Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.299
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.
PLoS One ; 15(12): e0232864, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33373369

RESUMO

Activation of the kappa opioid receptor (KOR) contributes to the aversive properties of stress, and modulates key neuronal circuits underlying many neurobehavioral disorders. KOR agonists directly inhibit ventral tegmental area (VTA) dopaminergic neurons, contributing to aversive responses (Margolis et al. 2003, 2006); therefore, selective KOR antagonists represent a novel therapeutic approach to restore circuit function. We used whole cell electrophysiology in acute rat midbrain slices to evaluate pharmacological properties of four novel KOR antagonists: BTRX-335140, BTRX-395750, PF-04455242, and JNJ-67953964. Each compound concentration-dependently reduced the outward current induced by the KOR selective agonist U-69,593. BTRX-335140 and BTRX-395750 fully blocked U-69,593 currents (IC50 = 1.2 ± 0.9 and 1.2 ± 1.3 nM, respectively). JNJ-67953964 showed an IC50 of 3.0 ± 4.6 nM. PF-04455242 exhibited partial antagonist activity asymptoting at 55% blockade (IC50 = 6.7 ± 15.1 nM). In 3/8 of neurons, 1 µM PF-04455242 generated an outward current independent of KOR activation. BTRX-335140 (10 nM) did not affect responses to saturating concentrations of the mu opioid receptor (MOR) agonist DAMGO or the delta opioid receptor (DOR) agonist DPDPE, while JNJ-67953964 (10 nM) partially blocked DAMGO and DPDPE responses. Importantly, BTRX-335140 (10 nM) rapidly washed out with complete recovery of U-69,593 responses within 10 min. Collectively, we show electrophysiological evidence of key differences amongst KOR antagonists that could impact their therapeutic potential and have not been observed using recombinant systems. The results of this study demonstrate the value of characterizing compounds in native neuronal tissue and within circuits implicated in the neurobehavioral disorders of interest.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Receptores Opioides kappa/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Benzamidas/farmacologia , Compostos de Bifenilo/farmacologia , Neurônios Dopaminérgicos/metabolismo , Eletrofisiologia , D-Penicilina (2,5)-Encefalina/farmacologia , Masculino , Mesencéfalo/metabolismo , Antagonistas de Entorpecentes/farmacologia , Oxidiazóis/farmacologia , Técnicas de Patch-Clamp/métodos , Piperidinas/farmacologia , Pirrolidinas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Sulfonamidas/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos
5.
Reprod Toxicol ; 93: 211-218, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32145291

RESUMO

The endogenous opioid peptides have been reported to be involved in the regulation of reproductive physiology. Many of the studies conclude with sentences around the harmful effect of opioids in male fertility but, actually, there is only one study regarding the real fertility potential of spermatozoa that have been exposed to mu specific opioids. The aim of the present study was to see if the modulation of delta (OPRD1) and kappa (OPRK1) opioid receptors in mouse sperm during capacitation was able to vary the embryo production after in vitro fertilization (IVF). The presence of OPRD1 and OPRK1 in mouse mature spermatozoa was analyzed by RT-PCR and immunofluorescence. Incubating the sperm with, on one hand, the delta specific agonist DPDPE and/or antagonist naltrindole, and, on the other hand, the kappa specific agonist U-50488 and antagonist nor-binaltorphimine, we analyzed the involvement of OPRD1 and OPRK1 on IVF and preimplantational embryo development. We verified the presence of OPRD1 and OPRK1 in mouse mature spermatozoa, not only at the mRNA level but also at protein level. Moreover, the sperm incubation with DPDPE, before the IVF, had an effect on the fertilization rate of sperm and reduced the number of reached blastocysts, which was reverted by naltrindole. Instead, the use of the kappa agonist U-50488 and the antagonist nor-binaltophimine did not have any effect on the amount and the quality of the achieved blastocysts. Although nowadays the pure delta or kappa opioid ligands are not used for the clinic, clinical trials are being conducted to be used in the near future, so it would be interesting to know if the modulation of these receptors in sperm would generate any consequence in relation to fertilization capacity.


Assuntos
Fertilização In Vitro , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Espermatozoides/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Blastocisto/fisiologia , Embrião de Mamíferos , Desenvolvimento Embrionário , D-Penicilina (2,5)-Encefalina/farmacologia , Masculino , Camundongos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oócitos/fisiologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Capacitação Espermática
7.
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
8.
Sci Rep ; 9(1): 5771, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30962495

RESUMO

Here, we report the chemical synthesis of two DPDPE analogues 7a (NOVA1) and 7b (NOVA2). This entailed the solid-phase synthesis of two enkephalin precursor chains followed by a CuI-catalyzed azide-alkyne cycloaddition, with the aim of improving in vivo analgesic efficacy versus DPDPE. NOVA2 showed good affinity and selectivity for the µ-opioid receptor (KI of 59.2 nM, EC50 of 12.9 nM, EMax of 87.3%), and long lasting anti-nociceptive effects in mice when compared to DPDPE.


Assuntos
Analgésicos/síntese química , Química Click/métodos , D-Penicilina (2,5)-Encefalina/análogos & derivados , Analgésicos/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Reação de Cicloadição/métodos , D-Penicilina (2,5)-Encefalina/síntese química , D-Penicilina (2,5)-Encefalina/farmacologia , Humanos , Masculino , Camundongos , Ligação Proteica , Receptores Opioides mu/metabolismo
9.
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
10.
Eur J Pharmacol ; 837: 88-95, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30086266

RESUMO

The nucleus accumbens contains delta-opioid receptors that may decrease inhibitory neurotransmission. As GABAB receptors inhibit dopamine release, decrease in activation of GABAB receptors may be a mediator of delta-opioid receptor-induced accumbal dopamine efflux. If so, accumbal dopamine efflux induced by delta-opioid receptor activation should be suppressed by stimulating GABAB receptors. As delta-opioid receptors are further subdivided into delta1- and delta2-opioid receptors, we analysed the effects of the GABAB receptor agonist baclofen on delta1- and delta2-opioid receptor-mediated accumbal dopamine efflux in freely moving rats using in vivo microdialysis. Drugs were applied intracerebrally through the dialysis probe. Doses of compounds show total amount administered (mol) during 25-50 min infusions. Baclofen (2.5 and 5.0 nmol), which did not alter basal dopamine levels, inhibited the delta1-opioid receptor agonist DPDPE (5.0 nmol)-induced dopamine efflux. Baclofen (2.5 and 5.0 nmol) also inhibited the delta2-opioid receptor agonist deltorphin II (25.0 nmol)-induced dopamine efflux. A low dose of the GABAB receptor antagonist 2-hydroxysaclofen (100.0 pmol), which failed to alter basal accumbal dopamine levels, counteracted the inhibitory effects of baclofen (5.0 nmol) on DPDPE (5.0 nmol)- and deltorphin II (25.0 nmol)-induced dopamine efflux. The present results show that reduction in accumbal GABAB receptor-mediated inhibition of accumbal dopaminergic activity facilitates activation of delta1- and delta2-opioid receptor-induced increases in accumbal dopamine efflux. This study suggests that activation of delta1- and delta2-opioid receptors on the cell bodies and/or terminals of accumbal GABAergic interneurons inhibits GABA release and, accordingly, decreases GABAB receptor-mediated inhibition of dopaminergic terminals, resulting in enhanced accumbal dopamine efflux.


Assuntos
Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Receptores de GABA-B/fisiologia , Receptores Opioides delta/fisiologia , Animais , Baclofeno/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Masculino , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/fisiologia
11.
Brain Res ; 1697: 53-58, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29902466

RESUMO

Placental Opioid Enhancing Factor (POEF) is found in amniotic fluid (AF) and placenta. When ingested, it enhances opioid-mediated pain relief. Our laboratory has shown that ingestion of AF specifically enhances the hypoalgesia associated with δ-opioid receptor activation in the brain. The specific biochemical compound in AF responsible for the enhancement of δ-opioid activity is of great interest as an analgesic adjunct for pain but is unknown at this time. Research efforts to isolate and characterize this biochemical compound are hampered by the lack of an algesiometric assay that allows repeated measurement of pain threshold and repeated exposure to δ-opioid receptor activation. The cold water tail-flick assay (CWTF) may be a sensitive and reliable pain threshold test of (a) all species of opioids that is (b) not subject to repeated-testing effects. Therefore the CWTF test is potentially ideal for the study of δ opioid systems in a repeated measures design. Here, we confirm these attributes of the CWTF test, and determined that (a) there are no repeated-exposure effects associated with the CWTF assay; (b) there are no repeated-exposure effects associated with repeated central injections of DPDPE ([D-Pen2,D-Pen5]-Enkephalin, a selective δ-opioid agonist) as measured by the CWTF assay; and (c) ingestion of AF in conjunction with a central injection of DPDPE produced the same hypoalgesic enhancement as previously found using another assay.


Assuntos
Líquido Amniótico/fisiologia , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Receptores Opioides delta/agonistas , Líquido Amniótico/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Temperatura Baixa/efeitos adversos , Ingestão de Alimentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Encefalinas/farmacologia , Feminino , Dor/metabolismo , Gravidez , Ratos , Ratos Long-Evans , Receptores Opioides delta/efeitos dos fármacos , Água
12.
Neuron ; 98(5): 963-976.e5, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29754753

RESUMO

Opioid receptors (ORs) precisely modulate behavior when activated by native peptide ligands but distort behaviors to produce pathology when activated by non-peptide drugs. A fundamental question is how drugs differ from peptides in their actions on target neurons. Here, we show that drugs differ in the subcellular location at which they activate ORs. We develop a genetically encoded biosensor that directly detects ligand-induced activation of ORs and uncover a real-time map of the spatiotemporal organization of OR activation in living neurons. Peptide agonists produce a characteristic activation pattern initiated in the plasma membrane and propagating to endosomes after receptor internalization. Drugs produce a different activation pattern by additionally driving OR activation in the somatic Golgi apparatus and Golgi elements extending throughout the dendritic arbor. These results establish an approach to probe the cellular basis of neuromodulation and reveal that drugs distort the spatiotemporal landscape of neuronal OR activation.


Assuntos
Analgésicos Opioides/metabolismo , Membrana Celular/metabolismo , Dendritos/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Neurônios/metabolismo , Peptídeos/metabolismo , Receptores Opioides/metabolismo , Animais , Técnicas Biossensoriais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , D-Penicilina (2,5)-Encefalina/metabolismo , Leucina Encefalina-2-Alanina/metabolismo , Células HEK293 , Células HeLa , Humanos , Espaço Intracelular , Microscopia de Fluorescência , Morfina/metabolismo , Naloxona , Antagonistas de Entorpecentes , Ratos , Análise Espaço-Temporal
13.
J Physiol Pharmacol ; 69(5)2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30683824

RESUMO

Steroid hormones play an important role in the regulation of cyclic changes in the uterus and preparation of intrauterine environment for the egg fertilization, embryo implantation and maintenance of pregnancy. Their secretion by porcine uterus has been demonstrated. The present study aimed to establish the effect of opioid receptors (µ, δ and κ) activation by selective agonists (DAMGO, DPLPE and U 50.488, respectively) on in vitro secretion of steroid hormones (during 6-h and 24-h incubations) by the endometrial explants of gilts on days 2 - 3, 10 - 11, 12 - 3, 15 - 16, 18 - 20 of the estrous cycle and 10 - 11, 12 - 13, 15 - 16 of pregnancy. The agonists at certain of tested concentrations (10-9, 10-8 and 10-7 M) affected secretion of steroid hormones. Progesterone secretion was increased by µ-opioid receptor agonist on days 18 - 20 (6 h) and by δ-agonist on days 2 - 3 and 18 - 20 (24 h) of the cycle. During pregnancy (days 15 - 16), κ-agonist increased it (6 h), but µ-opioid agonist decreased (24 h). Androstenedione secretion was decreased during shorter incubation; by µ- and δ-receptor agonists on days 2 - 3, by all agonists on days 12 - 13, and by κ-receptor agonist on days 18 - 20 of the cycle. However, it was increased during longer incubation with agonists of κ- and µ-opioid receptors on days 10 - 11 and 18 - 20 of the cycle, respectively. Estradiol secretion was elevated by κ- and µ-agonists (6 h) on days 2 - 3 and 15 - 16 of the cycle, respectively, as well as following 24-h incubation with µ-agonist on days 15 - 16, and µ- and κ-agonists on days 18 - 20 of the cycle. During pregnancy, its secretion was increased (24 h) ondays 15 - 16 by µ- and κ-opioid agonists. Cortisol secretion did not significantly change (versus control) in response to applied treatments. These results indicate a potential involvement of EOPs in the modulation of endometrial steroidogenesis in the pig during the estrous cycle and pregnancy.


Assuntos
Endométrio/metabolismo , Ciclo Estral/metabolismo , Gravidez/metabolismo , Receptores Opioides/agonistas , Esteroides/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Feminino , Suínos
14.
ACS Chem Neurosci ; 8(11): 2549-2557, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28796483

RESUMO

In an effort to expand the structure-activity relationship (SAR) studies of a series of mixed-efficacy opioid ligands, peptidomimetics that incorporate methoxy and hydroxy groups around a benzyl or 2-methylindanyl pendant on a tetrahydroquinoline (THQ) core of the peptidomimetics were evaluated. Compounds containing a methoxy or hydroxy moiety in the o- or m-positions increased binding affinity to the kappa opioid receptor (KOR), whereas compounds containing methoxy or hydroxy groups in the p-position decreased KOR affinity and reduced or eliminated efficacy at the mu opioid receptor (MOR). The results from a substituted 2-methylindanyl series aligned with the findings from the substituted benzyl series. Our studies culminated in the development of 8c, a mixed-efficacy MOR agonist/KOR agonist with subnanomolar binding affinity for both MOR and KOR.


Assuntos
Analgésicos Opioides/química , Peptidomiméticos/química , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/síntese química , Analgésicos Opioides/farmacologia , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Camundongos , Naloxona/farmacologia , Peptidomiméticos/síntese química , Peptidomiméticos/farmacologia , Ligação Proteica , Conformação Proteica , Ratos , Relação Estrutura-Atividade
15.
PLoS One ; 12(7): e0180998, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28700700

RESUMO

The involvement of heme oxygenase 1 (HO-1) in the modulation of the antinociceptive effects of opioids in type 1 diabetes has been demonstrated but the role played by the transcription factor Nrf2 in the regulation of painful neuropathy and in the effects and expression of δ-opioid receptors (DOR) in type 2 diabetes, has not been studied. In male BKS.Cg-m+/+Leprdb/J (db/db) mice, the anti-allodynic effects produced by a Nrf2 transcription factor activator, sulforaphane (SFN) administered alone and combined with two DOR agonists, [d-Pen(2),d-Pen(5)]-Enkephalin (DPDPE) and (+)-4-[(αR)-α-((2S,5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N diethylbenzamide (SNC-80), were evaluated. The effects of SFN on glucose levels and body weight as well as on the proteins levels of Nrf2, HO-1, NAD(P)H: quinone oxidoreductase 1 (NQO1), MAPKs (JNK) and DOR in sciatic nerve from db/db mice were also assessed. This study showed that the administration of SFN dose dependently reversed mechanical allodynia, reduced hyperglycemia and body weight gain associated to type 2 diabetes and significantly increased the anti-allodynic effects of DPDPE and SNC-80 in db/db mice. This treatment normalized the down regulation of Nrf2 and NQO1 and enhanced the protein levels of HO-1 in db/db mice. Moreover, the administration of SFN also inhibited the JNK phosphorylation and DOR down-regulation in the sciatic nerve of diabetic mice. Our data indicated that SFN treatment is effective in reversing mechanical allodynia and enhancing DOR antinociceptive effects in db/db mice which effects might be mediated by activating Nrf2 signaling, reducing hyperglycemia, inhibiting JNK phosphorylation and avoiding DOR down-regulation in the sciatic nerve of these animals. These results propose SFN, alone and/or combined with DOR agonists, as interesting approaches for the treatment of painful diabetic neuropathy associated to type 2 diabetes in mice.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Receptores Opioides delta/metabolismo , Animais , Benzamidas/farmacologia , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Western Blotting , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Peso Corporal/fisiologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Neuropatias Diabéticas/metabolismo , D-Penicilina (2,5)-Encefalina/farmacologia , Hiperalgesia/metabolismo , Isotiocianatos/farmacologia , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/genética , Piperazinas/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Sulfóxidos
16.
Future Med Chem ; 9(9): 859-869, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28635314

RESUMO

AIM: The conjugation of fluorescent labels to opioid peptides is an extremely challenging task, which needs to be overcome to create new classes of probes for biological assays. MATERIALS & METHODS: Three opioid peptide analogs of biphalin and [D-Pen2,5]-Enkephalin (DPDPE) containing a fluorescein-maleimide motif were synthesized. RESULTS & DISCUSSION: The biphalin analog 17 binds to opioid receptors with Kiµ = 530 ± 90 nM and Kiδ = 69.8 ± 16.4 nM. We then tested the ability of the compounds to stimulate G-protein-coupling, 17 activated µ-receptor expressing cells (EC50 = 16.7 ± 6.7 nM, EMax = 76 ± 4%) as well as δ-receptor expressing cells (EC50 = 42 ± 10 nM, EMax = 34 ± 8%). However, 17 was not able to fluorescently label receptor in live or fixed cells. CONCLUSION: Our data suggest that the biphalin scaffold could be employed to develop fluorescent ligands with the appropriate fluorescent motif, and suggest a means for further probe development.


Assuntos
D-Penicilina (2,5)-Encefalina/química , Encefalinas/química , Fluoresceína/química , Fluorescência , Corantes Fluorescentes/química , Maleimidas/química , Corantes Fluorescentes/síntese química , Humanos , Modelos Moleculares
17.
Sci Rep ; 6: 32799, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27605249

RESUMO

Neuropathic pain often results from peripheral nerve damage, which can involve immune response. Local leukocyte-derived opioid peptides or exogenous opioid agonists inhibit neuropathy-induced mechanical hypersensitivity in animal models. Since neuropathic pain can also be augmented by heat, in this study we investigated the role of opioids in the modulation of neuropathy-evoked heat hypersensitivity. We used a chronic constriction injury of the sciatic nerve in wild-type and opioid peptide-knockout mice, and tested opioid effects in heat and mechanical hypersensitivity using Hargreaves and von Frey tests, respectively. We found that although perineural exogenous opioid agonists, including peptidergic ligands, were effective, the endogenous opioid peptides ß-endorphin, Met-enkephalin and dynorphin A did not alleviate heat hypersensitivity. Specifically, corticotropin-releasing factor, an agent triggering opioid peptide secretion from leukocytes, applied perineurally did not attenuate heat hypersensitivity in wild-type mice. Exogenous opioids, also shown to release opioid peptides via activation of leukocyte opioid receptors, were equally analgesic in wild-type and opioid peptide-knockout mice, indicating that endogenous opioids do not contribute to exogenous opioid analgesia in heat hypersensitivity. Furthermore, exogenously applied opioid peptides were ineffective as well. Conversely, opioid peptides relieved mechanical hypersensitivity. Thus, both opioid type and sensory modality may determine the outcome of neuropathic pain treatment.


Assuntos
Analgésicos Opioides/farmacologia , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Peptídeos Opioides/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Hormônio Liberador da Corticotropina/farmacologia , Dinorfinas/genética , Dinorfinas/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Encefalina Metionina/genética , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Temperatura Alta/efeitos adversos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/genética , Peptídeos Opioides/genética , Sistema Nervoso Periférico/metabolismo , Receptores Opioides/agonistas , beta-Endorfina/genética , beta-Endorfina/metabolismo , beta-Endorfina/farmacologia
18.
Neurosci Lett ; 628: 52-8, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27246300

RESUMO

The insular cortex (IC) contributes to nociceptive information processing. IC neurons express opioid receptors, including the mu (MOR), kappa (KOR), and delta (DOR) subtypes. Opioidergic agonists suppress excitatory synaptic transmission in the cerebral cortex. In addition, morphine injection into the IC reduces responses to noxious thermal stimuli. However, the mechanisms of the opioid-dependent modulation of cortical excitation at the macroscopic level, which bridge the cellular and behavioral findings, have remained unknown. The present in vivo optical imaging study aimed to examine the effects of the agonists of each subtype on cortical excitatory propagation in the IC and the neighboring cortices, the primary (S1) and secondary somatosensory (S2) areas. To assess the opioidergic effects on the cortical circuits, we applied electrical stimulation to the maxillary 1st molar pulp, which induced excitation in the ventral part of S1 and the S2/insular oral region (IOR). The initial excitatory response was observed 10-14ms after stimulation, and then excitation propagated concentrically. DAMGO (10-100µM), an MOR agonist, suppressed the amplitude of cortical excitation and shrank the maximum excitation areas in S1 and S2/IOR. In contrast, 10-100µM DPDPE, a DOR agonist, increased the amplitude of excitation and expanded the area of maximum excitation. U50488 (10-100µM), a KOR agonist, had little effect on cortical excitation. These results suggest that MOR-induced suppression of excitatory propagation in the IC is an underlying mechanism of the powerful analgesic effects of MOR agonists. In contrast, DOR may play a minor role in suppressing acute pain.


Assuntos
Analgésicos Opioides/administração & dosagem , Córtex Cerebral/fisiologia , Excitabilidade Cortical/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Córtex Somatossensorial/fisiologia , Animais , Córtex Cerebral/efeitos dos fármacos , Polpa Dentária , Estimulação Elétrica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , D-Penicilina (2,5)-Encefalina/administração & dosagem , Masculino , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/efeitos dos fármacos , Imagens com Corantes Sensíveis à Voltagem
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...