The long-term objective of my research is to understand the influence of hematopoietic-specific developmental programs on the repair DNA damage such as double strand breaks (DSBs) and the initial molecular events that lead to translocations, which are a hallmark of leukemia, lymphoma, and soft-tissue sarcomas. DSBs are highly recombinogenic, increasing the exchange of information between two homologous DNA duplexes by several orders of magnitude;thus, mammalian cells are potentially at risk for rearrangements arising during DSB repair. Chromosomal DSBs result following exposure to irradiation, alkylating agents, and topoisomerase II (topoII) inhibitors that are common therapies in the treatment of human cancers. Treatment regimens that include the topoII inhibitor etoposide are associated with one class of therapy-related acute myeloid leukemia (t-AML) and chromosomal translocations involving the mixed lineage leukemia (MLL) gene on chromosome band 11q23. Similarity of 11q23 MLL breakpoints in t-AML and infant leukemias suggests an association between de novo infant leukemia and in utero exposure to topoII inhibitors. The list of potential topo II inhibitors is extensive, and it remains unclear which of these compounds have a direct potential to induce the chromosomal translocations observed in the clinical setting. Using a unique genetic system to determine the potential for repair of DSBs within the breakpoint cluster regions of the 11q23 MLL gene and common partner genes to result in chromosomal translocations, this proposal will (1) determine the potential for exposure to a range of topoII inhibitors to initiate chromosomal rearrangements within the breakpoint cluster region of the MLL and AF9 genes similar to those observed in the clinical setting;and (2) create a targeted mouse model to determine in vivo the potential for exposure to topoII inhibitors to initiate chromosomal rearrangements within the breakpoint cluster region of the MLL and AF9 genes as measured by the presence of MLL-AF9 genome rearrangements in bone marrow and peripheral blood. These approaches in both ex vivo cell culture and in vivo mouse models will provide significant insight into the initiation of potentially oncogenic chromosomal rearrangements and leukemogenesis. Unraveling the etiology and consequences of translocations may lead to new approaches to therapy and prevention.

Public Health Relevance

Exposure to the topoisomerase II (topoII) inhibitors is associated with chromosomal translocations involving the mixed lineage leukemia (MLL) gene on chromosome band 11q23, one class of therapy-related acute myeloid leukemia (t-AML), and possibly de novo infant leukemias following in utero exposure to topoII inhibitors. The list of potential topo II inhibitors is extensive, and it remains unclear which of these compounds have a direct potential to induce the chromosomal translocations observed in the clinical setting. Our approaches in both ex vivo cell culture and in vivo mouse models will provide significant insight into the initiation of these oncogenic chromosomal translocations and leukemogenesis and may lead to new approaches to therapy and prevention.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
4R01CA100159-07
Application #
8193254
Study Section
Cancer Genetics Study Section (CG)
Program Officer
Pelroy, Richard
Project Start
2003-05-19
Project End
2013-06-30
Budget Start
2011-07-01
Budget End
2012-06-30
Support Year
7
Fiscal Year
2011
Total Cost
$247,644
Indirect Cost
Name
University of North Carolina Charlotte
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
066300096
City
Charlotte
State
NC
Country
United States
Zip Code
28223
Bariar, Bhawana; Vestal, C Greer; Deem, Bradley et al. (2018) Bioflavonoids promote stable translocations between MLL-AF9 breakpoint cluster regions independent of normal chromosomal context: Model system to screen environmental risks. Environ Mol Mutagen :
Cupello, Steven; Richardson, Christine; Yan, Shan (2016) Cell-free Xenopus egg extracts for studying DNA damage response pathways. Int J Dev Biol 60:229-236
Richardson, Christine; Yan, Shan; Vestal, C Greer (2015) Oxidative stress, bone marrow failure, and genome instability in hematopoietic stem cells. Int J Mol Sci 16:2366-85
Bariar, Bhawana; Vestal, C Greer; Richardson, Christine (2013) Long-term effects of chromatin remodeling and DNA damage in stem cells induced by environmental and dietary agents. J Environ Pathol Toxicol Oncol 32:307-27
White, Ryan R; Sung, Patricia; Vestal, C Greer et al. (2013) Double-strand break repair by interchromosomal recombination: an in vivo repair mechanism utilized by multiple somatic tissues in mammals. PLoS One 8:e84379
Mouzannar, R; McCafferty, J; Benedetto, G et al. (2011) TRANSCRIPTIONAL AND PHOSPHO-PROTEOMIC SCREENS REVEAL STEM CELL ACTIVATION OF INSULIN-RESISTANCE AND TRANSFORMATION PATHWAYS FOLLOWING A SINGLE MINIMALLY TOXIC EPISODE OF ROS. Int J Genomics Proteomics 2:34-49
Pandita, Tej K; Richardson, Christine (2009) Chromatin remodeling finds its place in the DNA double-strand break response. Nucleic Acids Res 37:1363-77
Libura, Jolanta; Ward, Maureen; Solecka, Joanna et al. (2008) Etoposide-initiated MLL rearrangements detected at high frequency in human primitive hematopoietic stem cells with in vitro and in vivo long-term repopulating potential. Eur J Haematol 81:185-95
Koptyra, M; Cramer, K; Slupianek, A et al. (2008) BCR/ABL promotes accumulation of chromosomal aberrations induced by oxidative and genotoxic stress. Leukemia 22:1969-72
Francis, Richard; Richardson, Christine (2007) Multipotent hematopoietic cells susceptible to alternative double-strand break repair pathways that promote genome rearrangements. Genes Dev 21:1064-74

Showing the most recent 10 out of 12 publications