Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 340(3): 706-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22171093

ABSTRACT

Sodium-activated potassium (K(Na)) channels have been suggested to set the resting potential, to modulate slow after-hyperpolarizations, and to control bursting behavior or spike frequency adaptation (Trends Neurosci 28:422-428, 2005). One of the genes that encodes K(Na) channels is called Slack (Kcnt1, Slo2.2). Studies found that Slack channels were highly expressed in nociceptive dorsal root ganglion neurons and modulated their firing frequency (J Neurosci 30:14165-14172, 2010). Therefore, Slack channel openers are of significant interest as putative analgesic drugs. We screened the library of pharmacologically active compounds with recombinant human Slack channels expressed in Chinese hamster ovary cells, by using rubidium efflux measurements with atomic absorption spectrometry. Riluzole at 500 µM was used as a reference agonist. The antipsychotic drug loxapine and the anthelmintic drug niclosamide were both found to activate Slack channels, which was confirmed by using manual patch-clamp analyses (EC(50) = 4.4 µM and EC(50) = 2.9 µM, respectively). Psychotropic drugs structurally related to loxapine were also evaluated in patch-clamp experiments, but none was found to be as active as loxapine. Loxapine properties were confirmed at the single-channel level with recombinant rat Slack channels. In dorsal root ganglion neurons, loxapine was found to behave as an opener of native K(Na) channels and to increase the rheobase of action potential. This study identifies new K(Na) channel pharmacological tools, which will be useful for further Slack channel investigations.


Subject(s)
Antipsychotic Agents/pharmacology , Loxapine/pharmacology , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism , Action Potentials/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Loxapine/blood , Patch-Clamp Techniques , Potassium Channels, Sodium-Activated , Rats , Rats, Sprague-Dawley , Riluzole/pharmacology , Rubidium/metabolism
2.
Neuroscience ; 156(2): 353-64, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18691638

ABSTRACT

SR58611A is a selective beta(3)-adrenoceptor (Adrb3) agonist which has demonstrated antidepressant and anxiolytic properties in rodents. The present study confirmed the detection of Adrb3 mRNA transcript in rodent brain sub-regions and evaluated the effect of SR58611A on serotonergic and noradrenergic transmission in rats and mice in an attempt to elucidate the mechanism(s) underlying these properties. SR58611A (3 and 10 mg/kg, p.o.) increased the synthesis of 5-HT and tryptophan (Trp) levels in several rodent brain areas (cortex, hippocampus, hypothalamus, striatum). Moreover, SR58611A (10 mg/kg, p.o.) increased the release of 5-HT assessed by in vivo microdialysis in rat prefrontal cortex. Systemic (3 mg/kg, i.v.) or chronic administration of SR58611A (10 mg/kg, p.o.), in contrast to fluoxetine (15 mg/kg, p.o.), did not modify the activity of serotonergic neurons in the rat dorsal raphe nucleus. The increase in 5-HT synthesis induced by SR58611A was not observed in Adrb3s knockout mice, suggesting a selective involvement of Adrb3s in this effect. SR58611A (3 and 10 mg/kg, p.o.) did not modify norepinephrine synthesis and metabolism but increased its release in rat brain. Repeated administration of SR58611A (10 mg/kg, p.o.) did not modify basal norepinephrine release in rat prefrontal cortex whereas it prevented its tail-pinch stress-induced enhancement similarly to reboxetine (15 mg/kg, p.o.). Finally SR58611A increased the firing rate of noradrenergic neurons in the rat locus coeruleus following systemic (3 mg/kg, i.v.) or local (0.01 and 1 microM) but not chronic (10 mg/kg, p.o.) administration. These results suggest that the anxiolytic- and antidepressant-like activities of SR58611A involve an increase of brain serotonergic and noradrenergic neurotransmissions, triggered by activation of Adrb3s.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Brain/drug effects , Norepinephrine/metabolism , Serotonin/metabolism , Tetrahydronaphthalenes/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic beta-2 Receptor Agonists , Analysis of Variance , Animals , Brain/anatomy & histology , Brain/cytology , Brain/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Interactions , Fluoxetine/pharmacology , Male , Mice , Microdialysis , Morpholines/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Neurons/physiology , Rats , Reboxetine , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Tryptophan/metabolism
3.
Arch Mal Coeur Vaiss ; 100(5): 386-93, 2007 May.
Article in French | MEDLINE | ID: mdl-17646762

ABSTRACT

OBJECTIVES: Pulmonary artery banding is often required as a first palliative procedure in infants with congenital heart disease and high pulmonary blood flow or to retrain the left ventricle. The purpose of the study was to demonstrate the safety of a gastric banding system as an adjustable pulmonary artery banding in chronic implantation. METHODS: Five ewes underwent implantation of the banding system around the main pulmonary artery through a left thoracotomy. All had functional evaluation with progressive occlusion and opening of the device every two weeks for a total period of three months. Invasive pressure measurements in the right ventricle and aorta were carried out each time. RESULTS: Devices could be implanted easily. Progressive occlusion and re-opening were possible in all animals during each time point. All animals survived throughout the protocol. Retrieval of the device was achieved in all animals. In one, it was challenging because of the presence of a fibrotic reaction around the device. It died because of pulmonary artery perforation before the sacrifice. At autopsy, microscopic examination showed no signs of myocardial fibrosis. CONCLUSIONS: In animals, gastric banding system is a safe and effective implantable device to adjust pulmonary artery diameter over a prolonged period of time. This new device may be a valuable alternative to the repeated conventional pulmonary artery banding needed for ventricular retraining in humans.


Subject(s)
Prostheses and Implants , Prosthesis Implantation , Pulmonary Artery/surgery , Animals , Aorta/physiology , Blood Pressure/physiology , Constriction , Female , Fibrosis , Models, Animal , Prosthesis Design , Regional Blood Flow/physiology , Sheep , Thoracotomy , Ventricular Function, Right/physiology , Ventricular Pressure/physiology
4.
J Pharmacol Exp Ther ; 306(1): 407-20, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12682217

ABSTRACT

(5aS,8S,10aR)-5a,6,9,10-Tetrahydro,7H,11H-8,10a-methanopyrido[2',3':5,6]pyrano[2,3-d]azepine (SSR591813) is a novel compound that binds with high affinity to the rat and human alpha4beta2 nicotinic acetylcholine receptor (nAChR) subtypes (Ki = 107 and 36 nM, respectively) and displays selectivity for the alpha4beta2 nAChR (Ki, human alpha3beta4 > 1000, alpha3beta2 = 116; alpha1beta1deltagamma > 6000 nM and rat alpha7 > 6000 nM). Electrophysiological experiments indicate that SSR591813 is a partial agonist at the human alpha4beta2 nAChR subtype (EC50 = 1.3 micro M, IA =19% compared with the full agonist 1,1-dimethyl-4-phenyl-piperazinium). In vivo findings from microdialysis and drug discrimination studies confirm the partial intrinsic activity of SSR591813. The drug increases dopamine release in the nucleus accumbens shell (30 mg/kg i.p.) and generalizes to nicotine or amphetamine (10-20 mg/kg i.p.) in rats, with an efficacy approximately 2-fold lower than that of nicotine. Pretreatment with SSR591813 (10 mg/kg i.p.) reduces the dopamine-releasing and discriminative effects of nicotine. SSR591813 shows activity in animal models of nicotine dependence at doses devoid of unwanted side effects typically observed with nicotine (hypothermia and cardiovascular effects). The compound (10 mg/kg i.p.) also prevents withdrawal signs precipitated by mecamylamine in nicotine-dependent rats and partially blocks the discriminative cue of an acute precipitated withdrawal. SSR591813 (20 mg/kg i.p.) reduces i.v. nicotine self-administration and antagonizes nicotine-induced behavioral sensitization in rats. The present results confirm important role for alpha4beta2 nAChRs in mediating nicotine dependence and suggest that SSR591813, a partial agonist at this particular nAChR subtype, may have therapeutic potential in the clinical management of smoking cessation.


Subject(s)
Azepines/therapeutic use , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/metabolism , Smoking Cessation , Smoking/drug therapy , Animals , Behavior, Animal/drug effects , Body Temperature/drug effects , Brain/metabolism , Cardiovascular System/drug effects , Cells, Cultured , Dextroamphetamine/pharmacology , Discrimination Learning , Drug Interactions , Humans , Male , Mecamylamine/pharmacology , Microdialysis , Motor Activity/drug effects , Nicotine/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Self Administration , Substance Withdrawal Syndrome , Transfection , Xenopus laevis
5.
J Pharmacol Exp Ther ; 298(2): 753-68, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454940

ABSTRACT

SL651498 [6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyrido[3,4-b]indol-1-one] is a novel pyridoindole derivative that displays high affinity for rat native GABA(A) receptors containing alpha(1) (K(i) = 6.8 nM) and alpha2 (K(i) = 12.3 nM) subunits, and weaker affinity for alpha5-containing GABA(A) receptors (K(i) = 117 nM). Studies on recombinant rat GABA(A) receptors confirm these data (K(i), alpha1beta2gamma2 = 17, alpha2beta2gamma2 = 73, alpha5beta3gamma2 = 215 nM) and indicate intermediate affinity for the alpha3beta2gamma2 subtype (K(i) = 80 nM). SL651498 behaves as a full agonist at recombinant rat GABA(A) receptors containing alpha2 and alpha3 subunits and as a partial agonist at recombinant GABA(A) receptors expressing alpha1 and alpha5 subunits. SL651498 elicited anxiolytic-like activity similar to that of diazepam [minimal effective dose (MED): 1-10 mg/kg, i.p.] in three conflict models, in the elevated plus-maze, the light/dark test, and the defense test battery in rats and mice. Results from activity tests and electroencephalogram analysis indicated that SL651498 induced muscle weakness, ataxia, or sedation at doses much higher than those producing anxiolytic-like activity (MED > or = 30 mg/kg, i.p.). Repeated treatment for 10 days with SL651498 (30 mg/kg, i.p., b.i.d.) in mice was not associated with the development of tolerance to its anticonvulsant effects or physical dependence. Furthermore, SL651498 was much less active than diazepam in potentiating the depressant effects of ethanol in mice. The "anxioselective" profile of SL651498 points to a major role for GABA(A) alpha2 subtype in regulating anxiety and suggests that selectively targeting GABA(A) receptor subtypes can lead to drugs with increased clinical specificity.


Subject(s)
Anti-Anxiety Agents/pharmacology , Indoles/pharmacology , Pyrroles/pharmacology , Receptors, GABA-A/drug effects , Animals , Anticonvulsants/pharmacology , Anxiety/psychology , Central Nervous System Depressants/pharmacology , Discrimination, Psychological/drug effects , Drug Interactions , Drug Tolerance , Ethanol/pharmacology , Male , Membrane Potentials/drug effects , Mice , Motor Activity/drug effects , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Substance-Related Disorders/psychology
6.
Neuroreport ; 11(14): 3169-73, 2000 Sep 28.
Article in English | MEDLINE | ID: mdl-11043543

ABSTRACT

We analyzed the expression of native GABA(A) receptors in choline acetyltransferase and glutamic acid decarboxilase positive cells, from lamina IX of the lumbar region of rat spinal cord. More than one isoform of each subunit was detected within a single cell. The alpha3, alpha5, alpha1, beta3 and gamma2 subunit was the most frequent combination in both cell populations. However, the total number of subunit expressed by each cell type was different, being the ChAT positive cells the simplest. Interestingly, the ChAT and GAD positive cells also displayed a different pattern of distribution of both spliced isoforms of the gamma2 subunit. These results indicate that several GABA(A) receptors, with different molecular composition, are expressed in a single cell and that different cell types can express different GABA(A) receptors.


Subject(s)
Anterior Horn Cells/metabolism , Receptors, GABA-A/metabolism , Spinal Cord/metabolism , Animals , Animals, Newborn , Cell Count , Choline O-Acetyltransferase/metabolism , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Isoenzymes/metabolism , Protein Isoforms/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Reverse Transcriptase Polymerase Chain Reaction/statistics & numerical data , Spinal Cord/cytology
7.
J Biol Chem ; 275(18): 13856-62, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10788509

ABSTRACT

The neuronal glycine transporter GLYT2 takes up glycine from the extracellular space by an electrogenic process where this neurotransmitter is co-transported with sodium and chloride ions. We report in this paper that tyrosine at position 289 of GLYT2a is crucial for ion coupling, glycine affinity and sodium selectivity, stressing the essential role played by this residue of transmembrane domain III in the mechanism of transport. Substitution to tryptophan (Y289W), phenylalanine (Y289F), or serine (Y289S), renders transporters unable to catalyze glycine uptake. Measurements of glycine evoked steady-state currents in transfected HEK-293 cells reveal EC(50) values for glycine 17-fold (Y289F) and 45-fold (Y289S) higher than that of the wild type transporter. Sodium dependence is severely altered in tyrosine 289 mutants, both at the level of apparent affinity and cooperativity, with the more dramatic change corresponding to the less conservative substitution (Y289S). Accordingly, sodium selectivity is gradually lost in Y289F and Y289S mutants, and chloride dependence of glycine evoked currents is markedly decreased in Y289F and Y289S mutants. In the absence of three-dimensional information from these transporters, these results provide experimental evidence supporting the hypothesis of transmembrane domain III being part of a common permeation pathway for substrate and co-transported ions.


Subject(s)
Amino Acid Transport Systems, Neutral , Carrier Proteins/chemistry , Animals , Biological Transport , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Glycine/chemistry , Glycine/metabolism , Glycine Plasma Membrane Transport Proteins , Mutagenesis, Site-Directed , Protein Conformation , Structure-Activity Relationship , Tyrosine
8.
Psychopharmacology (Berl) ; 146(2): 205-13, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10525757

ABSTRACT

RATIONALE: It has been suggested that different BZ (omega) receptor subtypes may mediate distinct behavioural effects of BZ receptor ligands. OBJECTIVE: The present study examined this hypothesis further. METHODS: The antagonism exerted by the selective BZ(1) (omega(1)) receptor antagonist beta-CCT on the pharmacological effects of the selective BZ(1) (omega(1)) receptor agonist zolpidem and the non-selective BZ (omega) receptor agonist diazepam in behavioural, biochemical and electrophysiological experiments was assessed. RESULTS: beta-CCT which was devoid of activity per se, antagonized the effects of the non-selective BZ (omega) receptor full agonist diazepam and the selective BZ(1) (omega(1)) receptor full agonist zolpidem against seizures produced by isoniazid, but beta-CCT failed to affect their action on seizures produced by pentylenetetrazole (PTZ), suggesting that BZ(2) (omega(2)) receptors may be primarily involved in the convulsant action of PTZ. In the light/dark test, beta-CCT abolished the anxiolytic-like action of diazepam. In tests designed to investigate the central depressant activity of drugs, beta-CCT antagonized the sedative effects of diazepam and zolpidem, but failed to modify clearly the myorelaxant effects of diazepam. These differences may be related to the selectivity of beta-CCT for BZ(1) (omega(1)) sites as indicated by the preferential displacement of [(3)H]flumazenil in BZ(1) (omega(1))-enriched structures as compared to BZ(2) (omega(2))-enriched structures in the mouse. In in vitro experiments, beta-CCT antagonized the potentiation of the GABA-induced Cl(-) current produced by zolpidem in HEK cells expressing the alpha(1)beta(2)gamma(2) receptor or in cerebellar Purkinje neurones, while it failed to modify the diazepam potentiation at either alpha(3)beta(2)gamma(2) or alpha(5)beta(3)gamma(2) receptor subtypes. CONCLUSION: These results are consistent with the hypothesis that BZ(1) (omega(1)) receptors play an important role in the anxiolytic and sedative/hypnotic effects of BZ (omega) receptor ligands, whereas activity at BZ(2) (omega(2)) sites might be associated primarily with muscle relaxation.


Subject(s)
Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Receptors, GABA-A/drug effects , Animals , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Binding, Competitive/drug effects , Carbolines/pharmacology , Diazepam/pharmacokinetics , Diazepam/pharmacology , Flumazenil/pharmacokinetics , GABA Antagonists/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Ligands , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Muscle Relaxants, Central/pharmacology , Patch-Clamp Techniques , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Zolpidem
9.
J Biol Chem ; 274(19): 13370-4, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224099

ABSTRACT

gamma-aminobutyric acid type A (GABAA) receptors comprise a subfamily of ligand-gated ion channels whose activity can be modulated by ligands acting at the benzodiazepine binding site on the receptor. The benzodiazepine binding site was characterized using a site-directed mutagenesis strategy in which amino acids of the alpha5 subunit were substituted by their corresponding alpha1 residues. Given the high affinity and selectivity of alpha1-containing compared with alpha5-containing GABAA receptors for zolpidem, mutated alpha5 subunits were co-expressed with beta2 and gamma2 subunits, and the affinity of recombinant receptors for zolpidem was measured. One alpha5 mutant (bearing P162T, E200G, and T204S) exhibited properties similar to that of the alpha1 subunit, notably high affinity zolpidem binding and potentiation by zolpidem of GABA-induced chloride current. Two of these mutations, alpha5P162T and alpha5E200G, might alter binding pocket conformation, whereas alpha5T204S probably permits formation of a hydrogen bond with a proton acceptor in zolpidem. These three amino acid substitutions also influenced receptor affinity for CL218872. Our data thus suggest that corresponding amino acids of the alpha1 subunit, particularly alpha1-Ser204, are the crucial residues influencing ligand selectivity at the binding pocket of alpha1-containing receptors, and a model of this binding pocket is presented.


Subject(s)
Benzodiazepines/metabolism , Receptors, GABA-A/metabolism , Allosteric Site , Amino Acid Sequence , Animals , Binding Sites , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Pyridines/metabolism , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Sequence Homology, Amino Acid , Zolpidem
10.
J Affect Disord ; 51(3): 287-303, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10333983

ABSTRACT

Befloxatone, a novel oxazolidinone derivative, is a potent, selective and reversible monoamine oxidase A (MAO-A) inhibitor in vitro (K1A = 1.9-3.6 nM) and ex vivo (ED50 MAO-A = 0.02 mg/kg, p.o.). It does not interact with a large number of receptors, monoamine transporters or other amine oxidases. Binding studies with [3H]-befloxatone in rat brain sections show that it labels with high affinity (Kd = 1.3 nM) a single population of sites with the pharmacological characteristics and regional distribution of MAO-A. In the rat brain, befloxatone (0.75 mg/kg, i.p.) increases tissue levels of monoamines and decreases levels of their deaminated metabolites. Acute administration of befloxatone (0.75 mg/kg, i.p.) induces an increase in extracellular striatal dopamine and cortical norepinephrine but not cortical serotonin levels in the rat. Befloxatone (1 mg/kg, i.p.) potently inhibits the firing rate of serotonergic neurons, partially decreases the firing of noradrenergic neurons and has no effect on the firing of dopaminergic neurons (a mirror image of its effects on monoamine release in terminal regions), suggesting that the relative effects of befloxatone on monoamine release may be governed by autoreceptor-mediated control of monoaminergic neurons at the cell body level. Befloxatone (0.03-0.3 mg/kg, p.o.) exhibits potent activity in behavioural models predictive of antidepressant activity. Befloxatone (up to 1.5 mg/kg, p.o.) does not potentiate the pressor effects of orally administered tyramine at centrally active doses and duodenal [3H]-befloxatone binding is displaced by increasing doses of orally administered tyramine (0.1-40 mg/kg, i.p.). These results suggest that befloxatone is a potent reversible MAO-A inhibitor with antidepressant potential and a wide safety margin with regard to the potentiation of the pressor effect of tyramine.


Subject(s)
Monoamine Oxidase Inhibitors/pharmacology , Oxazoles/pharmacology , Animals , Autoradiography , Autoreceptors/drug effects , Autoreceptors/metabolism , Autoreceptors/physiology , Brain/diagnostic imaging , Brain/enzymology , Brain/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Locus Coeruleus/physiology , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/therapeutic use , Oxazoles/metabolism , Oxazoles/therapeutic use , Protein Binding , Radiography , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Rats , Receptors, Drug/drug effects , Receptors, Drug/metabolism , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/metabolism , Tissue Distribution
11.
Eur J Pharmacol ; 323(2-3): 277-81, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9128850

ABSTRACT

We have investigated, by using the whole-cell patch-clamp technique, the Ca2+ channel antagonist properties of eliprodil in cultured cerebellar granule cells which are known to express L-, N-, P- as well as Q- and R-type Ca2+ channels. Eliprodil maximally antagonized 50% of the voltage-dependent Ba2+ current with an IC50 of 4 microM. omega-Conotoxin-GVIA (3.2 microM) and omega-agatoxin-IVA (0.5 microM) blocked 28 and 43% of the current, respectively. When eliprodil (30 microM) was added to omega-conotoxin-GVIA or omega-agatoxin-IVA the magnitude of the maximal inhibition was identical to that obtained with eliprodil alone confirming a full blockade by eliprodil of N-, P- and Q-type Ca2+ channels. The L-type channel antagonist nimodipine (10 microM) blocked 24% of the current; this blockade was fully additive to that of eliprodil, indicating that the nimodipine-sensitive component of the current is eliprodil-insensitive. In the presence of eliprodil and nimodipine a residual Cd2+ sensitive current (25%), identified as the R-type current, remained unblocked. We conclude that in cerebellar granule neurons R- and L-type Ca2+ channels are insensitive to eliprodil. The nimodipine-sensitive channels present in cerebellar granule neurons may represent a neuronal subtype of L channels distinct from that (eliprodil-sensitive/nimodipine-sensitive) present in cortical or hippocampal neurons.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cerebellum/drug effects , Neurons/drug effects , Piperidines/pharmacology , Animals , Barium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Drug Interactions , Neurons/metabolism , Nimodipine/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Rats , Spider Venoms/pharmacology , omega-Agatoxin IVA , omega-Conotoxin GVIA
12.
Neurosci Lett ; 223(2): 133-6, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9089691

ABSTRACT

We have studied the effects of a variety of N-methyl-D-aspartate (NMDA) antagonists acting at different sites of the NMDA receptor complex on NMDA-induced currents in Xenopus oocytes expressing heteromeric NR1A/NR2 and NR1A/NR2B receptors. The polyamine site antagonists eliprodil (IC50 = 3.0 microM) and ifenprodil (IC50 = 0.27 microM) antagonized NMDA responses at NR1A/NR2B receptors but not at NR1A/NR2A receptors (IC50 > 100 microM). The channel blockers dizocilpine, memantine and phencyclidine (PCP) were equally potent antagonists at both receptor subtypes whereas dextromethorphan was four times more potent at NR1A/NR2A receptors. The glycine site antagonists L-689,560 and 7-Cl-kynurenate were 10 times more potent at NR1A/NR2A than at NR1A/NR2B receptor subtypes. The selectivity of eliprodil and ifenprodil for the NR1A/NR2B receptor subtype may, at least partially, explain their favorable side effects profile.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Binding Sites , Electric Conductivity , Female , Patch-Clamp Techniques , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Xenopus
13.
J Recept Signal Transduct Res ; 17(1-3): 99-113, 1997.
Article in English | MEDLINE | ID: mdl-9029483

ABSTRACT

The experiments reported here were motivated by our interest to express in stably-transfected cells large amounts of recombinant rat GABAA receptors. For this, we developed an original two step selection strategy, in which the first step consisted of transfecting HEK 293 cells with rat GABAA receptor alpha and beta subunits. G 418 resistant colonies isolated at this step were screened for [3H] muscimol binding to select for those that coexpressed alpha- and beta-subunits. The best alpha and beta subunit expressing colony was then supertransfected with a plasmid coding for the gamma rat GABAA receptor subunit and a mutant DHFR gene. After a second round of selection, this time in presence of methotrexate, those colonies that coexpressed ternary alpha beta gamma GABAA receptor combinations were distinguished using [3H] flumazenil as a probe. This strategy was applied to the isolation of 3 GABAA receptor clones, alpha 1 beta 2 gamma 2s, alpha 3 beta 2 gamma 2s and alpha 5 beta 3 gamma 2s, that expressed relatively high levels of these proteins. These 3 cell lines exhibited pharmacological and functional properties similar to cells transiently-transfected with equivalent subunit combinations. These cell lines therefore provide attractive models with which to evaluate the intrinsic activity and potency of compounds at recombinant GABAA receptor subtypes.


Subject(s)
Receptors, GABA-A/metabolism , Animals , Benzodiazepines/metabolism , Binding, Competitive , Cell Line , Chloride Channels/metabolism , Electrophysiology , Flumazenil/metabolism , Humans , Kinetics , Protein Conformation , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Recombinant Proteins/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transfection
14.
J Pharmacol Exp Ther ; 279(3): 1092-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8968329

ABSTRACT

Tolerance to benzodiazepines (BZs) is thought to involve alterations of the gamma-aminobutyric acid (GABA)A receptor as a result of the prolonged occupancy of its modulatory BZ recognition site. We used the whole-cell patch-clamp technique to compare the functional and pharmacological properties of GABAA receptors in acutely dissociated hippocampal neurons from the control or diazepam-tolerant rats. Administration of diazepam (15 mg/kg p.o.) twice a day for 10 days induced tolerance as demonstrated by the decreased potency of acute diazepam i.p. injections to protect against pentylenetetrazole-induced clonictonic convulsions (10.5% of tolerant rats protected by 0.1 mg/kg of diazepam against 55% of nontreated rats, 48 hr after the last dose of the chronic treatment). The specific current induced by 1 microM GABA in acutely dissociated hippocampal neurons 48 hr after withdrawal (10.5 +/- 1.3 microA/cm2) was similar to that observed in the control rats (8.7 +/- 0.8 microA/cm2). The EC50 value for GABA was unchanged by the chronic treatment [6.3 (5.4-7.1) and 7.5 (6.2-8.7) microM in neurons from the control and treated rats, respectively]. The potency of the nonselective allosteric modulator diazepam to stimulate Cl- currents was identical in cells from treated rats [EC50 values of 25 (20-30) and 34 (26-41) nM in the control and treated rats, respectively; P < .05], but the potency of the selective BZ1-site ligand zolpidem was decreased [EC50 values of 99 (88-111) and 267 (221-313) nM in the control and treated rats, respectively; P < .05]. The maximal potentiation of the GABA-induced current was significantly decreased with diazepam (maximal potentiation: 168.0 +/- 16.2 and 124.0 +/- 8.9% in the control and treated rats, respectively). These results suggest that tolerance to diazepam is accompanied in hippocampal neurons by a decrease in BZ1 binding sites and in the functional coupling of BZ/GABA recognition sites.


Subject(s)
Diazepam/administration & dosage , Hippocampus/drug effects , Neurons/drug effects , Receptors, GABA-A/drug effects , gamma-Aminobutyric Acid/metabolism , Allosteric Regulation , Animals , Diazepam/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Male , Neurons/metabolism , Patch-Clamp Techniques , Pentylenetetrazole/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Zolpidem
15.
Neuropharmacology ; 35(2): 137-45, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8734481

ABSTRACT

The whole-cell patch-clamp technique was used to compare the properties of native GABAA receptors in Purkinje and striatal neurons acutely dissociated from neonatal rat brains (7-11 days old). In symmetrical chloride concentrations and at a negative holding voltage, GABA induced inward currents in a concentration-dependent manner with EC50 values of 4 and 8 uM in Purkinje and striatal neurons, respectively. Diazepam potentiated the current induced by 1 uM GABA in Purkinje and striatal neurons with EC50 values of 28 and 42 nM and maximal potentiations of 128 and 182%, respectively. Zolpidem potentiated this GABA-induced current in Purkinje and striatal neurons with EC50 values of 33 and 195 nM and maximal potentiations of 189 and 236%, respectively. These results show that zolpidem, in contrast to diazepam, functionally discriminates subtypes of native GABAA receptors. Zolpidem has greater affinity for GABAA receptors containing omega 1 (Purkinje cells) than for those with omega 2 (striatum) sites and has higher intrinsic activity at these receptors than diazepam. These properties of zolpidem may contribute to its hypnoselective profile.


Subject(s)
Corpus Striatum/drug effects , Hypnotics and Sedatives/pharmacology , Neurons/drug effects , Purkinje Cells/drug effects , Pyridines/pharmacology , Receptors, GABA-A/drug effects , Animals , Corpus Striatum/physiology , Diazepam/pharmacology , Dose-Response Relationship, Drug , Neurons/physiology , Patch-Clamp Techniques , Purkinje Cells/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/physiology , Zolpidem , gamma-Aminobutyric Acid/pharmacology
16.
Eur J Pharmacol ; 296(2): 209-13, 1996 Jan 25.
Article in English | MEDLINE | ID: mdl-8838458

ABSTRACT

The NMDA receptor antagonist ifenprodil contains two asymmetric centres which give rise to four stereoisomeric forms of this molecule. The inhibitory effects of each of these stereoisomers on recombinant NMDA receptors expressed from NR1A/NR2A and NR1A/NR2B subunit combinations were studied in Xenopus oocytes by voltage-clamp recording. All four ifenprodil stereoisomers were potent antagonists at NR1A/NR2B (IC50 < 0.8 microM), but weak antagonists at NR1A/NR2A receptors (IC50 > 100 microM). In heteromeric NR1A/NR2B receptors, (+) erythro- and (-) threo-ifenprodil (IC50 0.21 and 0.22 microM, respectively) were about 4 times more potent than (-) erythro- and (+) threo-ifenprodil (IC50 0.81 and 0.76, respectively). These results show that the stereoisomers of ifenprodil exhibit a weak though significant stereoselectivity at the NR1A/NR2B NMDA receptor subtype.


Subject(s)
Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Oocytes , Piperidines/chemistry , Receptors, N-Methyl-D-Aspartate/classification , Receptors, N-Methyl-D-Aspartate/genetics , Stereoisomerism , Xenopus laevis
17.
Eur J Pharmacol ; 294(1): 91-100, 1995 Dec 27.
Article in English | MEDLINE | ID: mdl-8788420

ABSTRACT

The effect of eliprodil on P-type Ca2+ channels was investigated in acutely dissociated rat Purkinje neurons, by using the whole-cell patch-clamp technique. Eliprodil inhibited in a reversible manner the omega-agatoxin-IVA-sensitive Ba2+ current elicited by step depolarizations from a -80 mV holding voltage (IC50 = 1.9 microM). The Ba2+ current showed steady-state inactivation (V1/2 = -61 mV) which was shifted toward more positive values when the intracellular Ca2+ buffering was increased. In these conditions, the potency of eliprodil was decreased (IC50 = 8.2 microM), suggesting a modulation by intracellular Ca2+ of the eliprodil blockade. The potency of eliprodil was not modified at more depolarized holding potentials and was not dependent on the frequency at which the step-depolarizations were applied (0-0.2 Hz) indicating a lack of voltage and use dependence of the eliprodil blockade. When eliprodil was applied in the patch-pipette at a concentration which causes full block when applied externally, the Ba2+ current amplitude was not affected and external application of eliprodil was still efficacious, indicating an extracellular location of the binding site. Analysis of the time course of recovery from Ca2+ channel blockade obtained by concomitant application of eliprodil with Cd2+, omega-agatoxin-IVA or fluspirilene, indicated that these later compounds did not interact with eliprodil, suggesting that eliprodil acts at a different site. These results demonstrate that eliprodil blocks P-type Ca2+ channels in cerebellar Purkinje neurons and suggest that this property may contribute to its neuroprotective activity.


Subject(s)
Calcium Channel Blockers/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Piperidines/pharmacology , Purkinje Cells/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Binding Sites/drug effects , Cadmium/pharmacology , Dopamine Antagonists/pharmacology , Electrophysiology , Fluspirilene/pharmacology , In Vitro Techniques , Patch-Clamp Techniques , Piperidines/antagonists & inhibitors , Purkinje Cells/drug effects , Rats , Rats, Sprague-Dawley , Solutions , Spider Venoms/pharmacology , omega-Agatoxin IVA
18.
Mol Pharmacol ; 47(6): 1189-96, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7603459

ABSTRACT

We report here that carbamazepine and phenytoin, two widely used antiepileptic drugs, potentiate gamma-aminobutyric acid (GABA)-induced Cl- currents in human embryonic kidney cells transiently expressing the alpha 1 beta 2 gamma 2 subtype of the GABAA receptor and in cultured rat cortical neurons. In cortical neuron recordings, the current induced by 1 microM GABA was enhanced by carbamazepine and phenytoin with EC50 values of 24.5 nM and 19.6 nM and maximal potentiations of 45.6% and 90%, respectively. The potentiation by these compounds was dependent upon the concentration of GABA, suggesting an allosteric modulation of the receptor, but was not antagonized by the benzodiazepine (omega) modulatory site antagonist flumazenil. Carbamazepine and phenytoin did not modify GABA-induced currents in human embryonic kidney cells transiently expressing binary alpha 1 beta 2 recombinant GABAA receptors. The alpha 1 beta 2 recombinant is known to possess functional barbiturate, steroid, and picrotoxin sites, indicating that these sites are not involved in the modulatory effects of carbamazepine and phenytoin. When tested in cells containing recombinant alpha 1 beta 2 gamma 2, alpha 3 beta 2 gamma 2, or alpha 5 beta 2 gamma 2 GABAA receptors, carbamazepine and phenytoin potentiated the GABA-induced current only in those cells expressing the alpha 1 beta 2 gamma 2 receptor subtype. This indicates that the nature of the alpha subunit isoform plays a critical role in determining the carbamazepine/phenytoin pharmacophore. Our results therefore illustrate the existence of one or more new allosteric regulatory sites for carbamazepine and phenytoin on the GABAA receptor. These sites could be implicated in the known anticonvulsant properties of these drugs and thus may offer new targets in the search for novel antiepileptic drugs.


Subject(s)
Carbamazepine/pharmacology , Phenytoin/pharmacology , Receptors, GABA-A/drug effects , Animals , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Humans , Neurons/drug effects , Neurons/metabolism , Rats , Receptors, GABA-A/metabolism
19.
Eur J Pharmacol ; 257(3): 297-301, 1994 May 23.
Article in English | MEDLINE | ID: mdl-8088348

ABSTRACT

The effect of the non-competitive NMDA receptor antagonist eliprodil on NMDA receptor- and voltage-operated Ca2+ currents was investigated in rat cultured cortical neurons by using the whole-cell patch clamp technique. With neurons voltage-clamped at -40 mV, eliprodil reduced in a concentration-dependent manner the inward current induced by N-methyl-D-aspartate (NMDA) (10 microM) in the presence of D-serine with an IC50 of 0.67 microM (Imax = 83%). Eliprodil also blocked the total inward Ba2+ current carried in part by L- and N-type Ca2+ channels with an IC50 of 1.48 microM (Imax = 87%). These results suggest that the neuroprotective properties of eliprodil could be due to its combined ability to antagonize the NMDA receptor- and voltage-operated Ca2+ channels.


Subject(s)
Calcium Channels/drug effects , Cerebral Cortex/drug effects , Neurons/drug effects , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Electrophysiology , Neurons/cytology , Neurons/metabolism , Rats
20.
J Gen Physiol ; 100(5): 803-24, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1335477

ABSTRACT

The activity of taste cells maintained in the intact hamster tongue was monitored in response to acid stimulation by recording action currents from taste receptor cells with an extracellular "macro" patch pipette: a glass pipette was pressed over the taste pore of fungiform papillae and perfused with citric acid, hydrochloric acid, or NaCl. Because this technique restricted stimulus application to the small surface area of the apical membranes of the taste cells, many nonspecific, and potentially detrimental, effects of acid stimulation could be avoided. Acid stimulation reliably elicited fast transient currents (action currents of average amplitude, 9 pA) which were consistently smaller than those elicited by NaCl (29 pA). The frequency of action currents elicited by acid stimuli increased in a dose-dependent manner with decreasing pH from a threshold of about pH 5.0. Acid-elicited responses were independent of K+, Na+, Cl-, or Ca2+ at physiological (salivary) concentrations, and were unaffected by anthracene-9-carboxylic acid, tetraethylammonium bromide, diisothiocyanate-stilbene-2,2'-disulfonic acid, vanadate, or Cd2+. In contrast, amiloride (< or = 30 microM) fully and reversibly suppressed acid-evoked action currents. At submaximal amiloride concentrations, the frequency and amplitude of the action currents were reduced, indicating a reduction of the taste cell apical conductance concomitant with a decrease in cell excitation. Exposure to low pH elicited, in addition to transient currents, an amiloride-sensitive sustained d.c. current. This current is apparently carried by protons instead of Na+ through amiloride-sensitive channels. When citric acid was applied while the taste bud was stimulated by NaCl, the action currents became smaller and the response resembled that produced by acid alone. Because of the strong interdependence of the acid and salt (NaCl) responses when both stimuli are applied simultaneously, and because of the similarity in the concentration dependence of amiloride block, we conclude that amiloride-sensitive Na+ channels on hamster taste receptor cells are permeable to protons and may play a role in acid (sour) taste.


Subject(s)
Sodium Channels/metabolism , Taste Buds/metabolism , Action Potentials/drug effects , Amiloride/pharmacology , Animals , Citrates/pharmacology , Citric Acid , Cricetinae , Hydrogen-Ion Concentration , In Vitro Techniques , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Permeability , Protons , Saliva, Artificial , Sodium Channels/drug effects , Sodium Chloride/pharmacology , Taste Buds/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...