Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Med Hypotheses ; 70(3): 556-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17692472

RESUMEN

A substantial number of patients with chronic hepatitis C does not respond to treatment with interferon and ribavirin, the approved drugs to treat this viral infection. In vitro studies have shown that morphine, which exerts its effects by binding to opioid receptors, enhances the expression of hepatitis C RNA in hepatitis C-replicon containing liver cells, and that it interferes with the antiviral effect of interferon on the hepatitis C virus. Met-enkephalin, one of the endogenous opioid peptides, can bind to the same receptors to which morphine binds, triggering similar receptor-mediated effects. The liver in cholestasis can express Met-enkephalin immunoreactivity (MEIR). MEIR can also be detected in the liver of some patients with chronic hepatitis C. This finding suggests that Met-enkephalin is produced by or that it accumulates in the liver of patients with this viral hepatitis. Analogous to the effect of morphine on the hepatitis C-replicon containing liver cells, we hypothesize that Met-enkephalin enhances the replication of the hepatitis C virus in liver cells, and that it interferes with the antiviral effect of interferon, contributing to the lack of efficacy of this medication in the treatment of this viral infection in some patients. If this hypothesis is correct, the study of opiate antagonists in combination with antiviral therapy in patients with hepatitis C expressing MEIR in their livers merits consideration.


Asunto(s)
Hepatitis C Crónica/tratamiento farmacológico , Interferones/uso terapéutico , Hígado/fisiopatología , Péptidos Opioides/antagonistas & inhibidores , Encefalina Metionina/antagonistas & inhibidores , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Humanos , Interferones/efectos adversos , Morfina/farmacología , ARN Viral/efectos de los fármacos , ARN Viral/genética
2.
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
3.
Peptides ; 91: 40-48, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28363796

RESUMEN

We previously reported a series of novel endomorphin analogs with unnatural amino acid modifications. These analogs display good binding affinity and functional activity toward the µ opioid receptor (MOP). In the present study, we further investigated the spinal antinociceptive activity of these compounds. The analogs were potent in several nociceptive models. Opioid antagonists and antibodies against several endogenous opioid peptides were used to determine the mechanisms of action of these peptides. Intrathecal pretreatment with naloxone and ß-funaltrexamine (ß-FNA) effectively inhibited analog-induced analgesia, demonstrating that activity of the analogs is regulated primarily through MOP. Antinociception induced by analog 2 through 4 was not reversed by δ opioid receptor (DOP) or κ opioid receptor (KOP) antagonist; antibodies against dynorphin-A (1-17), dynorphin-B (1-13), and Leu5/Met5-enkephalin had no impact on the antinociceptive effects of these analogs. In contrast, antinociceptive effects induced by a spinal injection of the fluorine substituted analog 1 were significantly reversed by KOP antagonism. Furthermore, intrathecal pretreatment with antibodies against dynorphin-B (1-13) attenuated the antinociceptive effect of analog 1. These results indicate that the antinociceptive activity exerted by intrathecally-administered analog 1 is mediated, in part, through KOP with increased release of dynorphin-B (1-13). The chemical modifications used in the present study may serve as a useful tool to gain insight into the mechanisms of endomorphins activity.


Asunto(s)
Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Oligopéptidos/química , Oligopéptidos/farmacología , Péptidos Opioides/química , Péptidos Opioides/farmacología , Analgesia , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/antagonistas & inhibidores , Análisis de Varianza , Animales , Anticuerpos/inmunología , Dinorfinas/administración & dosificación , Dinorfinas/antagonistas & inhibidores , Dinorfinas/química , Dinorfinas/farmacología , Encefalina Leucina/administración & dosificación , Encefalina Leucina/antagonistas & inhibidores , Encefalina Leucina/química , Encefalina Leucina/farmacología , Encefalina Metionina/administración & dosificación , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/química , Encefalina Metionina/farmacología , Flúor/química , Inyecciones Espinales , Masculino , Ratones , Naloxona/administración & dosificación , Naloxona/farmacología , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Oligopéptidos/administración & dosificación , Oligopéptidos/antagonistas & inhibidores , Péptidos Opioides/administración & dosificación , Péptidos Opioides/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Receptores sigma/antagonistas & inhibidores
4.
J Natl Cancer Inst ; 75(2): 341-4, 1985 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3860686

RESUMEN

The antitumor effect of methionine-enkephalin [( Met]enkephalin) was demonstrated in C57BL/6J mice inoculated with B16-BL6 melanoma cells. Local subcutaneous tumor growth was inhibited with a 50-micrograms dose daily for 7 or 14 days. The antitumor effect of [Met]enkephalin was inhibited by the administration of the opioid receptor antagonist naloxone. Naloxone alone had no significant effect on tumor growth.


Asunto(s)
Encefalina Metionina/farmacología , Melanoma/patología , Animales , Encefalina Metionina/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Naloxona/farmacología , Trasplante de Neoplasias
5.
Cancer Res ; 51(3): 785-8, 1991 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-1988118

RESUMEN

The effect of the opioid receptor agonist methionine enkephalin (Met-enkephalin) and the opioid receptor antagonist naloxone on colonic carcinogenesis induced by azoxymethane was investigated in Wistar rats. Rats received ten weekly injections of 7.4 mg/kg of body weight of azoxymethane and injections of Met-enkephalin (50 micrograms/kg of body weight), naloxone (2 mg/kg of body weight), or Met-enkephalin (50 micrograms/kg of body weight) plus naloxone (2 mg/kg of body weight) once every 2 days. In wk 40, the group treated with Met-enkephalin had a significantly increased incidence of colonic tumors. A combination of Met-enkephalin and naloxone attenuated the enhancing effect by Met-enkephalin on the development of colonic tumors. Administration of naloxone alone had no influence on colonic tumorigenesis. During and after administration of the carcinogen, the bromodeoxyuridine-labeling indices of the colon mucosa and/or cancers were significantly increased in rats treated with Met-enkephalin. However, a combination of Met-enkephalin and naloxone significantly decreased the labeling indices of the colon mucosa and/or cancers. These findings indicate that Met-enkephalin enhanced colon carcinogenesis and that naloxone attenuated this enhancement. Because naloxone is an opioid receptor antagonist, these findings also indicate that the enhancing effect of Met-enkephalin on colon carcinogenesis may be mediated through opioid receptors.


Asunto(s)
Adenocarcinoma/inducido químicamente , Azoximetano/toxicidad , Neoplasias del Colon/inducido químicamente , Encefalina Metionina/farmacología , Adenocarcinoma/patología , Animales , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/patología , Esquema de Medicación , Sinergismo Farmacológico , Encefalina Metionina/antagonistas & inhibidores , Masculino , Naloxona/farmacología , Ratas , Ratas Endogámicas
6.
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
7.
Cell Prolif ; 36(3): 151-63, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12814431

RESUMEN

Opioid peptides are negative regulators of cell proliferation in several organs including the uterus. In the present study, the ontogeny of the direct inhibitory action of opioid peptides on the proliferation of cultured rat uterine cells was investigated. Uteri of 7, 14, 21, 28, 35 and 60-day-old rats were removed in a sterile way. Tissue blocks were dispersed by limited digestions with trypsin and collagenase. Cells were cultured in enriched Dulbecco's modified Eagle's medium (DMEM). Treatments were present during the entire culture period. Cell densities of the monolayers were determined by counting the cells following trypsinization and trypan blue exclusion. Rat uterine mixed cell cultures grew to confluence within 10 days. The average population doubling time gradually increased with the age of animals. Epidermal growth factor (EGF) increased cell densities of cultures from all age groups. The oestradiol (E2)-responsiveness appeared at 21 days of age. The effect of [D-Met2-Pro5]-enkephalinamide (ENK) was biphasic. ENK and [Met5]-enkephalin (OGF) decreased cell densities of both unstimulated and EGF-stimulated cultures from 7-day-old rats to the same extent. ENK failed to act in 14-day-old animals. From 21 days of age on, the E2- or EGF-stimulated proliferation was inhibited only by ENK and DAMGO, while 30 nm DPDPE, Dynorhin-A, OGF, [Leu5]-enkephalin, beta-endorphin, and morphiceptin were ineffective. The half-inhibitory concentration of ENK was 0.3 nm. The effects of ENK were prevented by concomitant treatment with naloxone. Our novel data demonstrate two different phases of the inhibitory action of opioid peptides on rat uterine cell proliferation during ontogeny with an insensitive interval in between.


Asunto(s)
Encefalina Metionina/análogos & derivados , Encefalinas/metabolismo , Péptidos Opioides/farmacología , Útero/efectos de los fármacos , Útero/crecimiento & desarrollo , Envejecimiento/metabolismo , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Dinorfinas/farmacología , Endorfinas/farmacología , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/farmacología , Encefalinas/farmacología , Factor de Crecimiento Epidérmico/farmacología , Estradiol/metabolismo , Femenino , Concentración 50 Inhibidora , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Ovariectomía , Ratas , Ratas Endogámicas , Ratas Wistar , Útero/metabolismo
8.
Neuropharmacology ; 33(11): 1333-8, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7532816

RESUMEN

To investigate the possible neuromodulatory role of nitric oxide (NO) in the gastrointestinal tract, an examination was made of the effects of NG-nitro-L-arginine (L-NOARG), an inhibitor of NO synthase, on the intestinal response to [Met5]-enkephalin (ENK) by recording the mechanical activity of the isolated duodenum from rats. [Met5]-enkephalin elicited a biphasic response of the duodenum, i.e. transient relaxation followed by contraction. The relaxation induced by ENK was blocked by naloxone, an opioid receptor antagonist, but not by tetrodotoxin (TTX). The contractile response of the duodenum to ENK was blocked by TTX but not by naloxone. The contractile response was not affected by hyoscine, a muscarinic antagonist, or guanethidine, an adrenergic neuron blocking agent, indicating mediation by non-adrenergic, non-cholinergic (NANC) nerves. The contractile but not the relaxant response to ENK was blocked by L- but not D-NOARG. The contractile response was also inhibited by methylene blue, an inhibitor of both NO synthase and guanylate cyclase, and by indomethacin, a cyclooxygenase inhibitor. Thus, endogenous NO and prostaglandins are involved in the contractile response to ENK. Endogenous NO may modulate the release of excitatory NANC transmitters via a prejunctional mechanism.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Encefalina Metionina/farmacología , Músculo Liso/fisiología , Óxido Nítrico/fisiología , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Arginina/farmacología , Duodeno/efectos de los fármacos , Duodeno/inervación , Duodeno/fisiología , Encefalina Metionina/antagonistas & inhibidores , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/inervación , Naloxona/farmacología , Óxido Nítrico Sintasa , Nitroarginina , Ratas , Ratas Wistar , Tetrodotoxina/farmacología
9.
J Neuroimmunol ; 31(2): 97-103, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1991823

RESUMEN

Opioid peptides and their analogs have been shown to stimulate adherence, conformational changes and locomotory activity in human as well as invertebrate granulocytes. The present study demonstrates that [Met]-enkephalin-Arg6-Phe7, an opioid substance thus far not included in these immunological tests, exhibits stimulatory effects comparable to those of [Met]-enkephalin in this regard. Furthermore, since neutral endopeptidase 24.11 (enkephalinase; CD10/NEP) exists in invertebrate immunocyte membranes, we demonstrate that its specific inhibitor, phosphoramidon, potentiates the effects of the heptapeptide in inducing conformational change in both human and invertebrate granulocytes. Additionally, the major metabolic products of NEP activity, Phe-Met-Arg-Phe and Tyr-Gly-Gly, appear to be potent antagonists of this enzyme activity, especially the tetrapeptide. The effects of heptapeptide stimulation showed a major difference between vertebrate and invertebrate immunocytes with respect to their time course, namely, the speed of their onset. [Met]-enkephalin-Arg6-Phe7 markedly stimulated the locomotory activity of these cells which becomes most noticeable within 15-45 min for Mytilus cells and in a 5-15 min period for human cells. It also enhanced the mobility and velocity of the responsive human (5 microns/min) and invertebrate cells (2.1 microns/min).


Asunto(s)
Encefalina Metionina/análogos & derivados , Granulocitos/inmunología , Sistema Inmunológico/fisiología , Animales , Bivalvos/inmunología , Movimiento Celular/efectos de los fármacos , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/inmunología , Granulocitos/citología , Humanos , Naloxona/farmacología , Oligopéptidos/farmacología
10.
J Neuroimmunol ; 15(3): 219-28, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-2954999

RESUMEN

Early events in an acute inflammatory response include the adherence of neutrophils (PMN) to capillary endothelial cells and the migration of these cells into tissues. This study was designed to determine if the opioid peptides beta-endorphin or met-enkephalin would induce a concentration-dependent increase in PMN adherence to serum-coated glass. Results show that PMN adherence is increased with both beta-endorphin and met-enkephalin and this increase may be partially blocked by the opiate antagonist naloxone. Binding of opioid peptides to the formyl peptide receptor was ruled out as a mechanism of increased adherence by showing that the opioids failed to block the binding of f-met-leu-phe-lys to PMN. These studies suggest that alterations in circulating opioid concentrations may modulate the adherence of PMN and thereby influence acute inflammatory reactions.


Asunto(s)
Endorfinas/farmacología , Encefalina Metionina/farmacología , Neutrófilos/efectos de los fármacos , Adulto , Adhesión Celular/efectos de los fármacos , Endorfinas/antagonistas & inhibidores , Encefalina Metionina/antagonistas & inhibidores , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , N-Formilmetionina Leucil-Fenilalanina/análogos & derivados , N-Formilmetionina Leucil-Fenilalanina/sangre , Naloxona/farmacología , Neutrófilos/inmunología , Neutrófilos/metabolismo , betaendorfina
11.
J Neuroimmunol ; 141(1-2): 30-9, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12965251

RESUMEN

Neuroimmune interactions control pain through activation of opioid receptors on sensory nerves by immune-derived opioid peptides. Here we evaluate mechanisms of intrinsic pain inhibition at different stages of Freund's adjuvant-induced inflammation of the rat paw. We use immunohistochemistry and paw pressure testing. Our data show that in early (6 h) inflammation leukocyte-derived beta-endorphin, met-enkephalin and dynorphin A activate peripheral mu-, delta- and kappa-receptors to inhibit nociception. In addition, central opioid mechanisms seem to contribute significantly to this effect. At later stages (4 days), antinociception is exclusively produced by leukocyte-derived beta-endorphin acting at peripheral mu and delta receptors. Corticotropin-releasing hormone (CRH) is an endogenous trigger of these effects at both stages. These findings indicate that peripheral opioid mechanisms of pain inhibition gain functional relevance with the chronicity of inflammation.


Asunto(s)
Dolor/patología , Dolor/prevención & control , Animales , Hormona Liberadora de Corticotropina/administración & dosificación , Dinorfinas/antagonistas & inhibidores , Dinorfinas/biosíntesis , Dinorfinas/fisiología , Edema/inmunología , Edema/metabolismo , Edema/fisiopatología , Endorfinas/antagonistas & inhibidores , Endorfinas/biosíntesis , Endorfinas/fisiología , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/biosíntesis , Encefalina Metionina/fisiología , Adyuvante de Freund/administración & dosificación , Miembro Posterior , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/fisiopatología , Inyecciones Subcutáneas , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/fisiología , Masculino , Naloxona/administración & dosificación , Dolor/inmunología , Umbral del Dolor , Ratas , Ratas Wistar , Estrés Fisiológico/inmunología , Estrés Fisiológico/metabolismo , Estrés Fisiológico/fisiopatología , Factores de Tiempo
12.
Neuroscience ; 11(4): 919-30, 1984 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6738859

RESUMEN

Neurotensin is an endogenous neuropeptide with neuronal perikarya or fibers distributed in the vicinity of the mesolimbic dopamine system. This observation, plus behavioral data showing that neurotensin injection into the nucleus accumbens blocks some behavioral effects of amphetamine, indicates that neurotensin may modulate the mesolimbic dopamine system. In this study it was shown that neurotensin given into the nucleus accumbens produces a dose-dependent blockade of locomotion and rearing initiated by dopamine injection into the nucleus accumbens. This effect is not mimicked by inactive neurotensin analogue nor some other endogenous neuropeptides. Since dopamine acts on postsynaptic dopamine receptors in the nucleus accumbens, neurotensin is acting, not on dopamine terminals, but on neurons or neuronal systems which are modulated by the mesolimbic dopamine system. This conclusion is supported by the facts that intra-accumbens injection of neurotensin does not alter accumbens levels of dopamine or its metabolites, nor does it affect the increase in dopamine metabolites produced by injection of neurotensin into the ventral tegmental area. Further, neurotensin was also found to block the dopamine-independent increase in locomotion and rearing produced by the injection of D-Ala2-Met- 5enkephalinamide into the nucleus accumbens. These data indicate that neurotensin acts on neurons in the nucleus accumbens to counteract the motor stimulant effects of dopamine or enkephalin. Therefore, in the nucleus accumbens, neurotensin is not acting to modulate the mesolimbic dopamine system, but rather appears to antagonize behavioral hyperactivity, regardless of the neurochemical initiation.


Asunto(s)
Antagonistas de Dopamina , Actividad Motora/efectos de los fármacos , Neurotensina/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleos Septales/efectos de los fármacos , Animales , Mapeo Encefálico , Encefalina Metionina/análogos & derivados , Encefalina Metionina/antagonistas & inhibidores , Inyecciones Intraventriculares , Masculino , Microinyecciones , Ratas , Ratas Endogámicas , Tegmento Mesencefálico/efectos de los fármacos
13.
Neuroscience ; 38(2): 373-80, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2263320

RESUMEN

Intracellular and extracellular recording techniques and in vitro preparation were used to examine the effect of [Met]enkephalin on the rat periaqueductal neurons. In the 20 cells that were recorded intracellularly, [Met]enkephalin caused an increase in the resting membrane conductance, hyperpolarization of the cell membrane, an increase in the firing threshold and a decrease in the spontaneous firing rate. This effect of [Met]enkephalin could be blocked by naloxone. The effect of [Met]enkephalin on 99 neurons was also examined using extracellular recording. In 59% of cells, pressure application of [Met]enkephalin caused a dose-dependent inhibition that could be blocked by naloxone; 15% of the cells were excited and the remaining neurons (26%) did not respond. Nineteen per cent of responsive cells were located in the dorsolateral subdivision; 41% in the ventrolateral and 13% in the dorsal regions. In 10 cells, perfusion with physiological saline solution/Mg did not alter the inhibitory effect of [Met]enkephalin. However, perfusion with physiological saline solution/Mg abolished the excitatory response to [Met]enkephalin in four cells. It is concluded that: (1) the major effect of [Met]enkephalin on periaqueductal gray cells is inhibition that occurs through a direct postsynaptic process. This inhibition is probably due to an increase in permeability to potassium; (2) a small population of periaqueductal gray cells are excited by [Met]enkephalin, probably through a presynaptic process.


Asunto(s)
Encefalina Metionina/farmacología , Neuronas/efectos de los fármacos , Sustancia Gris Periacueductal/efectos de los fármacos , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Relación Dosis-Respuesta a Droga , Electrofisiología , Encefalina Metionina/antagonistas & inhibidores , Técnicas In Vitro , Magnesio/farmacología , Masculino , Naloxona/farmacología , Sustancia Gris Periacueductal/citología , Ratas , Ratas Endogámicas , Cloruro de Sodio/farmacología
14.
Br J Pharmacol ; 83(3): 783-90, 1984 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6509263

RESUMEN

In intact rabbits anaesthetized with pentobarbitone, methionine enkephalin (Met enkephalin, 1-1,000 micrograms kg-1 i.v.) produced a dose-dependent bradycardia and hypotension. The bradycardia and hypotension were antagonized by naloxone hydrochloride (1 mg kg-1), but not by naloxone methobromide (1.3 mg kg-1). Phentolamine (1 and 4 mg kg-1 i.v.) blocked both the hypotension and bradycardia produced by Met enkephalin. The inhibitory effect of phentolamine was not due to a simple hypotensive action of this drug per se because a similar degree of hypotension induced by nitroprusside (15 micrograms kg-1, i.v.) caused a further reduction of pressure when Met enkephalin was added. Atropine (2 mg kg-1) reduced the bradycardia but not the hypotensive response to Met enkephalin. Met enkephalin did not antagonize the vasopressor effect of exogenous noradrenaline (2-8 micrograms kg-1, i.v.). Met enkephalin had no significant effects in superfused thoracic aortic strips and in isolated perfused hearts of rabbits. It is concluded that the cardiovascular effects of Met enkephalin are more probably due to an action on the central nervous system, although a peripheral site of action cannot be completely excluded.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Encefalina Metionina/antagonistas & inhibidores , Fentolamina/farmacología , Animales , Atropina/farmacología , Encefalina Metionina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Masculino , Naloxona/farmacología , Nitroprusiato/farmacología , Norepinefrina/farmacología , Perfusión , Conejos
15.
Int J Oncol ; 17(5): 1053-61, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11029512

RESUMEN

The native opioid growth factor (OGF), [Met5]-enkephalin, is a tonic inhibitory peptide that modulates cell proliferation and migration, as well as tissue organization, during development, cancer, homeostatic cellular renewal, wound healing, and angiogenesis. OGF action is mediated by the OGF receptor (OGFr). To investigate the target of OGF as to cell proliferation, the effects of excess OGF, and a deprivation of OGF-OGFr interaction by an opioid antagonist, naltrexone (NTX), were examined in 3 human cancer cell lines: pancreatic (BxPC-3), colon (HT-29), and head and neck (CAL-27). OGF exposure decreased growth, DNA synthesis, and mitosis, and increased the doubling time from control levels. FACS analysis revealed a marked increase in cells in the G0/G1 phase and compensatory reduction in cells in S and G2/M phases. Consistent with this observation, the percentage of labeled mitosis (PLM) analysis showed a notable increase in the time of the G0/G1 phase. Receptor blockade with NTX increased the rate of growth, length of DNA synthesis and mitotic phases, and decreased doubling time from control values. FACS analysis indicated an increase in the proportion of cells in S and G2/M phases, and a decrease in the number of cells in the G0/G1 phase. PLM evaluation demonstrated a shortening of the length of the S and G2 phases in the 3 cell lines, and decreases in the M and G0/G1 phases in some cancers. These results indicate that OGF action is directed at the G0/G1 phase, but interruption of OGF-OGFr interfacing has widespread repercussions on the cell cycle. The data on blockade of OGF-OGFr during log phase growth suggest a requisite escorting of the growth peptide and its receptor through the cell cycle.


Asunto(s)
Ciclo Celular/fisiología , Encefalina Metionina/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/patología , Receptores Opioides/fisiología , Adenocarcinoma/patología , Carcinoma de Células Escamosas/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Neoplasias del Colon/patología , Replicación del ADN/efectos de los fármacos , Encefalina Metionina/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/patología , Humanos , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neoplasias/metabolismo , Neoplasias Pancreáticas/patología , Receptores Opioides/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Ensayo de Tumor de Célula Madre
16.
Brain Res Mol Brain Res ; 33(1): 111-20, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8774952

RESUMEN

[Met5]-enkephalin, encoded by the preproenkephalin (PPE) gene, serves as a growth factor during brain development in addition to its role as a neurotransmitter. This study examined the relationship of gene and peptide expression in the developing (postnatal day 6) rat brain by disrupting peptide-receptor interaction with either a brief (4-6 h) or continuous opioid receptor blockade using a single injection of 1 or 50 mg/kg naltrexone (NTX), respectively; such perturbations result in growth inhibition or acceleration, respectively. In the caudate putamen, an area that has completed neurogenesis by postnatal day 6 and has an abundance of PPE mRNA and enkephalins in adulthood, NTX did not influence PPE mRNA in either NTX group, or the enkephalin levels in the 1 mg/kg NTX group. [Met5]-enkephalin values in the neostriatum, however, were 67-183% greater than controls in rats given 50 mg/kg NTX, beginning 5 min after drug injection. In the cerebellum, PPE mRNA expression was depressed from 5 min to 4 h in the 1 mg/kg NTX group, and was normal thereafter; mRNA levels in the 50 mg/kg NTX group were markedly subnormal for 24 h. Enkephalin levels were significantly depressed within 5 min of drug injection and remained so for 4 h in the 1 mg/kg NTX group, but were elevated to approximately 135% of control values at 8, 16, and 24 h. Enkephalin levels were not changed in the cerebellum of the 50 mg/kg NTX group, or in the plasma of either NTX group. These data suggest that a single exposure to NTX can affect transcriptional and translational mechanisms related to PPE mRNA and opioid peptide expression in a rapid and sustained manner, and that this treatment elicits a specific pattern of alterations dependent upon the brain region sampled, drug dosage, and/or the duration of opioid receptor blockade. Additionally, our results indicate that the decreased DNA synthesis in external germinal cells occurring after opioid receptor blockade as recorded earlier may be related to an increase in the potent opioid growth factor, [Met5]-enkephalin.


Asunto(s)
Encefalina Metionina/genética , Encefalinas/genética , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Precursores de Proteínas/genética , ARN Mensajero/biosíntesis , Animales , Núcleo Caudado/metabolismo , Cerebelo/metabolismo , Encefalina Metionina/antagonistas & inhibidores , Encefalinas/antagonistas & inhibidores , Femenino , Masculino , Precursores de Proteínas/antagonistas & inhibidores , Putamen/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
Peptides ; 14(3): 445-8, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8332544

RESUMEN

In the conscious dog, intravenous administration of methionine-enkephalin produces simultaneous increases in both heart rate (HR) and mean arterial blood pressure (MAP). This report describes both depressor and cardioaccelerator responses to methionine-enkephalin (10 micrograms/kg IV) in conscious dogs following acute hypotension induced by either bolus injection of isoproterenol (0.1-5.0 micrograms/kg IV) or infusion of sodium nitroprusside (SNP, 3-8 micrograms/kg/min). Cardiovascular responses to methionine-enkephalin were blocked by naloxone. Pretreatment of the dogs with the beta-adrenergic receptor antagonist propranolol failed to prevent the hypotensive response to methionine-enkephalin following SNP infusion. The results indicate that the hemodynamic responses to methionine-enkephalin can be altered by acute manipulation of blood pressure. These results may have implications relative to the role of endogenous opiates in regulation of blood pressure, especially in acute hypotensive states.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Encefalina Metionina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Animales , Perros , Encefalina Metionina/antagonistas & inhibidores , Femenino , Isoproterenol/farmacología , Masculino , Naloxona/farmacología , Nitroprusiato/farmacología , Propranolol/farmacología
18.
Regul Pept ; 47(2): 141-50, 1993 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-8234900

RESUMEN

We used the met-enkephalin analog (D-Met2,Pro5)-enkephalinamide (DMPEA) to investigate enkephalinergic control of alpha-melanocyte-stimulating hormone (alpha-MSH) secretion. Systemic (s.c.) administration of DMPEA elevated plasma titers of alpha-MSH in a dose- and time-related manner. Pretreatment with the opiate antagonist naltrexone had no effect on basal plasma levels of alpha-MSH but blocked DMPEA-induced alpha-MSH release. Treatment with a dose of naltrexone sufficient to block DMPEA-induced secretion of alpha-MSH had no effect on stress-induced secretion of alpha-MSH. Although pretreatment with the dopamine receptor agonist apomorphine prevented DMPEA-induced alpha-MSH secretion, DMPEA had no effect on the synthetic activity of tuberohypophysial dopamine neurons as gauged by measuring the accumulation of 3,4-dihydroxyphenylalanine in the neurointermediate lobe (NIL) following administration of NSD-1015. In vitro treatment of isolated NILs with DMPEA resulted in a significant increase in alpha-MSH release. Naltrexone completely blocked the stimulatory effects of DMPEA on alpha-MSH release in vitro. Our results indicate that DMPEA stimulates alpha-MSH secretion by acting directly through opiate receptors at the level of the NIL.


Asunto(s)
Encefalina Metionina/análogos & derivados , alfa-MSH/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/farmacología , Masculino , Naltrexona/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , alfa-MSH/sangre
19.
Regul Pept ; 12(4): 333-43, 1985 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-3867098

RESUMEN

The effects of Met-enkephalinamide (MET-ENKamide) on brain temperature (Tb) and metabolic rate (MR) were assessed following direct administration into the preoptic/anterior hypothalamus (PO/AH) of freely moving rats. Bilateral microinjections of saline or MET-ENKamide (1-25 micrograms/microliter) were delivered through cannula guide tubes previously implanted in nine animals. Thiorphan, an enkephalinase inhibitor, was microinjected into the PO/AH of two of the animals. All injections were made remotely at an ambient temperature of 22 +/- 1 degree C in a volume of 1 microliter. Measurements of Tb (via a brain-dwelling thermistor) and MR were recorded continuously. The ability of naloxone to antagonize the effects of MET-ENKamide was investigated by fashioning a double-barreled injection cannula to fit within each guide tube; 1 microliter of saline or naloxone (1-10 micrograms) was delivered bilaterally into the PO/AH followed by 1 microliter of MET-ENKamide (25 micrograms) 5-10 min later. PO/AH administration of MET-ENKamide (1-25 micrograms) produced dose-dependent increases in Tb preceded by dose-dependent increases in MR, with a characteristic time course of approximately 30 min. Naloxone antagonized the rise in Tb and MR, either partially or completely, depending on dose. When administered alone, naloxone had no effect on Tb or MR. Microinjection of thiorphan (10 micrograms) into the PO/AH evoked increases in Tb and MR that were similar to those responses induced by MET-ENKamide. These results support a role for endogenous Met-enkephalin in the regulation of Tb in the rat.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Encefalina Metionina/análogos & derivados , Área Preóptica/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Encefalina Metionina/administración & dosificación , Encefalina Metionina/antagonistas & inhibidores , Masculino , Tasa de Depuración Metabólica/efectos de los fármacos , Microinyecciones , Naloxona/administración & dosificación , Ratas , Ratas Endogámicas , Tiorfan , Tiopronina/administración & dosificación , Tiopronina/análogos & derivados
20.
Neuropeptides ; 16(3): 115-20, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2082199

RESUMEN

Opioid inhibition of phagocytosis in the protozoan ciliate Tetrahymena is antagonized by naloxone and this antagonism can be surmounted by increasing agonist concentration, which suggests a receptor-mediated mechanism. Desensitization of the opioid effect is time dependent in addition to concentration dependent. Chronic exposure to opioids results in the development of tolerance to the inhibitory effect of the agonists, and withdrawal of the latter results in a decrease in phagocytic capacity, which suggests that a state akin to dependence has been developed in these cells. Naloxone appears to behave as a partial agonist in tolerant cells, and there seems to exist cross-tolerance to mu and delta agonists.


Asunto(s)
Encefalina Metionina/análogos & derivados , Morfina/farmacología , Fagocitosis/efectos de los fármacos , Tetrahymena/efectos de los fármacos , Animales , Tolerancia a Medicamentos/fisiología , Encefalina Metionina/antagonistas & inhibidores , Encefalina Metionina/farmacología , Naloxona/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda