The distinct role of mGlu1 receptors in post-ischemic neuronal death

The distinct role of mGlu1 receptors in post-ischemic neuronal death

Review TRENDS in Pharmacological Sciences Vol.24 No.9 September 2003 461 The distinct role of mGlu1 receptors in post-ischemic neuronal death Dome...

1MB Sizes 5 Downloads 15 Views

Review

TRENDS in Pharmacological Sciences

Vol.24 No.9 September 2003

461

The distinct role of mGlu1 receptors in post-ischemic neuronal death Domenico E. Pellegrini-Giampietro Dipartimento di Farmacologia Preclinica e Clinica, Universita` di Firenze, Viale G. Pieraccini 6, 50139 Firenze, Italy

Metabotropic glutamate receptors of the mGlu1 and mGlu5 subtypes exhibit a high degree of sequence homology and are both coupled to phospholipase C and intracellular Ca21 mobilization. However, functional differences have been detected for these receptor subtypes when they are coexpressed in the same neuronal populations. Experimental evidence indicates that mGlu1 and mGlu5 receptors play a differential role in models of cerebral ischemia and that only mGlu1 receptors are implicated in the pathways leading to postischemic neuronal injury. The localization of mGlu1 receptors in GABA-containing interneurons rather than in hippocampal CA1 pyramidal cells that are vulnerable to ischemia has prompted studies that have provided a new viewpoint on the neuroprotective mechanism of mGlu1 receptor antagonists. The hypothesis predicts that these pharmacological agents attenuate post-ischemic injury by enhancing GABA-mediated neurotransmission. Glutamate exerts its modulatory effects on neuronal excitability and synaptic transmission by interacting with a family of G-protein-coupled receptors (GPCRs) termed metabotropic glutamate (mGlu) receptors [1,2]. Like all GPCRs, mGlu receptors have a seven-transmembrane domain (7TMD) region that appears to be structurally related to rhodopsin, although the sequence similarity with rhodopsin is very low. These receptors are, however, unique because unlike rhodopsin-like receptors their agonist binding site is not located in the 7TMD but instead is located in the large N-terminal extracellular domain (ECD), which is connected to the 7TMD by a cysteine-rich linker (Figure 1). Residues within intracellular loops 2 and 3 appear to be crucial for G-protein coupling selectivity and activation, respectively, whereas the C-terminal tail contains residues that are involved in the protein kinase C (PKC)-induced desensitization of the receptor and in the interaction with intracellular proteins such as calmodulin, Homer and PDZ domain-containing proteins. Eight mGlu receptor subtypes and multiple splice variants have been cloned to date from mammalian brain, and they have been subdivided into three groups on the basis of their structural homology, coupling mechanisms and agonist pharmacology [3,4] (Figure 1). Group I mGlu receptors include mGlu1 and mGlu5 receptors, are coupled to phospholipase C (PLC) and are Corresponding author: Domenico E. Pellegrini-Giampietro (domenico.pellegrini@ unifi.it).

selectively activated by (S)-3,5-dihydroxyphenylglycine [(S)-3,5-DHPG]. Group II (mGlu2 and mGlu3) and group III (mGlu4, mGlu6, mGlu7 and mGlu8) receptors are coupled via Gi/o proteins to adenylyl cyclase inhibition and can be pharmacologically distinguished because only members of group III are selectively stimulated by L -(þ)-2amino-4-phosphonobutyrate. Several splice variants of group I and III mGlu receptors have been identified: they differ mainly in their C-terminal domains and thus in their ability to interact with intracellular proteins [5]. Although members of each of the mGlu receptor groups can be located at presynaptic and postsynaptic sites in the brain, group I receptors have been shown to display a characteristic perisynaptic localization at the postsynaptic membrane of glutamatergic synapses [6], where they can regulate neuronal excitability by modulating a variety of Kþ channels and ionotropic glutamate (iGlu) receptors of the N-methyl-D -aspartic acid (NMDA) or a-amino-3hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) subtypes. Conversely, group II (which can also be postsynaptic) and group III mGlu receptors are expressed at presynaptic terminals, where they inhibit the release of glutamate and other neurotransmitters [7]. Some subtypes, and in particular mGlu3 and mGlu5 receptors, have also been detected in non-neuronal cells, including astrocytes and microglia. Their expression in these cells is dynamically regulated in response to brain stimulation and injury, which is suggestive of a mechanism for the modulation of glial function and glial – neuronal communication [8]. Following the initial observation that in native tissue mGlu5 receptors exist almost exclusively as homodimers stabilized by disulfide bonds, the concept has been extended to other mGlu subtypes and GPCRs (Figure 1). The mGlu1 receptor ECDs have been shown to form dimeric structures that are dramatically modified in their conformation following glutamate binding, thus indicating that the dimerization process might play a crucial role in mGlu receptor activation [9]. Evidence has accumulated in the past few years that mGlu receptors contribute to the neurotoxic effects of glutamate (also known as ‘excitotoxicity’) and are implicated in the mechanisms that lead to neurodegeneration in models of cerebral ischemia [8]. Because of their presynaptic inhibitory effects, group II and III mGlu receptor subtypes, which are negatively coupled to adenylyl cyclase, have also been proposed as potential targets for neuroprotective drugs in various disease models. However, mGlu1 and mGlu5 receptors, which are coupled to PLC

http://tips.trends.com 0165-6147/$ - see front matter q 2003 Elsevier Ltd. All rights reserved. doi:10.1016/S0165-6147(03)00231-1

462

Review

TRENDS in Pharmacological Sciences

(a)

Vol.24 No.9 September 2003

NH2

NH2 –S-S–

ECD

LB1 1

LB1

Glu

Glu

LB2

LB2

Cys-rich region 7TMD

1

2

3

4 α

Gq protein

AM

5 β

6

7

AM

1

2

γ

3 α

4 β

5

Receptor group

I

COOH

Receptor subtypes

Signal transduction

mGlu1α mGlu1β mGlu1d mGlu1e mGlu5a mGlu5b

7

γ

COOH

(b)

6

PtdIns(4,5)P2 Gq

PLC

+

DAG PKC + Ins(1,4,5)P3 Ca2+

mGlu2 II mGlu3 mGlu4a mGlu6

III

mGlu7a mGlu7b mGlu7c mGlu7d mGlu7e

Gi

AC cAMP

– ATP

mGlu8a mGlu8b TRENDS in Pharmacological Sciences

Fig. 1. Proposed secondary structure and classification of metabotropic glutamate (mGlu) receptors. (a) Schematic representation of a homodimeric group I mGlu receptor stabilized by a disulfide bond. Sharing structural similarities with bacterial perisplasmic binding proteins, the large N-terminal extracellular domain (ECD) of each protomer consists of two globular ligand binding (LB1 and LB2) lobes linked by a hinge region where the agonist glutamate (Glu) binds. Agonists are thought to stabilize an active closed conformation by making contacts with residues of both lobes, whereas competitive antagonists bind at the same site as agonists but are unable to stabilize the active conformation. The ECD is connected to the seven-transmembrane domain (7TMD) by a cysteine-rich region. Gq proteins are associated with intracellular loops 2 and 3, whereas allosteric modulators (AMs) interact with TMD 6 and 7 (the coexisting tridimensional interaction with TMD 3 is not shown). (b) The features of each mGlu receptor family group are shown. Greek letters are used for mGlu1a and mGlu1b splice variants, according to their original designation. Splice variants mGlu1c and mGlu4b are not included because data from different laboratories strongly suggest that they do not exist as full-length transcripts but probably represent recombination artifacts ([97] and F. Ferraguti, pers. commun.). Group I mGlu receptors are coupled to phospholipase C (PLC) via Gq proteins: their stimulation promotes the breakdown of phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P2] into inositol (1,4,5)-trisphosphate [Ins(1,4,5)P3] and diacylglycerol (DAG). Ins(1,4,5)P3 releases Ca2þ from intracellular stores, whereas DAG activates protein kinase C (PKC). Group II and III mGlu receptors are negatively coupled via Gi/o proteins to adenylyl cyclase (AC): their stimulation inhibits the formation of cAMP from its precursor ATP. http://tips.trends.com

Review

TRENDS in Pharmacological Sciences

activation and are associated with several potentially detrimental events, have received particular attention for their possible role in post-ischemic neuronal death [10,11]. The results of these studies indicate that group I mGlu receptor subtypes play a different role in models of cerebral ischemia and that mGlu1 rather than mGlu5 receptors are implicated in the pathways leading to post-ischemic neuronal injury. Group I mGlu receptor subtypes are attractive targets for anti-ischemic drugs According to the World Health Organization, stroke is the second leading cause of death and the most common cause of disability in the world. Unfortunately, clinical trials of neuroprotective drugs for the treatment of stroke have been generally unsuccessful so far. Therefore, together with fine-tuning of the design of these trials and of the development of more appropriate experimental animal models, drugs that possess a better therapeutic index and are aimed at alternative targets in the excitotoxic cascade that is responsible for neuronal death are required for the development of new and effective neuroprotective therapies for cerebral ischemia. Group I mGlu receptor antagonists represent a valid alternative as anti-ischemic drugs. In addition to the classical activation of PLC and release of Ca2þ from intracellular stores, stimulation of mGlu1 and mGlu5 receptors can trigger a variety of signaling cascades and modulate the activity of ion and ligand-gated channels through functional coupling with other transduction pathways such as adenylyl cyclase, phospholipase A2, phospholipase D, tyrosine kinase and mitogen-activated protein kinase [12,13]. Activation of mGlu1 and mGlu5 receptors might thus promote multiple processes that are known to participate in the pathological cascade leading to post-ischemic neuronal death, including: (i) an increase in neuronal excitability caused by the activation of inward cationic currents or the reduction of Kþ conductances; (ii) a rise in cytosolic free Ca2þ via a facilitatory coupling between ryanodine receptors and L-type Ca2þ channels or direct Ca2þ influx from the extracellular space through NMDA receptors and L-type channels [14]; (iii) an enhancement of the release of glutamate that correlates with the neurotoxic effects of group I mGlu receptor agonists [15]; (iv) a potentiation of NMDA and AMPA receptor responses that has been observed in a large number of brain areas; and (v) activation of the mitogenactivated protein kinase pathway via PKC [16]. mGlu1 but not mGlu5 receptor antagonists are neuroprotective in models of cerebral ischemia The assumption that pharmacological stimulation of group I mGlu receptors invariably leads to neurotoxic effects is too simplistic [11]. The poor selectivity of ligands (Table 1), the possible coupling of these receptors to alternative signaling pathways, and the observed neuroprotective effects of elevated cytosolic free Ca2þ [17] and PKC activation [18] under certain conditions are all factors that could explain the conflicting results observed with mGlu1 and/or mGlu5 receptor agonists. It has been proposed that the capability of group I mGlu receptors to http://tips.trends.com

Vol.24 No.9 September 2003

463

elicit neuroprotection rather than toxicity might depend on the expression of the NR2C subunit of NMDA receptors [19]. Activation of group I mGlu receptors in cerebellar granule cells attenuates the rise in intracellular Ca2þ and neuronal death driven by NMDA receptors, but when the expression of NR2C subunits is knocked down by antisense oligonucleotides the stimulation of both group I mGlu receptors and PKC enhances the effects of NMDA. Interestingly, the expression of the NR2C subunit is highest in the cerebellum and barely detectable in the hippocampus, striatum and neocortex [20], the regions that are most sensitive to ischemic damage. Another possible explanation has been suggested following studies in mixed cortical cells: two consecutive applications of the group I receptor agonist (S)-3,5-DHPG have been shown to induce a PKC-dependent functional switch of the receptors from a ‘neurotoxic’ to a ‘neuroprotective’ mode [21]. Contrary to the experimental observations with agonists, the use of mGlu1 receptor antagonists (Table 1) has consistently generated neuroprotective results, indicating that the endogenous activation of this receptor subtype is likely to play a major role in neurodegeneration. In the past few years, competitive and noncompetitive antagonists displaying increasing degrees of selectivity for mGlu1 receptors have been shown to reduce NMDA receptormediated neurotoxicity in vitro and in vivo [15,22 – 24] in addition to neuronal injury in a variety of models of cerebral ischemia (Table 2). Neuroprotection in cultured neocortical cells and in hippocampal slices exposed to oxygen– glucose deprivation (OGD) has been observed following the application of both mGlu1 receptor-preferring antagonists that are known to interact with other mGlu receptor subtypes, such as (S)-3-carboxy-4-hydroxyphenylglycine [(S)-4C3HPG], 1-aminoindan-1,5-dicarboxylic acid (AIDA) or (S)-(þ)-2-(30 -carboxybicyclo[1.1.1]pentyl)-glycine [(S)-CBPG] [25 –27], and more selective and potent compounds such as LY367385 (see Chemical names), 7-hydroxyiminocyclopropan[b]chromen-1a-carboxylic acid ethyl ester (CPCCOEt) or 3-methyl-aminothiophene dicarboxylic acid (3-MATIDA) [28,29]. In these in vitro models, the neuroprotective effects are evident even when Chemical names BAY367620: (3aS,6aS)-6a-naphtalen-2-ylmethyl-5-methyliden-hexahydro-cyclopental[c]furan-1-on LY344545: (2S)-2-amino-2-((1R,2R)-2-carboxycycloprop-1-yl)3-(9-xanthyl)propanoic acid LY367366: (RS)-a-thioxanthylmethyl-4-carboxyphenylglycine LY367385: (S)-2-methyl-4-carboxyphenylglycine LY393675: (S)-cis-a-thioxanthylmethyl-3-carboxycyclobutylglycine Ro016128: diphenylacetyl-carbamic acid ethyl ester Ro674853: 9H-xanthene-9-carbonyl-carbamic acid butyl ester Ro677476: (S)-2-(4-fluoro-phenyl)-1-(toluene-4-sulfonyl)-pyrrolidone SIB1757: 6-methyl-2-(phenylazo)pyridin-3-ol SIB1893: (E)-2-methyl-6-styrylpyridine SKF89976A: 1-(4,4-diphenyl-3-butenyl)-3-piperidinecarboxylic acid

464

Review

TRENDS in Pharmacological Sciences

Vol.24 No.9 September 2003

Table 1. Pharmacological tools to explore the function of group I mGlu receptorsa Ligand Agonists Nonselective L -Glutamate L -Quisqualate (1S-3R)-ACPD (S)-3HPG (S)-3,5-DHPG mGlu1-selective Ro016128b Ro677476b Ro674853b mGlu5-selective (RS)-CHPG (Z)-CBQA Antagonists Nonselective (S)-MCPG 4-BuHIB LY367366b LY393675b mGlu1-selective (S)-4C3HPG (S)-4CPG AIDA (S)-Homoquisqualate (S)-ACUDA (S)-TBPG (S)-CBPG LY367385b 3-MATIDA CPCCOEt BAY367620b EM-TBPC Dicarboxy-pyrrole n. 3 mGlu5-selective LY344545b SIB1757b SIB1893b MPEP MTEP

mGlu1

mGlu5

Notes

Refse

EC50 (mM) 1 – 184 0.03 –17 5 – 214 18 –600 6 – 60

1 –11 0.05 –0.3 5 –122 14 –35 0.7 –20

Endogenous, mGlu and iGlu receptor agonist AMPA receptor agonist Group II and III mGlu receptor agonist Weak group II mGlu receptor agonist –

0.21 0.17 0.07

. 10 .1 10

Positive allosteric modulatorc Positive allosteric modulatorc Positive allosteric modulatorc

. 10000 . 1000 IC50

750 11 (mM)

[90] [90] [90]

– –

40 –300 110 6.6 0.35

100 –500 97 5.6 0.48

Group II mGlu receptor antagonist – – –

11 –393 4 – 163 3 – 214 184 76 –232 69 25 –65 1.4 –12 6.3 7 – 23 0.16 0.015 –0.13 0.016

Partial agonist? Partial agonist? . 100 Agonist . 1000 . 1000 Partial agonist . 100 . 300 . 100 . 100 .1 . 16

mGlu2 receptor agonist Weak mGlu2 receptor agonist Weak mGlu2 receptor agonist – – – – – – Noncompetitive Noncompetitive, inverse agonistd Noncompetitive Noncompetitive

39.5 . 100 . 100 . 100 NP

5.5 0.37 0.29 0.032 0.005

Group II mGlu receptor antagonist Noncompetitive Noncompetitive Noncompetitive, inverse agonistd Noncompetitive, inverse agonistd

[91]

[29] [92] [93] [94] [95]

[96]

Abbreviations: (1S-3R)-ACPD, (1S-3R)-1-aminocyclopentane-1,3-dicarboxylic acid; (S)-ACUDA, (S)-2-(40 -carboxycubyl)glycine; (S)-AIDA, 1-aminoindan-1,5-dicarboxylic acid; AMPA, a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; 4-BuHIB, (S)-4-n-butylhomoibotenic acid; (S)-CBPG, (S)-(þ)-2-(30 -carboxybicyclo[1.1.1]pentyl)-glycine; (Z)-CBQA, 1-amino-3-[20 -(30 ,50 -dioxo-10 ,20 ,40 -oxadiazolidinyl)]cyclobutane-1-carboxylic acid; (S)-4C3HPG, (S)-3-carboxy-4-hydroxyphenylglycine; (RS)-CHPG, (RS)-2-chloro5-hydroxyphenylglycine; CPCCOEt, 7-hydroxyiminocyclopropan[b ]chromen-1a-carboxylic acid ethyl ester; 4CPG, (S)-4-carboxyphenylglycine; (S)-3,5-DHPG, (S)-3,5dihydroxyphenylglycine; dicarboxy-pyrrole n. 3, 3,5-dimethyl-pyrrole-2,4-dicarboxylic acid 2-propyl ester 4-(1,2,2-trimethyl-propyl) ester; EM-TBPC, 1-ethyl-2-methyl-6-oxo-4(1,2,4,5-tetrahydro-benzo[d ]azepin-3-yl)-1,6-dihydro-pyrimidine-5-carbonitrile; (S)-3HPG, (S)-3-hydroxyphenylglycine; iGlu receptor, ionotropic glutamate receptor; 3-MATIDA, 3-methyl-aminothiophene dicarboxylic acid; (S)-MCPG, (S)-a-methyl-4-carboxyphenylglycine; mGlu receptor, metabotropic glutamate receptor; MPEP, 2-methyl-6-(phenylethynyl)-pyridine; MTEP, 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine; NP, not published; (S)-TBPG, 2-(30 -(1H-tetrazol-5-yl)bicyclo[1.1.1]pentyl-1-yl)-glycine. b See Chemical names. c Positive allosteric modulators have no effect per se but potentiate the action of agonists by increasing their affinity, potency and efficacy. d Inverse agonists are compounds that inhibit agonist-independent constitutive activity of mGlu receptors in hetereologous cells. e Except where noted, data are from [4] and references therein. a

mGlu1 receptor antagonists are added to the incubation medium up to 60 min after OGD [29,30]. In addition, different laboratories have reported CA1 pyramidal cell neuroprotection using (S)-4C3HPG [31], LY367385 [22] and AIDA [26] in the transient two-vessel occlusion (2VO) gerbil model of global ischemia. Despite the initial observation that the targeted disruption of the gene encoding the mGlu1a receptor does not affect the extent of neuronal injury following permanent middle cerebral artery occlusion (MCAO) in mice [32], possibly as a result of compensatory mechanisms provided by other splice variants of mGlu1 receptors or perhaps mGlu5 receptors, more recent studies in the rat using selective and, in some cases, systemically active competitive and noncompetitive http://tips.trends.com

antagonists have shown that activation of mGlu1 receptors might also contribute to brain damage in models of focal ischemia [29,33,34]. Although mGlu5 receptors are physically and functionally associated with NMDA receptors [35,36] and their blockade has been demonstrated to be protective against NMDA- or b-amyloid-induced neurotoxicity [23,37], the results obtained using selective mGlu5 receptor antagonists in models of cerebral ischemia are scarce and not particularly encouraging (Table 2). Muralikrishna Rao and co-workers first demonstrated that the mGlu5 receptor-selective noncompetitive antagonist 2-methyl-6(phenylethynyl)-pyridine (MPEP) was more effective than AIDA in reducing CA1 pyramidal cell loss following

Review

TRENDS in Pharmacological Sciences

465

Vol.24 No.9 September 2003

Table 2. Effects of group I mGlu receptor antagonists in models of cerebral ischemia in vitro and in vivoa Drug mGlu1 receptor antagonists Oxygen –glucose deprivation (S)-4C3HPG AIDA, (S)-CBPG AIDA, (S)-CBPG (S)-4CPG LY367385b LY367385b CPCCOEt CPCCOEt 3-MATIDA 3-MATIDA Transient global ischemia (S)-4C3HPG LY367385b AIDA (S)-CBPG Focal ischemia (S)-4C3HPG BAY367620b LY367385b 3-MATIDA

Model

Effect

Refs

þ þ þ þ þ þ þ 2 þ þ

[25] [26 –28,30,60] [26,28,30] [27,60] [28,29] [28,29] [28] [28] [29] [29,57]

Gerbil 2VO Gerbil 2VO Gerbil 2VO Gerbil 2VO

þ þ þ þ

[31] [22] [26,28,30,57] [26,28,30,57]

pMCAO pMCAO pMCAO pMCAO

þ ^ þ þ

[33] [34] [29] [29]

Cortical cells Cortical cells Hippocampal Cortical cells Cortical cells Hippocampal Cortical cells Hippocampal Cortical cells Hippocampal

slices

slices slices slices

Drug

Model

Effect

Refs

mGlu5 receptor antagonists Oxygen –glucose deprivation MPEP MPEP

Cortical cells Hippocampal slices

2 2

[28] [28]

Transient global ischemia MPEP MPEP

Gerbil 2VO Gerbil 2VO

þ 2

[38] [28]

Focal ischemia MPEP MPEP

tMCAO pMCAO

þc 2

[39] [2]

Abbreviations: AIDA, 1-aminoindan-1,5-dicarboxylic acid; (S)-CBPG, (S)-(þ)-2-(30 -carboxybicyclo[1.1.1]pentyl)-glycine; (S)-4C3HPG, (S)-3-carboxy-4-hydroxyphenylglycine; CPCCOEt, 7-hydroxyiminocyclopropan[b ]chromen-1a-carboxylic acid ethyl ester; (S)-4CPG, (S)-4-carboxyphenylglycine; 3-MATIDA, 3-methyl-aminothiophene dicarboxylic acid; MPEP, 2-methyl-6-(phenylethynyl)-pyridine; pMCAO, permanent middle cerebral artery occlusion; tMCAO, transient middle cerebral artery occlusion; 2VO, two-vessel occlusion. Symbols: þ , neuroprotective;-, not neuroprotective; ^ , trend for a neuroprotective effect. b See Chemical names. c MPEP was protective only at a high dose, presumably via NMDA receptors. a

transient global ischemia in the gerbil [38], but this result could not be confirmed in a subsequent study, in which both the same (intraperitoneal) and a different (intracerebroventricular) route of administration were used [28]. Similarly, MPEP was not neuroprotective following OGD in vitro [28] (Figure 2) nor after permanent MCAO in the rat [2]. Moreover, incubation of cultured cortical cells and hippocampal slices with the nonselective group I mGlu receptor agonist (S)-3,5-DHPG but not with the selective mGlu5 receptor agonist (RS)-2-chloro-5-hydroxyphenylglycine [(S)-CHPG] significantly enhanced the extent of neuronal injury induced by OGD [28]. In rats subjected to transient focal ischemia, MPEP was able to reduce the size of the ischemic infarct when administered intracerebroventricularly at a high dose, but the protective effect was ascribed to a noncompetitive antagonism of NMDA receptors, rather than to an interaction with mGlu5 receptors [39]. Indeed, MPEP significantly reduces steady state NMDA-evoked whole-cell currents at high concentrations (20 –200 mM, 500– 5 000-fold above its IC50 value) that are neuroprotective against excitotoxicity and posttraumatic injury in cultured cortical neurons [40,41]. A differential pathophysiological role for mGlu1 and mGlu5 receptors in the CNS? Although mGlu1 and mGlu5 receptors are highly homologous and are both coupled to Gq proteins and phosphoinositide hydrolysis, they appear to serve different roles in the CNS [13]. Functional differences for mGlu1 and mGlu5 receptors are suggested by their distinct anatomical distribution but have also been detected when they are coexpressed in the same cells. For example, activation of mGlu1 receptors in transfected cells leads to a peaked rise in intracellular Ca2þ, whereas activation of mGlu5 http://tips.trends.com

receptors generates an oscillatory Ca2þ response, which is due to the presence of a threonine residue that is phosphorylated by PKC in the proximal region of the mGlu5 receptor C-terminal tail [42]. In hippocampal CA1 pyramidal cells, mGlu1 receptors mediate the depolarization and intracellular Ca2þ release evoked by the broad

255 201 147

94

40

TRENDS in Pharmacological Sciences

Fig. 2. CA1 pyramidal cell death induced by oxygen– glucose deprivation (OGD) in cultured hippocampal slices is attenuated by the metabotropic glutamate 1 (mGlu1) receptor antagonist 3-methyl-aminothiophene dicarboxylic acid (3-MATIDA) but not by the mGlu5 receptor antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP). Cultured slices were exposed to OGD for 30 min, incubated with propidium iodide (5 mg ml21) for 24 h and then photographed under fluorescence optics. The vertical scale shows different colors used to denote fluorescence intensity, in arbitrary units. Drugs were present in the medium during OGD and the subsequent 24 h recovery period. Scale bar ¼ 1.8 mm. Modified, with permission, from [28,29].

466

Review

TRENDS in Pharmacological Sciences

agonist the (S)-3,5-DHPG, whereas mGlu5 receptors are responsible for the (S)-3,5-DHPG-induced suppression of Ca2þ-activated Kþ currents and potentiation of NMDA receptor activation [43]. Despite a high level of mGlu1 and mGlu5 receptor coexpression, depolarization is evoked only by selective activation of mGlu5 receptors in the subthalamic nucleus [44] and of mGlu1 receptors in the substantia nigra pars reticulata [45] and in type II globus pallidus interneurons [46]. In striatal medium spiny neurons, induction of corticostriatal long-term depression requires the activation of mGlu1 but not mGlu5 receptors [47], whereas potentiation of NMDA receptor responses is mediated exclusively by mGlu5 receptors [48]. In cultured cortical neurons, stimulation of mGlu1 but not mGlu5 receptors induces potentiation of NMDA receptor currents through a Ca2þ – calmodulin-dependent and PKCindependent Pyk2/Src-family kinase pathway [49]. The specificity of mGlu1 and mGlu5 receptor functions in these neurons can also be explained by their diverse coupling to other downstream effectors or scaffolds. Recently, the activity of group I mGlu receptors has been shown to be tightly controlled by dynamic regulatory mechanisms such as the phosphorylation of specific mGlu1 or mGlu5 receptor domains by PKC and G-protein-coupled receptor kinases (GRKs), their interaction with regulators of G-protein signaling (RGS) proteins or their modulation by Homer and related proteins [13]. A distinct role for mGlu1 or mGlu5 receptors has been recognized not only in post-ischemic neuronal death (Table 2) but also in other experimental conditions that mimic human brain disease. For example, the use of antisense oligodeoxynucleotides directed to mGlu1 but not to mGlu5 receptors is neuroprotective in cortical neuronal cultures subjected to mechanical injury or following brain trauma in vivo [50]. In addition, selective mGlu1 receptor antagonists such as AIDA, CPCCOEt and LY367385 reduce traumatic neuronal injury in the same models [51,52], whereas the selective mGlu5 receptor antagonists MPEP and SIB1893 are neuroprotective only at high doses that antagonize NMDA receptors [41]. Studies with selective antagonists have established that mGlu1 and mGlu5 receptors play a complementary pathophysiological role in determining the histological and behavioral outcome following in vitro and in vivo spinal cord injury [53,54]. Although mGlu5 receptor antagonists suppress clonic seizures in vivo [55], a preferential role for mGlu1 receptors has also been shown in sustaining the prolongation of epileptiform bursts in hippocampal slices [56] and in cortical wedges [57] in vitro. By contrast, mGlu5 rather than mGlu1 receptor blockade appears to be beneficial in attenuating neurotoxicity induced by the active fragment of b-amyloid peptide [37,58] or methamphetamine [59]. Taken together, these experimental data show compelling evidence for the involvement of mGlu1 receptors in neurodegeneration but do not point to mGlu5 receptors as potentially important targets for neuroprotection against cerebral ischemia or traumatic injury. Differences in the release of arachidonic acid [38,60], the formation of cAMP [61] or the subtype of PKC [54] that is activated following activation of mGlu1 or mGlu5 receptors are among the possible explanations for http://tips.trends.com

Vol.24 No.9 September 2003

these findings. Also, the contribution of glial cells that express either mGlu1 or mGlu5 receptors [53,62] might account for the discrepant results obtained with antagonists in certain experimental models. Because activation of mGlu5 receptors supports neuronal survival during early development [63] and attenuates apoptosis in vitro [27], mGlu5 receptor antagonists might exacerbate apoptotic neuronal death following cerebral ischemia. On the contrary, mGlu5 receptors are likely to play a fundamental role in other CNS disorders, particularly in pain, anxiety, drug dependence and Parkinson’s disease [2,64]. Mechanisms of post-ischemic neuroprotection by mGlu1 receptor antagonists As discussed, activation of mGlu1 receptors might exacerbate post-ischemic neuronal injury through multiple noxious mechanisms, including an increase in intracellular free Ca2þ or the potentiation of NMDA receptor responses. However, the peculiar localization of this receptor subtype has prompted numerous studies in the past few years that have provided a new viewpoint on the neuroprotective mechanisms of mGlu1 receptor antagonists. In the CA1 hippocampal sub-region, the mGlu1a receptor splice variant is not expressed in vulnerable pyramidal cells, but is enriched in somatostatin-positive GABA-containing interneurons of the stratum oriensalveus [65], which appear to be resistant to global ischemia [66]. In a very recent study, mGlu1a receptor immunoreactivity was also identified in other types of interneurons that are located in various CA1 strata and target different pyramidal cell dendritic domains [67]. Similarly, mGlu1a receptors are expressed by GABA-containing interneurons in the neocortex [68], the striatum [69] and the substantia nigra pars reticulata [70]. These anatomical observations suggest that neuroprotection by mGlu1 receptor antagonists might be induced via changes in GABA-mediated transmission. Functional studies have shown that activation of mGlu1 receptors depresses inhibitory transmission in these brain areas [23,70,71] and that potentiation of GABA-mediated transmission is neuroprotective against post-ischemic injury [72]. The hypothesis that mGlu1 receptor antagonists attenuate post-ischemic injury by enhancing GABAmediated neurotransmission was first proposed by studies showing that perfusion with neuroprotective doses of AIDA enhanced the concentrations of GABA in the hippocampal dialysate of gerbils subjected to global ischemia [30]. In hippocampal slices exposed to OGD, the attenuation of CA1 damage observed with another mGlu1 receptor antagonist, 3-MATIDA, was mimicked by GABAA and GABAB receptor agonists and reduced by GABA receptor antagonists [57]. Likewise, LY367385 and CPCCOEt produced an increase in the extracellular levels of GABA in the corpus striatum of freely moving rats that was associated with a reduction in NMDA-mediated neurotoxicity [23]. In cultured neuronal cells exposed to NMDA, the neuroprotective effects of mGlu1 receptor antagonists were occluded by the previous application of GABA and SKF89976A (a GABA transporter inhibitor) and prevented by GABAA and GABAB receptor antagonists [23]. It is interesting to note that the reduction of

Review

TRENDS in Pharmacological Sciences

spontaneous epileptiform activity induced by 3-MATIDA was similarly reproduced by a GABA receptor agonist and reverted by an antagonist in mouse cortical wedges [57]. Taken together, these data suggest that a common GABA-mediated mechanism, which involves the release of GABA and stimulation of GABA receptors, might contribute to the neuroprotective effects of mGlu1 receptor antagonists (Figure 3). This hypothesis implies that mGlu1 receptors are located presynaptically and that their activation inhibits the release of GABA. Although most immunocytochemical studies have failed to detect a presynaptic localization of mGlu1 receptors, two recent reports provide electron microscopy evidence for mGlu1a receptors in GABAergic terminals that appear to be responsible for a decrease in

467

Vol.24 No.9 September 2003

inhibitory transmission in the substantia nigra [70,73]. Functional data support the existence of presynaptic mGlu1 receptors modulating neurotransmitter release in the hippocampus [74] and neocortex [75]. Their stimulation could lead to an inhibitory effect by suppressing Ca2þ currents through N- or P/Q-type channels, as observed in cortical neurons [76] and in heterologous systems [77], or by activation of a Ca2þ-dependent Kþ conductance [78]. In opposition to this inhibitory hypothesis, however, it has been reported that activation of mGlu1 receptors results in increased spike frequency and rhythmic firing activity in GABA-containing interneurons [79]. It is also possible that a presynaptic decrease in inhibitory transmission can be indirectly mediated by mGlu1 receptors located postsynaptically in principal

Principal neuron AMPA or NMDA receptor

Glutamatergic terminal

Na+, Ca2+

Glu

– GABAB receptor

mGlu1 receptor PtdIns (4,5)P2

mGlu1 receptor K+

PLC DAG DAG lipase

– Ca2+

+ K+

channel

+



Cl–

GABA GABAergic terminal

+



GABAA receptor

+ +

Ca2+ – 2-AG

K+ CB1 receptor

Ca2+ channel

TRENDS in Pharmacological Sciences

Fig. 3. Schematic model providing a possible explanation for the neuroprotective effects of metabotropic glutamate 1 (mGlu1) receptor antagonists. Excessive activation of postsynaptic AMPA and NMDA receptors by glutamate (Glu) produces a sustained depolarizing influx of Naþ and Ca2þ in principal neurons, which eventually leads to neurodegeneration. Conversely, activation of postsynaptic GABAA receptors produces an influx of Cl2, hyperpolarization and neuroprotection. GABA can also interact with presynaptic GABAB receptors that negatively control the release of glutamate from afferent terminals, thus leading to reduced excitation of postsynaptic neurons. The release of GABA from interneuron terminals is negatively controlled by mGlu1 receptors and cannabinoid CB1 receptors, via suppression of Ca2þ currents through N-type channels or activation of Kþ channels. Antagonists of mGlu1 receptors can lead to increased release of GABA, and therefore to neuroprotective hyperpolarization of principal neurons, by means of two mechanisms: direct blockade of presynaptic mGlu1 receptors or indirect inhibition of CB1 receptors located on GABAergic terminals. The latter mechanism is promoted by mGlu1 receptors located postsynaptically in principal neurons. Their activation leads to the formation of diacylglycerol (DAG), which can be converted to the cannabinoid 2-arachidonyl glycerol (2-AG) through the action of DAG lipase. 2-AG can diffuse through the membrane and act as a retrograde transmitter to activate CB1 receptors in presynaptic terminals. Abbreviations: PLC, phospholipase C; PtdIns(4,5)P2, phosphatidylinositol (4,5)-bisphosphate. http://tips.trends.com

468

Review

TRENDS in Pharmacological Sciences

neurons. Functional evidence exists for the presence of mGlu1 receptors in CA1 pyramidal cells [43] and, albeit not abundantly in the CA1 region, mGlu1b and mGlu1d splice variants are expressed in hippocampal principal neurons [80,81]. Activation of mGlu1 postsynaptic receptors is known to promote the release of endogenous cannabinoids from CA1 pyramidal cells [82] and cerebellar Purkinje cells [83]. Endocannabinoids could then act as retrograde transmitters to suppress the release of GABA following activation of cannabinoid CB1 receptors located on the presynaptic terminals of interneurons [84 – 87] (Figure 3). Concluding remarks Experimental evidence suggests that selective antagonism of mGlu1 receptors attenuates post-ischemic neuronal injury by enhancing GABA-mediated neurotransmission. This neuroprotective mechanism is unique, in that is not shared by the mGlu5 receptor, which is also coupled to phoshoinositide hydrolysis and Ca2þ mobilization, and resembles the ‘metabotropic’ mode of action of presynaptic kainate receptors [88]. Interestingly, mGlu1 receptor antagonists appear to act by increasing the release of GABA in other models of neurotoxicity, suggesting a more general applicability of this mechanism to other disorders involving glutamate-induced neuronal death. Although antibodies against mGlu1 receptors have been detected in two patients with severe ataxia [89], antagonists of group I mGlu receptors are expected to modulate neuronal excitability and modify the induction and progression of post-ischemic neuronal death without interfering with fast excitatory synaptic transmission, a deleterious mechanism that presumably accounts for most of the clinical sideeffects (e.g. sedation, ataxia and memory loss) that can be observed following the use of NMDA or AMPA receptor antagonists [17]. In addition, the knowledge of the detrimental mechanisms triggered by mGlu1 receptors can be helpful in designing new therapeutic approaches with drugs (including GABA-related drugs or cannabinoids) that intervene along the cascade of events that lead to neuronal death.

7

8

9 10 11

12

13

14

15

16

17 18

19

20 21

22

Acknowledgements I wish to thank Flavio Moroni, Guido Mannaioni and Alberto Chiarugi for critical discussions and Elena Meli and Andrea Cozzi for contributing ideas and for coordinating the experiments performed in our laboratory. I am grateful to Francesco Ferraguti for stimulating discussions and for permission to quote unpublished data.

23

24

References 1 De Blasi, A. et al. (2001) Molecular determinants of metabotropic glutamate receptor signaling. Trends Pharmacol. Sci. 22, 114 – 120 2 Gasparini, F. et al. (2002) Allosteric modulators of group I metabotropic glutamate receptors: novel subtype-selective ligands and therapeutic perspectives. Curr. Opin. Pharmacol. 2, 43 – 49 3 Conn, P.J. and Pin, J-P. (1997) Pharmacology and functions of metabotropic glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 37, 205 – 237 4 Schoepp, D.D. et al. (1999) Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology 38, 1431 – 1476 5 Dev, K.K. et al. (2001) Regulation of mglu(7) receptors by proteins that interact with the intracellular C-terminus. Trends Pharmacol. Sci. 22, 355 – 361 6 Luja´n, R. et al. (1996) Perisynaptic localization of metabotropic http://tips.trends.com

25

26

27

28

Vol.24 No.9 September 2003

glutamate receptors mGluR1 and mGluR5 on dendrites and dendritic spines in the rat hippocampus. Eur. J. Neurosci. 8, 1488 – 1500 Cartmell, J. and Schoepp, D.D. (2000) Regulation of neurotransmitter release by metabotropic glutamate receptors. J. Neurochem. 75, 889 – 907 Bruno, V. et al. (2001) Metabotropic glutamate receptor subtypes as targets for neuroprotective drugs. J. Cereb. Blood Flow Metab. 21, 1013– 1033 Kunishima, N. et al. (2000) Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature 407, 971– 977 Bordi, F. and Ugolini, A. (1999) Group I metabotropic glutamate receptors: implications for brain diseases. Prog. Neurobiol. 59, 55 – 79 Nicoletti, F. et al. (1999) Group-I metabotropic glutamate receptors: hypotheses to explain their dual role in neurotoxicity and neuroprotection. Neuropharmacology 38, 1477 – 1484 Hermans, E. and Challiss, R.A. (2001) Structural, signalling and regulatory properties of the group I metabotropic glutamate receptors: prototypic family C G-protein-coupled receptors. Biochem. J. 359, 465– 484 Valenti, O. et al. (2002) Distinct physiological roles of the Gq-coupled metabotropic glutamate receptors co-expressed in the same neuronal populations. J. Cell. Physiol. 191, 125 – 137 Fagni, L. et al. (2000) Complex interactions between mGluRs, intracellular Ca2 þ stores and ion channels in neurons. Trends Neurosci. 23, 80 – 88 Strasser, U. et al. (1998) Antagonists for group I mGluRs attenuate excitotoxic neuronal death in cortical cultures. Eur. J. Neurosci. 10, 2848– 2855 Calabresi, P. et al. (2001) Activation of metabotropic glutamate receptor subtype 1/protein kinase C/mitogen-activated protein kinase pathway is required for postischemic long-term potentiation in the striatum. Mol. Pharmacol. 60, 808 – 815 Lee, J-M. et al. (1999) The changing landscape of ischaemic brain injury mechanisms. Nature 399 (Suppl.), A7 – A14 Pizzi, M. et al. (1996) Metabotropic and ionotropic transducers of glutamate signal inversely control cytoplasmic Ca2þ concentration and excitotoxicity in cultured cerebellar granule cells: pivotal role of protein kinase C. Mol. Pharmacol. 49, 586– 594 Pizzi, M. et al. (1999) Reversal of glutamate excitotoxicity by activation of PKC-associated metabotropic glutamate receptors in cerebellar granule cells relies on NR2C subunit expression. Eur. J. Neurosci. 11, 2489– 2496 Monyer, H. et al. (1992) Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science 256, 1217– 1221 Bruno, V. et al. (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 Bruno, V. et al. (1999) Neuroprotective activity of the potent and selective mGlu1a metabotropic glutamate receptor antagonist, (þ )-2methyl-4-carboxyphenylglycine (LY367385): comparison with LY357366, a broader spectrum antagonist with equal affinity for mGlu1a and mGlu5 receptors. Neuropharmacology 38, 199 – 207 Battaglia, G. et al. (2001) Selective blockade of type-1 metabotropic glutamate receptors induces neuroprotection by enhancing gabaergic transmission. Mol. Cell. Neurosci. 17, 1071– 1083 Orlando, L.R. et al. (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 Buisson, A. and Choi, D.W. (1995) The inhibitory mGluR agonist, s-4carboxy-3-hydroxy-phenylglycine selectively attenuates NMDA neurotoxicity and oxygen-glucose deprivation-induced neuronal death. Neuropharmacology 34, 1081 – 1087 Pellegrini-Giampietro, D.E. et al. (1999) 1-Amino-1,5-dicarboxylic acid and (S)-(þ )-2-(30 -carboxybicyclo[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 Allen, J.W. et al. (2000) Activation of group I metabotropic glutamate receptors reduces neuronal apoptosis but increases necrotic cell death in vitro. Cell Death Differ. 7, 470 – 476 Meli, E. et al. (2002) Activation of mGlu1 but not mGlu5 metabotropic glutamate receptors contributes to post-ischemic neuronal injury in vitro and in vivo. Pharmacol. Biochem. Behav. 73, 439– 446

Review

TRENDS in Pharmacological Sciences

29 Moroni, F. et al. (2002) The novel and systemically active metabotropic glutamate 1 (mGlu1) receptor antagonist 3-MATIDA reduces postischemic neuronal death. Neuropharmacology 42, 741– 751 30 Pellegrini-Giampietro, D.E. et al. (1999) Protection with metabotropic glutamate 1 receptor antagonists in models of ischemic neuronal death: time-course and mechanisms. Neuropharmacology 38, 1607 – 1619 31 Henrich-Noack, P. et al. (1998) The mGlu receptor ligand (S)-4C3HPG protects neurons after global ischaemia in gerbils. Neuroreport 9, 985 – 988 32 Ferraguti, F. et al. (1997) Evidence against a permissive role of the metabotropic glutamate receptor 1 in acute excitotoxicity. Neuroscience 79, 1 – 5 33 Rauca, C. et al. (1998) (S)-4C3HPG reduces infarct size after focal cerebral ischemia. Neuropharmacology 37, 1649 – 1652 34 De Vry, J. et al. (2001) Neuroprotective and behavioral effects of the selective metabotropic glutamate mGlu(1) receptor antagonist BAY 36-7620. Eur. J. Pharmacol. 428, 203 – 214 35 Sheng, M. and Kim, E. (2000) The Shank family of scaffold proteins. J. Cell Sci. 113, 1851 – 1856 36 Alagarsamy, S. et al. (2001) Coordinate regulation of metabotropic glutamate receptors. Curr. Opin. Neurobiol. 11, 357 – 362 37 Bruno, V. et al. (2000) Selective blockade of metabotropic glutamate receptor subtype 5 is neuroprotective. Neuropharmacology 39, 2223 – 2230 38 Muralikrishna Rao, A. et al. (2000) Neuroprotection by group I metabotropic glutamate receptor antagonists in forebrain ischemia of gerbil. Neurosci. Lett. 293, 1 – 4 39 Bao, W.L. et al. (2001) Selective mGluR5 receptor antagonist or agonist provides neuroprotection in a rat model of focal cerebral ischemia. Brain Res. 922, 173– 179 40 O’Leary, D.M. et al. (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 41 Movsesyan, V.A. et al. (2001) mGluR5 antagonists 2-methyl-6(phenylethynyl)-pyridine and (E)-2-methyl-6-(phenylethynyl)-pyridine reduce traumatic neuronal injury in vivo and in vitro by antagonizing N-methyl-D-aspartate receptors. J. Pharmacol. Exp. Ther. 296, 41 – 47 42 Kawabata, S. et al. (1996) Control of calcium oscillations by phosphorylation of metabotropic glutamate receptors. Nature 383, 89 – 92 43 Mannaioni, G. et al. (2001) Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. J. Neurosci. 21, 5925 – 5934 44 Awad, H. et al. (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 45 Marino, M.J. et al. (2002) Haloperidol-induced alteration in the physiological actions of group I mGlus in the subthalamic nucleus and the substantia nigra pars reticulata. Neuropharmacology 43, 147– 159 46 Poisik, O.V. et al. (2003) Distinct functional roles of the metabotropic glutamate receptors 1 and 5 in the rat globus pallidus. J. Neurosci. 23, 122 – 130 47 Gubellini, P. et al. (2001) Selective involvement of mGlu1 receptors in corticostriatal LTD. Neuropharmacology 40, 839 – 846 48 Pisani, A. et al. (2001) Metabotropic glutamate receptor 5 mediates the potentiation of N-methyl-D-aspartate responses in medium spiny striatal neurons. Neuroscience 106, 579 – 587 49 Heidinger, V. et al. (2002) Metabotropic glutamate receptor 1-induced upregulation of NMDA receptor current: mediation through the Pyk2/ Src-family kinase pathway in cortical neurons. J. Neurosci. 22, 5452 – 5461 50 Mukhin, A. et al. (1996) Activation of metabotropic glutamate receptor mGluR1 contributes to post-traumatic neuronal injury. J. Neurosci. 16, 6012 – 6020 51 Faden, A.I. et al. (2001) Selective blockade of the mGluR1 receptor reduces traumatic neuronal injury in vitro and improves outcome after brain trauma. Exp. Neurol. 167, 435 – 444 52 Lyeth, B.G. et al. (2001) Group I metabotropic glutamate receptor antagonist reduces acute neuronal degeneration and behavioral deficits after traumatic brain injury in rats. Exp. Neurol. 169, 191– 199 53 Agrawal, S.K. et al. (1998) Role of group I metabotropic glutamate http://tips.trends.com

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72 73

74

75

Vol.24 No.9 September 2003

469

receptors in traumatic spinal cord white matter injury. J. Neurotrauma 15, 929 – 941 Mills, C.D. et al. (2002) Group I metabotropic glutamate receptors in spinal cord injury: roles in neuroprotection and the development of chronic central pain. J. Neurotrauma 19, 23 – 42 Chapman, A.G. et al. (2000) Anticonvulsant activity of two metabotropic glutamate group I antagonists selective for the mGlu5 receptor: 2-methyl-6- (phenylethynyl)-pyridine (MPEP), and (E)-6-methyl-2styryl-pyridine (SIB 1893). Neuropharmacology 39, 1567– 1574 Merlin, L.R. (2002) Differential roles for mGluR1 and mGluR5 in the persistent prolongation of epileptiform bursts. J. Neurophysiol. 87, 621– 625 Cozzi, A. et al. (2002) Metabotropic glutamate 1 (mGlu1) receptor antagonists enhance GABAergic neurotransmission: a mechanism for the attenuation of post-ischemic injury and epileptiform activity? Neuropharmacology 43, 119 – 130 Allen, J.W. et al. (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 Battaglia, G. et al. (2002) Selective blockade of mGlu5 metabotropic glutamate receptors is protective against methamphetamine neurotoxicity. J. Neurosci. 22, 2135 – 2141 Allen, J.W. et al. (2001) Exacerbation of neuronal death by activation of group I metabotropic glutamate receptors: role of NMDA receptors and arachidonic acid release. Exp. Neurol. 169, 449 – 460 Mao, L. and Wang, J.Q. (2002) Interactions between ionotropic and metabotropic glutamate receptors regulate cAMP response elementbinding protein phosphorylation in cultured striatal neurons. Neuroscience 115, 395 – 402 Catania, M.V. et al. (2001) Increased expression of neuronal nitric oxide synthase spliced variants in reactive astrocytes of amyotrophic lateral sclerosis human spinal cord. J. Neurosci. 21, RC148 Copani, A. et al. (1998) The metabotropic glutamate receptor mGlu5 controls the onset of developmental apoptosis in cultured cerebellar neurons. Eur. J. Neurosci. 10, 2173 – 2184 Spooren, W.P.J.M. et al. (2001) Novel allosteric antagonists shed light on mglu5 receptors and CNS disorders. Trends Pharmacol. Sci. 22, 331– 337 Baude, A. et al. (1993) The metabotropic glutamate receptor (mGluR1a) is concentrated at perisynaptic membrane of neuronal subpopulations as detected by immunogold reaction. Neuron 11, 771– 787 Blasco-Iba´n˜ez, J.M. and Freund, T.F. (1995) Synaptic input of horizontal interneurons in stratum oriens of the hippocampal CA1 subfield: structural basis of feed-back activation. Eur. J. Neurosci. 7, 2170– 2180 Ferraguti, F. et al. Immunolocalization of metabotropic glutamate receptor 1a (mGluR1a) in distinct classes of interneurons in the CA1 region of rat hippocampus. Hippocampus (in press) Stinehelfer, S. et al. (2000) Immunolocalization of mGluR1alpha in specific populations of local circuit neurons in the cerebral cortex. Brain Res. 861, 37 – 44 Tallaksen-Greene, S.J. et al. (1998) Localization of mGluR1a-like immunoreactivity and mGluR5-like immunoreactivity in identified populations of striatal neurons. Brain Res. 780, 210– 217 Marino, M.J. et al. (2001) Activation of group I metabotropic glutamate receptors produces a direct excitation and disinhibition of GABAergic projection neurons in the substantia nigra pars reticulata. J. Neurosci. 15, 7001 – 7012 Morishita, W. et al. (1998) Evidence for metabotropic glutamate receptor activation in the induction of depolarization-induced suppression of inhibition in hippocampal CA1. J. Neurosci. 18, 4870 – 4882 Schwartz-Bloom, R.D. and Sah, R. (2001) g-Aminobutyric acidA neurotransmission and cerebral ischemia. J. Neurochem. 77, 353 – 371 Hubert, G.W. et al. (2001) Differential subcellular localization of mGluR1a and mGluR5 in the rat and monkey substantia nigra. J. Neurosci. 21, 1838– 1847 Manahan-Vaughan, D. et al. (1999) Presynaptic group 1 metabotropic glutamate receptors may contribute to the expression of long-term potentiation in the hippocampal CA1 region. Neuroscience 94, 71– 82 Moroni, F. et al. (1998) Presynaptic mGlu1 type receptors potentiate transmitter output in the rat cortex. Eur. J. Pharmacol. 347, 189 – 195

470

Review

TRENDS in Pharmacological Sciences

76 Choi, S. and Lovinger, D.M. (1996) Metabotropic glutamate receptor modulation of voltage-gated Ca2þ-channels involves multiple receptor subtypes in cortical neurons. J. Neurosci. 16, 36 – 45 77 McCool, B.A. et al. (1998) Rat group I metabotropic glutamate receptors inhibit neuronal Ca2þ channels via multiple signal transduction pathways in HEK 293 cells. J. Neurophysiol. 79, 379– 391 78 Fiorillo, C.D. and Williams, J.T. (1998) Glutamate mediates an inhibitory postsynaptic potential in dopamine neurons. Nature 394, 78 – 82 79 van Hooft, J.A. et al. (2000) Differential expression of group I metabotropic glutamate receptors in functionally distinct hippocampal interneurons. J. Neurosci. 20, 3544– 3551 80 Berthele, A. et al. (1998) Differential expression of rat and human type I metabotropic glutamate receptor splice variant messenger RNAs. Neuroscience 85, 733– 749 81 Ferraguti, F. et al. (1998) Immunohistochemical localization of the mGluR1b metabotropic glutamate receptor in the adult rodent forebrain: evidence for a different distribution of mGluR1 splice variants. J. Comp. Neurol. 400, 391 – 407 82 Varma, N. et al. (2001) Metabotropic glutamate receptors drive the endocannabinoid system in hippocampus. J. Neurosci. 21, RC188 83 Maejima, T. et al. (2001) Presynaptic inhibition caused by retrograde signal from metabotropic glutamate to cannabinoid receptors. Neuron 31, 463 – 475 84 Alger, B.E. (2002) Retrograde signaling in the regulation of synaptic transmission: focus on endocannabinoids. Prog. Neurobiol. 68, 247–286 85 Diana, M.A. et al. (2002) Short-term retrograde inhibition of GABAergic synaptic currents in rat Purkinje cells is mediated by endogenous cannabinoids. J. Neurosci. 22, 200 – 208 86 Gerdeman, G.L. et al. (2002) Postsynaptic endocannabinoid release is critical to long-term depression in the striatum. Nat. Neurosci. 5, 446 – 451 87 Chevaleyre, V. and Castillo, P.E. (2003) Heterosynaptic LTD of

88

89

90

91

92

93

94

95

96 97

Vol.24 No.9 September 2003

hippocampal GABAergic synapses. A novel role of endocannabinoids in regulating excitability. Neuron 38, 461 – 472 Rodriguez-Moreno, A. and Lerma, J. (1998) Kainate receptor modulation of GABA release involves a metabotropic function. Neuron 20, 1211 – 1218 Sillevis Smitt, P. et al. (2000) Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N. Engl. J. Med. 342, 21 – 27 Knoflach, F. et al. (2001) Positive allosteric modulators of metabotropic glutamate 1 receptor: characterization, mechanism of action, and binding site. Proc. Natl. Acad. Sci. U. S. A. 98, 13402 – 13407 Costantino, G. et al. (2001) Synthesis and biological evaluation of 2-(30 -(1H-tetrazol-5-yl) bicyclo[1.1.1]pent-1-yl)glycine (S-TBPG), a novel mGlu1 receptor antagonist. Bioorg. Med. Chem. 9, 221– 227 Carroll, F.Y. et al. (2001) BAY36-7620: a potent non-competitive mGlu1 receptor antagonist with inverse agonist activity. Mol. Pharmacol. 59, 965– 973 Malherbe, P. et al. (2003) Mutational analysis and molecular modeling of the allosteric binding site of a novel, selective, noncompetitive antagonist of the metabotropic glutamate 1 receptor. J. Biol. Chem. 278, 8340– 8347 Micheli, F. et al. (2003) Synthesis and pharmacological characterisation of 2,4-dicarboxy-pyrroles as selective non-competitive mGluR1 antagonists. Bioorg. Med. Chem. 11, 171 – 183 Doherty, A.J. et al. (2000) A novel, competitive mGlu(5) 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 Brodkin, J. et al. (2002) Reduced stress-induced hyperthermia in mGluR5 knockout mice. Eur. J. Neurosci. 16, 2241 – 2244 Corti, C. et al. (2002) Distribution and synaptic localisation of the metabotropic glutamate receptor 4 (mGluR4) in the rodent CNS. Neuroscience 110, 403 – 420

Cytochrome P450 monoclonal antibodies For scientists interested in drug metabolism, reduction of adverse drug reactions and drug discovery, the National Cancer Institute (NCI) of the National Institutes of Health (NIH) is making available specific inhibitory monoclonal antibodies (mAbs) to human cytochrome P450 1A1, 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2C family, 2D6, 3A4/5 and 2E1. Requests for specific mAbs and the Material Transfer Agreement should be sent to Patrick Twomey, Technology Transfer Branch (e-mail: [email protected]). Requests for larger amounts of the mAbs can be obtained by license by contacting Fatima Sayyid, Office of Technology Transfer (e-mail: [email protected]). http://tips.trends.com