Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Neurosci Lett ; 563: 155-9, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24503508

RESUMO

It has been shown previously that norbinaltorphimine (norBNI) and 5'-guanidinonaltrindole (5'-GNTI), long-acting kappa opioid receptor (KOPR) antagonists, cause frenzied scratching in mice [1,2]. In the current study, we examined if zyklophin, a short-acting cyclic peptide KOPR antagonist, also elicited scratching behavior. When injected s.c. in the nape of the neck of male Swiss-Webster mice, zyklophin at doses of 0.1, 0.3 and 1mg/kg induced dose-related hindleg scratching of the neck between 3 and 15 min after injection. Pretreating mice with norBNI (20mg/kg, i.p.) at 18-20 h before challenge with zyklophin (0.3mg/kg) did not markedly affect scratching. Additionally, KOPR-/- mice given 0.3mg/kg of zyklophin displayed similar levels of scratching as wild-type animals. The absence of KOPR in KOPR-/- mice was confirmed with ex vivo radioligand binding using [(3)H]U69,593. Taken together, our data suggest that the presence of kappa receptors is not required for the excessive scratching caused by zyklophin. Thus, zyklophin, similar to the structurally different KOPR antagonist 5'-GNTI, appears to act at other targets to elicit scratching and potentially the sensation of itch.


Assuntos
Dinorfinas/efeitos adversos , Fragmentos de Peptídeos/efeitos adversos , Prurido/induzido quimicamente , Receptores Opioides kappa/antagonistas & inibidores , Animais , Benzenoacetamidas/farmacologia , Relação Dose-Resposta a Droga , Dinorfinas/administração & dosagem , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Pirrolidinas/farmacologia , Ensaio Radioligante , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética
2.
Neuroscience ; 223: 447-56, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22863678

RESUMO

Recently there has been a widespread interest in the development of kappa opioid receptor (KOPR) ligands for treatment of pain, depression and anxiety, and prevention of stress-induced drug relapse. However, most of these preclinical studies have been conducted using male experimental animals. In the present study we examined if sex differences exist in neural activity induced by the KOPR agonist trans-(±)-3,4-dichloro-N-methyl-N-(2-[1-pyrrolidinyl]-cyclohexyl) benzeneacetamide methanesulfonate (U50,488H). Here, we used immunohistochemistry to detect activation (phosphorylation) of p44/42 mitogen-activated protein kinase (MAPK) as an indicator of neural activity. Following habituation to injection for 3 days, adult guinea pigs received a single injection of U50,488H (5mg/kg, s.c.) and perfused 30-45 min later. U50,488H-induced an increase in the number of cells immuno-positive for phosphorylated p44/42 MAPK in subregions of the amygdala, thalamus, paraventricular nucleus of the hypothalamus, periaqueductal gray, and dorsal raphe nuclei. In contrast, U50,488H-induced a decrease in immuno-positive cells in the ventrolateral and lateral orbital cortex. Pretreatment with the KOPR antagonist norbinaltorphimine (10mg/kg, i.p.) 18 h prior to U50,488H significantly reversed the effects of U50,488H in most regions. In addition, we observed a notable sex difference in the basolateral amygdala; in males, U50,488H induced an increase in immuno-positive cell numbers but a decrease in females. However, across other brain regions males were generally more sensitive to U50,488H-induced alterations than females. These results suggest the need to include female subjects in studies examining emotional responses to KOPR ligands.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos não Narcóticos/farmacologia , Encéfalo/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Caracteres Sexuais , Animais , Encéfalo/enzimologia , Mapeamento Encefálico , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Cobaias , Masculino , Fosforilação/efeitos dos fármacos
3.
Neuroscience ; 205: 178-84, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22240251

RESUMO

OPRM1 A118G is a common single nucleotide polymorphism (SNP) in the coding region of the human mu opioid receptor (MOPR) gene OPRM1. This SNP is associated with higher morphine doses required for postoperative analgesia as well as a variety of drug addiction phenotypes. A mouse model possessing the equivalent substitution (A112G) in the Oprm1 gene was generated to facilitate mechanistic studies. Mice homozygous for the G112 allele (G/G) displayed lower antinociception to morphine compared with those homozygous for A112 allele (A/A), similar to humans, suggesting that the mice are a good model to further characterize underlying factors contributing to phenotypes associated with this SNP. Here, we compared [³H]DAMGO binding to the MOPR in the brains of A/A and G/G mice using quantitative in vitro autoradiography. A/A mice exhibited higher [³H]DAMGO binding than G/G in the cingulate, motor, and insular cortices, nucleus accumbens core and shell, hypothalamus, thalamus, amygdala, periaqueductal gray, superficial gray of superior colliculus, and ventral tegmental area. No genotype differences were observed in somatosensory cortex, caudate putamen, and hippocampus. When males and females were examined separately, A/A mice showed higher [³H]DAMGO binding than G/G mice in more brain regions in males than in females. Radioligand binding using brain membranes also showed higher [³H]DAMGO binding in the cortex and thalamus in A/A mice than G/G mice but no genotype differences in the caudate putamen or hippocampus. Thus, the A112G SNP is associated with reduced MOPR expression in some, but not all, brain regions, and appears to have some sex differences. The elevated MOPR expression in periaqueductal gray and thalamus in A/A mice are consistent with their higher antinociceptive responses to morphine. The higher MOPR levels in nucleus accumbens and/or ventral tegmental area of A/A mice is consistent with the higher morphine-induced hyperactivity and locomotor sensitization observed in these mice. Thus, these results provide some insights into the observed decreased clinical opioid potency in humans with the A118G SNP.


Assuntos
Regulação para Baixo/genética , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Alanina/genética , Substituição de Aminoácidos/efeitos dos fármacos , Substituição de Aminoácidos/genética , Analgésicos Opioides/farmacologia , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Feminino , Técnicas de Introdução de Genes , Glicina/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único/genética , Receptores Opioides mu/biossíntese , Transgenes/efeitos dos fármacos , Transgenes/genética
4.
Neuroscience ; 159(2): 492-500, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19373968

RESUMO

With the use of a rabbit polyclonal antiserum against a conserved region (54-118) of C-peptide of human preproinsulin-like peptide 7, referred to herein as C-INSL7, neurons expressing C-INSL7-immunoreactivity (irC-INSL7) were detected in the pontine nucleus incertus, the lateral or ventrolateral periaqueductal gray, dorsal raphe nuclei and dorsal substantia nigra. Immunoreactive fibers were present in numerous forebrain areas, with a high density in the septum, hypothalamus and thalamus. Pre-absorption of C-INSL7 antiserum with the peptide C-INSL7 (1 microg/ml), but not the insulin-like peptide 7 (INSL7; 1 microg/ml), also known as relaxin 3, abolished the immunoreactivity. Optical imaging with a voltage-sensitive dye bis-[1,3-dibutylbarbituric acid] trimethineoxonol (DiSBAC4(3)) showed that C-INSL7 (100 nM) depolarized or hyperpolarized a small population of cultured rat hypothalamic neurons studied. Ratiometric imaging studies with calcium-sensitive dye fura-2 showed that C-INSL7 (10-1000 nM) produced a dose-dependent increase in cytosolic calcium concentrations [Ca2+]i in cultured hypothalamic neurons with two distinct patterns: (1) a sustained elevation lasting for minutes; and (2) a fast, transitory rise followed by oscillations. In a Ca2+-free Hanks' solution, C-INSL7 again elicited two types of calcium transients: (1) a fast, transitory increase not followed by a plateau phase, and (2) a transitory rise followed by oscillations. INSL7 (100 nM) elicited a depolarization or hyperpolarization in a small population of hypothalamic neurons, and an increase of [Ca2+]i with two patterns that were dissimilar from that of C-INSL7. [125I]C-INSL7 bindings to rat brain membranes were inhibited by C-INSL7 in a dose-dependent manner; the Kd and Bmax. values were 17.7 +/- 8.2 nM and 45.4 +/- 20.5 fmol/mg protein. INSL7 did not inhibit [125I]C-INSL7 binding to rat brain membranes, indicating that C-INSL7 and INSL7 bind to distinct binding sites. Collectively, our result raises the possibility that C-INSL7 acts as a signaling molecule independent from INSL7 in the rat CNS.


Assuntos
Encéfalo/metabolismo , Peptídeo C/metabolismo , Animais , Encéfalo/anatomia & histologia , Peptídeo C/farmacologia , Cálcio/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Eletrofisiologia/métodos , Fura-2/metabolismo , Hipotálamo/citologia , Isótopos de Iodo/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Tiobarbitúricos/metabolismo
5.
Naunyn Schmiedebergs Arch Pharmacol ; 379(3): 263-70, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18925386

RESUMO

Kappa opioid receptor agonists induce water diuresis in animals and humans. We investigated the effects of s.c. nalfurafine, U50,488H, salvinorin A, and its longer-acting analog, 2-methoxymethyl-salvinorin B (MOM-sal B), on urinary output and sodium excretion over 5 h in euvolemic rats. Nalfurafine (0.005-0.02 mg/kg), U50,488H (0.1-10 mg/kg), and MOM-sal B (0.625-5 mg/kg) induced diuresis dose-dependently. Systemically (0.1-10 mg/kg) or centrally (50 microg, i.c.v.) administered salvinorin A was ineffective. 5'-Guanidinonaltrindole, a kappa receptor antagonist, inhibited nalfurafine- and MOM-sal B-induced diuresis. Nalfurafine and MOM-sal B had no effect on arginine vasopressin levels, measured at 2 h. Tolerance did not develop to the diuresis accompanying subchronic administration of nalfurafine (0.02 mg/kg). On the basis of our work, we (a) promote nalfurafine as a candidate diuretic to relieve water retention and (b) highlight salvinorin A as a kappa agonist that does not cause diuresis, probably because of its short duration of action.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Diterpenos Clerodânicos/farmacologia , Diuréticos/farmacologia , Morfinanos/farmacologia , Receptores Opioides kappa/agonistas , Compostos de Espiro/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/química , Animais , Diterpenos Clerodânicos/química , Diurese/efeitos dos fármacos , Diuréticos/química , Relação Dose-Resposta a Droga , Masculino , Morfinanos/química , Ratos , Ratos Sprague-Dawley , Compostos de Espiro/química , Relação Estrutura-Atividade , Fatores de Tempo
6.
Neuroscience ; 142(2): 481-91, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16893609

RESUMO

Numerous studies support a role for the endogenous opioid system in cocaine-influenced behavior. Few of these studies, however, selectively delineate a role for the mu opioid receptor (MOR) in this regard. This investigation examined if the MOR modulates cocaine-induced behavior in mice using a 17-base antisense oligodeoxynucleotide (AS ODN) directed against the MOR coding sequence 16-32. Specifically, cocaine-induced behavioral sensitization and conditioned reward were investigated. For the sensitization study, C57BL/6J mice received eight intermittent i.c.v. infusions of saline, mismatch oligodeoxynucleotide (ODN) (20 microg/4 microl) or AS ODN (20 microg/4 microl) over 20 days. Mice also received concomitant once daily i.p. injections of saline (4 ml/kg) or cocaine (15 mg/kg) for 10 days. There was a 7-day withdrawal period, after which all mice were challenged with cocaine (15 mg/kg) to test for behavioral sensitization. For the conditioned place preference (CPP) study, mice received five i.c.v. infusions of mismatch ODN or MOR AS ODN (days 1-5). An unbiased counterbalanced conditioning procedure was used where mice were conditioned with saline (4 ml/kg, i.p.) and cocaine (15 mg/kg, i.p.) on alternate days for four sessions (days 3-6). Mice were tested on day 7 for CPP. Immediately following testing, [3H]DAMGO (D-Ala2, N-Me-Phe4, Gly-ol5-enkephalin) receptor binding to brain homogenates was conducted. MOR AS attenuated cocaine-induced behavioral sensitization and conditioned reward. MOR AS ODN also reduced [3H]DAMGO binding. Collectively, these findings implicate the MOR as playing an important neuromodulatory role in the behavioral effects of cocaine in mice.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Receptores Opioides mu/fisiologia , Recompensa , Análise de Variância , Animais , Comportamento Animal , Transtornos Relacionados ao Uso de Cocaína/etiologia , Condicionamento Operante/efeitos dos fármacos , Vias de Administração de Medicamentos , Interações Medicamentosas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Entorpecentes/farmacologia , Ligação Proteica/efeitos dos fármacos , Radiografia/métodos , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética , Fatores de Tempo , Trítio/farmacocinética
7.
Neuroscience ; 140(4): 1265-76, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16650615

RESUMO

We have previously demonstrated that GEC1 interacts with the kappa opioid receptor and GEC1 expression enhances cell surface expression of the receptor in Chinese hamster ovary cells. In this study, we generated an antiserum (PA629) directed against GEC1 in rabbits, characterized its specificity, and investigated distribution of GEC1 in tissues and in brain regions and spinal cord and its subcellular localization in hypothalamic neurons in the rat. Immunofluorescence staining demonstrated that PA629 recognized HA-GEC1 transfected into Chinese hamster ovary cells, but not HA-GABARAP or HA-GATE-16, although the three share high homology. Pre-incubation of PA629 with GST-GEC1, but not GST, abolished the staining. In immunoblotting, affinity-purified PA629 (PA629p) recognized GEC1, GABARAP and GATE-16. GEC1 migrated slower than GABARAP and GATE-16, with a M(r) of 16 kDa for GEC1 and M(r) of 14 kDa for GABARAP and GATE-16. Immunoblotting results showed that GEC1 level was higher in liver and brain than in lung and heart, and very low in kidney and skeletal muscle. GEC1 was present in all rat brain regions examined and spinal cord. Immunohistochemistry demonstrated that GEC1 immunoreactivity was distributed ubiquitously in the rat CNS with highly intense immunoreactivity in various brain nuclei and motor neurons of the spinal cord. Ultrastructural examination of neurons in the paraventricular nucleus of the hypothalamus showed that GEC1 was associated with endoplasmic reticulum and Golgi apparatus and distributed along plasma membranes and in cytosol. Coupled with our previous observation that GEC1 interacts with N-ethylmaleimide-sensitive factor, these findings strongly suggest that GEC1 functions in intracellular trafficking in the biosynthesis pathway and perhaps also the endocytic pathway. The widespread distribution of GEC1 suggests that GEC1 may be associated with many proteins, in addition to the kappa opioid receptor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Química Encefálica/fisiologia , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Proteínas Associadas aos Microtúbulos/metabolismo , Medula Espinal/metabolismo , Medula Espinal/ultraestrutura , Animais , Sistema Nervoso Central/química , Sistema Nervoso Central/ultraestrutura , Masculino , Proteínas Associadas aos Microtúbulos/ultraestrutura , Ratos , Ratos Sprague-Dawley
8.
Biochemistry ; 40(45): 13501-9, 2001 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-11695897

RESUMO

Mutations within the "X1BBX2X3B" motif or its variants in the junction of the third intracellular (i3) loop and the sixth transmembrane domain (TM6) have been shown to lead to constitutive activation of several G protein-coupled receptors (GPCRs). In this study, T6.34(279) at the X3 locus of the rat mu opioid receptor was mutated to Lys and Asp, and the mutants were examined for binding and signaling properties. The T6.34(279)K mutant was poorly expressed, and pretreatment with naloxone greatly enhanced its expression. This construct exhibited properties identified previously with constitutive activation: (1) compared with the wild type, it produced much higher agonist-independent [35S]GTPgammaS binding, which was abolished by pertussis toxin treatment; (2) it displayed an enhanced affinity for the agonist DAMGO similar to that of the high-affinity state of the wild type, which was not altered by GTPgammaS, while having unchanged affinity for the antagonist diprenorphine. The T6.34(279)K mutant displayed a higher intracellular receptor pool than the wild type. Naloxone inhibited the basal [35S]GTPgammaS binding of the T6.34(279)K mutant, demonstrating inverse agonist activity at this mutant receptor. In contrast, the T6.34(279)D substitution did not increase basal [35S]GTPgammaS binding, greatly reduced agonist-promoted [35S]GTPgammaS binding, and markedly decreased affinity for DAMGO. Thus, the T6.34(279)D mutant adopts conformations corresponding to inactive states of the receptor. The results were interpreted in the structural context of a model for the mu opioid receptor that incorporates the information from the crystal structure of rhodopsin. The interaction of T6.34(279) with R3.50(165) in the mu opioid receptor is considered to stabilize the inactive conformations. The T6.34(279)K substitution would then disrupt this interaction and support agonist-free activation, while T6.34(279)D mutation should strengthen this interaction which keeps the receptor in inactive states. T6.34(279) may, in addition, interact with the neighboring R6.35(280) to help constrain the receptor in inactive states, and T6.34(279)K and T6.34(279)D mutations would affect this interaction by disrupting or strengthening it, respectively. To the best of our knowledge, the results presented here represent the first structurally rationalized demonstration that mutations of this locus can lead to dramatically different properties of a GPCR.


Assuntos
Motivos de Aminoácidos/fisiologia , Receptores Opioides mu/fisiologia , Motivos de Aminoácidos/genética , Substituição de Aminoácidos , Animais , Ácido Aspártico/genética , Células CHO , Sequência Conservada , Cricetinae , Diprenorfina/metabolismo , Expressão Gênica/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Lisina/genética , Modelos Moleculares , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Toxina Pertussis , Ratos , Receptores Opioides mu/química , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Radioisótopos de Enxofre , Treonina/genética , Trítio , Fatores de Virulência de Bordetella/farmacologia
9.
Biochemistry ; 40(40): 12039-50, 2001 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-11580279

RESUMO

The roles of conserved aspartates in the third transmembrane domain of the rat mu opioid receptor (RMOR) were explored with mutations of D3.32(147) and D3.49(164). D3.49(164) in the highly conserved DRY motif was mutated to 13 amino acids. Except for the D3.49(164)E mutant, each mutant displayed little or no detectable [(3)H]diprenorphine binding, and pretreatment with naloxone greatly enhanced binding. D3.49(164)H, -Q, -Y, -M, and -E mutants were further studied. D3.32(147) was substituted with A or N. All seven mutants exhibited similar binding affinities for the antagonist [(3)H]diprenorphine as the wild-type. The D3.49(164)H, -Q, -Y, and -M mutants, but not the D3.49(164)E and D3.32(147) mutants, exhibited enhanced basal [(35)S]GTPgammaS binding which was comparable to the maximally activated level of the wild-type and was related to expression levels. Naloxone, naltrexone, and naloxone methiodide significantly inhibited the basal [(35)S]GTPgammaS binding of the D3.49(164) mutants, indicating inverse agonist activities. Treatment of the D3.49(164)Y mutant with pertussis toxin greatly reduced the basal [(35)S]GTPgammaS binding, demonstrating constitutive activation of Galpha(i)/Galpha(o). The D3.49(164)H, -Y, -M, and -Q mutants had higher affinities for DAMGO than the wild-type, which were not significantly lowered by GTPgammaS. Thus, mutation of D3.49(164) to H, Y, M, or Q in RMOR resulted in receptor assuming activated conformations. In contrast, the D3.49(164)E mutant displayed significantly lower basal [(35)S]GTPgammaS binding and reduced affinity for DAMGO. Upon incubation of membranes at 37 degrees C, the constitutively active D3.49(164)Y mutant was structurally less stable, whereas the inactivated D3.49(164)E mutant was more stable, than the wild-type. Computational simulations showed that the E3.49 side chain interacted strongly with the conserved R3.50 in the DRY motif and stabilized the inactive form of the receptor. Taken together, these results indicate that D3.49 plays an important role in constraining the receptor in inactive conformations.


Assuntos
Receptores Opioides mu/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Diprenorfina/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Ligação de Hidrogênio , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Toxina Pertussis , Ligação Proteica , Ratos , Receptores Opioides mu/química , Receptores Opioides mu/genética , Homologia de Sequência de Aminoácidos , Fatores de Virulência de Bordetella/metabolismo , Fatores de Virulência de Bordetella/farmacologia
10.
Mol Pharmacol ; 60(5): 1064-75, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11641435

RESUMO

We demonstrated previously that D3.49(164) mutations resulted in constitutive activation of the rat mu-opioid receptor and abolished receptor expression unless cells were pretreated with naloxone, an inverse agonist. In this study, we investigated the properties of the D3.49(164)Q mutant and the mechanisms underlying the effect of naloxone. Naloxone pretreatment up-regulated [(3)H]diprenorphine binding and protein expression of the D3.49(164)Q mutant in a time- and dose-dependent manner without affecting its mRNA level. After naloxone removal, binding and protein expression of the mutant declined with time with no effect on its mRNA level. Naloxone methiodide (a quaternary ammonium analog) caused a maximal up-regulation about 50% of the naloxone effect, indicating that naloxone acts extracellularly and intracellularly. Expression of the mutant was enhanced by inverse agonists, a neutral antagonist, and agonists, with inverse agonists being most effective. In membranes, the mutant was structurally less stable than the wild type upon incubation at 37 degrees C, and naloxone and [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin stabilized the mutant. Coexpression of the dominant-negative mutants GRK2-K220R, arrestin-2(319-418), dynamin I-K44A, rab5A-N133I or rab7-N125I partially prevented the decline in binding of the mutant after naloxone removal. Chloroquine or proteasome inhibitor I reduced the down-regulation of the mutant. These results indicate that the D3.49(164)Q mutant is constitutively internalized via G protein coupled-receptor kinase-, arrestin-2-, dynamin-, rab5-, and rab7-dependent pathways and probably trafficked through early and late endosomes into lysosomes and degraded by lysosomes and proteasomes. Naloxone up-regulates the D3.49(164)Q mutant by stabilizing the mutant protein and blocking its constitutive internalization and down-regulation. To the best of our knowledge, this represents the first comprehensive analysis of the mechanisms involved in up-regulation of constitutively active mutants by an inverse agonist.


Assuntos
Diprenorfina/metabolismo , Naloxona/análogos & derivados , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/metabolismo , Animais , Sítios de Ligação , Transporte Biológico , Células CHO , Cricetinae , Cisteína Endopeptidases/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Endossomos/metabolismo , Lisossomos/metabolismo , Complexos Multienzimáticos/metabolismo , Mutação , Complexo de Endopeptidases do Proteassoma , Compostos de Amônio Quaternário , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Ratos , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética , Fatores de Tempo , Trítio , Regulação para Cima/efeitos dos fármacos
11.
Biochemistry ; 40(27): 8018-29, 2001 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-11434771

RESUMO

We have mapped the residues in the sixth transmembrane domains (TMs 6) of the mu, delta, and kappa opioid receptors that are accessible in the binding-site crevices by the substituted cysteine accessibility method (SCAM). We previously showed that ligand binding to the C7.38S mutants of the mu and kappa receptors and the wild-type delta receptor was relatively insensitive to methanethiosulfonate ethylammonium (MTSEA), a positively charged sulfhydryl-specific reagent. These MTSEA-insensitive constructs were used as the templates, and 22 consecutive residues in TM6 (excluding C6.47) of each receptor were mutated to cysteine, 1 at a time. Most mutants retained binding affinities for [3H]diprenorphine, a nonselective opioid antagonist, similar to that of the template receptors. Treatment with MTSEA significantly inhibited [3H]diprenorphine binding to 11 of 22 mutants of the rat mu receptor and 9 of 22 mutants of the human delta receptor and 10 of 22 mutants of the human kappa receptor. Naloxone or diprenorphine protected all sensitive mutants, except the A6.42(287)C mu mutant. Thus, V6.40, F6.44, W6.48, I6.51, Y6.54, V6.55, I6.56, I6.57, K6.58, and A6.59 of the mu receptor; F6.44, I6.51, F6.54, V6.55, I6.56, V6.57, W6.58, T6.59, and L6.60 of the delta receptor; and F6.44, W6.48, I6.51, F6.54, I6.55, L6.56, V6.57, E6.58, A6.59, and L6.60 of the kappa receptor are on the water-accessible surface of the binding-site crevices. The accessibility patterns of residues in the TMs 6 of the mu, delta, and kappa opioid receptors are consistent with the notion that the TMs 6 are in alpha-helical conformations with a narrow strip of accessibility on the intracellular side of 6.54 and a wider area of accessibility on the extracellular side of 6.54, likely due to a proline kink at 6.50 that bends the helix in toward the binding pocket and enables considerable motion in this region. The wider exposure of residues 6.55-6.60 to the binding-site crevice, combined with the divergent amino acid sequences, is consistent with the inferred role of residues in this region in determining ligand binding selectivity. The conservation of the accessibility pattern on the cytoplasmic side of 6.54 suggests that this region may be important for receptor activation. This accessibility pattern is similar to that of the D2 dopamine receptor, the only other GPCR in which TM6 has been mapped by SCAM. That opioid receptors and the remotely related D2 dopamine receptor have similar accessibility patterns in TM6 suggest that these segments of GPCRs in the rhodopsin-like subfamily not only share secondary structure but also are packed similarly into the transmembrane bundle and thus have similar tertiary structure.


Assuntos
Aminoácidos/metabolismo , Receptores Opioides/metabolismo , Substituição de Aminoácidos/genética , Aminoácidos/genética , Animais , Ligação Competitiva/genética , Cisteína/genética , Diprenorfina/metabolismo , Diprenorfina/farmacologia , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/farmacologia , Humanos , Naloxona/farmacologia , Antagonistas de Entorpecentes , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Estrutura Secundária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/genética , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Ratos , Receptores Opioides/genética , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
12.
J Pharmacol Exp Ther ; 297(2): 688-95, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11303059

RESUMO

Buprenorphine (BUP) is an oripavine analgesic that is beneficial in the maintenance treatment of opiate-dependent individuals. Although BUP has been studied extensively, relatively little is known about norbuprenorphine (norBUP), a major dealkylated metabolite of BUP. We now describe the binding of norBUP to opioid and nociceptin/orphanin FQ (ORL1) receptors, and its effects on [(35)S]guanosine-5'-O-(gamma-thio)triphosphate ([(35)S]GTP gamma S) binding mediated by opioid or ORL1 receptors and in the mouse acetic acid writhing test. Chinese hamster ovary cells stably transfected with each receptor were used for receptor binding and [(35)S]GTP gamma S binding. NorBUP exhibited high affinities for mu-, delta-, and kappa-opioid receptors with K(i) values in the nanomolar or subnanomolar range, comparable to those of BUP. NorBUP and BUP had low affinities for the ORL1 receptor with K(i) values in the micromolar range. In the [(35)S]GTP gamma S binding assay, norBUP displayed characteristics distinct from BUP. At the delta-receptor, norBUP was a potent full agonist, yet BUP had no agonist activity and antagonized actions of norBUP and DPDPE. At mu- and kappa-receptors, both norBUP and BUP were potent partial agonists, with norBUP having moderate efficacy and BUP having low efficacy. At the ORL1 receptor, norBUP was a full agonist with low potency, while BUP was a potent partial agonist. In the writhing test, BUP and norBUP both suppressed writhing in an efficacious and dose-dependent manner, giving A(50) values of 0.067 and 0.21 mg/kg, s.c., respectively. These results highlight the similarities and differences between BUP and norBUP, each of which may influence the unique pharmacological profile of BUP.


Assuntos
Analgésicos Opioides/farmacologia , Buprenorfina/farmacologia , Receptores Opioides/agonistas , Ácido Acético , Inibidores de Adenilil Ciclases , Animais , Buprenorfina/análogos & derivados , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colforsina/farmacologia , Cricetinae , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Camundongos , Antagonistas de Entorpecentes , Medição da Dor/efeitos dos fármacos , Receptores Opioides/biossíntese , Receptor de Nociceptina
13.
Biochemistry ; 39(45): 13904-15, 2000 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-11076532

RESUMO

Binding pockets of the opioid receptors are presumably formed among the transmembrane domains (TMDs) and are accessible from the extracellular medium. In this study, we determined the sensitivity of binding of [(3)H]diprenorphine, an antagonist, to mu, delta, and kappa opioid receptors to charged methanethiosulfonate (MTS) derivatives and identified the cysteine residues within the TMDs that conferred the sensitivity. Incubation of the mu opioid receptor expressed in HEK293 cells with MTS ethylammonium (MTSEA), MTS ethyltrimethylammonium (MTSET), or MTS ethylsulfonate (MTSES) inhibited [(3)H]diprenorphine binding with the potency order of MTSEA > MTSET > MTSES. Pretreatment of mu, delta, and kappa opioid receptors with MTSEA dose-dependently inhibited [(3)H]diprenorphine binding with MTSEA sensitivity in the order of kappa > mu >> delta. The effects of MTSEA occurred rapidly, reaching the maximal inhibition in 10 min. (-)-Naloxone, but not (+)-naloxone, prevented the MTSEA effect, demonstrating that the reaction occurs within or in the vicinity of the binding pockets. Each cysteine residue in the TMDs of the three receptors was mutated singly, and the effects of MTSEA treatment were examined. The mutants had similar affinities for [(3)H]diprenorphine, and C7. 38(321)S, C7.38(303)S, and C7.38(315)S mutations rendered mu, delta, and kappa opioid receptors less sensitive to the effect of MTSEA, respectively. These results indicate that the conserved Cys7.38 is differentially accessible in the binding-site crevice of these receptors. The second extracellular loop of the kappa receptor, which contains several acidic residues, appears to play a role, albeit small, in its higher sensitivity to MTSEA, whereas the negative charge of Glu6.58(297) did not. To the best of our knowledge, this is the first report to show that a conserved residue among highly homologous G protein-coupled receptors is differentially accessible in the binding-site crevice. In addition, this represents the first successful generation of MTSEA-insensitive mutants of mu, delta, and kappa opioid receptors, which will allow determination of residues accessible in the binding-site crevices of these receptors by the substituted cysteine accessibility method.


Assuntos
Sequência Conservada , Cisteína/metabolismo , Receptores Opioides/metabolismo , Sequência de Aminoácidos , Animais , Benzomorfanos/metabolismo , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Linhagem Celular , Sequência Conservada/efeitos dos fármacos , Cisteína/genética , Diprenorfina/antagonistas & inibidores , Diprenorfina/metabolismo , Relação Dose-Resposta a Droga , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/farmacologia , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Indicadores e Reagentes , Mesilatos/farmacologia , Metionina/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Naloxona/farmacologia , Antagonistas de Entorpecentes , Estrutura Secundária de Proteína/efeitos dos fármacos , Ratos , Receptores Opioides/genética , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Serina/metabolismo , Fatores de Tempo , Trítio
14.
Mol Pharmacol ; 58(4): 795-801, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10999950

RESUMO

Previously, we showed that the human kappa-opioid receptor (hkor) stably expressed in Chinese hamster ovary (CHO) cells underwent down-regulation after prolonged U50,488H treatment. In the present study, we determined the mechanisms underlying this process. U50, 488H caused a significant down-regulation of the hkor, although etorphine did not. Neither U50,488H nor etorphine caused down-regulation of the rat kappa-opioid receptor. Thus, similar to internalization, there are agonist and species differences in down-regulation of kappa-opioid receptors. Expression of the dominant negative mutants arrestin-2(319-418) or dynamin I-K44A significantly reduced U50,488H-induced down-regulation of the hkor. Coexpression of GRK2 or GRK2 and arrestin-2 permitted etorphine to induce down-regulation of the hkor, although expression of arrestin-2 or dynamin I alone did not. Expression of the dominant negative mutants rab5A-N133I or rab7-N125I blunted U50,488H-induced down-regulation. Pretreatment with lysosomal enzyme inhibitors [(2S, 3S)trans-epoxysuccinyl-L-leucylamido-3-methylbutane ethyl ester or chloroquine] or proteasome inhibitors (proteasome inhibitor I, MG-132, or lactacystin) decreased the extent of U50,488H-induced down-regulation. A combination of chloroquine and proteasome inhibitor I abolished U50,488H-induced down-regulation. These results indicate that U50,488H-induced down-regulation of the hkor involves GRK-, arrestin-2-, dynamin-, rab5-, and rab7-dependent mechanisms and receptors seem to be trafficked to lysosomes and proteasomes for degradation. Thus, U50,488H-induced internalization and down-regulation of the hkor share initial common mechanisms. To the best of our knowledge, these results represent the first report on the involvement of both rab5 and rab7 in agonist-induced down-regulation of a G protein-coupled receptor. In addition, this study is among the first to show the involvement of proteasomes in agonist-induced down-regulation of a G protein-coupled receptor.


Assuntos
Receptores Opioides kappa/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Arrestinas/genética , Arrestinas/fisiologia , Células CHO , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Cisteína Endopeptidases/fisiologia , Regulação para Baixo , Dinamina I , Dinaminas , Etorfina/farmacologia , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/fisiologia , Humanos , Lisossomos/fisiologia , Complexos Multienzimáticos/fisiologia , Mutação , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Pinocitose/efeitos dos fármacos , Pinocitose/fisiologia , Complexo de Endopeptidases do Proteassoma , Ratos , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Transfecção , Quinases de Receptores Adrenérgicos beta , Proteínas rab5 de Ligação ao GTP/fisiologia
15.
Mol Pharmacol ; 58(4): 669-76, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10999935

RESUMO

The steroid SC17599 (17alpha-acetoxy-6-dimethylaminomethyl-21-fluoro-3-ethoxypregna -3, 5-dien-20-one) has mu-opioid actions in vivo. The ability of SC17599 to interact with opioid receptors has been studied using radioligand and [(35)S]guanosine-5'-O-(3-thio)triphosphate (GTPgammaS) binding assays. SC17599 bound to mu-opioid receptors in SH-SY5Y neuroblastoma cells and to recombinant receptors expressed in rat C6 glioma cells and Chinese hamster ovary cells with good affinity and with greater than 100-fold selectivity for mu- over both delta- and kappa-opioid receptors. Binding was much reduced when aspartate 147 in the wild-type mu-opioid receptor was replaced with asparagine. The affinity of SC17599 for the mu-opioid receptor was decreased in the presence of sodium ions, indicating agonist activity. SC17599 stimulated the binding of [(35)S]GTPgammaS in a naloxone-reversible manner with good potency and maximal effect equivalent to that of the mu-opioid agonists fentanyl and [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin. In rat brain membranes, SC17599-mediated stimulation of [(35)S]GTPgammaS binding was reversed by the antagonist naltrexone. SC17599 lacks an aromatic ring and para-hydroxyl substituent considered critical in the pharmacophore for mu-opioids. The structural relationship between SC17599 and more traditional opioid ligands was investigated through genetic algorithm-based modeling techniques for pharmacophore generation (GASP) and ligand-receptor docking (GOLD). The relatively planar and electron-rich A ring of the steroid compensated for the lack of aromaticity. Modeling of ligand-receptor docking showed that both morphine and SC17599 occupy the same binding pocket within the transmembrane helix bundle of the mu-opioid receptor and that the relationship between their binding modes largely mimicked the pharmacophore alignment.


Assuntos
Encéfalo/efeitos dos fármacos , Pregnadienos/farmacologia , Receptores Opioides mu/agonistas , Animais , Encéfalo/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Cobaias , Humanos , Masculino , Modelos Moleculares , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
16.
J Biol Chem ; 275(14): 10443-52, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10744734

RESUMO

Agonist-dependent regulation of G protein-coupled receptors is dependent on their phosphorylation by G protein-coupled receptor kinases (GRKs). GRK2 and GRK3 are selectively regulated in vitro by free Gbetagamma subunits and negatively charged membrane phospholipids through their pleckstrin homology (PH) domains. However, the molecular binding determinants and physiological role for these ligands remain unclear. To address these issues, we generated an array of site-directed mutants within the GRK2 PH domain and characterized their interaction with Gbetagamma and phospholipids in vitro. Mutation of several residues in the loop 1 region of the PH domain, including Lys-567, Trp-576, Arg-578, and Arg-579, resulted in a loss of receptor phosphorylation, likely via disruption of phospholipid binding, that was reversed by Gbetagamma. Alternatively, mutation of residues distal to the C-terminal amphipathic alpha-helix, including Lys-663, Lys-665, Lys-667, and Arg-669, resulted in decreased responsiveness to Gbetagamma. Interestingly, mutation of Arg-587 in beta-sheet 3, a region not previously thought to interact with Gbetagamma, resulted in a specific and profound loss of Gbetagamma responsiveness. To further characterize these effects, two mutants (GRK2(K567E/R578E) and GRK2(R587Q)) were expressed in Sf9 cells and purified. Analysis of these mutants revealed that GRK2(K567E/R578E) was refractory to stimulation by negatively charged phospholipids but bound Gbetagamma similar to wild-type GRK2. In contrast, GRK2(R587Q) was stimulated by acidic phospholipids but failed to bind Gbetagamma. In order to examine the role of phospholipid and Gbetagamma interaction in cells, wild-type and mutant GRK2s were expressed with a beta(2)-adrenergic receptor (beta(2)AR) mutant that is responsive to GRK2 phosphorylation (beta(2)AR(Y326A)). In these cells, GRK2(K567E/R578E) and GRK2(R587Q) were largely defective in promoting agonist-dependent phosphorylation and internalization of beta(2)AR(Y326A). Similarly, wild-type GRK2 but not GRK2(K567E/R578E) or GRK2(R587Q) promoted morphinedependent phosphorylation of the mu-opioid receptor in cells. Thus, we have (i) identified several specific GRK2 binding determinants for Gbetagamma and phospholipids, and (ii) demonstrated that Gbetagamma binding is the limiting step for GRK2-dependent receptor phosphorylation in cells.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP , Fosfolipídeos/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células COS , Linhagem Celular , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfolipídeos/química , Fosforilação , Estrutura Secundária de Proteína , Spodoptera , Transfecção , Quinases de Receptores Adrenérgicos beta
17.
Synapse ; 36(1): 12-20, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10700022

RESUMO

Opiates active at the mu-opiate receptor (MOR) produce antinociception, in part, through actions involving substance P (SP), a peptide present in both unmyelinated primary afferents and interneurons within the dorsal horn. We examined potential functional sites for interactions between SP and MOR by using dual electron microscopic immunocytochemical localization of antisera against SP and a sequence-specific antipeptide antibody against MOR in rat cervical spinal dorsal horn. The distribution was compared with that of the functionally analogous dorsal horn of the trigeminal nucleus caudalis. Many of the SP-immunoreactive terminals in the dorsal horn contacted dendrites that contain MOR (53% in trigeminal; 70% in cervical spinal cord). Conversely, within the cervical spinal dorsal horn 79% of the MOR-labeled dendrites that received any afferent input were contacted by at least one SP-containing axon or terminal. Although SP-immunoreactive dendrites were rare, many of these (48%) contained MOR, suggesting that the activity of SP-containing spinal interneurons may be regulated by MOR ligands. A few SP-labeled terminals also contained MOR (12% in trigeminal; 6% in cervical spinal cord). These data support the idea that MOR ligands produce antinociception primarily through modulation of postsynaptic second-order nociceptive neurons in the dorsal horns of spinal cord and spinal trigeminal nuclei, some of which contain SP. They also suggest, however, that in each region, MOR agonists can act presynaptically to control the release of SP and/or glutamate from afferent terminals. The post- and presynaptic MOR sites are likely to account for the potency of MOR agonists as analgesics.


Assuntos
Células do Corno Posterior/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/ultraestrutura , Substância P/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/ultraestrutura , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Masculino , Microscopia Eletrônica , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/ultraestrutura , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Núcleo Espinal do Trigêmeo/citologia , Núcleo Espinal do Trigêmeo/efeitos dos fármacos , Núcleo Espinal do Trigêmeo/metabolismo
18.
Eur J Pharmacol ; 377(2-3): 223-31, 1999 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10456435

RESUMO

The role of nucleoside diphosphate kinase (NDKP), which converts GDP to GTP, in the coupling of mu-opioid receptors to G protein was investigated in membranes of Chinese hamster ovary cells stably transfected with the cloned rat mu-opioid receptor (rmor). Endogenous NDPK activity in membranes was determined to be 0.60+/-0.02 micromol/mg protein/30 min UDP (at 10 mM), a competitive substrate of NDPK for GDP with no effect on guanine nucleotide binding to G proteins, reduced basal [35S]GTPgammaS binding and unmasked morphine-stimulated [35S]GTPgammaS binding to pertussis toxin-sensitive G proteins, indicating that [35S]GTPgammaS binding to NDPK accounts for part of its high basal binding. UDP increased the extent of morphine-induced increase in [35S]GTPgammaS binding in the presence of GDP, most likely by reducing basal binding and inhibiting conversion of GDP to GTP. ATP greatly reduced morphine-induced increase in [35S]GTPgammaS binding, whereas AMP-PCP (adenylyl-(beta,gamma-methylene)-diphosphoate tetralithium salt), which cannot serve as the phosphate donor for NDPK, did not, demonstrating that effects of ATP is mediated by the NDPK product GTP. In addition, GDP and ATP increased the Kd and lowered the Bmax of the agonist [3H]DAMGO ([D-Ala2,N-Me-Phe4,Gly5ol]-Enkephalin) for the mu-opioid receptor and GDP alone increased Kd, most likely through their conversion to GTP by NDPK. Addition of exogenous NDPK enhanced the inhibitory effects of GDP and combined GDP and ATP on [3H]DAMGO binding. Thus, NDPK appears to play a role in modulating signal transduction of and agonist binding to mu-opioid receptors.


Assuntos
Membrana Celular/fisiologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Núcleosídeo-Difosfato Quinase/fisiologia , Receptores Opioides mu/metabolismo , Receptores Opioides mu/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Analgésicos Opioides/metabolismo , Animais , Células CHO , Células Cultivadas , Clonagem Molecular , Cricetinae , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Nucleotídeos de Guanina/metabolismo , Guanosina Difosfato/metabolismo , Toxina Pertussis , Ratos , Receptores Opioides mu/agonistas , Difosfato de Uridina/farmacologia , Fatores de Virulência de Bordetella/farmacologia
19.
Life Sci ; 65(2): 175-85, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10416823

RESUMO

We tested the hypotheses that the carboxylate side chain of Asp147 of the mu opioid receptor interacts with the protonated nitrogen of naltrexone and morphine and that this interaction is important for pharmacological properties of the two compounds. Mutation of Asp147 to Ala or Asn substantially reduced the affinity of naltrexone and the affinity, potency and efficacy of morphine, while the Glu mutant had similar properties as the wildtype, indicating the significant role of the carboxylate group of Asp147 in receptor binding and activation. This role could be due to its direct interaction with ligands or involvement in interhelical interactions. The unprotonated analogs of naltrexone and morphine, cyclopropylcarbonyl noroxymorphone (CPCNOM) and N-formylnormorphine (NFNM), respectively, were used to discriminate between these mechanisms. CPCNOM was much less potent as an antagonist and had substantially lower affinity for the mu receptor than naltrexone. Similarly, NFNM was unable to activate the mu receptor and had much lower affinity than morphine. These results indicate the importance of the protonated nitrogen. Notably, the D147A and D147N mutations did not appreciably affect the binding affinities of CPCNOM and NFNM. In addition, the D147E mutant had similar affinities for CPCNOM and NFNM as the D147A and D147N mu receptors. Thus, the carboxylate group of Asp147 is not important for binding of the two unprotonated compounds. These results indicate that the carboxylate group of Asp147 of the mu receptor interacts directly with the protonated nitrogen of naltrexone and morphine and this interaction is important for binding and receptor activation.


Assuntos
Ácido Aspártico/química , Morfina/química , Naltrexona/química , Receptores Opioides mu/química , Animais , Ácido Aspártico/genética , Células CHO , Ácidos Carboxílicos/metabolismo , Cricetinae , Relação Dose-Resposta a Droga , Ligantes , Mutagênese Sítio-Dirigida , Ligação Proteica , Ratos , Receptores Opioides mu/metabolismo
20.
FEBS Lett ; 447(2-3): 318-24, 1999 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-10214970

RESUMO

We examined whether a proposed spatial proximity between Asp114(2.50) and Asn332(7.49) affected the functional properties of the mu opioid receptor. The D114(2.50)N mutant had reduced binding affinities for morphine, DAMGO and CTAP, but not for naloxone and [3H]diprenorphine; this mutation also abolished agonist-induced increase in [35S]GTPgammaS binding. The N332(7.49)D mutation eliminated detectable binding of either [3H]diprenorphine or [3H]DAMGO. The combined D114(2.50)N-N332(7.49)D mutation restored high affinity binding for [3H]diprenorphine, CTAP and naloxone, and restored partially the binding affinities, potencies and efficacies of morphine and DAMGO. Thus, reciprocal mutations of Asp114(2.50) and Asn332(7.49) compensate for the detrimental effects of the single mutations, indicating that the residues are adjacent in space and that their chemical functionalities are important for ligand binding and receptor activation.


Assuntos
Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Animais , Asparagina/química , Ácido Aspártico/química , Sítios de Ligação/genética , Células CHO , Cricetinae , Diprenorfina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Técnicas In Vitro , Cinética , Mutagênese Sítio-Dirigida , Naloxona/metabolismo , Fragmentos de Peptídeos , Peptídeos/metabolismo , Estrutura Secundária de Proteína , Ratos , Receptores Opioides mu/genética , Somatostatina , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA