Associate editor: P. Molenaar
Regulation of metabotropic glutamate receptor signaling, desensitization and endocytosis

https://doi.org/10.1016/j.pharmthera.2005.01.008Get rights and content

Abstract

Metabotropic glutamate receptors (mGluRs) comprise a unique family of G protein-coupled receptors (GPCR) that can be classified into 3 groups based on G protein coupling specificity and sequence similarity. Group I mGluRs (mGluR1 and mGluR5) are coupled to the heterotrimeric G protein Gαq/11 and trigger the release of calcium from intracellular stores. In the present review, we discuss the molecular mechanisms involved in the desensitization and endocytosis of group I mGluRs. Group I mGluRs desensitize in response to both second-messenger-dependent protein kinases and G protein-coupled receptor kinases (GRK). However, GRK2-mediated mGluR1 desensitization appears to be both phosphorylation- and β-arrestin-independent. In addition to GRK-mediated uncoupling of mGluRs from heterotrimeric G proteins, the huntingtin-interacting protein, optineurin, also contributes to mGluR1 and mGluR5 desensitization. The G protein-uncoupling activity of optineurin appears to be facilitated by the presence of polyglutamine-expanded mutant huntingtin but not wild-type huntingtin. Group I mGluRs also undergo both agonist-dependent and -independent endocytosis in both heterologous cell expression systems and primary neuronal cultures. The present review overviews the current understanding of the contribution of second messenger-dependent protein kinases, β-arrestins and a novel Ral/phospholipase D2 (PLD2)-mediated endocytic pathway to the regulation of Group I mGluR endocytosis. Overall, the regulation of Group I mGluR desensitization and endocytosis appears to be mediated by the same molecular intermediates as have been described for more typical GPCR such as the β2-adrenergic receptor. However, there appears to be subtle, but important, differences in the mechanisms by which these intermediates are employed to regulate Group I mGluR desensitization and endocytosis.

Introduction

Glutamate is a major excitatory neurotransmitter in the central nervous system and it mediates its action through 2 distinct types of receptors: ionotropic and metabotropic (Pin & Duvosin, 1995). Ionotropic receptors are ion channels permeable to cations and are subdivided into N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors based on agonist preference (Nakanishi, 1994). Metabotropic glutamate receptors (mGluRs) are members of the G protein-coupled receptor (GPCR) superfamily and they have been shown to play an important role in processes requiring synaptic plasticity, such as learning and memory, neuronal development, and neurodegeneration (Nakanishi, 1994, Pin & Bockaert, 1995, Pin & Duvosin, 1995, Conn & Pin, 1997, Dale et al., 2003).

Eight distinct mammalian mGluRs have been reported that share 30–60% identity and have been divided into 3 subgroups based on sequence similarities, preferred signal transduction pathways and pharmacology (Nakanishi, 1994, Pin & Bockaert, 1995, Pin & Duvosin, 1995, Conn & Pin, 1997, Dale et al., 2003). They are Group I (mGluR1 and mGluR5), Group II (mGluR2 and mGluR3) and Group III (mGluR4, mGluR6, mGluR7, and mGluR8). This review focuses on Group I mGluRs. Group I mGluRs are positively coupled to phospholipase C (PLC) via Gαq/11 and inositol 1,4,5 trisphosphate (InsP3) formation (Abdul-Ghani et al., 1996). Their activation increases intracellular calcium levels by inducing calcium release from intracellular stores and stimulates protein kinase C (PKC) activation, potentiates L-type voltage-dependent calcium channels, and inhibits K+ conductances. These receptors also activate other G proteins, including Gαs and Gαi/o (Aramori & Nakanishi, 1992, Francesconi & Duvoisin, 2000). For example, mGluR1a, when transfected into CHO cells, stimulates both cAMP formation and arachidonic acid release (Aramori & Nakanishi, 1992), whereas mGluR5a does not couple with the stimulatory cAMP pathway (Abe et al., 1992). Group I mGluRs can also activate intracellular signals independent of G proteins, probably via Src (Heuss et al., 1999). Thus, these receptors can trigger a variety of signaling cascades and modulate the activity of ion and ligand-gated channels through functional coupling with transduction pathways such as PLC, adenylyl cyclase, phospholipase A2, phospholipase D (PLD), tyrosine kinases, extracellular signal-regulated protein kinases and mitogen-activated protein kinase kinase (Hermans & Challiss, 2001, Valenti et al., 2002).

Group I mGluRs are differentially distributed throughout the central nervous system. mGluR1 is highly expressed within CA3 pyramidal cells of the hippocampus, Purkinje cells of the cerebellum, the olfactory bulb, and thalamus. In contrast, mGluR5 is expressed in low levels in the cerebellum, but in high levels in the cortex, the striatum and nucleus accumbens of the basal ganglia, CA1 and CA3 pyramidal cells of the hippocampus and granule cells of the olfactory bulb (Bordi & Ugolini, 1999). There are also differences in the cortical distribution of Group I mGluRs among species, in particular between primates and rodents. The localization of mGluR1a to pyramidal cells in primate cortex contrasts with reports that mGluR1a is found almost exclusively in interneurons in rodent cortex (Muly et al., 2003, Kuwajima et al., 2004). Group I mGluRs are also found outside the brain in unmyelinated sensory afferent terminals in the skin, where they play an important role in pain sensation (Bhave et al., 2001). They are also found in melanocytes (Frati et al., 2000), osteoblasts (Gu & Publicover, 2000), heart cells (Gill et al., 1999), and hepatocytes (Storto et al., 2000). Although Group I mGluRs can be located at both presynaptic and postsynaptic sites in the brain, they primarily display a peri-synaptic localization at the postsynaptic membrane of glutamatergic neurons, where they often regulate neuronal excitability by modulating currents through ionotropic glutamate receptor channels (Shigermoto et al., 1993, Baude et al., 1997, Shigermoto et al., 1997).

Considering the variety of signal transduction pathways that can be activated by Group I mGluRs, it is not surprising that these receptors are involved in a large variety of physiological events. Group I mGluRs have been shown to play an important role in neuronal development (Catania et al., 1991, Plenz & Kitai, 1998, Flint et al., 1999, Hannan et al., 2001), synaptic plasticity (Zhong et al., 2000, Wu et al., 2001, Gubellini et al., 2003), neurodegeneration (Kingston et al., 1999, Bruno et al., 2001), and the induction of reactive astrocytes (Aronica et al., 2000). Although Group I mGluRs (mGluR1 and mGluR5) are highly homologous among themselves and are coupled to Gαq/11 proteins and phosphoinositide hydrolysis, they appear to fulfill different roles in the central nervous system (Valenti et al., 2002). Functional differences for mGluR1 and mGluR5 activity have been suggested by their distinct anatomical distribution, but have also been detected when they are coexpressed within the same cell. For example, activation of mGluR1 and mGluR5 in transfected cells generates oscillatory diacylglycerol, InsP3 and calcium responses at distinct frequencies that can be translated as oscillations in the activation of PKC (Dale et al., 2001a, Dale et al., 2001b, Babwah et al., 2003). These differences in oscillation frequency appear to be regulated by a single amino acid residue in the G protein-coupling domains of the mGluR1 (D854) and mGluR5 (T840) (Kawabata et al., 1996, Dale et al., 2001a, Dale et al., 2001b) (Fig. 1). The activation of mGluR5, but not of mGluR1 potentiates NMDA responses in striatal projection neurons (Pisani et al., 1997), whereas the presynaptic activation of mGluR1, but not of mGluR5, modulates GABAergic transmission and mediates neuroprotective effects against NMDA toxicity (Battaglia et al., 2001). Functional differences between mGluR1 and mGluR5 have also been reported in other cell types, including hippocampal CA1 pyramidal, globus pallidus, substantial-nigra pars reticulate, and midbrain dopaminergic neurons in the substantia nigra pars compacta and ventral tegmental area (Valenti et al., 2002).

The activation of Group I mGluRs has also been shown to play an important role in synaptic plasticity, which is defined as long- and short-term changes in the efficacy of synaptic transmission. Two classical forms of the synaptic plasticity long-term depression (LTD) and long-term potentiation (LTP) are observed in several brain regions and have been proposed to represent the cellular mechanisms for motor learning and behaviour. The expression of LTD has been shown to require the selective activation of mGluR1, as the amplitude of LTD is reduced in slices from mGluR1−/− mice (Conquet et al., 1994, Gubellini et al., 2001). The activation of Group I mGluRs also mediate the induction of corticostriatal LTP, as the blockade of both mGluR1 and mGluR5 can completely interfere with the induction of LTP (Gubellini et al., 2003). This is also supported by electrophysiological experiments utilizing mGluR1−/− and mGluR5−/− mice (Conquet et al., 1994, Chiamulera et al., 2001). Both groups of animals showed a reduced striatal LTP, but in different brain regions. In the hippocampus, mGluR1-deficient mice exhibit either normal or only slightly reduced CA1 and dentate LTP, but impaired CA3 mossy fiber LTP. These mice displayed a reduced performance in context-dependent fear conditioning tasks (Aiba et al., 1994). Conversely, mGluR5-deficient mice display a reduction in CA1 hippocampal LTP, but demonstrate normal CA3 mossy fiber LTP and have a reduced performance in spatial learning tasks (Lu et al., 1997). mGluR1-deficient mice display a reduced cerebellar LTD that is associated with a deficiency in eye blink reflex and motor coordination (Conquet et al., 1994, Gubellini et al., 2001). The role of Group I mGluRs on LTP has been proposed to be related to the enhancement of NMDA receptor-mediated responses through the activation of several calcium-dependent mechanisms (Calabresi et al., 1996, Dineley et al., 2001).

Altered glutamatergic transmission has been postulated as the pathophysiological basis of several neurological and neurodegenerative disorders, such as ischemia, brain trauma, chronic pain (Bhave et al., 2001, Karim et al., 2001, Zhou et al., 2001), epilepsy (Chapman et al., 2000), multiple sclerosis (Geurts et al., 2003), amyotropic lateral sclerosis (Laslo et al., 2001), diabetes (Berent-Spillson et al., 2003), Huntington's disease (Calabresi et al., 1999) and Parkinson's disease (Awad et al., 2000). In this context, the positive modulation of NMDA receptors by Group I mGluRs may lead to a massive influx of calcium ions and an enhanced glutamatergic calcium signal resulting in the selective loss of neurons involved in various diseases. However, it remains unknown whether the toxic effects of the glutamate receptors are the primary cause of the various pathological conditions or whether excessive glutamate levels are just consequences of the disorder leading to cell death. Nevertheless, the drugs targeted against the mGluRs (antagonists) are the focus of a number of therapeutic strategies for treating various neurological disorders.

Section snippets

Regulators of G protein signaling proteins

GPCR responsiveness is attenuated at the level of G proteins as well as at the level of receptor. At the level of G proteins, GPCR activity is attenuated by means of a family of proteins termed regulators of G protein signaling (RGS), which recognize specific members of G protein α subunits and accelerate the GTPase activity intrinsic to the α subunits of heterotrimeric G-proteins, thus playing crucial roles in the physiological control of G-protein signaling (Dohlman & Thorner, 1997,

β-Arrestin-dependent mGluR internalization

Group I mGluRs undergo rapid internalization after agonist exposure which is strongly inhibited by the expression of both β-arrestin- and dynamin dominant-negative mutants (Dale et al., 2001a, Dale et al., 2001b, Mundell et al., 2001) (Fig. 4). mGluR1a activation by glutamate increases GRK2/β-arrestin association with mGluR1a (Mundell et al., 2004) and promotes the membrane translocation of both β-arrestin1 and β-arrestin2 from cytosol to membrane, thereby contributing to mGluR1a

Summary

In conclusion, the present review overviews the multiple mechanisms by which the activity of Group I mGluRs can be regulated. Although Group I mGluRs utilize many of the same molecular intermediates as other more prototypical GPCR to effect their desensitization and endocytosis, these intermediates are utilized in a distinct manner. The desensitization of Group I mGluRs is mediated by a myriad of different second messenger-dependent protein kinases but the desensitization of these receptors can

Acknowledgments

G.D.K. is the recipient of a Canadian Institutes of Health Research (CIHR) studentship. S.S.G.F. is the recipient of a Canada Research Chair in Molecular Neuroscience and is a Heart and Stroke Foundation of Ontario Career Investigator. This work is supported by CIHR grants MA-15506 and CIHR grant MOP-62738 to S.S.G.F.

References (97)

  • C.V. Carman et al.

    Selective regulation of Gαq/11 by an RGS domain in the G protein-coupled receptor kinase, GRK2

    J Biol Chem

    (1999)
  • A.G. Chapman et al.

    Anticonvulsant activity of two metabotropic glutamate group I antagonists selective for the mGlu5 receptor: 2-methyl-6-(phenylethynyl)-pyridine (MPEP), and (E)-6-methyl-2-styryl-pyridine (SIB 1893)

    Neuropharmacology

    (2000)
  • F. Ciruela et al.

    Differential internalisation of mGluR1 splice variants in response to agonist and phorbol esters in permanently transfected BHK cells

    FEBS Lett

    (1997)
  • L.B. Dale et al.

    G protein-coupled receptor kinase-mediated desensitization of metabotropic glutamate receptor 1A protects against cell death

    J Biol Chem

    (2000)
  • L.B. Dale et al.

    Spatial-temporal dynamics of metabotropic glutamate receptor-mediated inositol 1,4,5-triphosphate, Ca2+, and protein kinase C oscillations

    J Biol Chem

    (2001)
  • G.K. Dhami et al.

    Phosphorylation-independent regulation of metabotropic glutamate receptor signaling by G protein-coupled receptor kinase 2

    J Biol Chem

    (2002)
  • G.K. Dhami et al.

    G Protein-coupled receptor kinase 2 regulator of G protein signaling homology domain binds to both metabotropic glutamate receptor 1a and Gαq to attenuate signaling

    J Biol Chem

    (2004)
  • G.K. Dhami et al.

    Phosphorylation-independent regulation of metabotropic glutamate receptor 1 signaling requires G protein-coupled receptor kinase 2 binding to the second intracellular loop

    J Biol Chem

    (2005)
  • H.G. Dohlman et al.

    RGS proteins and signaling by heterotrimeric G proteins

    J Biol Chem

    (1997)
  • L. Fourgeaud et al.

    The metabotropic glutamate receptor mGluR5 is endocytosed by a clathrin-independent pathway

    J Biol Chem

    (2003)
  • N.J. Freedman et al.

    Phosphorylation and desensitization of human endothelin A and B receptors. Evidence for G protein-coupled receptor kinase specificity

    J Biol Chem

    (1997)
  • R.W. Gereau et al.

    Role of protein kinase C phosphorylation in rapid desensitization of metabotropic glutamate receptor 5

    Neuron

    (1998)
  • S.S. Gill et al.

    Immunochemical localization of the metabotropic glutamate receptors in the rat heart

    Brain Res Bull

    (1999)
  • P. Gubellini et al.

    Selective involvement of mGlu1 receptors in corticostriatal LTD

    Neuropharmacology

    (2001)
  • P. Gubellini et al.

    Corticostriatal LTP requires combined mGluR1 and mGluR5 activation

    Neuropharmacology

    (2003)
  • P. Harjes et al.

    The hunt for huntingtin function: interaction partners tell many different stories

    Trends Biochem Sci

    (2003)
  • L. Iacovelli et al.

    Role of G protein-coupled receptor kinase 4 and β1 in agonist-stimulated metabotropic glutamate receptor 1 internalization and activation of mitogen-activated protein kinases

    J Biol Chem

    (2003)
  • P.J. Kammermeier et al.

    Expression of RGS2 alters the coupling of metabotropic glutamate receptor 1a to M-type K+ and N-type Ca2+ channels

    Neuron

    (1999)
  • W. Lu et al.

    Activation of synaptic NMDA receptors induces membrane insertion of new AMPA receptors and LTP in cultured hippocampal neurons

    Neuron

    (2001)
  • R. Minakami et al.

    Molecular cloninbg and the functional expression of two isoforms of human metaotropic receptor subtype 5

    Biochem Biophys Res Commun

    (1994)
  • S. Nakanishi

    Metabotropic glutamate receptors: synaptic transmission, modulation, and plasticity

    Neuron

    (1994)
  • M. Oppermann et al.

    Phosphorylation of the type 1A angiotensin II receptor by G protein-coupled receptor kinases and protein kinase C

    J Biol Chem

    (1996)
  • K. Palczewski et al.

    Identification of the N-terminal region in rhodopsin kinase involved in its interaction with rhodopsin

    J Biol Chem

    (1993)
  • J.P. Pin et al.

    Get receptive to metabotropic glutamate receptors

    Curr Opin Neurobiol

    (1995)
  • J.P. Pin et al.

    The metabotropic glutamate receptors: structure and functions

    Neuropharmacology

    (1995)
  • D.D. Schoepp et al.

    Selective inhibition of excitatory amino acid-stimulated phosphoinositide hydrolysis in the rat hippocampus by activation of protein kinase C

    Biochem Pharmacol

    (1988)
  • S.D. Sorensen et al.

    G protein-coupled receptor kinases regulate metabotropic glutamate receptor 5 function and expression

    Neuropharmacology

    (2003)
  • R. Sterne-Marr et al.

    Methods involved in the study of phosphorylation-independent regulation of G protein-coupled receptor signaling by GRK2

    Methods Enzymol

    (2004)
  • J. Zhong et al.

    Dual modulation of excitatory synaptic transmission by agonists at group I metabotropic glutamate receptors in the rat spinal dorsal horn

    Brain Res

    (2000)
  • S. Zhou et al.

    Metabotropic glutamate 1α receptors on peripheral primary afferent fibers: their role in nociception

    Brain Res

    (2001)
  • M.A. Abdul-Ghani et al.

    Metabotropic glutamate receptors coupled to IP3 production mediate inhibition of IAHP in rat dentate granule neurons

    J Neurophysiol

    (1996)
  • S. Alaluf et al.

    Agonist-mediated phosphorylation of metabotropic glutamate receptor 1a by protein kinase C in permanently by transfected BHK cells

    Biochem Soc Trans

    (1995)
  • E. Aronica et al.

    Upregulation of metabotropic glutamate receptor subtype mGluR3 and mGluR5 in reactive astrocytes in a rat model of mesial temporal lobe epilepsy

    Eur J Neurosci

    (2000)
  • H. Awad et al.

    Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus

    J Neurosci

    (2000)
  • A. Baude et al.

    The metabotropic glutamate receptor (mGluR1a) is concentrated at perisynaptic membrane of neuronal subpopulations as detected by immunogold reaction

    Neuron

    (1997)
  • A. Berent-Spillson et al.

    Protection against glucose-induced neuronal death by NAAG and GCP II inhibition is regulated by mGluR3

    J Neurochem

    (2003)
  • M. Bhattacharya et al.

    β-Arrestin regulates a RalGDS-Ral effector pathway mediating cytoskeletal reorganization

    Nat Cell Biol

    (2002)
  • M. Bhattacharya et al.

    Ral and phospholipase D2-dependent pathway for constitutive metabotropic glutamate receptor endocytosis

    J Neurosci

    (2004)
  • Cited by (169)

    • Glutamate receptor endocytosis and signaling in neurological conditions

      2023, Progress in Molecular Biology and Translational Science
    View all citing articles on Scopus
    View full text