Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Physiol ; 588(Pt 21): 4205-15, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20837644

RESUMEN

Neuropathic pain is a common diabetic complication affecting 8-16% of diabetic patients. It is characterized by aberrant symptoms of spontaneous and stimulus-evoked pain including hyperalgesia and allodynia. Magnesium (Mg) deficiency has been proposed as a factor in the pathogenesis of diabetes-related complications, including neuropathy. In the central nervous system, Mg is also a voltage-dependent blocker of the N-methyl-d-aspartate receptor channels involved in abnormal processing of sensory information. We hypothesized that Mg deficiency might contribute to the development of neuropathic pain and the worsening of clinical and biological signs of diabetes and consequently, that Mg administration could prevent or improve its complications. We examined the effects of oral Mg supplementation (296 mg l(-1) in drinking water for 3 weeks) on the development of neuropathic pain and on biological and clinical parameters of diabetes in streptozocin (STZ)-induced diabetic rats. STZ administration induced typical symptoms of type 1 diabetes. The diabetic rats also displayed mechanical hypersensitivity and tactile and thermal allodynia. The level of phosphorylated NMDA receptor NR1 subunit (pNR1) was higher in the spinal dorsal horn of diabetic hyperalgesic/allodynic rats. Magnesium supplementation failed to reduce hyperglycaemia, polyphagia and hypermagnesiuria, or to restore intracellular Mg levels and body growth, but increased insulinaemia and reduced polydipsia. Moreover, it abolished thermal and tactile allodynia, delayed the development of mechanical hypersensitivity, and prevented the increase in spinal cord dorsal horn pNR1. Thus, neuropathic pain symptoms can be attenuated by targeting the Mg-mediated blockade of NMDA receptors, offering new therapeutic opportunities for the management of chronic neuropathic pain.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/prevención & control , Hiperalgesia/prevención & control , Magnesio/uso terapéutico , Neuralgia/prevención & control , Receptores de N-Metil-D-Aspartato/metabolismo , Administración Oral , Animales , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Calor , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Magnesio/administración & dosificación , Masculino , Neuralgia/metabolismo , Neuralgia/fisiopatología , Fosforilación , Ratas , Ratas Sprague-Dawley , Estreptozocina , Estrés Mecánico
2.
FEBS Lett ; 496(1): 25-30, 2001 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-11343700

RESUMEN

We present evidence that gastrin, binding to a G protein-coupled receptor, activates the p38-mitogen-activated protein kinase (MAPK) pathway. Blockage of protein kinase C (PKC) by GF109203X, depletion of intracellular calcium by thapsigargin or inhibition of Src family kinases by PP2 prevented p38-MAPK activation and the Src kinase activity stimulated by gastrin. Inhibition of the PI 3-kinase by wortmannin or LY294002 did not affect these responses. In addition, the p38-MAPK inhibitor, SB203580, repressed gastrin-induced [(3)H]thymidine incorporation, indicating a major role of p38-MAPK in the growth-promoting effect of gastrin. Our results demonstrate that gastrin-induced DNA synthesis requires p38-MAPK activation through mechanisms that involve calcium mobilization, PKC and Src family kinases.


Asunto(s)
ADN/biosíntesis , Gastrinas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Familia-src Quinasas/metabolismo , Androstadienos/farmacología , Animales , Células CHO , Calcio/metabolismo , Cromonas/farmacología , Cricetinae , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP/metabolismo , Gastrinas/farmacología , Humanos , Líquido Intracelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Receptor de Colecistoquinina B , Receptores de Colecistoquinina/genética , Receptores de Colecistoquinina/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina , Transfección , Wortmanina , Proteínas Quinasas p38 Activadas por Mitógenos , Familia-src Quinasas/antagonistas & inhibidores
3.
J Biol Chem ; 274(29): 20657-63, 1999 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-10400698

RESUMEN

We have analyzed in Chinese hamster ovary cells the upstream mediators by which the G protein-coupled receptor, gastrin/CCKB, activates the extracellular-regulated kinases (ERKs) and p85/p110-phosphatidylinositol 3-kinase (PI 3-kinase) pathways. Overexpression of an inhibitory mutant of Shc completely blocked gastrin-stimulated Shc.Grb2 complex formation but partially inhibited ERK-1 activation by this peptide. Expression of Csk, which inactivates Src-family kinases, totally inhibited gastrin-induced Src-like activity detected in anti-Src and anti-Shc precipitates but diminished by 50% Shc phosphorylation and ERK-1 activation. We observed a rapid tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and an increase in Src-like kinase activity in anti-IRS-1 immunoprecipitates from gastrin-stimulated cells, suggesting that IRS-1 may be a direct substrate of Src. This hypothesis was supported by the inhibition of gastrin-induced Src. IRS-1 complex formation and IRS-1 phosphorylation in Csk-transfected cells. In addition, the increase in PI 3-kinase activity measured in anti-p85 or anti-IRS-1 precipitates following gastrin stimulation was abolished by Csk. Our results demonstrate the existence of two mechanisms in gastrin-mediated ERKs activation. One requires Shc phosphorylation by Src-family kinases, and the other one is independent of these two proteins. They also indicate that tyrosine phosphorylation of IRS-1 by Src-family kinases could lead to the recruitment and the activation of the p85/p110-PI 3-kinase in response to gastrin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Colecistoquinina/metabolismo , Familia-src Quinasas/metabolismo , Animales , Células CHO , Cricetinae , Replicación del ADN , Activación Enzimática , Proteína Adaptadora GRB2 , Gastrinas/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina , Proteína Quinasa 3 Activada por Mitógenos , Fosfoproteínas/metabolismo , Fosforilación , Proteínas/metabolismo , Receptor de Colecistoquinina B , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Tirosina/metabolismo
4.
FEBS Lett ; 445(2-3): 251-5, 1999 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-10094466

RESUMEN

The molecular events whereby gastrin occupancy of G/CCK(B) receptors leads to phosphatidylinositol (PI) 3-kinase activation have been examined. We report here that this peptide promotes the association between two non-receptor tyrosine kinases, p60Src and p125FAK, and elicits a parallel increase in tyrosine phosphorylation and activity of both kinases. Gastrin-induced PI 3-kinase activity was coprecipitated with p60Src and p125FAK and was inhibited by herbimycin A, the selective Src inhibitor PP-2 or cytochalasin D, which disrupts the actin cytoskeleton and prevents p125FAK activity. These results indicate, for the first time, that a p60Src/p125FAK complex acts upstream of the gastrin-stimulated PI 3-kinase pathway.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Gastrinas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Animales , Activación Enzimática , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Gastrinas/farmacología , Fosforilación , Pruebas de Precipitina , Ratas , Células Tumorales Cultivadas , Tirosina/metabolismo
5.
Biochem Biophys Res Commun ; 238(1): 202-6, 1997 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-9299479

RESUMEN

The gastrin/CCKB (G/CCKB) G protein-coupled receptor has been shown to mediate the proliferative effects of gastrin on normal and neoplastic gastro-intestinal tissues. In the present study, we examined the signal transduction mechanisms coupled to this receptor. We report here that phosphorylation and activity of the p70S6K, whose major substrate is the ribosomal S6 protein, are enhanced in response to gastrin. These effects were completely reversed by a commonly used PI-3-kinase inhibitor, wortmannin, suggesting that p70S6K may be a downstream target of PI-3-kinase in a signaling cascade induced by gastrin. In addition, blocking PI-3-kinase activity by wortmannin partially decreased gastrin-induced MAPK activation (42% +/- 3) as well as the tyrosine phosphorylation of She (50% +/- 6), an upstream regulator of the Ras-dependent MAPK pathway. These results indicate that at least two signaling pathways lead to MAPK activation by gastrin, only one of which is sensitive to PI-3-kinase inhibitors.


Asunto(s)
Androstadienos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de Unión al GTP/metabolismo , Gastrinas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Colecistoquinina/fisiología , Proteínas Ribosómicas/metabolismo , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Gastrinas/metabolismo , Neoplasias Pancreáticas , Fosfatidilinositol 3-Quinasas , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Polienos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Receptor de Colecistoquinina B , Proteínas Quinasas S6 Ribosómicas , Sirolimus , Células Tumorales Cultivadas , Wortmanina , Dominios Homologos src/efectos de los fármacos
6.
Biochem J ; 325 ( Pt 2): 383-9, 1997 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9230117

RESUMEN

The proliferative effects of gastrin on normal and neoplastic gastro-intestinal tissues have been shown to be mediated by the gastrin/CCKB (G/CCKB) G-protein-coupled receptors. We have recently reported that gastrin stimulates the tyrosine phosphorylation of Shc proteins and their subsequent association with the Grb2/Sos complex, leading to mitogen-activated protein kinase (MAPK) activation, a pathway known to play an important role in cell proliferation. We undertook the present study to characterize the signalling pathways used by this receptor to mediate the activation of the Shc/Grb2 complex. Since G/CCKB receptor occupancy leads to the activation of the phospholipase C (PLC)/protein kinase C (PKC) pathway, we examined whether PKC stimulation and Ca2+ mobilization contribute to the phosphorylation of Shc proteins and their association with Grb2 in response to gastrin. Our results indicate that Shc proteins are tyrosine phosphorylated and associate with Grb2 in response to phorbol esters, suggesting that activation of PKC is a potential signalling pathway leading to activation of the Shc/Grb2 complex. Inhibition of PKC by GF109203X completely blocked the effect of PMA on Shc tyrosine phosphorylation and its subsequent association with Grb2, but had a partial inhibitory effect on the response to gastrin. Depletion of the intracellular Ca2+ pools by treatment with thapsigargin blocked the increase in intracellular free calcium concentration induced by gastrin and diminished the ability of the peptide to stimulate Shc phosphorylation and recruitment of Grb2. In addition, removal of extracellular Ca2+ partially inhibited the effect of gastrin on Shc phosphorylation as well as its association with Grb2, indicating that the effects of gastrin are also mediated by Ca2+-dependent mechanisms. Furthermore, we show that blockage of the two major early signals generated by activation of PLC, which induced the activation of the Shc/Grb2 complex, also blocked gastrin-induced MAPK activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Gastrinas/farmacología , Proteínas Quinasas Activadas por Mitógenos , Proteína Quinasa C/metabolismo , Proteínas/metabolismo , Animales , Western Blotting , Activación Enzimática , Proteína Adaptadora GRB2 , Humanos , Proteína Quinasa 3 Activada por Mitógenos , Neoplasias Experimentales , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal/fisiología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/farmacología , Células Tumorales Cultivadas , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...