Your browser doesn't support javascript.
loading
Aberrant expression and localization of the RAP1 shelterin protein contribute to age-related phenotypes.
Stock, Amanda J; McDevitt, Ross A; Puligilla, Chandrakala; Wang, Yajun; Zhang, Yongqing; Wang, Kun; Sun, Chongkui; Becker, Kevin G; Lehrmann, Elin; Wood, William H; Gong, Yi; Aqdas, Mohammad; Sung, Myong-Hee; Hoffmann, Victoria; Liu, Chengyu; Gorospe, Myriam; Harrington, Lea; Ferrucci, Luigi; Liu, Yie.
Afiliación
  • Stock AJ; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • McDevitt RA; Comparative Medicine Section, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Puligilla C; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Wang Y; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Zhang Y; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Wang K; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Sun C; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Becker KG; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Lehrmann E; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Wood WH; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Gong Y; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Aqdas M; Laboratory of Molecular Biology and Immunology, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Sung MH; Laboratory of Molecular Biology and Immunology, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Hoffmann V; Division of Veterinary Resources, National Institutes of Health, Bethesda, Maryland, United States of America.
  • Liu C; Transgenic Core Facility, National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, Maryland, United States of America.
  • Gorospe M; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Harrington L; Institute for Research in Immunology & Cancer, Marcelle-Coutu Pavilion, Université de Montréal, Montreal, Quebec, Canada.
  • Ferrucci L; Translational Gerontology Branch, Biomedical Research Center, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
  • Liu Y; Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America.
PLoS Genet ; 18(11): e1010506, 2022 11.
Article en En | MEDLINE | ID: mdl-36441670
ABSTRACT
Short telomeres induce a DNA damage response (DDR) that evokes apoptosis and senescence in human cells. An extant question is the contribution of telomere dysfunction-induced DDR to the phenotypes observed in aging and telomere biology disorders. One candidate is RAP1, a telomere-associated protein that also controls transcription at extratelomeric regions. To distinguish these roles, we generated a knockin mouse carrying a mutated Rap1, which was incapable of binding telomeres and did not result in eroded telomeres or a DDR. Primary Rap1 knockin embryonic fibroblasts showed decreased RAP1 expression and re-localization away from telomeres, with an increased cytosolic distribution akin to that observed in human fibroblasts undergoing telomere erosion. Rap1 knockin mice were viable, but exhibited transcriptomic alterations, proinflammatory cytokine/chemokine signaling, reduced lifespan, and decreased healthspan with increased body weight/fasting blood glucose levels, spontaneous tumor incidence, and behavioral deficits. Taken together, our data present mechanisms distinct from telomere-induced DDR that underlie age-related phenotypes.
Asunto(s)

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Telómero / Complejo Shelterina Límite: Animals / Humans Idioma: En Revista: PLoS Genet Asunto de la revista: GENETICA Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Telómero / Complejo Shelterina Límite: Animals / Humans Idioma: En Revista: PLoS Genet Asunto de la revista: GENETICA Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos