Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Exp Cell Res ; 316(9): 1535-52, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20233592

RESUMEN

Human HspB1 (also denoted Hsp27) is an oligomeric anti-apoptotic protein that has tumorigenic and metastatic roles. To approach the structural organizations of HspB1 that are active in response to apoptosis inducers acting through different pathways, we have analyzed the relative protective efficiency induced by this protein as well its localization, oligomerization and phosphorylation. HeLa cells, that constitutively express high levels of HspB1 were treated with either etoposide, Fas agonist antibody, staurosporine or cytochalasin D. Variability in HspB1 efficiency to interfere with the different apoptotic transduction pathways induced by these agents were detected. Moreover, inducer-specific dynamic changes in HspB1 localization, native size and phosphorylation were observed, that differed from those observed after heat shock. Etoposide and Fas treatments gradually shifted HspB1 towards large but differently phosphorylated oligomeric structures. In contrast, staurosporine and cytochalasin D induced the rapid but transient formation of small oligomers before large structures were formed. These events correlated with inducer-specific phosphorylations of HspB1. Of interest, the formation of small oligomers in response to staurosporine and cytochalasin D was time correlated with the rapid disruption of F-actin. The subsequent, or gradual in the case of etoposide and Fas, formation of large oligomeric structures was a later event concomitant with the early phase of caspase activation. These observations support the hypothesis that HspB1 has the ability, through specific changes in its structural organization, to adapt and interfere at several levels with challenges triggered by different signal transduction pathways upstream of the execution phase of apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteínas de Choque Térmico HSP27/metabolismo , Transducción de Señal , Actinas/metabolismo , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Caspasa 3/metabolismo , Citocalasina D/farmacología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Técnica del Anticuerpo Fluorescente , Células HeLa , Proteínas de Choque Térmico , Respuesta al Choque Térmico , Humanos , Mitocondrias/efectos de los fármacos , Chaperonas Moleculares , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación/efectos de los fármacos , Estaurosporina/farmacología , Receptor fas/metabolismo
2.
Mol Cell Biol ; 22(3): 816-34, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11784858

RESUMEN

We previously showed that Hsp27 protects against apoptosis through its interaction with cytosolic cytochrome c. We have revisited this protective activity in murine cell lines expressing different levels of Hsp27. We report that Hsp27 also interferes, in a manner dependent on level of expression, with the release of cytochrome c from mitochondria. Moreover, a decreased level of endogenous Hsp27, which sensitized HeLa cells to apoptosis, reduced the delay required for cytochrome c release and procaspase 3 activation. The molecular mechanism regulating this function of Hsp27 is unknown. In our cell systems, Hsp27 is mainly cytosolic and only a small fraction of this protein colocalized with mitochondria. Moreover, we show that only a very small fraction of cytochrome c interacts with Hsp27, hence excluding a role of this interaction in the retention of cytochrome c in mitochondria. We also report that Bid intracellular relocalization was altered by changes in Hsp27 level of expression, suggesting that Hsp27 interferes with apoptotic signals upstream of mitochondria. We therefore investigated if the ability of Hsp27 to act as an expression-dependent modulator of F-actin microfilaments integrity was linked to the retention of cytochrome c in mitochondria. We show here that the F-actin depolymerizing agent cytochalasin D rapidly induced the release of cytochrome c from mitochondria and caspase activation. This phenomenon was delayed in cells pretreated with the F-actin stabilizer phalloidin and in cells expressing a high level of Hsp27. This suggests the existence of an apoptotic signaling pathway linking cytoskeleton damages to mitochondria. This pathway, which induces Bid intracellular redistribution, is negatively regulated by the ability of Hsp27 to protect F-actin network integrity. However, this upstream pathway is probably not the only one to be regulated by Hsp27 since, in staurosporine-treated cells, phalloidin only partially inhibited cytochrome c release and caspase activation. Moreover, in etoposide-treated cells, Hsp27 still delayed the release of cytochrome c from mitochondria and Bid intracellular redistribution in conditions where F-actin was not altered.


Asunto(s)
Grupo Citocromo c/metabolismo , Proteínas de Choque Térmico , Proteínas de Neoplasias/metabolismo , Células 3T3 , Actinas/metabolismo , Animales , Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Activación Enzimática , Etopósido/farmacología , Expresión Génica , Glutatión/metabolismo , Proteínas de Choque Térmico HSP27 , Células HeLa , Humanos , Cinética , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Chaperonas Moleculares , Proteínas de Neoplasias/genética , Estaurosporina/farmacología
3.
Antioxid Redox Signal ; 7(3-4): 414-22, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15706088

RESUMEN

Small stress proteins [small heat shock proteins (sHsps)] are molecular chaperones that modulate the ability of cells to respond to oxidative stress. The current knowledge concerning the protective mechanism generated by the expression of mammalian heat shock protein-27 (Hsp27) that allows cells to increase their resistance to oxidative stress is presented. We describe the effects mediated by Hsp27 expression toward crucial enzymes such as glucose-6-phosphate dehydrogenase and glutathione reductase that uphold glutathione in its reduced form. New data are presented showing that the expression of sHsps correlates with a drastic decrease in the intracellular level of iron, a catalyzer of hydroxyl radical (OH( . )) generation. A decreased ability of sHsps expressing cells to concentrate iron will therefore end up in a decreased level of oxidized proteins. In addition, we propose a role of Hsp27 in the presentation of oxidized proteins to the proteasome degradation machinery. We also present an analysis of several Hsp27 mutants that suggests that the C-terminal part of this stress protein is essential for its protective activity against oxidative stress.


Asunto(s)
Glutatión/metabolismo , Proteínas de Choque Térmico/fisiología , Hierro/metabolismo , Estrés Oxidativo , Animales , Regulación hacia Abajo , Proteínas de Choque Térmico/genética , Homeostasis , Ratones , Oxidación-Reducción
4.
PLoS One ; 8(8): e70545, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950959

RESUMEN

Several human small heat shock proteins (sHsps) are phosphorylated oligomeric chaperones that enhance stress resistance. They are characterized by their ability to interact and form polydispersed hetero-oligomeric complexes. We have analyzed the cellular consequences of the stable expression of either wild type HspB5 or its cataracts and myopathies inducing R120G mutant in growing and oxidative stress treated HeLa cells that originally express only HspB1. Here, we describe that wild type and mutant HspB5 induce drastic and opposite effects on cell morphology and oxidative stress resistance. The cellular distribution and phosphorylation of these polypeptides as well as the oligomerization profile of the resulting hetero-oligomeric complexes formed by HspB1 with the two types of exogenous polypeptides revealed the dominant effects induced by HspB5 polypeptides towards HspB1. The R120G mutation enhanced the native size and salt resistance of HspB1-HspB5 complex. However, in oxidative conditions the interaction between HspB1 and mutant HspB5 was drastically modified resulting in the aggregation of both partners. The mutation also induced the redistribution of HspB1 phosphorylated at serine 15, originally observed at the level of the small oligomers that do not interact with wild type HspB5, to the large oligomeric complex formed with mutant HspB5. This phosphorylation stabilized the interaction of HspB1 with mutant HspB5. A dominant negative effect towards HspB1 appears therefore as an important event in the cellular sensitivity to oxidative stress mediated by mutated HspB5 expression. These observations provide novel data that describe how a mutated sHsp can alter the protective activity of another member of this family of chaperones.


Asunto(s)
Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Mutación , Cadena B de alfa-Cristalina/genética , Cadena B de alfa-Cristalina/metabolismo , Expresión Génica , Proteínas de Choque Térmico HSP27/química , Células HeLa , Humanos , Estrés Oxidativo/genética , Fosforilación , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas , Cadena B de alfa-Cristalina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA