Open access peer-reviewed chapter

Metabotropic Glutamate Receptors in Anxiety Disorder

Written By

Jian Xu and Yongling Zhu

Submitted: 04 August 2023 Reviewed: 07 August 2023 Published: 09 October 2023

DOI: 10.5772/intechopen.1002630

From the Edited Volume

Anxiety and Anguish - Psychological Explorations and Anthropological Figures

Floriana Irtelli and Fabio Gabrielli

Chapter metrics overview

53 Chapter Downloads

View Full Metrics

Abstract

Anxiety disorders represent a prevalent group of mental health conditions characterized by patients experiencing excessive worry, fear, and distress. The neurobiological underpinnings of anxiety disorders are complex and involve multiple neurotransmitter systems. One such system is the glutamatergic system, which plays a critical role in anxiety regulation. Over the past few decades, much evidence has been gathered, substantiating the involvement of metabotropic glutamate receptors (mGluRs) in anxiety. Consequently, mGluRs have emerged as promising targets for treating anxiety disorders. This book chapter will provide an overview of the role of mGluRs in anxiety, focusing on their involvement in anxiety-related behaviors and their potential as therapeutic targets.

Keywords

  • anxiety disorder
  • depression
  • glutamate
  • metabotropic glutamate receptors (mGluRs)
  • ionotropic glutamate receptors (iGluRs)

1. Introduction

Anxiety disorders are among the most prevalent mental health conditions worldwide, affecting people of all ages. According to the World Health Organization (WHO), approximately 275 million people suffer from anxiety disorders globally. This staggering number reflects these conditions’ profound impact on individuals, families, and communities.

Although the causes of anxiety disorders are not yet fully understood, the condition is believed to arise from a complex interaction of genetic, biological, environmental, and psychological factors [1]. Converging lines of evidence from various branches of neuroscience indicate that anxiety disorders are frequently associated with imbalances in the brain’s neurotransmitter systems, including the glutamatergic system [2, 3, 4, 5, 6, 7]. Thus, understanding the involvement of the glutamatergic system in anxiety regulation can provide insights into potential therapeutic targets for treatment.

Glutamate is the most abundant excitatory neurotransmitter in the brain. It acts on two primary classes of glutamate receptors, ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). The iGluRs include three subfamilies: N-methyl-D-aspartate receptor (NMDA-Rs), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-Rs), and kainite receptors (KA-Rs). The mGluRs can be classified into group I, group II, and group III [8, 9, 10].

Among all glutamate receptors, mGluRs hold particular interest from a pharmacological standpoint for several reasons: (1) Diverse functions: Unlike iGluRs that primarily mediate fast synaptic transmission, mGluRs are involved in a wide range of cellular processes beyond simple neurotransmission. They can influence gene expression, intracellular signaling pathways, and various physiological responses, making them attractive targets for therapeutic intervention. (2) Broader therapeutic scope: Due to their involvement in numerous signaling pathways, targeting mGluRs presents promising opportunities for the development of drugs to address a wide range of conditions and neurological disorders. (3) Modulatory effects: mGluRs modulate synaptic transmission and neural circuitry in more nuanced and complex ways than iGluRs. This modulation allows for fine-tuning of neural activity, which could be beneficial in treating conditions where neural imbalances are involved. (4) Reduced risk of excitotoxicity: iGluRs can mediate excitotoxicity, a process that leads to neuronal damage and death due to excessive glutamate signaling. In contrast, mGluRs do not directly trigger such responses, reducing the risk of harmful side effects. (5) Drug specificity: Targeting mGluRs provides an opportunity to design drugs with better specificity, minimizing off-target effects and increasing therapeutic efficacy. Together, these reasons make mGluRs promising candidates for drug development, offering the potential to treat a wide range of neurological disorders, including anxiety and depression.

The objective of this chapter is to comprehensively examine the roles of mGluRs in anxiety disorders by reviewing the existing evidence. Additionally, we will summarize the findings from preclinical studies investigating the effects of targeting mGluRs for anxiety. Furthermore, this review will also assess results from clinical trials involving mGluR drugs for treating anxiety disorders.

Advertisement

2. Introduction of mGluRs

2.1 History of mGluRs’ discovery

In 1985, a groundbreaking discovery occurred when Sladeczek and coworkers demonstrated that glutamate possesses the capability to initiate the formation of molecules associated with a major second messenger system [11]. This finding unveiled the ability of glutamate to stimulate the production of inositol phosphates. Soon after, further evidence for the existence of mGluRs was discovered [12]. Built on these findings, Masu et al. successfully cloned the first mGluR, the mGluR1, in 1991 [13]. Subsequently, seven other mGluR subtypes were cloned by researchers. Together, a total of eight subtypes of mGluRs have been identified in the mammalian system [9]. Over the past three decades, tremendous strides have been made in comprehending the functions of these mGluRs.

2.2 Structure and function of mGluRs

mGluRs are a class of G-protein coupled receptors that bind glutamate. In contrast to iGluRs, mGluRs do not function as ion channels. Instead, their mode of operation involves initiating complex biochemical cascades. The eight subtypes of mGluRs exhibit distinct characteristics based on their sequence homology, signal transduction mechanisms, and pharmacological properties, leading to the categorization of mGluR subtypes into groups of groups I, II, and III mGluRs [14].

Structurally, all eight mGluRs contain an agonist-binding Venus fly trap (VFT) domain, which uses the cysteine-rich domain (CRDs) to connect to the highly conserved seven-pass trans-membrane domain (7TM) [15]. On cell membranes, mGluRs form obligate dimers. A recent structural study has revealed that when an agonist binds to these receptors’ VFT domain, it induces a compaction of the inter-subunit dimer interface. As a result, the CRDs come into close interactions, leading to the repositioning of the 7TM. This conformational change initiates the signaling process [16].

2.3 Insights from mGluR’s brain localization

Research on the localization of brain function highlights the association of specific brain regions with distinct functions. For example, anxiety has been linked to specific brain areas, including the hippocampus, prefrontal cortex, amygdala, bed nuclei of the stria terminalis, and hypothalamus [17, 18, 19]. Thus, when investigating the involvement of mGluRs in anxiety regulation and development, it becomes essential to examine mGluR’s precise expression patterns and localization within these brain regions. In this context, we have compiled mouse brain in situ hybridization data from the Allen Brain Institute and present these data in two figures, Figures 1 and 2. Please note that immunohistochemistry and in situ, mRNA hybridization have been extensively used to study the expression of mGluRs in the brain, and a wealth of literature is available [9] in addition to Allen Brain Atlas resources. Overall, analysis of mRNA in situ hybridization for different subtypes has led to several crucial observations:

  • Diverse expression patterns: The eight mGluRs exhibit distinct expression patterns in the brain, reflecting their varied functions.

  • Expression levels: The abundance of mGluRs varies significantly, with mGluR5 and mGluR4 being the most prevalent.

  • Anxiety-related brain regions: In line with their role in anxiety regulation, mGluR expression has been identified in various regions, including the hippocampus, prefrontal cortex, amygdala, bed nuclei of the stria terminalis, and hypothalamus. These brain areas are known to be associated with anxiety-related processes.

Figure 1.

In situ hybridization of mGluR1–8 in mouse brain sagittal slices. Images were exported from Allen brain institute (https://mouse.brain-map.org/). At the bottom is the brain atlas with arrows pointing to hippocampus and amygdala. The sagittal plane is about 3.0 mm from the midline. Scale Bar, 1200 μm.

Figure 2.

In situ hybridization of mGluR1–8 in mouse brain sagittal slices. Images were exported from Allen brain institute (https://mouse.brain-map.org/). At the bottom is the brain atlas with arrows pointing to hippocampus, hypothalamus, bed nucleus of the stria terminalis (BNST) and medial prefrontal cortex, and amygdala. The sagittal plane is about 0.5 mm from the midline. Scale Bar, 1200 μm.

While the understanding of brain localization of mGluRs in rodents is rather extensive, the localization of mGluRs in humans remains much less explored. However, novel imaging techniques are under active development to investigate the mGluRs in humans. These advancements in imaging studies offer promising avenues to examine the location and abundance of mGluRs in living individuals as well as in postmortem tissue [20, 21, 22]. Undoubtedly, future human studies will provide valuable insights into the brain localization of mGluRs, deepening our understanding of their potential roles in anxiety.

2.4 Subtypes of mGluRs

2.4.1 Group I mGluRs

Group I mGluRs consist of two subtypes: mGluR1 and mGluR5. Both mGluR5 and mGluR1 receptors are primarily located postsynaptically in the central nervous system (CNS). They play important roles in regulating synaptic transmission, neuronal excitability, and plasticity [9, 23, 24, 25, 26].

mGluR1 and mGluR5 display structural similarities and share common signaling mechanisms. Upon binding with glutamate, they both initiate the activation of the Gq/11 protein, subsequently leading to the activation of phospholipase C (PLC). PLC hydrolyzes phosphatidylinositol bisphosphate (PIP2) into inositol trisphosphate (IP3) and diacylglycerol (DAG). IP3 then promotes the release of calcium ions from intracellular stores, while DAG activates protein kinase C (PKC). These intracellular signaling pathways mediate the effects of group I mGluRs.

In the brain group I mGluRs are predominantly expressed in the hippocampus, cortex, and striatum, where they modulate synaptic transmission and synaptic plasticity. These receptors involve in many physiological processes, including motor activities, learning and memory, neuronal development, addiction, and emotion regulations [26, 27, 28, 29, 30, 31, 32]. Clinically, group I mGluRs have been associated with various neurological conditions, encompassing anxiety, depression, epilepsy, Parkinson’s disease, and fragile X syndrome. Consequently, these receptors have become potential targets for therapeutic interventions [33, 34, 35, 36].

2.4.2 Group II mGluRs

Group II mGluRs have two subtypes: mGluR2 and mGluR3 [37]. They are primarily located presynaptically in CNS where they act as autoreceptors [38]. Activation of mGluR2 or mGluR3 predominantly elicits an inhibitory response through a G-protein-coupled mechanism. When glutamate binds to group II mGluRs, it triggers the activation of G i/o proteins. The activated G i/o proteins inhibit adenylate cyclase (AC), leading to a decrease in cyclic AMP (cAMP) levels. The reduction in cAMP ultimately results in the inhibition of neurotransmitter release. Additionally, the dissociated beta-gamma subunits of the G-protein may modulate the activity of ion channels, leading to changes in membrane potential and ion flow across the cell membrane [39].

mGluR3 exhibits broad expression in CNS, whereas mGluR2 displays limited and overall low expression. It shows modest expression in the dentate gyrus and olfactory regions and weak expression in the thalamus, striatum, and cortex.

The functions of group II mGluRs have been associated with various physiological functions such as motor activities, learning and memory, emotion regulation, addiction, and neuroprotection [40, 41, 42, 43, 44, 45]. Group II mGluRs are also implicated in pathological conditions such as anxiety, depression, schizophrenia, pain, and neurodegenerative disorders [33, 46, 47].

2.4.3 Group III mGluRs

Group III mGluRs include four subtypes: mGluR4, mGluR6, mGluR7, and mGluR8. They are coupled to heteromeric Gi/Go protein. Activation of group III mGluRs also leads to the inhibition of adenylate cyclase [9].

Among the four group III mGluRs, mGluR6 is primarily located in the ON-bipolar cells of the retina [48]. In contrast, mGluR4, mGluR6, and mGluR7 are expressed in CNS with relatively widespread distributions. Specifically, mGluR4 is found in most brain regions, displaying the highest intensity in the cerebellum and moderate levels in the cortex, striatum, amygdala, and hippocampus. On the other hand, mGluR7 exhibits high expression in the hippocampus, and it also demonstrates a relatively strong presence in the amygdala and striatum. Although mGluR8 was initially identified in the retina [49], it is also expressed in the CNS, particularly in regions like the cerebellum, olfactory bulb, cortex, and hippocampus. However, the expression levels of mGluR8 in these CNS regions are lower than that of mGluR4.

Like group II mGluRs, group III mGluRs are also primarily located in the presynaptic terminals of neurons in the CNS [50], where they act as autoreceptors, responding to the release of glutamate from the same neuron to regulate neurotransmitter release. Group III mGluRs can also be found on postsynaptic neurons, which modulate postsynaptic responses to neurotransmitter signaling [51].

mGluR4 and mGluR8 have a much higher affinity to glutamate than mGluR7, which shows a low affinity for glutamate and is activated only by high glutamate concentrations [52, 53]. Overall, these versatile receptors play crucial roles in a wide range of physiological processes, including motor functions, learning and memory, fear extinction, anxiety, social behaviors, and epilepsy [54, 55, 56, 57, 58, 59, 60]. Furthermore, their implications extend to human neurological conditions, including epilepsy, anxiety disorders, depression, pain modulation, and addiction [33, 61, 62].

Advertisement

3. Preclinical studies for the roles of mGluRs in anxiety

3.1 History

Shortly after discovering mGluR genes in the 1990s, researchers began exploring the functions of mGluR subtypes at glutamatergic synapses and the possible roles of mGluRs in neurological disorders, including anxiety. Numerous preclinical studies have been conducted to examine the effects of different mGluR compounds in animal models of anxiety. Encouragingly, a considerable body of evidence has quickly emerged and confirmed that some mGluR compounds demonstrate anxiolytic and antidepressant-like properties in animal models. Particularly, studies in rodents have shown that antagonists of group I mGluRs, and agonists of group II mGluRs can act as anxiolytics and antidepressants [63, 64, 65, 66, 67, 68, 69]. These findings have laid a solid foundation for further research in this promising field.

Besides the pharmacological approach, mGluR mutant animal models have also been developed and tested extensively to explore mGluRs’ role in anxiety. Furthermore, through electrophysiological, cellular, and biochemical studies, optogenetics and chemogenetics, valuable insights have been gained into the specific modulation of synaptic transmission, intrinsic excitability, and synaptic plasticity of mGluRs in brain regions that govern emotions and anxiety, such as the amygdala, hippocampus, and prefrontal cortex [19, 70, 71].

Together, these collective data from preclinical research have significantly deepened our understanding of anxiety disorders’ molecular and neural foundations. Furthermore, these studies offer promising therapeutic avenues by targeting mGluRs for potential anxiety treatments.

3.2 Roles of group I mGluRs in anxiety

Group I mGluRs have long been implicated in regulating anxiety and anxiety disorders. These receptors are primarily expressed in brain regions associated with emotional processing, including the amygdala, prefrontal cortex, and hippocampus.

Numerous animal studies investigating drugs targeting group I mGluRs and their effects on anxiety-like behaviors have been conducted, yielding consistent findings. Overall, antagonistic treatment has shown significant anxiolytic responses in experimental animals, indicating their potential as therapeutic agents for anxiety-related disorders. These compounds have demonstrated a notable capacity to lower anxiety levels in preclinical models. This is supported by their ability to reduce fear-conditioned freezing, increase the time spent in the center of the open field, and decrease marble-burying behavior, among other positive indicators. However, it is important to point out that the anxiolytic effects of group I mGluR compounds may vary based on their specific brain region activation. Activation of mGluR1 or mGluR5 in particular brain regions, such as the hippocampus, amygdala, and the prefrontal cortex, may be particularly relevant to anxiety regulation. Also, the effects of group I mGluR compounds on anxiety may exhibit dose-dependent responses, with different outcomes observed at varying concentrations.

Owing to their significance and relevance, a plethora of exclusive reviews are available on animal studies focusing on drugs targeting group I mGluRs. Consequently, readers can refer to these articles to delve into comprehensive details regarding drug studies. Here are a few illustrative examples. Swanson et al. reviewed in 2005 animal studies on drugs targeting mGluRs on anxiety-like behaviors [72]. Krystal et al. reviewed in 2010 the preclinical animal studies that examined mGluR agonists and antagonists in rodent models of anxiety [73]. Of the studies examined in this 2010 review, about 90% of them reported an anxiolytic effect with mGluR5 antagonists. Riaza Bermudo-Soriano et al. provided another review [3]. In this comprehensive review conducted in 2012, the authors examined the effects of mGluR5 antagonists on anxiety through 43 animal studies. Remarkably, all but two of these studies revealed anxiolytic effects, indicating a strong potential for mGluR5 antagonists in anxiety treatment. Additionally, the authors assessed 20 animal studies involving mGluR1 antagonists and their impact on anxiety. Among these studies, an encouraging 13 of them demonstrated anxiolytic effects, further highlighting the promising therapeutic role of mGluR1 antagonists in addressing anxiety-related conditions.

3.3 Roles of group II mGluRs in anxiety

mGluR3 exhibits broad expression in many brain regions, including those known to be involved in emotional processing, such as the amygdala, prefrontal cortex, hippocampus, and bed nucleus of the stria terminalis. In contrast, mGluR2 shows a more limited expression pattern, with moderately strong presence in the dentate gyrus and olfactory regions and weaker expression in the thalamus, striatum, and cortex.

Group II mGluRs inhibit the release of glutamate, and by reducing excessive glutamate release, these receptors can help regulate neuronal activity and maintain a balance in the brain’s excitatory signaling, which may contribute to anxiety reduction. Indeed, many animal studies on drugs targeting the group II mGluRs on anxiety-like behaviors have been conducted, and rather consistent findings have been reported.

Overall, agonists and positive allosteric modulators of mGluR2 and/or mGluR3 receptors have been found to elicit anxiolytic responses in experimental animals. For example, mGluR2/3 agonists have been shown to reduce fear-potentiated startle, decrease stress-induced hyperthermia, and increase open-arm entries in the elevated plus maze [43, 74, 75]. Other studies showed that pharmacological activation of mGlu2/3 receptors shortens the time that was required for the conventional antidepressants to be effective as antidepressants in these rats, proposing the combination of mGluR2/3 agonists with other antianxiety agents as a potential treatment for anxiety [76, 77]. In line with the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist, Nasca et al. showed that L-acetylcarnitine causes rapid antidepressant effects through the epigenetic induction of mGlu2 receptors [78].

While it was somewhat expected that mGluR2/3 agonists might act as antianxiety agents, as mentioned earlier, some later studies brought about surprising findings. These studies revealed that negative allosteric modulators targeting mGluR2/3 also exhibited antidepressant and anxiolytic activity in rodents. The anxiolytic effect was demonstrated in various behavioral paradigms, including the learned helplessness (LH) paradigm [79], marble-burying, and forced swim test (FST) [80]. Therefore, these results suggest that the blockade mGluR2/3 may also hold promise as a treatment for depressive and anxiety disorders. In the 2012 review by Riaza Bermudo-Soriano et al., nine studies with mGluR2/3 antagonists were listed, with six demonstrated anxiolytic effects. Meanwhile, 28 studies with mGluR2/3 agonists were recorded, all but three demonstrated anxiolytic effects [3].

As such, the anxiolytic effects observed with the blockade or activation of mGluR2/3 have led to some conflicting findings. While the exact reasons for these discrepancies remain not fully understood, various factors, such as differences in behavioral assays, routes of drug administration, and dosages, may play a significant role in the observed outcomes. Therefore, it is imperative to conduct further research and engage in in-depth discussions to fully elucidate the potential of mGluR2/3 agonists or antagonists in anxiety treatment.

3.4 Roles of group III mGluRs in anxiety

The expression patterns of group III mGluRs indicate that they might also play significant roles in anxiety regulation. Notably, mGluR4, mGluR7, and mGluR8 are all present in the hippocampus, and there is a possibility of mGluR6 having low expression in this region as well. Additionally, these receptors, mGluR4, mGluR7, and mGluR8, are found in other crucial areas such as the hypothalamus, prefrontal cortex, and amygdala. Given their presence in these regions, the activation of these receptors could potentially influence synaptic transmission and impact anxiety-related processes.

While the specific roles of each subtype of group III mGluRs in anxiety are still an active area of research, evidence also suggests their involvement in anxiety regulation. Group III mGluR ligands have been comparatively less studied for their efficacy in anxiety disorders when compared to group I and II ligands; nevertheless, a substantial number of studies have been conducted thus far. Notably, the administration of group III mGluR agonists has demonstrated anxiolytic-like and antidepressant-like effects in experimental animal models. For example, Systemic administration of mGluR8 receptor agonist (S)-3,4-DCPG induces c-fos in stress-related brain regions in wild-type but not mGluR8 receptor knockout mice, suggesting that mGluR8 receptors are involved in anxiety regulation [81]. Several studies have demonstrated that the administration of group III mGluR agonists results in anxiolytic-like and antidepressant-like effects in behavioral tests [82, 83, 84]. In a separate study, mGluR4 PAM exhibited an anxiolytic effect but did not produce an antidepressant-like effect [85]. Recently, mGluR7 specific agonist was also found to be able to produce anxiolytic effects [86]. Compared to mGluR4, mGluR7, and mGluR8, the role of mGluR6 in anxiety remains uncertain. This uncertainty arises from its predominantly expressed location in the retina, with very low expression in the CNS. However, when tested in rats, pharmacological activation of mGluR6 in vivo using a selective agonist produced some anxiolytic-like effects, suggesting mGluR6 might also play a role in anxiety-related processes [82].

In summary, research indicates that among the four group III mGluRs, mGluR4, mGluR7, and mGluR8 are all associated with anxiety behaviors. Additionally, mGluR6 might also play a role in anxiety, though its involvement requires further investigation. Despite the progress over the years, the specific roles of the group III mGluR subtypes in anxiety remain largely unknown, emphasizing the need for further research to elucidate their exact functions and significance in anxiety behaviors.

3.5 mGluR animal models in anxiety

Alongside the pharmacological approach, researchers have extensively developed and tested mGluR mutant animal models to explore the roles of these receptors in anxiety. Constitutive knockouts for all mGluR genes have been generated [26, 42, 45, 56, 59, 87, 88, 89], and conditional knockout mice for specific mGluR genes have also been created [28, 34, 90]. Using mutant animals provides a significant advantage in terms of subtype precision, allowing researchers to target specific mGluR subtypes, which can be challenging to achieve with pharmacological compounds. However, it is crucial to acknowledge the limitations of knockout studies using mutant animals, such as potential gene compensation issues. Despite these challenges, using mutant animal models remains a valuable tool in advancing our understanding of mGluRs’ involvement in anxiety and related processes. Table 1 presents the findings from studies involving mGluR mutant mice.

GeneModelTestBehavior phenotypesAnxietyRef
Grm5koOFTGrm5 ko mice spent more time in the middle of the arena compared to the control.[27, 91, 92]
Grm5koMBTMarble burying is abolished in Grm5 ko mice.[27]
Grm5koEZMNo change in the time animals spent exploring the open area in Grm5 ko mice.[27]
Grm5koFEFear extinction is impaired in Grm5 ko mice.[28]
Grm5koEZM, DLBGrm5 ko mice showed increased anxiety accentuating with aging.[93]
Grm1koPPIGrm1 ko mice exhibited a significant PPI deficit despite their smaller body size and abnormal gait. Note: Evidence exists for the coupling of reduced PPI and certain anxiety disorders.[32]
Grm2koEPM, OFT, BWAno consistent effect on anxiety in Grm2 ko mice.[94]
Grm3koEPM, OFT
BWA
no consistent effect on anxiety Grm3 ko mice.[94]
Grm3 and Grm2double koEPM, OFT
BWA
No consistent effect on anxiety in Grm2/Grm3 double ko mice.[94]
Grm2koGrm2 ko and Grm3 ko mice were grossly normal. However, the anxiolytic-like activity of LY354740 (20 mg/kg, s.c.) was not evident in either Grm2 or Grm3 ko mice.[43]
Grm3koDLB, EPM, OFTNo difference on anxiety[40]
Grm4koOFT, EZMmiddle-aged Grm4 ko male mice showed increased measures of anxiety in the open field and elevated zero maze.[95]
Grm4koOFT, EZMNo changes in adult 6-month-old Grm4 male ko mice.[95]
Grm4koOFT, EZMfemale Grm4 ko mice showed reduced measures of anxiety.[95]
Grm6konot available[88]
Grm7koOFTGrm7 ko mice spent significantly more time exploring the open arms of the maze.[60]
Grm7koLDB, EPM, staircase test, SIHGrm7 ko mice displayed anxiolytic activity in four different behavioral tests, i.e., the light-dark box, the elevated plus maze, the staircase test, and the stress-induced hyperthermia test,[96]
Grm8koOF,EPMGrm8 ko mice showed increased measures of anxiety in the open field and the elevated plus maze, and an increased acoustic startle response were seen in 6- and 12-month-old Grm8 ko male mice.[58]

Table 1.

Anxiety-related behaviors in Grm1–8 ko mice.

Abbreviations: OFT: open field test, EZM: Elevated zero maze, DLB: Dark–Light Box test, FPS: Fear-potentiated startle, VC: Vogel conflict test, BWA: The Black & White Alley, SIH: stress-induced hyperthermia test.

Note: in mice, mGluR1–8 are encoded by Grm1–8 genes respectively.

As summarized above, Grm1 ko mice exhibit significant impairments in movement and gait [87], which pose challenges in interpreting behavioral measurements. Despite that, the Grm1 ko mice displayed a significant prepulse inhibition (PPI) deficit [32]. It is noteworthy that reduced PPI has been observed in patients with certain anxiety disorders [97, 98]. On the other hand, there is some disagreement in the effects of Grm5 ko mice when compared to the impact of mGluR5 agonists. Considering the consistent and robust anxiolytic effects of mGluR5 antagonists, it was expected for the Grm5 ko mice to display reduced anxiety. Indeed, in the open field test (OFT) and Marble-Burying Test (MBT), the Grm5 ko mice showed less anxiety. However, Grm5 ko mice also exhibited severe impairments in fear extinction [28], widely considered a key mechanism in posttraumatic stress disorder (PTSD). This finding suggests that an agonist of mGluR5, instead of an antagonist, may be helpful as a treatment option to treat severe anxiety in people who have experienced traumatic events [99, 100].

Despite the clear anxiolytic effects observed in mGlu2/3 ligands, the lack of an anxiety phenotype in Grm2 ko and Grm3 ko mice, as well as the double ko mice, was unexpected. But the anxiolytic-like activity of LY354740 in wild-type mice was not evident in either Grm2 or Grm3 ko mice, suggesting that anxiolytic-like activity was indeed meditated by mGluR2 and mGluR3 [43]. The exact reason for the unaltered anxiety in these knockout mice remains unknown. One possible explanation could be the presence of redundancy and compensation mechanisms. However, even in the Grm2/Grm3 double ko mice, the anxiety-like behaviors were not significantly affected, which suggests that redundancy and compensation alone may not fully account for the observation. Another plausible explanation could be that Grm2 and Grm3 ko mice may experience developmental changes, which could influence the outcome. Future testing with conditional knockout mice may prove helpful to further investigate and resolve this discrepancy.

The behavioral effects of knocking out genes for group III mGluRs, Grm4, Grm6, Grm7, and Grm8, were also quite intricate. In the case of Grm4 ko mice, the consequences of gene deletion were discovered to be dependent on age and gender [95]. Interestingly, Grm7 ko mice exhibited reduced anxiety [60, 96], while Grm8 ko mice displayed increased anxiety [58]. These outcomes do not entirely align with the pharmacological results, where the administration of group III mGluR agonists generally produces anxiolytic effects. As a result, further studies are necessary to address and clarify these discrepancies.

Advertisement

4. Human studies linking mGluRs in anxiety disorders

4.1 History

In 1982, a clinical study was published demonstrating the potential efficacy of fenobam in treating anxiety [101]. At that time, the specific target of fenobam was not yet known, and it was not until 20 years later that Porter et al. [102] discovered that fenobam actually acted as a selective and potent mGluR5 receptor antagonist. As a result, the 1982 study [101] provided the initial evidence for the involvement of mGluRs in anxiety, marking an essential milestone in comprehending the roles of these receptors in anxiety disorders. Since the 1982 report, numerous additional studies have been conducted, encompassing both animal studies and preclinical and clinical investigations. Collectively, these studies have overwhelmingly supported the roles of mGluRs in anxiety. The expanding body of evidence reinforce the initial findings and solidify our understanding of mGluR’s importance in anxiety disorders [33, 35, 73, 103, 104].

It is worth noting that mGluRs have been implicated in obsessive-compulsive disorder (OCD) and posttraumatic stress disorder (PTSD), both of which were previously classified as anxiety disorders before the introduction of the fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5). Additionally, while this chapter focuses on mGluR in anxiety disorder, it is important to highlight that mGluRs are also implicated in depression disorders. Although anxiety and depression are distinct disorders with unique characteristics, they often co-occur and share some similarities in symptoms and treatment approaches. Many individuals experience symptoms of both conditions simultaneously, leading to what is known as comorbid anxiety and depression. Therefore, this section also includes specific clinical evidence that implicates mGluRs in depression.

4.2 Findings from human genetic studies

Genetic linkage and association studies have been extensively conducted to identify chromosomal risk loci and susceptibility genes for anxiety and depression [105]. Among the findings, there is a multiple of evidence supporting the roles of GRM genes in anxiety (see Table 2 for details). Particularly, at least four studies have implicated either GRM7 or GRM8 in depression disorder [106, 107, 108, 109], giving strong support for the involvement of group III mGluRs in depression. Furthermore, genetic studies have strongly indicated the involvement of GRM5 in autism spectrum disorder (ASD) [110, 111, 112, 113]. Although ASD and anxiety are distinct conditions, it is worth noting that many ASD patients also experience anxiety disorder. As a result, these human genetic studies may offer insights into potential shared neural mechanisms underlying depression, anxiety, and even ASD, despite their different diagnostic classification.

StudiesFindingsReference
GRM7Linkage studiesThere is a genome-wide significant linkage to chromosome 3p26-3p25, with a peak signal near the gene GRM7 in depression.[106, 107]
GRM8GSAAThe glutamatergic synaptic transmission gene set (GO:0035249) includes 16 genes, of which six genes, SLC1A4, CACNA1A, GRM8, PARK2, UNC13A, and SHC3, showed nominal association with MDD.[108]
GRM8GWASA plausible biological association was found with SNPs within GRM8 in depression.[109]
GRM5WES, GWASHigh throughput sequencing has identified de novo GRM5 mutations and enrichment of rare variants of genes encoding components of mGluR signaling pathways. Increase in the prevalence GRM5 copy number variants in ASD was reported.[110, 111, 112, 113]

Table 2.

Evidence from genetic studies implicating GRM genes in anxiety.

Abbreviations: GSAA: gene-set-based association analysis, GWAS: genome-wide association scans, WES: whole-exome sequencing, ASD: autism spectrum disorder, MDD: major depression disorder, SNPs: single nucleotide polymorphisms.

4.3 Findings from emission tomography (PET)

PET has emerged as a valuable tool in diverse neurologic and psychiatric applications. Particularly, over the last two decades, significant advancements have been made in developing mGluR PET ligands, leading to an increasing number of PET radioligands that target mGluRs. These innovative ligands provide a noninvasive in vivo imaging technique, enabling the quantification of mGluR receptors in normal and disease-state conditions [7, 114].

For group I mGluRs, the pioneer tracer for both preclinical and clinical applications, [11C]ABP688, was developed in 2006 [115]. Since then, several more radioligands have been introduced, including [18F]FIMX for mGluR1 [116], [18F]FPEB [117], and [18F]SP203 [118] for mGluR5. These radioligands have been employed in clinical trials and have played a pivotal role in investigating the involvement of mGluRs in anxiety disorders. Among these ligands, mGluR5 has been the most extensively studied. The critical findings for these studies are summarized in Table 3. Interestingly, it appears that mGluR5 expression can vary significantly, showing either upregulation or downregulation depending on the specific disorder. For instance, individuals with MDD may exhibit lower levels of mGluR5, while those with PTSD may experience upregulation of this receptor (Table 3). As a result, when contemplating pharmacological interventions targeting mGluRs, it becomes crucial to meticulously justify their usage based on how the receptors are altered in different medical conditions.

ReceptorsLigandsDisordersFindingsmGluR5Ref
mGluR5[18F]FPEBMDDNo significant between-group differences were observed. Individuals with MDD had higher ACC glutamate, Importantly, the ACC mGluR5 DVR negatively correlated with glutamate.[22]
mGluR5[11C]ABP688MDDmGluR5 density reduced in the amygdala and prefrontal cortex in MDD.[21]
mGluR5[18F]FPEBPTSDThere is significantly higher mGluR5 availability in individuals with PTSD relative to matched controls across many brain regions.[119]
mGluR5[18F]FPEBSuicidal ideationThere is higher availability of mGluR5 in individuals with PTSD than healthy control and MDD groups. Furthermore, higher mGluR5 availability was associated with scan-day suicidal ideation among individuals with PTSD, but not MDD.[120]
mGluR5[11C]ABP688OCDThere is higher mGluR5 availability in OCD patients.[121]
mGluR5[11C]ABP688MDDNo significant difference in mGluR5 availability was observed between elderly subjects with MDD and healthy volunteers.[122]

Table 3.

Imaging studies of mGluRs in human.

Abbreviations: MDD: major depressive disorder, PTSD: posttraumatic stress disorder, OCD: obsessive-compulsive disorder, ACC: anterior cingulate, DVR: distribution volume ratio.

As of now, some PET ligands have also been developed for group II mGluRs, specifically targeting mGluR2 and mGluR3, with [11C]JNJ42491293 [123] being a notable ligand that has been used in clinical studies to probe mGluR2 in the human brain. On the other hand, the availability of group III PET ligands remains limited [124]. Although several ligands have been developed for PET imaging purposes, there is currently a lack of reports on their usage in human patients.

Advertisement

5. Clinical studies

The utilization of pharmacological modulation of glutamate transmission has long been regarded as a highly valuable therapeutic approach [3, 125, 126]. The mGluRs have emerged as potential targets for safely altering glutamate-driven excitation. Preclinical data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders [3, 72, 73, 127]. Numerous clinical trials have been conducted to explore the potential of targeting mGluRs for treating various neurological diseases. A recent review comprehensively summarized the findings from these clinical studies involving compounds that specifically interact with mGluRs [104].

Anxiety disorders are the most common mental disorders, affecting about 30% of the population at some point in life. Given the involvement of mGluRs in anxiety regulation, there has been tremendous interest in developing mGluR drugs for therapeutic use in anxiety disorders. Indeed, a number of mGluR antagonists or agonists have been used in clinical studies of anxiety disorder. These data are summarized in Table 4.

CompoundsReceptorStudyFindingsReference
FenobammGluR5 antagonistAnxiety disorderApproved by FDA for anxiety treatment.[101]
BasimglurantmGluR5 antagonistMDDThe primary endpoint (mean change in clinician-rated MADRS score from baseline to end of treatment) was not met.[128]
MavoglurantmGluR5 antagonistOCD patients that are unresponsive to SSRI therapyThis study of mavoglurant in OCD was terminated because of the lack of efficacy in the interim analysis.[129]
LY354740mGlu2/3 agonistPanic disorderLY354740 failed to show treatment effects that were different from placebo.[130]
LY544344mGlu2/3 agonistsPhase 2 GADImprovements in HA and CGI. However, the trial was discontinued early based on findings of convulsions in preclinical studies.[131]
JNJ-40411813mGlu2 agonistPhase 2a study in MDD patients with significant anxiety symptomsNo efficacy signal was detected on the primary endpoint, the 6-item Hamilton Anxiety Subscale.[132]
Pomaglumetad methionilmGlu2/3 agonistPTSD Fear-potentiated startleResult not disclosed.ClinicalTrials.gov Identifier: NCT02234687
DecoglurantmGlu2/3 antagonistMajor depressive disorder patients taking SSRI or SNRI antidepressantsNo significant separation from placebo on depression or cognition endpoints (high placebo response rate).[133]

Table 4.

Clinical studies on the effects of mGluR compounds on anxiety disorders and related conditions.

Abbreviations: HA and CGI: Hamilton Anxiety and Clinical Global Impression, MDD: major depressive disorder, OCD: obsessive-compulsive disorder, GAD: generalized anxiety disorder, HDR: Hamilton Depression Rating, MADRS: Montgomery-Asberg Depression Rating Scale, SSRI: selective serotonin reuptake inhibitors, SNRIs: serotonin and norepinephrine reuptake Inhibitors.

Currently, the only FDA-approved medication targeting mGluRs is fenobam, which initially received approval as an anxiolytic [101] before its characterization as a mGluR5 receptor antagonist [102]. Despite numerous clinical trials exploring mGluR-targeting compounds for treating anxiety, OCD, depression, and panic disorder, the results have not been as promising as anticipated. Notably, three studies targeting group II mGluRs [130, 131, 132] and two studies targeting mGluR5 [128, 129] did not yield the robust outcomes desired. The development of group III mGluRs as potential therapeutic targets has been relatively limited compared to other mGluR receptors. As of now, there have been no reports of human clinical trials involving group III compounds.

Rest assured, laboratories’ dedication to developing novel mGluR drugs will persist, and preclinical research will continue to advance our understanding of mGluR functions. Despite encountering various challenges, clinical inquiry into mGluRs will not cease. There remains a hopeful outlook that effective treatments can be developed based on the functions of mGluRs. With ongoing efforts and scientific exploration, we can aspire to find new therapeutic approaches for anxiety disorders.

Advertisement

6. Conclusion

This book chapter offers a comprehensive investigation into the roles of mGluRs in anxiety disorders. Through an exploration of their classification, neurobiological mechanisms, and potential therapeutic implications, the aim is to enhance our understanding of these receptors as potential targets for developing innovative treatments for anxiety disorders. The chapter begins with an exploration of the biology of mGluRs. It then transitions to an investigation of how mGluRs influence anxiety-related behaviors in animals, utilizing animal models as a foundation to understand the neurobiological mechanisms underlying the actions of mGluRs. In the subsequent section, the chapter delves into the clinical implications and therapeutic potential of mGluRs in anxiety disorders.

Preclinical data strongly supports the potential of mGluRs as promising therapeutic targets for anxiety disorders. A wealth of evidence demonstrates that certain mGluR compounds exhibit high efficacy as anxiolytic agents in animal models. However, from a mechanistic standpoint, many important questions remain unanswered, such as the specific roles of individual receptors and the underlying cellular mechanisms and neural circuits through which these receptors ultimately influence anxiety. Further research is needed to address these aspects and fully harness the therapeutic benefits of targeting mGluRs for anxiety disorders.

Unfortunately, despite initial expectations, the clinical studies on mGluR receptor ligands as anxiolytics have yielded somewhat disappointing results. However, we should approach this situation with cautious optimism. It is crucial to recognize that these ligands have been tested only in a limited range of anxiety disorders, and their full therapeutic potential remains yet to be defined. There is hope that through further exploration and broader clinical trials, more promising outcomes for these ligands may be revealed. Furthermore, ongoing advancements in developing new compounds with improved pharmacokinetic and safety profiles offer great potential for enhanced efficacy and better tolerability. This progress may ultimately pave the way for more effective mGluR-based treatments, providing renewed possibilities for individuals seeking relief from anxiety disorders.

References

  1. 1. Szuhany KL, Simon NM. Anxiety disorders: A review. JAMA. 2022;328(24):2431-2445
  2. 2. Nasir M et al. Glutamate systems in DSM-5 anxiety disorders: Their role and a review of glutamate and GABA psychopharmacology. Frontiers in Psychiatry. 2020;11:548505
  3. 3. Riaza Bermudo-Soriano C et al. New perspectives in glutamate and anxiety. Pharmacology, Biochemistry, and Behavior. 2012;100(4):752-774
  4. 4. Sanacora G et al. Targeting the glutamatergic system to develop novel, improved therapeutics for mood disorders. Nature Reviews. Drug Discovery. 2008;7(5):426-437
  5. 5. Altamura CA et al. Plasma and platelet excitatory amino acids in psychiatric disorders. The American Journal of Psychiatry. 1993;150(11):1731-1733
  6. 6. Frye MA et al. Low cerebrospinal fluid glutamate and glycine in refractory affective disorder. Biological Psychiatry. 2007;61(2):162-166
  7. 7. Kim JH et al. A review of molecular imaging of glutamate receptors. Molecules. 2020;25(20)
  8. 8. Traynelis SF et al. Glutamate receptor ion channels: Structure, regulation, and function. Pharmacological Reviews. 2010;62(3):405-496
  9. 9. Niswender CM, Conn PJ. Metabotropic glutamate receptors: Physiology, pharmacology, and disease. Annual Review of Pharmacology and Toxicology. 2010;50:295-322
  10. 10. Gereau RW, Swanson G. The Glutamate Receptors. The Receptors. Totowa, N.J: Humana Press. xi; 2008. p. 576
  11. 11. Sladeczek F et al. Glutamate stimulates inositol phosphate formation in striatal neurones. Nature. 1985;317(6039):717-719
  12. 12. Sugiyama H, Ito I, Hirono C. A new type of glutamate receptor linked to inositol phospholipid metabolism. Nature. 1987;325(6104):531-533
  13. 13. Masu M et al. Sequence and expression of a metabotropic glutamate receptor. Nature. 1991;349(6312):760-765
  14. 14. Conn PJ, Pin JP. Pharmacology and functions of metabotropic glutamate receptors. Annual Review of Pharmacology and Toxicology. 1997;37:205-237
  15. 15. Pin JP, Bettler B. Organization and functions of mGlu and GABA(B) receptor complexes. Nature. 2016;540(7631):60-68
  16. 16. Koehl A et al. Structural insights into the activation of metabotropic glutamate receptors. Nature. 2019;566(7742):79-84
  17. 17. Martin EI et al. The neurobiology of anxiety disorders: Brain imaging, genetics, and psychoneuroendocrinology. The Psychiatric Clinics of North America. 2009;32(3):549-575
  18. 18. Adhikari A. Distributed circuits underlying anxiety. Frontiers in Behavioral Neuroscience. 2014;8:112
  19. 19. Calhoon GG, Tye KM. Resolving the neural circuits of anxiety. Nature Neuroscience. 2015;18(10):1394-1404
  20. 20. Matosin N et al. Metabotropic glutamate receptor mGluR2/3 and mGluR5 binding in the anterior cingulate cortex in psychotic and nonpsychotic depression, bipolar disorder and schizophrenia: Implications for novel mGluR-based therapeutics. Journal of Psychiatry & Neuroscience. 2014;39(6):407-416
  21. 21. Deschwanden A et al. Reduced metabotropic glutamate receptor 5 density in major depression determined by [(11)C]ABP688 PET and postmortem study. The American Journal of Psychiatry. 2011;168(7):727-734
  22. 22. Abdallah CG et al. Metabotropic glutamate receptor 5 and glutamate involvement in major depressive disorder: A multimodal imaging study. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging. 2017;2(5):449-456
  23. 23. Luscher C, Huber KM. Group 1 mGluR-dependent synaptic long-term depression: Mechanisms and implications for circuitry and disease. Neuron. 2010;65(4):445-459
  24. 24. Xu J et al. Hippocampal metaplasticity is required for the formation of temporal associative memories. The Journal of Neuroscience. 2014;34(50):16762-16773
  25. 25. Chevaleyre V, Castillo PE. Heterosynaptic LTD of hippocampal GABAergic synapses: A novel role of endocannabinoids in regulating excitability. Neuron. 2003;38(3):461-472
  26. 26. Lu YM 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
  27. 27. Xu J et al. Genetic disruption of Grm5 causes complex alterations in motor activity, anxiety and social behaviors. Behavioural Brain Research. 2021;411:113378
  28. 28. Xu J et al. mGluR5 has a critical role in inhibitory learning. The Journal of Neuroscience. 2009;29(12):3676-3684
  29. 29. Chiamulera C et al. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nature Neuroscience. 2001;4(9):873-874
  30. 30. Wijetunge LS et al. mGluR5 regulates glutamate-dependent development of the mouse somatosensory cortex. The Journal of Neuroscience. 2008;28(49):13028-13037
  31. 31. Conquet F et al. Motor deficit and impairment of synaptic plasticity in mice lacking mGluR1. Nature. 1994;372(6503):237-243
  32. 32. Brody SA, Conquet F, Geyer MA. Disruption of prepulse inhibition in mice lacking mGluR1. The European Journal of Neuroscience. 2003;18(12):3361-3366
  33. 33. Crupi R, Impellizzeri D, Cuzzocrea S. Role of metabotropic glutamate receptors in neurological disorders. Frontiers in Molecular Neuroscience. 2019;12:20
  34. 34. Dogra S, Conn PJ. Metabotropic glutamate receptors As emerging targets for the treatment of schizophrenia. Molecular Pharmacology. 2022;101(5):275-285
  35. 35. Ferraguti F. Metabotropic glutamate receptors as targets for novel anxiolytics. Current Opinion in Pharmacology. 2018;38:37-42
  36. 36. Bear MF. Therapeutic implications of the mGluR theory of fragile X mental retardation. Genes, Brain, and Behavior. 2005;4(6):393-398
  37. 37. Tanabe Y et al. A family of metabotropic glutamate receptors. Neuron. 1992;8(1):169-179
  38. 38. Scanziani M et al. Use-dependent increases in glutamate concentration activate presynaptic metabotropic glutamate receptors. Nature. 1997;385(6617):630-634
  39. 39. Saugstad JA, Segerson TP, Westbrook GL. Metabotropic glutamate receptors activate G-protein-coupled inwardly rectifying potassium channels in Xenopus oocytes. The Journal of Neuroscience. 1996;16(19):5979-5985
  40. 40. Fujioka R et al. Comprehensive behavioral study of mGluR3 knockout mice: Implication in schizophrenia related endophenotypes. Molecular Brain. 2014;7:31
  41. 41. Linden AM et al. Use of MGLUR2 and MGLUR3 knockout mice to explore in vivo receptor specificity of the MGLUR2/3 selective antagonist LY341495. Neuropharmacology. 2009;57(2):172-182
  42. 42. Yokoi M et al. Impairment of hippocampal mossy fiber LTD in mice lacking mGluR2. Science. 1996;273(5275):645-647
  43. 43. Linden AM et al. 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. 2005;179(1):284-291
  44. 44. Morishima Y et al. Enhanced cocaine responsiveness and impaired motor coordination in metabotropic glutamate receptor subtype 2 knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(11):4170-4175
  45. 45. Corti C 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
  46. 46. Li SH, Abd-Elrahman KS, Ferguson SSG. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacology & Therapeutics. 2022;239:108275
  47. 47. Mazzitelli M et al. Group II metabotropic glutamate receptors: Role in pain mechanisms and pain modulation. Frontiers in Molecular Neuroscience. 2018;11:383
  48. 48. Nakajima Y 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
  49. 49. Duvoisin RM, Zhang C, Ramonell K. A novel metabotropic glutamate receptor expressed in the retina and olfactory bulb. The Journal of Neuroscience. 1995;15(4):3075-3083
  50. 50. Corti C et al. Distribution and synaptic localisation of the metabotropic glutamate receptor 4 (mGluR4) in the rodent CNS. Neuroscience. 2002;110(3):403-420
  51. 51. Nicoletti F et al. Metabotropic glutamate receptors: From the workbench to the bedside. Neuropharmacology. 2011;60(7-8):1017-1041
  52. 52. Schoepp DD, Jane DE, Monn JA. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology. 1999;38(10):1431-1476
  53. 53. Palazzo E et al. Metabotropic glutamate receptor 7: From synaptic function to therapeutic implications. Current Neuropharmacology. 2016;14(5):504-513
  54. 54. Holscher C et al. 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
  55. 55. Callaerts-Vegh Z 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
  56. 56. Pekhletski R 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
  57. 57. Iscru E et al. Improved spatial learning is associated with increased hippocampal but not prefrontal long-term potentiation in mGluR4 knockout mice. Genes, Brain, and Behavior. 2013;12(6):615-625
  58. 58. Duvoisin RM et al. Sex-dependent cognitive phenotype of mice lacking mGluR8. Behavioural Brain Research. 2010;209(1):21-26
  59. 59. Duvoisin RM 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
  60. 60. Fisher NM et al. Phenotypic profiling of mGlu(7) knockout mice reveals new implications for neurodevelopmental disorders. Genes, Brain, and Behavior. 2020;19(7):e12654
  61. 61. Bertaso F et al. PICK1 uncoupling from mGluR7a causes absence-like seizures. Nature Neuroscience. 2008;11(8):940-948
  62. 62. Goudet C et al. Metabotropic receptors for glutamate and GABA in pain. Brain Research Reviews. 2009;60(1):43-56
  63. 63. Cosford ND et al. 3-[(2-Methyl-1,3-thiazol-4-yl)ethynyl]-pyridine: A potent and highly selective metabotropic glutamate subtype 5 receptor antagonist with anxiolytic activity. Journal of Medicinal Chemistry. 2003;46(2):204-206
  64. 64. Helton DR et al. Anxiolytic and side-effect profile of LY354740: A potent, highly selective, orally active agonist for group II metabotropic glutamate receptors. The Journal of Pharmacology and Experimental Therapeutics. 1998;284(2):651-660
  65. 65. Klodzinska A et al. Potential anti-anxiety, anti-addictive effects of LY 354740, a selective group II glutamate metabotropic receptors agonist in animal models. Neuropharmacology. 1999;38(12):1831-1839
  66. 66. Pilc A et al. Multiple MPEP administrations evoke anxiolytic- and antidepressant-like effects in rats. Neuropharmacology. 2002;43(2):181-187
  67. 67. Spooren WP et al. Anxiolytic-like effects of the prototypical metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)pyridine in rodents. The Journal of Pharmacology and Experimental Therapeutics. 2000;295(3):1267-1275
  68. 68. Tatarczynska E 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
  69. 69. Tatarczynska E et al. The antianxiety-like effects of antagonists of group I and agonists of group II and III metabotropic glutamate receptors after intrahippocampal administration. Psychopharmacology. 2001;158(1):94-99
  70. 70. LeDoux JE. Emotion circuits in the brain. Annual Review of Neuroscience. 2000;23:155-184
  71. 71. Muir J, Lopez J, Bagot RC. Wiring the depressed brain: Optogenetic and chemogenetic circuit interrogation in animal models of depression. Neuropsychopharmacology. 2019;44(6):1013-1026
  72. 72. Swanson CJ et al. Metabotropic glutamate receptors as novel targets for anxiety and stress disorders. Nature Reviews. Drug Discovery. 2005;4(2):131-144
  73. 73. Krystal JH et al. Potential psychiatric applications of metabotropic glutamate receptor agonists and antagonists. CNS Drugs. 2010;24(8):669-693
  74. 74. Galici R 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
  75. 75. Muly EC et al. 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
  76. 76. Matrisciano F et al. Group-II metabotropic glutamate receptor ligands as adjunctive drugs in the treatment of depression: A new strategy to shorten the latency of antidepressant medication? Molecular Psychiatry. 2007;12(8):704-706
  77. 77. Fell MJ et al. 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-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039). The Journal of Pharmacology and Experimental Therapeutics. 2008;326(1):209-217
  78. 78. Nasca C et al. L-acetylcarnitine causes rapid antidepressant effects through the epigenetic induction of mGlu2 receptors. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(12):4804-4809
  79. 79. Yoshimizu T et al. An mGluR2/3 antagonist, MGS0039, exerts antidepressant and anxiolytic effects in behavioral models in rats. Psychopharmacology. 2006;186(4):587-593
  80. 80. Engers JL et al. Discovery of a selective and CNS penetrant negative allosteric modulator of metabotropic glutamate receptor subtype 3 with antidepressant and anxiolytic activity in rodents. Journal of Medicinal Chemistry. 2015;58(18):7485-7500
  81. 81. Linden AM et al. Systemic administration of the potent mGlu8 receptor agonist (S)-3,4-DCPG induces c-Fos in stress-related brain regions in wild-type, but not mGlu8 receptor knockout mice. Neuropharmacology. 2003;45(4):473-483
  82. 82. Palucha A et al. Group III mGlu receptor agonists produce anxiolytic- and antidepressant-like effects after central administration in rats. Neuropharmacology. 2004;46(2):151-159
  83. 83. Klak K et al. 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
  84. 84. Wieronska JM et al. Metabotropic glutamate receptor 4 novel agonist LSP1-2111 with anxiolytic, but not antidepressant-like activity, mediated by serotonergic and GABAergic systems. Neuropharmacology. 2010;59(7-8):627-634
  85. 85. Slawinska A et al. Anxiolytic- but not antidepressant-like activity of Lu AF21934, a novel, selective positive allosteric modulator of the mGlu(4) receptor. Neuropharmacology. 2013;66:225-235
  86. 86. Kotlinska JH et al. Impact of the metabotropic glutamate receptor7 (mGlu(7)) allosteric agonist, AMN082, on fear learning and memory and anxiety-like behavior. European Journal of Pharmacology. 2019;858:172512
  87. 87. Aiba A et al. Reduced hippocampal long-term potentiation and context-specific deficit in associative learning in mGluR1 mutant mice. Cell. 1994;79(2):365-375
  88. 88. Masu M et al. Specific deficit of the ON response in visual transmission by targeted disruption of the mGluR6 gene. Cell. 1995;80(5):757-765
  89. 89. Sansig G et al. Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7. The Journal of Neuroscience. 2001;21(22):8734-8745
  90. 90. Nakao H et al. Metabotropic glutamate receptor subtype-1 is essential for motor coordination in the adult cerebellum. Neuroscience Research. 2007;57(4):538-543
  91. 91. Hamilton A et al. Metabotropic glutamate receptor 5 knockout reduces cognitive impairment and pathogenesis in a mouse model of Alzheimer's disease. Molecular Brain. 2014;7:40
  92. 92. Olsen CM et al. Operant sensation seeking requires metabotropic glutamate receptor 5 (mGluR5). PLoS One. 2010;5(11):e15085
  93. 93. Inta D et al. Significant increase in anxiety during aging in mGlu5 receptor knockout mice. Behavioural Brain Research. 2013;241:27-31
  94. 94. De Filippis B et al. The role of group II metabotropic glutamate receptors in cognition and anxiety: Comparative studies in GRM2(−/−), GRM3(−/−) and GRM2/3(−/−) knockout mice. Neuropharmacology. 2015;89:19-32
  95. 95. Davis MJ et al. Measures of anxiety, sensorimotor function, and memory in male and female mGluR4(−)/(−) mice. Behavioural Brain Research. 2012;229(1):21-28
  96. 96. Cryan JF et al. 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
  97. 97. Ludewig S et al. Information-processing deficits and cognitive dysfunction in panic disorder. Journal of Psychiatry & Neuroscience. 2005;30(1):37-43
  98. 98. Grillon C et al. Baseline startle amplitude and prepulse inhibition in Vietnam veterans with posttraumatic stress disorder. Psychiatry Research. 1996;64(3):169-178
  99. 99. Fontanez-Nuin DE et al. Memory for fear extinction requires mGluR5-mediated activation of infralimbic neurons. Cerebral Cortex. 2011;21(3):727-735
  100. 100. Sethna F, Wang H. Pharmacological enhancement of mGluR5 facilitates contextual fear memory extinction. Learning & Memory. 2014;21(12):647-650
  101. 101. Pecknold JC et al. 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
  102. 102. Porter RH 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
  103. 103. Esterlis I et al. Metabotropic glutamatergic receptor 5 and stress disorders: Knowledge gained from receptor imaging studies. Biological Psychiatry. 2018;84(2):95-105
  104. 104. Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacology, Biochemistry, and Behavior. 2022;219:173446
  105. 105. Gottschalk MG, Domschke K. Genetics of generalized anxiety disorder and related traits. Dialogues in Clinical Neuroscience. 2017;19(2):159-168
  106. 106. Breen G et al. A genome-wide significant linkage for severe depression on chromosome 3: The depression network study. The American Journal of Psychiatry. 2011;168(8):840-847
  107. 107. Pergadia ML et al. A 3p26-3p25 genetic linkage finding for DSM-IV major depression in heavy smoking families. The American Journal of Psychiatry. 2011;168(8):848-852
  108. 108. Lee PH et al. Multi-locus genome-wide association analysis supports the role of glutamatergic synaptic transmission in the etiology of major depressive disorder. Translational Psychiatry. 2012;2(11):e184
  109. 109. Terracciano A et al. Genome-wide association scan of trait depression. Biological Psychiatry. 2010;68(9):811-817
  110. 110. Hadley D et al. The impact of the metabotropic glutamate receptor and other gene family interaction networks on autism. Nature Communications. 2014;5:4074
  111. 111. Iossifov I et al. De novo gene disruptions in children on the autistic spectrum. Neuron. 2012;74(2):285-299
  112. 112. Kelleher I et al. Prevalence of psychotic symptoms in childhood and adolescence: A systematic review and meta-analysis of population-based studies. Psychological Medicine. 2012;42(9):1857-1863
  113. 113. Wenger TL et al. The role of mGluR copy number variation in genetic and environmental forms of syndromic autism Spectrum disorder. Scientific Reports. 2016;6:19372
  114. 114. Fuchigami T, Nakayama M, Yoshida S. Development of PET and SPECT probes for glutamate receptors. Scientific World Journal. 2015;2015:716514
  115. 115. Ametamey SM et al. Radiosynthesis and preclinical evaluation of 11C-ABP688 as a probe for imaging the metabotropic glutamate receptor subtype 5. Journal of Nuclear Medicine. 2006;47(4):698-705
  116. 116. Zanotti-Fregonara P et al. The PET Radioligand 18F-FIMX images and quantifies metabotropic glutamate receptor 1 in proportion to the regional density of its gene transcript in human brain. Journal of Nuclear Medicine. 2016;57(2):242-247
  117. 117. Wong DF et al. 18F-FPEB, a PET radiopharmaceutical for quantifying metabotropic glutamate 5 receptors: A first-in-human study of radiochemical safety, biokinetics, and radiation dosimetry. Journal of Nuclear Medicine. 2013;54(3):388-396
  118. 118. Shetty HU et al. Radiodefluorination of 3-fluoro-5-(2-(2-[18F](fluoromethyl)-thiazol-4-yl)ethynyl)benzonitrile ([18F]SP203), a radioligand for imaging brain metabotropic glutamate subtype-5 receptors with positron emission tomography, occurs by glutathionylation in rat brain. The Journal of Pharmacology and Experimental Therapeutics. 2008;327(3):727-735
  119. 119. Holmes SE et al. Altered metabotropic glutamate receptor 5 markers in PTSD: In vivo and postmortem evidence. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(31):8390-8395
  120. 120. Davis MT et al. In vivo evidence for dysregulation of mGluR5 as a biomarker of suicidal ideation. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(23):11490-11495
  121. 121. Akkus F et al. Metabotropic glutamate receptor 5 binding in patients with obsessive-compulsive disorder. The International Journal of Neuropsychopharmacology. 2014;17(12):1915-1922
  122. 122. DeLorenzo C et al. Characterization of brain mGluR5 binding in a pilot study of late-life major depressive disorder using positron emission tomography and [(1)(1)C]ABP688. Translational Psychiatry. 2015;5(12):e693
  123. 123. Andres JI et al. Synthesis, evaluation, and radiolabeling of new potent positive allosteric modulators of the metabotropic glutamate receptor 2 as potential tracers for positron emission tomography imaging. Journal of Medicinal Chemistry. 2012;55(20):8685-8699
  124. 124. Kil KE et al. Radiosynthesis of N-(4-chloro-3-[(11)C]methoxyphenyl)-2-picolinamide ([(11)C]ML128) as a PET radiotracer for metabotropic glutamate receptor subtype 4 (mGlu4). Bioorganic & Medicinal Chemistry. 2013;21(19):5955-5962
  125. 125. Bittigau P, Ikonomidou C. Glutamate in neurologic diseases. Journal of Child Neurology. 1997;12(8):471-485
  126. 126. Javitt DC. Glutamate as a therapeutic target in psychiatric disorders. Molecular Psychiatry. 2004;9(11):984-997 979
  127. 127. Peterlik D, Flor PJ, Uschold- Schmidt N. The emerging role of metabotropic glutamate receptors in the pathophysiology of chronic stress-related disorders. Current Neuropharmacology. 2016;14(5):514-539
  128. 128. Quiroz JA et al. Efficacy and safety of basimglurant as adjunctive therapy for major depression: A Randomized clinical trial. JAMA Psychiatry. 2016;73(7):675-684
  129. 129. Rutrick D et al. Mavoglurant augmentation in OCD patients resistant to selective serotonin reuptake inhibitors: A proof-of-concept, Randomized, placebo-controlled, phase 2 study. Advances in Therapy. 2017;34(2):524-541
  130. 130. Bergink V, Westenberg HG. Metabotropic glutamate II receptor agonists in panic disorder: A double blind clinical trial with LY354740. International Clinical Psychopharmacology. 2005;20(6):291-293
  131. 131. Dunayevich E et al. Efficacy and tolerability of an mGlu2/3 agonist in the treatment of generalized anxiety disorder. Neuropsychopharmacology. 2008;33(7):1603-1610
  132. 132. Kent JM et al. Efficacy and safety of an adjunctive mGlu2 receptor positive allosteric modulator to a SSRI/SNRI in anxious depression. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2016;67:66-73
  133. 133. Umbricht D et al. Placebo-controlled trial of the mGlu2/3 negative allosteric modulator decoglurant in partially refractory major depressive disorder. The Journal of Clinical Psychiatry. 2020;81(4)

Written By

Jian Xu and Yongling Zhu

Submitted: 04 August 2023 Reviewed: 07 August 2023 Published: 09 October 2023