Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Salvador; s.n; 2015. 132 p. ilus, tab.
Tese em Português | LILACS | ID: biblio-1000963

RESUMO

Alguns estudos sugerem que as vias opioidérgicas centrais parecem desempenhar um papel regulatório no controle da ingestão de água e sal em mamíferos. As ações dos opioides centrais sobre a regulação do controle hidroeletrolítico são mediadas por vários dos subtipos de receptores opioides. O papel dos receptores delta e kappa-opioides centrais neste processo não está adequadamente elucidado sendo necessário mais estudos que o esclareçam. Objetivo: Este estudo investigou o envolvimento dos receptores delta e kappa-opioides centrais no apetite por sódio em ratos depletados deste íon e em rato ativados centralmente com angiotensina. Material e Métodos: Foram utilizados ratos Wistar (270 ± 20 g), submetidos à cirurgia estereotáxica para implante de cânula guia no ventrículo lateral esquerdo (VL), no órgão subfornical (OSF), no núcleo preóptico mediano (MnPO) e no núcleo basolateral da amígdala (BLA). No protocolo de depleção de sódio os animais foram submetidos à injeção subcutânea de furosemida combinada com dieta hipossódica quatro dias após a cirurgia. Neste modelo de estudo os animais receberam injeção intracerebroventricular (i.c.v.) do antagonista delta-opioide naltrindole no quinto dia pós-cirúrgico, nas doses de 5, 10 e 20 nmol/2 μL e do antagonista kappa-opioide, norbinaltorfimina, injetado no OSF, MnPO e BLA, nas doses de 0,5, 1,0 e 2,0 nmol/0,2 μL...


Central opioid pathways seem to have an important role on the control of water and salt intake in mammals, and brain opioid peptides may influence hydroelectrolyte balance through a myriad of actions mediated by distinct opioid receptors. The specific role of central delta and kappa-opioid receptors (DOR and KOR) in this process is far from being fully understood. In the present work, we investigated the role of those receptors in the control of water and salt intake, in sodium-depleted rats and rats with activation central angiotensinergic. Method: Wistar male rats (250 ± 20 g) were used in the experiment after stereotaxic cannulation of the VL left, SFO, MnPO and BLA. To study the effect of the blockade of central DOR and KOR on water and salt intake in rats were sodium depleted by the concomitant use of s.c. injections of furosemide and were kept in hypossodic diet, five days after surgery. In the sixth day, they received i.c.v. injections of a selective delta-opioid receptor antagonist (naltrindole) at the doses of 5, 10 and 20 nmol/2 μL and injections in the SFO, MnPO and BLA of a selective kappa-opioid receptor antagonist (norbinaltorphimine) at the doses of 0.5, 1.0 and 2.0 nmol/0.2 μL...


Assuntos
Animais , Apetite , Apetite/fisiologia , Apetite/imunologia , Receptores Opioides delta/análise , Receptores Opioides delta/classificação , Receptores Opioides delta/isolamento & purificação , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/análise , Receptores Opioides kappa/imunologia
2.
Neurochem Int ; 59(2): 192-201, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21672586

RESUMO

The goal of our work was a throughout characterization of the pharmacology of the TIPP-analog, Tyr-Tic-(2S,3R)-ß-MePhe-Phe-OH and see if putative δ-opioid receptor subtypes can be distinguished. Analgesic latencies were assessed in mouse tail-flick assays after intrathecal administration. In vitro receptor autoradiography, binding and ligand-stimulated [(35)S]GTPγS functional assays were performed in the presence of putative δ(1)-(DPDPE: agonist, BNTX: antagonist), δ(2)-(agonist: deltorphin II, Ile(5,6)-deltorphin II, antagonist: naltriben) and µ-(DAMGO: agonist) opioid ligands. The examined antagonist inhibited the effect of DPDPE by 60%, but did not antagonize δ(2)- and µ-agonist induced analgesia. The radiolabeled form identified binding sites with K(D)=0.18 nM and receptor densities of 102.7 fmol/mg protein in mouse brain membranes. The binding site distribution of the [(3)H]Tyr-Tic-(2S,3R)-ß-MePhe-Phe-OH agreed well with that of [(3)H]Ile(5,6)-deltorphin II as revealed by receptor autoradiography. Tyr-Tic-(2S,3R)-ß-MePhe-Phe-OH displayed 2.49±0.06 and 0.30±0.01 nM potency against DPDPE and deltorphin II in the [(35)S]GTPγS functional assay, respectively. The rank order of potency of putative δ(1)- and δ(2)-antagonists against DPDPE and deltorphin was similar in brain and CHO cells expressing human δ-opioid receptors. Deletion of the DOR-1 gene resulted in no residual binding of the radioligand and no significant DPDPE effect on G-protein activation. Tyr-Tic-(2S,3R)-ß-MePhe-Phe-OH is a highly potent and δ-opioid specific antagonist both in vivo and in vitro. However, the putative δ(1)- and δ(2)-opioid receptors could not be unequivocally distinguished in vitro.


Assuntos
Analgésicos Opioides/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Tetra-Hidroisoquinolinas/farmacologia , Animais , Autorradiografia , Células CHO , Cricetinae , Cricetulus , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/química , Receptores Opioides delta/classificação , Tetra-Hidroisoquinolinas/química
3.
Am J Physiol Heart Circ Physiol ; 291(5): H2246-54, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16782849

RESUMO

Ultra-low-dose methionine-enkephalin-arginine-phenylalanine improves vagal transmission (vagotonic) and decreases heart rate via delta(1)-opioid receptors within the sinoatrial (SA) node. Higher doses activate delta(2)-opioid receptors, interrupt vagal transmission (vagolytic), and reduce the bradycardia. Preconditioning-like occlusion of the nodal artery produced a vagotonic response that was reversed by the delta(1)-antagonist 7-benzylidenaltrexone (BNTX). The following study tested the hypothesis that extended delta(1)-opioid receptor stimulation reduces subsequent delta(2)-receptor responses. The delta(2)-agonist deltorphin II was introduced in the SA node by microdialysis to evaluate delta(2) responses before and after infusion of the delta(1)-agonist TAN-67. TAN-67 reduced the vagolytic effect of deltorphin by two-thirds. When the delta(1)-antagonist BNTX was combined with TAN-67, the deltorphin response was preserved, suggesting that attrition of the prior response was mediated by delta(1) activity. When TAN-67 was omitted in time control studies, some loss of delta(2) responses was apparent in the absence of the delta(1) treatment. This loss was also eliminated by BNTX, suggesting that the attenuation of the response after deltorphin alone was also the result of delta(1) activity. Additional studies tested TAN-67 alone in the absence of prior deltorphin. When time controls were conducted without the initial deltorphin treatment, a robust vagolytic response was observed. When TAN-67 preceded the delayed deltorphin, the vagolytic response was eroded, indicating an independent effect of TAN-67. BNTX infused afterward was unable to restore the delta(2) response. These data support the conclusion that the loss of the delta(2) response resulted from reduced delta(2) activity mediated by continued delta(1)-receptor stimulation and not the arithmetic consequence of increased competition from that same delta(1) receptor.


Assuntos
Receptores Opioides delta/metabolismo , Nó Sinoatrial/efeitos dos fármacos , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Bradicardia/tratamento farmacológico , Bradicardia/fisiopatologia , Cães , Relação Dose-Resposta a Droga , Encefalina Metionina/análogos & derivados , Encefalina Metionina/farmacologia , Feminino , Masculino , Microdiálise , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Quinolinas/farmacologia , Receptores Opioides delta/classificação , Receptores Opioides delta/efeitos dos fármacos , Nó Sinoatrial/inervação , Nó Sinoatrial/fisiologia , Estimulação Química , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
4.
Psychopharmacology (Berl) ; 182(3): 327-34, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16075288

RESUMO

RATIONALE: Recent studies have shown that endogenous opioid systems are associated with the regulation of emotional responses. In particular, it has been reported that delta-opioid receptors act naturally to inhibit stress and anxiety. OBJECTIVE: The present study was designed to examine the possible involvement of opioid delta-receptor subtypes in the anxiety-related behavior in the elevated-plus-maze test. METHODS: Six-week-old male Lewis rats were used. The total numbers of visits to the closed and open arms and the cumulative time spent and visits in the open arms were determined. Plasma corticosterone levels were measured by enzyme immunoassay. RESULTS: Naltrindole (NTI), a delta-opioid receptor antagonist (3 mg/kg s.c.), induced a significant decrease in the percentages of time spent and visits in the open arms. Naltriben (NTB), a delta2-opioid receptor antagonist (3 mg/kg s.c.), but not 7-benzylidenenaltrexone, a delta1-opioid receptor antagonist, produced similar anxiety-related behaviors in the elevated plus-maze. These effects of NTI and NTB were antagonized by pretreatment with (+)-4-[(aR)-a-((2S,5R)-4-allyl-2,5-dimethyl-1piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80), a delta-opioid receptor agonist. Furthermore, after exposure to the elevated plus-maze, the maximal increase in the plasma corticosterone level in NTI-treated rats was clearly higher than that in vehicle-treated rats. However, when NTI and SNC80 were coadministered, higher levels of plasma corticosterone were not seen after exposure to the elevated plus-maze. CONCLUSION: These results suggest that endogenous delta2-opioid-receptor-mediated systems are involved in the regulation of anxiety-related behaviors and might play a physiologically important role in the regulation of adrenocortical activity.


Assuntos
Ansiedade/psicologia , Aprendizagem em Labirinto/efeitos dos fármacos , Receptores Opioides delta/fisiologia , Animais , Benzamidas/farmacologia , Compostos de Benzilideno/farmacologia , Coleta de Amostras Sanguíneas , Corticosterona/sangue , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Piperazinas/farmacologia , Ratos , Ratos Endogâmicos Lew , Receptores Opioides delta/classificação
5.
Brain Res ; 1049(1): 61-9, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15936000

RESUMO

Despite insights into an increasingly significant role for delta opioid receptors in thermoregulation, it is unclear whether delta receptors located in the brain or periphery play the more critical role in body temperature regulation. Moreover, it is not entirely clear which delta receptor phenotype, delta1 or delta2, mediates the hypothermic actions of delta agonists. Because SNC-80 distributes into central and peripheral compartments and produces rapid hypothermia following systemic injection, the nonpeptide delta agonist is particularly useful in discriminating the site of action of delta receptor-mediated hypothermia. To determine the locus and phenotype of delta receptor which mediates SNC-80-induced hypothermia, we injected SNC-80 and phenotype selective delta antagonists to male Sprague-Dawley rats. SNC-80 (10-50 mg/kg, im) evoked hypothermia that peaked 30 min post-injection. Naltrexone (5 mg/kg, sc), an opioid antagonist, or naltrindole (5 mg/kg, sc), a delta antagonist, blocked the hypothermic response to SNC-80 (35 mg/kg, im). The hypothermia caused by SNC-80 (35 mg/kg, im) was blocked by a delta2 antagonist, naltriben (2.5 mg/kg, sc), but was not affected by BNTX (5 and 10 mg/kg, sc), a delta1 antagonist. The administration of naltriben (10 microg/rat, icv) 30 min before SNC-80 (35 mg/kg, im) prevented SNC-80-evoked hypothermia. In contrast, methylnaltrexone (5 mg/kg, sc), a peripherally restricted opioid antagonist, did not affect the hypothermia caused by SNC-80. The present data demonstrate that selective activation of brain delta2 receptors is a major mechanism of SNC-80-evoked hypothermia in rats.


Assuntos
Benzamidas/administração & dosagem , Regulação da Temperatura Corporal/efeitos dos fármacos , Regulação da Temperatura Corporal/fisiologia , Encéfalo/efeitos dos fármacos , Hipotermia/fisiopatologia , Piperazinas/administração & dosagem , Receptores Opioides delta/agonistas , Receptores Opioides delta/fisiologia , Animais , Área Sob a Curva , Relação Dose-Resposta a Droga , Hipotermia/induzido quimicamente , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/classificação
6.
Shock ; 23(3): 264-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15718926

RESUMO

delta opioid receptor agonists exert potent hemodynamic effects under ischemic conditions. This study was designed to assess the cardiovascular effects of Deltorphin-D(variant) (Delt-D(var)), a selective delta(2) opioid receptor agonist, in conscious, freely moving male rats during the posthemorrhage, recompensatory phase of a hemorrhagic trauma. Rats were fitted with femoral arterial and venous catheters for measurements of mean arterial pressure (MAP), heart rate (HR), and intravenous (i.v.) injections of isotonic saline, 1 mg/kg Delt-D(var), or 2 mg/kg Delt-D(var). During hemorrhaging, 30% (approximately 5 mL) of total blood volume was collected from the arterial catheter. MAP-HR was fitted to a logistic equation to determine baroreceptor reflex properties. Hemorrhaged rats progressed through three distinct phases: compensation, decompensation, and recompensation. Saline and 1 mg/kg Delt-D(var) rats treated posthemorrhage had similar MAP and HR after hemorrhage. In contrast, 2 mg/kg Delt-D(var) administered after hemorrhaging led to a faster and more complete recovery of MAP than compared with the other groups. In hemorrhaged rats, the average HR gain (bpm/mmHg) after 2 mg/kg Delt-D(var) treatment was greater and the BP(50) (BP at one-half the HR range) was significantly lower than after saline treatment. The results show that after hemorrhage, during the recompensatory period, stimulation of delta(2) opioid receptors leads to improved MAP, and this recovery may involve a change in baroreflex sensitivity.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hemorragia/tratamento farmacológico , Hemorragia/fisiopatologia , Receptores Opioides delta/agonistas , Sequência de Aminoácidos , Animais , Frequência Cardíaca/efeitos dos fármacos , Masculino , Oligopeptídeos/química , Oligopeptídeos/uso terapêutico , Ratos , Receptores Opioides delta/classificação
7.
J Cardiovasc Pharmacol Ther ; 6(4): 385-93, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11907641

RESUMO

BACKGROUND: Methionine-enkephalin-arginine-phenylalanine (MEAP) is an endogenous opiate derived from the C-terminal sequence of the larger precursor molecule proenkephalin. This heptapeptide is abundant in the myocardium and has significant vagolytic activity when infused systemically. MEAP interrupted vagal bradycardia when it was delivered directly into the sinoatrial node by local microdialysis. This study was conducted to determine the opioid receptor responsible for the vagolytic effect of MEAP. METHODS AND RESULTS: Microdialysis probes were placed in the sinoatrial node of mongrel dogs and perfused at 5 microL/min. Increasing doses of MEAP were included in the nodal perfusate and approximately two thirds of the vagal bradycardia was inhibited with a maximal effect at 0.3 nmoles/microL and a half-maximal response near 0.1 nmoles/microL. When deltorphin II (a delta opioid receptor agonist) was infused into the sinoatrial node, more than 95% of the vagal bradycardia was eliminated at 0.3 nmoles/microL with the half-maximal response near 0.1 nmoles/microL, indicating that deltorphin II was more efficacious than MEAP. The maximal deltorphin II and MEAP effects were both similarly reversed by the paired infusion of increasing doses of the delta opiate receptor antagonist, naltrindole. Selected mu (endomorphin, super DALDA) and kappa (dynorphin, U50488) receptor agonists and mu (CTAP) and kappa (norBNI) receptor antagonists were completely ineffective in this system. CONCLUSIONS: These data suggest that the vagolytic effect of MEAP involves the activation of delta opiate receptors within the sinoatrial node.


Assuntos
Bradicardia/fisiopatologia , Receptores Opioides delta/fisiologia , Nó Sinoatrial/fisiologia , Nervo Vago/fisiologia , Animais , Cães , Encefalina Metionina/análogos & derivados , Encefalina Metionina/farmacologia , Encefalinas/química , Encefalinas/farmacologia , Microdiálise , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/química , Precursores de Proteínas/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/classificação , Nó Sinoatrial/efeitos dos fármacos , Simpatomiméticos/farmacologia , Nervo Vago/efeitos dos fármacos
8.
Biochem Pharmacol ; 59(8): 915-25, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10692556

RESUMO

The two pharmacological delta-opioid receptor subtypes, delta1 and delta2, have been defined on the basis of pharmacological tools but remain to be characterized at the molecular level, since only a single cDNA has been cloned. The present study aimed to investigate the pharmacological properties of delta1- and delta2-opioid subtypes expressed in the human neuroblastoma cell line SK-N-BE and to characterize their putative corresponding mRNAs. Binding experiments using "selective" delta1- and delta2-opioid agonists and antagonists revealed the presence of two binding sites, demonstrating the presence of these delta1-opioid subtypes as they were previously described. The activation of these pharmacological subtypes by the selective agonists induced the incorporation of [alpha-(32)P]azidoanilide-GTP into Galpha(i2)/Galpha(0) subunits with the same efficiency and potency and inhibited adenosine 3', 5'-cyclic monophosphate (cAMP) accumulation with similar efficiency, while their sustained activation for 15 min induced a cross-desensitization. The "selective" delta1 and delta2 antagonists, 7-benzylidenenaltrexone and naltrindole benzofuran, respectively, were found to be as potent in blocking the inhibition of cAMP accumulation induced by both [D-Pen(2,5)]enkephalin and Tyr-D-Ala-Phe-Asp-Val-Val-Gly-NH(2). The possibility that delta-opioid subtypes could arise from alternative splicing was ruled out by reverse transcription-polymerase chain reaction (RT-PCR) experiments and the sequencing of PCR products, which revealed the presence of a single transcript encoding for the delta-opioid receptor. Different possibilities which could account for the delta-opioid receptor heterogeneity observed in the SN-N-BE cell line are discussed.


Assuntos
D-Penicilina (2,5)-Encefalina/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Analgésicos Opioides/farmacologia , Ligação Competitiva/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Humanos , Neuroblastoma , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/classificação , Receptores Opioides delta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
9.
Life Sci ; 65(16): PL233-9, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10573190

RESUMO

The ability of selective mu- ([D-Ala2, NHPhe4, Gly-ol]enkephalin: DAMGO), delta1- ([D-Pen2, Pen5]enkephalin: DPDPE) and delta2- ([D-Ala2]deltorphin II: DELT II) opioid receptor agonists to activate G-proteins in the midbrain and forebrain of mice and rats was examined by monitoring the binding of guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTPgammaS). The levels of [35S]GTPgammaS binding stimulated by DAMGO in the mouse and rat midbrain were significantly greater than those by DPDPE or DELT II. However, relatively lower levels of stimulation of [35S]GTPgammaS binding by all of the agonists than would have been predicted from the receptor densities were observed in either the limbic forebrain or striatum of mice and rats. The effects of DAMGO, DPDPE and DELT II in all three regions were completely reversed by selective mu-, delta1- and delta2-antagonists, respectively. The results indicate that the levels of mu-, delta1- and delta2-opioid receptor agonist-induced G-protein activation in the midbrain are in good agreement with the previously determined distribution densities of each receptor type. Furthermore, the discrepancies observed in the forebrain might reflect differential catalytic efficiencies of receptor-G-protein coupling.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Mesencéfalo/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Oligopeptídeos/farmacologia , Especificidade de Órgãos/efeitos dos fármacos , Prosencéfalo/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/classificação , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Radioisótopos de Enxofre
10.
Prog Neurobiol ; 57(4): 377-420, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10080383

RESUMO

The deltorphins are a class of highly selective delta-opioid heptapeptides from the skin of the Amazonian frogs Phyllomedusa sauvagei and P. bicolor. The first of these fascinating peptides came to light in 1987 by cloning of the cDNA of from frog skins, while the other members of this family were identified either by cDNA or isolation of the peptides. The distinctive feature of deltorphins is the presence of a naturally occurring D-enantiomer at the second position in their common N-terminal sequence, Tyr-D-Xaa-Phe, comparable to dermorphin, which is the prototype of a group of mu-selective opioids from the same source. The D-amino acid and the anionic residues, either Glu or Asp, as well as their unique amino acid compositions are responsible for the remarkable biostability, high delta-receptor affinity, bioactivity and peptide conformation. This review summarizes a decade of research from many laboratories that defined which residues and substituents in the deltorphins interact with the delta-receptor and characterized pharmacological and physiological activities in vitro and in vivo. It begins with a historical description of the topic and presents general schema for the synthesis of peptide analogues of deltorphins A, B and C as a means to document the methods employed in producing a myriad of analogues. Structure activity studies of the peptides and their pharmacological activities in vitro are detailed in abundantly tabulated data. A brief compendium of the current level of knowledge of the delta-receptor assists the reader to appreciate the rationale for the design of these analogues. Discussion of the conformation of these peptides addresses how structure leads to further hypotheses regarding ligand receptor interaction. The review ends with a broad discussion of the potential applications of these peptides in clinical and therapeutic settings.


Assuntos
Oligopeptídeos/química , Oligopeptídeos/fisiologia , Sequência de Aminoácidos , Animais , Anuros , Sítios de Ligação , Humanos , Entorpecentes/administração & dosagem , Entorpecentes/farmacologia , Oligopeptídeos/genética , Oligopeptídeos/farmacologia , Conformação Proteica , Receptores Opioides delta/classificação , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Sistemas do Segundo Mensageiro
11.
Br J Pharmacol ; 126(2): 451-6, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10077238

RESUMO

1. The ability of mu-opioid receptor agonists to activate G-proteins in the spinal cord of mu-opioid receptor knockout mice was examined by monitoring the binding to membranes of the non-hydrolyzable analogue of GTP, guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTPgammaS). 2. In the receptor binding study, Scatchard analysis of [3H][D-Ala2,NHPhe4,Gly-ol]enkephalin ([3H]DAMGO; mu-opioid receptor ligand) binding revealed that the heterozygous mu-knockout mice displayed approximately 40% reduction in the number of mu-receptors as compared to the wild-type mice. The homozygous mu-knockout mice showed no detectable mu-binding sites. 3. The newly isolated mu-opioid peptides endomorphin-1 and -2, the synthetic selective mu-opioid receptor agonist DAMGO and the prototype of mu-opioid receptor agonist morphine each produced concentration-dependent increases in [35S]GTPgammaS binding in wild-type mice. This stimulation was reduced by 55-70% of the wild-type level in heterozygous, and virtually eliminated in homozygous knockout mice. 4. No differences in the [35S]GTPgammaS binding stimulated by specific delta1- ([D-Pen2,5]enkephalin), delta2-([D-Ala2]deltorphin II) or kappa1-(U50,488H) opioid receptor agonists were noted in mice of any of the three genotypes. 5. The data clearly indicate that mu-opioid receptor gene products play a key role in G-protein activation by endomorphins, DAMGO and morphine in the mouse spinal cord. They support the idea that mu-opioid receptor densities could be rate-limiting steps in the G-protein activation by mu-opioid receptor agonists in the spinal cord. These thus indicate a limited physiological mu-receptor reserve. Furthermore, little change in delta1-, delta2- or kappa1-opioid receptor-G-protein complex appears to accompany mu-opioid receptor gene deletions in this region.


Assuntos
Proteínas de Ligação ao GTP/efeitos dos fármacos , Receptores Opioides mu/agonistas , Medula Espinal/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina , D-Penicilina (2,5)-Encefalina , Encefalinas/metabolismo , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Membranas/efeitos dos fármacos , Membranas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Morfina/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/classificação , Receptores Opioides kappa/agonistas , Medula Espinal/metabolismo , Radioisótopos de Enxofre , Trítio
12.
Methods Find Exp Clin Pharmacol ; 21(9): 595-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10669903

RESUMO

The purpose of this study was to elucidate the involvement of spinal delta-opioid receptor subtypes in forced walking stress-induced antinociception mice. We first confirmed that forced walking stress produced walking duration-dependent antinociception in mice as determined by the tail-flick test. Intrathecal treatment with 7-benzylidenenaltrexone, a selective delta 1-opioid receptor antagonist, significantly attenuated forced walking stress-induced antinociception. In contrast, intrathecal treatment with naltriben, a selective delta 2-opioid receptor antagonist, had no significant effect on forced walking stress-induced antinociception. Intracerebroventricular treatment with either 7-benzylidenenaltrexone or naltriben had no effect on the forced walking stress-induced antinociception. These results suggest that forced walking stress-induced antinociception is mediated by spinal delta 1-opioid receptors in mice.


Assuntos
Analgésicos/farmacologia , Compostos de Benzilideno/farmacologia , Naltrexona/análogos & derivados , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/fisiologia , Animais , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor , Receptores Opioides delta/classificação , Estresse Fisiológico/patologia , Fatores de Tempo , Caminhada/fisiologia
13.
Cardiovasc Drugs Ther ; 12(4): 365-73, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9825182

RESUMO

The present study aimed to examine (1) whether the role of the opioid receptor in ischemic preconditioning (PC) is consistent regardless of the duration of ischemic insult and (2) which opioid receptor subtype contributes to PC. In the first series of experiments, the effects of PC, a nonselective opioid receptor antagonist (naloxone), and their combination on the infarct size after various durations of ischemia were assessed. In anesthetized, open-chest rats, the coronary artery was occluded for 20, 30, or 40 minutes to induce infarction and was reperfused for 3 hours, PC was performed with two cycles of 5-minute ischemia followed by 5-minute reperfusion before the sustained ischemia. At 25 minutes before the ischemia, naloxone was injected alone or in combination with subsequent PC. Infarct size was determined by tetrazolium staining and was expressed as a percentage of the risk area size (%IS/RA). In the second series of experiments, the effects of a delta-receptor-selective antagonist, naltrindole (NTI), and a kappa-receptor selective antagonist, nor-binaltrophimine (nor-BNI), on PC before 30-minute coronary occlusion were assessed. In untreated controls, %IS/RA was 53.1 +/- 3.2 after 20 minutes, 67.9 +/- 3.9 after 30 minutes, and 87.8 +/- 2.0 after 40 minutes of ischemia, respectively. PC significantly reduced %IS/RA after 20, 30, and 40 minutes of ischemia to 3.1 +/- 0.8, 12.8 +/- 1.1, and 42.1 +/- 4.3, respectively (P < 0.05 vs. each control). Naloxone (6 mg/kg) partially attenuated the protection afforded by PC when the sustained ischemia was 30 minutes (%IS/RA = 27.4 +/- 4.5; P < 0.05 vs. PC), but this inhibitory effect of naloxone was not detected when the duration of the ischemia was 20 or 40 minutes. NTI (10 mg/kg) also attenuated infarct size limitation by PC after 30 minutes of ischemia (%IS/RA = 25.6 +/- 3.7), but nor-BNI (10 mg/kg) failed to modify infarct size limitation by PC (%IS/RA = 13.3 +/- 3.2). The present results suggest that activation of the opioid delta-receptor partly contributes to preconditioning against infarction in the rat and that there may be a time window (at around 30 minutes after the onset of ischemia) for this opioid receptor-mediated protective mechanism.


Assuntos
Coração/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/patologia , Naloxona/farmacologia , Receptores Opioides delta/fisiologia , Animais , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/classificação , Fatores de Tempo
14.
Eur J Pharmacol ; 354(1): R3-5, 1998 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-9726640

RESUMO

In this study we demonstrate allograft survival in a rat model of renal transplantation using the delta1-opioid receptor antagonist, 7-(benzospiroindanyl)naltrexone [corrected]. Treatment with 7-(benzospiroindanyl)naltrexone [corrected] caused 50% of the rats to survive longer than 100 days (untreated, 11 +/- 3 days). Naltrindole, a delta-opioid receptor antagonist without subtype selectivity, also promoted graft survival but was substantially less effective, suggesting that antagonism at delta1-opioid receptors is involved in allograft survival.


Assuntos
Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Rim/imunologia , Naltrexona/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Compostos de Espiro/farmacologia , Animais , Relação Dose-Resposta a Droga , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Endogâmicos BUF , Ratos Endogâmicos Lew , Receptores Opioides delta/classificação
15.
J Pharmacol Exp Ther ; 285(2): 820-7, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9580632

RESUMO

To analyze the selectivity of delta receptor subtypes to regulate different classes of G proteins, the expression of the alpha-subunits of Gi2, Gi3, Go1, Go2, Gq and G11 transducer proteins was reduced by administration of oligodeoxynucleotides (ODNs) complementary to sequences in their respective mRNAs. Mice receiving antisense ODNs to Gi2 alpha, Gi3 alpha, Go2 alpha and G11 alpha subunits showed an impaired antinociceptive response to all the delta agonists evaluated. An ODN to Go1 alpha specifically blocked the antinociceptive effect of the agonist of delta-1 receptors, [D-Pen2,5]enkephalin (DPDPE), without altering the activity of [D-Ala2]deltorphin II or [D-Ser2]-Leu-enkephalin-Thr (DSLET). In mice treated with an ODN to Gq alpha, the effects of the agonists of delta-2-opioid receptors were reduced, but not those of DPDPE. Thus, Go1 proteins are selectively linked to delta-1-mediated analgesia, and Gq proteins are related to delta-2-evoked antinociception. After impairing the synthesis of Go1 alpha subunits, DPDPE exhibited an antagonistic activity on the antinociception produced by [D-Ala2]deltorphin II. After treatment with ODNs complementary to sequences in Gq alpha or PLC-beta 1 mRNAs, the analgesic capacity of [D-Ala2]deltorphin II was diminished. However, the delta-2-agonist did not alter the antinociceptive activity of DPDPE. An ODN complementary to nucleotides 7 to 26 of the murine delta receptor reduced the analgesic potency of [D-Ala2]deltorphin II, but not that observed for DPDPE. In these mice, [D-Ala2]deltorphin II did not antagonize the effect of DPDPE. These results suggest the existence of different molecular forms of the delta opioid receptor, and the involvement of inositol-signaling pathways in the supraspinal antinociceptive effects of delta agonists.


Assuntos
Analgesia , Proteínas de Ligação ao GTP/análise , Inositol/metabolismo , Receptores Opioides delta/fisiologia , Analgésicos Opioides/farmacologia , Animais , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/imunologia , Masculino , Camundongos , Oligonucleotídeos Antissenso/farmacologia , Receptores Opioides delta/classificação , Fosfolipases Tipo C/fisiologia
16.
Life Sci ; 62(16): PL253-8, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9585172

RESUMO

The delta(delta)-opioid agonists [D-Pen2,5]enkephalin (DPDPE) and [D-Ala2]deltorphin II increased the formation of inositol phosphates (IPs) in mice periaqueductal gray matter (PAG) slices pre-labeled with myo-[3H]inositol. Both delta-agonists caused an increase in IP accumulation in a dose-dependent manner (1-100 microM) and which was pertussis toxin (0.5 microg/mouse, i.c.v.) sensitive. This effect was blocked by the delta-antagonist ICI-174.864 (10 microM). The presence of subtypes of the delta-opioid receptor (delta1 and delta2) in PAG has been suggested by pharmacological studies. In this brain structure, naltrindrole 5'-isothiocyanate (5'-NTII), but not 7-benzylidenenaltrexone (BNTX), antagonized the effects of DPDPE and [D-Ala2]deltorphin II, suggesting the involvement of a population of delta receptors sensitive to the delta2-antagonist NT II on this effect. To further investigate the participation of delta-receptor subtypes in the stimulation of IPs formation, mice were injected with antisense oligodeoxynucleotides (ODNs) directed to nucleotides 7-26 or 2946 of the cloned delta-receptor mRNA, and PAG slices from these animals were used in in vitro assays. The results demonstrate that the reported increase of phosphoinositide (PI) hydrolysis depends on the agonist activation of the delta2-opioid receptor subtype in the PAG.


Assuntos
Analgésicos/farmacologia , Encefalinas/farmacologia , Oligopeptídeos/farmacologia , Substância Cinzenta Periaquedutal/metabolismo , Substância Cinzenta Periaquedutal/ultraestrutura , Fosfatidilinositóis/biossíntese , Receptores Opioides delta/agonistas , Receptores Opioides delta/classificação , Animais , D-Penicilina (2,5)-Encefalina , Proteínas de Ligação ao GTP/fisiologia , Inositol/metabolismo , Masculino , Camundongos , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Toxina Pertussis , Receptores Opioides delta/fisiologia , Sensibilidade e Especificidade , Trítio , Fatores de Virulência de Bordetella/farmacologia
17.
Peptides ; 19(2): 343-50, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9493867

RESUMO

Quantitative ligand binding studies resolved two subtypes of the delta opioid receptor, termed delta(ncx1) and delta(ncx2), in mouse brain membranes depleted of mu receptors by pretreatment with the irreversible ligand, BIT. The purpose of the present study was to compare the binding parameters, ligand-selectivity profile and pharmacological properties of the cloned mouse delta receptor (MDOR) stably expressed in a cell line to the delta(ncx) binding sites of mouse brain. [3H][D-Ala2,D-Leu5]enkephalin labeled a single binding site in membranes prepared from MDOR cells under several different assay conditions including BIT-pretreatment. The MDOR had high affinity for delta agonists and antagonists. [3H][D-Ala2,D-Leu5]enkephalin labeled two binding sites in mouse brain membranes depleted of mu receptors by pretreatment with BIT: the delta(ncx1) site (high affinity for DPDPE and deltorphin) and the delta(ncx2) site (low affinity for DPDPE and deltorphin). Some agents were moderately selective for the delta(ncx2) site: [pCl]DPDPE (10.9-fold), JP41 (5.9-fold) and JP45 (3.8-fold). The Ki values of 12 opioids at the mouse MDOR were determined. These values were highly correlated with their values at the delta(ncx1) site but not the delta(ncx2) site. These data suggest that the delta(ncx2) site may be distinct from the cloned delta opioid receptor.


Assuntos
Encéfalo/metabolismo , Receptores Opioides delta/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Clonagem Molecular , Leucina Encefalina-2-Alanina/análogos & derivados , Leucina Encefalina-2-Alanina/metabolismo , Leucina Encefalina-2-Alanina/farmacologia , Técnicas In Vitro , Cinética , Ligantes , Camundongos , Receptores Opioides delta/classificação , Receptores Opioides delta/genética
18.
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
19.
Peptides ; 18(10): 1615-21, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9437724

RESUMO

A series of deltorphin I analogs containing D- or L-N-methylalanine (MeAla), D- or L-proline (Pro), alpha-aminoisobutyric acid (Aib), sarcosine (Sar) or D-tert-leucine (Tle) in place of D-Ala2, or phenylalanine in place of Tyr1, was synthesized. The opioid activity profiles of these peptides were determined in mu and delta opioid receptor-representative binding assays and bioassays in vitro as well as in the rat tail flick test in vivo. In comparison with the deltorphin I parent, both the L- and the D-MeAla2-analog were slightly more potent delta agonists in the mouse vas deferens (MDV) assay, and the D-MeAla2-analog showed two-fold higher antinociceptive potency in the analgesic test. In view of the fact that deltorphin analogs with an unsubstituted L-amino acid residue in the 2-position generally lack opioid activity, the observed high delta opioid potency of [L-MeAla2]deltorphin I is postulated to be due to the demonstrated presence of a conformer with a cis Tyr1-MeAla2 peptide bond, since the cis conformer allows for a spatial arrangement of the pharmacophoric moieties in the N-terminal tripeptide segment similar to that in active deltorphin analogs containing a D-amino acid residue in the 2-position. Substitution of Aib in the 2-position led to a compound, H-Tyr-Aib-Phe-Asp-Val-Val-Gly-NH2, which displayed lower delta receptor affinity than the parent peptide but higher delta selectivity and, surprisingly, three times higher antinociceptive potency. The D- and L-Pro2-, Sar2- and D-Tle2-analogs showed much reduced delta receptor affinities and were inactive in the tail flick test. Replacement of Tyr1 in deltorphin I with Phe produced a 32-fold decrease in delta receptor affinity but only a 7-fold drop in antinociceptive potency.


Assuntos
Analgésicos Opioides/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Animais , Encéfalo/metabolismo , Eletrofisiologia , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Contração Muscular/efeitos dos fármacos , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Medição da Dor , Ligação Proteica , Ratos , Receptores Opioides delta/agonistas , Receptores Opioides delta/classificação , Ducto Deferente/efeitos dos fármacos
20.
Eur J Pharmacol ; 338(1): 1-6, 1997 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-9407996

RESUMO

The effects of intracerebroventricular administration of delta1- and delta2-selective opioid receptor agonists on spontaneous alternation performance, elevated plus-maze behavior and passive avoidance learning including step-down and step-through types were examined in mice. Although the delta1-selective opioid receptor agonist, [D-Pen2,L-Pen5]enkephalin (DPLPE) (1-10 microg) or the delta2-selective opioid receptor agonist, [D-Ala2]deltorphin II (deltorphin) (1-10 microg) did not markedly affect spontaneous alternation performance or elevated plus-maze behavior, DPLPE (1, 3 and/or 10 microg) and deltorphin (3 and 10 microg) inhibited passive avoidance learning including step-down and step-through types. The delta1-selective opioid receptor antagonist, 7-benzylidenenaltrexone (3.5 ng), and the delta2-selective opioid receptor antagonist, naltriben (19 ng), significantly antagonized the inhibitory effects of DPLPE (3 microg) and deltorphin (3 microg) on passive avoidance learning, respectively. In contrast, DPLPE (3 microg) or deltorphin (3 microg) did not markedly influence behavioral responses induced by electroshocks during training of passive avoidance learning. Moreover, DPLPE (0.3-3 microg) or deltorphin (0.3-3 microg) failed to significantly affect the radiant heat-induced nociceptive responses. These results suggest that stimulation of delta1- and delta2-opioid receptors produces amnesia, depending on the learning tasks used.


Assuntos
Amnésia/induzido quimicamente , Aprendizagem da Esquiva/efeitos dos fármacos , Encefalinas/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides delta/agonistas , Analgesia , Animais , Comportamento Animal/efeitos dos fármacos , Eletrochoque , D-Penicilina (2,5)-Encefalina , Masculino , Camundongos , Receptores Opioides delta/classificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA