The long-range goal is to define the epithelial-stromal interactions responsible for aromatase expression and estrogen production in breast cancer tissue. The overall hypothesis is that upregulated aromatase expression in breast fibroblasts increases the tissue concentration of estradiol (E2), which enhances development and growth of malignant breast epithelial cells. This clinically pertains, since aromatase inhibitors (Als) are the most effective hormonal treatment of breast tumors. A single gene encodes aromatase, the key enzyme in estrogen biosynthesis, the inhibition of which by an Al effectively eliminates E2 production. We showed that 81% of estrogen production in breast cancer tissue was accounted for by the aberrant activation of the alternatively used promoter I.3/II region, which is coordinately regulated by PGE2 and its downstream signaling effectors/transcription factors p38/ATF-2, JNK/c-jun and BRCA1 in breast adipose fibroblasts. Selective inhibition of this promoter region may treat beast cancer while permitting aromatase expression at other body sites and thus obviate the key side effects of the current Als. The mechanisms that regulate the promoter I3/II region will be investigated under the following aims:1A. To identify p38 and JNK-dependent transcriptional regulators that stimulate aromatase expression in breast adipose fibroblasts. PGE2 secreted by malignant cells upregulates the aromatase promoters I.3/II significantly both in vivo in breast tumor tissue and in vitro in cultured breast adipose fibroblasts. We will test the hypothesis that the transcription factors ATF-2 and c-jun activated by p38 and JNK are necessary for PGE2-dependent transcriptional activation of aromatase via the promoter I.3/II region in breast adipose fibroblasts. 1B. To determine in vivo phosphorylation of p38, ATF-2, JNK and c-jun in fibroblasts in breast tumors. 2A. To define the role of BRCA1 in the regulation of aromatase expression in breast adipose fibroblasts. We will test the hypothesis that BRCA1 interacts with the transcriptional complex at promoters I.3/II to inhibit their transactivation, whereas reduced BRCA1 levels fail to repress aromatase expression. 2B. To determine in vivo differential expression of aromatase and other key PGE2/estradiol-related genes in BRCA1 haplo insufficient breast tissues from mutation carriers vs. noncarrier controls. Proposed studies may lead to new pilot trials of treatment and prevention using p38/JNK inhibitors in BRCA1 mutation carriers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA067167-13
Application #
7617649
Study Section
Molecular and Cellular Endocrinology Study Section (MCE)
Program Officer
Sathyamoorthy, Neeraja
Project Start
1996-05-03
Project End
2011-05-31
Budget Start
2009-06-01
Budget End
2010-05-31
Support Year
13
Fiscal Year
2009
Total Cost
$265,930
Indirect Cost
Name
Northwestern University at Chicago
Department
Obstetrics & Gynecology
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611
Moy, I; Todorovi?, V; Dubash, A D et al. (2015) Estrogen-dependent sushi domain containing 3 regulates cytoskeleton organization and migration in breast cancer cells. Oncogene 34:323-33
Zhao, Hong; Pearson, Elizabeth K; Brooks, David C et al. (2012) A humanized pattern of aromatase expression is associated with mammary hyperplasia in mice. Endocrinology 153:2701-13
Chen, Dong; Zhao, Hong; Coon 5th, John S et al. (2012) Weight gain increases human aromatase expression in mammary gland. Mol Cell Endocrinol 355:114-20
Bulun, Serdar E; Chen, Dong; Moy, Irene et al. (2012) Aromatase, breast cancer and obesity: a complex interaction. Trends Endocrinol Metab 23:83-9
Chen, Dong; Reierstad, Scott; Fang, Feng et al. (2011) JunD and JunB integrate prostaglandin E2 activation of breast cancer-associated proximal aromatase promoters. Mol Endocrinol 25:767-75
Demura, Masashi; Demura, Yoshiki; Ameshima, Shingo et al. (2011) Changes in aromatase (CYP19) gene promoter usage in non-small cell lung cancer. Lung Cancer 73:289-93
Lin, Z; Yin, P; Reierstad, S et al. (2010) Adenosine A1 receptor, a target and regulator of estrogen receptoralpha action, mediates the proliferative effects of estradiol in breast cancer. Oncogene 29:1114-22
Zhao, Hong; Innes, Joy; Brooks, David C et al. (2009) A novel promoter controls Cyp19a1 gene expression in mouse adipose tissue. Reprod Biol Endocrinol 7:37
Bulun, S E; Lin, Z; Zhao, H et al. (2009) Regulation of aromatase expression in breast cancer tissue. Ann N Y Acad Sci 1155:121-31
Bulun, Serdar E; Chen, Dong; Lu, Meiling et al. (2007) Aromatase excess in cancers of breast, endometrium and ovary. J Steroid Biochem Mol Biol 106:81-96

Showing the most recent 10 out of 11 publications