DNA topoisomerase I (top1) has become an essential target for anticancer research since the discovery that camptothecins are specific top1 poisons and exhibit promising anticancer activity. Camptothecins act by stabilizing top1-linked DNA breaks (cleavable complexes) which can be detected as protein-associated DNA breaks in drug-treated cells. Our studies are aimed at addressing several fundamental questions regarding the top1 cleavable complexes: (1) what are the molecular interactions between camptothecins and top1? (2) Can we discover and characterize new top1 inhibitors? (3) Are cleavable complexes trapped by other DNA alterations (base modifications, mismatches, DNA breaks, gaps...)? (4) How are the trapped cleavable complexes processed in cells and subcellular systems; and (3) what is the role(s) of topoisomerases in some cellular responses to DNA damage and apoptosis. Molecular interactions between camptothecins and top1 cleavable complexes have been addressed using an alkylating camptothecin derivative and a camptothecin- resistant point mutant top1. Both experimental approaches provide the most direct evidence that camptothecins block top1 by binding at the enzyme/DNA interface. Molecular modeling is consistent with this hypothesis. Two novel families of top1 inhibitors have been discovered in collaboration with outside groups: NSC 314622 by the COMPARE algorithm in the NCI Drug Screen, and aclarubicin by yeast screening. Cleavable complexes induce DNA damage after interference with replication and possibly transcription complexes. The resulting lesions are irreversible DNA breaks and top1 suicide complexes which are potentially recombinogenic. The processing (repair) of such lesions is yet unknown and is a goal of our studies. We are collaborating with Howard Nash (NIMH). In the absence of removal of cleavable complexes, top1 can kill cells (camptothecin-mimetic) or act as a DNA strand transferase and induce DNA recombinations. These effect are relevant to the cytotoxicity of camptothecins or other top1 poisons used in cancer treatment and to the cellular functions of top1.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Intramural Research (Z01)
Project #
1Z01BC006150-17
Application #
6100883
Study Section
Special Emphasis Panel (LMP)
Project Start
Project End
Budget Start
Budget End
Support Year
17
Fiscal Year
1998
Total Cost
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Yu, Le-Mao; Hu, Zhu; Chen, Yu et al. (2018) Synthesis and structure-activity relationship of furoquinolinediones as inhibitors of Tyrosyl-DNA phosphodiesterase 2 (TDP2). Eur J Med Chem 151:777-796
Tang, Sai-Wen; Thomas, Anish; Murai, Junko et al. (2018) Overcoming Resistance to DNA-Targeted Agents by Epigenetic Activation of Schlafen 11 (SLFN11) Expression with Class I Histone Deacetylase Inhibitors. Clin Cancer Res 24:1944-1953
Murai, Junko; Tang, Sai-Wen; Leo, Elisabetta et al. (2018) SLFN11 Blocks Stressed Replication Forks Independently of ATR. Mol Cell 69:371-384.e6
Lee, Hee-Sheung; Carmena, Mar; Liskovykh, Mikhail et al. (2018) Systematic Analysis of Compounds Specifically Targeting Telomeres and Telomerase for Clinical Implications in Cancer Therapy. Cancer Res 78:6282-6296
Kiselev, Evgeny; Dexheimer, Thomas S; Marchand, Christophe et al. (2018) Probing the evolutionary conserved residues Y204, F259, S400 and W590 that shape the catalytic groove of human TDP1 for 3'- and 5'-phosphodiester-DNA bond cleavage. DNA Repair (Amst) 66-67:64-71
Huang, Shar-Yin N; Dalla Rosa, Ilaria; Michaels, Stephanie A et al. (2018) Mitochondrial tyrosyl-DNA phosphodiesterase 2 and its TDP2S short isoform. EMBO Rep 19:
Tsuda, Masataka; Terada, Kazuhiro; Ooka, Masato et al. (2017) The dominant role of proofreading exonuclease activity of replicative polymerase ? in cellular tolerance to cytarabine (Ara-C). Oncotarget 8:33457-33474
Bruno, Peter M; Liu, Yunpeng; Park, Ga Young et al. (2017) A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress. Nat Med 23:461-471
Al Abo, Muthana; Sasanuma, Hiroyuki; Liu, Xiaojun et al. (2017) TDP1 is Critical for the Repair of DNA Breaks Induced by Sapacitabine, a Nucleoside also Targeting ATM- and BRCA-Deficient Tumors. Mol Cancer Ther 16:2543-2551
Thomas, Anish; Tanaka, Mamoru; Trepel, Jane et al. (2017) Temozolomide in the Era of Precision Medicine. Cancer Res 77:823-826

Showing the most recent 10 out of 81 publications