首页 | 本学科首页   官方微博 | 高级检索  
     


XRCC1 and base excision repair balance in response to nitric oxide
Authors:Mutamba James T  Svilar David  Prasongtanakij Somsak  Wang Xiao-Hong  Lin Ying-Chih  Dedon Peter C  Sobol Robert W  Engelward Bevin P
Affiliation:a Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
b Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
c University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA 15213, United States
d Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15213, United States
Abstract:Inflammation associated reactive oxygen and nitrogen species (RONs), including peroxynitrite (ONOO) and nitric oxide (NOradical dot), create base lesions that potentially play a role in the toxicity and large genomic rearrangements associated with many malignancies. Little is known about the role of base excision repair (BER) in removing these endogenous DNA lesions. Here, we explore the role of X-ray repair cross-complementing group 1 (XRCC1) in attenuating RONs-induced genotoxicity. XRCC1 is a scaffold protein critical for BER for which polymorphisms modulate the risk of cancer. We exploited CHO and human glioblastoma cell lines engineered to express varied levels of BER proteins to study XRCC1. Cytotoxicity and the levels of DNA repair intermediates (single-strand breaks; SSB) were evaluated following exposure of the cells to the ONOO donor, SIN-1, and to gaseous NOradical dot. XRCC1 null cells were slightly more sensitive to SIN-1 than wild-type cells. We used small-scale bioreactors to expose cells to NOradical dot and found that XRCC1-deficient CHO cells were not sensitive. However, using a molecular beacon assay to test lesion removal in vitro, we found that XRCC1 facilitates AAG-initiated excision of two key NOradical dot-induced DNA lesions: 1,N6-ethenoadenine and hypoxanthine. Furthermore, overexpression of AAG rendered XRCC1-deficient cells sensitive to NOradical dot-induced DNA damage. These results show that AAG is a key glycosylase for BER of NOradical dot-induced DNA damage and that XRCC1's role in modulating sensitivity to RONs is dependent upon the cellular level of AAG. This demonstrates the importance of considering the expression of other components of the BER pathway when evaluating the impact of XRCC1 polymorphisms on cancer risk.
Keywords:XRCC1   Base excision repair   Inflammation   Nitric oxide   SIN1   Glycosylase
本文献已被 ScienceDirect PubMed 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