Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(21): 11820-11828, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32393639

RESUMEN

Opioids, such as morphine and fentanyl, are widely used for the treatment of severe pain; however, prolonged treatment with these drugs leads to the development of tolerance and can lead to opioid use disorder. The "Opioid Epidemic" has generated a drive for a deeper understanding of the fundamental signaling mechanisms of opioid receptors. It is generally thought that the three types of opioid receptors (µ, δ, κ) are activated by endogenous peptides derived from three different precursors: Proopiomelanocortin, proenkephalin, and prodynorphin. Posttranslational processing of these precursors generates >20 peptides with opioid receptor activity, leading to a long-standing question of the significance of this repertoire of peptides. Here, we address some aspects of this question using a technical tour de force approach to systematically evaluate ligand binding and signaling properties ([35S]GTPγS binding and ß-arrestin recruitment) of 22 peptides at each of the three opioid receptors. We show that nearly all tested peptides are able to activate the three opioid receptors, and many of them exhibit agonist-directed receptor signaling (functional selectivity). Our data also challenge the dogma that shorter forms of ß-endorphin do not exhibit receptor activity; we show that they exhibit robust signaling in cultured cells and in an acute brain slice preparation. Collectively, this information lays the groundwork for improved understanding of the endogenous opioid system that will help in developing more effective treatments for pain and addiction.


Asunto(s)
Péptidos Opioides , Receptores Opioides/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Humanos , Masculino , Péptidos Opioides/agonistas , Péptidos Opioides/metabolismo , Proopiomelanocortina/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley
2.
Peptides ; 124: 170223, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31805297

RESUMEN

This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).


Asunto(s)
Analgésicos Opioides/farmacología , Péptidos Opioides/farmacología , Péptidos Opioides/fisiología , Receptores Opioides/metabolismo , Trastornos Relacionados con Sustancias/etiología , Animales , Dolor en Cáncer/tratamiento farmacológico , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/genética , Ingestión de Alimentos/efectos de los fármacos , Emociones , Femenino , Humanos , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Trastornos Relacionados con Opioides/etiología , Trastornos Relacionados con Opioides/prevención & control , Embarazo , Estrés Fisiológico , Receptor de Nociceptina
3.
Psychopharmacology (Berl) ; 234(9-10): 1371-1394, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28285326

RESUMEN

BACKGROUND: Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS: In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS: Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION: Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.


Asunto(s)
Analgésicos Opioides/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Receptores Opioides/metabolismo , Cese del Hábito de Fumar , Tabaquismo/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Sistemas de Liberación de Medicamentos/métodos , Predicción , Humanos , Nicotina/administración & dosificación , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/metabolismo , Receptores Opioides/agonistas , Recompensa , Cese del Hábito de Fumar/métodos , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Tabaquismo/tratamiento farmacológico
4.
J Pharmacol Exp Ther ; 349(3): 535-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24713140

RESUMEN

Cebranopadol (trans-6'-fluoro-4',9'-dihydro-N,N-dimethyl-4-phenyl-spiro[cyclohexane-1,1'(3'H)-pyrano[3,4-b]indol]-4-amine) is a novel analgesic nociceptin/orphanin FQ peptide (NOP) and opioid receptor agonist [Ki (nM)/EC50 (nM)/relative efficacy (%): human NOP receptor 0.9/13.0/89; human mu-opioid peptide (MOP) receptor 0.7/1.2/104; human kappa-opioid peptide receptor 2.6/17/67; human delta-opioid peptide receptor 18/110/105]. Cebranopadol exhibits highly potent and efficacious antinociceptive and antihypersensitive effects in several rat models of acute and chronic pain (tail-flick, rheumatoid arthritis, bone cancer, spinal nerve ligation, diabetic neuropathy) with ED50 values of 0.5-5.6 µg/kg after intravenous and 25.1 µg/kg after oral administration. In comparison with selective MOP receptor agonists, cebranopadol was more potent in models of chronic neuropathic than acute nociceptive pain. Cebranopadol's duration of action is long (up to 7 hours after intravenous 12 µg/kg; >9 hours after oral 55 µg/kg in the rat tail-flick test). The antihypersensitive activity of cebranopadol in the spinal nerve ligation model was partially reversed by pretreatment with the selective NOP receptor antagonist J-113397[1-[(3R,4R)-1-cyclooctylmethyl-3-hydroxymethyl-4-piperidyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one] or the opioid receptor antagonist naloxone, indicating that both NOP and opioid receptor agonism are involved in this activity. Development of analgesic tolerance in the chronic constriction injury model was clearly delayed compared with that from an equianalgesic dose of morphine (complete tolerance on day 26 versus day 11, respectively). Unlike morphine, cebranopadol did not disrupt motor coordination and respiration at doses within and exceeding the analgesic dose range. Cebranopadol, by its combination of agonism at NOP and opioid receptors, affords highly potent and efficacious analgesia in various pain models with a favorable side effect profile.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Indoles/uso terapéutico , Péptidos Opioides/agonistas , Dolor/tratamiento farmacológico , Receptores Opioides/agonistas , Compuestos de Espiro/uso terapéutico , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Artritis Experimental/complicaciones , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Conducta Animal/efectos de los fármacos , Neoplasias Óseas/complicaciones , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Femenino , Indoles/administración & dosificación , Indoles/efectos adversos , Indoles/farmacología , Masculino , Dolor/etiología , Dolor/metabolismo , Polineuropatías/complicaciones , Polineuropatías/tratamiento farmacológico , Polineuropatías/metabolismo , Unión Proteica , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Prueba de Desempeño de Rotación con Aceleración Constante , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/efectos adversos , Compuestos de Espiro/farmacología , Nociceptina
5.
J Pain ; 11(7): 663-71, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20071245

RESUMEN

UNLABELLED: We previously reported that endothelin A (ET-A) receptor antagonism attenuates carcinoma-induced pain in a cancer pain mouse model. In this study, we investigated the mechanism of ET-A receptor-mediated antinociception and evaluated the role of endogenous opioid analgesia. Squamous cell carcinoma (SCC) cell culture treated with the ET-A receptor antagonist (BQ-123) at 10(-6) M and 10(-5) M significantly increased production and secretion of beta-endorphin and leu-enkephalin, respectively. Behavioral studies were performed by inducing tumors in the hind paw of female nude mice with local injection of cells derived from a human oral SCC. Significant pain, as indicated by reduction in withdrawal thresholds in response to mechanical stimulation, began at 4 days after SCC inoculation and lasted to 18 days, the last day of measurement. Local administration of either naloxone methiodide (500 microg/kg), selective antagonists for mu-opioid receptor (CTOP, 500 microg/kg), or delta-opioid receptor (naltrindole, 11 mg/kg) but not kappa-opioid receptor (nor-BNI, 2.5 mg/kg) significantly reversed antinociception observed from ET-A receptor antagonism (BQ-123, 92 mg/kg) in cancer animals. These results demonstrate that antagonism of peripheral ET-A receptor attenuates carcinoma pain by modulating release of endogenous opioids to act on opioid receptors in the cancer microenvironment. PERSPECTIVE: This article proposes a novel mechanism for ET-A receptor antagonist drugs in managing cancer-induced pain. An improved understanding of the role of innate opioid analgesia in ET-A receptor-mediated antinociception might provide novel alternatives to morphine therapy for the treatment of cancer pain.


Asunto(s)
Carcinoma de Células Escamosas/complicaciones , Antagonistas de los Receptores de la Endotelina A , Péptidos Opioides/agonistas , Dolor Intratable/tratamiento farmacológico , Receptores Opioides/agonistas , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Desnudos , Antagonistas de Narcóticos , Trasplante de Neoplasias/métodos , Neoplasias Experimentales/complicaciones , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/fisiopatología , Péptidos Opioides/metabolismo , Dolor Intratable/metabolismo , Dolor Intratable/fisiopatología , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Receptores Opioides/metabolismo , Resultado del Tratamiento , Células Tumorales Cultivadas
6.
Endocrinology ; 148(10): 4993-5001, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17615146

RESUMEN

Orphanin FQ (OFQ), also known as nociceptin, is a member of the endogenous opioid peptide family that has been functionally implicated in the control of pain, anxiety, circadian rhythms, and neuroendocrine function. In the reproductive system, endogenous opioid peptides are involved in the steroid feedback control of GnRH pulses and the induction of the GnRH surge. The distribution of OFQ in the preoptic area and hypothalamus overlaps with GnRH, and in vitro evidence suggests that OFQ can inhibit GnRH secretion from hypothalamic fragments. Using the sheep as a model, we examined the potential anatomical colocalization between OFQ and GnRH using dual-label immunocytochemistry. Confocal microscopy revealed that approximately 93% of GnRH neurons, evenly distributed across brain regions, were also immunoreactive for OFQ. In addition, almost all GnRH fibers and terminals in the external zone of the median eminence, the site of neurosecretory release of GnRH, also colocalized OFQ. This high degree of colocalization suggested that OFQ might be functionally important in controlling reproductive endocrine events. We tested this possibility by examining the effects of intracerebroventricular administration of [Arg(14), Lys(15)] OFQ, an agonist to the OFQ receptor, on pulsatile LH secretion. The agonist inhibited LH pulse frequency in both luteal phase and ovariectomized ewes and suppressed pulse amplitude in the latter. The results provide in vivo evidence supporting a role for OFQ in the control of GnRH secretion and raise the possibility that it acts as part of an ultrashort, autocrine feedback loop controlling GnRH pulses.


Asunto(s)
Sistemas Neurosecretores/fisiología , Péptidos Opioides/fisiología , Reproducción/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Fase Luteínica/efectos de los fármacos , Hormona Luteinizante/metabolismo , Péptidos Opioides/agonistas , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Ovariectomía , Área Preóptica/metabolismo , Ovinos , Distribución Tisular , Nociceptina
7.
J Pept Res ; 66(5): 263-76, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16218994

RESUMEN

Dynorphin A (Dyn A), a 17 amino acid peptide H-Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg-Pro-Lys-Leu-Lys-Trp-Asp-Asn-Gln-OH, is a potent opioid peptide which interacts preferentially with kappa-opioid receptors. Research in the development of selective and potent opioid peptide ligands for the kappa-receptor is important in mediating analgesia. Several cyclic disulphide bridge-containing peptide analogues of Dyn A, which were conformationally constrained in the putative message or address segment of the opioid ligand, were designed, synthesized and assayed. To further investigate the conformational and topographical requirements for the residues in positions 5 and 11 of these analogues, a systematic series of Dyn A(1-11)-NH2 cyclic analogues incorporating the sulphydryl-containing amino acids L- and D-Cys and L- and D-Pen in positions 5 and 11 were synthesized and assayed. Cyclic lactam peptide analogues were also synthesized and assayed. Several of these cyclic analogues, retained the same affinity and selectivity (vs. the mu- and delta-receptors) as the parent Dyn A(1-11)-NH2 peptide in the guinea-pig brain (GPB), but exhibited a much lower activity in the guinea-pig ileum (GPI), thus leading to centrally vs. peripherally selective peptides. Studies of the structure-activity relationship of Dyn A peptide provide new insights into the importance of each amino acid residue (and their configurations) in Dyn A analogues for high potency and good selectivity at kappa-opioid receptors. We report herein the progress towards the development of Dyn A peptide ligands, which can act as agonists or antagonists at cell surface receptors that modulate cell function and animal behaviour using various approaches to rational peptide ligand-based drug design.


Asunto(s)
Diseño de Fármacos , Dinorfinas/síntesis química , Animales , Bioensayo , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Capsaicina/farmacología , Dinorfinas/antagonistas & inhibidores , Dinorfinas/farmacología , Cobayas , Íleon/efectos de los fármacos , Ligandos , Macaca mulatta , Ratones , Músculo Liso/efectos de los fármacos , Músculo Liso/enzimología , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/síntesis química , Dolor/tratamiento farmacológico , Péptidos/antagonistas & inhibidores , Péptidos/química , Conformación Proteica , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/efectos de los fármacos , Especificidad de la Especie
8.
Exp Cell Res ; 294(2): 434-45, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15023532

RESUMEN

Neuropeptides influence cancer cell replication and growth. Opioid peptides, and opiergic neurons are found in the prostate gland, and they are proposed to exert a role in tumor regulation, influencing cancer cell growth, as opioid agonists inhibit cell growth in several systems, including the human prostate cancer cell line LNCaP. In the same cell line, the existence of membrane testosterone receptors was recently reported, which increase, in a non-genomic manner, the secretion of PSA, and modify actin cytoskeleton dynamics, through the signaling cascade FAK-->PI-3 kinase-->Cdc42/Rac1. In the present work, we present data supporting that the general opioid agonist Ethylketocyclazocine (EKC) decreases testosterone-BSA (a non-internalizable testosterone analog) induced PSA secretion. Furthermore, we report that this opioid affects this non-genomic testosterone action, by modifying the distribution of the actin cytoskeleton in the cells, disrupting the above signaling cascade. In addition, after long (>24 h) incubation, opioids decrease the number of membrane testosterone receptors, and reverse their effect on the signaling molecules. In conclusion, our results provide some new insights of a possible action of opioids in prostate cancer control by interfering with the action and the expression of membrane testosterone receptors and signaling.


Asunto(s)
Carcinoma/metabolismo , Membrana Celular/metabolismo , Etilcetociclazocina/farmacología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Testosterona/análogos & derivados , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Analgésicos Opioides/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Carcinoma/tratamiento farmacológico , División Celular/efectos de los fármacos , División Celular/fisiología , Membrana Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Masculino , Péptidos Opioides/agonistas , Péptidos Opioides/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Receptores Androgénicos/efectos de los fármacos , Albúmina Sérica Bovina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Testosterona/metabolismo , Testosterona/farmacología , Células Tumorales Cultivadas
9.
J Steroid Biochem Mol Biol ; 74(1-2): 25-32, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11074353

RESUMEN

The effects of a single injection or continuous infusion of opioid peptide, [D-Met(2),pro(5)]enkephalinamide (ENK) on the hormone binding and transcriptional properties of estrogen receptors were investigated in estradiol (E(2)) treated rat uterus. The level of estrogen- (ER) and progesterone receptor (PR) proteins, the hormone binding of E(2) receptors and the effects of single injection of ENK with or without naltrexone (NAL) on the E(2)-induced changes in the level of Fos and Jun proteins and the binding of AP-1 proteins to DNA were studied. The receptor proteins levels were determined by Western blots and the binding of AP-1 to DNA by electrophoretic mobility shift assay. Both the ER and PR protein concentrations and the [3H]Estradiol binding to the high affinity nuclear receptors decreased after ENK treatment during the first two days. At 72 h the PR concentration decreased further, while no significant changes were found in the level of ER, however, at this time the former competitive E(2) binding turned into positive cooperativity. The E(2)-induced increase in the level of Fos proteins and the binding of AP-1 proteins to DNA was inhibited by a single injection of ENK. We conclude that the endogenous opioid peptides may interact with E(2) in the gene regulation of rat uterus.


Asunto(s)
Encefalina Metionina/análogos & derivados , Encefalina Metionina/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Péptidos Opioides/farmacología , Útero/efectos de los fármacos , Animales , ADN/genética , ADN/metabolismo , Encefalina Metionina/administración & dosificación , Estradiol/metabolismo , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/administración & dosificación , Femenino , Péptidos Opioides/administración & dosificación , Péptidos Opioides/agonistas , Ovariectomía , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Endogámicas , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Factor de Transcripción AP-1/metabolismo , Útero/metabolismo
10.
Peptides ; 21(7): 923-33, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10998526

RESUMEN

Nociceptin and its receptor (OP(4)) share sequence homologies with the opioid peptide ligand dynorphin A and its receptor OP(2). Cationic residues in the C-terminal sequence of both peptides seem to be required for selective receptor occupation, but the number and the distribution of these basic residues are different and quite critical. Both receptors are presumably activated by the peptides N-terminal sequence (Xaa-Gly Gly-Phe, where Xaa = Phe or Tyr); however, although OP(4) requires Phe(4) as a determinant pharmacophore, OP(2) requires Tyr(1) as do the other opioid receptors. An extensive structure-activity analysis of the N-terminal tetrapeptide has led to conclude that the presence of aromatic residues in position one and four, preferably Phe, as well as the distance between Phe(1) and Phe(4) are extremely critical for occupation and activation of OP(4) in contrast with other opioid receptors (e.g. OP(1), OP(3), OP(2)). Modification of distance between the side chains of Phe(1) and Phe(4) (as obtained with Nphe(1) substitution in both NC and NC(1-13)-NH(2)) and/or conformational orientation of Phe(1) (as in Phe(1)psi(CH(2)-NH)-Gly(2)) has brought to discovery of pure antagonist ([Nphe(1)]-NC(1-13)-NH(2)) and a partial agonist ([Phe(1) psi(CH(2)-NH)-Gly(2)]-NC(1-13)-NH(2)), which have allowed us to characterize and classify the OP(4) receptor in several species. Thus, although antagonist activities at the OP(4) receptor are obtained by chemical modification of Phe(1)-Gly(2) peptide bond or by a shift of Phe(1) side chain of NC peptides, antagonism at the OP(2) receptor requires the diallylation of the N-terminal amino function, for instance, of dynorphin A. These considerations support the interpretation that the two systems nociceptin/OP(4) and dynorphin A/OP(2) are distinct pharmacological entities that differs in both their active sites (Tyr(1) for Dyn A and Phe(4) for NC) and the number and position of cationic residues in the C-terminal portions of the molecules. The chemical features of novel OP(4) receptor ligands either pseudopeptides obtained by combinatorial library screening or molecules of nonpeptide structure are reported and discussed in comparison with NC and NC related peptides.


Asunto(s)
Dinorfinas/química , Dinorfinas/fisiología , Péptidos Opioides/química , Péptidos Opioides/fisiología , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Sitios de Unión , Células CHO , Técnicas Químicas Combinatorias , Cricetinae , Humanos , Cinética , Ratones , Datos de Secuencia Molecular , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Péptidos/química , Péptidos/fisiología , Conformación Proteica , Estructura Terciaria de Proteína , Receptores Opioides/química , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Vasodilatadores/agonistas , Vasodilatadores/antagonistas & inhibidores , Vasodilatadores/química , Receptor de Nociceptina , Nociceptina
11.
Pharmacol Biochem Behav ; 60(1): 1-5, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9610916

RESUMEN

Sensitivity of alpha2-adrenoceptors following repeated immobilization sessions plus morphine (MOR) or beta-endorphin (BETA) was assayed by examining clonidine (CLO)-induced hypoactivity in adult malnourished rats at perinatal age. As previously described, chronic restraint did not attenuate the hypoactivity elicited by CLO in malnourished rats, although chronic restraint did have such an effect on motor activity in control animals. MOR and BETA administration prior to each restraint session induced subsensitivity of alpha2-adrenoceptors in malnourished rats as determined by a blunted response to clonidine challenge. An injection of naloxone (NAL) prior to BETA before each stress session fully antagonized the subsensitivity to clonidine observed in malnourished animals. A possible deficiency in the functional role of the opiate system in the process of adaptation to chronic stress in perinatal malnourished rats is suggested.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Clonidina/farmacología , Actividad Motora/efectos de los fármacos , Trastornos Nutricionales/fisiopatología , Trastornos Nutricionales/psicología , Péptidos Opioides/agonistas , Animales , Animales Recién Nacidos , Femenino , Inmovilización , Morfina/farmacología , Actividad Motora/fisiología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Embarazo , Ratas , Ratas Wistar , Estrés Fisiológico/fisiopatología , Estrés Fisiológico/psicología , betaendorfina/farmacología
12.
Biochem Biophys Res Commun ; 246(1): 128-31, 1998 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-9600080

RESUMEN

Opioid agonists either potentiate or suppress basal cAMP production in SK-N-SH cells. The inhibitory effect is mediated by PTX-sensitive GTP-binding proteins, while the stimulatory effect involves Ca++ entry and calmodulin activation. Both pathways can be activated simultaneously by opioid agonists. Low (nM) concentrations of either mu (DAMGO) or delta (DPDPE) selective opioids potentiate cAMP formation. At higher (100 nM) concentrations, however, a net suppression takes over; this suppression can be eliminated by PTX, and the underlying stimulatory effect is disclosed. Micromolar concentrations of either mu or delta selective agonists cross-activate the other (delta or mu) receptors, and augment the stimulatory pathway. The overall outcome (either stimulation or inhibition of cAMP production) is dependent on the balance between the two overlapping pathways, and can be modified by blocking either of the two opposing mechanisms.


Asunto(s)
AMP Cíclico/biosíntesis , Neuroblastoma/metabolismo , Péptidos Opioides/farmacología , Calcio/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5) , Encefalina D-Penicilamina (2,5) , Encefalinas/farmacología , Proteínas de Unión al GTP/metabolismo , Humanos , Cinética , Péptidos Opioides/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Células Tumorales Cultivadas , Factores de Virulencia de Bordetella/farmacología
13.
Biochem Biophys Res Commun ; 238(1): 71-6, 1997 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-9299454

RESUMEN

The wild-type delta opioid receptor (DOR) and a carboxyl terminus-truncated mutant DOR lacking the last 31 amino acids (DOR-T) were expressed in neuroblastoma x glioma hybrid NG108-15 cells to investigate the role of the carboxyl terminus of DOR in agonist-dependent receptor phosphorylation. Stimulation of the cells with delta specific agonists significantly induced DOR phosphorylation whereas no phosphorylation of DOR-T was detected under the same conditions. Neither overexpression of G protein-coupled receptor kinases (GRK2 or GRK5) nor activation of protein kinase C promoted agonist-induced phosphorylation of DOR-T, in contrast to their strong stimulatory effect on the agonist-dependent phosphorylation of DOR. Furthermore, DOR-T failed to be internalized after agonist stimulation, probably due to its inability to be phosphorylated. Our results indicate that the carboxyl terminus of DOR is required for agonist-dependent receptor phosphorylation and the phosphorylation site(s) of DOR is likely located at its carboxyl terminus.


Asunto(s)
Péptidos Opioides/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides delta/fisiología , Animales , Diprenorfina/farmacología , Epítopos/metabolismo , Glioma , Ratones , Neuroblastoma , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/inmunología , Transfección , Células Tumorales Cultivadas
14.
Mol Pharmacol ; 50(4): 870-7, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8863832

RESUMEN

In neurons and transformed cell lines, opioid receptors are coupled to various signaling mechanisms involved in Ca2+ mobilization, including inhibition or activation of Ca2+ channels and phospholipase C-beta (PLC-beta), the enzyme responsible for generation of the Ca2+ mobilizing messenger inositol-1,4,5-trisphosphate [Ins(1,4,5)P3]. In the current study, we used selective PLC-beta and G protein antibodies to identify the PLC-beta isozyme activated by opioid receptors in intestinal smooth muscle and the G proteins to which the PLC-beta isozyme and adenylyl cyclase are coupled. [D-Pen2,D-Pen5]Enkephalin, a delta receptor agonist, stimulated Ins(1,4,5)P3 formation, Ca2+ release, and contraction; inhibited forskolin-stimulated cAMP formation in dispersed muscle cells; and stimulated phosphoinositide hydrolysis in plasma membranes; all of the effects were blocked by pertussis toxin. [D-Pen2,D-Pen5]Enkephalin-stimulated Ins(1,4,5)P3 formation, Ca2+ release, and contraction in permeabilized muscle cells and phosphoinositide hydrolysis in plasma membranes were selectively blocked by G beta antibody and PLC-beta 3 antibody; contractions stimulated by [D-Ala2,N-MePhe4,Gly-ol5]enkephalin, a mu receptor agonist, and U-69,593, a kappa receptor agonist, were also blocked by G beta and PLC-beta 3 antibodies. Inhibition of forskolin-stimulated cAMP formation by delta, mu, and kappa receptor agonists was partially blocked by G alpha i2 and G alpha o antibodies and additively blocked by a combination of the antibodies. The delta, mu, and kappa receptor agonists stimulated the binding of guanosine-5'-O-(3-thio)triphosphate to the alpha subunits of Gi2 and G(o) but not to the alpha subunits of other G proteins. We conclude that opioid mu, delta, and kappa receptors are selectively coupled to Gi2 and G(o) in intestinal smooth muscle. The beta gamma subunits of both G proteins activate PLC-beta 3, thereby stimulating Ins(1,4,5)P3-dependent Ca2+ release and smooth muscle contraction, whereas the alpha subunits inhibit adenylyl cyclase activity.


Asunto(s)
Inhibidores de Adenilato Ciclasa , Proteínas de Unión al GTP/fisiología , Isoenzimas/metabolismo , Músculo Liso/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores Opioides/fisiología , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/metabolismo , Animales , Calcio/metabolismo , Bovinos , Células Cultivadas , Activación Enzimática , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go , Proteínas de Unión al GTP/metabolismo , Cobayas , Mucosa Intestinal/metabolismo , Intestinos/fisiología , Intestinos/ultraestructura , Contracción Muscular/fisiología , Músculo Liso/enzimología , Músculo Liso/ultraestructura , Péptidos Opioides/agonistas , Fosfatidilinositoles/metabolismo , Fosfolipasa C beta , Receptores Opioides/metabolismo
15.
Endocrinology ; 135(5): 2137-43, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7956937

RESUMEN

Endogenous opiates exert a tonic restraint on LH release, which is centrally mediated through inhibition of hypothalamic GnRH secretion. The aim of the present study was, using an in vitro superfusion approach, to perform a detailed study of the effects of opioid receptor activation and blockade on the dimensions of pulsatile GnRH release from the isolated medial basal hypothalamus of the adult male guinea pig. The mu-receptor antagonist naloxone at 10(-3) M (n = 12), 10(-6) M (n = 12), and 10(-9) M (n = 12), the opioid agonist morphine sulfate at the same concentrations (n = 14 for each concentration), or morphine and naloxone together at 10(-3) M (n = 14), was added to the medium alternately either during the first or the second of two consecutive 3-h observation periods. Five-minute fractions (750 microliters) were collected for measurement of GnRH by RIA. Morphine suppressed total GnRH output from the medial basal hypothalami (P < or = 0.01 at 10(-3) M and 10(-6) M; P = NS at 10(-9) M), with mean pulse frequency, mean amplitude of all pulses, and mean sum of pulse amplitudes being significantly inhibited at all morphine concentrations, whether pulse analysis (Ultra pulse analysis algorithm) was performed at a two-coefficient of variation (2CV; P < or = 0.05, P < or = 0.01, and P < or = 0.01, respectively) or a 3CV (P < or = 0.05, P < or = 0.05, and P < or = 0.01, respectively) threshold. A different set of experiments (n = 14), with exposure to 10(-3) M morphine during the second of three consecutive 3-h observation periods, indicated that after morphine withdrawal a gradual recovery of GnRH output and pulse frequency to pretreatment values occurred, whereas pulse amplitude remained significantly suppressed during the posttreatment observation period. The inhibitory actions of 10(-3) M morphine were effectively prevented by the presence of 10(-3) M naloxone. Total GnRH output (P < or = 0.05), mean pulse frequency (P < or = 0.05), mean amplitude of all pulses (P < or = 0.05), and mean of the sum of pulse amplitudes (P < or = 0.01) were increased during opioid blockade with 10(-3) M and 10(-9) M naloxone (either 2CV or 3CV threshold for pulse analysis). A similar trend for 10(-6) M naloxone was only apparent after the exclusion of two outliers with unusually high basal GnRH release.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio/metabolismo , Péptidos Opioides/agonistas , Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Cobayas , Hipotálamo Medio/citología , Hormona Luteinizante/metabolismo , Masculino , Morfina/farmacología , Naloxona/farmacología , Narcóticos/metabolismo , Orquiectomía , Radioinmunoensayo , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA