Breast cancer risk is strongly determined by numerous genes, only a few of which have been identified. The female ACI rat exhibits a unique, genetically-conferred, susceptibility of to 172-estradiol (E2)-induced mammary cancer. Because estrogens have been inextricably implicated in the etiology of breast cancer, the ACI rat is rapidly gaining wide acceptance within the breast cancer research community as a physiologically relevant model. To define the genetic bases of susceptibility to E2-induced mammary cancer, we performed intercrosses between the susceptible ACI strain and the resistant Copenhagen (COP) or Brown Norway (BN) strains and revealed 9 quantitative trait loci (QTL) that significantly impact susceptibility to mammary cancer. Two of these QTL, Emca1 and Emca8, were mapped to an overlapping region of rat chromosome 5 (RNO5), and a third, Emca4, was mapped to RNO7. Data summarized herein strongly suggest that the regions of the human genome that are orthologous to these three Emca loci harbor as yet unidentified breast cancer susceptibility genes. The objectives of this research are to identify genes residing within Emca1, Emca4 and Emca8 that determine breast cancer risk and to define better the molecular mechanisms through which estrogens contribute to breast cancer development.
Aim 1 is to identify genes residing within Emca8 that determine susceptibility to E2-induced mammary cancer. A substitution mapping approach employing congenic rat strains will be used to fine map Emca8. Development of each congenic strain will be directed by preliminary data that are indicative of genetic heterogeneity across Emca8. Expression of genes residing within a minimal effective congenic interval will be evaluated at the mRNA and protein levels. Nucleotide sequence across the minimal congenic interval will be determined for the ACI and BN strains to identify the genetic variant responsible for the observed difference in susceptibility.
Aim 2 is to determine whether Emca1 and Emca8 harbor the same determinants of susceptibility to E2-induced mammary cancer. Genes identified in Aim 1 that mediate the actions of Emca8 on mammary cancer susceptibility will be evaluated in the COP strain to determine if the COP and BN rat strains share alleles for these genes.
Aim 3 is to identify the genetic variant residing within Emca4 that determines susceptibility to E2-induced mammary cancer.
This aim will be focused on the region of Emca4 that is orthologous to a recently mapped, but as yet unidentified, genetic determinant of breast cancer risk in humans. These proposed studies promise to greatly enhance our knowledge regarding how breast cancer risk is genetically determined, as well as to the mechanisms through which estrogens contribute to breast cancer development. Relevance to public health: This research utilizes a novel and physiologically relevant rat mammary cancer model to identify genes that determine mammary cancer susceptibility. Because of the numerous similarities between this model and breast cancer in humans, it is believe that the genes identified in this study will similarly impact breast cancer risk in humans. These studies will also reveal insight into the mechanisms through which estrogens contribute to breast cancer development.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA077876-16
Application #
8265709
Study Section
Cancer Genetics Study Section (CG)
Program Officer
Sharman, Anu
Project Start
1998-04-01
Project End
2014-05-31
Budget Start
2012-06-01
Budget End
2014-05-31
Support Year
16
Fiscal Year
2012
Total Cost
$339,939
Indirect Cost
$111,024
Name
University of Wisconsin Madison
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
161202122
City
Madison
State
WI
Country
United States
Zip Code
53715
Jerry, D Joseph; Shull, James D; Hadsell, Darryl L et al. (2018) Genetic variation in sensitivity to estrogens and breast cancer risk. Mamm Genome 29:24-37
Shull, James D; Dennison, Kirsten L; Chack, Aaron C et al. (2018) Rat models of 17?-estradiol-induced mammary cancer reveal novel insights into breast cancer etiology and prevention. Physiol Genomics 50:215-234
Dennison, Kirsten L; Chack, Aaron C; Hickman, Maureen Peters et al. (2018) Ept7, a quantitative trait locus that controls estrogen-induced pituitary lactotroph hyperplasia in rat, is orthologous to a locus in humans that has been associated with numerous cancer types and common diseases. PLoS One 13:e0204727
Das Gupta, Soumyasri; Sae-tan, Sudathip; Wahler, Joseph et al. (2015) Dietary ?-Tocopherol-Rich Mixture Inhibits Estrogen-Induced Mammary Tumorigenesis by Modulating Estrogen Metabolism, Antioxidant Response, and PPAR?. Cancer Prev Res (Phila) 8:807-16
Samanas, Nyssa Becker; Commers, Tessa W; Dennison, Kirsten L et al. (2015) Genetic etiology of renal agenesis: fine mapping of Renag1 and identification of Kit as the candidate functional gene. PLoS One 10:e0118147
Dennison, Kirsten L; Samanas, Nyssa Becker; Harenda, Quincy Eckert et al. (2015) Development and characterization of a novel rat model of estrogen-induced mammary cancer. Endocr Relat Cancer 22:239-48
Flister, Michael J; Endres, Bradley T; Rudemiller, Nathan et al. (2014) CXM: a new tool for mapping breast cancer risk in the tumor microenvironment. Cancer Res 74:6419-29
Kurz, Scott G; Dennison, Kirsten L; Samanas, Nyssa Becker et al. (2014) Ept7 influences estrogen action in the pituitary gland and body weight of rats. Mamm Genome 25:244-52
Colletti 2nd, John A; Leland-Wavrin, Kristin M; Kurz, Scott G et al. (2014) Validation of six genetic determinants of susceptibility to estrogen-induced mammary cancer in the rat and assessment of their relevance to breast cancer risk in humans. G3 (Bethesda) 4:1385-94
van Heesch, Sebastiaan; Mokry, Michal; Boskova, Veronika et al. (2013) Systematic biases in DNA copy number originate from isolation procedures. Genome Biol 14:R33

Showing the most recent 10 out of 32 publications