HNF1B variants associate with promoter methylation and regulate gene networks activated in prostate and ovarian cancer

Oncotarget. 2016 Nov 15;7(46):74734-74746. doi: 10.18632/oncotarget.12543.

Abstract

Two independent regions within HNF1B are consistently identified in prostate and ovarian cancer genome-wide association studies (GWAS); their functional roles are unclear. We link prostate cancer (PC) risk SNPs rs11649743 and rs3760511 with elevated HNF1B gene expression and allele-specific epigenetic silencing, and outline a mechanism by which common risk variants could effect functional changes that increase disease risk: functional assays suggest that HNF1B is a pro-differentiation factor that suppresses epithelial-to-mesenchymal transition (EMT) in unmethylated, healthy tissues. This tumor-suppressor activity is lost when HNF1B is silenced by promoter methylation in the progression to PC. Epigenetic inactivation of HNF1B in ovarian cancer also associates with known risk SNPs, with a similar impact on EMT. This represents one of the first comprehensive studies into the pleiotropic role of a GWAS-associated transcription factor across distinct cancer types, and is the first to describe a conserved role for a multi-cancer genetic risk factor.

Keywords: HNF1B; cancer; eQTL; ovarian; prostate.

MeSH terms

  • Alleles
  • Cell Line, Tumor
  • Cell Movement
  • Cell Proliferation
  • DNA Methylation*
  • Epithelial-Mesenchymal Transition
  • Female
  • Gene Expression Regulation, Neoplastic*
  • Gene Regulatory Networks*
  • Genetic Predisposition to Disease
  • Genetic Variation
  • Genome-Wide Association Study
  • Genotype
  • Hepatocyte Nuclear Factor 1-beta / genetics*
  • Humans
  • Linkage Disequilibrium
  • Male
  • Ovarian Neoplasms / genetics*
  • Ovarian Neoplasms / pathology
  • Polymorphism, Single Nucleotide
  • Promoter Regions, Genetic*
  • Prostatic Neoplasms / genetics*
  • Prostatic Neoplasms / pathology
  • Risk

Substances

  • HNF1B protein, human
  • Hepatocyte Nuclear Factor 1-beta