Integrative bioinformatics links HNF1B with clear cell carcinoma and tumor-associated thrombosis

PLoS One. 2013 Sep 9;8(9):e74562. doi: 10.1371/journal.pone.0074562. eCollection 2013.

Abstract

Clear cell carcinoma (CCC) is a histologically distinct carcinoma subtype that arises in several organ systems and is marked by cytoplasmic clearing, attributed to abundant intracellular glycogen. Previously, transcription factor hepatocyte nuclear factor 1-beta (HNF1B) was identified as a biomarker of ovarian CCC. Here, we set out to explore more broadly the relation between HNF1B and carcinomas with clear cell histology. HNF1B expression, evaluated by immunohistochemistry, was significantly associated with clear cell histology across diverse gynecologic and renal carcinomas (P<0.001), as was hypomethylation of the HNF1B promoter (P<0.001). From microarray analysis, an empirically-derived HNF1B signature was significantly enriched for computationally-predicted targets (with HNF1 binding sites) (P<0.03), as well as genes associated with glycogen metabolism, including glucose-6-phophatase, and strikingly the blood clotting cascade, including fibrinogen, prothrombin and factor XIII. Enrichment of the clotting cascade was also evident in microarray data from ovarian CCC versus other histotypes (P<0.01), and HNF1B-associated prothrombin expression was verified by immunohistochemistry (P = 0.015). Finally, among gynecologic carcinomas with cytoplasmic clearing, HNF1B immunostaining was linked to a 3.0-fold increased risk of clinically-significant venous thrombosis (P = 0.043), and with a 2.3-fold increased risk (P = 0.011) in a combined gynecologic and renal carcinoma cohort. Our results define HNF1B as a broad marker of clear cell phenotype, and support a mechanistic link to glycogen accumulation and thrombosis, possibly reflecting (for gynecologic CCC) derivation from secretory endometrium. Our findings also implicate a novel mechanism of tumor-associated thrombosis (a major cause of cancer mortality), based on the direct production of clotting factors by cancer cells.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenocarcinoma, Clear Cell / complications
  • Adenocarcinoma, Clear Cell / genetics*
  • Adenocarcinoma, Clear Cell / metabolism
  • Adenocarcinoma, Clear Cell / pathology
  • Blood Coagulation
  • Blood Coagulation Factors / genetics
  • Blood Coagulation Factors / metabolism
  • Carcinoma, Renal Cell / complications
  • Carcinoma, Renal Cell / genetics*
  • Carcinoma, Renal Cell / metabolism
  • Carcinoma, Renal Cell / pathology
  • Computational Biology
  • DNA Methylation
  • Endometrial Neoplasms / complications
  • Endometrial Neoplasms / genetics*
  • Endometrial Neoplasms / metabolism
  • Endometrial Neoplasms / pathology
  • Female
  • Gene Expression Regulation, Neoplastic*
  • Glucose-6-Phosphatase / genetics
  • Glucose-6-Phosphatase / metabolism
  • Glycogen / metabolism
  • Hepatocyte Nuclear Factor 1-beta / genetics*
  • Hepatocyte Nuclear Factor 1-beta / metabolism
  • Humans
  • Immunohistochemistry
  • Kidney Neoplasms / complications
  • Kidney Neoplasms / genetics*
  • Kidney Neoplasms / metabolism
  • Kidney Neoplasms / pathology
  • Oligonucleotide Array Sequence Analysis
  • Ovarian Neoplasms / complications
  • Ovarian Neoplasms / genetics*
  • Ovarian Neoplasms / metabolism
  • Ovarian Neoplasms / pathology
  • Risk
  • Venous Thrombosis / complications
  • Venous Thrombosis / genetics*
  • Venous Thrombosis / metabolism
  • Venous Thrombosis / pathology

Substances

  • Blood Coagulation Factors
  • HNF1B protein, human
  • Hepatocyte Nuclear Factor 1-beta
  • Glycogen
  • Glucose-6-Phosphatase

Grants and funding

This study was supported with funds from the Stanford University Department of Pathology. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.