Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Synapse ; 66(12): 1002-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22927303

ABSTRACT

The metabotropic glutamate 1 receptor (mGlu1) is an important protein in the regulation of glutamate transmission in the brain, and believed to be involved in disorders such as ischemia, epilepsy, neuropathic pain, anxiety, and schizophrenia. The goal of this study was to evaluate two selective mGlu1 antagonists [(11) C]3 and [(18) F]4 as potential PET radioligands for the in vivo imaging of the mGlu1 receptor. Biodistribution studies in rats indicated high uptake of [(11) C]3 and [(18) F]4 in the brain. The highest activity level was found in the cerebellum, followed by striatum, hippocampus, frontal cortex, and medulla, in a pattern consistent with the distribution of mGlu1 receptor in rat. At 30 min postinjection, the activity ratio of cerebellum to medulla was 4.5 for [(11) C]3, indicating a high degree of specific binding, while specific binding was lower for [(18) F]4 (cerebellum to medulla activity ratio of 2.0). Moreover, binding of the radioligands [(11) C]3 and [(18) F]4 in mGlu1 receptor-rich region such as cerebellum was blocked by pretreatment of the rats with their respective unlabeled compound or the selective mGlu1 antagonist (compound 5, 2 mg/kg each), but not by the selective mGlu2 antagonist LY341495, or the selective mGlu5 antagonist MPEP (2 mg/kg), thus indicating the binding specificity and selectivity of [(11) C]3 and [(18) F]4 to the mGlu1 receptor. However, in imaging experiments in baboons [(11) C]3 displayed a small specific binding signal only in the cerebellum, while the specific binding of [(18) F]4 was difficult to detect. Species differences in receptor density and affinity of the radioligands in large part account for the differences in the behavior of [(11) C]3 and [(18) F]4 in rats and baboons. Radioligands with higher affinity and/or lower lipophilicity are needed to successfully image the mGlu1 receptor in humans.


Subject(s)
Positron-Emission Tomography , Quinolines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, Metabotropic Glutamate/metabolism , Amino Acids/pharmacology , Animals , Brain/diagnostic imaging , CHO Cells , Carbon Radioisotopes/pharmacokinetics , Cricetinae , Cricetulus , Excitatory Amino Acid Antagonists/pharmacology , Fluorine Radioisotopes/pharmacokinetics , Ligands , Male , Papio , Pyridines/pharmacology , Quinolines/chemical synthesis , Quinolines/chemistry , Radiopharmaceuticals/chemical synthesis , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Tissue Distribution , Xanthenes/pharmacology
2.
J Psychopharmacol ; 26(9): 1265-70, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22182741

ABSTRACT

The α7 nicotinic acetylcholine receptors (nAChRs) have been highlighted as a target for cognitive enhancement in schizophrenia. Adult female hooded Lister rats received sub-chronic phencyclidine (PCP) (2 mg/kg) or vehicle i.p. twice daily for 7 days, followed by 7 days' washout. PCP-treated rats then received PNU-120596 (10 mg/kg; s.c.) or saline and were tested in the attentional set-shifting task. Sub-chronic PCP produced a significant cognitive deficit in the extra-dimensional shift (EDS) phase of the task (p < 0.001, compared with vehicle). PNU-120596 significantly improved performance of PCP-treated rats in the EDS phase of the attentional set-shifting task (p < 0.001). In conclusion, these data demonstrate that PNU-120596 improves cognitive dysfunction in our animal model of cognitive dysfunction in schizophrenia, most likely via modulation of α7 nACh receptors.


Subject(s)
Cognition Disorders/drug therapy , Isoxazoles/therapeutic use , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Nicotinic Agonists/therapeutic use , Phencyclidine/toxicity , Phenylurea Compounds/therapeutic use , Receptors, Nicotinic/chemistry , Allosteric Regulation , Animals , Attention/drug effects , Behavior, Animal/drug effects , Brain/metabolism , Cognition Disorders/chemically induced , Cognition Disorders/physiopathology , Discrimination Learning/drug effects , Disease Models, Animal , Drug Interactions , Female , Half-Life , Hallucinogens/administration & dosage , Hallucinogens/toxicity , Isoxazoles/metabolism , Isoxazoles/pharmacokinetics , Neurons/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neurotoxicity Syndromes/physiopathology , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacokinetics , Phencyclidine/administration & dosage , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacokinetics , Rats , Rats, Inbred Strains , Tissue Distribution , alpha7 Nicotinic Acetylcholine Receptor
3.
J Pharmacol Exp Ther ; 336(2): 560-74, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21084390

ABSTRACT

The α(7) nicotinic acetylcholine receptor (nAChR) is a potential therapeutic target for the treatment of cognitive deficits associated with schizophrenia, Alzheimer's disease, Parkinson's disease, and attention-deficit/hyperactivity disorder. Activation of α(7) nAChRs improved sensory gating and cognitive function in animal models and in early clinical trials. Here we describe the novel highly selective α(7) nAChR positive allosteric modulator, 2-[[4-fluoro-3-(trifluoromethyl)phenyl]amino]-4-(4-pyridinyl)-5-thiazolemethanol (JNJ-1930942). This compound enhances the choline-evoked rise in intracellular Ca(2+) levels in the GH4C1 cell line expressing the cloned human α(7) nAChR. JNJ-1930942 does not act on α4ß2, α3ß4 nAChRs or on the related 5-HT3A channel. Electrophysiological assessment in the GH4C1 cell line shows that JNJ-1930942 increases the peak and net charge response to choline, acetylcholine, and N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide (PNU-282987). The potentiation is obtained mainly by affecting the receptor desensitization characteristics, leaving activation and deactivation kinetics as well as recovery from desensitization relatively unchanged. Choline efficacy is increased over its full concentration response range, and choline potency is increased more than 10-fold. The potentiating effect is α(7) channel-dependent, because it is blocked by the α(7) antagonist methyllycaconitine. Moreover, in hippocampal slices, JNJ-1930942 enhances neurotransmission at hippocampal dentate gyrus synapses and facilitates the induction of long-term potentiation of electrically evoked synaptic responses in the dentate gyrus. In vivo, JNJ-1930942 reverses a genetically based auditory gating deficit in DBA/2 mice. JNJ-1930942 will be a useful tool to study the therapeutic potential of α(7) nAChR potentiation in central nervous system disorders in which a deficit in α(7) nAChR neurotransmission is hypothesized to be involved.


Subject(s)
Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Thiazoles/pharmacology , Allosteric Regulation , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Evoked Potentials, Auditory/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Humans , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred DBA , Nicotinic Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/physiology , Synaptic Transmission/drug effects , alpha7 Nicotinic Acetylcholine Receptor
4.
Eur Neuropsychopharmacol ; 21(4): 333-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20630711

ABSTRACT

RATIONALE: Nicotinic α7 acetylcholine receptors (nAChRs) have been highlighted as a target for cognitive enhancement in schizophrenia. AIM: To investigate whether the deficits induced by sub-chronic phencyclidine (PCP) in reversal learning and novel object recognition could be attenuated by the selective α7 nAChR full agonist, PNU-282987. METHODS: Adult female hooded-Lister rats received sub-chronic PCP (2mg/kg) or vehicle i.p. twice daily for 7days, followed by 7 days washout. In cohort 1, PCP-treated rats then received PNU-282987 (5, 10, 20mg/kg; s.c.) or vehicle and were tested in the reversal-learning task. In cohort 2, PCP-treated rats received PNU-282987 (10mg/kg; s.c.) or saline for 15days and were tested in the novel object recognition test on day 1 and on day 15, to test for tolerance. RESULTS: Sub-chronic PCP produced significant deficits in both cognitive tasks (P<0.01-0.001). PNU-282987 attenuated the PCP-induced deficits in reversal learning at 10mg/kg (P<0.01) and 20mg/kg (P<0.001), and in novel object recognition at 10mg/kg on day 1 (P<0.01) and on day 15 (P<0.001). CONCLUSIONS: These data show that PNU-282987 has efficacy to reverse PCP-induced deficits in two paradigms of relevance to schizophrenia. Results further suggest that 15-day once daily dosing of PNU-282987 (10mg/kg s.c.) does not cause tolerance in the rat. This study suggests that activation of α7 nAChRs, may represent a suitable strategy for improving cognitive deficits of relevance to schizophrenia.


Subject(s)
Benzamides/therapeutic use , Bridged Bicyclo Compounds/therapeutic use , Cognition Disorders/drug therapy , Excitatory Amino Acid Antagonists/toxicity , Nicotinic Agonists/therapeutic use , Phencyclidine/toxicity , Receptors, Nicotinic/metabolism , Schizophrenia/physiopathology , Animals , Behavior, Animal/drug effects , Benzamides/administration & dosage , Benzamides/blood , Benzamides/pharmacokinetics , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/blood , Bridged Bicyclo Compounds/pharmacokinetics , Cognition/drug effects , Cognition Disorders/chemically induced , Cognition Disorders/etiology , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Female , Half-Life , Learning/drug effects , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/blood , Nicotinic Agonists/pharmacokinetics , Random Allocation , Rats , Reversal Learning/drug effects , Tachyphylaxis , alpha7 Nicotinic Acetylcholine Receptor
5.
J Mol Neurosci ; 30(1-2): 109-10, 2006.
Article in English | MEDLINE | ID: mdl-17192651

ABSTRACT

The alpha7-nicotinic acetylcholine receptor (alpha7) is an important ionotropic receptor in the central nervous system, which becomes permeable to cations upon binding of its natural agonist acetylcholine (ACh). alpha7 kinetics are characterized by rapid activation, followed by fast desensitization of the current. As the wild-type (WT) alpha7 is difficult to express heterologously in mammalian cellular systems, frequently a more easily expressible chimera consisting of the extracellular domain of the alpha7 and the transmembrane domain of the 5HT3A receptor is used to study alpha7 pharmacology (chick alpha7/mouse 5HT3A [Eiselé et al., 1993]; human alpha7/mouse 5HT3A [Graig et al., 2004]). Desensitization characteristics of these chimera receptors have been described as intermediate compared with the fast desensitizing alpha7 and the more slowly desensitizing 5HT3A receptors. Here, we describe a fully human chimera receptor (h-alpha7/5HT3A), which is characterized by desensitization, and recovery kinetics that deviate from the human WT alpha7.


Subject(s)
Receptors, Nicotinic/physiology , Receptors, Serotonin, 5-HT3/physiology , Acetylcholine/physiology , Animals , Female , Mutant Chimeric Proteins/metabolism , Oocytes , Patch-Clamp Techniques , Receptors, Nicotinic/genetics , Receptors, Serotonin, 5-HT3/genetics , Transfection , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
6.
J Med Chem ; 48(16): 5096-9, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-16078827

ABSTRACT

A selective metabotropic glutamate 1 receptor (mGlu1) antagonist was labeled with the positron-emitting radioisotope carbon-11 and evaluated in ex vivo biodistribution studies and micro-positron emission tomography (micro-PET) imaging experiments in rats. Results from animal experiments demonstrate that the radioligand [11C]2 is the first PET tracer capable of labeling the rat mGlu1 receptor in vivo.


Subject(s)
Quinolines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Receptors, Metabotropic Glutamate/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Ligands , Positron-Emission Tomography , Quinolines/chemistry , Quinolines/pharmacology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Rats , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Structure-Activity Relationship , Tissue Distribution
7.
Behav Brain Res ; 164(1): 52-60, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16043241

ABSTRACT

Metabotropic glutamate receptors, including the mGlu1 receptor, have received considerable attention as potential targets for anxiolytic, antidepressant, antipsychotic and antinociceptive drugs. mGlu1 receptors have also been suggested to play a role in the modulation of cognitive processes, but knowledge is still very limited. In the present study the effects of the selective mGlu1 receptor antagonist 3,4-dihydro-2H-pyrano[2,3]beta-quinolin-7-yl)(cis-4-methoxycyclohexyl)methanone (JNJ16259685, 0.63-10 mg/kg s.c.) on more or less spatially demanding learning and spatial memory (retention and re-acquisition) were investigated in mice performing in a water maze. Selective mGlu1 receptor blockade with JNJ16259685 impaired spatial acquisition processes, irrespective of spatial load, as well as spatial re-acquisition, already at the lowest dose tested (0.63 mg/kg). In contrast, effects on spatial retention performance were relatively mild in mice that had learned to locate the position of the escape platform prior to treatment. Thigmotaxic behaviour and locomotor activity appeared to be unaffected by JNJ16259685. These data suggest that blockade of the mGlu1 receptor primarily affects learning of new information, but leaves retention of spatial information relatively unaffected. Blockade of the mGlu5 receptor with MPEP also impaired spatial learning, although only at the highest dose tested (10 mg/kg). An ex vivo receptor occupancy study in rats revealed that MPEP occupied central mGlu5 receptors with an ED(50) of 2.0 mg/kg one hour after subcutaneous administration. This is 50-150 times higher than the ED(50) reported for JNJ16259685 at central mGlu1 receptors and suggests that one reason why the two compounds cause cognitive effects at different doses might be due to differences in central mGlu receptor occupancy, rather than fundamentally different roles of mGlu1 and mGlu5 receptors in the modulation of cognitive function.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Maze Learning/drug effects , Quinolines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Space Perception/drug effects , Spatial Behavior/drug effects , Animals , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Receptors, Metabotropic Glutamate/physiology , Retention, Psychology/drug effects , Retention, Psychology/physiology , Space Perception/physiology , Spatial Behavior/physiology
8.
Eur J Neurosci ; 21(6): 1610-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15845088

ABSTRACT

We investigated the effect of 24 h sustained treatment with the mGlu1 receptor antagonist CPCCOEt on mGlu1 receptor signalling in primary cultures of rat cerebellar granule cells. In the absence of ionotropic glutamate (iGlu) blockers, the maximal inositol phosphate (IP) response (E(max)) but not the potency of glutamate was significantly increased when cells were pre-exposed for 24 h with CPCCOEt. When the contribution of iGlu receptors to the glutamate-induced IP response was eliminated with the use of DNQX, the E(max) was again increased but also the concentration eliciting 50% of the maximal glutamate stimulus was significantly decreased. In the absence of iGlu receptor inhibitors, the E(max) of quisqualate, which likely mediates IP accumulation only via the mGlu1 receptor, was significantly increased in CPCCOEt-pretreated cells. Also, less quisqualate was needed to reach the same IP effect. The potency of R193845, a selective mGlu1 receptor antagonist, was significantly decreased in antagonist-pretreated cells. These findings demonstrate that 24 h sustained antagonist treatment can render mGlu1 receptors in neurons supersensitive to agonists, with a concomitant decrease in the effectiveness of antagonists.


Subject(s)
Cerebellum/metabolism , Chromones/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Cells, Cultured , Cerebellum/drug effects , Dose-Response Relationship, Drug , Glutamic Acid/pharmacology , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Psychopharmacology (Berl) ; 179(1): 198-206, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15821950

ABSTRACT

RATIONALE: Group I metabotropic glutamate receptor antagonists, which block both the mGlu1 and mGlu5 receptors, have been shown to have anxiolytic effects in the lick suppression test in rats. OBJECTIVE: The anxiolytic potential of the selective mGlu1 antagonist 3,4-dihydro-2H-pyrano[2,3]beta-quinolin-7-yl)(cis-4-methoxycyclohexyl)methanone (JNJ16259685) was investigated and compared with the mGlu5 antagonist MPEP. METHODS: Anxiety-related behaviour was assessed in lick suppression and in the elevated zero maze in rats. Non-specific effects on pain threshold, water intake and locomotor activity were also measured. RESULTS: Acute administration of JNJ16259685 or MPEP increased the number of licks (lowest active dose 2.5 mg/kg IP for each compound). JNJ16259685 did not increase water intake or reduce acute pain threshold, suggesting that the anxiolytic-like properties are specific. However, acute administration decreased locomotor activity. The effects of chronic administration of JNJ16259685 over 14 days (5 mg/kg bid) on lick suppression were comparable to those seen after acute administration, arguing against development of behavioural tolerance or sensitisation. Yet, there was a tendency for an increase in locomotor activity after cessation of chronic treatment. Acute co-administration of both JNJ16259685 and MPEP had additive effects on the number of licks. No anxiolytic-like properties of JNJ16259685 were observed in the elevated zero maze. CONCLUSION: Our data suggest that the anxiolytic-like effects induced by group I metabotropic glutamate receptor antagonists are mediated through both mGlu1 and mGlu5 receptors. Rather than producing a general anxiolytic-like effect, the effects seen following mGlu1 antagonism seem task-dependent, as prominent effects were seen in a conflict procedure, but not in a task based on spontaneous exploration.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Anxiety/psychology , Dose-Response Relationship, Drug , Drinking/drug effects , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Wistar
10.
J Med Chem ; 48(6): 2134-53, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771457

ABSTRACT

We describe the discovery and the structure-activity relationship of a new series of quinoline derivatives acting as selective and highly potent noncompetitive mGlu1 antagonists. We first identified cis-10 as a fairly potent mGlu1 antagonist (IC(50) = 20 nM) in a cell-based signal transduction assay on the rat mGlu1 receptor expressed in CHO-K1 cells, and then we were able to design and synthesize highly potent compounds on both rat and human mGlu1 receptors as exemplified by compound cis-64a, which has an antagonist potency of 0.5 nM for the human mGlu1 receptor. We briefly present and discuss the in vitro metabolic stability of the compounds in human liver microsomes. We finally report the pharmacokinetic properties of our lead compound cis-64a.


Subject(s)
Quinolines/chemical synthesis , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Biological Availability , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Half-Life , Humans , In Vitro Techniques , Intracellular Fluid/metabolism , Microsomes, Liver/metabolism , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Stereoisomerism , Structure-Activity Relationship
11.
Neuropharmacology ; 47(7): 961-72, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15555631

ABSTRACT

We examined the pharmacological profile of (3,4-dihydro-2H-pyrano[2,3]b quinolin-7-yl) (cis-4-methoxycyclohexyl) methanone (JNJ16259685). At recombinant rat and human metabotropic glutamate (mGlu) 1a receptors, JNJ16259685 non-competitively inhibited glutamate-induced Ca2+ mobilization with IC50 values of 3.24+/-1.00 and 1.21+/-0.53 nM, respectively, while showing a much lower potency at the rat and human mGlu5a receptor. JNJ16259685 inhibited [3H]1-(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-2-phenyl-1-ethanone ([3H]R214127) binding to membranes prepared from cells expressing rat mGlu1a receptors with a Ki of 0.34+/-0.20 nM. JNJ16259685 showed no agonist, antagonist or positive allosteric activity toward rat mGlu2, -3, -4 or -6 receptors at concentrations up to 10 microM and did not bind to AMPA or NMDA receptors, or to a battery of other neurotransmitter receptors, ion channels and transporters. In primary cerebellar cultures, JNJ16259685 inhibited glutamate-mediated inositol phosphate production with an IC50 of 1.73+/-0.40 nM. Subcutaneously administered JNJ16259685 exhibited high potencies in occupying central mGlu1 receptors in the rat cerebellum and thalamus ( ED50=0.040 and 0.014 mg/kg, respectively). These data show that JNJ16259685 is a selective mGlu1 receptor antagonist with excellent potencies in inhibiting mGlu1 receptor function and binding and in occupying the mGlu1 receptor after systemic administration.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Quinolines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Animals , Autoradiography , CHO Cells , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cricetinae , Dose-Response Relationship, Drug , Female , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Inositol Phosphates/metabolism , Male , Pyrans/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/genetics , Recombinant Proteins/drug effects , Tetrahydrofolate Dehydrogenase/deficiency , Transfection
12.
Neuropharmacology ; 46(5): 609-19, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14996538

ABSTRACT

We used the selective metabotropic glutamate (mGlu) 1 receptor antagonist [3H]1-(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-2-phenyl-1-ethanone ([3H]R214127) to investigate the distribution of mGlu1 receptor binding sites in rat brain. We found high mGlu1 receptor binding in the cerebellum, thalamus, dentate gyrus and medial central gray, moderate binding within the CA3 of the hippocampus and hypothalamus, and low mGlu1 receptor binding in the basal ganglia and cortex. The mGlu1 receptor is also present in variable degree in the dorsal lateral septal nucleus, amygdala, interpeduncular nucleus and median raphe nucleus. Additionally, we employed [3H]R214127 autoradiography as a means of investigating the occupancy of central mGlu1 receptors following in vivo administration of mGlu1 receptor antagonists that prevent binding of this radioligand. We found that the mGlu1 receptor antagonist (3aS,6aS)-6a-naphtalan-2-ylmethyl-5-methyliden-hexahydro-cyclopenta[c]furan-1-on (BAY 36-7620), administered subcutaneously (s.c.) at 10 mg/kg, only occupied about 30% of cerebellar and thalamic mGlu1 receptors. The mGlu1/5 receptor antagonist 2-quinoxaline-carboxamide-N-adamantan-1-yl (NPS 2390) exhibited a relatively high potency in occupying mGlu1 receptors in rat cerebellum (ED50 = 0.75 mg/kg, s.c.) and thalamus (ED50 = 0.63 mg/kg, s.c). In the future, this method can be employed to gain more insight into the in vivo profile and central activity of potential therapeutic agents that act upon the mGlu1 receptor.


Subject(s)
Brain/metabolism , Pyrans/metabolism , Quinolines/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Autoradiography , Brain/drug effects , Dose-Response Relationship, Drug , Male , Naphthalenes/pharmacology , Protein Binding/physiology , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Tritium/metabolism
13.
Mol Pharmacol ; 63(5): 1082-93, 2003 May.
Article in English | MEDLINE | ID: mdl-12695537

ABSTRACT

R214127 was shown to be a potent and noncompetitive metabotropic glutamate 1 (mGlu1) receptor-selective antagonist. The kinetics and pharmacology of [(3)H]1-(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-2-phenyl-1-ethanone (R214127) binding to rat mGlu1a receptor Chinese hamster ovary (CHO)-dhfr(-) membranes was investigated, as well as the distribution of [(3)H]R214127 binding in rat brain tissue and sections. Specific binding to rat mGlu1a receptor CHO-dhfr(-) membranes was approximately 92% of total and was optimal at 4 degrees C. Full association was reached within 5 min, and [(3)H]R214127 bound to a single binding site with an apparent K(D) of 0.90 +/- 0.14 nM and a B(max) of 6512 +/- 1501 fmol/mg of protein. Inhibition experiments showed that [(3)H]R214127 binding was completely blocked by 2-quinoxaline-carboxamide-N-adamantan-1-yl (NPS 2390), (3aS,6aS)-6a-naphtalan-2-ylmethyl-5-methyliden-hexahydro-cyclopenta[c]furan-1-on (BAY 36-7620), and 7-(hydroxyimino)cyclo-propa[b]chromen-1a-carboxylate ethyl ester (CPCCOEt), but was not displaced by competitive mGlu1 receptor ligands such as glutamate and quisqualate, suggesting that R214127, NPS 2390, BAY 36-7620, and CPCCOEt bind to the same site or mutually exclusive sites. Experiments using rat cortex, striatum, hippocampus and cerebellum revealed that [(3)H]R214127 labeled a single high-affinity binding site (K(D) approximately 1 nM). B(max) values were highest in the cerebellum (4302 +/- 2042 fmol/mg of protein) and were 741 +/- 48, 688 +/- 125, and 471 +/- 68 fmol/mg of protein in the striatum, hippocampus, and cortex, respectively. The distribution of [(3)H]R214127 binding in rat brain was investigated in more detail by radioligand autoradiography. A high density of binding sites was detected in the molecular layer of the cerebellum. Moderate labeling was seen in the CA3 and dentate gyrus of the hippocampus, thalamus, olfactory tubercle, amygdala, and substantia nigra reticulata. The cerebral cortex, caudate putamen, ventral pallidum, and nucleus accumbens showed lower labeling. The high affinity and selectivity of [(3)H]R214127 for mGlu1 receptors renders this compound the ligand of choice to study the native mGlu1 receptor in brain.


Subject(s)
Adamantane/analogs & derivatives , Pyrans/pharmacology , Quinolines/pharmacology , Quinoxalines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Adamantane/pharmacology , Allosteric Regulation , Animals , Binding Sites , Binding, Competitive , Brain/drug effects , Brain/metabolism , CHO Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chromones/pharmacology , Cricetinae , Humans , Naphthalenes/pharmacology , Quisqualic Acid/pharmacology , Radioligand Assay , Rats , Transfection , Tritium
14.
Mol Pharmacol ; 61(5): 1244-54, 2002 May.
Article in English | MEDLINE | ID: mdl-11961143

ABSTRACT

The effect of antagonist pretreatment on the signaling properties of the human metabotropic glutamate 1a (hmGlu1a) receptor was examined in stably transfected L929sA cells. Pre-exposure of hmGlu1a receptor-expressing cells to the mGlu1 receptor antagonists (S)-4-carboxy-3-hydroxyphenylglycine and 7-(hydroxyimino)cyclo-propa[b]chromen-1a-carboxylate ethyl ester dramatically enhanced subsequent glutamate-induced phosphoinositide hydrolysis and intracellular [Ca(2+)] rise. We found clear indications that the antagonist-mediated enhancement of glutamate-evoked mGlu1a receptor signaling is caused by the development of mGlu1a receptor supersensitivity: the potency of glutamate was increased by 3-fold after 24 h antagonist pretreatment and the potency of the antagonists was significantly decreased in antagonist-pretreated cells. The kinetic profile of the antagonist-mediated enhancement showed that the maximal increase in intracellular [Ca(2+)] was already reached after 30-min pretreatment, suggesting that de novo receptor synthesis is not involved in the process of mGlu1a receptor supersensitization. Glutamate-mediated phosphoinositide hydrolysis increased up to 24 h after antagonist treatment. Although it seemed likely that the hmGlu1a receptor could desensitize after activation by endogenously present glutamate, removal of glutamate from the extracellular medium with GPT resulted in a much smaller enhancement of glutamate responsiveness. Moreover, the magnitude of antagonist-mediated receptor supersensitivity was much larger than the magnitude of agonist-induced receptor desensitization. These results suggest that antagonist-evoked mGlu1 receptor supersensitivity is not merely the result of a blockade of agonist-induced desensitization. Finally, we found that antagonist pretreatment doubled the amount of receptors at the cell surface. Our findings are the first lines of evidence that prolonged antagonist treatment can supersensitize the hmGlu1a receptor. In view of the potential therapeutic application of mGlu1 receptor antagonists, it will be important to know whether these phenomena occur in vivo.


Subject(s)
Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Humans , Interferon-beta/pharmacology , Kinetics , Rats , Rats, Wistar , Receptors, Cell Surface/biosynthesis , Receptors, Dopamine/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Signal Transduction/drug effects , Transaminases/pharmacology , Transfection
15.
J Org Chem ; 67(1): 227-33, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11777464

ABSTRACT

An efficient method for preparing conformationally restricted cyclopentenyl-glutamate analogues in a regioselective and diastereoselective manner has been developed using a formal [3 + 2] cycloaddition reaction of dehydroamino acids. Methods for preparing optically active versions of these compounds have also been devised. Of these compounds, (S)-2 is an agonist at the mGlu5 (EC(50) 18 microM) and mGlu2 (EC(50) 45 microM) receptors.


Subject(s)
Glutamates/chemical synthesis , Glutamates/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , CHO Cells , Calcium Signaling/drug effects , Cricetinae , Cyclopentanes/chemical synthesis , Cyclopentanes/metabolism , Cyclopentanes/pharmacology , Glutamates/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Molecular Conformation , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Transfection
16.
Mol Pharmacol ; 61(1): 85-96, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752209

ABSTRACT

This study documents differences in ligand binding and signal transduction properties between the human (h) 5-hydroxytryptamine (5-HT)4a and h5-HT4b receptor splice variants stably expressed in human embryonic kidney 293 cells. The fraction of the [3H]5-HT high-affinity site relative to the whole receptor population measured with [3H]GR113808 was higher for the h5-HT4a isoform (around 0.4) than for the 5-HT4b isoform (around 0.2) and was independent of the level of expression. The potency and efficacy of reference compounds tested for the cAMP response differed slightly but significantly between both variants. Most remarkably, 5-methoxytryptamine and prucalopride were found more potent on the 5-HT4b variant, whereas SDZ-HTF 919 and SB204070 were more potent on the 5-HT(4a) variant. Guanosine-5'-O-(3-[35S]thio)triphosphate binding on membranes and cAMP assays in whole cells revealed that only the h5-HT4b isoform coupled to Galphai/o-proteins in addition to its well-documented Galphas coupling. In contrast, the h5-HT4a receptor coupled only to Galphas-proteins, however, was able to trigger an increase in the intracellular calcium concentration ([Ca(2+)]i). The observed [Ca(2+)]i increase did not occur through inositol phosphate formation and was not sensitive to Bordetella pertussis toxin, forskolin, or 3-isobutyl-1-methylxanthine (pre)treatment but was due to Ca(2+) influx from the extracellular environment. Interestingly, the Ca(2+) pathway was dependent on high receptor expression levels and was compound-specific, because benzamide-like compounds triggered two to three times higher responses than indoleamines. Taken together, these data provide the first evidence for fine functional differences between C-terminal splice variants of the h5-HT4 receptor, which may contribute to a better understanding of the functional diversity of this receptor class.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Serotonin/physiology , Adenylyl Cyclases/metabolism , Alternative Splicing , Calcium/metabolism , Cell Line , Enzyme Activation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Indoles/pharmacology , Inositol Phosphates/metabolism , Pertussis Toxin , Receptors, Serotonin/drug effects , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT4 , Serotonin Antagonists/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Sulfur Radioisotopes , Transfection , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL