Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Pharmacol Exp Ther ; 376(3): 374-384, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33443077

RÉSUMÉ

Pharmacodynamic efficacy of drugs to activate their receptors is a key determinant of drug effects, and intermediate-efficacy agonists are often useful clinically because they retain sufficient efficacy to produce therapeutically desirable effects while minimizing undesirable effects. Molecular mechanisms of efficacy are not well understood, so rational drug design to control efficacy is not yet possible; however, receptor theory predicts that fixed-proportion mixtures of an agonist and antagonist for a given receptor can be adjusted to precisely control net efficacy of the mixture in activating that receptor. Moreover, the agonist proportion required to produce different effects provides a quantitative scale for comparing efficacy requirements across those effects. To test this hypothesis, the present study evaluated effectiveness of fixed-proportion agonist/antagonist mixtures to produce in vitro and in vivo effects mediated by µ-opioid receptors (MOR) and cannabinoid type 1 receptors (CB1R). Mixtures of 1) the MOR agonist fentanyl and antagonist naltrexone and 2) the CB1R agonist CP55,940 and antagonist/inverse agonist rimonabant were evaluated in an in vitro assay of ligand-stimulated guanosine 5'-O-(3-[35S]thio)triphosphate binding and an in vivo assay of thermal nociception in mice. For both agonist/antagonist pairs in both assays, increasing agonist proportions produced graded increases in maximal mixture effects, and lower agonist proportions were sufficient to produce in vivo than in vitro effects. These findings support the utility of agonist-antagonist mixtures as a strategy to control net efficacy of receptor activation and to quantify and compare efficacy requirements across a range of in vitro and in vivo endpoints. SIGNIFICANCE STATEMENT: Manipulation of agonist proportion in agonist/antagonist mixtures governs net mixture efficacy at the target receptor. Parameters of agonist/antagonist mixture effects can provide a quantitative metric for comparison of efficacy requirements across a wide range of conditions.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Cannabinoïdes/pharmacologie , Animaux , Cellules CHO , Cricetulus , Relation dose-effet des médicaments , Interactions médicamenteuses , Mâle , Souris , Récepteur cannabinoïde de type CB1/agonistes , Récepteur cannabinoïde de type CB1/antagonistes et inhibiteurs , Récepteur mu/agonistes , Récepteur mu/antagonistes et inhibiteurs
2.
Psychopharmacology (Berl) ; 237(9): 2777-2793, 2020 Sep.
Article de Anglais | MEDLINE | ID: mdl-32529265

RÉSUMÉ

RATIONALE: Cancer patients receiving the antineoplastic drug paclitaxel report higher incidences and longer duration of treatment-resistant depression than patients receiving other classes of chemotherapeutics. Rodents treated with paclitaxel exhibit a suite of changes in affect-like behaviors. Further, paclitaxel causes chemotherapy-induced peripheral neuropathy (CIPN) in humans and rodents. Kappa opioid receptors (KOR) have a well-established role in depression and neuropathy. The contributions of KOR signaling to paclitaxel-induced aversive-like state and CIPN in rodents remain to be explored. OBJECTIVES: We aimed to investigate whether dysregulation of the KOR/dynorphin system is associated with paclitaxel-mediated pain-like behavior and depression-like behavior. METHODS: Cancer-free male C57BL/6J mice were treated with four injections of vehicle or paclitaxel (32 mg/kg cumulative). The effects of the selective KOR antagonist norbinaltorphimine (norBNI) on paclitaxel-induced sucrose preference deficits and mechanical hypersensitivity were measured. Prodynorphin mRNA and receptor-mediated G protein activation were measured at two time points following the last paclitaxel injection using quantitative real-time polymerase chain reaction and agonist-stimulated [35S]guanosine-5'-O'-(γ-thio)-triphosphate ([35S]GTPγS) binding, respectively, in the nucleus accumbens (NAc), caudate-putamen, amygdala, and spinal cord. RESULTS: Paclitaxel produced a norBNI-reversible sucrose preference deficit, whereas mechanical hypersensitivity was not reversed by norBNI. Paclitaxel treatment increased the levels of mRNA for prodynorphin, a precursor for endogenous KOR agonists, in the NAc. Paclitaxel also had time-dependent effects on KOR-mediated G protein activation in the NAc. CONCLUSIONS: These results suggest that KOR signaling mediates an initial aversive component of paclitaxel, but not necessarily paclitaxel-induced mechanical hypersensitivity.


Sujet(s)
Antinéoplasiques d'origine végétale/toxicité , Apprentissage par évitement/effets des médicaments et des substances chimiques , Paclitaxel/toxicité , Neuropathies périphériques/induit chimiquement , Neuropathies périphériques/métabolisme , Récepteur kappa/métabolisme , Amygdale (système limbique)/effets des médicaments et des substances chimiques , Amygdale (système limbique)/métabolisme , Animaux , Apprentissage par évitement/physiologie , Relation dose-effet des médicaments , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Mâle , Souris , Souris de lignée C57BL , Noyau accumbens/effets des médicaments et des substances chimiques , Noyau accumbens/métabolisme
3.
J Pharmacol Exp Ther ; 359(2): 329-339, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-27535976

RÉSUMÉ

Synthetic cannabinoids (SCs) are an emerging class of abused drugs that differ from each other and the phytocannabinoid ∆9-tetrahydrocannabinol (THC) in their safety and cannabinoid-1 receptor (CB1R) pharmacology. As efficacy represents a critical parameter to understanding drug action, the present study investigated this metric by assessing in vivo and in vitro actions of THC, two well-characterized SCs (WIN55,212-2 and CP55,940), and three abused SCs (JWH-073, CP47,497, and A-834,735-D) in CB1 (+/+), (+/-), and (-/-) mice. All drugs produced maximal cannabimimetic in vivo effects (catalepsy, hypothermia, antinociception) in CB1 (+/+) mice, but these actions were essentially eliminated in CB1 (-/-) mice, indicating a CB1R mechanism of action. CB1R efficacy was inferred by comparing potencies between CB1 (+/+) and (+/-) mice [+/+ ED50 /+/- ED50], the latter of which has a 50% reduction of CB1Rs (i.e., decreased receptor reserve). Notably, CB1 (+/-) mice displayed profound rightward and downward shifts in the antinociception and hypothermia dose-response curves of low-efficacy compared with high-efficacy cannabinoids. In vitro efficacy, quantified using agonist-stimulated [35S]GTPγS binding in spinal cord tissue, significantly correlated with the relative efficacies of antinociception (r = 0.87) and hypothermia (r = 0.94) in CB1 (+/-) mice relative to CB1 (+/+) mice. Conversely, drug potencies for cataleptic effects did not differ between these genotypes and did not correlate with the in vitro efficacy measure. These results suggest that evaluation of antinociception and hypothermia in CB1 transgenic mice offers a useful in vivo approach to determine CB1R selectivity and efficacy of emerging SCs, which shows strong congruence with in vitro efficacy.


Sujet(s)
Évaluation préclinique de médicament/méthodes , Récepteur cannabinoïde de type CB1/agonistes , Récepteur cannabinoïde de type CB1/métabolisme , Animaux , Cannabinoïdes/pharmacologie , Femelle , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Mâle , Souris , Souris transgéniques , Récepteur cannabinoïde de type CB1/déficit
4.
Life Sci ; 92(8-9): 446-52, 2013 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-22940268

RÉSUMÉ

Cannabinoid CB1 receptors (CB1Rs) are expressed throughout the brain and mediate the central effects of cannabinoids, including Δ(9)-tetrahydrocannabinol (THC), the main psychoactive constituent of marijuana. Repeated THC administration produces tolerance to cannabinoid-mediated effects, although the magnitude of tolerance varies by effect. Consistent with this observation, CB1R desensitization and downregulation, as well as induction of immediate early genes (IEGs), vary by brain region. Zif268 and c-Fos are induced in the forebrain after acute THC administration. Phosphorylation of the cAMP response-element binding protein (CREB) is increased in a region-specific manner after THC administration. Results differ between acute versus repeated THC injection, and suggest that tolerance to IEG activation might develop in some regions. Repeated THC treatment produces CB1R desensitization and downregulation in the brain, although less adaption occurs in the striatum as compared to regions such as the hippocampus. Repeated THC treatment also induces expression of ΔFosB, a very stable isoform of FosB, in the striatum. Transgenic expression of ∆FosB in the striatum enhances the rewarding effects of several drugs, but its role in THC-mediated effects is not known. The inverse regional relationship between CB1R desensitization and ∆FosB induction suggests that these adaptations might inhibit each other, although this possibility has not been investigated. The differential regional expression of individual IEGs by acute or repeated THC administration suggests that regulation of target genes and effects on CB1R signaling will contribute to the behavioral effects of THC.


Sujet(s)
Chimie du cerveau/physiologie , Récepteur cannabinoïde de type CB1/physiologie , Chimie du cerveau/génétique , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/physiologie , Dronabinol/pharmacologie , Expression des gènes/génétique , Expression des gènes/physiologie , Gènes précoces , Gènes fos/génétique , Humains , Protéines proto-oncogènes c-fos/biosynthèse , Protéines proto-oncogènes c-fos/génétique , Récepteur cannabinoïde de type CB1/effets des médicaments et des substances chimiques , Récepteur cannabinoïde de type CB1/génétique , Récepteur cannabinoïde de type CB1/métabolisme
5.
Neuroimage ; 52(4): 1243-51, 2010 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-20451624

RÉSUMÉ

CB(1) receptors mediate the CNS effects of Delta(9)-tetrahydrocannabinol and synthetic cannabinoids. Previous studies have investigated cannabinoid-mediated G-protein activity in a subset of brain regions thought to mediate the behavioral effects of cannabinoids, but a detailed regional comparison of the effects of multiple ligands has not been conducted. This study used a novel approach, Statistical Parametric Mapping (SPM), to analyze 3D reconstructed brain images derived from agonist-stimulated [(35)S]GTPgammaS autoradiography in a whole-brain unbiased manner. SPM analysis demonstrated regional differences in the relative efficacies of cannabinoid agonists methanandamide (M-AEA), CP55,940 (CP) and WIN55,212-2 (WIN) in CB(1)(+/+) mouse brains. To assess the potential contribution of novel cannabinoid binding sites, experiments were performed in CB(1)(-/-) mouse brains. SPM analysis revealed that the aminoalkylindole WIN, but not the bicyclic cannabinoid CP or the endocannabinoid analogue M-AEA, stimulated [(35)S]GTPgammaS binding in cortex, hippocampus, hypothalamus, amygdala, cerebellum and certain brainstem areas (dorsal tegmental complex and locus coeruleus). No differences between WIN-stimulated G-protein activity and basal activity were found in basal ganglia. Pharmacological experiments using the CB(1) antagonist SR141716A in CB(1)(+/+) mice showed that SR141716A blocked WIN-stimulated G-protein activity in all brain regions, suggesting that it binds to both CB(1) and putative non-CB(1) sites. These studies show ligand and region-specific cannabinoid-mediated G-protein activity at both CB(1) and non-CB(1) sites and demonstrate that SPM is a powerful approach for the analysis of reconstructed brain imaging data derived from agonist-stimulated [(35)S]GTPgammaS autoradiography.


Sujet(s)
Encéphale/métabolisme , Protéines G/métabolisme , Imagerie tridimensionnelle/méthodes , Récepteur cannabinoïde de type CB1/métabolisme , Animaux , Encéphale/anatomie et histologie , Interprétation statistique de données , Souris , Souris de lignée C57BL , Souris transgéniques , Distribution tissulaire
6.
Synapse ; 62(5): 358-69, 2008 May.
Article de Anglais | MEDLINE | ID: mdl-18293355

RÉSUMÉ

The transcription factor DeltaFosB accumulates and persists in brain in response to chronic stimulation. This accumulation after chronic exposure to drugs of abuse has been demonstrated previously by Western blot most dramatically in striatal regions, including dorsal striatum (caudate/putamen) and nucleus accumbens. In the present study, we used immunohistochemistry to define with greater anatomical precision the induction of DeltaFosB throughout the rodent brain after chronic drug treatment. We also extended previous research involving cocaine, morphine, and nicotine to two additional drugs of abuse, ethanol and Delta(9)-tetrahydrocannabinol (Delta(9)-THC, the active ingredient in marijuana). We show here that chronic, but not acute, administration of each of four drugs of abuse, cocaine, morphine, ethanol, and Delta(9)-THC, robustly induces DeltaFosB in nucleus accumbens, although different patterns in the core vs. shell subregions of this nucleus were apparent for the different drugs. The drugs also differed in their degree of DeltaFosB induction in dorsal striatum. In addition, all four drugs induced DeltaFosB in prefrontal cortex, with the greatest effects observed with cocaine and ethanol, and all of the drugs induced DeltaFosB to a small extent in amygdala. Furthermore, all drugs induced DeltaFosB in the hippocampus, and, with the exception of ethanol, most of this induction was seen in the dentate. Lower levels of DeltaFosB induction were seen in other brain areas in response to a particular drug treatment. These findings provide further evidence that induction of DeltaFosB in nucleus accumbens is a common action of virtually all drugs of abuse and that, beyond nucleus accumbens, each drug induces DeltaFosB in a region-specific manner in brain.


Sujet(s)
Chimie du cerveau/effets des médicaments et des substances chimiques , Chimie du cerveau/génétique , Substances illicites/pharmacologie , Protéines proto-oncogènes c-fos/biosynthèse , Animaux , Dépresseurs du système nerveux central/pharmacologie , Cocaïne/pharmacologie , Troubles liés à la cocaïne/métabolisme , Dronabinol/pharmacologie , Éthanol/pharmacologie , Hallucinogènes/pharmacologie , Immunohistochimie , Mâle , Morphine/pharmacologie , Stupéfiants/pharmacologie , Protéines proto-oncogènes c-fos/génétique , Rats , Rat Sprague-Dawley , Autoadministration
7.
Br J Pharmacol ; 151(8): 1324-33, 2007 Aug.
Article de Anglais | MEDLINE | ID: mdl-17572699

RÉSUMÉ

BACKGROUND AND PURPOSE: Chronic morphine administration produces tolerance in vivo and attenuation of mu opioid receptor (MOR)-mediated G-protein activation measured in vitro, but the relationship between these adaptations is not clear. The present study examined MOR-mediated G-protein activation in the CNS of mice with different levels of morphine tolerance. EXPERIMENTAL APPROACH: Mice were implanted with morphine pellets, with or without supplemental morphine injections, to induce differing levels of tolerance as determined by a range of MOR-mediated behaviours. MOR function was measured using agonist-stimulated [(35)S]guanylyl-5'-O-(gamma-thio)-triphosphate ([(35)S]GTPgammaS) and receptor binding throughout the CNS. KEY RESULTS: Morphine pellet implantation produced 6-12-fold tolerance in antinociceptive assays, hypothermia and Straub tail, as measured by the ratio of morphine ED(50) values between morphine-treated and control groups. Pellet implantation plus supplemental injections produced 25-50-fold tolerance in these tests. In morphine pellet-implanted mice, MOR-stimulated [(35)S]GTPgammaS binding was significantly reduced only in the nucleus tractus solitarius (NTS) and spinal cord dorsal horn in tissue sections from morphine pellet-implanted mice. In contrast, MOR-stimulated [(35)S]GTPgammaS binding was significantly decreased in most regions examined in morphine pellet+morphine injected mice, including nucleus accumbens, caudate-putamen, periaqueductal gray, parabrachial nucleus, NTS and spinal cord. CONCLUSIONS AND IMPLICATIONS: Tolerance and the regional pattern of apparent MOR desensitization were influenced positively by the level of morphine exposure. These results indicate that desensitization of MOR-mediated G-protein activity is more regionally widespread upon induction of high levels of tolerance, suggesting that this response contributes more to high than low levels of tolerance to CNS-mediated effects of morphine.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Tolérance aux médicaments , Protéines G/métabolisme , Morphine/pharmacologie , Récepteur mu/métabolisme , Analgésiques morphiniques/administration et posologie , Animaux , Sites de fixation , Système nerveux central , Relation dose-effet des médicaments , Guanosine 5'-O-(3-thiotriphosphate) , Hypothermie/induit chimiquement , Mâle , Souris , Morphine/administration et posologie , Mesure de la douleur , Cellules de la corne dorsale , Noyau du tractus solitaire , Queue/effets des médicaments et des substances chimiques
8.
Biochem Pharmacol ; 62(4): 447-55, 2001 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-11448454

RÉSUMÉ

The efficacy of heroin metabolites for the stimulation of mu opioid receptor-mediated G-protein activation was investigated using agonist-stimulated [(35)S]guanosine-5'-O-(gamma-thio)-triphosphate binding. In rat thalamic membranes, heroin and its primary metabolite, 6-monoacetylmorphine (6-MAM), were more efficacious than morphine or morphine-6-beta D-glucuronide. This increased efficacy was not due to increased action of heroin and 6-MAM at delta receptors, as determined by competitive antagonism by naloxone, lack of antagonism by naltrindole, and competitive partial antagonism with morphine. In agreement with this interpretation, the same relative efficacy profile of heroin and its metabolites was observed at the cloned human mu opioid receptor expressed in C6 glioma cells. Moreover, these efficacy differences were GDP-dependent in a manner consistent with accepted mechanisms of receptor-mediated G-protein activation. The activity of heroin was attributed to in vitro deacetylation to 6-MAM, as confirmed by HPLC analysis. These results indicate that the heroin metabolite 6-MAM possesses higher efficacy than other heroin metabolites at mu opioid receptors, which may contribute to the higher efficacy of heroin compared with morphine in certain behavioral paradigms in vivo.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Protéines G/métabolisme , Héroïne/pharmacologie , Dérivés de la morphine/pharmacologie , Récepteur mu/métabolisme , Analgésiques morphiniques/métabolisme , Animaux , Relation dose-effet des médicaments , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Héroïne/métabolisme , Mâle , Morphine/pharmacologie , Rats , Rat Sprague-Dawley , Récepteur mu/agonistes , Radio-isotopes du soufre , Cellules cancéreuses en culture
9.
J Neurochem ; 77(4): 1048-57, 2001 May.
Article de Anglais | MEDLINE | ID: mdl-11359870

RÉSUMÉ

Heterozygous CB1 receptor knockout mice were used to examine the effect of reduced CB1 receptor density on G-protein activation in membranes prepared from four brain regions: cerebellum, hippocampus, striatum/globus pallidus (striatum/GP) and cingulate cortex. Results showed that CB1 receptor levels were approximately 50% lower in heterozygous mice in all regions examined. However, maximal stimulation of [(35)S]guanosine-5'-(gamma-O-thio) triphosphate ([(35)S]GTPgammaS) binding by the high efficacy agonist WIN 55,212-2 was reduced by only 20-25% in most brain regions, with the exception of striatum/GP where the decrease in stimulation was as predicted (approximately 50%). Furthermore, although the efficacies of the cannabinoid partial agonists, methanandamide and (9)-tetrahydrocannabinol, were similarly lower in heterozygous mice, their relative efficacies compared with WIN 55,212-2 were generally unchanged. Saturation analysis of net WIN 55,212-2-stimulated [(35)S]GTPgammaS binding showed that decreased stimulation by WIN 55,212-2 in striatum/GP of heterozygous mice was caused by a decrease in the apparent affinity of net-stimulated [(35)S]GTPgammaS binding. The apparent maximal number of binding sites (B(max)) values of net WIN 55,212-2-stimulated [(35)S]GTPgammaS binding were unchanged in cerebellum and striatum/GP of heterozygous mice, but decreased in cingulate cortex, with a similar trend in hippocampus. Moreover, in every region except cingulate cortex, the maximal number of net-stimulated [(35)S]GTPgammaS binding sites per receptor was significantly increased in heterozygous mice. These results indicate region-dependent increases in the apparent efficiency of CB1 receptor-mediated G-protein activation in heterozygous CB1 knockout mice.


Sujet(s)
Encéphale/métabolisme , Cannabinoïdes/pharmacologie , Protéines G/métabolisme , Récepteurs des médicaments/physiologie , Animaux , Acides arachidoniques/pharmacologie , Benzoxazines , Sites de fixation , Encéphale/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Cervelet/métabolisme , Corps strié/métabolisme , Croisements génétiques , Dronabinol/pharmacologie , Femelle , Globus pallidus/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Gyrus du cingulum/métabolisme , Hétérozygote , Hippocampe/métabolisme , Cinétique , Souris , Souris de lignée C57BL , Souris knockout , Morpholines/pharmacologie , Naphtalènes/pharmacologie , Spécificité d'organe , Récepteurs de cannabinoïdes , Récepteurs des médicaments/déficit , Récepteurs des médicaments/génétique
10.
Eur J Pharmacol ; 414(2-3): 135-43, 2001 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-11239913

RÉSUMÉ

N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride (SR141716A), a cannabinoid CB(1) receptor antagonist, has inverse agonist effects in cannabinoid CB(1) receptor-expressing cell lines, brain and peripheral organs. These studies characterized SR141716A-inhibited G-protein activity by measuring [35S]GTPgammaS binding. Maximal inhibition of basal [35S]GTPgammaS binding in cerebellar membranes was 50%. The EC(50) value for inhibition of [35S]GTPgammaS binding was 4.4 microM, whereas the K(e) for inhibition of R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone mesylate (WIN 55,212-2)-stimulated [35S]GTPgammaS binding was 0.6 nM. [35S]GTPgammaS autoradiography was used to examine the regional specificity of SR141716A inhibition. SR141716A inhibited basal [35S]GTPgammaS binding in all regions examined, with inhibition ranging from approximately 20% in caudate-putamen to 40% in hippocampus. These studies demonstrate that SR141716A is a competitive antagonist at nanomolar concentrations, whereas it inhibits basal receptor-mediated G-protein activity at micromolar concentrations. These data suggest that the apparent inverse agonist effect is either not cannabinoid CB(1) receptor-specific or that SR141716A is binding to different sites on the cannabinoid CB(1) receptor to produce inverse agonist versus competitive antagonist effects.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Guanosine 5'-O-(3-thiotriphosphate)/antagonistes et inhibiteurs , Pipéridines/pharmacologie , Pyrazoles/pharmacologie , Récepteurs des médicaments/antagonistes et inhibiteurs , Animaux , Benzoxazines , Encéphale/métabolisme , Inhibiteurs des canaux calciques/pharmacologie , Camphanes/pharmacologie , Relation dose-effet des médicaments , Protéines G/antagonistes et inhibiteurs , Protéines G/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Mâle , Morpholines/pharmacologie , Naphtalènes/pharmacologie , Rats , Rat Sprague-Dawley , Récepteurs de cannabinoïdes , Récepteurs des médicaments/métabolisme , Rimonabant , Chlorure de sodium/pharmacologie
11.
Br J Pharmacol ; 130(5): 987-96, 2000 Jul.
Article de Anglais | MEDLINE | ID: mdl-10882382

RÉSUMÉ

1. Sodium ions inhibit spontaneous G(i)/G(o)-coupled receptor activity and promote agonist-induced responses in vitro. The effects of sodium on the relative efficacy of opioid agonists for G-protein activation was measured by guanosine-5'-O-(gamma-(35)S)-triphosphate ([(35)S]-GTPgammaS) binding in membranes from two mu-opioid receptor-containing systems: CHO cells stably transfected with mouse mureceptors (mMOR-CHO cells) and rat thalamus. 2. NaCl inhibited basal [(35)S]-GTPgammaS binding in both systems, and this effect was partially mimicked by KCl. In mMOR-CHO membranes, net [(35)S]-GTPgammaS binding stimulated by partial but not full agonists was inhibited by NaCl with a potency that was inversely proportional to agonist efficacy. Monovalent cations were required for agonist-stimulated [(35)S]-GTPgammaS binding in this system, and increasing NaCl concentrations magnified relative efficacy differences among agonists. 3. In thalamic membranes, which contain a lower receptor:G-protein ratio than mMOR-CHO cells, similar monovalent cation effects were observed, with two exceptions: (1) [(35)S]-GTPgammaS binding stimulated by both full and partial agonists was inhibited by NaCl; and (2) monovalent cations were not required to observe agonist-stimulated [(35)S]-GTPgammaS binding. 4. Basal [(35)S]-GTPgammaS binding stimulated by the absence of monovalent cations resembled that of agonist-stimulated binding and was blocked by pretreatment of mMOR-CHO cells with pertussis toxin. 5. These results indicate that sodium inhibits spontaneous and agonist-occupied mu receptor-mediated G-protein activation in a manner inversely proportional to the efficacy of the agonist, and that spontaneous mu receptor activity and the relative efficacy of partial agonists acting at these receptors are both increased by increases in the stoichiometric ratio of receptors:G-proteins.


Sujet(s)
Protéines G/physiologie , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Récepteur mu/effets des médicaments et des substances chimiques , Chlorure de sodium/pharmacologie , Thalamus/effets des médicaments et des substances chimiques , Animaux , Cellules CHO , Cricetinae , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/pharmacologie , Mâle , Souris , Morphine/pharmacologie , Toxine pertussique , Rats , Rat Sprague-Dawley , Récepteur mu/physiologie , Thalamus/métabolisme , Transfection , Facteurs de virulence des Bordetella/pharmacologie
12.
J Neurosci ; 20(12): 4555-62, 2000 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-10844025

RÉSUMÉ

In previous studies from our laboratory, chronic noncontingent morphine administration decreased mu opioid receptor-activated G-proteins in specific brainstem nuclei. In the present study, mu opioid receptor binding and receptor-activated G-proteins were examined after chronic heroin self-administration. Rats were trained to self-administer intravenous heroin for up to 39 d, achieving heroin intake up to 366 mg. kg(-1). d(-1). mu opioid-stimulated [(35)S]GTPgammaS and [(3)H]naloxone autoradiography were performed in adjacent brain sections. Agonist-stimulated [(35)S]GTPgammaS autoradiography also examined other G-protein-coupled receptors, including delta opioid, ORL-1, GABA(B), adenosine A(1), cannabinoid, and 5-HT(1A). In brains from heroin self-administering rats, decreased mu opioid-stimulated [(35)S]GTPgammaS binding was observed in periaqueductal gray, locus coeruleus, lateral parabrachial nucleus, and commissural nucleus tractus solitarius, as previously observed in chronic morphine-treated animals. In addition, decreased mu opioid-stimulated [(35)S]GTPgammaS binding was found in thalamus and amygdala after heroin self-administration. Despite this decrease in mu-activated G-proteins, [(3)H]naloxone binding demonstrated increased mu opioid receptor binding in several brain regions after heroin self-administration, and there was a significant decrease in mu receptor G-protein efficiency as expressed as a ratio between agonist-activated G-proteins and mu receptor binding. No effects on agonist-stimulated [(35)S]GTPgammaS binding were found for any other receptor examined. The effect of chronic heroin self-administration to decrease mu-stimulated [(35)S]GTPgammaS binding varied between regions and was highest in brainstem and lowest in the cortex and striatum. These results not only provide potential neuronal mechanisms that may contribute to opioid tolerance and dependence, but also may explain why various chronic effects of opioids develop to different degrees.


Sujet(s)
Encéphale/métabolisme , Protéines G/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/pharmacocinétique , Dépendance à l'héroïne/physiopathologie , Héroïne/administration et posologie , Récepteur mu/effets des médicaments et des substances chimiques , Animaux , Autoradiographie , Encéphale/effets des médicaments et des substances chimiques , Protéines G/effets des médicaments et des substances chimiques , Héroïne/pharmacologie , Cinétique , Mâle , Naloxone/pharmacocinétique , Spécificité d'organe , Rats , Rats de lignée F344 , Récepteur mu/métabolisme , Autoadministration , Radio-isotopes du soufre , Tritium
13.
Eur J Pharmacol ; 389(2-3): 147-53, 2000 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-10688978

RÉSUMÉ

5-Hydroxytryptamine(1A) (5-HT(1A)) receptors, which activate inhibitory G-proteins, are implicated in psychiatric disorders including anxiety and depression. Studies suggest that chronic 5-HT(1A) receptor agonist administration alters 5-HT(1A) receptor function, but the effect of chronic treatment on 5-HT(1A) receptor-activated G-proteins is unclear. In this study, agonist-stimulated [35S]guanylyl-5'-O-(gamma-thio)-triphosphate (GTPgammaS) binding was examined following chronic administration of buspirone. Brains were processed for [35S]GTPgammaS autoradiography using R(+)-8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) for 5-HT(1A) receptors or baclofen for GABA(B) receptors. Net 8-OH-DPAT-stimulated [35S]GTPgammaS binding was decreased by 25-30% in the septum and dorsal raphe nucleus of buspirone-treated animals. No significant changes in 8-OH-DPAT-stimulated [35S]GTPgammaS binding were found in the prefrontal, entorhinal or cingulate cortices or hippocampus in buspirone-treated rats. GABA(B) receptor-stimulated [35S]GTPgammaS binding was increased by 25% in the hippocampus, with no significant changes in any other region examined. These results demonstrate region-specific alterations in 5-HT(1A) and GABA(B) receptor-activated G-proteins following chronic buspirone treatment, which may contribute to the clinical effects of this drug.


Sujet(s)
Chimie du cerveau/effets des médicaments et des substances chimiques , Buspirone/pharmacologie , Protéines G/analyse , Récepteurs sérotoninergiques/physiologie , 7-Dipropylamino-5,6,7,8-tétrahydro-1-naphtol/pharmacologie , Animaux , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Mâle , Rats , Rat Sprague-Dawley , Récepteurs GABA-B/physiologie , Récepteurs sérotoninergiques/effets des médicaments et des substances chimiques , Récepteurs de la sérotonine de type 5-HT1
14.
Biochem Pharmacol ; 59(11): 1395-401, 2000 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-10751548

RÉSUMÉ

This study investigated the relationship between mu receptor binding and mu agonist activation of G-proteins in the rat brain. To directly compare agonist potencies in receptor binding (K(i) values) and G-protein activation (K(s) values), both agonist-stimulated [(35)S]guanosine-5'-O-(gamma-thio)-triphosphate ([(35)S]GTPgammaS) and [(3)H]naloxone binding assays were conducted under identical conditions, using the full mu agonist [d-Ala(2), N-Me(4), Gly(5)-ol]-enkephalin (DAMGO). DAMGO exhibited biphasic competition of [(3)H]naloxone binding and stimulation of [(35)S]GTPgammaS binding in most regions. Whereas the high-affinity component represented a large percentage (50-80%) of total receptor sites, the high-affinity component of DAMGO-stimulated [(35)S]GTPgammaS binding was much lower, <30% of the total, and in most regions significant stimulation of [(35)S]GTPgammaS binding did not occur until the high-affinity binding sites were completely occupied. Moreover, the low-affinity potencies for DAMGO in receptor binding and G-protein activation were the same across different regions. Receptor-transducer amplification factors were calculated by the ratio of the apparent B(max) of net agonist-stimulated [(35)S]GTPgammaS binding to the B(max) of receptor binding. Amplification factors for the nine regions examined were relatively high and varied significantly across regions, from a ratio of 8 in the thalamus to 38 in the cortex, suggesting that the efficiency of mu opioid receptor coupling to G-proteins varies across brain regions.


Sujet(s)
Encéphale/métabolisme , Protéines G/métabolisme , Récepteur mu/métabolisme , Analgésiques morphiniques/pharmacologie , Animaux , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/pharmacologie , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Techniques in vitro , Mâle , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Rats , Rat Sprague-Dawley , Radio-isotopes du soufre , Tritium
15.
J Biol Chem ; 273(27): 16865-73, 1998 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-9642247

RÉSUMÉ

The relationship between GDP and cannabinoid-stimulated [35S]guanosine-5'-O-(3-thiotriphosphate) ([35S]GTPgammaS) binding was investigated in rat cerebellar membranes. Kinetic analyses showed that [35S]GTPgammaS binding reached steady-state levels and that the association rate was increased by the agonist WIN 55212-2 proportional to the concentration of GDP. Dissociation of [35S]GTPgammaS occurred with two rates (t1/2 = 7 and 170 min), and WIN 55212-2 increased the proportion of sites exhibiting the faster rate. Without GDP, [35S]GTPgammaS bound to membranes with high and low affinity, and WIN 55212-2 had no effect. With 30 microM GDP, [35S]GTPgammaS bound to low and intermediate affinity sites, and WIN 55212-2 induced high affinity [35S]GTPgammaS binding without affecting low affinity sites. GDP competed for high affinity [35S]GTPgammaS binding with high and intermediate affinity in the absence of WIN 55212-2 and with high and low affinity in the presence of WIN 55212-2. Cannabinoid ligands displayed differential abilities to maximally stimulate [35S]GTPgammaS binding in the presence of GDP. Efficacy differences among ligands increased with increasing GDP concentrations. GDP competition curves revealed that agonists induced low affinity GDP Ki values that were proportional to agonist Emax values, indicating that agonist efficacy is determined by displacement of GDP from G-proteins.


Sujet(s)
Cervelet/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Guanosine diphosphate/métabolisme , Morpholines/pharmacologie , Naphtalènes/pharmacologie , Récepteurs des médicaments/agonistes , Animaux , Benzoxazines , Membrane cellulaire/métabolisme , Protéines G/métabolisme , Période , Cinétique , Mâle , Liaison aux protéines , Rats , Rat Sprague-Dawley , Récepteurs de cannabinoïdes , Radio-isotopes du soufre
16.
Brain Res ; 791(1-2): 341-6, 1998 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-9593984

RÉSUMÉ

mu-Opioid receptors play a critical role in the regulation of the female reproductive cycle, and estrogen modulates the coupling of mu-opioid receptors to a potassium channel in the basal hypothalamus (BH) of the female guinea pig. Therefore, we ascertained the distribution of mu-opioid receptors in the BH with autoradiography using the mu-opioid selective agonist [3H]DAMGO. In addition, we investigated the effects of estrogen on DAMGO- or the GABAB receptor agonist baclofen-stimulated [35S]GTPgammaS binding in the BH. Based on the high density of mu-opioid receptors, but the lack of effects of estrogen on [35S]GTPgammaS binding, we conclude that mu-opioid receptor interaction with its G-protein is not the target of estrogen's actions.


Sujet(s)
Enképhalines/pharmacologie , Oestrogènes/pharmacologie , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Hypothalamus/effets des médicaments et des substances chimiques , Récepteur mu/agonistes , Animaux , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline , Enképhalines/métabolisme , Femelle , Cochons d'Inde , Hypothalamus/métabolisme , Système neuroendocrinien/effets des médicaments et des substances chimiques , Canaux potassiques/effets des médicaments et des substances chimiques , Dosage par compétition , Activation chimique , Radio-isotopes du soufre
17.
J Pharmacol Exp Ther ; 285(2): 496-505, 1998 May.
Article de Anglais | MEDLINE | ID: mdl-9580589

RÉSUMÉ

This study examined the signal transduction correlates of mu opioid agonist efficacy in two systems: mu receptor-transfected mMOR-CHO cell and rat thalamic membranes. The potency and maximal stimulation of [35S]GTP gamma S binding by various agonists was measured in the presence of excess GDP and compared with receptor binding affinity under identical assay conditions. Results showed that the relative maximal stimulation produced by these agonists was greater in mMOR-CHO cell than in rat thalamic membranes; some drugs that were full agonists in mMOR-CHO cells were partial agonists in the thalamus, and some partial agonists in the transfected cells were full antagonists in the thalamus. Furthermore, there was receptor reserve for G-protein activation by some agonists in mMOR-CHO cell membranes, but no receptor reserve was detected in rat thalamic membranes. Saturation analysis of agonist-stimulated [35S]GTP gamma S binding revealed that full agonists produced both a higher Bmax and apparent affinity of [35S]GTP gamma S binding than partial agonists. Correlation of the Bmax and KD of agonist-stimulated [35S]GTP gamma S binding with agonist intrinsic efficacy revealed only a moderate correlation with either parameter alone, but a highly significant correlation (r > 0.9) with a combination of the two parameters (Bmax/KD). These results suggest that the intrinsic efficacy of agonists at G-protein-coupled receptors is determined primarily by the ability of the agonist-occupied receptor to promote high-affinity GTP binding to the G-protein and to catalytically activate a maximal number G-proteins.


Sujet(s)
Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Récepteur mu/agonistes , Récepteur mu/physiologie , Transduction du signal , Thalamus/métabolisme , Animaux , Cellules CHO , Cricetinae , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline , Enképhalines/pharmacologie , Protéines G/physiologie , Souris , Rats , Récepteur mu/génétique
18.
J Neurochem ; 70(4): 1567-76, 1998 Apr.
Article de Anglais | MEDLINE | ID: mdl-9523574

RÉSUMÉ

Endomorphin-1 is a peptide whose binding selectivity suggests a role as an endogenous ligand at mu-opioid receptors. In the present study, the effect of endomorphin-1 on mu receptor-coupled G proteins was compared with that of the mu agonist DAMGO by using agonist-stimulated [35S]GTPgammaS binding in rat brain. [35S]GTPgammaS autoradiography revealed a similar localization of endomorphin-1- and DAMGO-stimulated [35S]GTPgammaS binding in areas including thalamus, caudate-putamen, amygdala, periaqueductal gray, parabrachial nucleus, and nucleus tractus solitarius. Naloxone blocked endomorphin-1-stimulated labeling in all regions examined. Although the distribution of endomorphin-1-stimulated [35S]GTPgammaS binding resembled that of DAMGO, the magnitude of endomorphin-1-stimulated binding was significantly lower than that produced by DAMGO. Concentration-effect curves of endomorphin-1 and DAMGO in thalamic membranes confirmed that endomorphin-1 produced only 70% of DAMGO-stimulated [35S]GTPgammaS binding. Differences in maximal stimulation of [35S]GTPgammaS binding between DAMGO and endomorphin-1 were magnified by increasing GDP concentrations, and saturation analysis of net endomorphin-1-stimulated [35S]GTPgammaS binding revealed a lower apparent Bmax value than that obtained with DAMGO. Endomorphin-1 also partially antagonized DAMGO stimulation of [35S]GTPgammaS binding. These results demonstrate that endomorphin-1 is a partial agonist for G protein activation at the mu-opioid receptor in brain.


Sujet(s)
Encéphale/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Oligopeptides/pharmacologie , Récepteur mu/agonistes , Animaux , Autoradiographie , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline , Enképhalines/pharmacologie , Guanosine diphosphate/pharmacologie , Mâle , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Rats , Rat Sprague-Dawley , Radio-isotopes du soufre
19.
J Recept Signal Transduct Res ; 18(1): 25-49, 1998 Jan.
Article de Anglais | MEDLINE | ID: mdl-9493566

RÉSUMÉ

Gi/Go proteins are uncoupled from receptors by ADP-ribosylation with pertussis toxin (PTX). However, PTX treatment of delta opioid receptor-containing NG108-15 cells reduces, but does not eliminate, opioid inhibition of adenylyl cyclase. The present study explored potential mechanisms of this residual inhibition. Overnight treatment of NG108-15 cells with 100 ng/ml PTX eliminated both PTX-catalyzed [adenylyl-32P]NAD+-labeling of G proteins and agonist stimulation of low Km GTPase in membranes. Although PTX-treatment decreased the maximal opioid inhibition of adenylyl cyclase by 50-65%, the inhibition that remained was concentration-dependent and antagonist-reversible. This inhibition persisted in the absence of GTP (even though opioid inhibition of adenylyl cyclase in untreated membranes was GTP-dependent), but was eliminated by hydrolysis-resistant guanine nucleotide analogs, indicating that G-proteins were still involved in the coupling mechanism. However, assays of agonist-stimulated [35S]GTPgammaS binding in the presence of excess GDP indicated that PTX pretreatment eliminated stimulation of guanine nucleotide exchange by opioid agonists. These results suggest that in membranes from PTX-treated NG108-15 cells, a subpopulation of G proteins may transduce an inhibitory signal from agonist-bound opioid receptors without involvement of guanine nucleotide exchange.


Sujet(s)
Toxine d'adénylate cyclase , Inhibiteurs des adénylate cyclases , Stupéfiants/pharmacologie , Toxine pertussique , Facteurs de virulence des Bordetella/pharmacologie , Adénosine diphosphate ribose/métabolisme , Animaux , Catalyse , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/enzymologie , Activation enzymatique , dGTPases/métabolisme , Protéines G/effets des médicaments et des substances chimiques , Protéines G/métabolisme , Nucléotides guanyliques/métabolisme , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Hydrolyse , Cinétique , Souris , Naltrexone/analogues et dérivés , Naltrexone/pharmacologie , Stupéfiants/agonistes , Neuroblastome , Cellules cancéreuses en culture
20.
Synapse ; 27(3): 177-82, 1997 Nov.
Article de Anglais | MEDLINE | ID: mdl-9329153

RÉSUMÉ

Intracerebroventricular administration of beta-funaltrexamine (beta-FNA) reduces the density of mu opioid receptors as measured by in situ autoradiography by 40-50% throughout the brain, with little regional variation [Martin et al. (1993) J. Pharmacol. Exp. Ther. 267:506-514] Recently an assay has been developed to study opioid stimulation of [35S]GTP-gamma-S binding autoradiographically in situ using slide-mounted brain sections [Sim et al. (1995) Proc. Natl. Acad. Sci. U.S.A. 92:7242-7246]. The present study was undertaken to determine the effect of mu opioid receptor alkylation on G protein activation by the mu opioid agonist DAMGO. Animals were injected intracerebroventricularly with 40 nmol of beta-FNA or saline and sacrificed 24 hours later. DAMGO stimulated [35S]GTP-gamma-S binding with an anatomical specificity consistent with the localization of mu opioid receptors. The percent stimulation by DAMGO ranged from approximately 50 to 100% in the regions studied. beta-FNA significantly decreased G protein activation by DAMGO in regions that are consistent with its reported long-lasting and insurmountable antagonism of the antinociceptive (medial thalamus, central gray) and reinforcing (nucleus accumbens) effects of mu opioid agonists [Adams et al. (1990) J. Pharmacol. Exp. Ther. 255:1027-1032; Martin et al. (1995) J. Pharmacol. Exp. Ther. 272:1135-1140]. However, the effects of beta-FNA were not equal in all brain regions. This may indicate regional differences in the coupling efficiency of mu opioid receptors with G proteins, or in the effects of beta-FNA on mu opioid receptor binding or on mu opioid receptor-stimulated G protein activity.


Sujet(s)
Encéphale/métabolisme , Enképhalines/pharmacologie , Guanosine 5'-O-(3-thiotriphosphate)/métabolisme , Naltrexone/analogues et dérivés , Antagonistes narcotiques/pharmacologie , Récepteur mu/agonistes , Récepteur mu/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline , Injections ventriculaires , Mâle , Naltrexone/administration et posologie , Naltrexone/pharmacologie , Antagonistes narcotiques/administration et posologie , Rats , Rats de lignée F344 , Radio-isotopes du soufre
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE