Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 396(2-3): 67-75, 2000 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-10822058

RESUMO

The collision-coupling model for receptor-G-protein interaction predicts that the rate of G-protein activation is dependent on receptor density, but not G-protein levels. C6 cells expressing mu- or delta-opioid receptors, or SH-SY5Y cells, were treated with beta-funaltrexamine (mu) or naltrindole-5'-isothiocyanate (delta) to decrease receptor number. The time course of full or partial agonist-stimulated ¿35SGTPgammaS binding did not vary in C6 cell membranes containing <1-25 pmol/mg mu-opioid receptor, or 1. 4-4.3 pmol/mg delta-opioid receptor, or in SHSY5Y cells containing 0. 16-0.39 pmol/mg receptor. The association of ¿35SGTPgammaS binding was faster in membranes from C6mu cells than from C6delta cells. A 10-fold reduction in functional G-protein, following pertussis toxin treatment, lowered the maximal level of ¿35SGTPgammaS binding but not the association rate. These data indicate a compartmentalization of opioid receptors and G protein at the cell membrane.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Proteínas de Ligação ao GTP/análise , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Receptores Opioides delta/análise , Receptores Opioides mu/análise , Células Tumorais Cultivadas
2.
J Neurochem ; 73(1): 289-300, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10386982

RESUMO

The influence of membrane microviscosity on mu-opioid agonist and antagonist binding, as well as agonist efficacy, was examined in membranes prepared from SH-SY5Y cells and from a C6 glioma cell line stably expressing the rat mu-opioid receptor (C6mu). Addition of cholesteryl hemisuccinate (CHS) to cell membranes increased membrane microviscosity and reduced the inhibitory effect of sodium and guanine nucleotides on the affinity of the full agonists sufentanil and [D-Ala2,N-MePhe4,Gly-ol5]enkephalin (DAMGO) for the mu-opioid receptor. Binding of the antagonists [3H]naltrexone and [3H]diprenorphine and the partial agonist nalbuphine was unaffected by CHS. The effect of CHS on agonist binding was reversed by subsequent addition of cis-vaccenic acid, suggesting that the effect of CHS is the result of increased membrane microviscosity and not a specific sterol-receptor interaction. CHS addition increased the potency of DAMGO to stimulate guanosine-5'-O-(3-[35S]thio)triphosphate binding by fourfold, whereas the potency of nalbuphine was unaffected. However, nalbuphine efficacy relative to that of the full agonist DAMGO was strongly increased in CHS-treated membranes compared with that in control membranes. Membrane rigidification also resulted in an increased efficacy for the partial agonists meperidine, profadol, and butorphanol relative to that of DAMGO as measured by agonist-stimulated GTPase activity in control and CHS-modified membranes. These findings support a regulatory role for membrane microviscosity in receptor-mediated G protein activation.


Assuntos
Membrana Celular/fisiologia , Fluidez de Membrana/fisiologia , Entorpecentes/metabolismo , Conformação Proteica , Receptores Opioides mu/química , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Ésteres do Colesterol/farmacologia , Diprenorfina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Glioma , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Nalbufina/metabolismo , Naltrexona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Neuroblastoma , Ratos , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Cloreto de Sódio/farmacologia , Sufentanil/metabolismo , Células Tumorais Cultivadas , Viscosidade
3.
J Pharmacol Exp Ther ; 288(2): 827-33, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9918595

RESUMO

A C6 glioma cell line stably transfected with the human kappa opioid receptor (kappaOR) was used to characterize receptor binding and G protein activation via the kappaOR by a comprehensive series of opioid ligands. The ligand-binding affinity for [3H]5alpha,7alpha, 8beta(-)-N-methyl-N-(7-Cl-pyrrolidinyl)-1-oxaspiro(4, 5)dec-8-yl)benzene acetamide (U69593) was similar to that observed in monkey brain membranes and was 10-fold lower in the presence of sodium and GDP. Both peptide and nonpeptide agonists maximally stimulated [35S]GTPgammaS binding. The stimulation of [35S]GTPgammaS binding was blocked by pretreatment of cells with pertussis toxin. Partial stimulation of [35S]GTPgammaS binding via the kappaOR was observed for several ligands that are antagonists at the mu opioid receptor, suggesting an additional mechanism of drug action. The ability of isomers of tifluadom and levallorphan to stimulate [35S]GTPgammaS binding indicates that the chiral carbon of levallorphan, a benzomorphan derivative, imparts a greater degree of stereoselectivity than does the chiral carbon in the benzodiazepine derivative tifluadom. In addition, (-)tifluadom, the less potent isomer of tifluadom, which is also a gamma-aminobutyric acidA receptor agonist, stimulated [35S]GTPgammaS binding. In contrast, d-pentazocine, (+)SKF10047, (+)cyclazocine, and d-ethylketocyclazocine displayed no agonist activity. kappaOR-selective antagonist norbinaltorphimine competitively inhibited the stimulation of [35S]GTPgammaS binding by the active isomers of ethylketocyclazocine, cyclazocine, and nalorphine to the same degree, indicating that all three ligands are eliciting an effect via the kappaOR. The results suggest that these cells express a homogeneous population of kappaOR, and that their [35S]GTPgammaS-binding properties make them an excellent means to assess kappaOR efficacy.


Assuntos
Analgésicos Opioides/farmacologia , Benzenoacetamidas , Glioma/ultraestrutura , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/metabolismo , Analgésicos Opioides/metabolismo , Animais , Glioma/genética , Glioma/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Ligantes , Naloxona/metabolismo , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Naltrexona/farmacologia , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Ratos , Receptores Opioides kappa/genética , Estereoisomerismo , Transfecção , Trítio , Células Tumorais Cultivadas
4.
J Pharmacol Exp Ther ; 287(2): 625-32, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9808689

RESUMO

Chronic treatment of C6 glioma cells stably expressing the rat delta opioid receptor (C6delta) with full agonists resulted in receptor down-regulation. Chronic [D-Ser2,L-Leu5]enkephalyl-Thr treatment caused a decrease in cell surface as well as a decrease in agonist-stimulated [35S]guanosine-5'-O-(3-thio)triphosphate binding. Treatment with full agonists for 12 hr resulted in a 90% decrease in receptor number that was paralleled by a decrease in the ability of agonist to stimulate [35S]guanosine-5'-O-(3-thio)triphosphate binding and inhibit forskolin-stimulated adenylyl cyclase. Of the remaining receptors, a smaller fraction of receptors (41 +/- 4 vs. 56 +/- 4% in control) exhibited high affinity for agonist as compared to receptors in control membranes. Elimination of functional guanosine triphosphate binding protein (G protein) by Pertussis toxin pretreatment did not alter the ability of agonist to down regulate receptor. We hypothesized that agonist affinity (not efficacy) would be a predictor of an agonist's ability to down-regulate receptor. However, we found that only full agonists were able to down-regulate receptor number, G protein activation and adenylyl cyclase inhibition. Chronic exposure to partial agonist 7-spiroindinooxymorphone, which has a very high affinity for the receptor, as well as morphine, did not cause receptor down-regulation. Taken together, these results suggest that full agonists alter receptor conformation such that the altered conformation is recognized by G protein as well as proteins involved in receptor down-regulation. In addition, down-regulation is independent of agonist-mediated G protein activation and subsequent down-stream signaling.


Assuntos
Regulação para Baixo , Proteínas de Ligação ao GTP/metabolismo , Receptores Opioides/metabolismo , Inibidores de Adenilil Ciclases , Animais , Colforsina/farmacologia , AMP Cíclico/metabolismo , D-Penicilina (2,5)-Encefalina , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacologia , Encefalinas/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ratos , Receptores Opioides/agonistas , Células Tumorais Cultivadas
6.
J Pharmacol Exp Ther ; 286(1): 282-8, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9655870

RESUMO

The ability of endogenous opioids to activate G proteins was measured in membranes from C6 rat glioma cells stably expressing a cloned rat mu receptor. Peptides representing each of the three known families of endogenous opioids (enkephalins, endorphins and dynorphins) were studied, as well as two recently discovered endogenous opioids, endomorphin-1 and -2, which are thought to represent a fourth family of endogenous opioid peptides. Stimulation of guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTP gamma S) binding to membranes was used as a measure of G protein activation. It was possible to differentiate high efficacy compounds such as Tyr-D-Ala-Gly-(Me)Phe-Gly-ol from lower-efficacy agonists such as morphine or meperidine. Met- and leu-enkephalin, beta endorphin and dynorphin A were all found to have high efficacy at the mu receptor, as were the peptide fragments beta endorphin-1(1-27) and dynorphin A-(1-13). Endomorphin-1 and -2 were found to be partial agonists, capable of both stimulating [35S]GTP gamma S binding and antagonizing the stimulation produced by the higher-efficacy agonist Tyr-D-Ala-Gly-(Me)Phe-Gly-ol. Binding affinities for the opioid agonists at the cloned mu receptor were measured by the displacement of radiolabeled antagonist. It was found that the Ki values closely matched the EC50 values for [35S]GTP gamma S binding stimulation, indicating that a large receptor reserve does not exist for the complete activation of G proteins in this system.


Assuntos
Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Peptídeos Opioides/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Ratos , Receptores Opioides mu/fisiologia , Proteínas Recombinantes/efeitos dos fármacos , Células Tumorais Cultivadas , beta-Endorfina/farmacologia
7.
Biochem Pharmacol ; 54(12): 1315-22, 1997 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9393674

RESUMO

A series of opioid ligands utilizing the 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) fluorophores 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene++ +-3-propionic acid or 4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza- s-indacene-3-propionic acid were synthesized and characterized for their ability to act as a suitable fluorescent label for the mu opioid receptor. All compounds displaced the mu opioid receptor binding of [3H]Tyr-D-Ala-Gly-(Me)Phe-Gly-ol in monkey brain membranes with high affinity. The binding of fluorescent ligands to delta and kappa receptors was highly variable. 5,7-Dimethyl-BODIPY naltrexamine, "6-BNX," displayed subnanomolar affinities for the mu and kappa opioid receptors (Ki 0.07 and 0.43 nM, respectively) and nanomolar affinity at the delta (Ki 1.4 nM) receptor. Using fluorescence spectroscopy, the binding of 6-BNX in membranes from C6 glioma cells transfected with the cloned mu opioid receptor was investigated. In these membranes containing a high receptor density (10-80 pmol/mg protein), 6-BNX labeling was saturable, mu opioid specific, stereoselective (as determined with the isomers dextrorphan and levorphanol), and more than 90% specific. The results describe a series of newly developed fluorescent ligands for the mu opioid receptor and the use of one of these ligands as a label for the cloned mu receptor. These ligands provide a new approach for studying the structural and biophysical nature of opioid receptors.


Assuntos
Compostos de Boro/metabolismo , Corantes Fluorescentes/metabolismo , Entorpecentes/metabolismo , Receptores Opioides mu/metabolismo , Animais , Corantes Fluorescentes/síntese química , Glioma/metabolismo , Entorpecentes/síntese química , Ratos , Receptores Opioides mu/análise , Células Tumorais Cultivadas
8.
Mol Pharmacol ; 52(5): 896-902, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9351981

RESUMO

In C6 glial cells stably expressing rat mu-opioid receptor, opioid agonist activation is negatively coupled to adenylyl cyclase through pertussis toxin-sensitive G proteins. In membranes, [D-Ala2, N-MePhe4,Gly-ol5]enkephalin (DAMGO) increases guanosine-5'-O-(3-[35S]thio)triphosphate (GTP[gamma-35S]) binding by 367% with an EC50 value of 28 nM. Prolonged exposure to agonists induced desensitization of the receptor as estimated by a reduction in the maximal stimulation of GTP[gamma-35S] binding by DAMGO and rightward shifts in the dose-response curves. In cells treated with 10 microM concentrations of etorphine, DAMGO, beta-endorphin, morphine, and butorphanol, DAMGO-stimulated GTP[gamma-35S] binding was 58%, 149%, 205%, 286%, and 325%, respectively. Guanine nucleotide regulation of agonist binding was correspondingly lower in membranes from tolerant cells. Furthermore, chronic opioid treatment increased forskolin-stimulated adenylyl cyclase activity, and potency of DAMGO to inhibit cAMP accumulation was lower in morphine- and DAMGO-tolerant cells (EC50 = 55 and 170 nM versus 18 nM for control). Chronic treatment with agonists reduced [3H]DAMGO binding in membranes with the rank order of etorphine > DAMGO = beta-endorphin > morphine > butorphanol, and the affinity of DAMGO in alkaloid- but not peptide-treated membranes was significantly lower in comparison with control. Pertussis toxin treatment of the cells before agonist treatment did not prevent the down-regulation by full agonists; DAMGO and etorphine exhibited approximately 80% internalization, whereas the ability of partial agonists was greatly impaired. In addition to establishing this cell line as a good model for further studies on the mechanisms of opioid tolerance, these results indicate important differences in the inactivation pathways of receptor triggered by full and partial agonists.


Assuntos
Analgésicos/farmacologia , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Adenilil Ciclases/metabolismo , Analgésicos/metabolismo , Animais , Ligação Competitiva , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Ligantes , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos
9.
J Pharmacol Exp Ther ; 283(2): 501-10, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9353363

RESUMO

A C6 glioma cell line stably transfected with the rat delta opioid receptor (C6delta) was used to characterize receptor binding and G protein activation by both peptide and nonpeptide delta opioid ligands. The ligand binding affinities for [3H]naltrindole and [3H]pCl-[D-Pen2,D-Pen5]enkephalin (DPDPE) were similar to those observed in monkey brain membranes. The nonpeptide agonists, BW373U86 and SNC80, as well as peptide agonist [D-Ser2, L-Leu5]enkephalyl-Thr maximally stimulated [35S]GTPgammaS binding by 640, 654 and 576%, respectively, over basal. The peptide agonists, DPDPE and deltorphin II, both stimulated [35S]GTPgammaS binding by 375%. Etorphine, diprenorphine, oxymorphindole and 7-spiroindanyloxymorphone were also partial agonists in this assay, although they were less efficacious than deltorphin II. Stimulation of [35S]GTPgammaS binding by agonists was blocked completely by pertussis toxin pretreatment. Both delta-1 and delta-2 selective antagonists 7-benzylidenenaltrexone and a benzofuran analog of naltrindole displayed high affinity for the cloned receptor (0.04 and 0.08 nM) and antagonized the stimulation of [35S]GTPgammaS binding by BW373U86 and DPDPE with similar potencies. Other evidence suggesting the lack of receptor subtypes includes the finding that stimulation of [35S]GTPgammaS binding by receptor subtype selective ligands DPDPE and deltorphin II was not additive. BW373U86, SNC80 and DPDPE maximally inhibited forskolin-stimulated adenylyl cyclase. These cells highly express a homogeneous population of delta opioid receptor that couple to inhibitory Go/Gi proteins. Ligand affinity for the delta opioid receptor correlates with ligand EC50 values for stimulation of [35S]GTPgammaS binding.


Assuntos
Glioma/metabolismo , Entorpecentes/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Toxina Adenilato Ciclase , Inibidores de Adenilil Ciclases , Animais , Toxina da Cólera/farmacologia , D-Penicilina (2,5)-Encefalina , Encefalinas/metabolismo , Encefalinas/farmacologia , Glioma/patologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Entorpecentes/metabolismo , Toxina Pertussis , Ratos , Receptores Opioides delta/classificação , Receptores Opioides delta/metabolismo , Células Tumorais Cultivadas , Fatores de Virulência de Bordetella/farmacologia
10.
J Neurochem ; 68(3): 1053-61, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9048750

RESUMO

SH-SY5Y neural cells expressing mu- and delta-opioid receptors were maintained viable in isotonic, sodium-free buffer in vitro. Intracellular sodium levels were manipulated by various methods, and ligand binding to intact cells was studied. In physiological buffer containing 118 mM sodium, [3H]Tyr-D-Ala-Gly-(Me)Phe-Gly-ol ([3H]-DAMGO) and [3H]naltrexone bound to mu receptor with KD values of 3.1 and 0.32 nM and Bmax values of 94 and 264 fmol/mg of protein, respectively. Replacement of sodium by choline decreased the affinity of the antagonist and increased Bmax for [3H]DAMGO, without significantly affecting the other corresponding binding parameters. Depolarizing concentrations of KCl (34 mM) in physiological buffer decreased the intracellular sodium levels by 67%, but this did not decrease the [3H]DAMGO binding to the cells. Incubation of cells with monensin and ouabain increased the intracellular sodium levels dramatically (from 78 to 250 and 300 nmol/mg, respectively), with no changes in agonist binding parameters. Ethylisopropylamiloride inhibited [3H]DAMGO and [3H]naloxone binding to intact cells with EC50 values of 24 and 3,600 nM, respectively. Adenylyl cyclase activities measured in intact cells, at different concentrations of sodium, showed the physiological significance of this ion in signal transduction. Potency of DAMGO in inhibiting the forskolin-stimulated adenylyl cyclase activity was significantly higher at lower concentrations of sodium. However, inhibition reached the maximal level only at 50 mM sodium, and typical sigmoidal dose-response curves were obtained only in the presence of 118 mM sodium. Furthermore, even at low or high intracellular sodium levels, DAMGO inhibition of cyclic AMP levels was normal. These results support a role for extracellular sodium in regulating not only the ligand interactions with the receptor, but also the signal transduction through the mu receptor.


Assuntos
Espaço Extracelular/metabolismo , Neurônios/metabolismo , Receptores Opioides mu/metabolismo , Sódio/fisiologia , Inibidores de Adenilil Ciclases , Amilorida/análogos & derivados , Amilorida/farmacologia , AMP Cíclico/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Monensin/farmacologia , Naltrexona/metabolismo , Concentração Osmolar , Ouabaína/farmacologia , Sódio/metabolismo , Células Tumorais Cultivadas
11.
Drug Alcohol Depend ; 42(2): 93-104, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8889408

RESUMO

Zipeprol was evaluated in a number of in vitro and in vivo assays predictive of stimulant, depressant, or opioid abuse potential. Zipeprol had affinity for mu and kappa opioid binding sites as well as sigma binding sites. However, it failed to exert opioid-like agonist actions in rodents, and did not attenuate withdrawal signs in morphine- or pentobarbital-dependent rats. Zipeprol did not substitute for either amphetamine or pentobarbital in drug discrimination assays in rhesus monkeys. On the other hand, it suppressed morphine withdrawal signs in rhesus monkeys in two assays, and it acted as a quadazocine-sensitive reinforcer in monkeys trained to self-inject alfentanil. Zipeprol also acted as a reinforcer in monkeys trained to self-inject methohexital. In a dose range of 10-18 mg/kg, zipeprol induced convulsions in monkeys. Zipeprol appears to have abuse potential and a novel spectrum of action involving both opioid and non-opioid effects.


Assuntos
Antitussígenos/farmacologia , Transtornos Relacionados ao Uso de Opioides/psicologia , Piperazinas/farmacologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Animais , Encéfalo/efeitos dos fármacos , Aprendizagem por Discriminação , Sinergismo Farmacológico , Feminino , Cobaias , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos ICR , Motivação , Ratos , Ratos Sprague-Dawley , Receptores Opioides/efeitos dos fármacos , Risco , Abuso de Substâncias por Via Intravenosa/psicologia , Síndrome de Abstinência a Substâncias/psicologia
12.
J Pharmacol Exp Ther ; 278(3): 1121-7, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8819494

RESUMO

In C6 glioma cells stably expressing a homogeneous population of the cloned rat mu opioid receptor, the binding affinities of opioid agonists and subsequent activation of G protein were examined. Opioid receptor number in membranes of these cells was high (10-30 pmol/mg protein [3H]diprenorphine binding sites). Opioids were found to bind to the receptor with high affinity [Tyr-D-Ala-Gly-(Me)Phe-Gly-ol (DAMGO) 0.23 nM; sufentanil 0.034 nM; morphine 0.16 nM]. Activation of G protein by opioid agonists was examined by measuring the stimulation of guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTP gamma S) binding. Sufentanil increased [35S]GTP gamma S binding by 326% with an EC50 value of 2.39 nM. Agonist stimulation of [35S]GTP gamma S binding was stereoselective, naltrexone-reversible, and pertussis toxin-sensitive. The "intrinsic activity" of opioids at the mu receptor was reflected by the magnitude of agonist-mediated activation of G protein. The rank order of the stimulation of [35S]GTP gamma S binding was etonitazene = sufentanil = DAMGO = PLO17 = fentanyl > morphine > profadol > meperidine > butorphanol = nalbuphine = pentazocine > cyclazocine = nalorphine > levallorphan > naltrexone. High affinity binding of ligands to the mu opioid receptor was reduced by the addition of sodium and guanosine diphosphate at concentrations used in the [35S]GTP gamma S binding assay. Ligand affinity was reduced in a manner correlating with "intrinsic activity". DAMGO, 1229-fold, nalbuphine 35-fold, naltrexone, 3-fold. The results presented show that the stable expression of the rat mu opioid receptor in C6 cells provides an effective tool to examine opioid receptor signal transduction mechanisms and evaluate the activity of novel opioids at the mu receptor.


Assuntos
Receptores Opioides mu/agonistas , Animais , Ligação Competitiva , Membrana Celular/metabolismo , Ativação Enzimática , Proteínas de Ligação ao GTP/metabolismo , Glioma , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ensaio Radioligante , Ratos , Receptores Opioides mu/fisiologia , Proteínas Recombinantes , Transdução de Sinais , Sufentanil/metabolismo , Células Tumorais Cultivadas
13.
Mol Pharmacol ; 48(4): 690-5, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7476895

RESUMO

Incubation of SH-SY5Y neural cells with mycophenolic acid (MPA), an inhibitor of inosine monophosphate dehydrogenase (the key enzyme in purine nucleotide biosynthesis), reduced the cellular content of GTP by 94% relative to its concentration in control cells (43 nmol/mg protein) without altering the level of GDP. Although in GTP-depleted intact cells the receptor binding parameters (Kd and Bmax) of the opioid antagonist [3H]naltrexone were unchanged from those in untreated cells, the binding affinity of the mu-selective opioid agonist [3H]Tyr-D-Ala-Gly-(Me)- Phe-Gly-ol ([3H]DAMGO) was enhanced 2-fold. Furthermore, the kinetics of ligand/receptor interaction revealed that in the nucleotide-depleted cells, the dissociation rate constant for [3H]DAMGO was reduced by 44%. Initial exposure of SH-SY5Y cells to pertussis toxin reduced high-affinity ligand binding by 95% and abolished the effect of MPA treatment. Renewed incubation of the GTP-depleted cells with guanosine restored the original GTP levels and agonist binding. Neither MPA nor guanosine treatment changed the Bmax of [3H]DAMGO binding. Forskolin- and prostaglandin E1-stimulated adenylyl cyclase activities were decreased significantly in GTP-depleted cells. DAMGO and [D-Pen2,D-Pen5]enkephalin inhibitions of adenylyl cyclase were also affected with MPA treatment. Maximal inhibition of forskolin-stimulated adenylyl cyclase activity by both of the agonists was reduced, whereas MPA caused a 2-fold reduction in potency for DAMGO. The results show that reduction in endogenous GTP levels leads to noticeable changes in agonist, receptor, and G protein interactions, as measured by agonist binding, and to subsequent diminution of the signal transduction, as reflected by the cAMP levels.


Assuntos
Guanosina Trifosfato/fisiologia , Neurônios/metabolismo , Receptores Opioides mu/metabolismo , Adenilil Ciclases/análise , Sequência de Aminoácidos , Analgésicos/metabolismo , Analgésicos/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Cinética , Dados de Sequência Molecular , Ácido Micofenólico/farmacologia , Naltrexona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Neuroblastoma , Neurônios/fisiologia , Neurônios/ultraestrutura , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Trítio , Células Tumorais Cultivadas
14.
J Pharmacol Exp Ther ; 274(1): 17-28, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7616395

RESUMO

These studies examined the opioid receptor binding affinities and behavioral effects of several fentanyl derivatives in rhesus monkeys. OHM3295, OHM3296, OHM3326 and OHM3463 displayed high affinity for mu (IC50 = 7-66 nM) as compared to kappa (IC50 = 263-3255 nM) or delta (IC50 = 480-4500 nM) receptors as measured by their ability to displace [3H](D-Ala2-Me-Phe4,Glyol5)enkephalin, [3H](5,7,8[beta])-N-[2- (1-pyrrolidinyl)1-oxaspiro[4,5]dec-8-yl]benzeneacetamide and [3H](D-Pen2-D-Pen5)enkephalin, respectively. All four compounds maintained i.v. self-administration responding at rates above those maintained by the mu agonist alfentanil. In drug discrimination studies, OHM3463, OHM3326 and OHM3296 substituted completely for nalbuphine whereas OHM3295, and a related compound, mirfentanil, substituted partially for nalbuphine. In morphine-treated monkeys, OHM3295 substituted for naltrexone; in monkeys acutely deprived of morphine, only OHM3463 reversed naltrexone-lever responding. All four compounds had antinociceptive effects, although the extent to which these effects were accompanied by respiratory depression or modified by naltrexone, as well as the interactions between antinociceptive effects of fentanyl derivatives and alfentanil, varied markedly among compounds. Thus, OHM3463 shared effects with mu agonists (e.g., alfentanil) under all conditions; the other three compounds had opioid agonist effects under only a subset of conditions. Moreover, one of these compounds (OHM3295) antagonized the discriminative stimulus and antinociceptive effects of other mu agonists. Collectively, these compounds appear to vary on two dimensions: opioid efficacy and the contribution of nonopioid actions to their antinociceptive effects. Together with results obtained with other fentanyl derivatives (mirfentanil) under similar conditions, results of the current study suggest this chemical class might be especially fertile for the development of novel analgesics that might have reduced toxicity and abuse liability as compared to fentanyl and related compounds that are currently used in medicine.


Assuntos
Comportamento Animal/efeitos dos fármacos , Fentanila/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Animais , Fentanila/administração & dosagem , Fentanila/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Macaca mulatta , Nalbufina/farmacologia , Naltrexona/farmacologia , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Respiração/efeitos dos fármacos , Autoadministração
15.
J Pharmacol Exp Ther ; 271(3): 1630-7, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7996478

RESUMO

The binding parameters of radiolabeled DAMGO (mu), DPDPE and pCl-DPDPE (delta) and 5 alpha, 7 alpha, 8 beta-N-methyl-N-[7-(1- pyrrolidinyl)-1-oxaspiro(4,5)dec-8-yl]benzeneacetamide (also known as U69593, kappa) and the affinity and selectivity profiles of various opioid agonists and antagonists at the three opioid receptor types were determined in membranes from brain cortex of rhesus monkey. Among the 10 opioids with established mu-selective actions, etonitazene inhibited the binding of [3H]DAMGO with a Ki of 0.02 nM (0.01 nM without sodium) and exhibited mu/delta and mu/kappa selectivities of 8800 and 11,650, respectively. DAMGO had a Ki of 1.23 nM and was about 500-fold more selective at mu receptors compared with delta and kappa sites. Other mu opioids with higher than 100-fold binding selectivity were fentanil and sufentanil. Highly selective delta opioids were DPDPE, deltorphin II and naltrindole. With the exception of N,N-diallyl-Tyr-Aib-Aib-Phe-Leu-OH, all investigated putative delta opioids bound to delta sites with low Kis, i.e., 0.04 nM, 0.13 nM and 1.4 nM for naltrindole, (+/-)-4-[(alpha-R*)-alpha-((2S*,5R*)-4-allyl-2,5-dimethyl-1-piperazinyl) -3- hydroxybenzyl]-N,N-diethylbenzamide and DPDPE, respectively. In this series, the displacement of [3H]pCl-DPDPE yielded results similar to those obtained with [3H]DPDPE. With nanomolar Kis of 0.70, 0.89, 0.25 and 0.06, respectively, the highest kappa selectivity was displayed by (trans)-(+/-)-3,4-dichloro-N-methyl- N-[2-(1-pyrrolidinyl)-cyclohexyl]benzeneacetamide and U69593, followed by dynorphin 1-13 and norbinaltorphimine.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Encéfalo/metabolismo , Entorpecentes/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina , D-Penicilina (2,5)-Encefalina , Encefalinas/metabolismo , Macaca mulatta , Masculino , Relação Estrutura-Atividade
16.
J Pharmacol Exp Ther ; 271(2): 715-21, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7965787

RESUMO

The effects of the systemically active irreversible opioid receptor antagonist clocinnamox (C-CAM; 14 beta-(p-chlorocinnamoylamino)-7,8-dihydro-N- cyclopropylmethyl normorphinone mesylate) on mu receptor binding to cerebral membranes and on mu opioid analgesia were assessed using mice. After systemic administration, C-CAM produced a dose-dependent decrease in the Bmax values of both [3H]DAMGO ([D-Ala2, N-MePhe4, Gly5-ol][tyrosyl-3,5-3H]enkephalin) and [3H]naltrexone without affecting the Kd value of either ligand. After administration of 3.2 mg/kg of C-CAM, [3H]DAMGO binding recovered gradually, returning to control levels by 8 days. This time course of recovery was similar to that observed with 3.2 mg/kg of C-CAM against morphine analgesia in the warm-water tail-withdrawal assay. The analgesic effect of the mu agonist etonitazene also was assessed in the assay. C-CAM produced dose-dependent rightward and slight downward shifts of the etonitazene dose-effect curve. The analgesic activity of etonitazene had still not returned to base-line levels 12 days after administration of 32 mg/kg of C-CAM, a time at which [3H]DAMGO binding had returned to control levels. In addition, the apparent pA2 values of etonitazene with naltrexone in the tail-withdrawal assay were assessed at 4, 8 and 12 days after the administration of 32 mg/kg of C-CAM, and none were found to be different from the control pA2 value. These results support the notion that C-CAM is an irreversible mu receptor antagonist and suggest that post-treatment, perhaps newly synthesized, mu receptors are similar to mu receptors in control membranes.


Assuntos
Analgesia , Cinamatos/farmacologia , Derivados da Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Animais , Benzimidazóis/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Masculino , Camundongos , Naltrexona/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo
17.
J Med Chem ; 37(20): 3408-18, 1994 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-7932569

RESUMO

The enantiomeric (-)- and (+)-N-(methyl through decyl) normetazocines (5,9 alpha-dimethyl-2'-hydroxy-6,7-benzomorphans) were synthesized and their in vitro and in vivo activities determined. Increasingly bulky enantiomeric N-alkyl homologs were prepared until their interaction with the sigma 1 receptor decreased and their insolubility became a hindrance to their evaluation in vivo and/or in vitro. The (-)-methyl, -pentyl, -hexyl, and -heptyl homologs were essentially as potent as, or more potent than, morphine in the tail-flick, phenylquinone, and hot-plate assays for antinociceptive activity; the (-)-propyl homolog had narcotic antagonist activity between that of nalorphine and naloxone in the tail-flick vs morphine assay, and it also displayed antagonist properties in the single-dose suppression assay in the rhesus monkey. The antinociceptively potent (-)-heptyl homolog did not substitute for morphine in monkeys but did show morphine-like properties in a primary physical-dependence study in continuously infused rats. All five potent compounds showed high affinity for the mu opioid receptor from both rat and monkey preparations and the kappa opioid receptor (< 0.05 microM), and all except the (-)-methyl homolog interacted reasonably well at the delta receptor (K(i) < 0.1 microM). The (-)-propyl compound was equipotent (K(i) 1.5-2.0 nM) at mu and kappa receptors. The pattern of interaction of the (-)-enantiomeric homologs with mu receptors from rat and monkey preparations was similar, but not identical. The enantioselectivity of the homologs for mu receptors was greater in the rat than in the monkey preparation for all but the N-H and butyl compounds, and the enantioselectivity of the lower homologs (methyl through butyl) for the mu (monkey) receptor was greater than for the kappa or delta receptors. However, bulkier homologs (hexyl through decyl) displayed higher enantioselectivity at kappa or delta receptors than at the mu (monkey) receptor. The (+)-butyl through (+)-octyl homologs were essentially equipotent with, or more potent than, (+)-pentazocine at the sigma receptor. Only the (+)-H and (+)-methyl homologs had high affinity (< 0.05 microM) at PCP binding sites.


Assuntos
Ciclazocina/análogos & derivados , Entorpecentes/síntese química , Sequência de Aminoácidos , Analgesia , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Ciclazocina/química , Ciclazocina/metabolismo , Ciclazocina/farmacologia , Feminino , Cobaias , Macaca mulatta , Masculino , Camundongos , Dados de Sequência Molecular , Entorpecentes/metabolismo , Entorpecentes/farmacologia , Medição da Dor , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Receptores sigma/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
18.
J Pharmacol Exp Ther ; 268(1): 47-58, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8301589

RESUMO

Bremazocine, [5R-(5,7,8 beta)]-N-methyl-N-[7-(1-pyrrolidinyl)1-oxaspiro [4,5]dec-8-yl]-4-benzofuranacetamide (Cl-977), (+-)-trans-3,4-dichloro-N- methyl-(2-(pyrrolidin-1-yl)-5-methoxy-1,2,3,4-tetrahydronapth++ +-1-yl benzeneacetamide methanesulfonate (DUP 747), ethylketocyclazocine (EKC), nalorphine, (+/-)-trans-N-methyl-N-[2-(1- pyrrolidnyl)-cyclohexyl]benzo[b]thiophene-4-acetamide (PD117302), trans-(+/-)-3,4-dichloro-N-methyl-[2-(1-pyrrolidinyl)- cyclohexyl]benzeneacetamide (U-50,488), (5,7,8 beta)-N-methyl-N[2-(1- pyrrolidinyl), 1-oxaspiro[4,5]dec-8-yl benzeneacetamide (U-69,593) and spiradoline were compared in rhesus monkeys for their discriminative stimulus, analgesic and respiratory effects. Selected compounds also were studied for their binding affinities at mu [[3H](D-Ala2-Me-Phe4,Glyol5)enkephalin], kappa ([3H]U-69,593) and delta [[3H](D-Pen2-D-Pen5) enkephalin], opioid receptors in monkey brain membranes. All compounds substituted completely (> or = 90%) for EKC in monkeys discriminating between EKC and saline, with the exception that DUP 747 produced a maximum of 74% EKC responding. None of the compounds reversed naltrexone responding in morphine-abstinent monkeys; all of the compounds substituted for naltrexone in morphine-treated monkeys discriminating between naltrexone and saline, with the exception that spiradoline produced a maximum of 68% naltrexone responding. Eight compounds produced maximum analgesic effects in a tail withdrawal procedure and quadazocine antagonized these effects; nalorphine did not have analgesic effects, but it antagonized analgesic effects of several other compounds. U-50,488 did not decrease respiratory function, whereas U-69,593 decreased frequency of respiration and volume of respiration to less than 40% of control values; Cl-977, DUP 747, PD117302 and spiradoline had limited effects on respiratory function. Larger doses of each compound increased both respiration and motor activity.


Assuntos
Comportamento Animal/efeitos dos fármacos , Entorpecentes/farmacologia , Receptores Opioides kappa/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Sítios de Ligação , Etilcetociclazocina/farmacologia , Feminino , Macaca mulatta , Masculino , Estrutura Molecular , Naltrexona/farmacologia , Entorpecentes/classificação , Entorpecentes/metabolismo , Receptores Opioides kappa/metabolismo , Respiração/efeitos dos fármacos
20.
J Pharmacol Exp Ther ; 267(2): 896-903, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8246165

RESUMO

Rhesus monkeys were trained to respond under a fixed-ratio 30 schedule of food reinforcement. The mu opioid agonists alfentanil and fentanyl, the kappa opioid agonists ethylketocyclazocine (EKC) and U69,593, the delta opioid agonist BW373U86 [(+-)-4-((R*)-a-((2S*5R*)-4-allyl-2,5-dimethyl-1-piperazinal)-3-h ydroxy- benzyl)-N,N-diethylbenzamide dihydrochloride] and the nonopioid, noncompetitive N-methyl-D-aspartate antagonist ketamine all produced a dose-dependent decrease in rates of responding. Quadazocine (0.1-10 mg/kg) antagonized the rate-decreasing effects of all the opioid agonists, but not of ketamine. The in vivo apparent pA2 values (95% CL) for quadazocine in combination with each agonist were: alfentanil, 7.7 (7.6-7.8); fentanyl, 7.7 (7.6-7.8); EKC, 6.3 (5.9-6.7); U69,593, 6.5 (5.9-7.0); and BW373U86, 5.5 (5.3-5.8). Additionally, antagonist effects of individual doses of quadazocine in combination with each agonist were evaluated by using in vivo apparent pKB analysis, and pKB values were found to be similar to the more rigorously determined pA2 values. The relative pA2 and pKB values of quadazocine in antagonizing the rate-decreasing effects of mu, kappa and delta opioid agonists corresponded to the relative potency of quadazocine in displacing the specific binding of the mu agonist [3H]Tyr-D-Ala-Gly-(Me)-Phe-Gly-ol (IC50 = 0.080 nM), the kappa agonist [3H]U69,593 (IC50 = 0.52 nM) and the delta agonist [D-Pen2,D-Pen5]-[3H]enkephalin (IC50 = 4.6 nM) from binding sites in membranes from monkey brain cortex.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Azocinas/farmacologia , Condicionamento Operante/efeitos dos fármacos , Antagonistas de Entorpecentes , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides/fisiologia , Sequência de Aminoácidos , Animais , Azocinas/metabolismo , Comportamento Animal/efeitos dos fármacos , Benzamidas/farmacologia , Ligação Competitiva , Relação Dose-Resposta a Droga , Feminino , Cinética , Macaca mulatta , Masculino , Dados de Sequência Molecular , Antagonistas de Entorpecentes/metabolismo , Piperazinas/farmacologia , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...