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

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Genes Dev ; 28(24): 2712-25, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512559

RESUMEN

Cellular senescence is a stable proliferation arrest that suppresses tumorigenesis. Cellular senescence and associated tumor suppression depend on control of chromatin. Histone chaperone HIRA deposits variant histone H3.3 and histone H4 into chromatin in a DNA replication-independent manner. Appropriately for a DNA replication-independent chaperone, HIRA is involved in control of chromatin in nonproliferating senescent cells, although its role is poorly defined. Here, we show that nonproliferating senescent cells express and incorporate histone H3.3 and other canonical core histones into a dynamic chromatin landscape. Expression of canonical histones is linked to alternative mRNA splicing to eliminate signals that confer mRNA instability in nonproliferating cells. Deposition of newly synthesized histones H3.3 and H4 into chromatin of senescent cells depends on HIRA. HIRA and newly deposited H3.3 colocalize at promoters of expressed genes, partially redistributing between proliferating and senescent cells to parallel changes in expression. In senescent cells, but not proliferating cells, promoters of active genes are exceptionally enriched in H4K16ac, and HIRA is required for retention of H4K16ac. HIRA is also required for retention of H4K16ac in vivo and suppression of oncogene-induced neoplasia. These results show that HIRA controls a specialized, dynamic H4K16ac-decorated chromatin landscape in senescent cells and enforces tumor suppression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Senescencia Celular/fisiología , Chaperonas de Histonas/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Senescencia Celular/genética , Cromatina/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Chaperonas de Histonas/genética , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Papiloma/patología , Neoplasias Cutáneas/patología , Tamoxifeno/farmacología , Factores de Transcripción/genética
2.
Proc Natl Acad Sci U S A ; 110(40): 16009-14, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24043806

RESUMEN

Cellular senescence is a stable proliferation arrest associated with an altered secretory pathway (senescence-associated secretory phenotype). Cellular senescence is also a tumor suppressor mechanism, to which both proliferation arrest and senescence-associated secretory phenotype are thought to contribute. The melanocytes within benign human nevi are a paradigm for tumor-suppressive senescent cells in a premalignant neoplasm. Here a comparison of proliferating and senescent melanocytes and melanoma cell lines by RNA sequencing emphasizes the importance of senescence-associated proliferation arrest in suppression of transformation. Previous studies showed that activation of the Wnt signaling pathway can delay or bypass senescence. Consistent with this, we present evidence that repression of Wnt signaling contributes to melanocyte senescence in vitro. Surprisingly, Wnt signaling is active in many senescent human melanocytes in nevi, and this is linked to histological indicators of higher proliferative and malignant potential. In a mouse, activated Wnt signaling delays senescence-associated proliferation arrest to expand the population of senescent oncogene-expressing melanocytes. These results suggest that Wnt signaling can potentiate nevogenesis in vivo by delaying senescence. Further, we suggest that activated Wnt signaling in human nevi undermines senescence-mediated tumor suppression and enhances the probability of malignancy.


Asunto(s)
Senescencia Celular/fisiología , Melanocitos/fisiología , Melanoma/etiología , Nevo/fisiopatología , Vía de Señalización Wnt/fisiología , Animales , Línea Celular Tumoral , Cartilla de ADN/genética , Células HEK293 , Humanos , Immunoblotting , Inmunohistoquímica , Melanocitos/citología , Ratones , Análisis por Micromatrices , Nevo/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
3.
Cell Rep ; 12(9): 1483-96, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26299965

RESUMEN

Oncogene-induced senescence (OIS) is a tumor suppression mechanism that blocks cell proliferation in response to oncogenic signaling. OIS is frequently accompanied by multinucleation; however, the origin of this is unknown. Here, we show that multinucleate OIS cells originate mostly from failed mitosis. Prior to senescence, mutant H-RasV12 activation in primary human fibroblasts compromised mitosis, concordant with abnormal expression of mitotic genes functionally linked to the observed mitotic spindle and chromatin defects. Simultaneously, H-RasV12 activation enhanced survival of cells with damaged mitoses, culminating in extended mitotic arrest and aberrant exit from mitosis via mitotic slippage. ERK-dependent transcriptional upregulation of Mcl1 was, at least in part, responsible for enhanced survival and slippage of cells with mitotic defects. Importantly, mitotic slippage and oncogene signaling cooperatively induced senescence and key senescence effectors p21 and p16. In summary, activated Ras coordinately triggers mitotic disruption and enhanced cell survival to promote formation of multinucleate senescent cells.


Asunto(s)
Senescencia Celular , Células Gigantes/citología , Mitosis , Proteínas ras/metabolismo , Línea Celular , Células Cultivadas , Células Gigantes/metabolismo , Humanos , Mutación , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas ras/genética
4.
Acta Biomater ; 7(7): 2919-25, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21459166

RESUMEN

Biomimicry is being used in the next generation of biomaterials. Tuning material surface features such as chemistry, stiffness and topography allow the control of cell adhesion, proliferation, growth and differentiation. Here, microtopographical features with nanoscale depths, similar in scale to osteoclast resorption pits, were used to promote in vitro bone formation in basal medium. Primary human osteoblasts were used to represent an orthopaedically relevant cell type and analysis of adhesions, cytoskeleton, osteospecific proteins (phospho-Runx2 and osteopontin) and mineralisation (alizarin red) was performed. The results further demonstrate the potential for biomimicry in material design and show that the osteoblast response can be tuned from changes in feature size.


Asunto(s)
Biomimética , Osteogénesis/fisiología , Materiales Biocompatibles/metabolismo , Adhesión Celular/fisiología , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Humanos , Osteoblastos/citología , Osteoblastos/fisiología , Osteopontina/metabolismo , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA