Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 226(1-2): 59-66, 2004 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-15489006

RESUMEN

Both transcription factors albumin site d-binding protein (DBP) and thyrotroph embryonic factor (TEF) are elements of the "cell-clock". Their circadian accumulation in suprachiasmatic nucleus (SCN) and peripheral tissues such as liver, kidney and lung is thought to participate in controlling circadian regulation of downstream genes. TEF and DBP control elements have never been investigated in the insulin-secreting cells, but impairment of the circadian rhythm of the beta-cells might be involved in the development of diabetic state as type 2 diabetics have lost daily temporal variations of insulin secretion. We investigated the expression pattern of TEF and DBP in insulin-secreting cells. TEF and DBP transcripts are expressed at extremely high levels in human pancreatic islets compared to other tissues, suggesting a potentially important circadian regulation of these cells. Both TEF and DPB accumulate in a circadian way in insulin-secreting cells after a serum shock known to restore circadian rhythms in cultured cells. In addition, the expression of islet-specific genes involved in glucose sensing (glucose transporter 2 (Glut2), glucokinase), insulin production (insulin) and secretion (migration inhibitory factor (MIF), somatostatin and syntaxin 1A) were modulated in the same daily rhythm as well. The circadian deregulation of these genes could therefore participate in the diabetic state development.


Asunto(s)
Ritmo Circadiano , Proteínas de Unión al ADN/metabolismo , Insulina/biosíntesis , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factores de Transcripción/metabolismo , Antígenos de Superficie/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Regulación de la Expresión Génica , Glucoquinasa/metabolismo , Transportador de Glucosa de Tipo 2 , Humanos , Secreción de Insulina , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas , Somatostatina/metabolismo , Núcleo Supraquiasmático/metabolismo , Sintaxina 1
2.
Cytokine ; 24(1-2): 13-24, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14561487

RESUMEN

The stress-activated protein kinase c-Jun NH2-terminal kinase (JNK) is a central signal for interleukin-1beta (IL-1beta)-induced apoptosis in insulin-producing beta-cells. The cell-permeable peptide inhibitor of JNK (JNKI1), that introduces the JNK binding domain (JBD) of the scaffold protein islet-brain 1 (IB1) inside cells, effectively prevents beta-cell death caused by this cytokine. To define the molecular targets of JNK involved in cytokine-induced beta-cell apoptosis we investigated whether JNKI1 or stable expression of JBD affected the expression of selected pro- and anti-apoptotic genes induced in rat (RIN-5AH-T2B) and mouse (betaTC3) insulinoma cells exposed to IL-1beta. Inhibition of JNK significantly reduced phosphorylation of the specific JNK substrate c-Jun (p<0.05), IL-1beta-induced apoptosis (p<0.001), and IL-1beta-mediated c-fos gene expression. However, neither JNKI1 nor JBD did influence IL-1beta-induced NO synthesis or iNOS expression or the transcription of the genes encoding mitochondrial manganese superoxide dismutase (MnSOD), catalase (CAT), glutathione peroxidase (GPx), glutathione-S-transferase rho (GSTrho), heat shock protein (HSP) 70, IL-1beta-converting enzyme (ICE), caspase-3, apoptosis-inducing factor (AIF), Bcl-2 or Bcl-xL. We suggest that the anti-apoptotic effect of JNK inhibition by JBD is independent of the transcription of major pro- and anti-apoptotic genes, but may be exerted at the translational or posttranslational level.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis/fisiología , Islotes Pancreáticos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Sitios de Unión , Insulina/metabolismo , Interleucina-1/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Óxido Nítrico , Óxido Nítrico Sintasa/metabolismo , Estructura Terciaria de Proteína , Ratas
3.
Diabetes ; 50(1): 77-82, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11147798

RESUMEN

Stress conditions and proinflammatory cytokines activate the c-Jun NH2-terminal kinase (JNK), a member of the stress-activated group of mitogen-activated protein kinases (MAPKs). We recently demonstrated that inhibition of JNK signaling with the use of the islet-brain (IB) 1 and 2 proteins prevented interleukin (IL)-1beta-induced pancreatic beta-cell death. Bioactive cell-permeable peptide inhibitors of JNK were engineered by linking the minimal 20-amino acid inhibitory domains of the IB proteins to the 10-amino acid HIV-TAT sequence that rapidly translocates inside cells. Kinase assays indicate that the inhibitors block activation of the transcription factor c-Jun by JNK. Addition of the peptides to the insulin-secreting betaTC-3 cell line results in a marked inhibition of IL-1beta-induced c-jun and c-fos expression. The peptides protect betaTC-3 cells against apoptosis induced by IL-1beta. All-D retro-inverso peptides penetrate cells as efficiently as the L-enantiomers, decrease c-Jun activation by JNK, and remain highly stable inside cells. These latter peptides confer full protection against IL-1beta-induced apoptosis for up to 2 weeks of continual treatment with IL-1beta. These data establish these bioactive cell-permeable peptides as potent pharmacological compounds that decrease intracellular JNK signaling and confer long-term protection to pancreatic beta-cells from IL-1beta-induced apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Péptidos/farmacología , Péptidos/farmacocinética , Secuencia de Aminoácidos/genética , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular , Secuencia Conservada/genética , Humanos , Interleucina-1/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Péptidos/síntesis química , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Transactivadores/genética
5.
Genomics ; 64(3): 324-30, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10756100

RESUMEN

IB1/JIP-1 is a scaffold protein that regulates the c-Jun NH(2)-terminal kinase (JNK) signaling pathway, which is activated by environmental stresses and/or by treatment with proinflammatory cytokines including IL-1beta and TNF-alpha. The JNKs play an essential role in many biological processes, including the maturation and differentiation of immune cells and the apoptosis of cell targets of the immune system. IB1 is expressed predominantly in brain and pancreatic beta-cells where it protects cells from proapoptotic programs. Recently, a mutation in the amino-terminus of IB1 was associated with diabetes. A novel isoform, IB2, was cloned and characterized. Overall, both IB1 and IB2 proteins share a very similar organization, with a JNK-binding domain, a Src homology 3 domain, a phosphotyrosine-interacting domain, and polyacidic and polyproline stretches located at similar positions. The IB2 gene (HGMW-approved symbol MAPK8IP2) maps to human chromosome 22q13 and contains 10 coding exons. Northern and RT-PCR analyses indicate that IB2 is expressed in brain and in pancreatic cells, including insulin-secreting cells. IB2 interacts with both JNK and the JNK-kinase MKK7. In addition, ectopic expression of the JNK-binding domain of IB2 decreases IL-1beta-induced pancreatic beta-cell death. These data establish IB2 as a novel scaffold protein that regulates the JNK signaling pathway in brain and pancreatic beta-cells and indicate that IB2 represents a novel candidate gene for diabetes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cromosomas Humanos Par 22 , Proteínas Nucleares/genética , Transactivadores/genética , Apoptosis , Secuencia de Bases , Northern Blotting , Mapeo Cromosómico , Clonación Molecular , Humanos , Insulina/metabolismo , Secreción de Insulina , Interleucina-1/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalme del ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo , Células Tumorales Cultivadas
6.
J Biol Chem ; 275(22): 16466-72, 2000 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-10748095

RESUMEN

IB1/JIP-1 is a scaffold protein that interacts with upstream components of the c-Jun N-terminal kinase (JNK) signaling pathway. IB1 is expressed at high levels in pancreatic beta cells and may therefore exert a tight control on signaling events mediated by JNK in these cells. Activation of JNK by interleukin 1 (IL-1beta) or by the upstream JNK constitutive activator DeltaMEKK1 promoted apoptosis in two pancreatic beta cell lines and decreased IB1 content by 50-60%. To study the functional consequences of the reduced IB1 content in beta cell lines, we used an insulin-secreting cell line expressing an inducible IB1 antisense RNA that lead to a 38% IB1 decrease. Reducing IB1 levels in these cells increased phosphorylation of c-Jun and increased the apoptotic rate in presence of IL-1beta. Nitric oxide production was not stimulated by expression of the IB1 antisense RNA. Complementary experiments indicated that overexpression of IB1 in insulin-producing cells prevented JNK-mediated activation of the transcription factors c-Jun, ATF2, and Elk1 and decreased IL-1beta- and DeltaMEKK1-induced apoptosis. These data indicate that IB1 plays an anti-apoptotic function in insulin-producing cells probably by controlling the activity of the JNK signaling pathway.


Asunto(s)
Apoptosis/fisiología , Insulina/metabolismo , Interleucina-1/farmacología , Islotes Pancreáticos/efectos de los fármacos , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Apoptosis/efectos de los fármacos , Doxiciclina/farmacología , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de la radiación , Óxido Nítrico/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , ARN sin Sentido/genética , Rayos Ultravioleta
7.
Plant Physiol ; 87(1): 46-9, 1988 May.
Artículo en Inglés | MEDLINE | ID: mdl-16666124

RESUMEN

Nitrogen effects on the regulation of photosynthesis in wheat (Triticum aestivum L., cv Remia) seedlings were examined. Ribulose 1,5-bisphosphate carboxylase/oxygenase was rapidly extracted and tested for initial activity and for activity after incubation in presence of CO(2) and Mg(2+). Freeze clamped leaf segments were extracted for determinations of foliar steady state levels of ribulose 1,5-bisphosphate, triose phosphate, 3-phosphoglycerate, ATP, and ADP. Nitrogen deficient leaves showed increased ATP/ADP and triose phosphate/3-phosphoglycerate ratios suggesting increased assimilatory power. Ribulose 1,5-bisphosphate levels were decreased due to reduced pentose phosphate reductive cycle activity. Nevertheless, photosynthesis appeared to be limited by ribulose 1,5-bisphosphate carboxylase/oxygenase, independent of nitrogen nutrition. Its degree of activation was increased in nitrogen deficient plants and provided for maximum photosynthesis at decreased enzyme protein levels. It is suggested that ribulose 1,5-bisphosphate carboxylase/oxygenase activity is regulated according to the amount of assimilatory power.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...