We generated a novel model of mice heterozygous for a targeted null mutation of Tgfbr1. When crossed with mice carrying a mutation in the Apc tumor suppressor gene, these mice develop twice as many intestinal tumors as wild-type littermates. Invasive adenocarcinoma with features of human colon cancer is only identified among ApcMin/+;Tgfbr1+/- mice, not among ApcMin/+;Tgfbr1+/+ mice and tumors do not exhibit loss of heterozygosity at the Tgfbr1 locus. TGF-?-mediated growth inhibition, phosphorylation of Smad2 and Smad3, and overall TGF-? signaling are mildly decreased in haploinsufficient embryonic fibroblasts. Decreased Smad2 and Smad3 phosphorylation is observed in the colonic epithelium crypts of ApcMin/+;Tgfbr1+/- mice. Decreased Smad signaling is associated with increased cell proliferation in the crypts of the intestinal mucosa and intestinal tumors. Thus, constitutively reduced Tgfbr1-mediated signaling is a potent modifier of colorectal cancer development. To determine the relevance of these findings in humans, we analyzed germline peripheral blood for TGFBR1 expression. We found that 29 of 242 (12.0%) patients with colorectal cancer but only three of 195 (1.5%) controls had evidence of germline allele-specific expression of TGFBR1 (TGFBR1 ASE). These results indicate that TGFR1 ASE is one of the most commonly inherited cause of colorectal cancer, which increases cancer risk by approximately 870%. Differences in TGF-?-mediated phosphorylation of SMAD2 and SMAD3 between human lymphoid cells from individuals with wild-type TGFBR1 and individuals with TGFBR1 ASE were comparable to those found between Tgbr1+/+ and Tgfbr1+/- mouse embryonic fibroblasts. It is the purpose of the studies outlined in this application to unravel the molecular mechanisms of Tgfbr1 haploinsufficiency in colorectal cancer through execution of three Specific Aims: First: To characterize the phenotype of Tgfbr1+/+ and Tgfbr1+/- mice with respect to colon cancer: The phenotype of Tgfbr1+/+ and Tgfbr1+/- mice will be evaluated in three different backgrounds (129Sv/J, C57BL/6 and FVB/N). Colon tumors will be induced using the azoxymethane protocol and by crossing the transgenic strains with Apc1638N/+ and KRASV12G;Apc1638N/+ mice. Second: To determine the role of Tgfbr1 haploinsufficiency in development of cancer stem cells and intestinal stem cells as well as the contribution of stromal and lymphoepithelial Tgfbr1 signaling to tumor formation: We will determine the growth of Tgfbr1+/+ and Tgfbr1+/- tumors in nude mice to evaluate the contribution of Tgfbr1 haploinsufficiency on tumor stem cell development. Using the Lgr5 mouse model, we will also assess whether Tgfbr1 haploinsufficiency enhances the development of intestinal stem cells. The role of Tgfbr1 haploinsufficiency within stromal and lymphoepithelial cells will be evaluated as it relates to colon cancer development. Third: To assess Tgfbr1 haploinsufficiency oncogenic properties: Using MEFs from three distinct genetic backgrounds (129Sv/J, C57BL/6 and FVB/N) we will determine the extent to which Tgfbr1 haploinsufficiency enhances oncogenesis.

Public Health Relevance

We have shown that decreased expression of the type I TGF-? receptor gene is emerging as the most common cause of colorectal cancer reported to date. The primary goal of this project is to understand the mechanisms linking decreased gene expression and colorectal cancer development. Experiments will be conducted with mice that only express one copy of the gene, which will be bred together with mice that spontaneously develop colon cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA137000-05
Application #
8597530
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Mietz, Judy
Project Start
2010-01-01
Project End
2014-12-31
Budget Start
2014-01-01
Budget End
2014-12-31
Support Year
5
Fiscal Year
2014
Total Cost
$276,589
Indirect Cost
$86,348
Name
University of Alabama Birmingham
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Principe, Daniel R; Doll, Jennifer A; Bauer, Jessica et al. (2014) TGF-?: duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst 106:djt369
Pasche, Boris; Pennison, Michael J; Jimenez, Hugo et al. (2014) TGFBR1 and cancer susceptibility. Trans Am Clin Climatol Assoc 125:300-12
Borate, Uma; Absher, Devin; Erba, Harry P et al. (2012) Potential of whole-genome sequencing for determining risk and personalizing therapy: focus on AML. Expert Rev Anticancer Ther 12:1289-97
Pasche, Boris; Absher, Devin (2011) Whole-genome sequencing: a step closer to personalized medicine. JAMA 305:1596-7
Kaklamani, Virginia; Yi, Nengjun; Zhang, Kui et al. (2011) Polymorphisms of ADIPOQ and ADIPOR1 and prostate cancer risk. Metabolism 60:1234-43
Moore-Smith, Lakisha; Pasche, Boris (2011) TGFBR1 signaling and breast cancer. J Mammary Gland Biol Neoplasia 16:89-95
Yi, Nengjun; Kaklamani, Virginia G; Pasche, Boris (2011) Bayesian analysis of genetic interactions in case-control studies, with application to adiponectin genes and colorectal cancer risk. Ann Hum Genet 75:90-104
Pasche, Boris; Yi, Nengjun (2010) Candidate gene association studies: successes and failures. Curr Opin Genet Dev 20:257-61
Pasche, Boris (2010) Cancer genetics. Introduction. Cancer Treat Res 155:xi-xii
Pasche, Boris; Wisinski, Kari B; Sadim, Maureen et al. (2010) Constitutively decreased TGFBR1 allelic expression is a common finding in colorectal cancer and is associated with three TGFBR1 SNPs. J Exp Clin Cancer Res 29:57