Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Photodermatol Photoimmunol Photomed ; 33(2): 84-91, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28039905

RESUMEN

BACKGROUND: We previously reported that ultraviolet (UV) A eye irradiation reduces the ulcerative colitis induced by dextran sodium sulfate (DSS). This study examined the effects of UVA on colon carcinoma induced by azoxymethane (AOM) and DSS. METHODS: We irradiated the eyes of ICR mice with UVA at a dose of 110 kJ/m2 using an FL20SBLB-A lamp for the experimental period. RESULTS: In mice treated with these drugs, the symptom of colon carcinoma was reduced by UVA eye irradiation. The levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α in the blood were increased in AOM + DSS-treated mice; however, those levels were reduced by UVA eye irradiation. The expression of ß-endorphin, methionine-enkephalin (OGF), µ-opioid receptor, and opioid growth factor receptor (OGFR) of the colon was increased in the AOM + DSS-treated mice, and these levels were increased further following UVA eye irradiation. When ß-endorphin inhibitor was administered, the ameliorative effect of UVA eye irradiation was reduced, and the effect of eye irradiation disappeared entirely following the administration of naltrexone (inhibitor of both opioid receptor and OGFR). CONCLUSIONS: These results suggested that UVA eye irradiation exerts major effects on AOM + DSS-induced colon carcinoma.


Asunto(s)
Colon/metabolismo , Neoplasias del Colon/metabolismo , Encefalina Metionina/metabolismo , Ojo/efectos de la radiación , Rayos Ultravioleta , betaendorfina/metabolismo , Aminoquinolinas/farmacología , Animales , Azoximetano , Benzamidas/farmacología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Sulfato de Dextran , Femenino , Interleucina-6/sangre , Antígeno Ki-67/metabolismo , Ratones Endogámicos ICR , Naltrexona/farmacología , Receptores Opioides/metabolismo , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Factor de Necrosis Tumoral alfa/sangre , betaendorfina/antagonistas & inhibidores
2.
Alcohol Clin Exp Res ; 37(7): 1133-42, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23413810

RESUMEN

BACKGROUND: Prenatal exposure to ethanol (EtOH) reduces the expression of hypothalamic proopiomelanocortin (POMC) gene, known to control various physiological functions including the organismal stress response. In this study, we determined whether the changes in POMC neuronal functions are associated with altered expressions of histone-modifying and DNA-methylating enzymes in POMC-producing neurons, because these enzymes are known to be involved in regulation of gene expression. In addition, we tested whether gestational choline supplementation prevents the adverse effects of EtOH on these neurons. METHODS: Pregnant rat dams were fed with alcohol-containing liquid diet or control diet during gestational days 7 and 21 with or without choline, and their male offspring rats were used during the adult period. Using double-immunohistochemistry, real-time reverse transcription polymerase chain reaction (RT-PCR) and methylation-specific RT-PCR, we determined protein and mRNA levels of histone-modifying and DNA-methylating enzymes and the changes in POMC gene methylation and expression in the hypothalamus of adult male offspring rats. Additionally, we measured the basal- and lipopolysaccharide (LPS)-induced corticosterone levels in plasma by enzyme-linked immunosorbent assay. RESULTS: Prenatal EtOH treatment suppressed hypothalamic levels of protein and mRNA of histone activation marks (H3K4me3, Set7/9, acetylated H3K9, phosphorylated H3S10), and increased the repressive marks (H3K9me2, G9a, Setdb1), DNA-methylating enzyme (Dnmt1), and the methyl-CpG-binding protein (MeCP2). The treatment also elevated the level of POMC gene methylation, while it reduced levels of POMC mRNA and ß-EP and elevated corticosterone response to LPS. Gestational choline normalized the EtOH-altered protein and the mRNA levels of H3K4me3, Set7/9, H3K9me2, G9a, Setdb1, Dnmt1, and MeCP2. It also normalizes the changes in POMC gene methylation and gene expression, ß-EP production, and the corticosterone response to LPS. CONCLUSIONS: These data suggest that prenatal EtOH modulates histone and DNA methylation in POMC neurons that may be resulting in hypermethylation of POMC gene and reduction in POMC gene expression. Gestational choline supplementation prevents the adverse effects of EtOH on these neurons.


Asunto(s)
Colina/administración & dosificación , Metilación de ADN/efectos de los fármacos , Histonas/antagonistas & inhibidores , Efectos Tardíos de la Exposición Prenatal/prevención & control , Proopiomelanocortina/antagonistas & inhibidores , betaendorfina/antagonistas & inhibidores , Animales , Metilación de ADN/fisiología , Suplementos Dietéticos , Femenino , Histonas/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proopiomelanocortina/biosíntesis , Ratas , betaendorfina/biosíntesis
3.
Brain Behav Immun ; 24(7): 1045-53, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20385224

RESUMEN

Neuropathic pain is a debilitating consequence of nerve injuries and is frequently resistant to classical therapies. T lymphocytes mediate adaptive immune responses and have been suggested to generate neuropathic pain. In contrast, in this study we investigated T cells as a source of opioidergic analgesic ß-endorphin for the control of augmented tactile sensitivity following neuropathy. We employed in vivo nociceptive (von Frey) testing, flow cytometry and immunofluorescence in wild-type and mice with severe combined immunodeficiency (SCID) subjected to a chronic constriction injury of the sciatic nerve. In wild-type mice, T lymphocytes constituted approximately 11% of all immune cells infiltrating the injury site, and they expressed ß-endorphin and receptors for corticotropin-releasing factor (CRF), an agent releasing opioids from leukocytes. CRF applied at the nerve injury site fully reversed neuropathy-induced mechanical hypersensitivity in wild-type animals. In SCID mice, T cells expressing ß-endorphin and CRF receptors were absent at the damaged nerve. Consequently, these animals had substantially reduced CRF-mediated antinociception. Importantly, the decreased antinociception was fully restored by transfer of wild-type mice-derived T lymphocytes in SCID mice. The re-established CRF antinociception could be reversed by co-injection of an antibody against ß-endorphin or an opioid receptor antagonist with limited access to the central nervous system. We propose that, in response to CRF stimulation, T lymphocytes accumulating at the injured nerves utilize ß-endorphin for activation of local neuronal opioid receptors to reduce neuropathy-induced mechanical hypersensitivity. Our findings reveal ß-endorphin-containing T cells as a crucial component of beneficial adaptive immune responses associated with painful peripheral nerve injuries.


Asunto(s)
Neuralgia/inmunología , Neuralgia/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Nervio Ciático/lesiones , Linfocitos T/metabolismo , betaendorfina/metabolismo , Analgésicos/metabolismo , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Antagonistas de Narcóticos , Dimensión del Dolor/métodos , Umbral del Dolor/psicología , Linfocitos T/inmunología , betaendorfina/antagonistas & inhibidores
4.
Dev Comp Immunol ; 32(4): 330-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17651799

RESUMEN

In this in vitro study, the role of beta-endorphin in the control of phagocytic and cytotoxic activities of fish splenic phagocytes was investigated. Further, the involvement of specific opioid receptor was explored. beta-Endorphin stimulated phagocytosis, whereas inhibited nitric oxide production as assessed by nitrite release. However, it had concentration-related biphasic effects on superoxide production, stimulatory at low and inhibitory at high concentration. Naltrexone, non-selective opioid receptor antagonist, antagonized the effect of beta-endorphin on phagocyte functions. Moreover, CTAP, selective mu-receptor antagonist, completely blocked the effect of beta-endorphin on phagocytosis and nitrite release. With regard to superoxide production, CTAP blocked the stimulatory effect of beta-endorphin at low concentration, while the inhibitory effect at high concentration was completely antagonized by selective delta-receptor antagonist, NTI. In conclusion, beta-endorphin acting via mu-receptor stimulated phagocytosis and inhibited nitric oxide production, while its biphasic effect on superoxide production seems to be mediated by mu- and delta-receptors.


Asunto(s)
Fagocitos/metabolismo , Fagocitosis , Receptores Opioides/metabolismo , betaendorfina/metabolismo , Animales , Peces , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Óxido Nítrico/metabolismo , Fragmentos de Péptidos , Péptidos/farmacología , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Somatostatina , Bazo/citología , Bazo/metabolismo , Superóxidos/metabolismo , betaendorfina/antagonistas & inhibidores
5.
East Mediterr Health J ; 13(2): 257-65, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17684846

RESUMEN

To address the role of the opioid system in the pathogenesis of hepatic encephalopathy (HE) we measured plasma met-enkephalin, beta-endorphin and leu-enkephalin in patients with different grades of HE compared to control subjects and patients with cirrhosis. Plasma met-enkephalin levels were significantly higher in patients with cirrhosis and all grades of HE than controls. Plasma beta-endorphin levels were similar in the 3 groups. Plasma leu-enkephalin levels were significantly higher in HE grades II, III and IV than in controls, patients with cirrhosis and HE grade I patients. Our results support data on the involvement of met-enkephalin and leu-enkephalin in the pathogenesis of HE and provide a rationale for the use of opioid receptor antagonists in the treatment of HE.


Asunto(s)
Encefalina Leucina/sangre , Encefalina Metionina/sangre , Encefalopatía Hepática/sangre , Encefalopatía Hepática/etiología , Neurotransmisores/sangre , betaendorfina/sangre , Amoníaco/sangre , Análisis de Varianza , Estudios de Casos y Controles , Causalidad , Egipto , Encefalina Leucina/antagonistas & inhibidores , Encefalina Metionina/antagonistas & inhibidores , Encefalopatía Hepática/clasificación , Encefalopatía Hepática/tratamiento farmacológico , Hepatitis B/complicaciones , Hepatitis C/complicaciones , Hepatitis Autoinmune/complicaciones , Humanos , Cirrosis Hepática/sangre , Cirrosis Hepática/clasificación , Cirrosis Hepática/etiología , Tasa de Depuración Metabólica , Naltrexona/análogos & derivados , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Neurotransmisores/antagonistas & inhibidores , Radioinmunoensayo , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas , betaendorfina/antagonistas & inhibidores
6.
Arch Pharm Res ; 28(2): 227-31, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15789756

RESUMEN

We examined the effect of the subcutaneous (s.c.) pretreatment of formalin into both hind paws of mice on the antinociception induced by the intracerebroventricularly (i.c.v.) or intrathecally (i.t.) administration of beta-endorphin using the tail-flick test. Pretreatment with formalin (5%) for 5 h had no affect on the i.c.v. administered beta-endorphin-induced tail-flick response. However, pretreatment with formalin for 40 h attenuated the tail-flick inhibition induced by i.c.v. administered beta-endorphin. This antinociceptive tolerance to i.c.v. beta-endorphin continued up to 1 week, but to a lesser extent. Pretreatment with formalin for 5 and 40 h significantly reduced the i.t. beta-endorphin-induced inhibition of the tail-flick response, which continued up to 1 week. The s.c. formalin treatment increased the hypothalamic pro-opiomelanocortin (POMC) mRNA level at 2 h, but this returned to the basal level after 40 h. Our results suggest that the increase in the POMC mRNA level in the hypothalamus appears to be involved in the supraspinal or spinal beta-endorphin-induced antinociceptive tolerance in formalin-induced inflammatory pain.


Asunto(s)
Formaldehído , Dimensión del Dolor/efectos de los fármacos , Tiempo de Reacción/efectos de los fármacos , betaendorfina/antagonistas & inhibidores , betaendorfina/farmacología , Animales , Northern Blotting , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Inyecciones Espinales , Masculino , Ratones , Ratones Endogámicos ICR , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/fisiología , ARN Mensajero/biosíntesis , betaendorfina/administración & dosificación
7.
Hypertension ; 29(1 Pt 1): 105-10, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9039088

RESUMEN

To investigate the involvement of endogenous opioids in acute increases in blood pressure and their functional relationship with atrial natriuretic factor and endothelin-1, we assessed plasma levels of beta-endorphin, met-enkephalin, dynorphin B, catecholamines, atrial natriuretic factor, and endothelin-1 before and after administration of the opioid antagonist naloxone hydrochloride (8 mg i.v.) in 28 hypertensive patients with a stress-induced acute increase in blood pressure. Ten patients with established mild or moderate essential hypertension and 10 normotensive subjects served as control groups. Opioids, atrial natriuretic factor, and endothelin-I were radioimmunoassayed after chromatographic preextraction; catecholamines were determined by high-performance liquid chromatography with electrochemical detection. Patients with an acute increase in blood pressure (systolic, 203.2 +/- 2.2 mm Hg; diastolic, 108.4 +/- 1.3) had plasma opioid, catecholamine, and atrial natriuretic factor levels significantly (P < .01) higher than hypertensive control patients (systolic pressure, 176.4 +/- 1.0 mm Hg; diastolic, 100.0 +/- 1.4), who had a hormonal pattern similar to that of normotensive subjects (systolic pressure, 123.2 +/- 1.5 mm Hg; diastolic, 75.0 +/- 2.0). Endothelin-1 did not differ in any group. In patients with an acute increase in blood pressure, naloxone significantly (P < .01) reduced blood pressure, heart rate, opioids, catecholamines, and atrial natriuretic factor 10 minutes after administration. Naloxone effects on blood pressure, heart rate, opioids, and catecholamines wore off within 20 minutes. In control groups, naloxone failed to modify any of the considered parameters. Our findings suggest that pressor effects of opioid peptides mediated by the autonomic nervous system during stress-induced acute episodes of blood pressure increase in hypertensive patients.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Péptidos Opioides/fisiología , Factor Natriurético Atrial/antagonistas & inhibidores , Factor Natriurético Atrial/sangre , Factor Natriurético Atrial/fisiología , Presión Sanguínea/efectos de los fármacos , Dinorfinas/antagonistas & inhibidores , Dinorfinas/sangre , Dinorfinas/fisiología , Endorfinas/antagonistas & inhibidores , Endorfinas/sangre , Endorfinas/fisiología , Endotelina-1/antagonistas & inhibidores , Endotelina-1/sangre , Endotelina-1/fisiología , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/sangre , Encefalina Metionina/fisiología , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Humanos , Hipertensión/sangre , Hipertensión/etiología , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Naloxona/administración & dosificación , Naloxona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Norepinefrina/antagonistas & inhibidores , Norepinefrina/sangre , Norepinefrina/fisiología , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/sangre , Estrés Fisiológico/complicaciones , Estrés Psicológico/complicaciones , betaendorfina/antagonistas & inhibidores , betaendorfina/sangre , betaendorfina/fisiología
8.
Pain ; 104(1-2): 55-63, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12855314

RESUMEN

Although exogenous administration of beta-endorphin to the arcuate nucleus of hypothalamus (ARC) had been shown to produce antinociception, the role of endogenous beta-endorphin of the ARC in nociceptive processing has not been studied directly. The aim of the present study was to investigate the effect of endogenous beta-endorphin in the ARC on nociception in rats with carrageenan-induced inflammation. The hindpaw withdrawal latency (HWL) to noxious thermal and mechanical stimulation was assessed by the hot-plate test and the Randall Selitto Test. Intra-ARC injection of naloxone had no significant influence on the HWL to thermal and mechanical stimulation in intact rats. The HWL decreased significantly after intra-ARC injection of 1 or 10 microg of naloxone in rats with inflammation, but not with 0.1 microg of naloxone. Furthermore, intra-ARC administration of the selective mu-opioid receptor antagonist beta-funaltrexamine (beta-FNA) decreased the nociceptive response latencies to both stimulation in a dose-dependent manner in rats with inflammation, while intra-ARC administration of the selective delta-opioid receptor antagonist naltrindole or the selective kappa-opioid receptor antagonist nor-binaltorphimine (nor-BNI) showed no influences on the nociceptive response latency. The antiserum against beta-endorphin, administered to the ARC, also dose-dependently reduced the HWL in rats with inflammation. The results indicate that endogenous beta-endorphin in the ARC plays an important role in the endogenous antinociceptive system in rats with inflammation, and that its effect is predominantly mediated by the mu-opioid receptor.


Asunto(s)
Analgésicos/uso terapéutico , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Inflamación/tratamiento farmacológico , betaendorfina/antagonistas & inhibidores , betaendorfina/uso terapéutico , Analgésicos/antagonistas & inhibidores , Animales , Núcleo Arqueado del Hipotálamo/fisiología , Sueros Inmunes/farmacología , Inflamación/inducido químicamente , Masculino , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Wistar , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/fisiología , betaendorfina/fisiología
9.
Neuropharmacology ; 27(9): 957-63, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2972939

RESUMEN

beta-Endorphin-(1-27), administered intraventricularly has been previously reported to block the analgesia induced by beta-endorphin injected intraventricularly. The present study was to determine if the blocking effect of beta-endorphin-(1-27) was specific to beta-endorphin which stimulates epsilon receptors, but not to other opioids with activity at different opioid receptors. The antagonistic effects of beta-endorphin-(1-27) on the analgesia induced by beta-endorphin (epsilon-opioid receptor agonist), D-Ala2-NMePhe4-Gly-ol-enkephalin(DAGO) and morphine, (mu-opioid receptor agonists), D-Pen2-D-Pen5-enkephalin(DPDPE) and D-Ala2-D-Leu5-enkephalin(DADLE) (delta-opioid receptor agonists) and U-50, 488H (kappa-opioid receptor agonist) were studied. beta-Endorphin-(1-27) injected intraventricularly, at doses which, when injected alone did not produce analgesia, antagonized the analgesia induced by beta-endorphin given intraventricularly. However, the analgesia induced by DAGO, morphine, DPDPE, DADLE and U-50, 488H given intraventricularly was not antagonized by beta-endorphin-(1-27). The data suggest that beta-endorphin-(1-27) selectively blocks the analgesia induced by the stimulation of epsilon receptors but not by the stimulation of mu, delta, and kappa receptors. The results support the previously proposed hypothesis that beta-endorphin produces its analgesia by stimulating specific epsilon receptors.


Asunto(s)
Analgésicos/antagonistas & inhibidores , Encefalinas/antagonistas & inhibidores , Morfina/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Pirrolidinas/antagonistas & inhibidores , betaendorfina/antagonistas & inhibidores , betaendorfina/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5) , Encefalina D-Penicilamina (2,5) , Encefalina Leucina/análogos & derivados , Encefalina Leucina/antagonistas & inhibidores , Leucina Encefalina-2-Alanina , Masculino , Ratones , Ratones Endogámicos ICR , Tiempo de Reacción/efectos de los fármacos
10.
Neuroscience ; 78(2): 461-7, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9145802

RESUMEN

The antinociception induced by beta-endorphin given supraspinally has been demonstrated previously to be mediated by the release of Met-enkephalin acting on delta2-opioid receptors in the spinal cord. The present study was designed to determine the role of nitric oxide in the spinal cord on beta-endorphin-induced release of Met-enkephalin and antinociception. The experiments were performed in pentobarbital-anesthetized rats. The release of Met-enkephalin was performed using a spinal cord perfusion technique and the Met-enkephalin released in the spinal perfusates was measured by radioimmunoassay. Antinociception was assessed by the tail-flick test. beta-Endorphin (2 microg) given intraventricularly induced the release of Met-enkephalin from the spinal cord. The release of Met-enkephalin was dose-dependently attenuated by N(omega)-nitro-L-arginine (0.1 nM-1 microM) added into spinal perfusates and the attenuation was reversed by intrathecally applied L-arginine. The stereoisomer N(omega)-nitro-D-arginine given intrathecally, however, did not inhibit the release of Met-enkephalin induced by intraventricularly administered beta-endorphin. beta-Endorphin (4 microg) given intraventricularly produced antinociception in rats pretreated intrathecally with saline. The antinociception induced by beta-endorphin was blocked by intrathecally administered N(omega)-nitro-L-arginine (5 microg) and the blockade of antinociception was reversed by intrathecal injection of L-arginine (50 microg). N(omega)-Nitro-D-arginine (5 microg) given intrathecally did not block the intraventricularly administered beta-endorphin-induced antinociception. N(omega)-Nitro-L-arginine (10 microg) given intraventricularly did not affect intraventricularly administered beta-endorphin-induced Met-enkephalin release nor did it affect intraventricular beta-endorphin-induced antinociception, indicating that the effect of N(omega)-nitro-L-arginine is not at supraspinal sites. Intrathecal pretreatment with N(omega)-nitro-L-arginine did not affect intrathecally administered [D-Ala2]deltorphin II-induced antinociception. Our results indicate that N(omega)-nitro-L-arginine given intrathecally attenuates intraventricular beta-endorphin-administered inhibition of the tail-flick response by presynaptically inhibiting the release of Met-enkephalin.


Asunto(s)
Analgésicos/antagonistas & inhibidores , Encefalina Metionina/metabolismo , Inhibidores Enzimáticos/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitroarginina/farmacología , betaendorfina/antagonistas & inhibidores , Analgésicos/administración & dosificación , Analgésicos/farmacología , Animales , Inhibidores Enzimáticos/administración & dosificación , Inyecciones Intraventriculares , Inyecciones Espinales , Masculino , Nitroarginina/administración & dosificación , Oligopéptidos/farmacología , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , betaendorfina/administración & dosificación , betaendorfina/farmacología
11.
Neuroscience ; 115(3): 715-21, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12435410

RESUMEN

beta-Endorphin is a non-selective opioid peptide which binds mu-, delta- and putative epsilon (beta-endorphin-sensitive non-mu-, non-delta- and non-kappa(1)-)-opioid receptors. We have previously reported that beta-endorphin-produced G-protein activation is mediated by the stimulation of both mu- and putative epsilon-opioid receptors. The present study was designed to further characterize this putative epsilon-opioid receptor-mediated G-protein activation in the pons/medulla membrane obtained from mice lacking mu-opioid receptor, using a guanosine-5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS)-binding assay. beta-Endorphin and the mu-opioid receptor agonist [D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO) increased the [(35)S]GTPgammaS binding in a concentration-dependent manner (0.001-10 microM), and at 10 microM beta-endorphin and DAMGO produced approximately 250 and 120% increases of [(35)S]GTPgammaS binding in the pons/medulla membrane obtained from wild-type mice, respectively. In the pons/medulla membrane obtained from mu-opioid receptor knockout mice, beta-endorphin-stimulated [(35)S]GTPgammaS binding was only partially attenuated and a more than 100% increase by 10 microM beta-endorphin still remained, while DAMGO failed to produce any increase in [(35)S]GTPgammaS binding. The residual increase in [(35)S]GTPgammaS binding by 10 microM beta-endorphin in mu-opioid receptor knockout mice was partially but significantly attenuated by the putative epsilon-opioid receptor partial agonist beta-endorphin (1-27), but not by the delta-opioid receptor antagonist naltrindole or the kappa(1)-receptor antagonist norbinaltorphimine. Furthermore, buprenorphine significantly attenuated the residual increase in [(35)S]GTPgammaS binding by 10 microM beta-endorphin in mu-opioid receptor knockout mice. The present results indicate that beta-endorphin activates G-protein by stimulation of putative epsilon-opioid receptors in the condition lacking the mu-opioid receptor, and buprenorphine acts as an antagonist for putative epsilon-opioid receptors in this condition.


Asunto(s)
Buprenorfina/farmacología , Proteínas de Unión al GTP/metabolismo , Naltrexona/análogos & derivados , Narcóticos/farmacología , Receptores Opioides mu/deficiencia , Receptores Opioides/metabolismo , Rombencéfalo/metabolismo , betaendorfina/metabolismo , Animales , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Femenino , Proteínas de Unión al GTP/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato) , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/metabolismo , Ratones , Ratones Noqueados , Naltrexona/farmacología , Antagonistas de Narcóticos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Puente/efectos de los fármacos , Puente/metabolismo , Ensayo de Unión Radioligante , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/genética , Rombencéfalo/efectos de los fármacos , Radioisótopos de Azufre , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , betaendorfina/antagonistas & inhibidores
12.
Br J Pharmacol ; 113(4): 1416-22, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7889299

RESUMEN

1. Endogenous corticosteroids and opioids are involved in many functions of the organism, including analgesia, cerebral excitability, stress and others. Therefore, we considered it important to gain information on the functional interaction between corticosteroids and specific opioid receptor subpopulations. 2. We have found that systemic administration (i.p.) of the potent synthetic corticosteroid, dexamethasone, reduced the antinociception induced by the highly selective mu agonist, DAMGO or by less selective mu agonists morphine and beta-endorphin administered i.c.v.. On the contrary dexamethasone exerted little or no influence on the antinociception induced by a delta 1 agonist, DPDPE and a delta 2 agonist deltorphin II. Dexamethasone potentiated the antinociception induced by the kappa agonist, U50,488. 3. In experiments performed in an in vitro model of cerebral excitability in the rat hippocampal slice, dexamethasone strongly prevented both the increase of the duration of the field potential recorded in CA1, and the appearance and number of additional population spikes induced by mu receptor agonists. 4. In both models pretreatment with cycloheximide, a protein synthesis inhibitor, prevented the antagonism by dexamethasone of responses to the mu opioid agonists. 5. Our data indicate that in the rodent brain there is an important functional interaction between the corticosteroid and the opioid systems at least at the mu receptor level, while delta and kappa receptors are modulated in different ways.


Asunto(s)
Analgésicos/antagonistas & inhibidores , Dexametasona/farmacología , Receptores Opioides mu/antagonistas & inhibidores , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero , Analgésicos/farmacología , Animales , Electrofisiología , Encefalina Ala(2)-MeFe(4)-Gli(5) , Encefalina D-Penicilamina (2,5) , Encefalinas/antagonistas & inhibidores , Encefalinas/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Masculino , Ratones , Morfina/antagonistas & inhibidores , Morfina/farmacología , Oligopéptidos/antagonistas & inhibidores , Oligopéptidos/farmacología , Dimensión del Dolor/efectos de los fármacos , Pirrolidinas/farmacología , Ratas , Ratas Wistar , betaendorfina/antagonistas & inhibidores , betaendorfina/farmacología
13.
Neuroreport ; 15(3): 519-21, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15094515

RESUMEN

Endogenous beta-endorphin levels in the brain are elevated in response to cocaine and are downstream of the mesolimbic dopaminergic system. However, beta-endorphin's direct involvement in cocaine reinforcement has not been demonstrated. In the present study, a single bilateral microinjection of anti-beta-endorphin antibodies (4 microg) to the nucleus accumbens during the maintenance phase of cocaine self-administration (1 mg/kg/infusion) significantly increased the number of active and inactive lever responses. The increase in lever responses is reminiscent of rat behavior during extinction of cocaine self-administration. Further, a cocaine dose-response demonstrates that the increased lever presses in anti-beta-endorphin antibody-injected rats was still present after substitution with a lower dose of cocaine. These findings support a critical role for beta-endorphin in the cocaine brain reward system.


Asunto(s)
Conducta Animal/fisiología , Trastornos Relacionados con Cocaína/psicología , betaendorfina/fisiología , Animales , Anticuerpos Bloqueadores/farmacología , Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Microinyecciones , Núcleo Accumbens , Ratas , Ratas Sprague-Dawley , Recompensa , Autoadministración , betaendorfina/antagonistas & inhibidores
14.
J Physiol Paris ; 95(1-6): 215-20, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11595440

RESUMEN

Neonatal monosodium glutamate treatment reduced immunoreactive beta-endorphin content in the mediobasal hypothalamus by 50% in adult, male Wistar rats as compared to hypertonic saline-treated littermates; there was also a moderate (approx. 25%) reduction in the rostral part of the nucleus of the solitary tract. In sham-treated adults the intracisternally injected alpha-2 adenoceptor stimulant clonidine (0.47 nmol/rat) and the delta opioid receptor type agonist (D-Ala(2), D-Leu(5))-enkephalin (0.8 nmol/rat) reduced acidified ethanol-induced mucosal lesions in the stomach by 84.1 and 77.5%, respectively, whereas the same doses were completely ineffective in rats treated neonatally by monosodium glutamate. The data taken together with the results of previous studies with the same substances in rats with retroarcuate knife cuts suggest that neuronal damage in the nucleus of the solitary tract region rather than in the arcuate nucleus is responsible for the changes seen in the pharmacological responsiveness.


Asunto(s)
Animales Recién Nacidos/fisiología , Tronco Encefálico/fisiología , Citoprotección/efectos de los fármacos , Receptores Adrenérgicos alfa/fisiología , Receptores Opioides delta/fisiología , Glutamato de Sodio/farmacología , Estómago/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Tronco Encefálico/efectos de los fármacos , Clonidina/farmacología , Leucina Encefalina-2-Alanina/farmacología , Crecimiento/efectos de los fármacos , Masculino , Neurotransmisores/metabolismo , Pupila/efectos de la radiación , Ratas , Ratas Wistar , betaendorfina/antagonistas & inhibidores , betaendorfina/metabolismo
15.
Regul Pept ; 63(2-3): 79-83, 1996 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-8837214

RESUMEN

This study investigates the role of the opiod receptors and of the opioid peptide beta-endorphin in the development of yeast-induced inflammation in the rat paw. Pretreatment with the opioid receptor antagonist naltrexone (10 mg/kg i.p.) exacerbates the paw edema, while morphine pretreatment (5 and 10 mg/kg) reduces it. In addition, the intravenous injection of a specific anti-beta-endorphin antibody aggravates the yeast-induced inflammation. On the contrary, both the kappa-opioid receptor antagonist MR 1452 (2.5, 5 and 10 mg/kg i.p.) and the delta-opioid receptor antagonist ICI 174-864 (2.5, 5 and 10 mg/kg i.p.) do not interfere with the inflammatory process. After intraplantar injection, naltrexone, morphine and the anti-beta-endorphin antibody do not interfere with the yeast-induced inflammatory edema. Our data suggest that beta-endorphin exerts an inhibitory regulation on the inflammatory responses through the activation of mu-opioid receptors probably located on immune cells, rather than in the paw.


Asunto(s)
Edema/metabolismo , Inflamación/metabolismo , Receptores Opioides/metabolismo , betaendorfina/farmacología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Benzomorfanos/farmacología , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacología , Pie , Masculino , Morfina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley , Levaduras/metabolismo , betaendorfina/antagonistas & inhibidores , betaendorfina/inmunología
16.
Neuropeptides ; 21(3): 131-7, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1321361

RESUMEN

The effects of cholecystokinin octapeptide (CCK8s) given intrathecally (i.t.) on antinociception and the release of immunoreactive Met-enkephalin in the spinal perfusate induced by intraventricular (i.vt.) injection of beta-endorphin were studied in anesthetized rats. beta-Endorphin (5 micrograms) given i.vt. inhibited the tail-flick response. The inhibition of the tail-flick response induced by beta-endorphin was blocked dose-dependently by CCK8s (0.1-7 micrograms) given i.t. The antagonistic effect of CCK8s on beta-endorphin-induced inhibition was blocked dose dependently by co-intrathecal injection of proglumide (3 and 10 micrograms), a CCK8s receptor antagonist. beta-Endorphin (5 micrograms) given i.vt. elicited a release of immunoreactive Met-enkephalin in the spinal perfusate. Repeated injections of the same dose of beta-endorphin released about the same amount of the immunoreactive Met-enkephalin in the spinal perfusate. CCK8s at concentrations from 1 x 10(-9) to 1 x 10(-6) M added into the spinal perfusate decreased the release of Met-enkephalin induced by beta-endorphin given i.vt. in a dose-dependent manner. The results suggest that CCK8s may attenuate beta-endorphin-induced inhibition of the tail-flick response by inhibiting the release of Met-enkephalin from the spinal cord.


Asunto(s)
Encefalina Metionina/metabolismo , Dolor/fisiopatología , Sincalida/farmacología , Animales , Depresión Química , Inyecciones Intraventriculares , Inyecciones Espinales , Antagonistas de Narcóticos , Proglumida/farmacología , Ratas , Ratas Endogámicas , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Receptores Opioides/fisiología , Receptores Opioides delta , Reflejo/efectos de los fármacos , Tasa de Secreción/efectos de los fármacos , Médula Espinal/fisiopatología , Cola (estructura animal) , betaendorfina/administración & dosificación , betaendorfina/antagonistas & inhibidores , betaendorfina/farmacología
17.
Brain Res ; 661(1-2): 19-24, 1994 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-7834370

RESUMEN

Previously, we have shown that intracisternal (i.c.) administration of beta-endorphin suppresses brain and liver DNA synthesis in rat pups. This finding is consistent with the view that endogenous CNS beta-endorphin plays an important role in controlling postnatal growth. Recent evidence suggests that brain CCK8, the sulfated carboxyterminal octapeptide fragment of cholecystokinin, may function physiologically as an endogenous opioid antagonist. We now report that CCK8 injected i.c. together with beta-endorphin effectively prevented beta-endorphin from inhibiting brain and liver DNA synthesis in 10-day-old rats. CCK8 blocked the liver DNA effect of beta-endorphin via actions within the brain, as subcutaneous administration of CCK8 was ineffective. In contrast to CCK8, i.c. administration of CCK8U (the unsulfated form of CCK8) together with beta-endorphin did not prevent beta-endorphin from inhibiting liver DNA synthesis, and only slightly reversed the brain DNA effect. The results obtained support a role for endogenous brain CCK8 in the modulation of tissue DNA responses to CNS beta-endorphin and possibly to other endogenous opioids. If so, interference with brain CCK function could disrupt tissue growth. Thus, normal mammalian development may require a close functional interaction between the cholecystokinin and beta-endorphin systems in the brain.


Asunto(s)
Encéfalo/metabolismo , Cerebelo/fisiología , ADN/biosíntesis , Hígado/metabolismo , Sincalida/farmacología , betaendorfina/farmacología , Animales , Encéfalo/efectos de los fármacos , Cerebelo/efectos de los fármacos , Femenino , Inyecciones Subcutáneas , Lactancia , Hígado/efectos de los fármacos , Microinyecciones , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Sincalida/administración & dosificación , Sincalida/antagonistas & inhibidores , betaendorfina/administración & dosificación , betaendorfina/antagonistas & inhibidores
18.
Brain Res ; 707(2): 189-95, 1996 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-8919295

RESUMEN

We evaluated the effects of intracerebroventricular (i.c.v.) administration of beta-endorphin and naloxone, an opioid antagonist, on the induction of c-fos and corticotropin-releasing factor (CRF) mRNA to clarify the effects of beta-endorphin on cellular activity and CRF gene expression in the paraventricular nucleus (PVN) of the rat using in situ hybridization. A significant induction of c-fos mRNA was noted in the PVN after i.c.v. injection of beta-endorphin, compared to control. This induction was inhibited by the administration of naloxone. A significant increase in CRF mRNA levels in the PVN was observed 120 min after the i.c.v. injection of beta-endorphin. This increase was partially, but significantly, inhibited by naloxone administration. In addition, i.c.v. administration of beta-endorphin increased plasma ACTH concentration in freely moving rats, which was inhibited by intravenous injection of CRF antiserum. These results suggest that the i.c.v. injection of beta-endorphin increases the neuronal activity and the biosynthesis of CRF in the PVN, and stimulates the secretion of ACTH by increasing CRF secretion. This effect on the PVN was mediated, at least in part, via the opioid receptor.


Asunto(s)
Hormona Liberadora de Corticotropina/biosíntesis , Expresión Génica/efectos de los fármacos , Genes fos/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/biosíntesis , betaendorfina/farmacología , Animales , Autorradiografía , Densitometría , Hibridación in Situ , Inyecciones Intraventriculares , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Wistar , betaendorfina/administración & dosificación , betaendorfina/antagonistas & inhibidores
19.
Brain Res ; 741(1-2): 13-26, 1996 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-9001699

RESUMEN

In addition to brainstem sites of action, analgesia can be elicited following amygdala microinjections of morphine and mu-selective opioid agonists. The present study examined whether opioid analgesia elicited by either morphine or beta-endorphin in the amygdala could be altered by either the general opioid antagonist, naltrexone, the mu-selective antagonist, beta-funaltrexamine (BFNA) or the delta 2 antagonist, naltrindole isothiocyanate (Ntii) in the periaqueductal gray (PAG). Both morphine (2.5-5 micrograms) and beta-endorphin (2.5-5 micrograms) microinjected into either the baso-lateral or central nuclei of the amygdala significantly increased tail-flick latencies and jump thresholds in rats. The increases were far more pronounced on the jump test than on the tail-flick test. Placements dorsal and medial to the amygdala were ineffective. Naltrexone (1-5 micrograms) in the PAG significantly reduced both morphine (tail-flick: 70-75%; jump: 60-81%) and beta-endorphin (tail-flick: 100%; jump: 93%) analgesia elicited from the amygdala, indicating that an opioid synapse in the PAG was integral for the full expression of analgesia elicited from the amygdala by both agonists. Both BFNA (68%) and Ntii (100%) in the PAG significantly reduced morphine, but not beta-endorphin analgesia in the amygdala on the tail-flick test. Ntii in the PAG was more effective in reducing morphine (60%) and beta-endorphin (79%) analgesia in the amygdala on the jump test than BFNA (15-24%). Opioid agonist-induced analgesia in the amygdala was unaffected by opioid antagonists administered into control misplacements in the lateral mesencephalon, and the small hyperalgesia elicited by opioid antagonists in the PAG could not account for the reductions in opioid agonist effects in the amygdala. These data indicate that PAG delta 2, and to a lesser degree, mu opioid receptors are necessary for the full expression of morphine and beta-endorphin analgesia elicited from the amygdala.


Asunto(s)
Amígdala del Cerebelo/fisiología , Analgésicos Opioides/antagonistas & inhibidores , Morfina/antagonistas & inhibidores , Antagonistas de Narcóticos/farmacología , Sustancia Gris Periacueductal/fisiología , betaendorfina/antagonistas & inhibidores , Amígdala del Cerebelo/anatomía & histología , Analgésicos Opioides/farmacología , Animales , Masculino , Microinyecciones , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/anatomía & histología , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , betaendorfina/farmacología
20.
Brain Res ; 747(1): 52-9, 1997 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-9042527

RESUMEN

Glycyl-L-glutamine (Gly-Gln; beta-endorphin 30-31) is an endogenous dipeptide that is synthesized through the post-translational processing of beta-endorphin. Previously, we showed that Gly-Gln inhibits the hypotension and respiratory depression produced by central beta-endorphin administration. In this study, we tested whether cyclo(Gly-Gln), a non-polar, cyclic Gly-Gln derivative, was similarly effective following intracerebro-ventricular (i.c.v.) or intra-arterial (i.a.) administration to pentobarbital-anesthetized rats pretreated with beta-endorphin (0.5 nmol i.c.v.). Intracerebroventricular cyclo(Gly-Gln) (0.3, 0.6 or 1.0 nmol) injection produced a dose-dependent inhibition of beta-endorphin-induced hypotension, but not bradycardia, with a potency similar to that of Gly-Gln. Cyclo(Gly-Gln) (5 mg/kg) was also effective following i.a. injection and significantly attenuated the fall in arterial pressure elicited by i.c.v. beta-endorphin, consistent with evidence that cyclic dipeptides permeate the blood-brain barrier; i.a. Gly-Gln was ineffective. Intra-arterial cyclo(Gly-Gln) (5 mg/kg) and i.c.v. Gly-Gln (10 nmol) also attenuated the hypotension and respiratory depression induced by morphine (50 or 100 nmol i.c.v.). Cyclo(Gly-Gln) (0.5, 5.0 or 50.0 mg/kg i.a.) had no effect on arterial pressure or heart rate when given alone. These findings indicate that cyclo(Gly-Gln) is a biologically active peptide capable of reversing the cardiorespiratory depression produced by beta-endorphin or morphine.


Asunto(s)
Hemodinámica/efectos de los fármacos , Morfina/antagonistas & inhibidores , Narcóticos/farmacología , Mecánica Respiratoria/efectos de los fármacos , betaendorfina/antagonistas & inhibidores , Animales , Presión Sanguínea/efectos de los fármacos , Depresión Química , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Inyecciones Intraarteriales , Inyecciones Intraventriculares , Masculino , Morfina/administración & dosificación , Morfina/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/prevención & control , betaendorfina/administración & dosificación , betaendorfina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA