Tachpyridine, a metal chelator, induces G2 cell-cycle arrest, activates checkpoint kinases, and sensitizes cells to ionizing radiation

Iron is critical for cell growth and proliferation. Iron chelators are being explored for a number of clinical applications, including the treatment of neurodegenerative disorders, heart disease, and cancer. To uncover mechanisms of action of tachpyridine, a chelator currently undergoing preclinical...

Full description

Saved in:
Bibliographic Details
Published inBlood Vol. 106; no. 9; pp. 3191 - 3199
Main Authors Turner, JoLyn, Koumenis, Constantinos, Kute, Timothy E., Planalp, Roy P., Brechbiel, Martin W., Beardsley, Dillon, Cody, Brooke, Brown, Kevin D., Torti, Frank M., Torti, Suzy V.
Format Journal Article
LanguageEnglish
Published Washington, DC Elsevier Inc 01.11.2005
The Americain Society of Hematology
2005 by The American Society of Hematology
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Iron is critical for cell growth and proliferation. Iron chelators are being explored for a number of clinical applications, including the treatment of neurodegenerative disorders, heart disease, and cancer. To uncover mechanisms of action of tachpyridine, a chelator currently undergoing preclinical evaluation as an anticancer agent, cell-cycle analysis was performed. Tachpyridine arrested cells at G2, a radiosensitive phase of the cell cycle, and enhanced the sensitivity of cancer cells but not nontransformed cells to ionizing radiation. G2 arrest was p53 independent and was accompanied by activation of the checkpoint kinases CHK1 and CHK2. G2 arrest was blocked by UCN-01, a CHK1 inhibitor, but proceeded in CHK2 knock-out cells, indicating a critical role for CHK1 in G2 arrest. Tachpyridine-induced cell-cycle arrest was abrogated in cells treated with caffeine, an inhibitor of the ataxia-telangiectasia mutated/ataxia-telangiectasia-mutated and Rad3-related (ATM/ATR) kinases. Further, G2 arrest proceeded in ATM-deficient cells but was blocked in ATR-deficient cells, implicating ATR as the proximal kinase in tachpyridine-mediated G2 arrest. Collectively, our results suggest that iron chelators may function as antitumor and radioenhancing agents and uncover a previously unexplored activity of iron chelators in activation of ATR and checkpoint kinases.
Bibliography:Reprints: Suzy V. Torti, Department of Biochemistry, Medical Center Blvd, Winston Salem, NC 27157; e-mail: storti@wfubmc.edu.
The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.
Prepublished online as Blood First Edition Paper, July 12, 2005; DOI 10.1182/blood-2005-03-1263.
Supported by the National Institutes of Health (grant no. DK 57781) (S.V.T.); preclinical development of tachpyridine was supported by a grant from the National Cancer Institute (NCI) Rapid Access to Intervention Development (RAID) and the NCI Rapid Access to NCI Discovery Resources (RAND) programs of the National Institutes of Health (S.V.T.); and grant CA102289 (K.B.).
ISSN:0006-4971
1528-0020
DOI:10.1182/blood-2005-03-1263