Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Cell Cycle ; 7(9): 1262-8, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18418057

RESUMEN

Telomere attrition, DNA damage and constitutive mitogenic signaling can all trigger cellular senescence in normal cells and serve as a defense against tumor progression. Cancer cells may circumvent this cellular defense by acquiring genetic mutations in checkpoint proteins responsible for regulating permanent cell cycle arrest. A small family of tumor suppressor genes encoding the retinoblastoma susceptibility protein family (Rb, p107, p130) exerts a partially redundant control of entry into S phase of DNA replication and cellular proliferation. Here we report that activation of the p53-dependent DNA damage response has been found to accelerate senescence in human prostate cancer cells lacking a functional Rb protein. This novel form of irradiation-induced premature cellular senescence reinforces the notion that other Rb family members may compensate for loss of Rb protein in the DNA damage response pathway. Consistent with this hypothesis, depletion of p107 potently inhibits the irradiation-induced senescence observed in DU145 cells. In contrast, p130 depletion triggers a robust and unexpected form of premature senescence in unirradiated cells. The dominant effect of depleting both p107 and p130, in the absence of Rb, was a complete blockade of irradiation-induced cellular senescence. Onset of the p107-dependent senescence was temporally associated with p53-mediated stabilization of the cyclin-dependent kinase inhibitor p27 and decreases in c-myc and cks1 expression. These results indicate that p107 is required for initiation of accelerated cellular senescence in the absence of Rb and introduces the concept that p130 may be required to prevent the onset of terminal growth arrest in unstimulated prostate cancer cells lacking a functional Rb allele.


Asunto(s)
Carcinoma/genética , Senescencia Celular/genética , Proteína Sustrato Asociada a CrK/genética , Neoplasias de la Próstata/genética , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/efectos de la radiación , Senescencia Celular/efectos de la radiación , Proteína Sustrato Asociada a CrK/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/genética , Regulación hacia Abajo/genética , Genes cdc/fisiología , Genes cdc/efectos de la radiación , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Radiación , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
2.
Cancer Res ; 68(19): 7864-71, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829542

RESUMEN

Males of advanced age represent a rapidly growing population at risk for prostate cancer. In the contemporary setting of earlier detection, a majority of prostate carcinomas are still clinically localized and often treated using radiation therapy. Our recent studies have shown that premature cellular senescence, rather than apoptosis, accounts for most of the clonogenic death induced by clinically relevant doses of irradiation in prostate cancer cells. We show here that this treatment-induced senescence was associated with a significantly increased release of exosome-like microvesicles. In premature senescence, this novel secretory phenotype was dependent on the activation of p53. In addition, the release of exosome-like microvesicles also increased during proliferative senescence in normal human diploid fibroblasts. These data support the hypothesis that senescence, initiated either by telomere attrition (e.g., aging) or DNA damage (e.g., radiotherapy), may induce a p53-dependent increase in the biogenesis of exosome-like vesicles. Ultrastructural analysis and RNA interference-mediated knockdown of Tsg101 provided significant evidence that the additional exosomes released by prematurely senescent prostate cancer cells were principally derived from multivesicular endosomes. Moreover, these exosomes were enriched in B7-H3 protein, a recently identified diagnostic marker for prostate cancer, and an abundance of what has recently been termed "exosomal shuttle RNA." Our findings are consistent with the proposal that exosomes can transfer cargos, with both immunoregulatory potential and genetic information, between cells through a novel mechanism that may be recruited to increase exosome release during accelerated and replicative cellular senescence.


Asunto(s)
Carcinoma/metabolismo , Senescencia Celular/fisiología , Neoplasias de la Próstata/metabolismo , Vesículas Secretoras/metabolismo , Carcinoma/patología , Endosomas/metabolismo , Endosomas/efectos de la radiación , Exocitosis/fisiología , Humanos , Masculino , Neoplasias de la Próstata/patología , Vesículas Secretoras/efectos de la radiación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
3.
J Biol Chem ; 282(35): 25884-92, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17604278

RESUMEN

Gelsolin regulates the dynamic assembly and disassembly of the actin-based cytoskeleton in non-muscle cells and clears the circulation of filaments released following cell death. Gelsolin is a six-domain (G1-G6) protein activated by calcium via a multi-step process that involves unfolding from a compact form to a more open form in which the three actin-binding sites (on the G1, G2, and G4 subdomains) become exposed. To follow the global structural changes that accompany calcium activation of gelsolin, small-angle x-ray scattering (SAXS) data were collected for full-length human plasma gelsolin at nanomolar to millimolar concentrations of free Ca2+. Analysis of these data showed that, upon increasing free Ca2+ levels, the radius of gyration (Rg) increased nearly 12 A, from 31.1+/-0.3 to 43+/-2 A, and the maximum linear dimension (Dmax) of the gelsolin molecule increased 55 A, from 100 to 155A. Structural reconstruction of gelsolin from these data provided a striking visual tracking of the gradual Ca2+-induced opening of the gelsolin molecule and highlighted the critical role played by the flexible linkers between homologous domains. The tightly packed architecture of calcium-free gelsolin, seen from both SAXS and x-ray crystallographic models, is already partially opened up in as low as 0.5 nM Ca2+. Our data confirm that, although the molecule springs open from 0 to 1 microM free Ca2+, even higher calcium concentrations help to stabilize a more open structure, with increases in Rg and Dmax of approximately 2 and approximately 15 A, respectively. At these higher calcium levels, the SAXS-based models provide a molecular shape that is compatible with that of the crystal structures solved for Ca2+/gelsolin C-terminal and N-terminal halves+/-monomeric G-actin. Placement of these crystal structures within the boundaries of the SAXS-based model suggests a movement of the G1/G2 subunits that would be required upon binding to actin.


Asunto(s)
Actinas/química , Calcio/química , Gelsolina/química , Modelos Moleculares , Actinas/metabolismo , Calcio/metabolismo , Cristalografía por Rayos X , Citoesqueleto/química , Citoesqueleto/metabolismo , Gelsolina/metabolismo , Humanos , Unión Proteica/fisiología , Estructura Cuaternaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología
4.
Cell Cycle ; 6(5): 595-605, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17351335

RESUMEN

Because p53 inactivation may limit the effectiveness of radiation therapy for localized prostate cancer, it is important to understand how this gene regulates clonogenic survival after an exposure to ionizing radiation. Here, we show that premature cellular senescence is the principal mode of cell death accounting for the radiosensitivity of human prostate cancer cell lines retaining p53 function. Alternative stress response pathways controlled by this tumor suppressor, including cell cycle arrest, DNA damage repair, mitotic catastrophe and apoptosis, contributed significantly less to radiation-induced clonogenic death. Using a dominant negative C-terminal fragment of p53, we present the first evidence that a complete loss of endogenous p53 function is sufficient to limit the irradiation-induced senescence and clonogenic death of prostate cancer cells. Conversely, inheritance of wild-type p53 by prostate cancer cells lacking a functional allele of this gene (i.e., DU145) significantly increases clonogenic death through p53-dependent cellular senescence and apoptotic pathways. Our data provide evidence that mutations of even one p53 allele may be sufficient to alter their clonogenic fate. In addition, they support the idea that the p53 pathway can be used as a specific target for enhancing the radiosensitivity of prostate cancer cells. Activation of p53 by the drug nutlin-3 is shown to be an effective radiosensitizer of prostate cancer cells retaining functional alleles of p53 and this effect was entirely attributable to an increased induction of p53-dependent cellular senescence.


Asunto(s)
Senescencia Celular/efectos de la radiación , Neoplasias de la Próstata , Tolerancia a Radiación/efectos de la radiación , Proteína p53 Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/efectos de la radiación , Línea Celular Tumoral , Senescencia Celular/fisiología , Relación Dosis-Respuesta en la Radiación , Humanos , Masculino , Neoplasias de la Próstata/fisiopatología , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda