ERBB2 increases metastatic potentials specifically in androgen-insensitive prostate cancer cells

PLoS One. 2014 Jun 17;9(6):e99525. doi: 10.1371/journal.pone.0099525. eCollection 2014.

Abstract

Despite all the blood-based biomarkers used to monitor prostate cancer patients, prostate cancer remains as the second common cause of cancer mortality in men in the United States. This is largely due to a lack of understanding of the molecular pathways that are responsible for the aggressive forms of prostate cancers, the castrate-resistant prostate cancer and the metastatic prostate cancer. Cell signaling pathways activated by the ERBB2 oncogene or the RAS oncogene are frequently found to be altered in metastatic prostate cancers. To evaluate and define the role of the ERBB2/RAS pathway in prostate cancer metastasis, we have evaluated the impact of ERBB2- or RAS-overexpression on the metastatic potentials for four prostate cancer cell lines derived from tumors with different androgen sensitivities. To do so, we transfected the human DU145, LnCaP, and PC3 prostate cancer cells and the murine Myc-CaP prostate cancer cells with the activated form of ERBB2 or H-RAS and assessed their metastatic potentials by three complementary assays, a wound healing assay, a transwell motility assay, and a transwell invasion assay. We showed that while overexpression of ERBB2 increased the metastatic potential of the androgen-insensitive prostate cancer cells (i.e. PC3 and DU145), it did not affect metastatic potentials of the androgen-sensitive prostate cancer cells (i.e. LnCaP and Myc-CaP). In contrast, overexpression of H-RAS only increased the cell motility of Myc-CaP cells, which overexpress the human c-MYC oncogene. Our data suggest that ERBB2 collaborates with androgen signaling to promote prostate cancer metastasis, and that although RAS is one of the critical downstream effectors of ERBB2, it does not phenocopy ERBB2 for its impact on the metastatic potentials of prostate cancer cell lines.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Androgens / physiology
  • Animals
  • Cell Line, Tumor
  • Cell Movement
  • Cell Proliferation
  • Cellular Senescence
  • Gene Expression
  • Humans
  • MAP Kinase Signaling System
  • Male
  • Mice
  • Neoplasm Metastasis
  • Phosphatidylinositol 3-Kinases / metabolism
  • Prostatic Neoplasms, Castration-Resistant / metabolism*
  • Prostatic Neoplasms, Castration-Resistant / pathology
  • Proto-Oncogene Proteins c-akt / metabolism
  • Receptor, ErbB-2 / physiology*
  • ras Proteins / genetics
  • ras Proteins / metabolism

Substances

  • Androgens
  • Phosphatidylinositol 3-Kinases
  • ERBB2 protein, human
  • Receptor, ErbB-2
  • Proto-Oncogene Proteins c-akt
  • ras Proteins

Grants and funding

Start-up funds from Rutgers New Jersey Medical School. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.