021). Additionally, there is evidence to suggest that mGlu7 can directly regulate synaptic release dynamics through translocation of Munc 13-1, an essential component of synaptic release machinery (Martín et al., 2010). This variety of direct and indirect mechanisms could give mGlu7 considerable control over synaptic function depending upon its agonist-mediated or constitutive activation status.
βγ-mediated mGlu7 signaling also activates potassium efflux through G protein-coupled inwardly rectifying potassium channels (Saugstad et al., 1996; Niswender et al., 2010), causing hyperpolarization of the presynaptic terminal to provide an additional mechanism of neurotransmitter release control (Fig. 1). Although activation of mGlu7 is generally associated with depression of synaptic release (Gigg et al., 1994; Okamoto et al., 1994; Saugstad et al., 1996; Schoepp, 2001; Pelkey et al., 2005; Martín et al., 2007; Summa et al., 2013; Klar et al., 2015; Gogliotti et al., 2017; Girard et al., 2019), its localization and stimulation patterns can determine the type of plasticity induced after its activation. As an autoreceptor, short-term activation of mGlu7 leads to synaptic depression, while repeated mGlu7 stimulation at the presynapse can lead to synaptic potentiation (Pelkey et al., 2005, 2008). Additionally, mGlu7 functions as a heteroreceptor on γ-aminobutyric acid (GABA)-releasing interneurons of the hippocampus and amygdala, and its activation at these locations is essential for neuronal plasticity (Gee et al., 2014; Klar et al., 2015). Activation of mGlu7 at these heteroreceptor locations reduces GABA-mediated depression of excitatory neurons, allowing for enhanced plasticity and activity of local excitatory neurons and synapses (Gee et al., 2014; Klar et al., 2015). This function is directly contrary to mGlu7’s canonical role when expressed at excitatory presynapses, which represses continued glutamate release.
In addition to these mechanisms, mGlu7-expressing synapses can also be controlled via noncanonical signaling. For example, orthosteric activation of mGlu7 can induce signaling through β-arrestins 1 and 2 following receptor phosphorylation by G-protein coupled receptor kinases (GRKs). These proteins activate (β-arrestin 1) or inhibit (β-arrestin 2) the mitogen-associated protein kinase cascade, respectively (Fig. 1) (Iacovelli et al., 2014). Evidence also suggests that mGlu7 exhibits constitutive activity that contributes to the regulation of tonic neurotransmitter levels (Kammermeier, 2015), and mGlu7 has been shown to potentiate glutamate release upon prolonged stimulation (Martín et al., 2010, 2011). These extensive intracellular signaling pathways highlight the variety and complexity of mechanisms by which mGlu7 can modulate synaptic release and provide evidence that these mechanisms may play a role in vital cognitive processes.
mGlu7 is encoded by the GRM7 gene, which is located at Chr3p26 and consists of 880,419 base pairs and 10 exons (O’Leary et al., 2016). GRM7 has many splice variants containing 3′ alterations to the mRNA (Schulz et al., 2002), but the main isoforms expressed in the human brain are splice variants 1 and 2, encoding mGlu7a and mGlu7b, respectively (O’Leary et al., 2016). mGlu7b mRNA includes a 92-nucleotide insertion at the 3′ end of the gene, which translates to a 7 amino acid addition at the C-terminus of the protein (Flor et al., 1997). Although mGlu7a and mGlu7b have slightly different temporal and spatial expression patterns (Flor et al., 1997; Monn et al., 1997; Linden et al., 2004), to date, this insertion has not been observed to confer any functional changes to the mGlu7 receptor. This extensive gene also contains numerous, clinically significant single nucleotide polymorphism (SNP) or mutation sites within both introns and exons (O’Leary et al., 2016). These nucleotide alterations are often correlated with neurodevelopmental disorders, psychiatric disorders, or learning disabilities, even when present in gene introns where it is not clear how/if they affect protein sequence or structure (Elia et al., 2011; Park et al., 2013; Tomioka et al., 2014; Liu et al., 2015; Bedogni et al., 2016; Charng et al., 2016; Reuter et al., 2017; Dark et al., 2018; Noroozi et al., 2019; Marafi et al., 2020; Fisher et al., 2021; Zhang et al., 2021; Song et al., 2021; Chaumette et al., 2022; Januel et al., 2024).
Cognitive Disorders and Dysfunction
Cognitive processes are applications of attention, memory, and learning and the ability to call upon formed memories in the future (Millan et al., 2012). Control of cognitive function arises from a variety of sources at the molecular, cellular, and histologic levels within the brain. This is driven by various mechanisms including phosphorylation imbalance, redox states, and neurotransmitter release/tone (Cui et al., 2008; Giralt et al., 2012; Yabuki et al., 2014; Kaur and Sharma, 2022). Cognitive dysfunction (used interchangeably with the terms cognitive deficit or cognitive impairment) encompasses impacts on cognitive processes associated with underlying conditions such as psychiatric disorders, neurodevelopmental disorders, neurodegenerative diseases, and aging. These conditions and their effects on cognition have been reviewed on numerous occasions (Levy, 1994; Gräff and Mansuy, 2009; Giralt et al., 2012; Millan et al., 2012; Yabuki et al., 2014; Kim et al., 2015; Kaur and Sharma, 2022; Dhakal and Bobrin, 2023; Hoglund et al., 2023). Cognitive disorders differ from cognitive dysfunction in that they are standalone diagnoses unassociated with an underlying condition (Dhakal and Bobrin, 2023). The current International Classification of Diseases (ICD) codes identify various levels of cognitive disorder severity, each categorized with its own respective code (https://www.cdc.gov/nchs/icd/icd10cm_browsertool.htm). However, there is often difficulty in translation of these codes internationally, as each nation adopts and revises them to fit their own medical system (Jetté et al., 2010). Despite these differences, patients diagnosed with cognitive disorders often have variable or unknown origins of the disorders, which complicates treatment strategies (Srivastava and Schwartz, 2014). mGlu7 has been directly associated with effects on cognition in mouse models (Bushell et al., 2002; Hölscher et al., 2004; O’Connor et al., 2010; Fisher et al., 2020), and, as mentioned, SNPs or mutations in the GRM7 gene result in intellectual disability and developmental delay in humans. These observations suggest that the receptor is an underexploited target in the treatment of these disorders. Here we focus on how mGlu7 specifically modulates cognition.
Current State of the Field
Models of mGlu7-Related Cognitive Impairments
The consequences of knockout of the Grm7 locus have been studied in animal models for their effects on cognitive function both alone as well as in the context of other phenotypes related to various disease states. Grm7−/− mice exhibit diminished anxiety behaviors, as well as deficits in associative fear learning (Masugi et al., 1999; Sansig et al., 2001; Bushell et al., 2002; Fisher et al., 2020), suggesting that abnormalities induced by Grm7 deletion stem from receptor activity in both the hippocampus and amygdala. Attenuation of mGlu7 function or expression also alters behavioral and electrochemical function in the thalamocortical network (Tassin et al., 2016). Reduced mGlu7 expression during mouse development increases phospho-cAMP response element binding protein (CREB) and Yes-associated protein (YAP) levels, leading to increased neural progenitor cell proliferation and reduced terminal differentiation (Xia et al., 2015). Mutations in the mouse Grm7 gene have also been reported to reduce the GABA-synthesizing enzymes Gad65 and Gad67 and increase reelin expression in the hippocampal formation (Wierońska et al., 2010a), which suggests dysfunctional GABA signaling and increased synaptic connectivity in these animals (Sansig et al., 2001; Bushell et al., 2002; Gogliotti et al., 2017; Fisher et al., 2020, 2021). Interestingly, proper mGlu7 expression and intracellular protein interactions have been shown to be essential in preventing epileptic activity in the brain of mice (El Far et al., 2000; Perroy et al., 2002; Bertaso et al., 2008; Zhang et al., 2008; Girard et al., 2019), suggesting a mechanism for mGlu7 dysregulation in other contexts that display severe abnormalities in neuronal excitability. Mouse models in which mGlu7 function is altered by disrupting protein-protein interactions with Pick1 and Elfn1 also display deficits in cognitive domains (Bertaso et al., 2008; Zhang et al., 2008; Tomioka et al., 2014).
While the face validity of Grm7 knockout models is strong, these models lack construct validity regarding GRM7 mutations seen in patient populations. A clinically relevant Grm7 mutation induced in an exon sequence in rodents results in reduced surface expression of mGlu7, as well as diminished axon outgrowth and presynaptic terminal development (Fisher et al., 2021; Song et al., 2021). This mutation (I154T) appears to cause severe developmental delay and other neurologic phenotypes via a reduction in receptor dimerization (Fisher et al., 2021). This decreased dimerization results in reduced surface expression of mGlu7 receptor protein, although expression levels of Grm7 mRNA remain normal (Fisher et al., 2021). There have been other GRM7 mutations associated with neurodevelopmental disorders as well, such as R622Q, R658W, and T675K, which have been examined for neuronal morphology and function (Song et al., 2021). Of these observed mutations, a dual mutant R658W/T675K mGlu7 receptor has been tested in vitro using isolated rat neurons, and these mutations cause degradation of the mGlu7 protein as well as diminished axon outgrowth that is not able to be rescued by cell treatment with mGlu7 agonists (Song et al., 2021).
Other translational models, such as mouse models of autism spectrum disorder (ASD; associated with numerous cognitive deficits in humans), exhibit reduced mGlu7 expression in the prefrontal cortex, which can be ameliorated by virally increasing mGlu7 expression in this area (Wang et al., 2021). Furthermore, patients with the neurodevelopmental disorder Rett syndrome (RTT) express reduced levels of mGlu7 in the brain, and mice modeling RTT also express low levels of mGlu7 protein (Bedogni et al., 2016; Gogliotti et al., 2017; Freitas and Niswender, 2023). An additional nongenetic model of pain-induced cognitive deficits has also been used to assess the effects of mGlu7 on cognition, as well as algesia (Palazzo et al., 2015). Interestingly, this study identified dramatic changes in mGlu7 expression patterns in response to chronic pain, as well as analgesic, anxiolytic, and procognitive effects of an mGlu7 negative allosteric modulator. These mouse models are useful in probing the functional and behavioral effects of Grm7 coding mutations, neurodevelopmental-related dysfunctions, and changes in mGlu7 expression or signaling in response to external effects. However, mice have limited ability to recapitulate models of intronic SNPs in the human GRM7 gene, which have been correlated with cognitive impairments (Park et al., 2013, 2014; Chaumette et al., 2022).
Clinical Examples of mGlu7-Related Cognitive Impairments
GRM7 SNPs Associated with Cognitive Impairments
Various clinical studies have identified GRM7 SNPs and correlated their possible connection to cognitive deficits and disorders (Park et al., 2013; Yang and Pan, 2013; Liu et al., 2015; Charng et al., 2016; Reuter et al., 2017; Marafi et al., 2020; Zhang et al., 2021; Chaumette et al., 2022; Januel et al., 2024). SNPs rs9826579 and rs3792452 have been associated with attention deficit hyperactivity disorder (ADHD) in Chinese and Korean sample populations, respectively (Park et al., 2013; Zhang et al., 2021), and an association of rs3792452 with methylphenidate responses in ADHD patients has also been reported (Park et al., 2014). While the mechanisms of how these SNPs contribute to the development of ADHD have yet to be fully elucidated, it is hypothesized that they alter mGlu7 expression, dysregulating the balance of GABA and glutamate within the brain (Zhang et al., 2021). Novel de novo GRM7 intronic SNPs, chromosomal deletions, and coding mutations have also been correlated with ASD and developmental delay in patient populations (Yang and Pan, 2013; Liu et al., 2015; Fisher et al., 2018; Marafi et al., 2020). The intronic SNP rs1396409 was also recently shown to be associated with cognitive deficits in schizophrenia patients early in their diagnosis (Chaumette et al., 2022). Marafi et al. conducted an extensive study of GRM7 variants associated with neurodevelopmental disorders, identifying three novel variations, as well as characterization of the clinical phenotypes associated with 11 affected individuals (Marafi et al., 2020). All affected patients suffered from developmental delay, early-onset epilepsy, and microcephaly, with a majority of cases also presenting with cerebral atrophy and hypomyelination as revealed by neuroimaging. Recently, five additional patients have been reported with similar phenotypes (Januel et al., 2024).
RTT
RTT is caused by pathogenic mutations in the methyl CpG-binding protein 2 (MECP2) gene and is characterized by a period of normal development for approximately 6 months after birth, followed by developmental regression accompanied by numerous cognitive, behavioral, and physical impairments (Amir et al., 1999; Neul et al., 2010). As a transcriptional modulator through binding of methylated DNA, MeCP2 regulates the expression of numerous genes (Chahrour et al., 2008). mGlu7 receptor levels are reduced in RTT patient brain samples, and Grm7 transcript is reduced in Mecp2+/− mouse models of RTT (Bedogni et al., 2016; Gogliotti et al., 2017; Freitas and Niswender, 2023). Our laboratory has shown that, in both male (Mecp2−/y) and female (Mecp2+/−) mice, hippocampal long-term potentiation (LTP) deficits, behavioral abnormalities, and cognitive deficits can be corrected with mGlu7 positive allosteric modulators (Gogliotti et al., 2017). These findings directly implicate mGlu7 dysregulation in the cognitive pathophysiology of RTT and demonstrate evidence that potentiation or activation of mGlu7 could be used to rescue these deficits.
ADHD
ADHD is a common learning disability in children, with an incidence of approximately 5% (Polanczyk et al., 2007). This disorder results in reduced attention, as well as hyperlocomotion in some individuals (Dark et al., 2018); these phenotypes reduce children’s effectiveness in school and can greatly impact learning if left untreated during childhood (Pfiffner and Haack, 2014). Genome-wide association studies have established a connection between GRM7 expression and ADHD incidence in the human population (Elia et al., 2011), which is hypothesized to be connected to mGlu7’s role in synaptic plasticity as an essential part of memory and learning (Hölscher et al., 2004; Klar et al., 2015; Bushell et al., 2002). There is also evidence to implicate the ELFN1 gene in ADHD development, as Elfn1 knockout model mice exhibit seizures and hyperactivity phenotypes and mutations in ELFN1 are correlated with ADHD in clinical populations (Tomioka et al., 2014). In mice, Elfn1 plays a major role in recruitment of mGlu7 to the presynaptic active zone (Park et al., 2020; Matsunaga and Aruga, 2021). The similar phenotypes seen in mice and humans with either GRM7 or ELFN1 mutations suggest a crucial level of regulation imparted by the mGlu7/Elfn1 transynaptic interaction. Elfn1 has also been shown to act as an allosteric regulator of group III mGlu receptor activity in vitro (Dunn et al., 2018; Stachniak et al., 2019); therefore, loss of Elfn1, dysregulation of mGlu7 activity, or a combination of these factors could play an essential role in ADHD. Further, it will be important to determine if the interactions of Elfn1 with mGlu7 in vivo complicate the pharmacology of mGlu7 ligands when the two are coexpressed, not only in ADHD but in any scenario where mGlu7 is a potential drug target.
Current Research Areas and Future Directions
Receptor Dimerization and Localization
As previously mentioned, receptor dimerization of mGlu7 is essential for surface expression and function (Pin et al., 2003; Habrian et al., 2019; Lee et al., 2020, 2023; Du et al., 2021; Fisher et al., 2021; Thibado et al., 2021; Lin et al., 2022; Wang et al., 2023). The critical nature of dimerization for mGlu7 is evident from our findings that the mGlu7 I154T point mutation, one of the first mutations observed in patients (Charng et al., 2016; Marafi et al., 2020; Fisher et al., 2021), results in reduced surface expression in vitro due to impaired dimerization and almost no receptor expression in mice (Fisher et al., 2021). These effects were studied solely in the context of mGlu7 receptor homodimerization. As recent research has begun to elucidate patterns of heterodimerization of mGlu7 and its effects on receptor function and pharmacology (Yin and Niswender, 2014; Liu et al., 2017; Habrian et al., 2019; Du et al., 2021; McCullock and Kammermeier, 2021; Xiang et al., 2021; Lin et al., 2022), it will be interesting to revisit the effects of various clinical mGlu7 mutations on heterodimerization of the receptor with other mGlus.
The ability of mGlu receptors to heterodimerize was first characterized by the Pin laboratory in 2011 (Doumazane et al., 2011), where it was determined that group I receptors could heterodimerize within their group (mGlu1 and mGlu5), while group II and group III mGlu receptors could heterodimerize between groups. These heterodimers have high biologic relevance, as they have been physically (Meng et al., 2022) and pharmacologically (Yin and Niswender, 2014; Moreno Delgado et al., 2017; Habrian et al., 2019, 2023; Xiang et al., 2021; Lin et al., 2022) observed throughout the brain. These patterns of heterodimerization also have implications for further therapeutic development, as they confer pharmacological changes to the receptors compared with their homodimeric counterparts (Fig. 2). For example, mGlu2/7 heterodimers have been observed in the hippocampus (Habrian et al., 2019), and these receptors display cooperativity that is not present in homodimers. Additionally, L-2-amino-4-phosphonobutyric acid (L-AP4), a group III mGlu receptor agonist, exhibits threefold higher efficacy at an mGlu2/7 heterodimer when compared with an mGlu7/7 homodimer (Du et al., 2021). An mGlu7 negative allosteric modulator (NAM) can even restrict downstream signaling of heterodimer activation in the presence of an mGlu2 positive allosteric modulator (PAM) at mGlu2/7 heterodimers when treated with group 2-specific agonist LY354740 (Du et al., 2021). These findings suggest there is biased association of G-proteins to heterodimer subunits. Separate studies have also concluded that mGlu3/7 heterodimers have higher basal signaling and increased activation kinetics compared with either mGlu3/3 or mGlu7/7 homodimers, and this activity is sensitive to orthosteric antagonists (Habrian et al., 2019; Kukaj et al., 2023). Recently, we identified a signature consistent with mGlu7/8 heterodimers at Schaffer collateral-CA1 (SC-CA1) synapses of the hippocampus that segregates compounds into those that block mGlu7/8 heterodimers and those that do not (Lin et al., 2022), providing additional challenges in understanding mGlu7 pharmacology and drug discovery efforts to target this receptor therapeutically.
There are also numerous post-translational modifications recently identified that contribute to mGlu7 surface expression and synaptic localization. For example, N-linked glycosylation at 4 asparagine residues of mGlu7 is essential for its release from the endoplasmic reticulum and further trafficking to the cell membrane (Park et al., 2020). Once expressed in the outer cell membrane, the activities of neuronal precursor cell-expressed developmentally downregulated 4 (Nedd4) E3 ubiquitin ligase, as well as β-arrestins, are essential for endocytosis and degradation of the receptor, both tonically and in response to agonist stimulation (Lee et al., 2019). Recent advances have been made in understanding active zone clustering of mGlu7, which depends on both cis- and trans-acting mechanisms (Boudin et al., 2000; Suh et al., 2008; Tomioka et al., 2014; Lee et al., 2019; Park et al., 2020; Kang et al., 2021). As discussed earlier, Elfn1 functions trans-synaptically to facilitate mGlu7 clustering, as well as modulate its pharmacology and inducing constitutive activity (Stachniak et al., 2019). This localization provided by Elfn1 is exceptionally important considering mGlu7’s low affinity for glutamate (Okamoto et al., 1994; Niswender and Conn, 2010). Interestingly, the trans-synaptic interaction of Elfn1 with mGlu7 enhances constitutive activity of mGlu7, reducing synaptic glutamate release (Stachniak et al., 2019). This anchoring function of Elfn1 has been observed to occur with various group III mGlu receptors, reducing cAMP concentrations and modifying agonist pharmacology, as well as increasing constitutive activity of these receptors (Dunn et al., 2018). These dimerization and localization mechanisms are essential to mGlu7 surface expression and function, implicating numerous proteins as alternative mechanisms potentially modulating mGlu7 signaling in various disease states.
Intracellular Signaling Pathways
It has recently been elucidated that neuromodulatory and retrograde synaptic signaling in the CNS and peripheral nervous system is capable of increasing phosphorylation of synapsin-1 through cAMP-dependent pathways, directly modulating vesicular release potentials (Patzke et al., 2019; Polishchuk et al., 2023). These findings suggest that synapsin-1 phosphorylation could therefore be attenuated by reduction of cAMP via neuromodulatory signals (such as via mGlu7 activation) as well. Coincidentally, mGlu7 has been previously implicated in modulation of synapsin-1 phosphorylation; this study attributed this effect to reductions in Ca2+ channel activation, and likely reduction of CaMKII activity, although the specific effector was not identified (Hay et al., 2000). These findings suggest a potentially significant role of mGlu7 downstream signaling in the direct modulation of synaptic release through synapsin-1 via two separate pathways. These mechanisms warrant further investigation and pharmacological exploitation, as synapsin-1 has been shown to play a role in learning, memory, and developmental deficits in animal models (Kao et al., 2002; Cui et al., 2008). Evidence suggests that mGlu7 can also directly modulate synaptic release through interactions with Munc 13-1, a protein required for synaptic vesicle priming (Martín et al., 2010). These pathways, paired with mGlu7’s known and extensive effects on various other cellular and synaptic regulatory mechanisms discussed previously, may help fully elucidate mGlu7’s observed effects within the hippocampus in attenuating GABA release and facilitating LTP, which is essential at this location for memory formation (Klar et al., 2015). The impact of mGlu7 activation on synaptic release machinery function is a novel area of research requiring further investigation. The use of models knocking out or manipulating various components of the synaptic release machinery, or studies employing mGlu7 ligands to study specific aspects of synaptic release mechanisms, could provide new insight into how mGlu7 activation has a direct effect on synaptic vesicle release.
Novel findings also suggest differential roles of β-arrestin-mediated signaling and internalization regarding the mGlu family (Lee et al., 2023), which have identified different mechanisms of internalization, recycling, degradation, and signaling based on C-terminal sequences of mGlu receptors. Findings also show that these effects are further modulated by the formation of mGlu heterodimers. While mGlu7 was not extensively interrogated in this study, previous results indicate a major role of β-arrestins in mGlu7 expression and function (Iacovelli et al., 2014; Lee et al., 2019). This suggests unknown β-arrestin-mediated modulation of mGlu7 heterodimer function that could have implications on pharmacological activity at these receptors. Utilizing models to interrogate patterns of G protein-coupled receptor kinase phosphorylation and β-arrestin binding of mGlu7 heterodimers would provide more insight into how stimulation patterns can affect surface presence and downstream signaling of these heterodimers and build upon current findings reported for other mGlu heterodimers (Lee et al., 2023), as well as mGlu7 homodimers (Iacovelli et al., 2014). Elucidating these processes would increase understanding of how mGlu7-targeting compounds exert their effects in cell-based assays as well as in in vivo models.
mGlu7 Specific Ligands
The field of mGlu pharmacology relies heavily on the use of tool compounds to agonize, antagonize, and modulate the receptors to observe their physiologic function and validate their potential as drug targets (Fig. 3). Agonists have been used for decades to stimulate the mGlu receptors, with the prototypical group III mGlu receptor agonist being L-AP4 (Graham and Burgoyne, 1994; Schoepp et al., 1996; Frauli et al., 2007). A series of compounds was later developed to probe the effects of group III mGlus both in vitro and in vivo. LSP1-2111, and the related LSP1-3801, were the first of these compounds, displaying anxiolytic effects in mouse models (Acher et al., 2010, 2012; Wierońska et al., 2010b; Amalric et al., 2013; Cajina et al., 2013). These compounds were followed by LSP4-2022 and LSP3-2156 (Goudet et al., 2012; Podkowa et al., 2015) and, most recently, LSP2-9166 (Hajasova et al., 2018), which is selective for mGlu4 and mGlu7 compared with the broad group III activity of the previous iterations of LSP compounds as well as L-AP4. The previously mentioned agonists act orthosterically, which imparts difficulty in terms of group III mGlu targeting selectivity. Allosteric agonists have also been developed to increase this selectivity. An mGlu7-specific allosteric agonist, AMN082, has been extensively investigated (Mitsukawa et al., 2005), although it undergoes rapid metabolism in vivo and exhibits significant off-target activity (Sukoff Rizzo et al., 2011). Two additional studies have recently reported allosteric agonists of the mGlu7 receptor as well. Cid et al. developed a series of allosteric agonists utilizing computational modeling, which have comparable potencies to AMN082 (Cid et al., 2019). Dickson et al. have also reported an mGlu7 allosteric agonist, CVN636, with high potency, selectivity, and favorable CNS penetrance (Dickson et al., 2023).
Fig. 3.
Metabotropic glutamate receptor group III and mGlu7-selective tool compounds organized by their mode of pharmacology. All orthosteric agonists are active at all group III receptors, aside from LSP2-9166, which is active at mGlu4 and mGlu7 and only active at mGlu8 at high concentrations (Acher et al., 2012; Amalric et al., 2013; Hajasova et al., 2018). AMN082 is a selective allosteric agonist (Mitsukawa et al., 2005), while CVN636 (Dickson et al., 2023) and the series of compounds published by Cid et al. (Cid et al., 2019) are also selective, with more favorable in vivo parameters. NAMs shown are active at mGlu7 homodimers, with some displaying activity at mGlu7/8 heterodimers as well (Reed et al., 2020; Lin et al., 2022). VU0155094 and VU0422288 are group III mGlu receptor-selective PAMs (Jalan-Sakrikar et al., 2014), VU6005649 is a dual mGlu7/mGlu8 PAM, and VU6027459 is an mGlu7-selective PAM (Reed et al., 2020). VU6046980 is a highly mGlu7-selective PAM derived from VU6027459 (Kalbfleisch et al., 2023). Figure created with Biorender.com.
The development of allosteric modulators has also provided new tools and therapeutic opportunities to target specific mGlu receptor subtypes. A breakthrough for the group III mGlu receptors was the report of the PAM, N-phenyl-7-(hydroxyamino) cyclopropa[b]chromen-1a-carboxamide, published in 2003 (Maj et al., 2003; Marino et al., 2003), which was the first reported modulator of mGlu4. Two PAMs with activity at mGlu7 were reported in 2014: VU0155094 and VU0422288 (Jalan-Sakrikar et al., 2014). However, these compounds are not selective for mGlu7, with activity at mGlu4 and mGlu8 as well. Further compound screening and optimization has highlighted the difficulty of producing useful in vivo PAM tool compounds (Reed et al., 2020; Kalbfleisch et al., 2023). We recently reported several PAM ligands for mGlu7: VU6027459 (Reed et al., 2020) and the optimized VU6046980 (Kalbfleisch et al., 2023). While these compounds are CNS penetrant and display the necessary pharmacokinetic properties to serve as tool compounds, the scaffold appears to exhibit an off-target sedation activity that is retained in Grm7−/− mice (Kalbfleisch et al., 2023). These challenges in the development of PAMs suggest that further work is needed to identify selective mGlu7 PAMs for preclinical use.
A NAM with specific mGlu7 activity was reported in 2007 with the development of MMPIP (Suzuki et al., 2007). These early allosteric compounds laid the groundwork for the current field of subtype-specific mGlu allosteric modulators. ADX71743 was later reported as an mGlu7-specific NAM, with characterization in vitro as well as in vivo (Kalinichev et al., 2013). We reported VU6010608, a structurally distinct NAM from either the MMPIP or ADX71743 scaffolds, which had potent mGlu7 NAM activity but was metabolically labile (Reed et al., 2019a; Reed et al., 2019b). VU6012962, derived from VU6010608, displays favorable CNS penetrance and high potency (Reed et al., 2019b). VU6012962 was further optimized using iterative libraries of compounds replacing labile amide linkers with oxadiazole bioisosteres characterized through structure-activity relationships to identify VU6019278, a potent mGlu7-specific NAM with improved pharmacokinetic properties for use in vivo (Reed et al., 2019a).
Context-Selective Ligands
Traditional investigation of small molecules acting on mGlu7 has focused on the targeting of mGlu7/7 homodimers. However, numerous mGlu heterodimers have been pharmacologically characterized, and these various combinations exhibit differential pharmacological responses when compared with homodimers (Kammermeier, 2012; Yin and Niswender, 2014; Wang et al., 2023). Considering the extensive expression of mGlu7 across the brain, and the propensity of mGlu receptors to form heterodimers (Okamoto et al., 1994; Conn and Pin, 1997; Doumazane et al., 2011; Du et al., 2021; Lin et al., 2022; Meng et al., 2022; Habrian et al., 2023), tool compounds selective for specific dimer structures of mGlu7 could be exceptionally helpful in elucidating the function of heterodimers versus homodimers, as well as their regional expression and specificity. Two identified mGlu7 NAMs have been shown to attenuate receptor activation in vitro and block hippocampal LTP ex vivo regardless of dimerization with mGlu8, while two similar compounds were shown to only act on mGlu7/7 homodimers in vitro and have no effect on LTP measurements (Lin et al., 2022). These data suggest that allosteric modulators can be used to differentiate the function of mGlu7 homodimers and heterodimers, allowing for a deeper understanding of complex heterodimer expression and function as well as the opportunity to specifically target therapeutic effects to unique receptor conformations with spatial selectivity.
Conclusions
mGlu receptors, including mGlu7, have been well studied for decades as drug targets for numerous neurologic diseases. mGlu7, as the most conserved and highly expressed of the family, poses unique opportunities (and challenges) as a drug target. Observed behavioral alterations in animal models, as well as in humans with knockout or loss-of-function mutations, have identified mGlu7 function as essential in proper neural development and cognitive function, an area that is affected in the pathophysiology of a multitude of diseases yet has few efficacious treatments. The numerous tools for exploring mGlu7 pharmacology have been useful in understanding its basic signaling characteristics and potential therapeutic effects, but there has not yet been an mGlu7-targeting compound studied in the clinic. This is likely a result of our limited understanding of the wide expanse of complex interactions implicating mGlu7 in vivo, an area we are just beginning to understand more in depth. Novel findings in these understudied areas, as well as an increased focus on translational research in the biologic sciences as a whole, create a unique opportunity for more selective, efficacious, and safe treatments to be developed at the bench and transitioned to the bedside where they can benefit patients suffering from debilitating cognitive conditions associated with mGlu7 dysregulation.
Data Availability
This article contains no datasets generated or analyzed during the current study.
Abbreviations
- ADHD
attention deficit hyperactivity disorder
- Ca2+
calcium ion
- CNS
central nervous system
- ELFN1/2
extracellular leucine-rich repeat and fibronectin type III domain-containing 1/2
- GABA
γ-aminobutyric acid
- L-AP4
L-2-amino-4-phosphonobutyric acid
- LTP
long term potentiation
- mGlu
metabotropic glutamate
- NAM
negative allosteric modulator
- PAM
positive allosteric modulator
- PKA
protein kinase A
- RTT
Rett syndrome
- SNP
single-nucleotide polymorphism
Authorship Contributions
Wrote or contributed to the writing of the manuscript: Parent, Niswender.
Footnotes
This work was supported by National Institutes of Health National Institute of Mental Health [Grants MH124671 and MH062646] and National Institute of Neurological Disorders and Stroke [Grants NS132060 and NS031373].
C.M.N. receives royalties and financial support from Acadia Pharmaceuticals Inc. and Boehringer Ingelheim. H.H.P. has no interests to disclose.
References
- Acher F, Pin J-P, Goudet C, Eschalier A, Busserolles J, Rigault D, Lemasson I, Cesarini S, Commare B (2012) inventors, Université d’Auvergne Université Paris Cité, assignee. Hypophosphorous acid derivatives having antihyperalgic activity and biological applications thereof. Patent WO2012156931/US20140107078. 2012 May 16.
- Acher F, Selvam C, Pin J-P (2010) inventors, Centre National de la Recherche Scientifique (CNRS), assignee. Diastereoisomers of hypophosphorous acid derivatives. Patent WO2010106526/EP2408789/US20120016155/JP2012520870/CA2755544. 2010 Mar 19.
- Alam TAgrawal SSeverin JYoung RSAndersson RArner EHasegawa ALizio MRamilowski JAAbugessaisa I, et al. (2020) Comparative transcriptomics of primary cells in vertebrates. Genome Res 30:951–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amalric M, Lopez S, Goudet C, Fisone G, Battaglia G, Nicoletti F, Pin J-P, Acher FC (2013) Group III and subtype 4 metabotropic glutamate receptor agonists: discovery and pathophysiological applications in Parkinson’s disease. Neuropharmacology 66:53–64. [DOI] [PubMed] [Google Scholar]
- Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY (1999) Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet 23:185–188. [DOI] [PubMed] [Google Scholar]
- Bedogni F, Cobolli Gigli C, Pozzi D, Rossi RL, Scaramuzza L, Rossetti G, Pagani M, Kilstrup-Nielsen C, Matteoli M, Landsberger N (2016) Defects during Mecp2 null embryonic cortex development precede the onset of overt neurological symptoms. Cereb Cortex 26:2517–2529. [DOI] [PubMed] [Google Scholar]
- Bertaso FZhang CScheschonka Ade Bock FFontanaud PMarin PHuganir RLBetz HBockaert JFagni L, et al. (2008) PICK1 uncoupling from mGluR7a causes absence-like seizures. Nat Neurosci 11:940–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boudin H, Doan A, Xia J, Shigemoto R, Huganir RL, Worley P, Craig AM (2000) Presynaptic clustering of mGluR7a requires the PICK1 PDZ domain binding site. Neuron 28:485–497. [DOI] [PubMed] [Google Scholar]
- Bushell TJ, Sansig G, Collett VJ, van der Putten H, Collingridge GL (2002) Altered short-term synaptic plasticity in mice lacking the metabotropic glutamate receptor mGlu7. ScientificWorldJournal 2:730–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cajina MNattini MSong DSmagin GJørgensen EBChandrasena GBundgaard CToft DBHuang XAcher F, et al. (2013) Qualification of LSP1-2111 as a brain penetrant group III metabotropic glutamate receptor orthosteric agonist. ACS Med Chem Lett 5:119–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chahrour M, Jung SY, Shaw C, Zhou X, Wong STC, Qin J, Zoghbi HY (2008) MeCP2, a key contributor to neurological disease, activates and represses transcription. Science 320:1224–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charng W-LKaraca ECoban Akdemir ZGambin TAtik MMGu SPosey JEJhangiani SNMuzny DMDoddapaneni H, et al. (2016) Exome sequencing in mostly consanguineous Arab families with neurologic disease provides a high potential molecular diagnosis rate. BMC Med Genomics 9:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaumette BSengupta SMLepage MMalla AIyer SNKebir ODion PARouleau GAKrebs M-OShah JL, et al. ; ICAAR study group (2022) A polymorphism in the glutamate metabotropic receptor 7 is associated with cognitive deficits in the early phases of psychosis. Schizophr Res 249:56–62. [DOI] [PubMed] [Google Scholar]
- Cho RW, Buhl LK, Volfson D, Tran A, Li F, Akbergenova Y, Littleton JT (2015) Phosphorylation of complexin by PKA regulates activity-dependent spontaneous neurotransmitter release and structural synaptic plasticity. Neuron 88:749–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cid JM, Lavreysen H, Tresadern G, Pérez-Benito L, Tovar F, Fontana A, Trabanco AA (2019) Computationally guided identification of allosteric agonists of the metabotropic glutamate 7 receptor. ACS Chem Neurosci 10:1043–1054. [DOI] [PubMed] [Google Scholar]
- Conn PJ, Pin J-P (1997) Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol 37:205–237. [DOI] [PubMed] [Google Scholar]
- Cui Y, Costa RM, Murphy GG, Elgersma Y, Zhu Y, Gutmann DH, Parada LF, Mody I, Silva AJ (2008) Neurofibromin regulation of ERK signaling modulates GABA release and learning. Cell 135:549–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dark C, Homman-Ludiye J, Bryson-Richardson RJ (2018) The role of ADHD associated genes in neurodevelopment. Dev Biol 438:69–83. [DOI] [PubMed] [Google Scholar]
- Dhakal A, Bobrin BD (2023) Cognitive deficits, in StatPearls, StatPearls Publishing, Treasure Island, FL. [PubMed] [Google Scholar]
- Dickson LTeall MChevalier ECheung TLiwicki GMMack SStephenson AWhite KFosbeary RHarrison DC, et al. (2023) Discovery of CVN636: a highly potent, selective, and CNS penetrant mGluR7 allosteric agonist. ACS Med Chem Lett 14:442–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doumazane E, Scholler P, Zwier JM, Trinquet E, Rondard P, Pin J-P (2011) A new approach to analyze cell surface protein complexes reveals specific heterodimeric metabotropic glutamate receptors. FASEB J 25:66–77. [DOI] [PubMed] [Google Scholar]
- Du JWang DFan HXu CTai LLin SHan STan QWang XXu T, et al. (2021) Structures of human mGlu2 and mGlu7 homo- and heterodimers. Nature 594:589–593. [DOI] [PubMed] [Google Scholar]
- Dunn HA, Patil DN, Cao Y, Orlandi C, Martemyanov KA (2018) Synaptic adhesion protein ELFN1 is a selective allosteric modulator of group III metabotropic glutamate receptors in trans. Proc Natl Acad Sci USA 115:5022–5027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El Far O, Airas J, Wischmeyer E, Nehring RB, Karschin A, Betz H (2000) Interaction of the C-terminal tail region of the metabotropic glutamate receptor 7 with the protein kinase C substrate PICK1. Eur J Neurosci 12:4215–4221. [DOI] [PubMed] [Google Scholar]
- El Far O, Betz H (2002) G-protein-coupled receptors for neurotransmitter amino acids: C-terminal tails, crowded signalosomes. Biochem J 365:329–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elia JGlessner JTWang KTakahashi NShtir CJHadley DSleiman PMAZhang HKim CERobison R, et al. (2011) Genome-wide copy number variation study associates metabotropic glutamate receptor gene networks with attention deficit hyperactivity disorder. Nat Genet 44:78–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferraguti F, Shigemoto R (2006) Metabotropic glutamate receptors. Cell Tissue Res 326:483–504. [DOI] [PubMed] [Google Scholar]
- Fisher NMAlHashim ABuch ABBadivuku HSamman MMWeiss KMCestero GIDoes MDRook JMLindsley CW, et al. (2021) A GRM7 mutation associated with developmental delay reduces mGlu7 expression and produces neurological phenotypes. JCI Insight 6:143324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher NMGould RWGogliotti RGMcDonald AJBadivuku HChennareddy SBuch ABMoore AMJenkins MTRobb WH, et al. (2020) Phenotypic profiling of mGlu7 knockout mice reveals new implications for neurodevelopmental disorders. Genes Brain Behav 19:e12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher NM, Seto M, Lindsley CW, Niswender CM (2018) Metabotropic glutamate receptor 7: a new therapeutic target in neurodevelopmental disorders. Front Mol Neurosci 11:387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flor PJ, Van Der Putten H, Rüegg D, Lukic S, Leonhardt T, Bence M, Sansig G, Knöpfel T, Kuhn R (1997) A novel splice variant of a metabotropic glutamate receptor, human mGluR7b. Neuropharmacology 36:153–159. [DOI] [PubMed] [Google Scholar]
- Frauli M, Hubert N, Schann S, Triballeau N, Bertrand H-O, Acher F, Neuville P, Pin J-P, Prézeau L (2007) Amino-pyrrolidine tricarboxylic acids give new insight into group III metabotropic glutamate receptor activation mechanism. Mol Pharmacol 71:704–712. [DOI] [PubMed] [Google Scholar]
- Freitas GA, Niswender CM (2023) GRM7 gene mutations and consequences for neurodevelopment. Pharmacol Biochem Behav 225:173546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gee CEPeterlik DNeuhäuser CBouhelal RKaupmann KLaue GUschold-Schmidt NFeuerbach DZimmermann KOfner S, et al. (2014) Blocking metabotropic glutamate receptor subtype 7 (mGlu7) via the Venus flytrap domain (VFTD) inhibits amygdala plasticity, stress, and anxiety-related behavior. J Biol Chem 289:10975–10987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gigg J, Tan AM, Finch DM (1994) Glutamatergic hippocampal formation projections to prefrontal cortex in the rat are regulated by GABAergic inhibition and show convergence with glutamatergic projections from the limbic thalamus. Hippocampus 4:189–198. [DOI] [PubMed] [Google Scholar]
- Giralt A, Saavedra A, Alberch J, Pérez-Navarro E (2012) Cognitive dysfunction in Huntington’s disease: humans, mouse models and molecular mechanisms. J Huntingtons Dis 1:155–173. [DOI] [PubMed] [Google Scholar]
- Girard BTuduri PMoreno MPSakkaki SBarboux CBouschet TVarrault AVitre JMcCort-Tranchepain IDairou J, et al. (2019) The mGlu7 receptor provides protective effects against epileptogenesis and epileptic seizures. Neurobiol Dis 129:13–28. [DOI] [PubMed] [Google Scholar]
- Gogliotti RGSenter RKFisher NMAdams JZamorano RWalker AGBlobaum ALEngers DWHopkins CRDaniels JS, et al. (2017) mGlu7 potentiation rescues cognitive, social, and respiratory phenotypes in a mouse model of Rett syndrome. Sci Transl Med 9:eaai7459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goudet CVilar BCourtiol TDeltheil TBessiron TBrabet IOueslati NRigault DBertrand H-OMcLean H, et al. (2012) A novel selective metabotropic glutamate receptor 4 agonist reveals new possibilities for developing subtype selective ligands with therapeutic potential. FASEB J 26:1682–1693. [DOI] [PubMed] [Google Scholar]
- Gräff J, Mansuy IM (2009) Epigenetic dysregulation in cognitive disorders. Eur J Neurosci 30:1–8. [DOI] [PubMed] [Google Scholar]
- Graham ME, Burgoyne RD (1994) Activation of metabotropic glutamate receptors by L-AP4 stimulates survival of rat cerebellar granule cells in culture. Eur J Pharmacol 288:115–123. [DOI] [PubMed] [Google Scholar]
- Habrian C, Latorraca N, Fu Z, Isacoff EY (2023) Homo- and hetero-dimeric subunit interactions set affinity and efficacy in metabotropic glutamate receptors. Nat Commun 14:8288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Habrian CH, Levitz J, Vyklicky V, Fu Z, Hoagland A, McCort-Tranchepain I, Acher F, Isacoff EY (2019) Conformational pathway provides unique sensitivity to a synaptic mGluR. Nat Commun 10:5572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hajasova Z, Canestrelli C, Acher F, Noble F, Marie N (2018) Role of mGlu7 receptor in morphine rewarding effects is uncovered by a novel orthosteric agonist. Neuropharmacology 131:424–430. [DOI] [PubMed] [Google Scholar]
- Hay M, Hoang CJ, Hasser EM, Price EM (2000) Activation of metabotropic glutamate receptors inhibits synapsin I phosphorylation in visceral sensory neurons. J Membr Biol 178:195–204. [DOI] [PubMed] [Google Scholar]
- Hoglund BK, Carfagno V, Olive MF, Leyrer-Jackson JM(2023) Metabotropic glutamate receptors and cognition: from underlying plasticity and neuroprotection to cognitive disorders and therapeutic targets, in International Review of Neurobiology (Knackstedt LA, and Schwendt M eds) pp 367–413, Academic Press, New York. [DOI] [PubMed] [Google Scholar]
- Hölscher C, Schmid S, Pilz PKD, Sansig G, van der Putten H, Plappert CF (2004) Lack of the metabotropic glutamate receptor subtype 7 selectively impairs short-term working memory but not long-term memory. Behav Brain Res 154:473–481. [DOI] [PubMed] [Google Scholar]
- Houamed KM, Kuijper JL, Gilbert TL, Haldeman BA, O’Hara PJ, Mulvihill ER, Almers W, Hagen FS (1991) Cloning, expression, and gene structure of a G protein-coupled glutamate receptor from rat brain. Science 252:1318–1321. [DOI] [PubMed] [Google Scholar]
- Iacovelli L, Felicioni M, Nisticò R, Nicoletti F, De Blasi A (2014) Selective regulation of recombinantly expressed mGlu7 metabotropic glutamate receptors by G protein-coupled receptor kinases and arrestins. Neuropharmacology 77:303–312. [DOI] [PubMed] [Google Scholar]
- Jalan-Sakrikar NField JRKlar RMattmann MEGregory KJZamorano REngers DWBollinger SRWeaver CDDays EL, et al. (2014) Identification of positive allosteric modulators VU0155094 (ML397) and VU0422288 (ML396) reveals new insights into the biology of metabotropic glutamate receptor 7. ACS Chem Neurosci 5:1221–1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Januel LChatron NRivier-Ringenbach CCabet SLabalme ASahin YDarvish HKruer MBakhtiari SSanlaville D, et al. (2024) GRM7-related disorder: five additional patients from three independent families and review of the literature. Eur J Med Genet 67:104893. [DOI] [PubMed] [Google Scholar]
- Jetté NQuan HHemmelgarn BDrosler SMaass CMoskal LPaoin WSundararajan VGao SJakob R, et al. ; IMECCHI Investigators (2010) The development, evolution, and modifications of ICD-10: challenges to the international comparability of morbidity data. Med Care 48:1105–1110. [DOI] [PubMed] [Google Scholar]
- Jingami H, Nakanishi S, Morikawa K (2003) Structure of the metabotropic glutamate receptor. Curr Opin Neurobiol 13:271–278. [DOI] [PubMed] [Google Scholar]
- Kalbfleisch JJ, Rodriguez AL, Lei X, Weiss K, Blobaum AL, Boutaud O, Niswender CM, Lindsley CW (2023) Persistent challenges in the development of an mGlu7 PAM in vivo tool compound: the discovery of VU6046980. Bioorg Med Chem Lett 80:129106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalinichev MRouillier MGirard FRoyer-Urios IBournique BFinn TCharvin DCampo BLe Poul EMutel V, et al. (2013) ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. J Pharmacol Exp Ther 344:624–636. [DOI] [PubMed] [Google Scholar]
- Kammermeier PJ (2015) Constitutive activity of metabotropic glutamate receptor 7. BMC Neurosci 16:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kammermeier PJ (2012) Functional and pharmacological characteristics of metabotropic glutamate receptors 2/4 heterodimers. Mol Pharmacol 82:438–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaneko M, Takahashi T (2004) Presynaptic mechanism underlying cAMP-dependent synaptic potentiation. J Neurosci 24:5202–5208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang M, Lee D, Song JM, Park S, Park DH, Lee S, Suh YH (2021) Neddylation is required for presynaptic clustering of mGlu7 and maturation of presynaptic terminals. Exp Mol Med 53:457–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kao H-T, Song HJ, Porton B, Ming GL, Hoh J, Abraham M, Czernik AJ, Pieribone VA, Poo MM, Greengard P (2002) A protein kinase A-dependent molecular switch in synapsins regulates neurite outgrowth. Nat Neurosci 5:431–437. [DOI] [PubMed] [Google Scholar]
- Kaur M, Sharma S (2022) Molecular mechanisms of cognitive impairment associated with stroke. Metab Brain Dis 37:279–287. [DOI] [PubMed] [Google Scholar]
- Kim Y-S, Kwak SM, Myung S-K (2015) Caffeine intake from coffee or tea and cognitive disorders: a meta-analysis of observational studies. Neuroepidemiology 44:51–63. [DOI] [PubMed] [Google Scholar]
- Kinoshita A, Shigemoto R, Ohishi H, van der Putten H, Mizuno N (1998) 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. J Comp Neurol 393:332–352. [PubMed] [Google Scholar]
- Klar R, Walker AG, Ghose D, Grueter BA, Engers DW, Hopkins CR, Lindsley CW, Xiang Z, Conn PJ, Niswender CM (2015) Activation of metabotropic glutamate receptor 7 is required for induction of long-term potentiation at SC-CA1 synapses in the hippocampus. J Neurosci 35:7600–7615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kniazeff J, Prézeau L, Rondard P, Pin J-P, Goudet C (2011) Dimers and beyond: the functional puzzles of class C GPCRs. Pharmacol Ther 130:9–25. [DOI] [PubMed] [Google Scholar]
- Koehl AHu HFeng DSun BZhang YRobertson MJChu MKobilka TSPardon ESteyaert J, et al. (2019) Structural insights into metabotropic glutamate receptor activation. Nature 566:79–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kukaj T, Sattler C, Zimmer T, Schmauder R, Benndorf K (2023) Kinetic fingerprinting of metabotropic glutamate receptors. Commun Biol 6:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi S, Jingami H, Morikawa K (2000) Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature 407:971–977. [DOI] [PubMed] [Google Scholar]
- Lee J, Gonzalez-Hernandez AJ, Kristt M, Abreu N, Roßmann K, Arefin A, Marx DC, Broichhagen J, Levitz J (2023) Distinct beta-arrestin coupling and intracellular trafficking of metabotropic glutamate receptor homo- and heterodimers. Sci Adv 9:eadi8076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J, Munguba H, Gutzeit VA, Singh DR, Kristt M, Dittman JS, Levitz J (2020) Defining the homo- and heterodimerization propensities of metabotropic glutamate receptors. Cell Rep 31:107605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee S, Park S, Lee H, Han S, Song JM, Han D, Suh YH (2019) Nedd4 E3 ligase and beta-arrestins regulate ubiquitination, trafficking, and stability of the mGlu7 receptor. eLife 8:e44502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levitz J, Habrian C, Bharill S, Fu Z, Vafabakhsh R, Isacoff EY (2016) Mechanism of assembly and cooperativity of homomeric and heteromeric metabotropic glutamate receptors. Neuron 92:143–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin XFisher NMDogra SSenter RKReed CWKalbfleisch JJLindsley CWAsher WBXiang ZNiswender CM, et al. (2022) Differential activity of mGlu7 allosteric modulators provides evidence for mGlu7/8 heterodimers at hippocampal Schaffer collateral-CA1 synapses. J Biol Chem 298:102458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linden A-M, Greene SJ, Bergeron M, Schoepp DD (2004) Anxiolytic activity of the MGLU2/3 receptor agonist LY354740 on the elevated plus maze is associated with the suppression of stress-induced c-Fos in the hippocampus and increases in c-Fos induction in several other stress-sensitive brain regions. Neuropsychopharmacology 29:502–513. [DOI] [PubMed] [Google Scholar]
- Liu JZhang ZMoreno-Delgado DDalton JARovira XTrapero AGoudet CLlebaria AGiraldo JYuan Q, et al. (2017) Allosteric control of an asymmetric transduction in a G protein-coupled receptor heterodimer. eLife 6:e26985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Zhang Y, Zhao D, Dong R, Yang X, Tammimies K, Uddin M, Scherer SW, Gai Z (2015) Rare de novo deletion of metabotropic glutamate receptor 7 (GRM7) gene in a patient with autism spectrum disorder. Am J Med Genet B Neuropsychiatr Genet 168B:258–264. [DOI] [PubMed] [Google Scholar]
- Maj MBruno VDragic ZYamamoto RBattaglia GInderbitzin WStoehr NStein TGasparini FVranesic I, et al. (2003) (-)-PHCCC, a positive allosteric modulator of mGluR4: characterization, mechanism of action, and neuroprotection. Neuropharmacology 45:895–906. [DOI] [PubMed] [Google Scholar]
- Marafi DMitani TIsikay SHertecant JAlmannai MManickam KAbou Jamra REl-Hattab AWRajah JFatih JM, et al. (2020) Biallelic GRM7 variants cause epilepsy, microcephaly, and cerebral atrophy. Ann Clin Transl Neurol 7:610–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marino MJWilliams DL JrO’Brien JAValenti OMcDonald TPClements MKWang RDiLella AGHess JFKinney GG, et al. (2003) Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson’s disease treatment. Proc Natl Acad Sci USA 100:13668–13673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martín R, Bartolomé-Martín D, Torres M, Sánchez-Prieto J (2011) Non-additive potentiation of glutamate release by phorbol esters and metabotropic mGlu7 receptor in cerebrocortical nerve terminals. J Neurochem 116:476–485. [DOI] [PubMed] [Google Scholar]
- Martín R, Durroux T, Ciruela F, Torres M, Pin J-P, Sánchez-Prieto J (2010) The metabotropic glutamate receptor mGlu7 activates phospholipase C, translocates munc-13-1 protein, and potentiates glutamate release at cerebrocortical nerve terminals. J Biol Chem 285:17907–17917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martín R, Torres M, Sánchez-Prieto J (2007) mGluR7 inhibits glutamate release through a PKC-independent decrease in the activity of P/Q-type Ca2+ channels and by diminishing cAMP in hippocampal nerve terminals. Eur J Neurosci 26:312–322. [DOI] [PubMed] [Google Scholar]
- Masugi M, Yokoi M, Shigemoto R, Muguruma K, Watanabe Y, Sansig G, van der Putten H, Nakanishi S (1999) Metabotropic glutamate receptor subtype 7 ablation causes deficit in fear response and conditioned taste aversion. J Neurosci 19:955–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsunaga H, Aruga J (2021) Trans-synaptic regulation of metabotropic glutamate receptors by Elfn proteins in health and disease. Front Neural Circuits 15:634875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCullock TW, Kammermeier PJ (2021) The evidence for and consequences of metabotropic glutamate receptor heterodimerization. Neuropharmacology 199:108801. [DOI] [PubMed] [Google Scholar]
- Meng JXu CLafon P-ARoux SMathieu MZhou RScholler PBlanc EBecker JAJLe Merrer J, et al. (2022) Nanobody-based sensors reveal a high proportion of mGlu heterodimers in the brain. Nat Chem Biol 18:894–903. [DOI] [PubMed] [Google Scholar]
- Millán C, Luján R, Shigemoto R, Sánchez-Prieto J (2002) The inhibition of glutamate release by metabotropic glutamate receptor 7 affects both [Ca2+]c and cAMP: evidence for a strong reduction of Ca2+ entry in single nerve terminals. J Biol Chem 277:14092–14101. [DOI] [PubMed] [Google Scholar]
- Millan MJAgid YBrüne MBullmore ETCarter CSClayton NSConnor RDavis SDeakin BDeRubeis RJ, et al. (2012) Cognitive dysfunction in psychiatric disorders: characteristics, causes and the quest for improved therapy. Nat Rev Drug Discov 11:141–168. [DOI] [PubMed] [Google Scholar]
- Mitsukawa KYamamoto ROfner SNozulak JPescott OLukic SStoehr NMombereau CKuhn RMcAllister KH, et al. (2005) A selective metabotropic glutamate receptor 7 agonist: activation of receptor signaling via an allosteric site modulates stress parameters in vivo. Proc Natl Acad Sci USA 102:18712–18717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monn JAValli MJMassey SMWright RASalhoff CRJohnson BGHowe TAlt CARhodes GARobey RL, et al. (1997) Design, synthesis, and pharmacological characterization of (+)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY354740): a potent, selective, and orally active group 2 metabotropic glutamate receptor agonist possessing anticonvulsant and anxiolytic properties. J Med Chem 40:528–537. [DOI] [PubMed] [Google Scholar]
- Moreno Delgado DMøller TCSter JGiraldo JMaurel DRovira XScholler PZwier JMPerroy JDurroux T, et al. (2017) Pharmacological evidence for a metabotropic glutamate receptor heterodimer in neuronal cells. eLife 6:e25233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagy G, Reim K, Matti U, Brose N, Binz T, Rettig J, Neher E, Sørensen JB (2004) Regulation of releasable vesicle pool sizes by protein kinase A-dependent phosphorylation of SNAP-25. Neuron 41:417–429. [DOI] [PubMed] [Google Scholar]
- Nakajima Y, Yamamoto T, Nakayama T, Nakanishi S (1999) A relationship between protein kinase C phosphorylation and calmodulin binding to the metabotropic glutamate receptor subtype 7. J Biol Chem 274:27573–27577. [DOI] [PubMed] [Google Scholar]
- Neul JLKaufmann WEGlaze DGChristodoulou JClarke AJBahi-Buisson NLeonard HBailey MESSchanen NCZappella M, et al. ; RettSearch Consortium (2010) Rett syndrome: revised diagnostic criteria and nomenclature. Ann Neurol 68:944–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niswender CM, Conn PJ (2010) Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol 50:295–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niswender CM, Johnson KA, Miller NR, Ayala JE, Luo Q, Williams R, Saleh S, Orton D, Weaver CD, Conn PJ (2010) Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Mol Pharmacol 77:459–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noroozi R, Taheri M, Omrani MD, Ghafouri-Fard S (2019) Glutamate receptor metabotropic 7 (GRM7) gene polymorphisms in mood disorders and attention deficit hyperactive disorder. Neurochem Int 129:104483. [DOI] [PubMed] [Google Scholar]
- O’Connor RM, Finger BC, Flor PJ, Cryan JF (2010) Metabotropic glutamate receptor 7: at the interface of cognition and emotion. Eur J Pharmacol 639:123–131. [DOI] [PubMed] [Google Scholar]
- O’Connor V, El Far O, Bofill-Cardona E, Nanoff C, Freissmuth M, Karschin A, Airas JM, Betz H, Boehm S (1999) Calmodulin dependence of presynaptic metabotropic glutamate receptor signaling. Science 286:1180–1184. [DOI] [PubMed] [Google Scholar]
- Okamoto N, Hori S, Akazawa C, Hayashi Y, Shigemoto R, Mizuno N, Nakanishi S (1994) Molecular characterization of a new metabotropic glutamate receptor mGluR7 coupled to inhibitory cyclic AMP signal transduction. J Biol Chem 269:1231–1236. [PubMed] [Google Scholar]
- O’Leary NAWright MWBrister JRCiufo SHaddad DMcVeigh RRajput BRobbertse BSmith-White BAko-Adjei D, et al. (2016) Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res 44(D1):D733–D745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levy R (1994) Aging-associated cognitive decline. Working Party of the International Psychogeriatric Association in collaboration with the World Health Organization. Int Psychogeriatr 6:63–68. [PubMed] [Google Scholar]
- Palazzo ERomano RLuongo LBoccella SDe Gregorio DGiordano MERossi FMarabese IScafuro MAde Novellis V, et al. (2015) MMPIP, an mGluR7-selective negative allosteric modulator, alleviates pain and normalizes affective and cognitive behavior in neuropathic mice. Pain 156:1060–1073. [DOI] [PubMed] [Google Scholar]
- Park DH, Park S, Song JM, Kang M, Lee S, Horak M, Suh YH (2020) N-linked glycosylation of the mGlu7 receptor regulates the forward trafficking and transsynaptic interaction with Elfn1. FASEB J 34:14977–14996. [DOI] [PubMed] [Google Scholar]
- Park SJung S-WKim B-NCho S-CShin M-SKim J-WYoo HJCho D-YChung U-SSon J-W, et al. (2013) Association between the GRM7 rs3792452 polymorphism and attention deficit hyperactivity disorder in a Korean sample. Behav Brain Funct 9:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park S, Kim B-N, Cho S-C, Kim J-W, Kim JI, Shin M-S, Yoo H-J, Han DH, Cheong JH (2014) The metabotropic glutamate receptor subtype 7 rs3792452 polymorphism is associated with the response to methylphenidate in children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 24:223–227. [DOI] [PubMed] [Google Scholar]
- Patzke C, Brockmann MM, Dai J, Gan KJ, Grauel MK, Fenske P, Liu Y, Acuna C, Rosenmund C, Südhof TC (2019) Neuromodulator signaling bidirectionally controls vesicle numbers in human synapses. Cell 179:498–513.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patzke C, Dai J, Brockmann MM, Sun Z, Fenske P, Rosenmund C, Südhof TC (2021) Cannabinoid receptor activation acutely increases synaptic vesicle numbers by activating synapsins in human synapses. Mol Psychiatry 26:6253–6268. [DOI] [PubMed] [Google Scholar]
- Pelkey KA, Lavezzari G, Racca C, Roche KW, McBain CJ (2005) mGluR7 is a metaplastic switch controlling bidirectional plasticity of feedforward inhibition. Neuron 46:89–102. [DOI] [PubMed] [Google Scholar]
- Pelkey KA, Topolnik L, Yuan X-Q, Lacaille J-C, McBain CJ (2008) State-dependent cAMP sensitivity of presynaptic function underlies metaplasticity in a hippocampal feedforward inhibitory circuit. Neuron 60:980–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perroy J, El Far O, Bertaso F, Pin JP, Betz H, Bockaert J, Fagni L (2002) PICK1 is required for the control of synaptic transmission by the metabotropic glutamate receptor 7. EMBO J 21:2990–2999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perroy J, Prezeau L, De Waard M, Shigemoto R, Bockaert J, Fagni L (2000) Selective blockade of P/Q-type calcium channels by the metabotropic glutamate receptor type 7 involves a phospholipase C pathway in neurons. J Neurosci 20:7896–7904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pfiffner LJ, Haack LM (2014) Behavior management for school-aged children with ADHD. Child Adolesc Psychiatr Clin N Am 23:731–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pin J-P, Galvez T, Prézeau L (2003) Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacol Ther 98:325–354. [DOI] [PubMed] [Google Scholar]
- Podkowa K, Rzeźniczek S, Marciniak M, Acher F, Pilc A, Pałucha-Poniewiera A (2015) A novel mGlu4 selective agonist LSP4-2022 increases behavioral despair in mouse models of antidepressant action. Neuropharmacology 97:338–345. [DOI] [PubMed] [Google Scholar]
- Polanczyk G, de Lima MS, Horta BL, Biederman J, Rohde LA (2007) The worldwide prevalence of ADHD: a systematic review and metaregression analysis. Am J Psychiatry 164:942–948. [DOI] [PubMed] [Google Scholar]
- Polishchuk A, Cilleros-Mañé V, Just-Borràs L, Balanyà-Segura M, Vandellòs Pont G, Silvera Simón C, Tomàs M, Garcia N, Tomàs J, Lanuza MA (2023) Synaptic retrograde regulation of the PKA-induced SNAP-25 and synapsin-1 phosphorylation. Cell Mol Biol Lett 28:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reed CW, Kalbfleisch JJ, Wong MJ, Washecheck JP, Hunter A, Rodriguez AL, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW (2020) Discovery of VU6027459: a first-in-class selective and CNS penetrant mGlu7 positive allosteric modulator tool compound. ACS Med Chem Lett 11:1773–1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reed CW, Washecheck JP, Quitlag MC, Jenkins MT, Rodriguez AL, Engers DW, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW (2019a) Surveying heterocycles as amide bioisosteres within a series of mGlu7 NAMs: discovery of VU6019278. Bioorg Med Chem Lett 29:1211–1214. [DOI] [PubMed] [Google Scholar]
- Reed CWYohn SEWashecheck JPRoenfanz HFQuitalig MCLuscombe VBJenkins MTRodriguez ALEngers DWBlobaum AL, et al. (2019b) Discovery of an orally bioavailable and central nervous system (CNS) penetrant mGlu7 negative allosteric modulator (NAM) in vivo tool compound: N-(2-(1 H-1,2,4-triazol-1-yl)-5-(trifluoromethoxy)phenyl)-4-(cyclopropylmethoxy)-3-methoxybenzamide (VU6012962). J Med Chem 62:1690–1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reuter MSTawamie HBuchert RHosny Gebril OFroukh TThiel CUebe SEkici ABKrumbiegel MZweier C, et al. (2017) Diagnostic yield and novel candidate genes by exome sequencing in 152 consanguineous families with neurodevelopmental disorders. JAMA Psychiatry 74:293–299. [DOI] [PubMed] [Google Scholar]
- Sansig GBushell TJClarke VRRozov ABurnashev NPortet CGasparini FSchmutz MKlebs KShigemoto R, et al. (2001) Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7. J Neurosci 21:8734–8745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saugstad JA, Segerson TP, Westbrook GL (1996) Metabotropic glutamate receptors activate G-protein-coupled inwardly rectifying potassium channels in Xenopus oocytes. J Neurosci 16:5979–5985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoepp DD (2001) Unveiling the functions of presynaptic metabotropic glutamate receptors in the central nervous system. J Pharmacol Exp Ther 299:12–20. [PubMed] [Google Scholar]
- Schoepp DD, Salhoff CR, Wright RA, Johnson BG, Burnett JP, Mayne NG, Belagaje R, Wu S, Monn JA (1996) The novel metabotropic glutamate receptor agonist 2R,4R-APDC potentiates stimulation of phosphoinositide hydrolysis in the rat hippocampus by 3,5-dihydroxyphenylglycine: evidence for a synergistic interaction between group 1 and group 2 receptors. Neuropharmacology 35:1661–1672. [DOI] [PubMed] [Google Scholar]
- Schulz HL, Stohr H, Weber BHF (2002) Characterization of three novel isoforms of the metabotrobic glutamate receptor 7 (GRM7). Neurosci Lett 326:37–40. [DOI] [PubMed] [Google Scholar]
- Sjöstedt EZhong WFagerberg LKarlsson MMitsios NAdori COksvold PEdfors FLimiszewska AHikmet F, et al. (2020) An atlas of the protein-coding genes in the human, pig, and mouse brain. Science 367:eaay5947. [DOI] [PubMed] [Google Scholar]
- Somogyi P, Dalezios Y, Luján R, Roberts JDB, Watanabe M, Shigemoto R (2003) High level of mGluR7 in the presynaptic active zones of select populations of GABAergic terminals innervating interneurons in the rat hippocampus. Eur J Neurosci 17:2503–2520. [DOI] [PubMed] [Google Scholar]
- Song J-M, Kang M, Park D-H, Park S, Lee S, Suh YH (2021) Pathogenic GRM7 mutations associated with neurodevelopmental disorders impair axon outgrowth and presynaptic terminal development. J Neurosci 41:2344–2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastava AK, Schwartz CE (2014) Intellectual disability and autism spectrum disorders: causal genes and molecular mechanisms. Neurosci Biobehav Rev 46:161–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stachniak TJ, Sylwestrak EL, Scheiffele P, Hall BJ, Ghosh A (2019) Elfn1-induced constitutive activation of mGluR7 determines frequency-dependent recruitment of somatostatin interneurons. J Neurosci 39:4461–4474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suh YH, Pelkey KA, Lavezzari G, Roche PA, Huganir RL, McBain CJ, Roche KW (2008) Corequirement of PICK1 binding and PKC phosphorylation for stable surface expression of the metabotropic glutamate receptor mGluR7. Neuron 58:736–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sukoff Rizzo SJLeonard SKGilbert ADollings PSmith DLZhang M-YDi LPlatt BJNeal SDwyer JM, et al. (2011) The metabotropic glutamate receptor 7 allosteric modulator AMN082: a monoaminergic agent in disguise? J Pharmacol Exp Ther 338:345–352. [DOI] [PubMed] [Google Scholar]
- Summa M, Di Prisco S, Grilli M, Usai C, Marchi M, Pittaluga A (2013) Presynaptic mGlu7 receptors control GABA release in mouse hippocampus. Neuropharmacology 66:215–224. [DOI] [PubMed] [Google Scholar]
- Suzuki G, Tsukamoto N, Fushiki H, Kawagishi A, Nakamura M, Kurihara H, Mitsuya M, Ohkubo M, Ohta H (2007) In vitro pharmacological characterization of novel isoxazolopyridone derivatives as allosteric metabotropic glutamate receptor 7 antagonists. J Pharmacol Exp Ther 323:147–156. [DOI] [PubMed] [Google Scholar]
- Tanabe Y, Masu M, Ishii T, Shigemoto R, Nakanishi S (1992) A family of metabotropic glutamate receptors. Neuron 8:169–179. [DOI] [PubMed] [Google Scholar]
- Tassin V, Girard B, Chotte A, Fontanaud P, Rigault D, Kalinichev M, Perroy J, Acher F, Fagni L, Bertaso F (2016) Phasic and tonic mGlu7 receptor activity modulates the thalamocortical network. Front Neural Circuits 10:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thibado JKTano J-YLee JSalas-Estrada LProvasi DStrauss AMarcelo Lamim Ribeiro JXiang GBroichhagen JFilizola M, et al. (2021) Differences in interactions between transmembrane domains tune the activation of metabotropic glutamate receptors. eLife 10:e67027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomioka NHYasuda HMiyamoto HHatayama MMorimura NMatsumoto YSuzuki TOdagawa MOdaka YSIwayama Y, et al. (2014) Elfn1 recruits presynaptic mGluR7 in trans and its loss results in seizures. Nat Commun 5:4501. [DOI] [PubMed] [Google Scholar]
- Trudeau LE, Emery DG, Haydon PG (1996) Direct modulation of the secretory machinery underlies PKA-dependent synaptic facilitation in hippocampal neurons. Neuron 17:789–797. [DOI] [PubMed] [Google Scholar]
- Uhlén MFagerberg LHallström BMLindskog COksvold PMardinoglu ASivertsson ÅKampf CSjöstedt EAsplund A, et al. (2015) Proteomics. tissue-based map of the human proteome. Science 347:1260419. [DOI] [PubMed] [Google Scholar]
- Wang X, Gao C, Zhang Y, Hu S, Qiao Y, Zhao Z, Gou L, Song J, Wang Q (2021) Overexpression of mGluR7 in the prefrontal cortex attenuates autistic behaviors in mice. Front Cell Neurosci 15:689611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang XWang MXu TFeng YShao QHan SChu XXu YLin SZhao Q, et al. (2023) Structural insights into dimerization and activation of the mGlu2-mGlu3 and mGlu2-mGlu4 heterodimers. Cell Res 33:762–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wierońska JM, Brański P, Siwek A, Dybala M, Nowak G, Pilc A (2010a) GABAergic dysfunction in mGlu7 receptor-deficient mice as reflected by decreased levels of glutamic acid decarboxylase 65 and 67kDa and increased reelin proteins in the hippocampus. Brain Res 1334:12–24. [DOI] [PubMed] [Google Scholar]
- Wierońska JM, Stachowicz K, Pałucha-Poniewiera A, Acher F, Brański P, Pilc A (2010b) Metabotropic glutamate receptor 4 novel agonist LSP1-2111 with anxiolytic, but not antidepressant-like activity, mediated by serotonergic and GABAergic systems. Neuropharmacology 59:627–634. [DOI] [PubMed] [Google Scholar]
- Xia W, Liu Y, Jiao J (2015) GRM7 regulates embryonic neurogenesis via CREB and YAP. Stem Cell Reports 4:795–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiang Z, Lv X, Lin X, O’Brien DE, Altman MK, Lindsley CW, Javitch JA, Niswender CM, Conn PJ (2021) Input-specific regulation of glutamatergic synaptic transmission in the medial prefrontal cortex by mGlu2/mGlu4 receptor heterodimers. Sci Signal 14:eabd2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yabuki Y, Shioda N, Maeda T, Hiraide S, Togashi H, Fukunaga K (2014) Aberrant CaMKII activity in the medial prefrontal cortex is associated with cognitive dysfunction in ADHD model rats. Brain Res 1557:90–100. [DOI] [PubMed] [Google Scholar]
- Yang Y, Pan C (2013) Role of metabotropic glutamate receptor 7 in autism spectrum disorders: a pilot study. Life Sci 92:149–153. [DOI] [PubMed] [Google Scholar]
- Yin S, Niswender CM (2014) Progress toward advanced understanding of metabotropic glutamate receptors: structure, signaling and therapeutic indications. Cell Signal 26:2284–2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C-S, Bertaso F, Eulenburg V, Lerner-Natoli M, Herin GA, Bauer L, Bockaert J, Fagni L, Betz H, Scheschonka A (2008) Knock-in mice lacking the PDZ-ligand motif of mGluR7a show impaired PKC-dependent autoinhibition of glutamate release, spatial working memory deficits, and increased susceptibility to pentylenetetrazol. J Neurosci 28:8604–8614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Q, Chen X, Li S, Yao T, Wu J (2021) Association between the group III metabotropic glutamate receptor gene polymorphisms and attention-deficit/hyperactivity disorder and functional exploration of risk loci. J Psychiatr Res 132:65–71. [DOI] [PubMed] [Google Scholar]