Greenwood RS, Fan Z, McHugh R, Meeker R (2000) Inhibition of hippocampal kindling by .... Lafon-Cazal M, Fagni L, Guiraud MJ, Mary S, Lerner-Natoli M, Pin.
Journal of Cerebral Blood Flow and Metabolism 21:1013–1033 © 2001 The International Society for Cerebral Blood Flow and Metabolism Published by Lippincott Williams & Wilkins, Inc., Philadelphia
Review Article
Metabotropic Glutamate Receptor Subtypes as Targets for Neuroprotective Drugs *Valeria Bruno, *Giuseppe Battaglia, †Agata Copani, *Mara D’Onofrio, ‡P. Di Iorio, *Antonio De Blasi, §Daniela Melchiorri, 储Peter J. Flor, and *§Ferdinando Nicoletti *I.N.M. Neuromed, Pozzilli, †Department of Pharmaceutical Sciences, University of Catania, ‡Department of Biomedical Sciences, University of Chieti, and §Department of Human Physiology and Pharmacology, University of Rome “La Sapienza”, Italy; and 储Novartis Pharma AG, Nervous System Research, Basel, Switzerland
Summary: Metabotropic glutamate (mGlu) receptors have been considered as potential targets for neuroprotective drugs, but the lack of specific drugs has limited the development of neuroprotective strategies in experimental models of acute or chronic central nervous system (CNS) disorders. The advent of potent and centrally available subtype-selective ligands has overcome this limitation, leading to an extensive investigation of the role of mGlu receptor subtypes in neurodegeneration during the last 2 years. Examples of these drugs are the noncompetitive mGlu1 receptor antagonists, CPCCOEt and BAY36-7620; the noncompetitive mGlu5 receptor antagonists, 2-methyl-6-(phenylethynyl)pyridine, SIB-1893, and SIB-1757; and the potent mGlu2/3 receptor agonists, LY354740 and LY379268. Pharmacologic blockade of mGlu1 or mGlu5 receptors or pharmacologic activation of mGlu2/3 or mGlu4/7/8 receptors produces neuroprotection in a variety of in vitro or in vivo models. MGlu1 receptor antagonists are promising drugs for the treatment of brain ischemia or for the prophylaxis of neuronal damage induced by synaptic hyperactivity. MGlu5 receptor antagonists may limit neuronal damage induced by a hyperactivity of N-methyl-d-aspartate (NMDA) receptors, be-
cause mGlu5 and NMDA receptors are physically and functionally connected in neuronal membranes. A series of observations suggest a potential application of mGlu5 receptor antagonists in chronic neurodegenerative disorders, such as amyotrophic lateral sclerosis and Alzheimer disease. MGlu2/3 receptor agonists inhibit glutamate release, but also promote the synthesis and release of neurotrophic factors in astrocytes. These drugs may therefore have a broad application as neuroprotective agents in a variety of CNS disorders. Finally, mGlu4/7/8 receptor agonists potently inhibit glutamate release and have a potential application in seizure disorders. The advantage of all these drugs with respect to NMDA or AMPA receptor agonists derives from the evidence that mGlu receptors do not “mediate,” but rather “modulate” excitatory synaptic transmission. Therefore, it can be expected that mGlu receptor ligands are devoid of the undesirable effects resulting from the inhibition of excitatory synaptic transmission, such as sedation or an impairment of learning and memory. Key Words: mGlu receptors—Neuroprotection—Subtype-selective ligands.
Neurodegeneration is a common outcome of a variety of acute or chronic central nervous system (CNS) disorders, such as stroke, temporal lobe epilepsy, Parkinson disease, Alzheimer disease, Huntington disease, and the AIDS-dementia complex. In most of these disorders, the current therapy is symptomatic and does not prevent or arrest the ongoing neurodegeneration. Like many other
cells, neurons die by necrosis or apoptosis, and both phenotypes of death are commonly found in acute disorders such as stroke. Apoptotic death can develop as a direct consequence of the causative agent—such as the lack of oxygen and glucose in stroke or the presence of ß-amyloid fibrils in Alzheimer disease)—but also can be secondary to the loss of neurotrophic inputs normally released from neurons that primarily die. The coexistence of different processes of neuronal death must be outlined because the same therapeutic intervention that protects against necrotic death can aggravate apoptosis by trophic deprivation and vice versa (Lee et al., 1999).
Received May 23, 2001; final revision received June 18, 2001; accepted June 21, 2001. Address correspondence and reprint requests to Dr. Ferdinando Nicoletti, Dept. of Human Physiology and Pharmacology, University of Rome “La Sapienza,” Piazzale Aldo Moro, 5, 00185 Roma, Italy.
1013
1014
V. BRUNO ET AL.
The intracellular events underlying the execution phase of cell death have been extensively dissected and ultimately involve the activation of caspases. However, these processes are not cell-specific; therefore, they cannot be easily targeted by neuroprotective drugs. For example, a long-term neuroprotective therapy with a caspase inhibitor will impair the occurrence of “helpful” apoptosis in cells infected by viruses or harboring genomic damage. A better strategy is to intervene against extracellular insults that specifically contribute to neuronal death. An excessive activation of ionotropic glutamate (iGlu) receptors, particularly N-methyl-d-aspartate (NMDA) receptors, has been widely implicated in the pathophysiology of neuronal damage in most of acute and chronic neurodegenerative disorders (Choi, 1992; Doble, 1999). A number of NMDA or alpha-amino-3hydroxyl-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonists have been tested as neuroprotectants in experimental animals and humans. However, in spite of the promising results obtained in animal models, all clinical trials with iGlu receptor antagonists in stroke have been unsuccessful because of the follwing: 1) a narrow therapeutic window, 2) the occurrence of undesirable side effects, and 3) lack of efficacy (Lee et al., 1999). The impairment of fast excitatory synaptic transmission caused by iGlu receptor antagonists is certainly responsible for side effects such as sedation, ataxia, and memory loss, and also might contribute to the lack of efficacy of these drugs. According to the hypothesis of a dual role of Ca2+ in cell survival (Johnson et al., 1992), neuronal death may be triggered not only by a Ca2+ overload, but also by the reduction of free cytosolic Ca2+ below a critical threshold. This threshold is set by trophic factors produced by neighboring neurons or glial cells. A selective blockade of iGlu receptors therefore may deprive susceptible neurons from the amount of Ca2+ that could compensate for the loss of neurotrophic agents. Therefore, it can be expected that iGlu receptor antagonists protect neurons that die from an excessive increase in cytosolic free Ca2+, but facilitate apoptotic death in neurons that no longer receive neurotrophic input. There are two strategies to overcome this limitation. First, one can try to limit the intracellular consequences of an excessive activation of iGlu receptors without impairing the “physiologic effects” of receptor activation. Examples are provided by the receptor abuse–dependent antagonists (RADA drugs), such as melatonin or the gangliosides GM1 and GT1b, which were proven to be protective against excitotoxic neuronal death (Manev et al., 1990a,b, 1997; Costa et al., 1994). Second, neuroprotective drugs can target specific categories of membrane receptors that “modulate” rather than mediate excitatory synaptic transmission but are nonetheless implicated in the pathophysiology of neuronal damage. Metabotropic glutamate (mGlu) receptors meet these criteria (Nicoletti et al., 1996) and will be discussed in this review. J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
STRUCTURAL AND FUNCTIONAL FEATURES OF mGlu RECEPTORS MGlu receptors belong to family 3 of G-protein– coupled receptors, which also include gamma aminobutyric acid type B receptor (GABA-B), Ca2+-sensing, and some olfactory, pheromone, and taste receptors. The extracellular domain of mGlu receptors harbors the agonist binding site and is structurally related to bacterial periplasmic amino acid binding proteins (O’Hara et al., 1993). X-ray crystallographic studies of the mGlu1 receptor support the original hypothesis (O’Hara et al., 1993) that glutamate binds to a cleft separating two globular lobes, switching the equilibrium toward the closed conformation of the two lobes. In addition, the receptor acts as a dimer in such a way that the closure of one of the two extracellular domains resulting from glutamate binding produces a drastic change in conformation that reduces the distance of the two C-terminus domains of the promoters (Kunishima et al., 2000). The second intracellular loop is the least conserved among the mGlu receptor subtypes and is critical for the selectivity of G-protein coupling, whereas the third intracellular loop is mostly involved in G protein activation and controls the coupling efficacy in cooperation with the first loop and the C-terminal tail (De Blasi et al., 2001). mGlu receptors form a family of 8 subtypes (mGlu1 to mGlu8), which are subdivided into 3 groups on the basis of structural homology, pharmacologic profile, and transduction pathways (Fig. 1). Group-I includes mGlu1 (splice variants: mGlu1a, -b, -c, -d, -e) and mGlu5 (splice variants: mGlu5a and -b) receptors, which are coupled to polyphosphoinositide (PI) hydrolysis (Tanabe et al., 1992; Minakami et al., 1994; Joly et al., 1995; Laurie et al., 1996; Flor et al., 1996; Schoepp et al., 1999). Activation of these receptors generates inositol-1,4,5trisphoshate (InsP3) and diacylglycerol, which releases intracellular Ca2+ and activates protein kinase C (PKC), respectively (Fig. 1). However, in transfected cells, activation of mGlu1a receptors produces a single peaked intracellular Ca2+ response, whereas activation of mGlu5 receptors generates an oscillatory increase in intracellular Ca2+. The latter property relies on the presence of a threonine residue just distal to the seventh TM domain of the mGlu5 receptor, which is phosphorylated by PKC (Kawabata et al., 1996). Whether or not this difference is physiologically relevant is unclear. Native mGlu1 and mGlu5 receptors also can negatively modulate a variety of K+ channels and activate inward cationic currents (Pin and Duvoisin, 1995; Chuang et al., 2000), with the net effect of increasing neuronal excitability. MGlu5 receptors also are expressed in astrocytes and microglia (Biber et al., 1999). Group-II mGlu receptors include mGlu2 and mGlu3 (Flor et al., 1995a; Emile et al., 1996), which are coupled to Gi proteins in heterologous expression
mGlu RECEPTORS AND NEUROPROTECTION
1015
FIG. 1. General features of metabotropic glutamate (mGlu) receptors. PLC, phospholipase C; PKC, protein kinase C; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; PDE, phosphodiesterase.
systems. Activation of these receptors reduces cyclic adenosine monophosphate formation (Fig. 1), but also can produce pleiotropic effects mediated by the betagamma subunits of the G protein, such as the activation of mitogen-activated protein kinase and the phosphatidylinositol-3-kinase (PI-3-K) pathways (Ferraguti et al., 1999). Both receptor subtypes are preferentially localized in the preterminal region of axons, where their activation attenuates glutamate release. mGlu3 receptors, however, are widely expressed on astrocytes, where their functional role is becoming increasingly important (see below). Group-III mGlu receptors include the subtypes mGlu4 (splice variants: mGlu4a and -b), mGlu6, mGlu7 (splice variants mGlu7a and -b), and mGlu8 (splices variants: mGlu8a and -b), which are all coupled to Gi proteins in transfected cells (Okamoto et al., 1994; Flor et al., 1995b; 1997; Duvoisin et al., 1995; Corti et al., 1997). mGlu4, and mGlu7 and mGlu8 receptors are all presynaptically localized, and their activation inhibits the release of glutamate or GABA. mGlu6 receptors are exclusively expressed by ON bipolar cells in the retina, and are physiologically involved in the amplification of visual inputs. In these cells, mGlu6 receptors are coupled to a cyclic guanosine monophosphate (cGMP) phospho-
diesterase (Fig. 1) (Nakanishi, 1994). There is no evidence of mGlu receptor localization in oligodendrocytes. RECENT ADVANCES IN THE PHARMACOLOGY OF mGlu RECEPTORS mGlu can be pharmacologically differentiated from iGlu with the agonist 1S,3R-1-amino-cyclopentane-1,3dicarboxylic acid (1S,3R-ACPD), which, however, is not subtype-selective and activates both group-I and group-II mGlu receptors (Schoepp et al., 1999) (Table 1). Group-I mGlu receptors can be selectively activated by 3,5dihydroxyphenylglycine (DHPG) or 3-hydroxyphenylglycine (3-HPG), which cannot readily discriminate between mGlu1 and mGlu5 receptors. 2-Chlorohydroxyphenylglycine (CHPG) selectively activates mGlu5 receptors, but is less potent than DHPG or 1S,3R-ACPD (Schoepp et al., 1999). The phenylglycine derivatives alpha-methyl-4-carboxyphenylglycine (MCPG), 4-carboxyphenylglycine (4CPG), and 4-carboxy-3hydroxyphenylglycine (4C3HPG) have been widely used to antagonize group-I mGlu receptors with a certain degree of selectivity. All three drugs preferentially antagonize mGlu1 over mGlu5 receptors, but also can recruit J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1016
V. BRUNO ET AL. TABLE 1. Subtype-selective agonists and antagonists of mGlu receptors Drug
Potency (mol/L)
3,5-DHPG (Ag.) 3-HPG (Ag.) AIDA (C Ant.) LY367385 (C. Ant.) CPCCOEt (NC Ant.) BAY36-7620 (NC Ant.) 3,5-DHPG (Ag.) 3-HPG (Ag.) CHPG (Ag.) MPEP (NC Ant.) SIB-1757 (NC Ant.) SIB-1893 (NC Ant.) 2R,4R-APDC (Ag.) LY354740 (Ag.) LY379268 (Ag.) DCG-IV (Ag.) NAAG (Ag.) ADED (Ant.) ADBD (Ant.) LY341495 (Ant.) L-AP4 (Ag.) SOP (Ag.) (R,S)-PPG (Ag.) MSOP (Ant.) MAP4 (Ant.) L-AP4 (Ag.) SOP (Ag.) (R,S)-PPG (Ag.) L-AP4 (Ag.) SOP (Ag.) (R,S)-PPG (Ag.) L-AP4 (Ag.) SOP (Ag.) (R,S)-PPG (Ag.) MAP4 (Ant.)
6–374 18–600 3.4–214 7 4.76–23 0.16 2–20 14–35 750 0.032 0.4 0.3 0.3–10 0.005–0.1 0.003–0.014 0.06–0.1 65 6.1–18 30–50 0.014–0.03 0.2–1 1–2 5.2 19 36–190 0.2–0.9 0.4–3 4.7 160–1300 31–1600 185 0.06–0.9 0.3–2 0.2 25
Subtype mGlu1
mGlu5
mGlu2/3
mGlu4
mGlu6 mGlu7 mGlu8
Comment Group-I selective weak mGlu2 agonist weak mGlu2 agonist mGlu1-selective mGlu1-selective mGlu1-selective mGlu5-selective mGlu5-selective mGlu5-selective mGlu5-selective Group-II selective weaker mGlu6/8 agonist weaker mGlu6/8 agonist mGlu4/6/7/8 ant., NMDA agonist mGlu3-selective weaker mGlu1/5/6/7/8 agonist Group-III selective, mGlu2 ant. Group-III selective Group-III selective mGlu2 ant.
Ag., agonist; Ant., antagonist; C Ant., competitive antagonist; NC Ant., noncompetitive antagonist; DHPG, 3,5-dihydroxyphenylglycine; 3-HPG, 3-hydroxyphenylglycine; AIDA, (R,S)-1-aminoindan-1,5-dicarboxylic acid; LY367385, 2-methyl-4-carboxyphenylglycine; CPCCOEt, 7-(Hydroxyimino)cyclopropa[b]chromen-1acarboxylate ethyl ester; BAY36-7620, [(3aS,6aS)-6a-Naphtalen-2-ylmethyl-5-methyliden-hexahydrocyclopental[c]furan-1-on]; CHPG, 2-chlorohydroxyphenylglycine; MPEP, 2-methyl-6-(phenylethynyl)pyridine; SIB-1757, 6-methyl-2-(phenylazo)pyridin-3-ol; SIB-1893, (E)-2-methyl-6-stryrylpyridine; 2R,4R-APDC, 2R,4R-4-aminopyrrolidine-2,4-dicarboxylic acid; LY354740, (1S,2S,5R,6S)-(+)-2-aminobicyclo[3.1.0]hexane2,6-dicarboxylic acid; LY379268, (−)-2-oxa-4-aminocyclo[3.1.0]hexane-4,6-dicarboxylic acid; DCG-IV, (2S,2⬘R,3⬘R)-2-(2⬘,3⬘-dicarboxycyclopropyl)glycine; NAAG, N-acetylaspartylglutamate; ADED, (2S,4S)-2amino-4-(2,2-diphenylethyl)pentane-1,5-dioic acid; ADBD, (2S,4S)-2-amino-4-(4,4-diphenylbut-1-yl)pentane1,5-dioic acid; LY341495, (2S,1⬘S,2⬘S)-2-(9-xanthylmethyl)-2-(2⬘-carboxycyclopropyl)glycine; L-AP4, L-2amino-4-phosphonobutanoate; SOP, L-serine-O-phosphate; (R,S)-PPG, (+)-4-phosphonophenylglycine; MSOP, (RS)-␣-methylserine-O-phosphate; MAP4, (S)-␣-methyl-2-amino-4-phosphonobutanoic acid.
mGlu2 and mGlu3 receptors (MCPG as an antagonist, 4C3HPG as an agonist). Novel subtype-selective antagonists are now available that help to efficiently discriminate effects produced by the activation of mGlu1 and mGlu5 receptors. The compounds 2-methyl-4carboxyphenylglycine (LY367385), (R,S)-1aminoindan-1,5-dicarboxylic acid (AIDA), and (S)-(+)2-(3⬘-carboxybicyclo[1.1.1]pentyl)-glycine (CBPG) behave as competitive mGlu1 antagonists (CBPG is also an mGlu5 receptor agonist), whereas (2S,1⬘R,2⬘R)-2-(9xanthylmethyl)-2-(2⬘-carboxycyclopropyl)glycine (LY344545) selectively antagonize mGlu5 receptors. 7-(Hydroxyimino)cyclopropa[b]chromen-1a-carboxylate J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
ethyl ester (CPCCOEt) and [(3aS,6aS)-6a-Naphtalen-2ylmethyl-5-methyliden-hexahydro-cyclopental[c]furan1-on] (BAY36-7620) are nonamino acid molecules acting as noncompetitive antagonists of mGlu1 receptors with a high degree of selectivity (in the low micromolar range) (Annoura et al., 1996; Litschig et al., 1999; Carroll et al., 2001). Interestingly, BAY36-7620 can cross the blood–brain barrier; therefore, it is suitable for in vivo studies (Carroll et al., 2001). A series of phenylpyridine derivatives, including 2-methyl-6-(phenylethynyl)pyridine (MPEP), (E)-2-methyl-6-stryrylpyridine (SIB1893), and 6-methyl-2-(phenylazo)pyridin-3-ol (SIB1757), behave as highly potent and selective noncom-
mGlu RECEPTORS AND NEUROPROTECTION petitive mGlu5 receptor antagonists, and at least MPEP is proven to be centrally available (Gasparini et al., 1999b; Spooren et al., 2001). mGlu2 and mGlu3 receptors can be activated by 2R,4R-4-aminopyrrolidine-2,4dicarboxylic acid (2R,4R-APDC), which is so far the most selective agonist at these receptor subtypes. Last generation agonists of mGlu2/3 receptors include the Lilly compounds (1S,2S,5R,6S)-(+)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY354740), (−)-2oxa-4-aminocyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY379268), and (−)-2-thia-4-aminobicyclo[3.1.0] hexane-4,6-dicarboxylic acid (LY389795), which are highly potent (Ki value in the low nanomolar range) and are systemically available. (2S,2⬘R,3⬘R)-2-(2⬘,3⬘dicarboxycyclopropyl)glycine (DCG-IV) and its congener (2S,1⬘S,2⬘S)-2-(carboxycyclopropyl)glycine (LCCG-I) have been widely used as mGlu2/3 receptor agonists. However, DCG-IV also can activate NMDA receptors and behaves as a group-III mGlu receptor antagonist, whereas L-CCG-I also can activate mGlu1, -5, and -8 receptors (Brabet et al., 1998; Schoepp et al., 1999). N-acetylaspartylglutamate (NAAG) is an interesting compound because it shows selectivity for mGlu3 over mGlu2 receptors (Wroblewska et al., 1997). mGlu2 and mGlu3 receptors can be antagonized by a series of compounds, of which (2S,4S)-2-amino-4-(2,2diphenylethyl)pentane-1,5-dioic acid (ADED) and (2S,4S)-2-amino-4-(4,4-diphenylbut-1-yl)pentane1,5-dioic acid (ADBD) (Schoepp et al., 1999) show the highest degree of selectivity. (2S,1⬘S,2⬘S)-2-(9xanthylmethyl)-2-(2⬘-carboxycyclopropyl)glycine (LY341495) shows a high potency in antagonizing mGlu2/3 receptors, but can recruit all other receptor subtypes at micromolar concentrations (Schoepp et al., 1999). Finally, all group-III mGlu receptors are activated by phosphonoamino acids, such as L-2-amino-4phosphonobutanoate (L-AP4, the prototypic ligand) and (+)-4-phosphonophenylglycine (PPG), as well as by Lserine-O-phosphate (L-SOP), which is an endogenous compound. All these compounds show a greater affinity for mGlu4, -6, and -8 than for mGlu7. PPG has a greater affinity for mGlu8 than for mGlu4 and mGlu6 receptors (Gasparini et al., 1999a; 2000). Subtype-selective antagonists of group-III mGlu receptors are lacking. (S)-␣methyl-2-amino-4-phosphonobutanoic acid (MAP4) and (RS)-␣-methylserine-O-phosphate (MSOP) are generally used to inhibit mGlu4 receptors (Jane et al., 1994; Thomas et al., 1996). ROLE OF GROUP-I mGlu RECEPTORS IN NEURODEGENERATION/ NEUROPROTECTION Two issues will be addressed in this section: 1) how pharmacologic activation of group-I mGlu receptors affects excitotoxic neurodegeneration, and (2) how endog-
1017
enous activation of mGlu1 or mGlu5 receptors affects neurodegeneration in a variety of cellular or animal models of CNS disorders. As a corollary of the second point, the authors will comment on the potential use of subtypeselective mGlu1 or mGlu5 receptor antagonists in acute or chronic neurodegenerative disorders. Pharmacologic activation of group-I mGlu receptors in neurodegeneration/neuroprotection As activation of mGlu1 or mGlu5 receptors produces excitatory effects in neurons (Pin and Duvoisin, 1995), it is reasonable to assume that these receptors facilitate the induction and/or progression of excitotoxic neuronal death. Early in vivo experiments with 1S,3R-ACPD, which also can activate mGlu2 and -3 receptors, are consistent with this view. 1S,3R-ACPD locally injected into the rat hippocampus or caudate nucleus produces neurodegeneration that requires the endogenous activation of NMDA receptors in the adult, but exclusively depends on intracellular Ca2+ mobilization in neonates (McDonald and Schoepp, 1992; Sacaan and Schoepp, 1992; McDonald et al., 1993). Accordingly, the efficacy of mGlu receptor agonists to stimulate PI hydrolysis is much greater in brain slices from newborn than from adult animals (Nicoletti et al., 1986). In more recent in vivo studies, DHPG has shown neurotoxic and proconvulsant activities when injected intracerebroventricularly (Ong and Balcar, 1997; Camon et al., 1998; Merlin and Wong, 1997; Burke and Hablitz, 1996). The in vitro counterpart of these data has been obtained by using mixed cultures of mouse cortical cells, in which neurons are grown over a monolayer of confluent astrocytes. In this model, a brief pulse with NMDA produces delayed neuronal death that can be detected by assessing trypan blue staining or extracellular lactate dehydrogenase activity 20 to 24 hours after the pulse. 3HPG, DHPG, or quisqualate (a mixed agonist of group-I mGlu and AMPA receptors) coapplied with submaximal concentrations of NMDA amplify excitotoxic neuronal death (Buisson and Choi, 1995; Bruno et al., 1995b). Similar results are obtained with CHPG (Bruno et al., unpublished). Interestingly, at least DHPG and quisqualate retain their neurotoxic effects when applied immediately after the NMDA pulse and maintained in the medium afterwards (Bruno et al., 1995b). Other studies, however, unequivocally show that group-I mGlu receptor activation produces neuroprotection rather than neurotoxicity in a variety of in vitro models, including cultured cerebellar granule cells challenged with glutamate (Pizzi et al., 1993, 1996a, 1999), cultured spinal motor neurons challenged with kainate (Pizzi et al., 2000) or brain slices challenged with excitotoxins or oxygen-glucose deprivation (Opitz and Reymann, 1993; Pizzi et al., 1996b; Schroeder et al., 1999). Possible explanations for these contrasting results have been discussed in a recent review (Nicoletti et al., 1999) J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1018
V. BRUNO ET AL.
and include: (i) the heteromeric composition of NMDA receptors; (ii) the existence of an “activity-dependent switch” from facilitatory to inhibitory group-I mGlu receptors; and (iii) the absence or presence of astrocytes expressing mGlu5 receptors. Two additional aspects will be discussed, i.e. the role of mGlu5 receptors in supporting the survival of developing neurons; and the activitydependent changes in the subcellular expression of mGlu5 receptors. The presence of the NR2C subunit in NMDA receptors of mature cultured cerebellar granule cells has been considered as a major determinant for the neuroprotective response to group-I mGlu receptor agonists. Accordingly, 3HPG is highly protective against glutamate toxicity in control cultured granule cells, but not in cells treated with NR2C antisense oligonucleotides (Pizzi et al., 1999). This is an interesting observation, particularly because the NR2C subunit is highly expressed in the human brain (Lin et al., 1996; Daggett et al., 1998). The lack of the NR2C in the substantia nigra could predispose nigral neurons to excitotoxic damage. An additional explanation (which is not mutually exclusive with the NR2C hypothesis) is that pharmacologic activation of group-I mGlu receptors may either facilitate or attenuate excitotoxic neuronal death depending on the functional state of the receptors. Dynamic changes in the activity of group-I mGlu receptors have been described by J. Sanchez-Prieto and his collaborators (Herrero et al., 1992, 1994, 1998; Sistiaga et al., 1998, 2000), who examined the modulation of glutamate release in corticostriatal synaptosomes. In this preparation, a first application of group-I mGlu receptor agonists facilitates glutamate release, whereas a second application (within a restricted timeframe) inhibits release. This “functional switch” in receptor activity involves receptor phosphorylation by PKC, similarly to what happens during classical receptor desensitization, and depends on changes in the G-protein coupling. The same phenomenon has been observed by recording excitatory postsynaptic currents (EPSCs) in CA1 pyramidal cells exposed to DHPG (Rodriguez-Moreno et al., 1998). The authors examined whether the model of the functional switch also can be applied to excitotoxic neuronal death. In mixed cortical cultures, DHPG amplifies NMDA toxicity when applied only once (either before or during the NMDA pulse), but becomes neuroprotective when applied for the second time shortly after a brief preexposure. Neuroprotection is lost when less than 45 minutes are allowed between the first and the second exposure to DHPG, or when DHPG is applied for the first time in combination with PKC inhibitors or mGlu5 receptor antagonists. The release of endogenous glutamate stimulated by the NMDA pulse in culture also is amplified by a unique application, but is reduced by two consecutive applications of DHPG (Bruno et al., 2001). Thus, one expects that the final effect of group-I mGlu receptor agonists on excitotoxic J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
neuronal death is related to the functional state of group-I mGlu receptors, which depends on how frequently these receptors are activated by endogenous glutamate. The authors believe that this dual regulation of excitotoxic neuronal death is exclusively performed by presynaptic mGlu receptors (perhaps coinciding with mGlu5 receptors) regulating glutamate release (Fig. 2). Although classical electron microscopy studies show that mGlu1 and mGlu5 receptors are preferentially localized in the peripheral portions of the postsynaptic densities (Baude et al., 1993; Lujan et al., 1997; Shigemoto et al., 1997), evidence for a presynaptic localization of group-I mGlu receptors is emerging (PJ Conn, unpublished observation), and the possibility exists that mGlu5 receptors may be targeted to nerve endings depending on the availability of particular variants of Homer proteins (see below). The final effect of group-I mGlu receptor agonists on neuronal death may also critically depend on the presence and functional state of glial cells. Interestingly, reactive astrocytes in the injured brain tissue, as well as their equivalent in culture (that is, astrocytes cultivated in chemically defined medium deprived of serum and enriched in endothelial growth factor or transforming growth factor-alpha (TGF-alpha)) express high levels of functional mGlu5 receptors (Miller et al., 1993). The physiologic significance of this phenomenon is unknown, but one can predict that activation of mGlu5 receptors by the endogenously released glutamate controls some aspects related to the responsiveness of reactive astrocytes to neuronal injury. Interestingly, DHPG (which is otherwise neuroprotective) amplifies excitotoxicity when applied to cultured granule cells on which a coverslip with a monolayer of confluent astrocytes has been flipped (G Barbagallo and MV Catania, unpublished observation). Among the putative neurotoxic factors released by astrocytes in response to mGlu5 receptor activation, attention should be focused on nitric oxide. Notably, the neuronal isoform of constitutive nitric oxide synthase (nNOS) is expressed in cultured astrocytes grown in chemically defined medium (Nicoletti et al., 1999) and reactive astrocytes in the spinal cord of individuals who have died from amytrophic lateral sclerosis express high amounts of the beta and gamma variants of nNOS (Catania et al., 2001). Interestingly, pharmacologic activation of mGlu5 receptors protects neurons against apoptosis by trophic deprivation. Copani et al. (1998) have shown that in granule cells grown under suboptimal conditions—that is, in medium containing 10 rather than 25 mmol/L K+)—the onset of apoptotic death coincides with the developmental decline in mGlu5 receptors, and cells are rescued by the addition of group-I mGlu receptor agonists. In addition, pharmacologic activation of group-I mGlu receptors is protective in cortical or hippocampal neurons deprived of trophic support (Sagara and Schubert, 1998); hence,
mGlu RECEPTORS AND NEUROPROTECTION
1019
crosslinking-capable isoforms of Homer—that is, Homer-1b, -1c, -2 and -3—can allow mGlu receptor clustering and a functional coupling of mGlu1 or mGlu5 receptors with other cell receptors, ion channels, or cytoskeleton proteins (Brakeman et al., 1997; Tu et al., 1999; Kammermeier et al., 2000; Fagni et al., 2000; Minakami et al., 2000). Homer-1a, the short homer isoform that is encoded by an early gene switched on by synaptic hyperactivity, is unable to form crosslinking with other Homer proteins and can act as a negative dominant by disrupting mGlu receptor clusters (Xiao et al., 2000). A number of studies show that Homer proteins critically affect the cell surface targeting of mGlu receptors (Ciruela et al., 2000; Ango et al., 2000). In cultured granule cells, for example, cotransfection of mGlu5 receptors with Homer-1a disrupts receptor clustering but allows the axonal targeting of the receptor (Ango et al., 2000). Thus, synaptic hyperactivity (with ensuing induction of Homer-1a) may substantially affect the subcellular distribution of group-I mGlu receptors, and therefore the functional response to mGlu1 or mGlu5 receptor agonists. All these factors make the final response to a group-I mGlu receptor agonist highly unpredictable and rule out a possible clinical application for these drugs in neurodegenerative disorders. This pessimistic view is strengthened by the low central bioavailability of group-I mGlu receptor agonists and by their propensity to induce seizures. FIG. 2. (A) Functional switch in the modulation of glutamate release by presynaptic group-I metabotropic glutamate “mGlu” receptors. Activation of “naïve” mGlu receptors enhances glutamate release, whereas activation of “experienced” receptors inhibits release. This mechanism also has been implicated to explain the dual regulation of N-methyl-d-aspartate (NMDA) toxicity by group-I mGlu receptor agonists (Bruno et al., 2001). (B) Activation of mGlu1 receptors inhibits gamma aminobutyric acid (GABA) release. This mechanism explains why neuroprotection by mGlu1 receptor antagonists is affected by drugs that interfere with GABAergic transmission. (C) Modulation of NMDA receptors by mGlu5 receptors depends on the specific expression of NMDA receptor subunits. The presence of the NR2C or NR2D subunit prevents the positive modulation of NMDA receptors by PKC (see text). PLC, phosphodiesterase C.
one expects that the final effect of mGlu5 receptor agonists on neuronal death depends on the proportion of neurons that primarily die in response to the toxic insult and those that die because of the lack of trophic inputs. Obviously, this is impossible to predict in human pathology. Finally, the diversity in the response to group-I mGlu receptor agonists may depend on the subcellular localization of mGlu1 or mGlu5 receptors in a particular neuronal population. Group-I mGlu receptors interact with members of a family of synaptic proteins named Homer, which bind to a proline-rich motif of the C-terminus of mGlu receptors through their PDZ domain. The longer
Effect of endogenous activation of group-I mGlu receptors on neurodegeneration: mGlu1 and mGlu5 receptor antagonists as neuroprotective agents mGlu1 receptor antagonists and neuroprotection. In spite of the contrasting effects of group-I mGlu receptor agonists, mGlu1 receptor antagonists are always neuroprotective, indicating that endogenous activation of mGlu1 receptors contributes to the induction and/or progression of neuronal death. Both competitive and noncompetitive mGlu1 receptor antagonists (such as AIDA, LY367385, 4-carboxypheylglycine, and CPCCOEt) (Pellicciari et al., 1995; Clark et al., 1997; Watkins and Collingridge, 1994; Litshig et al., 1999) reduce excitotoxic neuronal death in mouse cortical cultures challenged with NMDA (Strasser et al., 1998; Bruno et al., 1999; Battaglia et al., 2001) and also are effective in protecting striatal neurons against NMDA or quinolinic acid toxicity (Bruno et al., 1999; Orlando et al., 2001; Battaglia et al., 2001). Neuroprotection by LY367385, AIDA, and CBPG also is observed in models that are closer to human pathology, such as oxygen glucose deprivation in mouse cortical cultures or rat organotypic hippocampal cultures, and the 4-vessel occlusion model in gerbils (Pellegrini-Giampietro et al., 1999a,b; Bruno et al., 1999). Notably, no mGlu receptor ligands have J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1020
V. BRUNO ET AL.
been proven to be effective in animal models of permanent or transient focal ischemia. Interestingly, mGlu1 receptor expression is reduced after ischemic preconditioning in gerbil hippocampus (Sommer et al., 2000). Recent studies have gained new insights into the mechanism whereby mGlu1 receptor antagonists attenuate neuronal death. mGlu1 receptors are expressed in GABAergic neurons, and their activation depresses inhibitory synaptic transmission (Baude et al., 1993; Gereau and Conn, 1995; Morishita et al., 1998; Bushell et al., 1999; Paquet and Smith, 2000); hence, Battaglia et al. (2001) have hypothesized that mGlu1 receptor antagonists are neuroprotective by enhancing GABA release (Fig. 2). This hypothesis is supported by the following observations: 1) neuroprotection by LY367385 or CPCCOEt against NMDA toxicity in culture is obliterated by GABA or SKF89976A (an inhibitor of GABA transporter) and is prevented by bicuculline and 2-hydroxysaclophen, which antagonize GABAA and GABAB receptors, respectively; 2) the same cocktail of GABA receptor blockers prevents the protective activity of CPCCOEt against excitotoxic death of striatal neurons in in vivo experiments; 3) toxic concentrations of NMDA can stimulate GABA release in the caudate nucleus of freely moving rats only if LY367385 or CPCCOEt are present in the perfusate; and 4) in cortico-striatal slices, DHPG depresses GABAergic IPSCs through the activation of mGlu1 receptors (Battaglia et al., 2001). This contrasts with the protective action of mGlu5 receptor antagonists, which does not involve a GABAergic component (Battaglia et al., 2001). Based on these findings, one expects that mGlu1 receptor antagonists are particularly helpful in the treatment of CNS disorders that are primarily caused by an impairment of inhibitory synaptic transmission, such as temporal lobe epilepsy associated with Ammon horn sclerosis and mossy fiber sprouting. Under these particular conditions, seizure activity results from the development of recurrent excitatory synapses within neighbouring dentate gyrus granule cells that overcome the control of inhibitory GABAergic synapses (McNamara, 1999). Seizure activity causes further damage of granule cells, thus generating a vicious cycle that might be interrupted by enhancing GABA release with mGlu1 receptor antagonists. Interestingly, the expression of mGlu1a receptors is up-regulated in the molecular layer of the dentate gyrus of patients with temporal lobe epilepsy, as well as in the dentate gyrus of kainatetreated and kindled rats (Blumcke et al., 2000). The relevance of mGlu1 receptors in the pathophysiology of temporal lobe epilepsy is strengthened by the evidence that DHPG induces long-lasting epileptiform discharges in the hippocampus (Merlin, 1999; Galoyan and Merlin, 2000), whereas mGlu1 antisense oligonucleotides are protective against hippocampal kindling (Greenwood et al., 2000). In addition, the mGlu1 receptor antagonists J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
LY367385 and AIDA reduce sound-induced seizures in DBA/2 mice and in genetically epilepsy prone rats, and exert antiepileptic activity in lethargic mice (Chapman et al., 1999). A potential drawback associated with the use of mGlu1 receptor antagonists is the induction of ataxia. mGlu1a receptors are located in the dendritic spines of Purkinje cells, which receive excitatory inputs from both parallel and climbing fibers (Nusser et al., 1994). MGlu1 knockout mice are ataxic and do not develop long-term depression (LTD) at the parallel fiber-Purkinje cell synapses (Aiba et al., 1994). In two patients in remission from Hodgkin disease, paraneoplastic ataxia has been associated to neutralizing autoantibodies directed against mGlu1a receptors; injection of these antibodies in mice induces reversible ataxia (Sillevis Smitt et al., 2000). In vivo studies with centrally available mGlu1 receptor antagonists are necessary to establish whether pharmacologic inhibition of mGlu1 receptors impairs motor coordination in experimental animals. mGlu5 receptor antagonists and neuroprotection. The noncompetitive mGlu5 receptor antagonists— MPEP, SIB-1893, and SIB-1757—are currently used as powerful tools to examine how endogenous activation of mGlu5 receptors affects neurodegeneration and neuronal function in general. However, the recent report that relatively high concentrations of MPEP or SIB-1893 (20 or 100 mol/L) reduce NMDA currents in cultured cortical neurons (O’Leary et al., 2000; Movsesyan et al., 2001) casts doubts on the usefulness of these pharmacologic agents, and therefore deserves some comments. In oocytes expressing NR1 and NR2A or NR2B subunits, in which no partnership exists between NMDA and mGlu5 receptors, MPEP has no effect on NMDA currents. A slight inhibition (22% of controls) is only observed in cells expressing NR1 + NR2B subunits, but this effect is not statistically significant (Gasparini et al., 1999b). Neither SIB-1893 nor SIB-1757 has any significant antagonistic activity at recombinant NMDA receptors (Varney et al., 1999). The inhibitory effects of MPEP or SIB1893 on NMDA currents in cortical neurons might reflect different regulatory properties of native versus recombinant NMDA receptors, or more likely, the existence of the functional link between NMDA and mGlu5 receptors (see below). One cannot exclude that inhibition of NMDA currents by MPEP and SIB-1893 derives from their ability to antagonize mGlu5 receptors that are endogenously activated by the glutamate released extracellularly or trapped in the plasma membrane. The authors can safely rely on experiments in which MPEP or SIB1893 is used at concentrations less than 20 mol/L, although it is their belief that even at 20 mol/L the two drugs predominantly, if not exclusively, antagonize mGlu5 receptors.
mGlu RECEPTORS AND NEUROPROTECTION MPEP, SIB-1757, and SIB-1893 are highly potent (substantial activity in the nanomolar range) in reducing NMDA toxicity in mixed cultures of cortical cells, and relatively low doses of MPEP (5 to 20 nmol) are protective against NMDA or quinolinic acid toxicity when locally injected into the rat caudate nucleus (Bruno et al., 2000b). To strengthen the specificity of this effect, it is noteworthy that iso-MPEP (an isomer of MPEP that does not interact with mGlu5 receptors) fails to protect against NMDA toxicity (Bruno et al., 2000b). Neuroprotection by MPEP in vitro and in vivo is not affected by GABAergic drugs (Battaglia et al., 2001) and can be ascribed to the ability of the drug to disrupt the functional coupling between mGlu5 and NMDA receptors. A growing body of evidence indicates that mGlu5 receptors positively modulate NMDA receptors (Awad et al., 2000; Ugolini et al., 1999; Salt and Binns, 2000; Doherty et al., 2000; Jia et al., 1998; Pisani et al., 1997), and that the two receptors are coexpressed in most of the neurons. The lack of both mGlu5 and NMDA receptors in Purkinje cells supports this view. A chain of anchoring proteins may physically link NMDA and mGlu5 receptors. The NR2 subunits of NMDA receptors interact with a series of “membrane-associated guanlyl kinase” (MAGUK) proteins (such as PSD-95, PSD-93, and SAP102), which, through their PDZ domains, may assemble with the SHANK proteins. These proteins can interact with the long isoforms of Homer proteins, thus linking NMDA receptors with mGlu5 receptors (Tu et al., 1999). One mechanism by which mGlu5 receptors positively modulate NMDA receptors is mediated by the formation of diacylglycerol with ensuing activation of PKC. Protein kinase C can phosphorylate NMDA receptors, thus relieving the Mg2+ blockade of the NMDA-gated ion channel (Chuang et al., 2000). This modulation mediated by PKC has some structural requirements. In recombinant cells, only NMDA receptors containing the NR2A or NR2B subunits are positively modulated by PKC, whereas receptors containing the NR2C or NR2D subunits are not (Kutsuwada et al., 1992; Mori et al., 1993) (Fig. 2). The Ca2+ released from intracellular stores through the InsP3-sensitive channels (that also bind to Homer proteins) can induce changes in NMDA receptor activity after binding to calmodulin. Ca2+-calmodulin can compete with alfa-actinin-2 for a common binding site on the NR1 subunit, thus affecting the anchorage of NMDA receptors to the cytoskeleton (Niethammer et al., 1996; Wyszynski et al., 1997; Dunah et al., 2000). The interaction between NMDA and mGlu5 receptors is reciprocal. Activation of NMDA enhances mGlu5 receptor responses by preventing the homologous desensitization of mGlu5 receptors (De Blasi et al., 2001). This effect is mediated by calcineurin, which is activated by Ca2+ and dephosphorylates mGlu5 receptors at a PKC phosphorylation site (Alagarsamy et al., 1999). Thus, a vicious
1021
cycle exists in which NMDA and mGlu5 receptors potentiate each other. This particular form of receptor crosstalk has important implications in synaptic plasticity and in neurotoxicity. The expression of long-term potentiation is mediated by an increased activity of AMPA receptors, which results, at least in part, from the phosphorylation of Ser 831 of the GluR1 subunit by PKC and CaM kinase II (Scannevin and Huganir, 2000). A coactivation of mGlu5 and NMDA receptors might be required for a sustained activity of PKC and CaM kinase II, and, therefore, for the induction of a long-lasting, nondecremental form of long-term potentiation (Jia et al., 1998). A similar scenario can be proposed for the induction and progression of excitotoxic damage (Fig. 3). In cultured cortical cells challenged with NMDA, for example, it is known that the amount of glutamate released during and after the NMDA pulse contributes to the development of neuronal damage (Monyer et al., 1992). One can speculate that the combined activation of NMDA and mGlu5 receptors leads to the phosphorylation of AMPA receptors, and that activation of “hyperactive” AMPA receptors by the endogenous glutamate facilitates the progression of excitotoxic damage. Pharmacologic blockade of mGlu5 receptors during the induction phase of excitotoxic death would “isolate” NMDA receptors from its natural partner, thus reducing the strength of the excitotoxic insult below the death threshold. If this model is correct, then one expects that mGlu5 receptor antagonists are highly effective as neuroprotective agents in all CNS disorders in which activation of NMDA receptors is critically involved in the pathophysiology of neurodegeneration. Focal ischemia is one of these disorders, but no data on the effect of mGlu5 receptor antagonists are currently available. This may simply suggest that results are not encouraging. In the gerbil model of transient global ischemia, which is more refractory to NMDA receptor antagonists, MPEP is shown to be more effective than AIDA in increasing neuronal survival (Rao et al., 2000). However, this is not confirmed in another study in which only mGlu1 receptor antagonists and not MPEP are neuroprotective in ischemic gerbils and in cultured neurons subjected to oxygen–glucose deprivation (D. Pellegrini-Giampietro and F. Moroni, unpubished observation). The idea that mGlu5 receptor antagonists are promising drugs in the treatment of stroke is met with skepticism because of the heterogeneity of neuronal death, which includes necrosis (and/or apoptosis) directly caused by the shortfall in oxygen and glucose supply, and apoptosis secondary to the loss of synaptic inputs or neurotrophic factors (Lee et al., 1999). Knowing that activation of mGlu5 receptors supports neuronal survival during early development (Copani et al., 1998), it is easy to predict that mGlu5 receptor antagonists are detrimental for neurons that develop apoptosis by trophic deprivation in response to brain ischJ Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1022
V. BRUNO ET AL.
FIG. 3. Speculative model on the interaction between N-methyl-d-aspartate (NMDA) and metabotropic glutamate 5 (mGlu5) receptors in the induction of excitotoxic damage (from Soderling and Derkach, 2000). The two receptors are physically linked by a chain of anchoring proteins, and act synergistically to activate a number of Ca2+-regulated proteins, such as Ca2+/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), and type-I adenylyl cyclase (AC-I). This leads to the phosphorylation of specific Ser residues in the GluR1 subunit of alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, thus increasing the activity of AMPA receptors. The possibility that AMPA receptors lose the GluR2 subunit (Pellegrini-Giampietro et al., 1992) or move from the cytosol to the membrane also is considered. Activation of AMPA receptors by endogenous glutamate may contribute to the progression of excitotoxic damage. ER, endoplasmic reticulum; cAMP, cyclic adenosine monophosphate; PKA, protein kinase A; DAG, diacylglycerol.
emia (Allen et al., 2000). One also should take into account that mGlu5 receptors are expressed in endothelial cells (Lin and Maiese, 2001; Morley et al., 1998), where their physiologic role is still unknown. Clearly, more studies are needed to clarify this issue. The authors believe that mGlu5 receptor antagonists may be beneficial during those conditions in which a toxic insult homogeneously impairs the survival of a specific neuronal population (that is, when the component of apoptosis by trophic deprivation is lacking). This is typified by toxicologic parkinsonism induced by 1-methyl4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or by some amphetamine derivatives (such as metamphetamine), which selectively destroy the nigro-striatal dopaminergic pathway. The authors recently have found that systemically administered MPEP or SIB-1893 (but not CPCCOEt) is protective against degeneration of striatal dopaminergic terminals produced by three consecutive injections of metamphetamine in mice. MPEP also can reduce the formation of reactive oxygen species induced by metamphetamine in the striatum of freely moving mice without interfering with the uptake of metamphetamine into dopaminergic terminals (G. Battaglia et al., manuscript in preparation). This raises the intriguing possibility that endogenous activation of mGlu5 receptors contributes to J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
the oxidative damage of the nigro-striatal dopaminergic pathway in response to amphetamine derivatives. mGlu5 receptor antagonists also can act as symptomatic drugs in experimental models of nigro-striatal degeneration. Activation of mGlu5 receptors induces direct excitation, and selectively potentiates NMDA currents in neurons of the subthalamic nucleus (Awad et al., 2000). As an increased activity of the subthalamic nucleus is implicated in the pathophysiology of bradykinesia, one can predict that mGlu5 receptor antagonists can improve motor symptoms resulting from the loss of nigro-striatal dopaminergic fibers. It would be interesting to study the efficacy of mGlu5 antagonists in the MPTP model of parkinsonism in mice or primates. The evidence that activation of mGlu5 receptors enhances NMDA responses in striatal medium spiny neurons (Pisani et al., 2000) and restores NMDA toxicity in the striatum of decorticated animals (Orlando et al., 2001) suggests a potential application for mGlu5 receptor antagonists in the experimental treatment of Huntington chorea. However, the role of excitotoxic mechanisms in the pathophysiology of Huntington disease (DiFiglia, 1990) has been questioned by the unexpected finding that transgenic mice carrying an expanded CAG repeat in the Huntington gene are less sensitive to kainic acid tox-
mGlu RECEPTORS AND NEUROPROTECTION icity (Hansson et al., 1999). It is certainly interesting to examine whether an early treatment with a centrally available mGlu5 receptor antagonist (like MPEP or SIB1893) delays the onset of neurologic symptoms and reduces the neuropathologic hallmarks of the disease in these animals. Whether or not mGlu5 receptor antagonists may be beneficial in the experimental treatment of experimental amyotrophic lateral sclerosis (ALS) is currently unknown. Immunocytochemical analysis shows that spinal cord motoneurons express mGlu1 but not mGlu5 receptors (Alvarez et al., 2000), and this may explain the resistance of this particular neuronal type to NMDA toxicity. However, mGlu5 receptors may still be linked to the pathophysiology of ALS because they are heavily expressed on reactive astrocytes, which, in a mouse model of ALS, play an active role in motoneuron degeneration (Levine et al., 1999). Catania et al. (2001) have shown that reactive astrocytes in the ventral horns and white matter of autoptic spinal cord samples from sporadic or familiar ALS patients express high levels of nNOS. Activation of mGlu5 receptors in reactive astrocytes might provide a potential source for the Ca2+-stimulated formation of NO, thus contributing to neuronal damage through a process of glial-neuronal interaction. Finally, studies performed on cortical cultures show that MPEP, SIB-1893, and SIB-1757 are highly potent against neurotoxicity induced by the active fragment of ß-amyloid peptide (fragment 25-35) (Bruno et al., 2000b). These experiments are performed in the presence of a cocktail of NMDA and AMPA receptor antagonists (MK-801 and DNQX), thus providing a model in which NMDA receptors do not contribute to neuronal death. Interestingly, inhibition of mGlu1 receptors by AIDA does not reduce but rather exacerbates ß-amyloid toxicity in cortical cells (Allen et al., 1999). Although the mechanism(s) underlying the neuroprotective effect of mGlu5 receptor antagonists is unknown, this evidence encourages the use of MPEP or its congeners in experimental animal models of Alzheimer disease, such as mice carrying a double mutation in the amyloid precursor protein and presenilin-1 genes. Role group-II mGlu receptors in neuroprotection mGlu2/3 receptor agonists are protective in experimental paradigms of neurodegeneration. Activation of mGlu2/3 receptors produces inhibitory effects mediated by a reduction in intracellular cyclic adenosine monophosphate levels or by a decreased activity of voltagegated Ca2+ channels (Pin and Duvoisin, 1995). This has encouraged the search for potent and selective agonists of potential use as neuroprotective agents. The search led to the discovery of a number of compounds, such as LY354740 and LY379268, which are potent and sys-
1023
temically active. LY354740 is currently in clinical trials but, curiously, it has been indicated for the treatment of anxiety rather than for the treatment of stroke or other neurodegenerative disorders (Helton et al., 1998; Klodzinska et al., 1999). The recent finding that mGlu3 receptors expressed by astrocytes control the production of putative neurotrophic agents (such as nerve growth factor or TGF-ß) leads to a serious reconsideration of these particular receptor subtypes in the therapy of neurodegenerative disorders. Initial studies on the role of mGlu2/3 receptors in neurodegeneration were performed in in vitro models—mixed cortical cultures, cultures of mesencephalic neurons, and cultures of cerebellar granule cells—using “old generation” agonists, such as DCGIV, 4C3HPG, L-CCG-I, and 1S,3R-ACPD (Pizzi et al., 1993; Ambrosini et al., 1995; Bruno et al., 1994, 1995a, Buisson et al., 1995; Thomsen et al., 1996). In cortical cultures, these drugs were neuroprotective against neurotoxicity induced by a brief NMDA pulse and, interestingly, they retained their activity when applied up to 1 hour after the NMDA pulse (Bruno et al., 1995a). The authors have shown that group-II mGlu receptor agonists also are protective against neuronal toxicity induced by a prolonged exposure to kainic acid (Bruno et al., 1995a), although in another study this effect was restricted to the small subpopulation of cultured neurons that respond to kainate with an enhanced influx of Co2+ (Gottron et al., 1995). More recently, neuroprotection studies have been extended to “second generation” mGlu2/3 receptor agonists, such as 2R,4R-APDC, LY354740, LY379268, and LY389795. All these drugs were proven to protect neurons against NMDA toxicity in culture (Battaglia et al., 1998; Kingston et al., 1999a,b; Behrens et al., 1999; D’Onofrio et al., 2001). However, the relatively low potency of LY354740 and LY379268 in the study of Behrens et al. (1999) led to the suggestion that neuroprotection was mediated by the activation of mGlu4 or mGlu8 receptors, which can be recruited by micromolar concentrations of both compounds (Schoepp et al., 1999). An alternative explanation is that native mGlu2/3 receptors have a lower affinity to LY354740 or LY379268 or that some unknown factor limits the access of these compounds to mGlu2/3 receptors in cortical cells. Initially, it was believed that mGlu2/3 receptor agonists were neuroprotective by reducing the release of endogenous glutamate. It was consistent with this hypothesis that an enhanced glutamate release occurring during or after the NMDA pulse contributes to the progression of neuronal damage (Monyer et al., 1992). However, although DCGIV could reduce the release of glutamate in cultured cortical cells challenged with NMDA, this effect was lost when DCG-IV was applied 1 hour after the NMDA pulse, although the drug was still neuroprotective (Bruno et al., 1995a). The idea that some additional mechanisms could take place was supported by the evidence that the J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1024
V. BRUNO ET AL.
protein synthesis inhibitor, cycloheximide, prevented neuroprotection by DCG-IV in cultured cortical cells (Bruno et al., 1997). Unexpectedly, cycloheximide was interfering with a novel form of glial–neuronal interaction that involved mGlu3 receptors present in astrocytes (Petralia et al., 1996). The medium collected from cultured astrocytes challenged with any group-II mGlu receptor agonist (including the mGlu3-selective agonist, NAAG) was highly protective when transferred to cultured neurons challenged with NMDA (Bruno et al., 1997; Wroblewska et al., 1997). This protection was abolished when protein synthesis in astrocytes was inhibited by cycloheximide or when the glial medium was heated (Bruno et al., 1997). Thus, it appeared that activation of glial mGlu3 receptors promoted a novel form of glial–neuronal interaction mediated by the synthesis and release of a putative neuroprotective factor (Fig. 4). This is fully consistent with more recent evidence that LY354740 and LY379260 are much more effective against excitotoxic death in mixed cultures than in pure neuronal cultures (Kingston et al., 1999b). Different factors including nerve growth factor and protein S-100ß (Ciccarelli et al., 1999) are released from astrocytes in response to mGlu3 receptor agonists, but neuroprotection appears to be mediated by the synthesis and release of TGF-ß. Application of neutralizing anti–TGF-ß1 or anti–
TGF-ß2 antibodies prevents neuroprotection induced by mGlu2/3 receptor agonists in mixed cultures and abolishes the protective activity of the glial medium. In addition, mGlu2/3 receptor agonists (including LY379268) enhance the de novo synthesis and release of TGF-ß1 and TGF-ß2 in cultured astrocytes (Bruno et al., 1998; D’Onofrio et al., 2001). The transduction pathway activated in response to glial mGlu3 receptors does not involve a reduction in cyclic adenosine monophosphate formation (as one would expect), but rather activation of the mitogen-activated protein (MAP) kinase and the PI3-kinase pathways. Inhibitors of these pathways (that is, the compounds PD98059 and LY294002) reduce the ability of LY379268 to enhance the de novo synthesis of TGF-ß in astrocytes and to protect neurons grown in mixed cultures against excitotoxic death (D’Onofrio et al., 2001). This mechanism has been confirmed in in vivo studies, where LY379268 enhances the expression of TGF-ß1 in the corpus striatum and cerebral cortex and protects striatal neurons against NMDA toxicity through a mechanism that involves the activation of the mitogenactivated protein kinase pathway (D’Onofrio et al., 2001). Mitogen-activated protein kinases, P90rsk and P70S6k, which regulate gene expression and protein synthesis, could represent the downstream effectors of the de novo synthesis of TGF-ß (Fig. 4). The evidence that glial
FIG. 4. The neuroprotective effect of group-II mGlu receptors is mediated by glial metabotropic glutamate 3 (mGlu3) receptors and requires glial production of transforming growth factor- (TGF-), through the activation of mitogen-activated protein kinase (MAPK) and PI-3-K pathways. TGF- released from astrocytes may exert its neuroprotective effect by an autocrine/paracrine mechanism on the glial cell inducing an overexpression of the serpin PAI-1 (plasminogen activator inhibitor type 1), which is then released from the astrocyte (left panel), and by acting in neurons through a set of high affinity serine-threonine kinases (TGF-ßRI and TGF-ßRII), which signal to the nucleus through the activation of SMAD2/3 and SMAD4 transcriptional factors or through the activation of transforming growth factor kinase 1 (TAK1). Downstream events include the induction of cell cycle proteins (p15, p21, and p27) or the inhibition of cyclooxygenase-2 (COX-2) expression. Other factors, still unknown, might contribute to TGF-–induced neuroprotection (right panel).
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
mGlu RECEPTORS AND NEUROPROTECTION mGlu3 receptors regulate the production of TGF-ß extends the potential application of group-II mGlu receptor agonists to multiple forms of neuronal degeneration. Tumor growth factor-ß is protective against neuronal degeneration induced by excitotoxins, oxygen-glucose deprivation, ß-amyloid peptide, and the HIV capside protein gp 120 (Chao et al., 1994; Prehn et al., 1994, 1996; Copani et al., 1995a,b, 1999; Henrich-Noak et al., 1996; Meucci and Miller, 1996; Prehn and Miller, 1996; Tomoda et al., 1996; Ren et al., 1997; Bruno et al., 1998; Flanders et al., 1998; Ruocco et al., 1999; Scorziello et al., 1997). Its mechanism of action includes induction of serpins (Buisson et al., 1998; Docagne et al., 1999), induction of cell-cycle inhibitors (Datto et al., 1995; Copani et al., 1999), and inhibition of cyclooxygenase-2 (Pruzanski et al., 1998) (Fig. 4). Finally, the existence of this particular form of glial–neuronal interaction may explain the unexpected protective activity of mGlu2/3 receptor agonists against neuronal apoptosis induced by staurosporine (Buisson et al., 1995; Kingston et al., 1999a), or by ß-amyloid peptide in the presence of ionotropic glutamate receptor antagonists (Copani et al., 1995a). Interestingly, the latter effect is seen in mixed cultures of cortical cells, but not in cultured cerebellar granule cells, where only few astrocytes are present (Copani et al., 1995a). mGlu2/3 receptor agonists as potential drugs in acute and chronic neurodegeneration. Because of their particular mechanism of action, mGlu2/3 receptor agonists become serious candidates as neuroprotective agents. First, these drugs produce a “delayed rescue” effect in culture (Bruno et al., 1995a), which suggests a favourable therapeutic window in the treatment of brain ischemia. Second, the mechanism mediated by the release of neurotrophic factors extends the potential spectrum of these drugs to most of the acute and chronic neurodegenerative disorders, and may circumvent the major limitation associated with the systemic injection of neurotrophic factors, that is, the low penetration across the blood–brain barrier. Third, the impact of these drugs on fast excitatory synaptic transmission should be minimal because presynaptic mGlu2/3 receptors are less important than group-III mGlu receptors in the regulation of glutamate release, and glial mGlu3 receptors are not involved in the regulation of synaptic transmission. Finally, glial mGlu3 receptors appear to be localized in the vascular side of glial cell membranes (Shigemoto R, unpublished observation); therefore, they should not be saturated by the synaptically released glutamate. How are these predictions met by current evidence in in vivo models of neuronal degeneration? The first evidence that activation of mGlu2/3 receptors is neuroprotective in vivo was provided by a series of studies performed in the laboratory of Dr. H. Shinozaki in Japan. Intracerebroventricular injection of DCG-IV
1025
was shown to protect vulnerable neurons against kainate toxicity (Miyamoto et al., 1994). This finding was remarkable because DCG-IV also can activate NMDA receptors, albeit at greater concentrations (Ishida et al., 1993). Data with “second generation” agonists on in vivo excitotoxicity are controversial. D’Onofrio et al. (2001) have shown that LY379268 protects striatal neurons against NMDA toxicity when injected locally or systemically, whereas no effect of LY354740 on striatal NMDA toxicity was reported by Behrens et al. (1999). A greater potency of LY379268 at native mGlu2/3 receptors or, more likely, the different doses of drugs used in the two experiments (10 nmol of LY354740 vs. 25 nmol of LY379268) may account for this difference. The effect of mGlu2/3 receptor agonists on ischemic neuronal damage has been studied in animal models of global or focal ischemia. In the gerbil model of global ischemia, intracerebroventricular injection of 4C3HPG protects hippocampal CA1 neurons when administered before or immediately after the induction of ischemia (HenrichNoack et al., 1998). This drug, however, also behaves as a mGlu1 receptor antagonist (see above). Systemically injected LY354740 (Bond et al., 1998) or LY379268 (Bond et al., 1999) also are neuroprotective in the ischemic gerbil and in a neonatal rat model of hypoxia– ischemia (Cai et al., 1999). The protective activity of LY379268 against ischemic degeneration of CA1 pyramidal cells in gerbils is not mediated by an increase in the expression of TGF-ß (Bond et al., 1999, 2000). Interestingly, the hippocampus is the only brain region examined where local infusion of mGlu2/3 receptor agonists does not enhance the expression of TGF-ß, raising the possibility that the response of astrocytes to these drugs is region-specific (D’Onofrio et al., 2001). LY354740 and LY379268 do not reduce the infarct size in rat models of focal ischemia, that is, electrocoagulation, (Lam et al., 1998) and endothelin-1–induced (Bond et al., 1999) middle cerebral artery occlusion. This casts doubt on the possible efficacy of mGlu2/3 receptor agonists in the experimental therapy of stroke. Recently, group-II mGlu receptors have been proposed as targets for the therapy of Parkinson disease. Overactive glutamatergic afferents from the subthalamic nucleus could cause both an excitotoxic loss of dopaminergic neurons in the substantia nigra and hyperactivation of GABAergic neurons in the globus pallidus, which leads to a reduction of motor activity (Albin et al., 1989). Drugs that can reduce glutamate release by activating presynaptic mGlu2/3 receptors might at one time delay the degeneration of substantia nigra neurons and improve motor activity (Konieczny et al., 1999; Wolfarth et al., 2000). No studies have been performed on the efficacy of systemically active mGlu2/3 receptor agonists in animal models of Huntington disease. However, the observation of an early reduction of mGlu2 receptors in the striatum J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1026
V. BRUNO ET AL.
of transgenic mice carrying expanded CAG repeats in the Huntington gene (Cha et al., 1998) raises the intriguing possibility that the loss of these receptors contributes to the pathophysiology of neuronal damage in Huntington chorea. The evidence that mGlu2/3 receptor agonists protect cultured spinal motoneurons against kainate toxicity (Pizzi et al., 2000) encourages the use of these drugs in animal models of amyotrophic lateral sclerosis, such as transgenic mice carrying mutations of superoxide dismutase. Group-II mGlu receptor agonists are protective against seizures in experimental animal models of epilepsy. Microinjections of 4C3HPG and 1S,3S-ACPD into the inferior colliculus reduce sound-induced seizures in genetically epilepsy-prone rats (Tang et al., 1997). Systemic injection of LY354740 exerts protective activity against seizures induced by ACPD, pentylenetetrazole, or picrotoxin (Monn et al., 1997; Klodzinska et al., 1999, 2000; Suzuki et al., 1999). It would be interesting to examine whether activation of mGlu2/3 receptors protects against neuronal damage that is secondary to neuronal hyperactivity, as observed in models of temporal lobe epilepsy. GROUP-III mGlu RECEPTOR AGONISTS AS NEUROPROTECTIVE DRUGS The pharmacology of group-III mGlu receptors (mGlu4, -6, -7, and -8) is less well developed than the pharmacology of the other subtypes because of the requirement for an omega-phosphonic group in agonist molecules, such as L-AP4 or PPG (see above). In addition, the strong acidic moiety limits the penetration of the drugs across the blood–brain barrier, and no centrally available agonists or antagonists have been developed so far. Thus, most of the available information derives from in vitro studies or from the effect of intracerebral injections of group-III mGlu receptor agonists. Group-III mGlu receptor agonists protect cultured cortical cells and cultured cerebellar granule cells against excitotoxic death (Bruno et al., 1995a, 1996, 2000a; Gasparini et al., 1999a; Lafon-Cazal et al., 1999) and apoptosis induced by the active fragment of ß-amyloid peptide (Copani et al., 1995a). In addition, group-III mGlu receptor agonists are protective against “mechanical injury” in neuronal cultures (Faden et al., 1997). In mixed cortical cultures challenged with NMDA, neuroprotection by L-AP4 or PPG is mediated by mGlu4 receptors, because it is no longer observed in cultures prepared from mGlu4 knockout mice (Bruno et al., 2000a). Interestingly, cultures from mGlu4 knockout mice are more sensitive to NMDA toxicity and release greater amounts of glutamate under basal conditions and in response to NMDA. High concentrations of PPG, which can recruit mGlu7 receptors, reduce NMDA-stimulated glutamate release in cultures lacking mGlu4 receptors, but never less than the levels J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
observed in wild-type cultures challenged with NMDA (Bruno et al., 2000a). These data suggest that mGlu4 receptors are critically involved in the homeostatic regulation of glutamate release, whereas mGlu7 receptors are recruited only in response to high levels of extracellular glutamate. In vivo studies support a role for mGlu4 receptors in the control of glutamate release and excitotoxic neuronal death. mGlu4 receptor knockout mice are refractory to the protective activity of low doses of PPG coinjected with NMDA into the caudate nucleus. In addition, these animals show a much greater stimulation of striatal glutamate release by NMDA in microdialysis studies (Bruno et al., 2000a). Thus, mGlu4 receptor agonists should effectively prevent the component of excitotoxic neuronal death mediated by an excessive release of glutamate. Other possible mechanisms underlying neuroprotection include the inhibition of NMDA responses by postsynaptic group-III mGlu receptors (Martin et al., 1997) and the modulation of nitric oxide formation (Maiese et al., 1995, 2000). (R,S)-PPG improves the functional recovery of CA1 pyramidal cells of hippocampal slices subjected to a severe hypoxic/hypoglycemic insult (Henrich-Noak et al., 2000; Sabelhaus et al., 2000), providing evidence that is indicative of a protective activity against ischemic brain damage. Studies on experimental animal models of transient or focal ischemia await the synthesis of potent and centrally available receptor agonists. Interestingly, activation of group-III mGlu receptors supports the survival of cultured cerebellar granule cells undergoing apoptosis by trophic deprivation (Graham and Burgoyne, 1994). This effect cannot be explained by any of the mechanisms outlined above, and suggests that activation of group-III mGlu receptors supports neuronal survival through some additional intracellular mechanisms. The authors recently have found that activation of group-III mGlu receptors stimulates the PI-3-kinase pathway in cultured cerebellar granule cells (Iacovelli et al., manuscript in preparation). The PI-3-kinase pathway has an established role in supporting cell survival and is activated in response to neurotrophic agents (such as insulin-like growth factor-I) that prevent apoptosis of cultured cerebellar granule cells (D’Mello et al., 1997). Whether this intracellular response is exclusive of groupIII mGlu receptors expressed on cultured granule cells or can be extended to other neuronal types remains to be established. Similar to group-I antagonists or group-II agonists, group-III mGlu receptor agonists exert anticonvulsant activity (Abdul-Ghani et al., 1997; Tang et al., 1997; Chapman et al., 1999; Gasparini et al., 1999a; Claudio et al., 2000). Although these effects are common to many drugs, a growing body of evidence suggests that mGlu4 receptors are directly involved in the pathophysiology of epilepsy (Lie et al., 2000).
mGlu RECEPTORS AND NEUROPROTECTION
1027
TABLE 2. Possible applications of subtype-selective mGlu receptor agonists or antagonists in human pathology Subtype
Drug
Drug mechanism
mGlu1
Antagonist
↑ GABA release
mGlu5
Antagonist
↓ NMDA currents
mGlu2/3
Agonist
mGlu4/7/8
Agonist
↓ Glutamate release Glial–neuronal interaction ↓ Glutamate release
Clinical application Ischemia Ammon horn sclerosis Toxicologic Parkinsonism? Huntington disease, ALS, Alzheimer disease Broad Epilepsy
mGlu, metabotropic glutamate; GABA, gamma aminobutyric acid; NMDA, N-methyl-D-aspartate; ALS, amyothrophic lateral sclerosis.
Pentylenetetrazol-induced seizures lead to an increase in (3H)-L-AP4 binding in the cerebral cortex (Thomsen, 1999), whereas electrical kindling is associated with a lower ability of group-III mGlu receptor agonists to inhibit transmission at the lateral perforant path–dentate gyrus synapses (Klapstein et al., 1999). Human studies show that mGlu4 receptors are up-regulated in dentate granule cells and CA4 neurons in surgical hippocampal specimen obtained from patients with temporal lobe epilepsy (Lie et al., 2000), whereas a reduced function of L-AP4–sensitive mGlu receptors is observed in the epileptic sclerotic hippocampus (Dietrich et al., 1999). An attractive hypothesis is that seizure vulnerability of distinct neuronal populations is regulated by the levels of expression and function of mGlu4 receptors. A reduced activity of mGlu4 receptors may predispose vulnerable neurons to excitotoxic degeneration, thus contributing to the development of Ammon horn sclerosis. Interestingly, mGlu4 knockout mice show an increase in seizure susceptibility, and the human mGlu4 receptor gene falls within a susceptible focus for juvenile myoclonic epilepsy in the chromosome 6, band p21.3 (Wong et al., 2001). mGlu7 and mGlu8 also have been implicated in the pathophysiology of epilepsy. Although no association between polymorphisms in genes encoding mGlu7 and mGlu8 receptors and idiopathic generalized epilepsy has been found so far (Goodwin et al., 2000), mGlu7 knockout mice show increased seizure susceptibility (H. van der Putten, personal communication). CONCLUSIONS Based on recent studies with subtype-selective ligands, the authors can comment on which mGlu receptor subtype should be targeted by neuroprotective drugs in different acute or chronic neurodegenerative disorders (Table 2). For all of the reasons outlined above, mGlu1/5 receptor agonists should not be used as neuroprotective agents. mGlu1 receptor antagonists have a potential application in the treatment of brain ischemia or might be used for the prophylaxis of neuronal damage induced by synaptic hyperactivity, as occurs in temporal lobe epilepsy. Ataxia may represent a serious side effect of these drugs. mGlu5 receptor antagonists should be particularly
indicated in all conditions in which excitotoxicity resulting from a sustained activation of NMDA receptors contributes to neuronal death. The impairment of apoptosis by trophic deprivation may limit the clinical efficacy of these drugs in focal ischemia. In the authors’ opinion, mGlu5 receptor antagonists may be beneficial in the experimental treatment of chronic neurodegenerative disorders, such as Huntington chorea, ALS, or Alzheimer disease (see above). Inhibition of glial mGlu5 receptors could contribute to the neuroprotective activity of these drugs in chronic disorders. mGlu2/3 receptor agonists induce neuroprotection through two distinct mechanisms—the inhibition of glutamate release and the production of neurotrophic factors (such as TGF-ß) by astrocytes. The latter mechanism suggests a broad application for these drugs in neurodegenerative disorders and may allow circumventing the major limitation associated with the peripheral administration of neurotrophic factors, that is, their poor penetration across the blood–brain barrier. mGlu4/7/8 receptor agonists induce neuroprotection by reducing glutamate release. These drugs have a potential application in the experimental treatment of seizure disorders. mGlu4 and mGlu7 receptors seem to be seriously implicated in the pathophysiology of epilepsy. REFERENCES Abdul-Ghani AS, Attwell PJ, Singh-Kent N, Bradford HF, Croucher MJ, Jane DE (1997) Anti-epileptogenic and anticonvulsant activity of L-2-amino-4-phosphonobutyrate, a presynaptic glutamate receptor agonist. Brain Res 755:202–212 Aiba A, Kano M, Chen C, Stanton ME, Fox GD, Herrup K, Zwingman TA, Tonegawa S (1994) Deficient cerebellar long-term depression and impaired motor learning in mGluR1 mutant mice. Cell 79:377–388 Alagarsamy S, Marino MJ, Rouse ST, Gereau IV RW, Heinemann SF, Conn PJ (1999) Activation of NMDA receptors reverses desensitization of mGluR5 in native and recombinant systems. Nature Neurosci 2:234–240 Albin RL, Young AB, Penney JB (1989) The functional anatomy of basal ganglia disorders. Trends Neurosci 12:366–375 Allen JW, Eldadah BA, Faden AI (1999) Beta-amyloid-induced apoptosis of cerebellar granule cells and cortical neurons: exacerbation by selective inhibition of group-I metabotropic glutamate receptors. Neuropharmacology 38:1243–1252 Allen JW, Knoblach SM, Faden AI (2000) Activation of group I metabotropic glutamate receptors reduces neuronal apoptosis but increases necrotic cell death in vitro. Cell Death Differ 7:470–476 Alvarez FJ, Villalba RM, Carr PA, Grandes P, Somohano PM (2000)
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1028
V. BRUNO ET AL.
Differential distribution of metabotropic glutamate receptors 1a, 1b, and 5 in the rat spinal cord. J Comp Neurol 422:464–487 Ambrosini A, Bresciani L, Fracchia S, Brunello N, Racagni G (1995) Metabotropic glutamate receptors negatively coupled to adenylate cyclase inhibit N-methyl-D-aspartate receptor activity and prevent neurotoxicity in mesencephalic neurons in vitro. Mol Pharmacol 47:1057–1064 Ango F, Pin JP, Tu JC, Xiao B, Worley PF Bockaert J, Fagni L (2000) Dendritic and axonal targeting of type 5 metabotropic glutamate receptor is regulated by homer 1 protein and neuronal excitation. J Neurosci 20:8710–8716 Annoura H, Fukunaga A, Uesugi M, Tatsuoka T, Horikawa Y (1996) A novel class of antagonists for metabotropic glutamate receptors, 7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxylates. Bioorg Med Chem Lett 6:7763–7766 Awad H, Hubert GW, Smith Y, Levey AI, Conn PJ (2000) Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci 20:7871–7879 Battaglia G, Bruno V, Ngomba RT, Di Grezia R, Copani A, Nicoletti F (1998) Selective activation of group-II metabotropic glutamate receptors is protective against excitotoxic neuronal death. Eur J Pharmacol 356:271–274 Battaglia G, Bruno V, Pisani A, Centone D, Catania MV, Calabresi P, Nicoletti F (2001) Selective blockade of type-I metabotropic glutamate receptors induce neuroprotection by enhancing GABAergic transmission. Mol Cell Neurosci 17:1071–1083 Baude A, Nusser Z, Roberts JD, Mulvihill E, McIlhinney RA, Somogyi P (1993) The metabotropic glutamate receptor (mGluR1 alpha) is concentrated at perisynaptic membrane of neuronal subpopulations as detected by immunogold reaction. Neuron 11:771–787 Behrens MM, Strasser U, Heidinger V, Lobner D, Yu SP, McDonald JW, Won M, Choi DW (1999) Selective activation of group II mGluRs with LY354740 does not prevent neuronal excitotoxicity. Neuropharmacology 38:1621–1630 Biber K, Laurie DJ, Berthele A, Sommer B, Tolle TR, Gebicke-Harter PJ, van Calker D, Boddeke HW (1999) Expression and signaling of group I metabotropic glutamate receptors in astrocytes and microglia. J Neurochem 72:1671–1680 Blumcke I, Suter B, Behle K, Kuhn R, Schramm J, Elger CE, Wiestler OD (2000) Loss of hilar mossy cells in Ammon’s horn sclerosis. Epilepsia 6:S174–180 Bond A, O’Neill MJ, Hicks CA, Monn JA, Lodge D (1998) Neuroprotective effects of a systemically active group II metabotropic glutamate receptor agonist LY354740 in a gerbil model of global ischaemia. Neuroreport 9:1191–1193 Bond A, Ragumoorthy N, Monn JA, Hicks CA, Ward MA, Lodge D, O’Neill MJ (1999) LY379268, a potent and selective group II metabotropic glutamate receptor agonist, is neuroprotective in gerbil global, but not focal, cerebral ischaemia. Neurosci Lett 273:191–194 Bond A, Jones NM, Hicks CA, Whiffin GM, Ward MA, O’Neill MF, Kingston AE, Monn JA, Ornstein PL, Schoepp DD, Lodge D, O’Neill MJ (2000) Neuroprotective effects of LY379268, a selective mGlu2/3 receptor agonist: investigations into possible mechanism of action in vivo. J Pharmacol Exp Ther 294:800–809 Brabet I, Parmentier M-L, De Colle C, Bockaert J, Acher F, Pin JP (1998) Comparative effect of L-CCG-I, DCG-IV and ␥-carboxylL-glutamate on all cloned metabotropic glutamate receptor subtypes. Neuropharmacology 37:1043–1051 Brakeman PR, Lanahan AA, O’Brien R, Roche K, Barnes CA, Huganir RL, Worley PF (1997) Homer: a protein that selectively binds metabotropic glutamate receptors. Nature 386:284–288 Bruno V, Copani A, Battaglia G, Raffaele R, Shinozaki H, Nicoletti F (1994) Protective effect of the metabotropic glutamate receptor agonist, DCG-IV, against excitotoxic neuronal death. Eur J Pharmacol 256:109–112 Bruno V, Battaglia G, Copani A, Giffard RG, Raciti G, Raffaele R, Shinozaki H, Nicoletti F (1995a) Activation of class II or class III metabotropic glutamate receptors protects cultured cortical neurons against excitotoxic degeneration. Eur J Neurosci 7:1906– 1913 Bruno V, Copani A, Knöpfel T, Kuhn R, Casabona G, Dell’Albani P,
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
Condorelli DF, Nicoletti F (1995b) Activation of metabotropic glutamate receptors coupled to inositol phospholipid hydrolysis amplifies NMDA-induced neuronal degeneration in cultured cortical cells. Neuropharmacology 34:1089–1098 Bruno V, Copani A, Bonanno L, Knöpfel T, Kuhn R, Roberts PJ, Nicoletti F (1996) Activation of group III metabotropic glutamate receptors is neuroprotective in cortical cultures. Eur J Pharmacol 310:61–66 Bruno V, Sureda FX, Storto M, Casabona G, Caruso A, Knöpfel T, Kuhn R, Nicoletti F (1997) The neuroprotective activity of groupII metabotropic glutamate receptors requires new protein synthesis and involves a glial-neuronal signaling. J Neurosci 17:1891–1897 Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, Nicoletti F (1998) Neuroprotection by glial metabotropic glutamate receptors is mediated by transforming growth factor-beta. J Neurosci 18:9594–9600 Bruno V, Battaglia G, Kingston A, O’Neill MJ, Catania MV, Di Grezia R, Nicoletti F (1999) Neuroprotective activity of the potent and selective mGlu1a metabotropic glutamate receptor antagonist, (+)2-methyl-4-carboxyphenylglycine (LY367385): comparison with LY367366, a broader spectrum antagonist with equal affinity for mGlu1a and mGlu5 receptors. Neuropharmacology 38:199–207 Bruno V, Battaglia G, Ksiazek I, van der Putten H, Catania MV, Giuffrida R, Lukic S, Leonhardt T, Inderbitzin W, Gasparini F, Kuhn R, Hampson DR, Nicoletti F, Flor PJ (2000a) Selective activation of mGlu4 metabotropic glutamate receptors is protective against excitotoxic neuronal death. J Neurosci 20:6413–6420 Bruno V, Ksiazek I, Battaglia G, Lukic S, Leonhardt T, Sauer D, Gasparini F, Kuhn R, Nicoletti F, Flor PJ (2000b) Selective blockade of metabotropic glutamate receptor subtype 5 is neuroprotective. Neuropharmacology 39:2223–2230 Bruno V, Battaglia G, Copani A, Cespedes VM, Galindo MF, Cena V, Sanchez-Prieto J, Gasparini F, Kuhn R, Flor PJ, Nicoletti F (2001) An activity-dependent switch from facilitation to inhibition in the control of excitotoxicity by group-I metabotropic glutamate receptors. Eur J Neurosci 13:1469–1478 Buisson A, Choi DW (1995) The inibitory mGluR agonist, S-4carboxy-3-hydroxy-phenylglycine selectively attenuates NMDA neurotoxicity and oxygen-glucose deprivation-induced neuronal death. Neuropharmacology 34:1081–1087 Buisson A, Nicole O, Docagne F, Sartelet H, MacKenzie ET, Vivien D (1998) Up-regulation of a serine protease inhibitor in astrocytes mediates the neuroprotective activity of transforming growth factor-1. FASEB J 12:1683–1691 Burke JP, Hablitz JJ (1996) G-protein activation by metabotropic glutamate receptors reduces spike frequency adaptation in neocortical neurons. Neuroscience 75:123–131 Bushell TJ, Lee CC, Shigemoto R, Miller RJ (1999) Modulation of synaptic transmission and differential localisation of mGluRs in cultured hippocampal autapses. Neuropharmacology 38:1553– 1567 Cai Z, Xiao F, Fratkin JD, Rhodes PG (1999) Protection of neonatal rat brain from hypoxic-ischemic injury by LY379268, a group II metabotropic glutamate receptor agonist. Neuroreport 10:3927– 3931 Camon L, Vives P, de Vera N, Martinez E (1998) Seizures and neuronal damage induced in the rat by activation of group I metabotropic glutamate receptors with their selective agonist 3,5dihydroxyphenylglycine. J Neurosci Res 51:339–348 Carroll FY, Stolle A, Beart PM, Voerste A, Brabet I, Mauler F, Joly C, Antonicek H, Bockaert J, Miller T, Pin JP, Prezeau L (2001) BAY36-7620: a potent non-competitive mGlu1 receptor antagonist with inverse agonist activity. Mol Pharmacol 59:965–973 Catania MV, Aronica E, Yankaya B, Troost D (2001) Increased expression of neuronal nitric oxide synthase splice variants in reactive astrocytes of amyotrophic lateral sclerosis human spinal Cord. J Neurosci 21:RC148 Cha JH, Kosinski CM, Kerner JA, Alsdorf SA, Mangiarini L, Davies SW, Penney JB, Bates GP, Young AB (1998) Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc Natl Acad Sci U S A 95:6480–6485 Chao CC, Hu S, Kravitz FH, Tsang M, Anderson WR, Peterson PK
mGlu RECEPTORS AND NEUROPROTECTION (1994) Transforming growth factor-beta protects human neurons against beta-amyloid-induced injury. Mol Chem Neuropathol 23:159–178 Chapman AG, Nanan K, Yip P, Meldrum BS (1999) Anticonvulsant activity of a metabotropic glutamate receptor 8 preferential agonist, (R,S)-4-phosphonophenylglycine. Eur J Pharmacol 383:23–27 Choi DW (1992) Excitotoxic cell death. J Neurobiol 23:1261–1276 Chuang SC, Bianchi R, Wong RK (2000) Group-I mGluR activation turns on a voltage-gated inward current in hippocampal pyramidal cells. J Neurophysiol 83:2844–2853 Ciccarelli R, Di Iorio P, Bruno V, Battaglia G, D’Alimonte I, D’Onofrio M, Nicoletti F, Caciagli F (1999) Activation of A1 adenosine and mGlu3 metabotropic glutamate receptors enhances the release of nerve growth factor and S-100ß protein from cultured astrocytes. Glia 27:275–281 Ciruela F, Soloviev MM, Chan WY, McIlhinney RA (2000) Homer1c/Vesl-1L modulates the cell surface targeting of metabotropic glutamate receptor type 1 alpha: evidence for an anchoring function. Mol Cell Neurosci 15:36–50 Clark BP, Baker SR, Goldsworthy J, Harris JR, Kingston AE (1997) 2-Methyl-4-carboxyphenylglycine (LY367385) selectively antagonizes metabotropic glutamate mGluR1 receptors. Bioorg Med Chem Lett 7:2777–2780 Claudio OI, Ferchmin P, Velisek L, Sperber EF, Moshe SL, Ortiz JG (2000) Plasticity of excitatory amino acid transporters in experimental epilepsy. Epilepsia 41:S104-S110 Copani A, Bruno V, Battaglia G, Leanza G, Pellitteri R, Russo A, Stanzani S, Nicoletti F (1995a) Activation of metabotropic glutamate receptors protects cultured neurons against apoptosis induced by beta-amyloid peptide. Mol Pharmacol 47:890–897 Copani A, Bruno V, Barresi V, Battaglia G, Condorelli DF, Nicoletti F (1995b) Activation of metabotropic glutamate receptors prevents neuronal apoptosis in culture. J Neurochem 64:101–108 Copani A, Casabona G, Bruno V, Caruso A, Condorelli DF, Messina A, Di Giorgi-Gerevini V, Pin JP, Kuhn R, Knöpfel T, Nicoletti F (1998) The metabotropic glutamate receptor mGlu5 controls the onset of developmental apoptosis in cultured cerebellar neurons. Eur J Neurosci 10:2173–2184 Copani A, Condorelli F, Caruso A, Vancheri C, Sala A, Giuffrida Stella AM, Canonico PL, Nicoletti F, Sortino MA (1999) Mitotic signalling by ß-amyloid causes neuronal death. FASEB J 13:2225–2234 Costa E, Armstrong DM, Guidotti A, Kharlamov A, Kiedrowski L, Manev H, Polo A, Wroblewski JT (1994) Gangliosides in the protection against glutamate excitotoxicity. Prog Brain Res 101:357–373 Corti C, Rimland JM, Corsi M, Ferraguti F (1997) Isolation and analysis of splice variants of the rat metabotropic glutamate receptors, mGluR7 and mGluR8 [abstract]. Soc Neurosci Abs 23:939 Daggett LP, Johnson EC, Varney MA, Lin FF, Hess SD, Deal CR, Jachec C, Lu CC, Kerner JA, Landwehrmeyer GB, Standaert DG, Young AB, Harpold MM, Velicelebi G (1998) The human Nmethyl-D-aspartate receptor 2C subunit: genomic analysis, distribution in human brain, and functional expression. J Neurochem 71:1953–1968 Datto MB, Li Y, Panus JF, Howe DJ, Xiong Y, Wang XF (1995) Transforming growth factor beta induces the cyclin-dependent kinase inhibitor p21 through a p53-independent mechanism. Proc Natl Acad Sci U S A 92:5545–5549 De Blasi A, Conn PJ, Pin JP, Nicoletti F (2001) Molecular determinant of metabotropic glutamate receptor signaling. Trends Pharmacol Sci 22:114–120 Dietrich D, Kral T, Clusmann H, Friedl M, Schramm J (1999) Reduced function of L-AP4-sensitive metabotropic glutamate receptors in human epileptic sclerotic hippocampus. Eur J Neurosci 11:1109– 1113 DiFiglia M (1990) Excitotoxic injury of the neostriatum: a model for Huntington’s disease. Trends Neurosci 13:286–289 D’Mello SR, Borodezt K, Soltoff SP (1997) Insulin-like growth factor and potassium depolarization maintain neuronal survival by distinct pathways: possible involvement of PI 3-kinase in IGF-1 signaling. J Neurosci 17:1548–1560
1029
Doble A (1999) The role of excitotoxicity in neurodegenerative disease: implications for therapy. Pharmacol Ther 81:163–221 Docagne F, Nicole O, Marti HH, MacKenzie ET, Buisson A, Vivien D (1999) Transforming growth factor-beta1 as a regulator of the serpins/t-PA axis in cerebral ischemia. FASEB J 13:1315–1324 Doherty AJ, Palmer MJ, Bortolotto ZA, Hargreaves A, Kingston AE, Ornstein PL, Schoepp DD, Lodge D, Collingridge GL (2000) A novel, competitive mGlu5 receptor antagonist (LY344545) blocks DHPG-induced potentiation of NMDA responses but not the induction of LTP in rat hippocampal slices. Br J Pharmacol 131:239–244 D’Onofrio M, Cuomo L, Battaglia G, Ngomba RT, Storto M, Kingston AE, Orzi F, De Blasi A, Nicoletti F, Bruno V (2001) Neuroprotection mediated by group-II metabotropic glutamate receptors involves the activation of the MAP kinase pathway and requires the induction of TGF-ß1. J Neurochem (in press) Dunah AW, Wyszynski M, Martin DM, Sheng M, Standaert DG (2000) Alpha-actinin-2 in rat striatum: localization and interaction with NMDA glutamate receptor subunits. Brain Res Mol Brain Res 79:77–87 Duvoisin RM, Zhang C, Ramonell K (1995) A novel metabotropic glutamate receptor expressed in the retina and olfactory bulb. J Neurosci 15:3075–3083 Emile L, Mercken L, Apiou F, Pradier L, Bock MD, Menager J, Clot J, Doble A, Blanchard JC (1996) Molecular cloning, functional expression, pharmacological characterization and chromosomal localization of the human metabotropic glutamate receptor type 3. Neuropharmacology 35:523–530 Faden AI, Ivanova SA, Yakovlev AG, Mukhin AG (1997) Neuroprotective effects of group III mGluR in traumatic neuronal injury. J Neurotrauma 14:885–895 Fagni L, Chavis P, Ango F, Bockaert J (2000) Complex interactions between mGluRs, intracellular Ca2+ stores and ion channels in neurons. Trends Neurosci 23:80–88 Ferraguti F, Baldani-Guerra B, Corsi M, Nakanishi S, Corti C (1999) Activation of the extracellular signal-regulated kinase 2 by metabotropic glutamate receptors. Eur J Neurosci 11:2073–2082 Flanders KC, Ren RF, Lippa CF (1998) Transforming growth factorbetas in neurodegenerative disease. Prog Neurobiol 54:71–85 Flor PJ, Lindauer K, Püttner I, Rüegg D, Lukic S, Knöpfel T, Kuhn R (1995a) Molecular cloning, functional expression and pharmacological characterization of the human metabotropic glutamate receptor type 2. Eur J Neurosci 7:622–629 Flor PJ, Lukic S, Rüegg D, Leonhardt T, Knöpfel T, Kuhn R (1995b) Molecular cloning, functional expression and pharmacological characterization of the human metabotropic glutamate receptor type 4. Neuropharmacology 34:149–155 Flor PJ, Gomeza J, Tones MA, Kuhn R, Pin JP, Knöpfel T (1996) The C-terminal domain of the mGluR1 metabotropic glutamate receptor affects sensitivity to agonists. J Neurochem 67:58–63 Flor PJ, van der Putten H, Rüegg D, Lukic S, Leonhardt T, Bence M, Sansig G, Knöpfel T, Kuhn R (1997) A novel splice variant of a metabotropic glutamate receptor, human mGluR7b. Neuropharmacology 36:153–159 Galoyan SM, Merlin LR (2000) Long-lasting potentiation of epileptiform bursts by group I mGluRs is NMDA receptor independent. J Neurophysiol 83:2463–2467 Gasparini F, Bruno V, Battaglia G, Lukic S, Leonhardt T, Inderbitzin W, Laurie D, Sommer B, Varney MA, Hess SD, Johnson EC, Kuhn R, Urwyler S, Sauer D, Portet C, Schmutz M, Nicoletti F, Flor PJ (1999a) (R,S)-4-phosphonophenylglycine, a potent and selective group III metabotropic glutamate receptor agonist is anticonvulsive and neuroprotective in vivo. J Pharmacol Exp Ther 290:1678–1687 Gasparini F, Lingenhohl K, Stoehr N, Flor PJ, Heinrich M, Vranesic I, Biollaz M, Allgeier H, Heckendorn R, Urwyler S, Varney MA, Johnson EC, Hess SD, Rao SP, Sacaan AI, Santori EM, Velicelebi G, Kuhn R (1999b) 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology 38:1493–1503 Gasparini F, Inderbitzin W, Francotte E, Lecis G, Richert P, Dragic Z, Kuhn R, Flor PJ (2000) (+)-4-Phosphonophenylglycine (PPG) a
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1030
V. BRUNO ET AL.
new group III selective metabotropic glutamate receptor agonist. Bioorg Med Chem Lett 10:1241–1244 Gereau IV RW, Conn PJ (1995) Multiple presynaptic metabotropic glutamate receptors modulate excitatory and inhibitory synaptic transmission in hippocampal area CA1. J Neurosci 15:6879–6889 Goodwin H, Curran N, Chioza B, Blower J, Nashef L, Asherson P, Makoff AJ (2000) No association found between polymorphisms in genes encoding mGluR7 and mGluR8 and idiopathic generalised epilepsy in a case control study. Epilepsy Res 39:27–31 Gottron F, Turetsky D, Choi DW (1995) SMI-32 antibody against non-phosphorylated neurofilaments identifies a subpopulation of cultured cortical neurons hypersensitive to kainate toxicity. Neurosci Lett 194:1–4 Graham ME, Burgoyne RD (1994) Activation of metabotropic glutamate receptors by L-AP4 stimulates survival of rat cerebellar granule cells in culture. Eur J Pharmacol 288:115–123 Greenwood RS, Fan Z, McHugh R, Meeker R (2000) Inhibition of hippocampal kindling by metabotropic glutamate receptor antisense oligonucleotides. Mol Cell Neurosci 16:233–243 Hansson O, Petersen A, Leist M, Nicotera P, Castilho RF, Brundin P (1999) Transgenic mice expressing a Huntington’s disease mutation are resistant to quinolinic acid-induced striatal excitotoxicity. Proc Natl Acad Sci U S A 96:8727–8732 Helton DR, Tizzano JP, Monn JA, Schoepp DD, Kallman MJ (1998) Anxiolytic and side-effect profile of LY354740: a potent, highly selective, orally active agonist for group II metabotropic glutamate receptors. J Pharmacol Exp Ther 284:651–660 Henrich-Noack P, Prehn JH, Krieglstein J (1996) TGF-beta 1 protects hippocampal neurons against degeneration caused by transient global ischemia. Dose-response relationship and potential neuroprotective mechanisms. Stroke 27:1609–1614 Henrich-Noack P, Hatton CD, Reymann KG (1998) The mGlu receptor ligand (S)-4C3HPG protects neurons after global ischaemia in gerbils. NeuroReport 9:985–988 Henrich-Noack P, Flor PJ, Sabelhaus CF, Prass K, Dirnagl U, Gasparini F, Sauter A, Rudin M, Reymann KG (2000) Distinct influence of the group III metabotropic glutamate receptor agonist (R,S)-4phosphonophenylglycine [(R,S)-PPG] on different forms of neuronal damage. Neuropharmacology 39:911–917 Herrero I, Miras-Portugal MT, Sanchez-Prieto J (1992) Positive feedback of glutamate exocytosis by metabotropic presynaptic receptor stimulation. Nature 360:163–166 Herrero I, Miras-Potugal MT, Sanchez-Prieto J (1994) Rapid desensitization of the presynaptic metabotropic receptor that facilitates glutamate exocytosis. Eur J Neurosci 6:115–120 Herrero I, Miras-Portugal MT, Sanchez-Prieto J (1998) Functional switch from facilitation to inhibition in the presynapticcontrol of glutamate release by metabotropic glutamate receptors. J Biol Chem 273:1951–1958 Ishida M, Saitoh T, Shimamoto K, Ohfune Y, Shinozaki H (1993) A novel metabotropic glutamate receptor agonist: marked depression of monosynaptic excitation in the newborn rat isolated spinal cord. Br J Pharmacol 109:1169–1177 Jane DE, Jones PL, Pook PC, Tse HW, Watkins JC (1994) Actions of two new antagonists showing selectivity for different sub-types of metabotropic glutamate receptor in the neonatal rat spinal cord. Br J Pharmacol 112:809–816 Jia Z, Lu Y, Henderson J, Taverna F, Romano C, Abramow-Newerly W, Wojtowicz JM, Roder J (1998) Selective abolition of the NMDA component of long-term potentiation in mice lacking mGluR5. Learn Mem 5:331–343 Johnson EM J, Koike T, Franklin J (1992) A “calcium set-point hypothesis” of neuronal dependence on neurotrophic factors. Exp Neurol 115:163–166 Joly C, Gomeza J, Brabet I, Curry K, Bockaert J, Pin JP (1995) Molecular, functional, and pharmacological characterization of the metabotropic glutamate receptor type 5 splice variants: comparison with mGluR1. J Neurosci 15:3970–3981 Kammermeier PJ, Xiao B, Tu JC, Worley PF, Ikeda SR (2000) Homer proteins regulate coupling of group I metabotropic glutamate receptors to N-type calcium and M-type potassium channels. J Neurosci 20:7238–7245 Kawabata S, Tsutsumi R, Kohara A, Yamaguchi T, Naknishi S, Okada
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
M (1996) Control of calcium oscillations by phosphorylation of metabotropic glutamate receptors. Nature 383:89–92 Kingston AE, O’Neill MJ, Bond A, Bruno V, Battaglia G, Nicoletti F, Harris JR, Clark BP, Monn JA, Lodge D, Schoepp DD (1999a) Neuroprotective action of novel and potent ligands of group-I and group-II metabotropic glutamate receptors. Ann N Y Acad Sci 890:438–439 Kingston AE, O’Neill MJ, Lam A, Bales KR, Monn JA, Schoepp DD (1999b) Neuroprotection by metabotropic glutamate receptor agonists: LY354740, LY379268 and LY389795. Eur J Pharmacol 377:155–165 Klapstein GJ, Meldrum BS, Mody I (1999) Decreased sensitivity to group III mGluR agonists in the lateral perforant path following kindling. Neuropharmacology 38:927–933 Klodzinska A, Chojnacka-Wojcik E, Pilc A (1999) Selective group II glutamate metabotropic receptor agonist LY354740 attenuates pentylentetrazole- and picrotoxin-induced seizures. Pol J Pharmacol 51:543–545 Klodzinska A, Bijak M, Chojnacka-Wojcik E, Kroczka B, Swiader M, Czuczwar SJ, Pilc A (2000) Roles of group II metabotropic glutamate receptors in modulation of seizure activity. Naunyn Schmiedebergs Arch Pharmacol 361:283–288 Konieczny J, Ossowska K, Wolfarth S, Pilc, A (1999) LY354740, a group II metabotropic glutamate receptor agonist with potential antiparkinsonian properties in rats. Naunyn-Schmiedebergs Arch Pharmacol 358:500–502 Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi S, Jingami H, Morikawa K (2000) Structural basis of glutamate recognition by dimeric metabotropic glutamate receptor. Nature 407:971–977 Kutsuwada T, Kashiwabuchi N, Mori H, Sakimura K, Kushiya E, Araki K, Meguro H, et al (1992) Molecular diversity of the NMDA receptor channel. Nature 358:36–41 Lafon-Cazal M, Fagni L, Guiraud MJ, Mary S, Lerner-Natoli M, Pin JP, Shigemoto R, Bockaert J (1999) mGluR7-like metabotropic glutamate receptors inhibit NMDA-mediated excitotoxicity in cultured mouse cerebellar granule neurons. Eur J Neurosci 11:663– 672 Lam AG, Soriano MA, Monn JA, Schoepp DD, Lodge D, McCulloch J (1998) Effects of the selective metabotropic glutamate agonist LY354740 in a rat model of permanent ischaemia. Neurosci Lett 254:121–123 Laurie DJ, Boddeke HW, Hiltscher R, Sommer B (1996) HmGlu1d, a novel splice variant of the human type I metabotropic glutamate receptor. Eur J Pharmacol 296:R1-R3 Lee JM, Zipfel GJ, Choi DW (1999) The changing landscape of ischemic brain injury mechanisms. Nature 399:A7–A14 Levine JB, Kong J, Nadler M, Xu Z (1999) Astrocytes interact intimately with degenerating motor neurons in mouse amyotrophic lateral sclerosis (ALS). Glia 28:215–224 Lie AA, Becker A, Behle K, Beck H, Malitschek B, Conn PJ, Kuhn R, Nitsch R, Plaschke M, Schramm J, Elger CE, Wiestler OD, Blumcke I (2000) Up-regulation of the metabotropic glutamate receptor mGluR4 in hippocampal neurons with reduced seizure vulnerability. Ann Neurol 47:26–35 Lin YJ, Bovetto S, Carver JM, Giordano T (1996) Cloning of the cDNA for the human NMDA receptor NR2C subunit and its expression in the central nervous system and periphery. Brain Res Mol Brain Res 43:57–64 Lin SH, Maiese K (2001) The metabotropic glutamate receptor system protects against ischemic free radical programmed cell death in rat brain endothelial cells. J Cereb Blood Flow Metab 21:262–275 Litschig S, Gasparini F, Rüegg D, Munier N, Flor PJ, Vranesic I-T, Prezeau L, Pin J-P, Thomsen C, Kuhn R (1999) CPCCOEt, a noncompetitive metabotropic glutamate receptor 1 antagonist, inhibits receptor signaling without affecting glutamate binding. Mol Pharmacol 55:453–461 Lujan R, Roberts JD, Shigemoto R, Ohishi H, Somogyi P (1997) Differential plasma membrane distribution of metabotropic glutamate receptors mGluR1 alpha, mGluR2 and mGluR5, relative to neurotransmitter release sites. J Chem Neuroanat 13:219–241 Maiese K, Greenberg R, Boccone L, Swiriduk M (1995) Activation of
mGlu RECEPTORS AND NEUROPROTECTION the metabotropic glutamate receptor is neuroprotective during nitric oxide toxicity in primary hippocampal neurons of rats. Neurosci Lett 194:173–176 Maiese K, Vincent A, Lin SH, Shaw T (2000) Group I and group III metabotropic glutamate receptor subtypes provide enhanced neuroprotection. J Neurosci Res 62:257–272 Manev H, Favaron M, Vicini S, Guidotti A, Costa E (1990a) Glutamate-induced neuronal death in primary cultures of cerebellar granule cells: protection by synthetic derivatives of endogenous sphingolipids. J Pharmacol Exp Ther 252:419–427 Manev H, Costa E, Wroblewski JT, Guidotti A (1990b) Abusive stimulation of excitatory amino acid receptors: a strategy to limit neurotoxicity. FASEB J 4:2789–2797 Manev H, Uz T, Giusti P (1997) Neuroprotective action of the pineal hormone melatonin against excitotoxicity. Receptor abusedependent antagonism (RADA). Ann N Y Acad Sci 825:85–89 Martin G, Nie Z, Siggins GR (1997) Metabotropic glutamate receptors regulate N-methyl-D-aspartate-mediated synaptic transmission in nucleus accumbens. J Neurophysiol 78:3028–3038 McDonald JW, Schoepp DD (1992) The metabotropic glutamate receptor agonist 1S,3R-ACPD selectively potentiates N-methyl-Daspartate-induced brain injury. Eur J Pharmacol 215:353–354 McDonald JW, Fix AS, Tizzano JP, Scoepp DD (1993) Seizures and brain injury in neonatal rats induced by 1S, 3R-ACPD, a metabotropic glutamate receptor agonist. J Neurosci 13:4445–4455 McNamara JO (1999) Emerging insights into the genesis of epilepsy. Nature 399:A15–A22 Merlin LR, Wong RK (1997) Role of group-I metabotropic glutamate receptors in the pattering of epileptiform activities in vitro. J Neurophysiol 78:539–544 Merlin LR (1999) Group I mGluR-mediated silent induction of longlasting epileptiform discharges. J Neurophysiol 82:1078–1081 Meucci O, Miller RJ (1996) Gp120-induced neurotoxicity in hippocampal pyramidal neuron cultures: protective action of TGF-beta1. J Neurosci 16:4080–4088 Miller S, Bridges RJ, Cotman CW (1993) Stimulation of phosphoinositide hydrolysis by trans-(+/−)-ACPD is greatly enhanced when astrocytes are cultured in a serum-free defined medium. Brain Res 618:175–178 Minakami R, Katsuki F, Yamamoto T, Nakamura K, Sugiyama H (1994) Molecular cloning and the functional expression of two isoforms of human metabotropic glutamate receptor subtype 5. Biochem Biophys Res Commun 199:1136–1143 Minakami R, Kato A, Sugiyama H (2000) Interaction of Vesl1L/Homer 1c with syntaxin 13. Biochem Biophys Res Commun 272:466–471 Miyamoto M, Ishida M, Kwak S, Shinozaki H (1994) Agonists for metabotropic glutamate receptors in the rat delay recovery from halothane anesthesia. Eur J Pharmacol 260:99–102 Monn JA, Valli MJ, Massey SM, Wright RA, Salhoff CR, Johnson BG, Howe T, Alt CA, Rhodes GA, Robey RL, Griffey KR, Tizzano JP, Kallman MJ, Helton DR, Schoepp DD (1997) Design, synthesis, and pharmacological characterization of (+)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY354740): a potent, selective, and orally active group 2 metabotropic glutamate receptor agonist possessing anticonvulsant and anxiolytic properties. J Med Chem 40:528–537 Monyer H, Giffard RG, Hartley DM, Dugan LL, Goldberg MP, Choi DW (1992) Oxygen or glucose deprivation-induced neuronal injury in cortical cell cultures is reduced by tetanus toxin. Neuron 8:967–973 Mori H, Yamakura T, Masaki H, Mishina M (1993) Involvement of the carboxy-terminal region in modulation by TPA of the NMDA receptor channel. Neuroreport 4:519–522 Morishita W, Kirov SA, Alger BE (1998) Evidence for metabotropic glutamate receptor activation in the induction of depolarizationinduced suppression of inhibition in hippocampal CA1. J Neurosci 18:4870–4882 Morley P, Small DL, Murray CL, Mealing GA, Poulter MO, Durkin JP, Stanimirovic DB (1998) Evidence that functional glutamate receptors are not expressed on rat or human cerebromicrovascular endothelial cells. J Cereb Blood Flow Metab 18:396–406 Movsesyan VA, O’Leary DM, Fan L, Bao W, Mullins PG, Knoblach
1031
SM, Faden AI (2001) mGluR5 antagonists 2-methyl-6(phenylethynyl)-pyridine and (E)-2-methyl-6-(2-phenylethenyl)pyridine reduce traumatic neuronal injury in vitro and in vivo by antagonizing N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 296:41–47 Nakanishi S (1994) Metabotropic glutamate receptors: synaptic transmission, modulation, and plasticity. Neuron 13:1031–1037 Nicoletti F, Iadarola MJ, Wroblewski JT, Costa E (1986) Excitatory amino acid recognition sites coupled with inositol phospholipid metabolism: developmental changes and interaction with a1adrenoceptors. Proc Natl Acad Sci U S A 83:1931–1935 Nicoletti F, Bruno V, Copani A, Casabona G, Knöpfel T (1996) Metabotropic glutamate receptors: a new target for the therapy of neurodegenerative disorders? Trends Neurosci 1:267–271 Nicoletti F, Bruno V, Catania MV, Battaglia G, Copani A, Barbagallo G, Ceña V, Sanchez-Prieto J, Spano PF, Pizzi M (1999) Group-I metabotropic glutamate receptors: hypotheses to explain their dual role in neurotoxicity and neuroprotection. Neuropharmacology 38:1477–1484 Niethammer M, Kim E, Sheng M (1996) Interaction between the C terminus of NMDA receptor subunits and multiple members of the PSD-95 family of membrane-associated guanylate kinases. J Neurosci 16:2157–2163 Nusser Z, Mulvihill E, Streit P, Somogyi P (1994) Subsynaptic segregation of metabotropic and ionotropic glutamate receptors as revealed by immunogold localization. Neuroscience 61:421–427 O’Hara PJ, Sheppard PO, Thojersen H, Venezia D, Haldeman BA, McGrane V, Houamed KM, Thomsen C, Gilbert TL, Mulvihill ER (1993) The ligand-binding domain in metabotropic glutamate receptors is related to bacterial peryplasmic binding proteins. Neuron 11:41–52 Okamoto N, Hori S, Akazawa C, Hayashi Y, Shigemoto R, Mizuno N, Nakanishi S (1994) Molecular characterization of a new metabotropic glutamate receptor mGluR7 coupled to inhibitory cyclic AMP signal transduction. J Biol Chem 269:1231–1236 O’Leary DM, Movsesyan V, Vicini S, Faden AI (2000) Selective mGluR5 antagonists MPEP and SIB-1893 decrease NMDA or glutamate-mediated neuronal toxicity through actions that reflect NMDA receptor antagonism. Br J Pharmacol 131:1429–1437 Ong WY, Balcar VJ (1997) Group I metabotropic glutamate receptor agonist causes neurodegeneration in rat hippocampus. J Hirnforsch 3:317–322 Opitz T, Reymann KG (1993) (1S,3R)-ACPD protects synaptic transmission from hypoxia in hippocampal slices. Neuropharmacology 32:103–104 Orlando LR, Alsdorf SA, Penney JB Jr, Young AB (2001) The role of group I and group II metabotropic glutamate receptors in modulation of striatal NMDA and quinolinic acid toxicity. Exp Neurol 167:196–204 Paquet M, Smith Y (2000) Subsynaptic localization of group I metabotropic glutamate receptors at glutamatergic synapses in monkey striatum [abstract]. Soc Neurosci Abs 26:740 Pellegrini-Giampietro DE, Zukin RS, Bennett MV, Cho S, Pulsinelli WA (1992) Switch in glutamate receptor subunit gene expression in CA1 subfield of hippocampus following global ischemia in rats. Proc Natl Acad Sci U S A 89:10499–10503 Pellegrini-Giampietro DE, Cozzi A, Peruginelli F, Leonardi P, Meli E, Pellicciari R, Moroni F (1999a) 1-Aminoindan-1,5-dicarboxylic acid and (S)-(+)-2-(3’-carboxyicyclo(1.1.1.)pentyl)-glycine, two mGlu1 receptor-preferring antagonists, reduce neuronal death in in vitro and in vivo models of cerebral ischemia. Eur J Neurosci 11:3637–3647 Pellegrini-Giampietro DE, Peruginelli F, Meli E, Cozzi A, AlbaniTorregrossa S, Pellicciari R, Moroni F (1999b) Protection with metabotropic glutamate 1 receptor antagonists in models of ischemic neuronal death: time-course and mechanisms. Neuropharmacology 38:1607–1619 Pellicciari R, Luneia R, Costantino G, Marinozzi M, Natalini B, Jakobsen I, Kanstrup A, Lombardi G, Moroni F, Thomsen C (1995) 1-Aminoindan-1,5-dicarboxilic acid: a novel antagonist of phospholipase C-linked metabotropic glutamate receptors. J Med Chem 38:3717–3719 Petralia RS, Wang YX, Niedzielski AS, Wenthold RJ (1996) The
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
1032
V. BRUNO ET AL.
metabotropic glutamate receptors, mGluR2 and mGluR3, show unique postsynaptic, presynaptic and glial localizations. Neuroscience 71:949–976 Pin JP, Duvoisin R (1995) The metabotropic glutamate receptors: structure and functions. Neuropharmacology 34:1–26 Pisani A, Calabresi P, Centonze D, Bernardi G (1997) Enhancement of NMDA responses by group-I metabotropic glutamate receptor activation in striatal neurones. Br J Pharmacol 120:1007–1014 Pisani A, Bernardi G, Bonsi P, Centonze D, Giacomini P, Calabresi P (2000) Cell-type specificity of mGluR activation in striatal neuronal subtypes. Amino Acids 19:119–129 Pizzi M, Fallacara C, Arrighi V, Memo M, Spano PF (1993) Attenuation of excitatory amino acid toxicity by metabotropic glutamate receptor agonists and aniracetam in primary cultures of cerebellar granule cells. J Neurochem 61:683–689 Pizzi M, Galli P, Consolandi O, Arrighi V. Memo M. Spano PF (1996a) Metabotropic and ionotropic transducers of glutamate signals inversely control cytoplasmic Ca2+ concentration and excitotoxicity in cultured cerebellar graniule cells: pivotal role of protein kinase C. Mol Pharmacol 49: 586–594 Pizzi M, Consolandi O, Memo M, Spano PF (1996b) Activation of multiple metabotropic glutamate receptor subtypes prevents NMDA-induced excitotoxicity in rat hippocampal slices. Eur J Neurosci 8:1516–1521 Pizzi M, Boroni F, Moraitis K, Bianchetti A, Memo M, Spano PF (1999) Reversal of glutamate excitotoxicity by activation of PKCassociated metabotropic glutamate receptors in cerebellar granule cells relies on NR2C subunit expression. Eur J Neurosci 11:1–8 Pizzi M, Benarese M, Boroni F, Goffi F, Valerio A, Spano PF (2000) Neuroprotection by metabotropic glutamate receptor agonists on kainate-induced degeneration of motor neurons in spinal cord slices from adult rat. Neuropharmacology 39:903–910 Prehn JHM, Bindokas VP, Marcuccilli CJ, Krajewski SW, Reed JC, Miller RJ (1994) Regulation of neuronal Bcl-2 protein expression and calcium homeostasis by transforming growth factor type beta confers wide-ranging protection on rat hippocampal neurons. Proc Natl Acad Sci U S A 91:12599–12603 Prehn JH, Miller RJ (1996) Opposite effects of TGF-beta1 on rapidlyand slowly triggered excitotoxic injury. Neuropharmacology 35:249–256 Prehn JHM, Bindokas VP, Jordan J, Galindo MF, Ghadge GD, Roos RP, Boise LH, Thomson CB, Krajewski SW, Reed JC, Miller RJ (1996) Protective effect of transforming growth factor-ß1 on ßamyloid neurotoxicity in rat hippocampal neurons. Mol Pharmacol 49:319–328 Pruzanski W, Stefanski E, Vadas P, Kennedy BP, van den Bosch H (1998) Regulation of the cellular expression of secretory and cytosolic phospholipases A2, and cyclooxygenase-2 by peptide growth factors. Biochim Biophys Acta 1403:47–56 Rao AM, Hatcher JF, Dempsey RJ (2000) Neuroprotection by group I metabotropic glutamate receptor antagonists in forebrain ischemia of gerbil. Neurosci Lett 293:1–4 Ren RF, Hawver DB, Kim RS, Flanders KC (1997) Transforming growth factor-ß protects human hNT cells from degeneration induced by ß-amyloid peptide: involvement of the TGF-ß type II receptor. Brain Res Mol Brain Res 48:315–322 Rodriguez-Moreno A, Sistiaga A, Lerma J, Sanchez-Prieto J (1998) Switch from facilitation to inhibition of excitatory synaptic transmission by the desensitization of group-I mGluRs in the rat hippocampus. Neuron 21:1477–1486 Ruocco A, Nicole O, Docagne F, Ali C, Chazalviel L, Komesly S, Yablonsky F, Roussel S, MacKenzie ET, Vivien D, Buisson A (1999) A transforming growth factor-beta antagonist unmasks the neuroprotective role of this endogenous cytokine in excitotoxic and ischemic brain injury. J Cereb Blood Flow Metab 19:1345–1353 Sabelhaus CF, Schroder UH, Breder J, Henrich-Noack P, Reymann KG (2000) Neuroprotection against hypoxic/hypoglycaemic injury after the insult by the group III metabotropic glutamate receptor agonist (R, S)-4-phosphonophenylglycine. Br J Pharmacol 131:655–658 Sacaan AI, Schoepp DD (1992) Activation of hippocampal metabotropic excitatory amino acid receptors leads to seizures and neuronal damage. Neurosci Lett 139:77–82
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001
Sagara Y, Schubert D (1998) The activation of metabotropic glutamate receptors protects nerve cells from oxidative stress. J Neurosci 18:6662–6671 Salt TE, Binns KE (2000) Contributions of mGlu1 and mGlu5 receptors to interactions with N-methyl-D-aspartate receptor-mediated responses and nociceptive sensory responses of rat thalamic neurons. Neuroscience 100:375–380 Scannevin RH, Huganir RL (2000) Postsynaptic organization and regulation of excitatory synapses. Nat Rev Neurosci 1:133–141 Schoepp DD, Jane DE, Monn AJ (1999) Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology 38:1431–1476 Schroeder UH, Opiz T, Jager T, Sabelhaus CF, Breder J, Reymann KG (1999) Protective effects of group-I metabotropic glutamate receptor activation against hypoxic/hypoglicemic injury in rat hippocampal slices: timing and involvement of protein kinase C. Neuropharmacology 38:209–216 Scorziello A, Florio T, Bajetto A, Thellung S, Schettini G (1997) TGF-beta1 prevents gp120-induced impairment of Ca2+ homeostasis and rescues cortical neurons from apoptotic death. J Neurosci Res 49:600–607 Shigemoto R, Kinoshita A, Wada E, Nomura S, Ohishi H, Takada M, Flor PJ, Neki A, Abe T, Nakanishi S, Mizuno N (1997) Differential presynaptic localization of metabotropic glutamate receptor subtypes in the rat hippocampus. J Neurosci 17:7503–7522 Sillevis Smitt P, Kinoshita A, De Leeuw B, Moll W, Coesmans M, Jaarsma D, Henzen-Logmans S, Vecht C, De Zeeuw C, Sekiyama N, Nakanishi S, Shigemoto R (2000) Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 342:21–27 Sistiaga A, Herrero I, Conquet F, Sanchez-Prieto J (1998) The metabotropic glutamate receptor 1 is not involved in the facilitation of glutamate release in cerebro-cortical nerve terminals. Neuropharmacology 3:1485–1492 Sistiaga A, Sanchez-Prieto J (2000) Protein phosphatase 1 and 2A inhibitors prolong the switch in the control of glutamate release by group I metabotropic glutamate receptors: characterization of the inhibitory pathway. J Neurochem 75:1566–1574 Soderling TR, Derkach VA (2000) Postsynaptic protein phosphorilation and LTP. Trends Neurosci 23:75–80 Sommer C, Roth SU, Kuhn R, Kiessling M (2000) Metabotropic glutamate receptor subtypes are differentially expressed after transient cerebral ischemia without, during and after tolerance induction in the gerbil hippocampus. Brain Res 872:172–180 Spooren WPJM, Gasparini F, Salt TE, Kuhn R (2001) Novel allosteric antagonists shed light on mGlu5 receptors and CNS disorders. Trends Pharmacol Sci (in press) Strasser U, Lobner D, Berhens MM, Canzoniero LM, Choi DW (1998) Antagonists of group-I mGluRs attenuate exicitotoxic neuronal death in cortical cultures. Eur J Neurosci 10:2848–2855 Suzuki T, Shimizu N, Tsuda M, Soma M, Misawa M (1999) Role of metabotropic glutamate receptors in the hypersusceptibility to pentylentetrazole-induced seizures during diazepam withdrawal. Eur J Pharmacol 369:163–168 Tanabe Y, Masu M, Ishii T, Shigemoto R, Nakanishi S (1992) A family of metabotropic glutamate receptors. Neuron 8:169–179 Tang E, Yip PK, Chapman AG, Jane DE, Meldrum BS (1997) Prolonged anticonvulsant action of glutamate metabotropic receptor agonists in inferior colliculus of genetically epilepsy-prone rats. Eur J Pharmacol 327:109–115 Thomas NK, Jane DE, Tse HW, Watkins JC (1996) Alpha-methyl derivatives of serine-O-phosphate as novel, selective competitive metabotropic glutamate receptor antagonists. Neuropharmacology 35:637–642 Thomsen C, Bruno V, Nicoletti F, Marinozzi M, Pellicciari R (1996) (2S,1’S,2’S,3’R)-2-(2’-Carboxy-3’-phenylcyclopropyl)glycine, a potent and selective antagonist of type 2 metabotropic glutamate receptors. Mol Pharmacol 50:6–9 Thomsen C (1999) Pentylenetetrazol-induced seizures increase 3H-L2-amino-4-phosphonobutyrate binding in discrete regions of the rat bran. Neurosci Lett 266:5–8 Tomoda T, Shirasawa T, Yahagi YI, Ishii K, Takagi H, Furiva Y, Arai
mGlu RECEPTORS AND NEUROPROTECTION KI, Mori H, Muramatsu MA (1996) Transforming growth factorbeta is a survival factor for neonate cortical neurons: coincident expression of type-1 receptors in developing cerebral cortices. Dev Biol 179:79–90 Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakman P, Doan A, Aakalu VK, Lanahan AA, Sheng M, Worley PF (1999) Coupling of mGluR/Homer and PSD-95 coplexes by the Shank family of postsynaptic density proteins. Neuron 23:583–592 Ugolini A, Corsi M, Bordi F (1999) Potentiation of NMDA and AMPA responses by the specific mGluR5 agonist CHPG in spinal cord motoneurons. Neuropharmacology 38:1569–1576 Varney MA, Cosford N, Jachec C, Rao SP, Sacaan A, Lin FF, Bleicher L, Santori EM, Flor PJ, Allgeier H, Gasparini F, Kuhn R, Hess SD, VeliÁelebi G, Jonhson EC (1999) SIB-1757 and SIB-1893: selective, noncompetitive antagonists of metabotropic glutamate receptor type 5. J Pharmacol Exp Ther 290:170–181 Watkins J, Collingridge G (1994) Phenylglycine derivatives as antago-
1033
nists of metabotropic glutamate receptors. Trends Pharmacol Sci 15:333–342 Wolfarth S, Konieczny J, Lorenc-Koci E, Ossowaska K, Pilc A (2000) The role of metabotropic glutamate receptor (mGluR) ligands in parkinsonian muscle rigidity. Amino Acids 19:95–101 Wong CG, Scherer SW, Snead OC 3rd, Hampson DR (2001) Localization of the human mGluR4 gene within an epilepsy susceptibility locus (1). Brain Res Mol Brain Res 87:109–116 Wroblewska B, Wroblewski JT, Pschenichkin S, Surin A, Sullivan SE, Neale JH (1997) N-acetylaspartylglutamate selectively activates mGluR3 receptors in transfected cells. J Neurochem 69:174–181 Wyszynski M, Lin J, Rao A, Nigh E, Beggs AH, Craig AM, Sheng M (1997) Competitive binding of alpha-actinin and calmodulin to the NMDA receptor. Nature 385:439–42 Xiao B, Tu JC, Worley PF (2000) Homer: a link between neural activity and glutamate receptor function. Curr Opin Neurobiol 10:370–374
J Cereb Blood Flow Metab, Vol. 21, No. 9, 2001