Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Life Sci Space Res (Amst) ; 20: 101-112, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30797427

RESUMEN

There exists a wide degree of genetic variation within the normal human population which includes disease free individuals with heterozygote defects in major DNA repair genes. A lack of understanding of how this genetic variation impacts cellular phenotypes that inform cancer risk post heavy ion exposure poses a major limitation in developing personalized cancer risk assessment astronauts. We initiated a pilot study with Human Mammary Epithelial Cell strains (HMEC) derived from wild type, a p16 silenced derivative of wild type, and various genetic variants that were heterozygote for DNA repair genes; BRCA1, BRCA2 and ATM. Cells strains were exposed to different high and low LET radiation qualities to generate both simple and complex lesions and centrosome aberrations were examined as a surrogate marker of genomic instability and cancer susceptibility post different exposures. Our results indicate that centrosome aberration frequency is higher in the genetic variants under study. The aberration frequency increases with dose, complexity of the lesion generated by different radiation qualities and age of the individual. This increase in genomic instability correlates with elevated check-point activation post radiation exposure. These studies suggest that the influence of individual genetics on cell cycle regulation could modify the degree of early genomic instability in response to complex lesions and potentially define cancer predisposition in response to HZE exposure. These results will have significant implications in estimating cancer susceptibility in genetically variant individuals exposed to HZE particles.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/patología , Mama/patología , Aberraciones Cromosómicas , Radiación Cósmica , Variación Genética , Mama/metabolismo , Mama/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Células Cultivadas , Daño del ADN , Femenino , Humanos , Fenotipo , Fosfoproteínas , Proyectos Piloto
2.
Aging (Albany NY) ; 9(3): 665-686, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28245431

RESUMEN

Exposures to various DNA damaging agents can deregulate a wide array of critical mechanisms that maintain genome integrity. It is unclear how these processes are impacted by one's age at the time of exposure and the complexity of the DNA lesion. To clarify this, we employed radiation as a tool to generate simple and complex lesions in normal primary human mammary epithelial cells derived from women of various ages. We hypothesized that genomic instability in the progeny of older cells exposed to complex damages will be exacerbated by age-associated deterioration in function and accentuate age-related cancer predisposition. Centrosome aberrations and changes in stem cell numbers were examined to assess cancer susceptibility. Our data show that the frequency of centrosome aberrations proportionately increases with age following complex damage causing exposures. However, a dose-dependent increase in stem cell numbers was independent of both age and the nature of the insult. Phospho-protein signatures provide mechanistic clues to signaling networks implicated in these effects. Together these studies suggest that complex damage can threaten the genome stability of the stem cell population in older people. Propagation of this instability is subject to influence by the microenvironment and will ultimately define cancer risk in the older population.


Asunto(s)
Envejecimiento/patología , Neoplasias de la Mama/patología , Centrosoma/efectos de la radiación , Células Epiteliales/efectos de la radiación , Células Madre/efectos de la radiación , Adulto , Anciano , Células Cultivadas , Daño del ADN/efectos de la radiación , Susceptibilidad a Enfermedades , Células Epiteliales/patología , Femenino , Inestabilidad Genómica , Humanos , Persona de Mediana Edad , Células Madre/fisiología
3.
Cell Metab ; 22(5): 895-906, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26456335

RESUMEN

Many genes that affect replicative lifespan (RLS) in the budding yeast Saccharomyces cerevisiae also affect aging in other organisms such as C. elegans and M. musculus. We performed a systematic analysis of yeast RLS in a set of 4,698 viable single-gene deletion strains. Multiple functional gene clusters were identified, and full genome-to-genome comparison demonstrated a significant conservation in longevity pathways between yeast and C. elegans. Among the mechanisms of aging identified, deletion of tRNA exporter LOS1 robustly extended lifespan. Dietary restriction (DR) and inhibition of mechanistic Target of Rapamycin (mTOR) exclude Los1 from the nucleus in a Rad53-dependent manner. Moreover, lifespan extension from deletion of LOS1 is nonadditive with DR or mTOR inhibition, and results in Gcn4 transcription factor activation. Thus, the DNA damage response and mTOR converge on Los1-mediated nuclear tRNA export to regulate Gcn4 activity and aging.


Asunto(s)
Envejecimiento/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Longevidad/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Saccharomyces cerevisiae/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Caenorhabditis elegans/genética , Restricción Calórica , Daño del ADN/genética , Eliminación de Gen , Regulación de la Expresión Génica/genética , Genoma , ARN de Transferencia/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA