traductor

viernes, 8 de mayo de 2015

A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging

A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging

  1. Juan Carlos Izpisua Belmonte3,
+Author Affiliations
  1. 1National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
  2. 2Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China.
  3. 3Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
  4. 4State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
  5. 5Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
  6. 6Diagnosis and Treatment Center for Oral Disease, the 306th Hospital of the PLA, Beijing, China.
  7. 7College of Life Sciences, Peking University, Beijing 100871, China.
  8. 8The Center for Anti-aging and Regenerative Medicine, Shenzhen University, Shenzhen 518060, China.
  9. 9Universidad Católica San Antonio de Murcia, Campus de los Jerónimos s/n, 30107 Guadalupe, Murcia, Spain.
  10. 10Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China.
  11. 11Center for Molecular and Translational Medicine (CMTM), Beijing 100101, China.
  12. 12Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
  13. 13Beijing Institute for Brain Disorders, Beijing100069, China.
  1. Corresponding author. E-mail: ghliu@ibp.ac.cn (G.-H.L.); tangfuchou@pku.edu.cn (F.T.); belmonte@salk.edu (J.C.I.B.)
  1. * These authors contributed equally to this work.
Werner syndrome (WS) is a premature aging disorder caused by WRN protein deficiency. Here, we report on the generation of a human WS model in human embryonic stem cells (ESCs). Differentiation of WRN-null ESCs to mesenchymal stem cells (MSCs) recapitulates features of premature cellular aging, a global loss of H3K9me3, and changes in heterochromatin architecture. We show that WRN associates with heterochromatin proteins SUV39H1 and HP1α and nuclear lamina-heterochromatin anchoring protein LAP2β. Targeted knock-in of catalytically inactive SUV39H1 in wild-type MSCs recapitulates accelerated cellular senescence, resembling WRN-deficient MSCs. Moreover, decrease in WRN and heterochromatin marks are detected in MSCs from older individuals. Our observations uncover a role for WRN in maintaining heterochromatin stability and highlight heterochromatin disorganization as a potential determinant of human aging.

No hay comentarios: