4.3 Article

XRCC1 and base excision repair balance in response to nitric oxide

Journal

DNA REPAIR
Volume 10, Issue 12, Pages 1282-1293

Publisher

ELSEVIER SCIENCE BV
DOI: 10.1016/j.dnarep.2011.10.008

Keywords

XRCC1; Base excision repair; Inflammation; Nitric oxide; SIN1; Glycosylase

Funding

  1. MIT Center for Environmental Health Sciences [NIEHS P30-ES002109]
  2. Pittsburgh Foundation
  3. National Institutes of Health (NIH) [GM087798, CA148629, P01-CA026731, 2-R01-CA079827-05A1, ES019498, P30 CA047904]
  4. University of Pittsburgh Department of Pharmacology & Chemical Biology
  5. [U01-ES016045]

Ask authors/readers for more resources

Inflammation associated reactive oxygen and nitrogen species (RONs), including peroxynitrite (ONOO-) and nitric oxide (NO center 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 NO center dot. XRCC1 null cells were slightly more sensitive to SIN-1 than wild-type cells. We used small-scale bioreactors to expose cells to NO center 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 NO center dot-induced DNA lesions: 1,N-6-ethenoadenine and hypoxanthine. Furthermore, overexpression of AAG rendered XRCC1-deficient cells sensitive to NO center dot-induced DNA damage. These results show that AAG is a key glycosylase for BER of NO center 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. Published by Elsevier B.V.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.3
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available