The mismatch repair-mediated cell cycle checkpoint response to fluorodeoxyuridine

The loss of DNA mismatch repair (MMR) is responsible for hereditary nonpolyposis colorectal cancer and a subset of sporadic tumors. Acquired resistance or tolerance to some anti‐cancer drugs occurs when MMR function is impaired. 5‐Fluorouracil (FU), an anti‐cancer drug used in the treatment of advan...

Full description

Saved in:
Bibliographic Details
Published inJournal of cellular biochemistry Vol. 105; no. 1; pp. 245 - 254
Main Authors Liu, Angen, Yoshioka, Ken-ichi, Salerno, Vincenzo, Hsieh, Peggy
Format Journal Article
LanguageEnglish
Published Hoboken Wiley Subscription Services, Inc., A Wiley Company 01.09.2008
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:The loss of DNA mismatch repair (MMR) is responsible for hereditary nonpolyposis colorectal cancer and a subset of sporadic tumors. Acquired resistance or tolerance to some anti‐cancer drugs occurs when MMR function is impaired. 5‐Fluorouracil (FU), an anti‐cancer drug used in the treatment of advanced colorectal and other cancers, and its metabolites are incorporated into RNA and DNA and inhibit thymidylate synthase resulting in depletion of dTTP and incorporation in DNA of uracil. Although the MMR deficiency has been implicated in tolerance to FU, the mechanism of cell killing remains unclear. Here, we examine the cellular response to fluorodeoxyuridine (FdU) and the role of the MMR system. After brief exposure of cells to low doses of FdU, MMR mediates DNA damage signaling during S‐phase and triggers arrest in G2/M in the first cell cycle in a manner requiring MutSα, MutLα, and DNA replication. Cell cycle arrest is mediated by ATR kinase and results in phosphorylation of Chk1 and SMC1. MutSα binds FdU:G mispairs in vitro consistent with its being a DNA damage sensor. Prolonged treatment with FdU results in an irreversible arrest in G2 that is independent of MMR status and leads to the accumulation of DNA lesions that are targeted by the base excision repair (BER) pathway. Thus, MMR can act as a direct sensor of FdU‐mediated DNA lesions eliciting cell cycle arrest via the ATR/Chk1 pathway. However, at higher levels of damage, other damage surveillance pathways such as BER also play important roles. J. Cell. Biochem. 105: 245–254, 2008. © 2008 Wiley‐Liss, Inc.
Bibliography:istex:FDF76102C51F760DD196E595D7C5BAB50DC2387D
ArticleID:JCB21824
ark:/67375/WNG-ZH0N6SF9-B
Intramural Research Program of the National Institute of Diabetes and Digestive and Kidney Diseases
NIH
ObjectType-Article-2
SourceType-Scholarly Journals-1
ObjectType-Feature-1
content type line 23
Angen Liu's present address is Fox Chase Cancer Center, Genomics Core Facility, 333 Cottman Avenue, Philadelphia, PA 19111−2497.
Ken-ichi Yoshioka's present address is Department of Pathological Biochemistry, Tokyo Medical and Dental University, 2−3−10 Kandasurugadai, Chiyoda-ku, Tokyo 101−0062, Japan.
ISSN:0730-2312
1097-4644
DOI:10.1002/jcb.21824