首页 | 本学科首页   官方微博 | 高级检索  
     


The oncogenic phosphatase WIP1 negatively regulates nucleotide excision repair
Authors:Thuy-Ai Nguyen  Scott D. Slattery  Sung-Hwan Moon  Yolanda F. Darlington  Xiongbin Lu  Lawrence A. Donehower
Affiliation:1. Interdepartmental Graduate Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, United States;2. Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030, United States;3. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States;4. Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, United States;1. DuPont Haskell Global Centers for Health & Environmental Sciences, P.O. Box 30, 1090 Elkton Road, Newark, DE, USA;2. University of the Sciences, Department of Biological Sciences, Cell and Molecular Biology Graduate Program, Philadelphia, PA, USA;1. Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan;2. Gastrointestinal Cancer Research Laboratory, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas;3. Department of Pathology, Chonnam National University Medical School, Gwangju, Korea;4. Department of Surgery, Chonnam National University Medical School, Gwangju, Korea;5. Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan;6. Department of Molecular Immunology, Toho University Faculty of Medicine, Tokyo, Japan;7. Department of Gastroenterological Surgery and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan;8. Department of Gastrointestinal and Pediatric Surgery, Graduate School of Medicine Mie University, Mie, Japan;9. Department of Biostatistics School of Public Health, University of Michigan, Ann Arbor, Michigan;1. Centro de Biotecnologia, Instituto Butantan, São Paulo, 05503-900, Brazil;2. Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;3. Global Antibiotics Research and Development Partnership (GARDP), Drugs for Neglected Diseases initiative (DNDi), Chemin Louis-Dunant 15, 1202 Geneva, Switzerland;4. Laboratório de Imunogenética, Instituto Butantan, São Paulo, 05503-900, Brazil;5. Laboratório Especial de Inovação e Desenvolvimento Industrial, Instituto Butantan, São Paulo, 05503-900, Brazil
Abstract:Nucleotide excision repair (NER) is the only mechanism in humans to repair UV-induced DNA lesions such as pyrimidine (6-4) pyrimidone photoproducts and cyclobutane pyrimidine dimers (CPDs). In response to UV damage, the ataxia telangiectasia mutated and Rad3-related (ATR) kinase phosphorylates and activates several downstream effector proteins, such as p53 and XPA, to arrest cell cycle progression, stimulate DNA repair, or initiate apoptosis. However, following the completion of DNA repair, there must be active mechanisms that restore the cell to a prestressed homeostatic state. An important part of this recovery must include a process to reduce p53 and NER activity as well as to remove repair protein complexes from the DNA damage sites. Since activation of the damage response occurs in part through phosphorylation, phosphatases are obvious candidates as homeostatic regulators of the DNA damage and repair responses. Therefore, we investigated whether the serine/threonine wild-type p53-induced phosphatase 1 (WIP1/PPM1D) might regulate NER. WIP1 overexpression inhibits the kinetics of NER and CPD repair, whereas WIP1 depletion enhances NER kinetics and CPD repair. This NER suppression is dependent on WIP1 phosphatase activity, as phosphatase-dead WIP1 mutants failed to inhibit NER. Moreover, WIP1 suppresses the kinetics of UV-induced damage repair largely through effects on NER, as XPD-deficient cells are not further suppressed in repairing UV damage by overexpressed WIP1. Wip1 null mice quickly repair their CPD and undergo less UV-induced apoptosis than their wild-type counterparts. In vitro phosphatase assays identify XPA and XPC as two potential WIP1 targets in the NER pathway. Thus WIP1 may suppress NER kinetics by dephosphorylating and inactivating XPA and XPC and other NER proteins and regulators after UV-induced DNA damage is repaired.
Keywords:
本文献已被 ScienceDirect 等数据库收录!
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号