Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 332(3): 1064-71, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19940105

ABSTRACT

Metabotropic glutamate (mGlu) receptors modulate synaptic transmission in the central nervous system and represent promising therapeutic targets for symptomatic treatment of Parkinson's disease (PD). Among the eight mGlu receptor subtypes, mGlu7 receptor is prominently expressed in the basal ganglia, but its role in restoring motor function in animal models of PD is not known. The effects of N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082), the first selective allosteric activator of mGlu7 receptors, were thus tested in different rodent models of PD. Here, we show that oral (5 mg/kg) or intrastriatal administration (0.1 and 0.5 nmol) of AMN082 reverses haloperidol-induced catalepsy in rats. AMN082 (2.5 and 5 mg/kg) reduces apomorphine-induced rotations in unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats. In a more complex task commonly used to evaluate major akinetic symptoms of PD patients, 5 mg/kg AMN082 reverses the increased reaction time to respond to a cue of bilateral 6-OHDA-lesioned rats. In addition, AMN082 reduces the duration of haloperidol-induced catalepsy in a mGlu7 receptor-dependent manner in wild-type but not mGlu7 receptor knockout mice. Higher doses of AMN082 (10 and 20 mg/kg p.o.) have no effect on the same models of PD. Overall these findings suggest that mGlu7 receptor activation can reverse motor dysfunction associated with reduced dopamine activity. Selective ligands of mGlu7 receptor subtypes may thus be considered as promising compounds for the development of antiparkinsonian therapeutic strategies.


Subject(s)
Parkinson Disease, Secondary/physiopathology , Receptors, Metabotropic Glutamate/physiology , Allosteric Regulation , Animals , Apomorphine/pharmacology , Benzhydryl Compounds/pharmacology , Catalepsy/chemically induced , Catalepsy/physiopathology , Disease Models, Animal , Haloperidol , Male , Mice , Mice, Knockout , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Wistar , Reaction Time/drug effects , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Stereotyped Behavior/drug effects
2.
Mol Psychiatry ; 13(10): 970-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-17712315

ABSTRACT

Formation and extinction of aversive memories in the mammalian brain are insufficiently understood at the cellular and molecular levels. Using the novel metabotropic glutamate receptor 7 (mGluR7) agonist AMN082, we demonstrate that mGluR7 activation facilitates the extinction of aversive memories in two different amygdala-dependent tasks. Conversely, mGluR7 knockdown using short interfering RNA attenuated the extinction of learned aversion. mGluR7 activation also blocked the acquisition of Pavlovian fear learning and its electrophysiological correlate long-term potentiation in the amygdala. The finding that mGluR7 critically regulates extinction, in addition to acquisition of aversive memories, demonstrates that this receptor may be relevant for the manifestation and treatment of anxiety disorders.


Subject(s)
Amygdala/physiology , Avoidance Learning/physiology , Extinction, Psychological/physiology , Memory/physiology , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/physiology , Amygdala/cytology , Amygdala/drug effects , Animals , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electric Stimulation , Extinction, Psychological/drug effects , Glutamic Acid/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Protein Binding/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Transfection
3.
Neurobiol Dis ; 26(3): 623-33, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17446080

ABSTRACT

Altered expression and distribution of neurotransmitter receptors, including metabotropic glutamate receptors (mGluRs), constitute key aspects in epileptogenesis, impaired hippocampal excitability and neuronal degeneration. mGluR1 mediates predominantly excitatory effects, whereas mGluR4 acts as inhibitory presynaptic receptor. Increased hippocampal expression of mGluR1 and mGluR4 has been observed in human temporal lobe epilepsy (TLE). In this study, we address whether genetic mGluR1 upregulation and mGluR4 knock-down influence seizure susceptibility and/or vulnerability of hippocampal neurons by analyzing transgenic animals in the pilocarpine TLE model. Therefore, we generated transgenic mice expressing mGluR1-enhanced green fluorescent protein (EGFP) fusion protein under control of the human cytomegalovirus (CMV) immediate early promoter. Status epilepticus (SE) was induced in (a) mice overexpressing mGluR1-EGFP and (b) mice deficient for mGluR4 (mGluR4 KO) as well as littermate controls. In the acute epileptic stage after pilocarpine application, mGluR4 KO mice showed a significant increase of severe seizure activity, in contrast to mGluR1 transgenics. Analysis of both transgenic mouse lines in the chronic epileptic phase, using a telemetric EEG-/video-monitoring system, revealed a significant increase in seizure frequency only in mGluR1-EGFP mice. In contrast, enhanced neuronal cell loss was only present in the hippocampus of epileptic mGluR4 KO mice. Our results suggest a role for mGluR1 in promoting seizure susceptibility as well as for mGluR4 to counteract excitatory activity and seizure-associated vulnerability of hippocampal neurons. Therefore, our data strongly recommend both mGluRs as potential drug targets to interfere with the development of hippocampal damage and seizure activity in TLE.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Genetic Predisposition to Disease/genetics , Hippocampus/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Convulsants , Disease Models, Animal , Down-Regulation/physiology , Epilepsy/metabolism , Epilepsy/physiopathology , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/physiopathology , Gene Expression Regulation/genetics , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Hippocampus/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/pathology , Pilocarpine , Receptors, Metabotropic Glutamate/genetics , Recombinant Fusion Proteins/genetics , Up-Regulation/physiology
4.
Neuropharmacology ; 45(7): 895-906, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14573382

ABSTRACT

Group-III metabotropic glutamate receptors (mGluR4, -6, -7, and -8) modulate neurotoxicity of excitatory amino acids and beta-amyloid-peptide (betaAP), as well as epileptic convulsions, most likely via presynaptic inhibition of glutamatergic neurotransmission. Due to the lack of subtype-selective ligands for group-III receptors, we previously utilized knock-out mice to identify mGluR4 as the primary receptor mediating neuroprotection of unselective group-III agonists such as L-AP(4) or (+)-PPG, whereas mGluR7 is critical for anticonvulsive effects. In a recent effort to find group-III subtype-selective drugs we identified (+/-)-PHCCC as a positive allosteric modulator for mGluR4. This compound increases agonist potency and markedly enhances maximum efficacy and, at higher concentrations, directly activates mGluR4 with low efficacy. All the activity of (+/-)-PHCCC resides in the (-)-enantiomer, which is inactive at mGluR2, -3, -5a, -6, -7b and -8a, but shows partial antagonist activity at mGluR1b (30% maximum antagonist efficacy). Chimeric receptor studies showed that the binding site of (-)-PHCCC is localized in the transmembrane region.Finally, (-)-PHCCC showed neuroprotection against betaAP- and NMDA-toxicity in mixed cultures of mouse cortical neurons. This neuroprotection was additive to that induced by the highly efficacious mGluR1 antagonist CPCCOEt and was blocked by MSOP, a group-III mGluR antagonist. Our data provide evidence for a novel pharmacological site on mGluR4, which may be used as a target-site for therapeutics.


Subject(s)
Benzopyrans/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Calcium/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cloning, Molecular , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/toxicity , Excitatory Amino Acids/toxicity , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/toxicity , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/drug effects
5.
Amino Acids ; 23(1-3): 207-11, 2002.
Article in English | MEDLINE | ID: mdl-12373539

ABSTRACT

There is a need to identify subtype-specific ligands for mGlu receptors to elucidate the potential of these receptors for the treatment of nervous system disorders. To date, most mGlu receptor antagonists are amino acid-like compounds acting as competitive antagonists at the glutamate binding site located in the large extracellular N-terminal domain. We have characterized novel subtype-selective mGlu(5) receptor antagonists which are structurally unrelated to competitive mGlu receptor ligands. Using a series of chimeric receptors and point mutations we demonstrate that these antagonists act as inverse agonists with a novel allosteric binding site in the seven-transmembrane domain. Recent studies in animal models implicate mGlu(5) receptors as a potentially important therapeutic target particularly for the treatment of pain and anxiety.


Subject(s)
Excitatory Amino Acid Antagonists/metabolism , Ligands , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Anti-Anxiety Agents/metabolism , Anxiety/drug therapy , Anxiety/metabolism , Binding Sites , Brain/metabolism , Excitatory Amino Acid Antagonists/therapeutic use , Pain/drug therapy , Pain/metabolism , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
6.
J Neurosci ; 21(22): 8734-45, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698585

ABSTRACT

To study the role of mGlu7 receptors (mGluR7), we used homologous recombination to generate mice lacking this metabotropic receptor subtype (mGluR7(-/-)). After the serendipitous discovery of a sensory stimulus-evoked epileptic phenotype, we tested two convulsant drugs, pentylenetetrazole (PTZ) and bicuculline. In animals aged 12 weeks and older, subthreshold doses of these drugs induced seizures in mGluR7(-/-), but not in mGluR7(+/-), mice. PTZ-induced seizures were inhibited by three standard anticonvulsant drugs, but not by the group III selective mGluR agonist (R,S)-4-phosphonophenylglycine (PPG). Consistent with the lack of signs of epileptic activity in the absence of specific stimuli, mGluR7(-/-) mice showed no major changes in synaptic properties in two slice preparations. However, slightly increased excitability was evident in hippocampal slices. In addition, there was slower recovery from frequency facilitation in cortical slices, suggesting a role for mGluR7 as a frequency-dependent regulator in presynaptic terminals. Our findings suggest that mGluR7 receptors have a unique role in regulating neuronal excitability and that these receptors may be a novel target for the development of anticonvulsant drugs.


Subject(s)
Genetic Predisposition to Disease , Glycine/analogs & derivatives , Receptors, Metabotropic Glutamate/deficiency , Seizures/genetics , Animals , Anticonvulsants/pharmacology , Bicuculline , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Convulsants , Drug Resistance/genetics , Electroencephalography , Excitatory Amino Acid Agonists/pharmacology , Gene Targeting , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/physiopathology , Homozygote , In Vitro Techniques , Mice , Mice, Knockout , Pentylenetetrazole , Phenotype , Physical Chromosome Mapping , Receptors, Metabotropic Glutamate/genetics , Seizures/chemically induced , Seizures/physiopathology , Seizures/prevention & control
7.
J Cereb Blood Flow Metab ; 21(9): 1013-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524608

ABSTRACT

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 BAY-36-7620; 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, because 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.


Subject(s)
Brain Diseases/drug therapy , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Humans
8.
Eur J Neurosci ; 13(8): 1469-78, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11328342

ABSTRACT

Activation of group I metabotropic glutamate receptors (mGlu1 or -5 receptors) is known to either enhance or attenuate excitotoxic neuronal death depending on the experimental conditions. We have examined the possibility that these receptors may switch between two different functional modes in regulating excitotoxicity. In mixed cultures of cortical cells, the selective mGlu1/5 agonist, 3,5-dihydroxyphenylglycine (DHPG), amplified neurodegeneration induced by a toxic pulse of NMDA. This effect was observed when DHPG was either combined with NMDA or transiently applied to the cultures prior to the NMDA pulse. However, two consecutive applications of DHPG consistently produced neuroprotection. Similar effects were observed with DHPG or quisqualate (a potent agonist of mGlu1/5 receptors) in pure cultures of cortical neurons virtually devoid of astrocytes. In cultures of hippocampal pyramidal neurons, however, only protective effects of DHPG were seen suggesting that, in these particular cultures, group I mGlu receptors were endogenously switched into a "neuroprotective mode". The characteristics of the activity-dependent switch from facilitation to inhibition were examined in mixed cultures of cortical cells. The switch in the response to DHPG was observed when the two applications of the drug were separated by an interval ranging from 1-45 min, but was lost when the interval was extended to 90 min. In addition, this phenomenon required the initial activation of mGlu5 receptors (as indicated by the use of subtype-selective antagonists) and was mediated by the activation of protein kinase C. We conclude that group I mGlu receptors are subjected to an activity-dependent switch in regulating excitotoxic neuronal death and, therefore, the recent "history" of these receptors is critical for the response to agonists or antagonists.


Subject(s)
Neural Inhibition/physiology , Neurotoxins/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Enzyme Activation/physiology , Excitatory Amino Acid Agonists/poisoning , Excitatory Amino Acid Antagonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Mice , N-Methylaspartate/poisoning , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Protein Kinase C/metabolism , Quisqualic Acid/pharmacology , Receptor, Metabotropic Glutamate 5 , Resorcinols/pharmacology , Time Factors
9.
Farmaco ; 56(1-2): 95-9, 2001.
Article in English | MEDLINE | ID: mdl-11347975

ABSTRACT

We have investigated the mechanism of inhibition of the new group I mGluR antagonists CPCCOEt and MPEP and determined that both compounds have a non-competitive mode of inhibition. Furthermore using chimeric/mutated receptors constructs we have found that these antagonists act at a novel pharmacological site located in the trans-membrane (TM). Specific non-conserved amino acid residues in the TM domain have been identified which are necessary for the inhibition by CPCCOEt and MPEP of the mGlul and mGlu5 receptors, respectively. Using molecular modeling a model of the TM domain was built for both mGlu1 and mGlu5 receptor subtypes. Docking of CPCCOEt and MPEP into their respective model allowed the modelisation of the novel binding site.


Subject(s)
Receptors, Metabotropic Glutamate/antagonists & inhibitors , Binding Sites , Models, Molecular , Receptors, Metabotropic Glutamate/chemistry
10.
Neuropharmacology ; 40(1): 1-9, 2001.
Article in English | MEDLINE | ID: mdl-11077065

ABSTRACT

The excitatory neurotransmitter, glutamate, is particularly important in the transmission of pain information in the nervous system through the activation of ionotropic and metabotropic glutamate receptors. A potent, subtype-selective antagonist of the metabotropic glutamate-5 (mGlu5) receptor, 2-methyl-6-(phenylethynyl)-pyridine (MPEP), has now been discovered that has effective anti-hyperalgesic effects in models of inflammatory pain. MPEP did not affect rotarod locomotor performance, or normal responses to noxious mechanical or thermal stimulation in naïve rats. However, in models of inflammatory pain, systemic administration of MPEP produced effective reversal of mechanical hyperalgesia without affecting inflammatory oedema. In contrast to the non-steroidal anti-inflammatory drugs, indomethacin and diclofenac, the maximal anti-hyperalgesic effects of orally administered MPEP were observed without acute erosion of the gastric mucosa. In contrast to its effects in models of inflammatory pain, MPEP did not produce significant reversal of mechanical hyperalgesia in a rat model of neuropathic pain.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Nociceptors/drug effects , Pain/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Chronic Disease , Excitatory Amino Acid Antagonists/adverse effects , Hyperalgesia/drug therapy , Male , Motor Activity/drug effects , Pain/psychology , Pain Measurement/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology
11.
J Med Chem ; 43(23): 4428-36, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11087567

ABSTRACT

Racemic CPCCOEt ((1aRS,7aRS)-2-hydroxyimino-1a, 2-dihydro-1H-7-oxacyclopropa[b]naphthalene-7a-carboxylic acid ethyl ester, (+/-)-1) derivatives have been shown to be subtype-selective metabotropic glutamate (mGlu) 1 receptor antagonists (Annoura et al. Bioorg. Med. Chem. Lett. 1996, 6, 763-766). The optical isomers of (+/-)-1 have been separated by chromatography on a chiral stationary phase. The absolute configuration at the C-1a and C-7a positions was determined using X-ray crystallography of an amide derivative with the methyl ester of L-phenylalanine (L-PheOMe) ((+)-6). In a phosphoinositol (PI) turnover assay at the cloned human mGlu1b receptor, (-)-1 and the new amide derivatives (-)-5 and (-)-6, all of which have (1aS,7aS)-stereochemistry on the chromane ring system, showed IC(50) values of 1.5, 0.43, and 0.93 microM, respectively. In contrast, (+)-1 and the new amide derivatives (+)-5 and (+)-6were found to be inactive up to a concentration of 30 microM indicating a selectivity for the (-)-enantiomers of at least 70-fold. In a previous study (Litschig et al. Mol. Pharmacol. 1999, 55, 453-461) we demonstrated using site-directed mutagenesis that the interaction site of (+/-)-1 is located in the transmembrane (TM) domain of hmGlu1b. To suggest a plausible binding mode of (-)-1, we have built a molecular mechanics model of the putative seven TM domain of hmGlu1 based on the alpha-carbon template of the TM helices of rhodopsin. A receptor docking hypothesis suggests that the OH of T815 (TMVII) comes in close contact with the oxime OH of (-)-1 and (-)-5, whereas no such close interactions could be demonstrated by docking of (+)-1.


Subject(s)
Chromones/chemical synthesis , Excitatory Amino Acid Antagonists/chemical synthesis , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Chromones/chemistry , Chromones/pharmacology , Cricetinae , Crystallography, X-Ray , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , Hydrolysis , Inositol Phosphates/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Metabotropic Glutamate/chemistry , Stereoisomerism , Structure-Activity Relationship
12.
Neuropharmacology ; 39(12): 2223-30, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10974306

ABSTRACT

We have used potent and selective non-competitive antagonists of metabotropic glutamate receptor subtype 5 (mGlu5) -- 2-methyl-6-phenylethynylpyridine (MPEP), [6-methyl-2-(phenylazo)-3-pyridinol] (SIB-1757) and [(E)-2-methyl-6-(2-phenylethenyl)pyridine] (SIB-1893) - to examine whether endogenous activation of this particular metabotropic glutamate receptor subtype contributes to neuronal degeneration. In cortical cultures challenged with N-methyl-D-aspartate (NMDA), all three mGlu5 receptor antagonists were neuroprotective. The effect of MPEP was highly specific because the close analogue, 3-methyl-6-phenylethynylpyridine (iso-MPEP), which did not antagonize heterologously expressed mGlu5 receptors, was devoid of activity on NMDA toxicity. Neuroprotection by mGlu5 receptor antagonists was also observed in cortical cultures challenged with a toxic concentration of beta-amyloid peptide. We have also examined the effect of mGlu5 receptor antagonists in in vivo models of excitotoxic degeneration. MPEP and SIB-1893 were neuroprotective against neuronal damage induced by intrastriatal injection of NMDA or quinolinic acid. These results indicate that mGlu5 receptors represent a suitable target for novel neuroprotective agents of potential application in neurodegenerative disorders.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Male , Mice , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/toxicity , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Neuroglia/drug effects , Neurons/pathology , Phenazopyridine/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5
13.
J Neurosci ; 20(17): 6413-20, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10964947

ABSTRACT

Activation of group III metabotropic glutamate receptors (mGluR4, mGluR6, mGluR7, and mGluR8) has been established to be neuroprotective in vitro and in vivo. To disclose the identity of the receptor subtype(s) that exert(s) the protective effect, we have used group III agonists in combination with mGluR4 subtype-deficient mice (-/-). In cortical cultures prepared from wild-type (+/+) mice and exposed to a toxic pulse of NMDA, the selective group III agonist (+)-4-phosphonophenylglycine [(+)-PPG] reversed excitotoxicity with an EC(50) value of 4.9 microm, whereas its enantiomer (-)-PPG was inactive. This correlated closely with the potency of (+)-PPG in activating recombinant mGluR4a. In cortical neurons from -/- mice, (+)-PPG showed no protection against the NMDA insult up to 300 microm, whereas group I/II mGluR ligands still retained their protective activity. Classical group III agonists (l-2-amino-4-phosphonobutyrate and l-serine-O-phosphate) were also substantially neuroprotective against NMDA toxicity in +/+ and heterozygous (+/-) cultures but were inactive in -/- cultures. Interestingly, -/- cultures were more vulnerable to low concentrations of NMDA and showed higher extracellular glutamate levels compared with +/+ cultures. We have also examined neurodegeneration induced by intrastriatal infusion of NMDA in wild-type or mGluR4-deficient mice. Low doses of (R,S)-PPG (10 nmol/0.5 microl) substantially reduced NMDA toxicity in +/+ mice but were ineffective in -/- mice. Higher doses of (R,S)-PPG were neuroprotective in both strains of animals. Finally, microdialysis studies showed that intrastriatal infusion of NMDA increased extracellular glutamate levels to a greater extent in -/- than in +/+ mice, supporting the hypothesis that the mGluR4 subtype is necessary for the maintenance of the homeostasis of extracellular glutamate levels.


Subject(s)
Aminobutyrates/pharmacology , Cerebral Cortex/cytology , Glycine/analogs & derivatives , N-Methylaspartate/toxicity , Neurons/physiology , Neurotoxins/pharmacology , Receptors, Metabotropic Glutamate/physiology , Animals , Cells, Cultured , Cerebral Cortex/physiology , Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/metabolism , Glycine/pharmacology , Heterozygote , Mice , Mice, Knockout , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Stereoisomerism
14.
J Biol Chem ; 275(43): 33750-8, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10934211

ABSTRACT

We have investigated the mechanism of inhibition and site of action of the novel human metabotropic glutamate receptor 5 (hmGluR5) antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), which is structurally unrelated to classical metabotropic glutamate receptor (mGluR) ligands. Schild analysis indicated that MPEP acts in a non-competitive manner. MPEP also inhibited to a large extent constitutive receptor activity in cells transiently overexpressing rat mGluR5, suggesting that MPEP acts as an inverse agonist. To investigate the molecular determinants that govern selective ligand binding, a mutagenesis study was performed using chimeras and single amino acid substitutions of hmGluR1 and hmGluR5. The mutants were tested for binding of the novel mGluR5 radioligand [(3)H]2-methyl-6-(3-methoxyphenyl)ethynyl pyridine (M-MPEP), a close analog of MPEP. Replacement of Ala-810 in transmembrane (TM) VII or Pro-655 and Ser-658 in TMIII with the homologous residues of hmGluR1 abolished radioligand binding. In contrast, the reciprocal hmGluR1 mutant bearing these three residues of hmGluR5 showed high affinity for [(3)H]M-MPEP. Radioligand binding to these mutants was also inhibited by 7-hydroxyiminocyclopropan[b]chromen-1a-carboxylic acid ethyl ester (CPCCOEt), a structurally unrelated non-competitive mGluR1 antagonist previously shown to interact with residues Thr-815 and Ala-818 in TMVII of hmGluR1. These results indicate that MPEP and CPCCOEt bind to overlapping binding pockets in the TM region of group I mGluRs but interact with different non-conserved residues.


Subject(s)
Chromones/metabolism , Excitatory Amino Acid Antagonists/metabolism , Pyridines/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , COS Cells , Cricetinae , Models, Molecular , Molecular Sequence Data , Rats , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 10(13): 1447-50, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10888329

ABSTRACT

On the basis of a pharmacophore definition of mGlu4 agonists, the two novel semi-rigid derivatives 12 and 13 were designed and synthesized. The preliminary biological evaluation demonstrated that both compounds interact with hmGlu4a, while ineffective at group II receptor subtypes. In particular, derivative 13 is a full hmGlu4a agonist with an EC50 = 17 microM.


Subject(s)
Cyclopentanes/chemical synthesis , Excitatory Amino Acid Agonists/chemical synthesis , Receptors, Metabotropic Glutamate/agonists , Animals , Biological Assay , CHO Cells , Colforsin/pharmacology , Cricetinae , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Humans , Molecular Structure , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
16.
Bioorg Med Chem Lett ; 10(11): 1241-4, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10866390

ABSTRACT

A new synthesis of (R,S)-PPG (4-phosphonophenylglycine) and the separation of the protected enantiomers leading after deprotection to (+)- and (-)-PPG are described. Pharmacological characterization at the group III metabotropic glutamate receptors hmGluR4a and hmGluR7b revealed (+)-PPG as the active enantiomer.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Glycine/analogs & derivatives , Receptors, Metabotropic Glutamate/agonists , Glycine/chemical synthesis , Glycine/chemistry , Glycine/pharmacology
17.
Neuropharmacology ; 39(5): 911-7, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10699457

ABSTRACT

With this study we evaluated the influence of (R, S)-4-phosphonophenylglycine [(R,S)-PPG], a selective group III metabotropic glutamate receptor agonist, on excitotoxic, hypoxic/hypoglycaemic and ischaemic cerebral damage in rodents. Consistent with previous data showing neuroprotective and anticonvulsive effects (Gasparini, F., Bruno, V., Battaglia, G., Lukic, S., Leonhardt, T., Inderbitzin, W., et al., 1999. (R, S)-4-Phosphonophenylglycine, a potent and selective group III metabotropic glutamate receptor agonist, is anticonvulsive and neuroprotective in vivo. Journal of Pharmacology and Experimental Therapeutics 290, 1678-1687), we found pronounced neuroprotective effects with (R,S)-PPG (300 nmol) in a model of excitotoxicity, i.e. quinolinic acid-induced striatal lesions in rats. However, neither in focal cerebral ischaemia in mice nor in global cerebral ischaemia in gerbils or rats did (R,S)-PPG have any significant influence on the extent of neuronal damage. In a model of hypoxia/hypoglycaemia in acutely isolated hippocampal slices, however, (R,S)-PPG led to an improved recovery of population spike amplitude. As acutely isolated hippocampal slices are only viable for a few hours, these electrophysiological recordings can only be performed in a limited time window after the challenge-when most probably excitotoxicity is still the predominant influence in hypoxic pathophysiology. From this we conclude that group III mGluR agonists might be promising drugs against damage mediated mainly by excitotoxicity, but less likely against development of neuronal death due to ischaemia.


Subject(s)
Brain Diseases/drug therapy , Glycine/analogs & derivatives , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Action Potentials/drug effects , Action Potentials/physiology , Animals , Brain Diseases/pathology , Brain Diseases/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell Hypoxia/drug effects , Corpus Striatum/drug effects , Corpus Striatum/pathology , Cytoprotection/drug effects , Disease Models, Animal , Gerbillinae , Glycine/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiopathology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Quinolinic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors
18.
Neuropharmacology ; 38(10): 1493-503, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10530811

ABSTRACT

In the present paper we describe 2-methyl-6-(phenylethynyl)-pyridine (MPEP) as a potent, selective and systemically active antagonist for the metabotropic glutamate receptor subtype 5 (mGlu5). At the human mGlu5a receptor expressed in recombinant cells, MPEP completely inhibited quisqualate-stimulated phosphoinositide (PI) hydrolysis with an IC50 value of 36 nM while having no agonist or antagonist activities at cells expressing the human mGlu1b receptor at concentrations up to 30 microM. When tested at group II and III receptors, MPEP did not show agonist or antagonist activity at 100 microM on human mGlu2, -3, -4a, -7b, and -8a receptors nor at 10 microM on the human mGlu6 receptor. Electrophysiological recordings in Xenopus laevis oocytes demonstrated no significant effect at 100 microM on human NMDA (NMDA1A/2A), rat AMPA (Glu3-(flop)) and human kainate (Glu6-(IYQ)) receptor subtypes nor at 10 microM on the human NMDA1A/2B receptor. In rat neonatal brain slices, MPEP inhibited DHPG-stimulated PI hydrolysis with a potency and selectivity similar to that observed on human mGlu receptors. Furthermore, in extracellular recordings in the CA1 area of the hippocampus in anesthetized rats, the microiontophoretic application of DHPG induced neuronal firing that was blocked when MPEP was administered by iontophoretic or intravenous routes. Excitations induced by microiontophoretic application of AMPA were not affected.


Subject(s)
Brain/physiology , Excitatory Amino Acid Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/physiology , Animals , Animals, Newborn , Brain/drug effects , Cell Line , Cyclic AMP/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Lithium Chloride/pharmacology , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Oocytes/physiology , Phosphatidylinositols/metabolism , Quisqualic Acid/pharmacology , Radioligand Assay , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/antagonists & inhibitors , Sulfur Radioisotopes , Transfection , Xenopus laevis
19.
J Pharmacol Exp Ther ; 290(1): 170-81, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381773

ABSTRACT

Cell lines expressing the human metabotropic glutamate receptor subtype 5a (hmGluR5a) and hmGluR1b were used as targets in an automated high-throughput screening (HTS) system that measures changes in intracellular Ca2+ ([Ca2+]i) using fluorescence detection. This functional screen was used to identify the mGluR5-selective antagonist, SIB-1757 [6-methyl-2-(phenylazo)-3-pyridinol], which inhibited the glutamate-induced [Ca2+]i responses at hmGluR5 with an IC50 of 0.37 microM compared with an IC50 of >100 microM at hmGluR1. Schild analysis demonstrated a noncompetitive mechanism of inhibition. Pharmacophore mapping was used to identify an additional compound, SIB-1893 [(E)-2-methyl-6-(2-phenylethenyl)pyridine], which was also shown to block glutamate-induced increases in [Ca2+]i at hmGluR5 with an IC50 of 0.29 microM compared with an IC50 of >100 microM at hmGluR1. SIB-1757 and SIB-1893 showed little or no activity when tested for agonist and antagonist activity at the other recombinant human mGluR subtypes, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, kainate, and N-methyl-D-aspartate receptors. In rat neonatal brain slices, SIB-1757 and SIB-1893 inhibited (S)-3,5-dihydroxyphenylglycine (DHPG)-evoked inositol phosphate accumulation in hippocampus and striatum by 60% to 80%, with a potency similar to that observed on recombinant mGluR5. However, in the cerebellum, a brain region with low mGluR5 expression, SIB-1757 failed to inhibit DHPG-evoked inositol phosphate accumulation. In cultured rat cortical neurons, SIB-1757 and SIB-1893 largely inhibited DHPG-evoked [Ca2+]i signals, revealing a population of neurons that were less sensitive to SIB-1757 and SIB-1893. This is the first description of highly selective, noncompetitive mGluR5 antagonists. These compounds will be useful tools in evaluating the role of mGluR5 in normal physiology and in animal models of disease.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Phenazopyridine/analogs & derivatives , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Animals, Newborn , Binding, Competitive , Brain/cytology , Brain/drug effects , Brain/metabolism , CHO Cells , Calcium/metabolism , Cells, Cultured , Cricetinae , Excitatory Amino Acid Antagonists/chemistry , Humans , In Vitro Techniques , Inositol Phosphates/metabolism , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/antagonists & inhibitors , Neurons/drug effects , Phenazopyridine/chemistry , Phenazopyridine/pharmacology , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Recombinant Proteins/metabolism , Structure-Activity Relationship
20.
J Pharmacol Exp Ther ; 289(3): 1678-87, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10336568

ABSTRACT

Group III metabotropic glutamate receptors (mGluRs) are thought to modulate neurotoxicity of excitatory amino acids, via mechanisms of presynaptic inhibition, such as regulation of neurotransmitter release. Here, we describe (R,S)-4-phosphonophenylglycine (PPG) as a novel, potent, and selective agonist for group III mGluRs. In recombinant cell lines expressing the human receptors hmGluR4a, hmGluR6, hmGluR7b, or hmGluR8a, EC50 values for (R,S)-PPG of 5.2 +/- 0.7 microM, 4.7 +/- 0.9 microM, 185 +/- 42 microM, and 0.2 +/- 0.1 microM, respectively, were measured. The compound showed EC50 and IC50 values of >/=200 microM at group I and II hmGluRs and was inactive at cloned human N-methyl-D-aspartate, alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionate, and kainate receptors (>300 microM). On the other hand, it showed micromolar affinity for a Ca2+/Cl--dependent L-glutamate binding site in rat brain, similar to other phosphono-substituted amino acids like L-2-amino-4-phosphonobutyrate. In cultured cortical neurons, (R, S)-PPG provided protection against a toxic pulse of N-methyl-D-aspartate (EC50 = 12 microM), which was reversed by the group III mGluR antagonist (R,S)-alpha-methylserine-O-phosphate but not by the group II antagonist (2S)-alpha-ethylglutamate. Moreover, (R,S)-PPG protected against N-methyl-D-aspartate- and quinolinic acid-induced striatal lesions in rats and was anticonvulsive in the maximal electroshock model in mice. In contrast to the group III mGluR agonists L-2-amino-4-phosphonobutyrate and L-serine-O-phosphate, (R,S)-PPG showed no proconvulsive effects (2200 nmol i.c.v.). These data provide novel in vivo evidence for group III mGluRs as attractive targets for neuroprotective and anticonvulsive therapy. Also, (R,S)-PPG represents an attractive tool to analyze the roles of group III mGluRs in nervous system physiology and pathology.


Subject(s)
Anticonvulsants/pharmacology , Brain/metabolism , Glycine/analogs & derivatives , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , Cell Line , Cell Membrane/metabolism , Colforsin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiology , Cyclic AMP/metabolism , Electroshock , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/pharmacology , Humans , Kinetics , Male , Mice , Mice, Inbred Strains , N-Methylaspartate/toxicity , Phosphatidylinositols/metabolism , Protein Isoforms/agonists , Protein Isoforms/genetics , Quinolinic Acid/toxicity , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/agonists , Second Messenger Systems/drug effects , Seizures/physiopathology , Seizures/prevention & control , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...