Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.294
Filtrar
1.
Bull Exp Biol Med ; 176(3): 338-341, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38340196

RESUMO

A comparative analysis of the infarct-limiting activity of δ- and κ-opioid receptors (OR) agonists was carried out on a model of coronary occlusion (45 min) and reperfusion (120 min) in male Wistar rats. We used selective δ2-OR agonist deltorphin II (0.12 mg/kg), δ-OR agonists BW373U86 and p-Cl-Phe DPDPE (0.1 and 1 mg/kg), selective agonists of δ1-OR DPDPE (0.1 and 0.969 mg/kg), κ1-OR U-50,488 (0.1 and 1 mg/kg), κ2-OR GR-89696 (0.1 mg/kg), and κ-OR ICI-199,441 (0.1 mg/kg). All drugs were administered intravenously 5 min before reperfusion. Deltorphin II, BW373U86 (1 mg/kg), p-Cl-Phe DPDPE (1 mg/kg), U-50,488 (1 mg/kg), and ICI-199,441 had a cardioprotective effect. The most promising compounds for drug development are ICI-199,441 and deltorphin II.


Assuntos
Benzamidas , Reperfusão Miocárdica , Piperazinas , Receptores Opioides delta , Ratos , Animais , Masculino , Ratos Wistar , D-Penicilina (2,5)-Encefalina , Infarto
2.
Molecules ; 26(22)2021 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-34833919

RESUMO

BACKGROUND: Recent studies suggest that lipids, including free fatty acids (FFAs), are necessary for proper µ opioid receptor (MOR) binding and that activation of opioid receptors (ORs) improves intestinal inflammation. The objective of the study was to investigate a possible interaction between the ORs and FFA receptors (FFARs) ligands in the colitis. METHODS: The potential synergistic effect of ORs and FFARs ligands was evaluated using mouse model of acute colitis induced by dextran sulfate sodium (DSS, 4%). Compounds were injected intraperitoneally (i.p.) once or twice daily at the doses of 0.01 or 0.02 mg/kg body weight (BW) (DAMGO-an MOR agonist), 0.3 mg/kg BW (DPDPE-a δ OR (DOR) agonist) and 1 mg/kg BW (naloxone-a non-selective OR antagonist, GLPG 0974-a FFAR2 antagonist, GSK 137647-a FFAR4 agonist and AH 7614-a FFAR4 antagonist) for 4 days. RESULTS: Myeloperoxidase (MPO) activity was significantly decreased after DAMGO (0.02 mg/kg BW) and GSK 137647 (1 mg/kg BW) administration and co-administration as compared to DSS group. CONCLUSIONS: Treatment with ligands of ORs and FFARs may affect the immune cells in the inflammation; however, no significant influence on the severity of colitis and no synergistic effect were observed.


Assuntos
Colite/tratamento farmacológico , Colite/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/metabolismo , Compostos de Anilina/administração & dosagem , Animais , Butiratos/administração & dosagem , Colite/imunologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , D-Penicilina (2,5)-Encefalina/administração & dosagem , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Peroxidase/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Opioides/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sulfonamidas/administração & dosagem , Tiofenos/administração & dosagem , Xantenos/administração & dosagem
3.
Bull Exp Biol Med ; 170(5): 604-607, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33792818

RESUMO

The study examined the effects of δ-opioid receptor (OR) agonists on infarct size on cardiac ischemia (45 min) and reperfusion (120 min) in vivo model in rats narcotized with α-chloralose. The OR agonists were injected intravenously 5 min prior to reperfusion, OR antagonists were administered 10 min before reperfusion. A selective δ-OR agonist BW373U86 (1 mg/kg) reduced the infarct size/area at risk ratio. A selective δ1-OR agonist DPDPE injected in the doses of 0.1 or 0.969 mg/kg produced no effect on infarct size. The selective δ2-OR agonist deltorphin II (0.12 mg/kg) reduced infarct size/area at risk ratio by 2 times. The δ-OR agonist p-Cl-Phe-DPDPE (1 mg/kg) reduced infarct size/area at risk ratio by 40%. Naltrexone and naloxone methiodide, the peripheral OR antagonists, and selective δ2-OR antagonist naltriben prevented the infarct size limiting effect of deltorphin II. Therefore, activation of peripheral δ2-OR enhanced cardiac resistance against toxic action of reperfusion. During reperfusion, deltorphin II demonstrated the most pronounced cardioprotective activity.


Assuntos
Cardiotônicos/uso terapêutico , D-Penicilina (2,5)-Encefalina/uso terapêutico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Antagonistas de Entorpecentes/uso terapêutico , Receptores Opioides delta/metabolismo , Animais , Benzamidas/uso terapêutico , Masculino , Piperazinas/uso terapêutico , Ratos , Ratos Wistar
4.
Am J Physiol Regul Integr Comp Physiol ; 316(6): R727-R734, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30943058

RESUMO

A reflex arising from contracting hindlimb muscle is responsible in part for the increases in arterial pressure and heart rate evoked by exercise. The afferent arm of this reflex comprises group III and IV afferents. δ-Opioid receptors are expressed predominately on the spinal endings of group III afferents, whereas µ-opioid receptors are expressed predominately on the spinal endings of group IV afferents. Using stimuli that activated group III afferents, namely static contraction, calcaneal tendon stretch, and lactic acid injection into the superficial epigastric artery, we tested the hypothesis that, in rats with either patent or ligated femoral arteries, activation of pre- and postsynaptic δ-opioid receptors in the dorsal horn attenuated pressor reflex responses to these stimuli. In rats with patent arteries or ligated femoral arteries, [d-Pen2,5]enkephalin (DPDPE), a δ-opioid agonist injected intrathecally (10 µg in 10 µl), significantly attenuated the pressor responses to contraction, stretch, and lactic acid (all P < 0.05). Naltrindole, a δ-opioid receptor antagonist, prevented the attenuation. In contrast, DPDPE did not attenuate the pressor response to capsaicin injection into the superficial epigastric artery in either group of rats (both P > 0.05). Intrathecal injection of saline (10 µl), the vehicle for DPDPE, had no effect on the pressor responses in either group of rats. We conclude that activation of spinal δ-opioid receptors attenuates reflexes evoked by group III afferents in both freely perfused and ligated rats.


Assuntos
D-Penicilina (2,5)-Encefalina/farmacologia , Condicionamento Físico Animal/fisiologia , Receptores Opioides delta/efeitos dos fármacos , Reflexo/fisiologia , Animais , Estado de Descerebração/fisiopatologia , Artéria Femoral/fisiopatologia , Frequência Cardíaca/fisiologia , Masculino , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Esforço Físico/fisiologia , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos
5.
Anesth Analg ; 128(5): 1013-1021, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30801358

RESUMO

BACKGROUND: The use of regional and other opioid-sparing forms of anesthesia has been associated with a decrease in the recurrence of certain malignancies. Direct suppression of human natural killer cells by opioids has been postulated to explain this observation. However, the effect of different classes of opioids on suppression of natural killer cell cytotoxicity has not been systematically characterized. METHODS: After confirming that freshly isolated natural killer cells from peripheral human blood express opioid receptors, cells were incubated with increasing concentrations of clinically used or receptor-specific opioid agonists. We also evaluated the effect of pretreatment with receptor-specific antagonists or naloxone. Treated natural killer cells were then coincubated with a carboxyfluorescein succinimidyl ester-labeled target tumor cell line, K562. Annexin V staining was used to compare the percent of tumor cell apoptosis in the presence of opioid-pretreated and untreated natural killer cells. Treated samples were compared to untreated samples using Kruskal-Wallis tests with a post hoc Dunn correction. RESULTS: Morphine, methadone, buprenorphine, loperamide, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin, and U-50488 significantly decreased natural killer cell cytotoxicity. When natural killer cells were pretreated with naloxone, cyprodime, and nor-binaltorphimine before exposure to morphine, there was no difference in natural killer cytotoxicity, compared to the amount observed by untreated natural killer cells. Fentanyl, O-desmethyltramadol, and [D-Pen2,D-Pen5] enkephalin did not change natural killer cell cytotoxicity compare to untreated natural killer cells. CONCLUSIONS: Incubation of isolated natural killer cells with certain opioids causes a decrease in activity that is not observed after naloxone pretreatment. Suppression of natural killer cell cytotoxicity was observed with µ- and κ-receptor agonists but not δ-receptor agonists. These data suggest that the effect is mediated by µ- and κ-receptor agonism and that suppression is similar with many clinically used opioids.


Assuntos
Analgésicos Opioides/administração & dosagem , Células Matadoras Naturais/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/administração & dosagem , Anestesia , Buprenorfina/administração & dosagem , D-Penicilina (2,5)-Encefalina/administração & dosagem , Fentanila/administração & dosagem , Fluoresceínas/administração & dosagem , Humanos , Terapia de Imunossupressão , Células K562 , Loperamida/administração & dosagem , Metadona/administração & dosagem , Morfinanos/administração & dosagem , Morfina/administração & dosagem , Naloxona/administração & dosagem , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Succinimidas/administração & dosagem , Receptor 4 Toll-Like/metabolismo , Tramadol/administração & dosagem , Tramadol/análogos & derivados
6.
Cell Physiol Biochem ; 38(5): 1815-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27160211

RESUMO

BACKGROUND/AIMS: To develop a suitable hepatocyte-like cell model that could be a substitute for primary hepatocytes with essential transporter expression and functions. Induced hepatocyte-like (iHep) cells directly reprogrammed from mice fibroblast cells were fully characterized. METHODS: Naïve iHep cells were transfected with nuclear hepatocyte factor 4 alpha (Hnf4α) and treated with selected small molecules. Sandwich cultured configuration was applied. The mRNA and protein expression of transporters were determined by Real Time PCR and confocal. The functional transporters were estimated by drug biliary excretion measurement. The inhibition of bile acid efflux transporters by cholestatic drugs were assessed. RESULTS: The expression and function of p-glycoprotein (P-gp), bile salt efflux pump (Bsep), multidrug resistance-associated protein 2 (Mrp2), Na+-dependent taurocholate cotransporting polypeptide (Ntcp), and organic anion transporter polypedtides (Oatps) in iHep cells were significantly improved after transfection of hepatocyte nuclear factor 4 alpha (Hnf4α) and treatment with selected inducers. In vitro intrinsic biliary clearances (CLb,int) of optimized iHep cells for rosuvastatin, methotrexate, d8-TCA (deuterium-labeled sodium taurocholate acid) and DPDPE ([D-Pen2,5] enkephalin hydrate) correlated well with that of sandwich-cultured primary mouse hepatocytes (SCMHs) (r2 = 0.984). Cholestatic drugs were evaluated and the results were compared well with primary mice hepatocytes. CONCLUSION: The optimized iHep cells expressed functional drug transporters and were comparable to primary mice hepatocytes. This study suggested direct reprogramming could provide a potential alternative to primary hepatocytes for drug candidate hepatobiliary disposition and hepatotoxicity screening.


Assuntos
Anticolesterolemiantes/metabolismo , Reprogramação Celular , Fator 4 Nuclear de Hepatócito/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Anticolesterolemiantes/análise , Anticolesterolemiantes/toxicidade , Ácidos e Sais Biliares/metabolismo , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , D-Penicilina (2,5)-Encefalina/análise , D-Penicilina (2,5)-Encefalina/metabolismo , D-Penicilina (2,5)-Encefalina/toxicidade , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/citologia , Hepatócitos/metabolismo , Proteínas de Membrana Transportadoras/genética , Metotrexato/análise , Metotrexato/metabolismo , Metotrexato/toxicidade , Camundongos , Camundongos Endogâmicos ICR , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Rosuvastatina Cálcica/análise , Rosuvastatina Cálcica/metabolismo , Rosuvastatina Cálcica/toxicidade , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Simportadores/genética , Simportadores/metabolismo
7.
Spinal Cord ; 54(10): 767-777, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26927293

RESUMO

OBJECTIVE: The current study aimed to evaluate the contribution(s) of specific opioid receptor systems to the analgesic and detrimental effects of morphine, observed after spinal cord injury in prior studies. STUDY DESIGN: We used specific opioid receptor agonists to assess the effects of µ- (DAMGO), δ- (DPDPE) and κ- (GR89696) opioid receptor activation on locomotor (Basso, Beattie and Bresnahan scale, tapered beam and ladder tests) and sensory (girdle, tactile and tail-flick tests) recovery in a rodent contusion model (T12). We also tested the contribution of non-classic opioid binding using [+]- morphine. METHODS: First, a dose-response curve for analgesic efficacy was generated for each opioid agonist. Baseline locomotor and sensory reactivity was assessed 24 h after injury. Subjects were then treated with an intrathecal dose of a specific agonist and re-tested after 30 min. To evaluate the effects on recovery, subjects were treated with a single dose of an agonist and both locomotor and sensory function were monitored for 21 days. RESULTS: All agonists for the classic opioid receptors, but not the [+]- morphine enantiomer, produced antinociception at a concentration equivalent to a dose of morphine previously shown to produce strong analgesic effects (0.32 µmol). DAMGO and [+]- morphine did not affect long-term recovery. GR89696, however, significantly undermined the recovery of locomotor function at all doses tested. CONCLUSIONS: On the basis of these data, we hypothesize that the analgesic efficacy of morphine is primarily mediated by binding to the classic µ-opioid receptor. Conversely, the adverse effects of morphine may be linked to activation of the κ-opioid receptor. Ultimately, elucidating the molecular mechanisms underlying the effects of morphine is imperative to develop safe and effective pharmacological interventions in a clinical setting. SETTING: USA. SPONSORSHIP: Grant DA31197 to MA Hook and the NIDA Drug Supply Program.


Assuntos
Analgésicos Opioides/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/uso terapêutico , D-Penicilina (2,5)-Encefalina/uso terapêutico , Locomoção/efeitos dos fármacos , Masculino , Morfina/uso terapêutico , Medição da Dor , Piperazinas/uso terapêutico , Pirrolidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Traumatismos da Medula Espinal/fisiopatologia
8.
J Neurochem ; 132(2): 206-17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25330347

RESUMO

Physical exercise stimulates the release of endogenous opioid peptides supposed to be responsible for changes in mood, anxiety, and performance. Exercise alters sensitivity to these effects that modify the efficacy at the opioid receptor. Although there is evidence that relates exercise to neuropeptide expression in the brain, the effects of exercise on opioid receptor binding and signal transduction mechanisms downstream of these receptors have not been explored. Here, we characterized the binding and G protein activation of mu opioid receptor, kappa opioid receptor or delta opioid receptor in several brain regions following acute (7 days) and chronic (30 days) exercise. As regards short- (acute) or long-term effects (chronic) of exercise, overall, higher opioid receptor binding was observed in acute-exercise animals and the opposite was found in the chronic-exercise animals. The binding of [(35) S]GTPγS under basal conditions (absence of agonists) was elevated in sensorimotor cortex and hippocampus, an effect more evident after chronic exercise. Divergence of findings was observed for mu opioid receptor, kappa opioid receptor, and delta opioid receptor receptor activation in our study. Our results support existing evidence of opioid receptor binding and G protein activation occurring differentially in brain regions in response to diverse exercise stimuli. We characterized the binding and G protein activation of mu, kappa, and delta opioid receptors in several brain regions following acute (7 days) and chronic (30 days) exercise. Higher opioid receptor binding was observed in the acute exercise animal group and opposite findings in the chronic exercise group. Higher G protein activation under basal conditions was noted in rats submitted to chronic exercise, as visible in the depicted pseudo-color autoradiograms.


Assuntos
Química Encefálica , Condicionamento Físico Animal , Receptores Opioides/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Benzenoacetamidas/metabolismo , Córtex Cerebral/metabolismo , Eletrochoque , D-Penicilina (2,5)-Encefalina/metabolismo , Ativação Enzimática , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Naloxona/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos Opioides/metabolismo , Ligação Proteica , Pirrolidinas/metabolismo , Ratos , Transdução de Sinais , Fatores de Tempo
9.
J Pharmacol Exp Ther ; 353(1): 44-51, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25637601

RESUMO

The regulation of opioid receptor system function in peripheral sensory neurons is not well understood. Opioid agonist efficacy to inhibit nociceptor function and to promote antinociception is generally weak under basal conditions and frequently no response occurs. However, in response to a cyclooxygenase-dependent metabolite of arachidonic acid (AA) after exposure to inflammatory mediators, such as bradykinin (BK) or exogenous AA, peripheral opioid receptor systems become much more responsive to opioid agonists. In this study, we examined the time course for the induction and maintenance of functional competence of the δ-opioid receptor (DOR) system in adult rat nociceptors in culture and in vivo. We found that the responsive state of DOR after pretreatment with BK or exogenous AA is transient (30-60 minutes) and persists for 15-30 minutes after a 15-minute exposure of nociceptors to BK or AA. Interestingly, whereas functional competence of the DOR system could be reinduced with a second application of BK 60 minutes after the first, responsiveness of the DOR system could not be reinduced after an initial exposure to AA. This nonresponsive state of DOR after exogenous AA was mediated by a lipoxygenase (LOX)-dependent metabolite of AA. Intraplantar carrageenan also produced transient DOR functional competence and responsiveness was also reinduced by inhibition of LOX. Thus, the DOR system expressed by peripheral sensory neurons is under dual regulation by cyclooxygenase- and LOX-dependent metabolites of AA.


Assuntos
Ácido Araquidônico/metabolismo , Receptores Opioides delta/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Ácido Araquidônico/farmacologia , Bradicinina/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Lipoxigenase/metabolismo , Inibidores de Lipoxigenase/farmacologia , Masculino , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Células Receptoras Sensoriais/efeitos dos fármacos , Gânglio Trigeminal/citologia
10.
Cell Mol Life Sci ; 71(8): 1529-46, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24022593

RESUMO

Signaling bias refers to G protein-coupled receptor ligand ability to preferentially activate one type of signal over another. Bias to evoke signaling as opposed to sequestration has been proposed as a predictor of opioid ligand potential for generating tolerance. Here we measured whether delta opioid receptor agonists preferentially inhibited cyclase activity over internalization in HEK cells. Efficacy (τ) and affinity (KA) values were estimated from functional data and bias was calculated from efficiency coefficients (log τ/KA). This approach better represented the data as compared to alternative methods that estimate bias exclusively from τ values. Log (τ/KA) coefficients indicated that SNC-80 and UFP-512 promoted cyclase inhibition more efficiently than DOR internalization as compared to DPDPE (bias factor for SNC-80: 50 and for UFP-512: 132). Molecular determinants of internalization were different in HEK293 cells and neurons with ßarrs contributing to internalization in both cell types, while PKC and GRK2 activities were only involved in neurons. Rank orders of ligand ability to engage different internalization mechanisms in neurons were compared to rank order of E max values for cyclase assays in HEK cells. Comparison revealed a significant reversal in rank order for cyclase E max values and ßarr-dependent internalization in neurons, indicating that these responses were ligand-specific. Despite this evidence, and because kinases involved in internalization were not the same across cellular backgrounds, it is not possible to assert if the magnitude and nature of bias revealed by rank orders of maximal responses is the same as the one measured in HEK cells.


Assuntos
Dor Crônica/tratamento farmacológico , AMP Cíclico/metabolismo , Tolerância a Medicamentos/fisiologia , Neurônios/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Análise de Variância , Benzamidas/farmacologia , Benzimidazóis/farmacologia , Western Blotting , Relação Dose-Resposta a Droga , D-Penicilina (2,5)-Encefalina , Imunofluorescência , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas de Fluorescência Verde , Células HEK293 , Humanos , Ligantes , Oligopeptídeos/farmacologia , Piperazinas/farmacologia
11.
Eksp Klin Farmakol ; 78(1): 27-9, 2015.
Artigo em Russo | MEDLINE | ID: mdl-25826871

RESUMO

It is established that activation of delta1 opioid receptors with their selective agonist DPDPE(100 µg/kg) significantly increases IgM immune response not only in C57BL/6J mice with unchanged psychoemotional state, but also in mice displaying aggressive or depressive-like behavior in the social stress model (10 days of agonistic confrontations). SCH-23390 (1.0 mg/kg), selective antagonist of dopamine D1 receptors, and selective D2 receptor blocker haloperidol (1 mg/kg) prevented the immunostimulating effect of DPDPE in animals not subjected to social stress. At the same time, both SCH-23390 and haloperidol did not affect DPDPE-induced immunostimulation in mice engaged in aggressive or depressive-like behaviors.


Assuntos
Agressão/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Depressão/imunologia , Antagonistas de Dopamina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Imunoglobulina M/biossíntese , Estresse Psicológico/imunologia , Agressão/psicologia , Animais , Benzazepinas/farmacologia , Depressão/tratamento farmacológico , Depressão/psicologia , Eritrócitos/imunologia , Haloperidol/farmacologia , Imunização , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Opioides delta/metabolismo , Ovinos , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Estresse Psicológico/tratamento farmacológico
12.
Am J Physiol Cell Physiol ; 307(3): C266-77, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24898587

RESUMO

At birth, asphyxial stressors such as hypoxia and hypercapnia are important physiological stimuli for adrenal catecholamine release that is critical for the proper transition to extrauterine life. We recently showed that chronic opioids blunt chemosensitivity of neonatal rat adrenomedullary chromaffin cells (AMCs) to hypoxia and hypercapnia. This blunting was attributable to increased ATP-sensitive K(+) (KATP) channel and decreased carbonic anhydrase (CA) I and II expression, respectively, and involved µ- and δ-opioid receptor signaling pathways. To address underlying molecular mechanisms, we first exposed an O2- and CO2-sensitive, immortalized rat chromaffin cell line (MAH cells) to combined µ {[d-Arg(2),Ly(4)]dermorphin-(1-4)-amide}- and δ ([d-Pen(2),5,P-Cl-Phe(4)]enkephalin)-opioid agonists (2 µM) for ∼7 days. Western blot and quantitative real-time PCR analysis revealed that chronic opioids increased KATP channel subunit Kir6.2 and decreased CAII expression; both effects were blocked by naloxone and were absent in hypoxia-inducible factor (HIF)-2α-deficient MAH cells. Chronic opioids also stimulated HIF-2α accumulation along a time course similar to Kir6.2. Chromatin immunoprecipitation assays on opioid-treated cells revealed the binding of HIF-2α to a hypoxia response element in the promoter region of the Kir6.2 gene. The opioid-induced regulation of Kir6.2 and CAII was dependent on protein kinase A, but not protein kinase C or calmodulin kinase, activity. Interestingly, a similar pattern of HIF-2α, Kir6.2, and CAII regulation (including downregulation of CAI) was replicated in chromaffin tissue obtained from rat pups born to dams exposed to morphine throughout gestation. Collectively, these data reveal novel mechanisms by which chronic opioids blunt asphyxial chemosensitivity in AMCs, thereby contributing to abnormal arousal responses in the offspring of opiate-addicted mothers.


Assuntos
Células Cromafins/metabolismo , Canais KATP/biossíntese , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Córtex Suprarrenal/citologia , Córtex Suprarrenal/metabolismo , Medula Suprarrenal/citologia , Medula Suprarrenal/metabolismo , Analgésicos Opioides/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Anidrase Carbônica I/biossíntese , Anidrase Carbônica II/biossíntese , Hipóxia Celular , Linhagem Celular , Células Cromafins/citologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dopamina/metabolismo , D-Penicilina (2,5)-Encefalina/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Hipercapnia , Indóis/farmacologia , Isoquinolinas/farmacologia , Canais KATP/genética , Maleimidas/farmacologia , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Norepinefrina/metabolismo , Oligopeptídeos/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Gravidez , Regiões Promotoras Genéticas , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Sulfonamidas/farmacologia
13.
J Pharmacol Exp Ther ; 351(1): 224-32, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25204546

RESUMO

The administration of µ-opioid receptor (MOR), δ-opioid receptor (DOR), and cannabinoid 2 receptor (CB2R) agonists attenuates inflammatory pain. We investigated whether treatment with the heme oxygenase 1 (HO-1) inducer, cobalt protoporphyrin IX (CoPP), could modulate the local effects and expression of MOR, DOR, or CB2R during chronic inflammatory pain. In mice with inflammatory pain induced by the subplantar administration of complete Freund's adjuvant, we evaluated the effects of the intraperitoneal administration of 10 mg/kg CoPP on the antiallodynic and antihyperalgesic actions of locally administered MOR (morphine), DOR (DPDPE {[d-Pen(2),d-Pen(5)]-enkephalin}), or CB2R [JWH-015 {(2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone}] agonists and its reversion with the HO-1 inhibitor, tin protoporphyrin IX (SnPP). The effect of CoPP treatment on the dorsal root ganglia expression of HO-1, MOR, DOR, and CB2R was also assessed. The results show that treatment with CoPP increased the local antinociceptive effects produced by morphine, DPDPE, or JWH-015 during chronic inflammatory pain, and these effects were blocked by the subplantar administration of SnPP, indicating the participation of HO-1 in the antinociceptive actions. CoPP treatment, apart from inducing the expression of HO-1, also enhanced the expression of MOR, did not alter CB2R, and avoided the decreased expression of DOR induced by inflammatory pain. This study shows that the HO-1 inducer (CoPP) increased the local antinociceptive effects of MOR, DOR, and CB2R agonists during inflammatory pain by altering the peripheral expression of MOR and DOR. Therefore, the coadministration of CoPP with local morphine, DPDPE, or JWH-015 may be a good strategy for the management of chronic inflammatory pain.


Assuntos
Analgésicos/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Dor Nociceptiva/tratamento farmacológico , Protoporfirinas/farmacologia , Analgésicos/uso terapêutico , Animais , D-Penicilina (2,5)-Encefalina/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/metabolismo , Protoporfirinas/uso terapêutico , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Receptores sigma/agonistas , Receptores sigma/genética , Receptores sigma/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 306(4): R265-72, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24401991

RESUMO

The central opioid system is involved in a broadly distributed neural network that regulates food intake. Here, we show that activation of central δ-opioid receptor not only stimulated normal diet intake but conversely suppressed high-fat diet intake as well. [D-Pen(2,5)]-enkephalin (DPDPE), an agonist selective for the δ-receptor, increased normal diet intake after central administration to nonfasted male mice. The orexigenic activity of DPDPE was inhibited by blockade of cyclooxygenase (COX)-2, lipocalin-type prostaglandin D synthase (L-PGDS), D-type prostanoid receptor 1 (DP(1)), and neuropeptide Y (NPY) receptor type 1 (Y1) for PGD(2) and NPY, respectively, suggesting that this was mediated by the PGD(2)-NPY system. In contrast, DPDPE decreased high-fat diet intake in mice fed a high-fat diet. DPDPE-induced suppression of high-fat diet intake was blocked by antagonists of melanocortin 4 (MC(4)) and corticotropin-releasing factor (CRF) receptors but not by knockout of the L-PGDS gene. These results suggest that central δ-opioid receptor activation suppresses high-fat diet intake via the MC-CRF system, independent of the orexigenic PGD(2) system. Furthermore, orally administered rubiscolin-6, an opioid peptide derived from spinach Rubisco, suppressed high-fat diet intake. This suppression was also blocked by centrally administered naltrindole, an antagonist for the δ-receptor, suggesting that rubiscolin-6 suppressed high-fat diet intake via activation of central δ-opioid receptor.


Assuntos
Analgésicos Opioides/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , D-Penicilina (2,5)-Encefalina/farmacologia , Receptores Opioides delta/agonistas , Animais , Celecoxib , Inibidores de Ciclo-Oxigenase/farmacologia , Dieta Hiperlipídica , Ingestão de Alimentos/fisiologia , Masculino , Camundongos , Pirazóis/farmacologia , Receptores de Prostaglandina/antagonistas & inibidores , Sulfonamidas/farmacologia
15.
Biopolymers ; 101(1): 21-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23564013

RESUMO

Delta-opioid (DOP) receptors are members of the G protein-coupled receptor (GPCR) sub-family of opioid receptors, and are evolutionarily related, with homology exceeding 70%, to cognate mu-opioid (MOP), kappa-opioid (KOP), and nociceptin opioid (NOP) receptors. DOP receptors are considered attractive drug targets for pain management because agonists at these receptors are reported to exhibit strong antinociceptive activity with relatively few side effects. Among the most potent analgesics targeting the DOP receptor are the linear and cyclic enkephalin analogs known as DADLE (Tyr-D-Ala-Gly-Phe-D-Leu) and DPDPE (Tyr-D-Pen-Gly-Phe-D-Pen), respectively. Several computational and experimental studies have been carried out over the years to characterize the conformational profile of these penta-peptides with the ultimate goal of designing potent peptidomimetic agonists for the DOP receptor. The computational studies published to date, however, have investigated only a limited range of timescales and used over-simplified representations of the solvent environment. We provide here a thorough exploration of the conformational space of DADLE and DPDPE in an explicit solvent, using microsecond-scale molecular dynamics and bias-exchange metadynamics simulations. Free-energy profiles derived from these simulations point to a small number of DADLE and DPDPE conformational minima in solution, which are separated by relatively small energy barriers. Candidate bioactive forms of these peptides are selected from identified common spatial arrangements of key pharmacophoric points within all sampled conformations.


Assuntos
D-Penicilina (2,5)-Encefalina , Receptores Opioides delta , Encefalinas , Receptores Opioides , Receptores Opioides mu , Rubiaceae
16.
Alcohol Clin Exp Res ; 38(1): 195-203, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24033469

RESUMO

BACKGROUND: While there is a growing body of evidence that the delta opioid receptor (DOR) modulates ethanol (EtOH) consumption, development of DOR-based medications is limited in part because there are 2 pharmacologically distinct DOR subtypes (DOR-1 and DOR-2) that can have opposing actions on behavior. METHODS: We studied the behavioral influence of the DOR-1-selective agonist [D-Pen(2) ,D-Pen(5) ]-Enkephalin (DPDPE) and the DOR-2-selective agonist deltorphin microinjected into the ventral tegmental area (VTA) on EtOH consumption and conditioned place preference (CPP) and the physiological effects of these 2 DOR agonists on GABAergic synaptic transmission in VTA-containing brain slices from Lewis rats. RESULTS: Neither deltorphin nor DPDPE induced a significant place preference in EtOH-naïve Lewis rats. However, deltorphin (but not DPDPE) induced a significant CPP in EtOH-drinking rats. In contrast to the previous finding that intra-VTA DOR-1 activity inhibits EtOH consumption and that this inhibition correlates with a DPDPE-induced inhibition of GABA release, here we found no effect of DOR-2 activity on EtOH consumption nor was there a correlation between level of drinking and deltorphin-induced change in GABAergic synaptic transmission. CONCLUSIONS: These data indicate that the therapeutic potential of DOR agonists for alcohol abuse is through a selective action at the DOR-1 form of the receptor.


Assuntos
D-Penicilina (2,5)-Encefalina/administração & dosagem , Etanol/administração & dosagem , Oligopeptídeos/administração & dosagem , Receptores Opioides delta/agonistas , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/psicologia , Analgésicos Opioides/administração & dosagem , Animais , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Injeções Intraventriculares , Masculino , Ratos , Ratos Endogâmicos Lew , Receptores Opioides delta/fisiologia , Área Tegmentar Ventral/fisiologia
17.
Am J Physiol Heart Circ Physiol ; 305(8): H1246-55, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23934854

RESUMO

In rats with ligated femoral arteries, the exercise pressor reflex is exaggerated, an effect that is attenuated by stimulation of peripheral µ-opioid receptors on group IV metabosensitive afferents. In contrast, δ-opioid receptors are expressed mostly on group III mechanosensitive afferents, a finding that prompted us to determine whether stimulation of these opioid receptors could also attenuate the exaggerated exercise pressor reflex in "ligated" rats. We found femoral arterial injection of [D-Pen2,D-Pen5]enkephalin (DPDPE; 1.0 µg), a δ-opioid agonist, significantly attenuated the pressor and cardioaccelerator components of the exercise pressor reflex evoked by hindlimb muscle contraction in both rats with ligated and patent femoral arteries. DPDPE significantly decreased the pressor responses to muscle mechanoreflex activation, evoked by tendon stretch, in ligated rats only. DPDPE (1.0 µg) had no effect in either group on the pressor and cardioaccelerator responses to capsaicin (0.2 µg), which primarily stimulates group IV afferents. DPDPE (1.0 µg) had no effect on the pressor and cardioaccelerator responses to lactic acid (24 mM), which stimulates group III and IV afferents, in rats with patent femoral arteries but significantly decreased the pressor response in ligated rats. Western blots revealed the amount of protein comprising the δ-opioid receptor was greater in dorsal root ganglia innervating hindlimbs with ligated femoral arteries than in dorsal root ganglia innervating hindlimbs with patent femoral arteries. Our findings support the hypothesis that stimulation of δ-opioid receptors on group III afferents attenuated the exercise pressor reflex.


Assuntos
Mecanorreceptores/fisiologia , Músculo Esquelético/inervação , Condicionamento Físico Animal/fisiologia , Receptores Opioides delta/fisiologia , Reflexo/fisiologia , Analgésicos Opioides/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , D-Penicilina (2,5)-Encefalina/farmacologia , Artéria Femoral/cirurgia , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Ligadura , Masculino , Mecanorreceptores/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Reflexo/efeitos dos fármacos
18.
J Pharmacol Exp Ther ; 346(3): 535-44, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23820126

RESUMO

Activation of opioid receptors on peripheral sensory neurons has the potential for safe pain control, as it lacks centrally mediated side effects. While this approach often only partially suppressed neuropathic pain in animal models, opioids were mostly applied to animal paws although neuropathy was induced at the nerve trunk. Here we aimed to identify the most relevant peripheral site of opioid action for efficient antinociception in neuropathy. On days 2 and 14 following a chronic constriction injury (CCI) of the sciatic nerve in mice, we evaluated dose and time relationships of the effects of µ-, δ-, and κ-opioid receptor agonists injected either at the CCI site or intraplantarly (i.pl.) into the lesioned nerve-innervated paw, on spontaneous paw lifting and heat and mechanical hypersensitivity (using Hargreaves and von Frey tests, respectively). We found that neither agonist diminished spontaneous paw lifting, despite the application site. Heat hypersensitivity was partially attenuated by i.pl. µ-receptor agonist only, while it was improved by all three agonists applied at the CCI site. Mechanical hypersensitivity was slightly diminished by all agonists administered i.pl., whereas it was completely blocked by all opioids injected at the CCI site. These antinociceptive effects were opioid receptor type-selective and site-specific. Thus, opioids might not be effective against spontaneous pain, but they improve heat and mechanical hypersensitivity in neuropathy. Importantly, efficient alleviation of hypersensitivity is governed by peripheral opioid receptors at the injured nerve trunk rather than at its peripheral terminals. Identifying the primary action site of analgesics is important for the development of adequate pain therapies.


Assuntos
Analgésicos Opioides/farmacologia , Terminações Nervosas/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Nervos Periféricos/patologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Constrição Patológica/patologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Pé/inervação , Pé/patologia , Temperatura Alta , Hiperalgesia/tratamento farmacológico , Hiperalgesia/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/patologia , Medição da Dor/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/patologia , Nervos Periféricos/efeitos dos fármacos , Estimulação Física , Receptores Opioides/agonistas
19.
Anesthesiology ; 118(5): 1180-97, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23358127

RESUMO

BACKGROUND: The administration of µ-opioid receptors (MOR) and δ-opioid receptors (DOR) as well as cannabinoid-2 receptor (CB2R) agonists attenuates neuropathic pain. We investigated if treatment with two carbon monoxide-releasing molecules (CORM-2 and CORM-3) or an inducible heme oxygenase inducer (cobalt protoporphyrin IX, CoPP) could modulate the local and systemic effects and expression of MOR, DOR, and CB2R during neuropathic pain. METHODS: In C57BL/6 mice, at 10 days after the chronic constriction of sciatic nerve, we evaluated the effects of the intraperitoneal administration of 10 mg/kg of CORM-2, CORM-3, or CoPP on the antiallodynic and antihyperalgesic actions of a locally or systemically administered MOR (morphine), DOR ([d-Pen(2),d-Pen(5)]-enkephalin) or CB2R ((2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone ) agonist. The effects of CORM-2 and CoPP treatments on the expression of MOR, DOR, CB2R, inducible and constitutive heme oxygenases, microglia activation marker (CD11b/c), and neuronal and inducible nitric oxide synthases were also assessed. RESULTS: Treatments with CO-RMs and CoPP reduced the mechanical and thermal hypersensitivity induced by sciatic nerve injury, increased the local, but not systemic, antinociceptive effects of morphine, and decreased those produced by DPDPE and JWH-015. Both CORM-2 and CoPP treatments enhanced MOR and inducible heme oxygenase expression, unaltered DOR and constitutive heme oxygenase expression, and decreased the overexpression of CB2R, CD11b/c, and neuronal and inducible nitric oxide synthases induced by sciatic nerve injury. CONCLUSIONS: This study shows that CO-RMs and CoPP treatments increase the local antinociceptive effects of morphine through enhancing MOR peripheral expression and inhibiting spinal microglial activation and overexpression of neuronal/inducible nitric oxide synthases.


Assuntos
Monóxido de Carbono/uso terapêutico , Heme Oxigenase-1/biossíntese , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Compostos Organometálicos/uso terapêutico , Receptores Opioides mu/efeitos dos fármacos , Analgésicos Opioides/uso terapêutico , Animais , Monóxido de Carbono/administração & dosagem , Constrição Patológica/complicações , D-Penicilina (2,5)-Encefalina/uso terapêutico , Indução Enzimática/efeitos dos fármacos , Temperatura Alta , Indóis/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/uso terapêutico , Medição da Dor/efeitos dos fármacos , Estimulação Física , Protoporfirinas/uso terapêutico , Receptor CB2 de Canabinoide/agonistas , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/biossíntese
20.
J Biol Chem ; 286(10): 7854-7864, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21209077

RESUMO

Regulator of G protein signaling protein 4 (RGS4) acts as a GTPase accelerating protein to modulate µ- and δ- opioid receptor (MOR and DOR, respectively) signaling. In turn, exposure to MOR agonists leads to changes in RGS4 at the mRNA and/or protein level. Here we have used human neuroblastoma SH-SY5Y cells that endogenously express MOR, DOR, and RGS4 to study opioid-mediated down-regulation of RGS4. Overnight treatment of SH-SY5Y cells with the MOR agonist DAMGO or the DOR agonist DPDPE decreased RGS4 protein by ∼60% accompanied by a profound loss of opioid receptors but with no change in RGS4 mRNA. The decrease in RGS4 protein was prevented by the pretreatment with pertussis toxin or the opioid antagonist naloxone. The agonist-induced down-regulation of RGS4 proteins was completely blocked by treatment with the proteasome inhibitors MG132 or lactacystin or high concentrations of leupeptin, indicating involvement of ubiquitin-proteasome and lysosomal degradation. Polyubiquitinated RGS4 protein was observed in the presence of MG132 or the specific proteasome inhibitor lactacystin and promoted by opioid agonist. The loss of opioid receptors was not prevented by MG132, demonstrating a different degradation pathway. RGS4 is a GTPase accelerating protein for both Gα(i/o) and Gα(q) proteins. After overnight treatment with DAMGO to reduce RGS4 protein, signaling at the Gα(i/o)-coupled DOR and the Gα(q)-coupled M(3) muscarinic receptor (M(3)R) was increased but not signaling of the α(2) adrenergic receptor or bradykinin BK(2) receptor, suggesting the development of cross-talk between the DOR and M(3)R involving RGS4.


Assuntos
Analgésicos Opioides/farmacologia , Regulação para Baixo/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Proteínas RGS/biossíntese , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Ubiquitinação/efeitos dos fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Leupeptinas/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Toxina Pertussis/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Receptor Muscarínico M3/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores da Bradicinina/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA