Chapter Three - Modulation of Chronic Pain by Metabotropic Glutamate Receptors

https://doi.org/10.1016/bs.apha.2015.11.001Get rights and content

Abstract

Metabotropic glutamate receptors (mGluRs) belong to class C G-protein-coupled receptors. They are expressed throughout the nervous system on both neurons and glial cells. In the central nervous system (CNS), mGluRs are mainly located in the proximity of the synaptic cleft where they regulate glutamatergic transmission in addition to a number of other neurotransmitters. To date, eight subtypes of mGluRs (mGluR1–mGluR8) have been cloned and classified into three groups on the basis of sequence similarities, and pharmacological and biochemical properties. Consequently, group I mGluRs includes mGluR1 and mGluR5, group II mGluRs includes mGluR2 and mGluR3, and group III mGluRs consists of mGluR4, mGluR6, mGluR7, and mGluR8. With the exception of mGluR6, whose localization is restricted within the retina, all mGluRs are ubiquitously expressed throughout the peripheral and CNS with some subtype specificity in different anatomical regions. mGluRs participate in many physiological processes and play important roles in a number of neurological conditions including anxiety, depression, schizophrenia, and neurodegenerative disorders. mGluRs also participate in the physiological transmission of pain stimuli as well as to mechanisms involved in the establishment of chronic pain. Therefore, these receptors are attractive targets for therapeutic intervention in several neurological disorders including chronic pain. Thus, understanding the physiological function and role of each mGluR subtype in the development of chronic pain will provide a better insight into the potential use of subtype-selective drugs currently being developed as orthosteric or allosteric ligands.

Introduction

Pain is usually classified as either nociceptive, inflammatory, or neuropathic, according to its origin, symptoms, and underlying mechanisms (Costigan, Scholz, & Woolfet, 2009). Although this classification is very useful from a didactic point of view, it does not reflect the complexity of pain disorders in clinical conditions. In chronic pain patients, the presence of more than one component extremely complicates the assessment and management of pain (Baron & Binder, 2004). In this chapter, recent literature describing the role of glutamate transmission via metabotropic glutamate receptor (mGluR) subtypes in the pathophysiology of chronic pain is reviewed.

Glutamate is the main excitatory amino acid in the central nervous system (CNS) and is involved in brain function and pathology. As neurotransmitter, glutamate activates two different types of receptors: ionotropic glutamate receptors (iGluRs) and mGluRs. According to their response to different agonists, iGluRs have been further subdivided into three distinct types: N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-isoxazole-4-propionate (AMPA), and kainate (KA) receptors (Hollmann and Heinemann, 1994, Monaghan et al., 1989). iGluRs are directly coupled to a membrane ion channel and mediate fast excitatory synaptic transmission. On the contrary, mGluRs are cell membrane receptors belonging to the class C of the G-protein-coupled receptor (GPCR) superfamily that modulate rather than mediate neuronal excitability and neurotransmitter release via G-protein-dependent and non-G-protein-dependent signaling pathways (Anwyl, 1999).

By activating iGluRs and/or mGluRs, glutamate participates not only in the physiological transmission of nociceptive information but also in the development of peripheral and central mechanisms of pain hypersensitivity (Latremoliere & Woolf, 2009). Glutamate plays a primary role in the increased synaptic efficacy occurring after injury, a mechanism that has been referred to activity-dependent central sensitization (Woolf, 1983). As for the hippocampal long-term potentiation (LTP) of the synaptic strength (Ji, Kohno, Moore, & Woolf, 2003) which is a key mechanism for memory consolidation (Nicoll, 2003), glutamate-induced plasticity is a fundamental step for the development of the so-called “memory of pain” that is the basis for the establishment of chronic pain symptoms such as hyperalgesia, allodynia, and spontaneous pain (Woolf, 1983). Both NMDA and AMPA receptors, together with a number of metabotropic receptors, can trigger central sensitization through an increase in intracellular calcium levels that activate intracellular pathways strengthening the excitatory synapse (Latremoliere & Woolf, 2009). The blockade of NMDA receptors consistently resulting in the prevention of hyperexcitability of nociceptive neurons suggests the potential use of NMDA antagonist in the treatment of pain hypersensitivity (Woolf & Thompson, 1991). However, besides the selective antagonists of the NMDA receptor NR2B subunit (Boyce et al., 1999), the direct blockade of glutamate transmission by iGluRs antagonists is known to have detrimental effects on learning and memory processes (Chizh, 2007, Nakazato et al., 2005, Wei et al., 2001, Zhang et al., 2009); therefore, an NMDA antagonist-based therapy for chronic pain is unlikely to be developed. Consequently, understanding the function, expression, and role of mGluRs in pain modulation and their involvement in the induction and maintenance of central sensitization may help to find a practicable alternative to reduce hypersensitivity in chronic pain conditions.

Section snippets

Classification of mGluRs

mGluRs belong to the class C of GPCRs (Schiöth & Fredriksson, 2005). There are eight types of mGluRs (mGluR1–8) that can be divided into three different groups according to their sequence similarity, pharmacology and intracellular signal transduction pathways (Fig. 1; Conn and Pin, 1997, Pin and Duvoisin, 1995). Group I include mGluR1 and mGluR5; group II include mGluR2 and mGluR3; and group III is formed by mGluR4, mGluR6, mGluR7, and mGluR8. Moreover, virtually all mGluR genes have been

Orthosteric Ligands and Allosteric Modulators of mGluRs

During the last decades, a number of synthetic compounds have been developed with the ability to modulate glutamatergic transmission by either activating or inhibiting specific mGluR groups or subtypes. Among these several compounds, orthosteric ligands and allosteric modulators have been employed to investigate the specific role of mGluRs in pain modulation (Chiechio and Nicoletti, 2012, Montana and Gereau, 2011). Since the majority of allosteric modulators bind to a site within the 7TMD,

mGluR Subtypes in Pain

The assessment and management of chronic pain syndromes can be extremely complicated by the coexistence of mixed pain forms (Baron & Binder, 2004). By counteracting both nociceptive and persistent forms of pain, mGluRs are excellent candidates for chronic pain treatment. mGluRs are able to reduce pain hypersensitivity acting at different levels, from the periphery to brain regions involved in the perception and modulation of pain. With the exception of the mGluR6 subtype, whose localization is

Role of Group I mGluRs in Pain Modulation

Pharmacological interventions for pain relief are usually aimed at inhibiting group I mGluRs and/or potentiating group II and III mGluR signaling. Glutamate released from primary afferent neurons is a key event for persistent activation of spinal neurons leading to hypersensitivity in chronic pain states. Among group I mGluRs, the mGluR5 subtype, together with NMDA receptors, gives an important contribution in nociceptive processing and in the development of central sensitization by activating

Role of Group II mGluRs in Pain Modulation

The role of group II mGluRs has been extensively investigated for their ability to reduce hyperalgesia in animal models of persistent pain (Fisher and Coderre, 1996, Fisher et al., 2002, Sharpe et al., 2000, Simmons et al., 2002). mGluR2 and mGluR3 are generally located presynaptically on sensory nerve terminals. In the synapse, these receptors function as presynaptic auto- or heteroreceptors, regulate neurotransmitter release, and mainly respond to glutamate released from astrocytes (Kalivas,

Role of Group III mGluRs in Pain Modulation

mGluR subtypes belonging to group III are also expressed throughout the pain neuraxis from peripheral nerves to the CNS. In particular, the expression of mGluR8 has been shown in unmyelinated fibers of digital nerves where they negatively modulate the activity of TRPV1 receptors on nociceptors via the inhibition of adenylyl cyclase (Govea, Zhou, & Carlton, 2012). Consistently, the intraplantar injection of the group III mGluRs agonist, l-AP4, is able to attenuate the hyperalgesia induced by the

Conclusion

The treatment of chronic pain can be extremely complicated due to the coexistence of mixed pain forms that often occur in patients. Over the past decades, the role of mGluRs has been investigated in different forms of pain. The increasing availability of novel subtype-selective pharmacological ligands acting at mGluRs has helped to clarify the precise role of each mGluR subtype in pain processing and in mechanisms of nociceptive sensitization suggesting that these receptors can serve as

Conflict of Interest

No conflicts of interest to declare.

References (167)

  • G.H. Cho et al.

    Synthesis and biological evaluation of aryl isoxazole derivatives as metabotropic glutamate receptor 1 antagonists: A potential treatment for neuropathic pain

    Bioorganic & Medicinal Chemistry Letters

    (2015)
  • M.K. Chung et al.

    Peripheral group I metabotropic glutamate receptor activation leads to muscle mechanical hyperalgesia through TRPV1 phosphorylation in the rat

    The Journal of Pain

    (2015)
  • V. de Novellis et al.

    Periaqueductal grey CB1 cannabinoid and metabotropic glutamate subtype 5 receptors modulate changes in rostral ventromedial medulla neuronal activities induced by subcutaneous formalin in the rat

    Neuroscience

    (2005)
  • A.J. Doherty et al.

    (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) activates mGlu5, but no mGlu1, receptors expressed in CHO cells and potentiates NMDA responses in the hippocampus

    Neuropharmacology

    (1997)
  • S. Dolan et al.

    Up-regulation of metabotropic glutamate receptor subtypes 3 and 5 in spinal cord in a clinical model of persistent inflammation and hyperalgesia

    Pain

    (2003)
  • G.M. Drew et al.

    Multiple metabotropic glutamate receptor subtypes modulate GABAergic neurotransmission in rat periaqueductal grey neurons in vitro

    Neuropharmacology

    (2004)
  • R.M. Duvoisin et al.

    Acute pharmacological modulation of mGluR8 reduces measures of anxiety

    Behavioural Brain Research

    (2010)
  • S.A. Eaton et al.

    Thalamic NMDA receptors and nociceptive sensory synaptic transmission

    Neuroscience Letters

    (1990)
  • K. Fisher et al.

    The contribution of metabotropic glutamate receptors (mGluRs) to formalin-induced Nociception

    Pain

    (1996)
  • K. Fisher et al.

    Antinociceptive effects following intrathecal pretreatment with selective metabotropic glutamate receptor compounds in a rat model of neuropathic pain

    Pharmacology, Biochemistry, and Behavior

    (2002)
  • L. Gama et al.

    Heterodimerization of calcium sensing receptors with metabotropic glutamate receptors in neurons

    The Journal of Biological Chemistry

    (2001)
  • F. Gasparini et al.

    2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist

    Neuropharmacology

    (1999)
  • C.E. Gee et al.

    Blocking metabotropic glutamate receptor subtype 7 (mGlu7) via the Venus flytrap domain (VFTD) inhibits amygdala plasticity, stress, and anxiety-related behavior

    The Journal of Biological Chemistry

    (2014)
  • G. Gerber et al.

    Group II and group III metabotropic glutamate receptor agonists depress synaptic transmission in the rat spinal cord dorsal horn

    Neuroscience

    (2000)
  • C. Goudet et al.

    Group III metabotropic glutamate receptors inhibit hyperalgesia in animal models of inflammation and neuropathic pain

    Pain

    (2008)
  • C. Goudet et al.

    Metabotropic receptors for glutamate and GABA in pain

    Brain Research Reviews

    (2009)
  • R.M. Govea et al.

    Group III metabotropic glutamate receptors and transient receptor potential vanilloid 1 co-localize and interact on nociceptors

    Neuroscience

    (2012)
  • K.J. Gregory et al.

    Pharmacology of metabotropic glutamate receptor allosteric modulators: Structural basis and therapeutic potential for CNS disorders

    Progress in Molecular Biology and Translational Science

    (2013)
  • J. Hao et al.

    Discovery of (1R,2R)-N-(4-(6-isopropylpyridine-2-yl)-3-(2-methyl-2H-indazol-5-yl)isothiazol-5-yl)-2-methylcyclopropanecarboxamide, a potent and orally efficacious mGlu5 receptor negative allosteric modulator

    Bioorganic & Medicinal Chemistry Letters

    (2013)
  • J. Hu et al.

    Human Ca2 + receptor cysteine-rich domain. Analysis of function of mutant and chimeric receptors

    The Journal of Biological Chemistry

    (2000)
  • D.E. Jane et al.

    Potent antagonists at the L-AP4- and (1S,3S)-ACPD-sensitive presynaptic metabotropic glutamate receptors in the neonatal rat spinal cord

    Neuropharmacology

    (1996)
  • R.R. Ji et al.

    Central sensitization and LTP: Do pain and memory share similar mechanisms?

    Trends in Neurosciences

    (2003)
  • B.G. Johnson et al.

    [3H]-LY341495 as a novel antagonist radioligand for group II metabotropic glutamate (mGlu) receptors: Characterization of binding to membranes of mGlu receptor subtype expressing cells

    Neuropharmacology

    (1999)
  • Y. Kim et al.

    Novel thienopyrimidinones as mGluR1 antagonists

    European Journal of Medicinal Chemistry

    (2014)
  • A.E. Kingston et al.

    Pharmacological analysis of 4-carboxyphenylglycine derivatives: Comparison of effects on mGluR1 alpha and mGluR5a subtypes

    Neuropharmacology

    (1995)
  • T. Kiritoshi et al.

    Group II mGluRs modulate baseline and arthritis pain-related synaptic transmission in the rat medial prefrontal cortex

    Neuropharmacology

    (2015)
  • T. Kiritoshi et al.

    Modulation of pyramidal cell output in the medial prefrontal cortex by mGluR5 interacting with CB1

    Neuropharmacology

    (2013)
  • J. Kniazeff et al.

    Dimers and beyond: The functional puzzles of class C GPCRs

    Pharmacology & Therapeutics

    (2011)
  • R. Kolhekar et al.

    Thalamic NMDA receptors modulate inflammation-produced hyperalgesia in the rat

    Pain

    (1997)
  • A. Latremoliere et al.

    Central sensitization: A generator of pain hypersensitivity by central neural plasticity

    The Journal of Pain

    (2009)
  • H. Lavreysen et al.

    JNJ16259685, a highly potent, selective and systemically active mGlu1 receptor antagonist

    Neuropharmacology

    (2004)
  • K.S. Lee et al.

    Peripheral group II and III metabotropic glutamate receptors in the knee joint attenuate carrageenan-induced nociceptive behavior in rats

    Neuroscience Letters

    (2013)
  • H. Li et al.

    Localization of a metabotropic glutamate receptor, mGluR7, in axon terminals of presumed nociceptive, primary afferent fibers in the superficial layers of the spinal dorsal horn: An electron microscope study in the rat

    Neuroscience Letters

    (1997)
  • E. Lindström et al.

    Involvement of metabotropic glutamate 5 receptor in visceral pain

    Pain

    (2008)
  • L. Luongo et al.

    Role of metabotropic glutamate receptor 1 in the basolateral amygdala-driven prefrontal cortical deactivation in inflammatory pain in the rat

    Neuropharmacology

    (2013)
  • S. Maione et al.

    Periaqueductal gray matter metabotropic glutamate receptors modulate formalin-induced nociception

    Pain

    (2000)
  • I. Marabese et al.

    Effects of (S)-3,4-DCPG, an mGlu8 receptor agonist, on inflammatory and neuropathic pain in mice

    Neuropharmacology

    (2007)
  • N. Ai et al.

    Identification of nitazoxanide as a group I metabotropic glutamate receptor negative modulator for the treatment of neuropathic pain: An in silico drug repositioning study

    Pharmaceutical Research

    (2015)
  • E. Akgün et al.

    Ligands that interact with putative MOR-mGluR5 heteromer in mice with inflammatory pain produce potent antinociception

    Proceedings of the National Academy of Sciences of the United States of America

    (2013)
  • J.J. Azkue et al.

    Immunoreactivity for the group III metabotropic glutamate receptor subtype mGluR4a in the superficial laminae of the rat spinal dorsal horn

    The Journal of Comparative Neurology

    (2001)
  • Cited by (37)

    • Glutamatergic systems in neuropathic pain and emerging non-opioid therapies

      2022, Pharmacological Research
      Citation Excerpt :

      Physiologically, they generate slower post-synaptic potentials than iGluRs and are implicated in long-term changes, particularly in synaptic plasticity and the formation of learning and memory [154]. All mGluR subtypes (except for mGluR6, which is localized in the retina only) are expressed within the nociceptive pathways where they play roles modulating pain transmission[135,155]. Many selective compounds for the different mGluR subtypes have been identified in the last few years, contributing to our understanding of their specific roles in nociception and suggesting that these receptors are promising targets for the treatment of chronic pain.

    • Ah-type Baroreceptor Neurons Expressing Estrogen Dependent mGluR7 Mediate Descending Inhibition of Cardiac Nociception

      2022, Neuroscience
      Citation Excerpt :

      Metabotropic glutamate receptors (mGluRs) are a group of G-protein-coupled receptors (GPCRs) that bind glutamate and modulate synaptic transmission (Niswender and Conn, 2010, Palazzo et al., 2017), including baroreflex to balance of autonomic control of BP. Moreover, mGluR activation is involved in pain initiation and transmission (Chiechio, 2016, Boccella et al., 2020). The aim of this investigation was to verify whether glutamate mediates baroreceptor activation partly through afferent-specific expression of mGluRs in the baroreflex afferent pathway and produces more pronounced descending inhibition to cardiac nociception.

    • N-acetylaspartylglutamate (NAAG) and glutamate carboxypeptidase II: An abundant peptide neurotransmitter-enzyme system with multiple clinical applications

      2020, Progress in Neurobiology
      Citation Excerpt :

      While NAAG activates a limited subpopulation of NMDA receptors under some conditions (Khacho et al., 2015) but not others (Lea et al., 2001; Losi et al., 2004; Fricker et al., 2009), its most studied functions are mediated by activation of the Group II metabotropic glutamate receptor mGluR3 and the subsequent inhibition of adenylate or guanylate cyclase (Wroblewska et al., 1998, 2006). This receptor is expressed by astrocytes (Wroblewska et al., 1998; Minoshima and Nakanishi, 1999) and has been localized on pre and post synaptic neurons as well (Chiechio, 2016). While the actions of NAAG on astrocytes and post-synaptic neuronal sites have not been extensively studied, NAAG activation of presynaptic metabotropic receptors reduces corelease of small amine transmitters, including glutamate (Zhao et al., 2001; Sanabria et al., 2004; Romei et al., 2013) (Fig. 1).

    View all citing articles on Scopus
    View full text