Met receptor-induced Grb2 or Shc signals both promote transformation of intestinal epithelial cells, albeit they are required for distinct oncogenic functions

BMC Cancer. 2014 Apr 4:14:240. doi: 10.1186/1471-2407-14-240.

Abstract

Background: Deregulation of receptor tyrosine kinases (RTK) contributes to the initiation and progression of intestinal-derived epithelial cancers, including colorectal cancer (CRC). However, the roles of the proximal signaling molecules engaged by RTKs in different oncogenic functions of CRC remain unclear.

Methods: Herein, the functional impact of expressing variant forms of the oncogenic Met receptor (Tpr-Met) that selectively recruit the adaptor proteins Grb2 or Shc was investigated in a model derived from normal intestinal epithelial cells (IEC-6). An RNA interference (RNAi) approach was used to define the requirement of Grb2 or Shc in Tpr-Met-transformed IEC-6 cells. Since Grb2 and Shc couple RTKs to the activation of the Ras/MEK/Erk and PI3K/Akt pathways, Erk and Akt phosphorylation/activation states were monitored in transformed IEC-6 cells, and a pharmacological approach was employed to provide insights into the roles of these pathways in oncogenic processes evoked by activated Met, and downstream of Grb2 and Shc.

Results: We show, for the first time, that constitutive activation of either Grb2 or Shc signals in IEC-6 cells, promotes morphological transformation associated with down-regulation of E-cadherin, as well as increased cell growth, loss of growth contact inhibition, anchorage-independent growth, and resistance to serum deprivation and anoikis. Oncogenic activation of Met was revealed to induce morphological transformation, E-cadherin down-regulation, and protection against anoikis by mechanisms dependent on Grb2, while Shc was shown to be partly required for enhanced cell growth. The coupling of activated Met to the Ras/MEK/Erk and PI3K/Akt pathways, and the sustained engagement of Grb2 or Shc in IECs, was shown to trigger negative feedback, limiting the extent of activation of these pathways. Nonetheless, morphological alterations and E-cadherin down-regulation induced by the oncogenic Tpr-Met, and by Grb2 or Shc signals, were blocked by MEK, but not PI3K, inhibitors while the enhanced growth and resistance to anoikis induced by Tpr-Met were nearly abolished by co-treatment with both inhibitors.

Conclusion: Overall, these results identify Grb2 and Shc as central signaling effectors of Met-driven progression of intestinal epithelial-derived cancers. Notably, they suggest that Grb2 may represent a promising target for the design of novel CRC therapies.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Cadherins / metabolism
  • Cell Line
  • Cell Transformation, Neoplastic / genetics
  • Colorectal Neoplasms / genetics*
  • Colorectal Neoplasms / pathology
  • Colorectal Neoplasms / therapy
  • Epithelial Cells / metabolism
  • GRB2 Adaptor Protein / biosynthesis*
  • GRB2 Adaptor Protein / metabolism
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Intestinal Mucosa / metabolism
  • Intestines / pathology
  • Proto-Oncogene Proteins c-met / genetics
  • Proto-Oncogene Proteins c-met / metabolism*
  • Shc Signaling Adaptor Proteins / biosynthesis*
  • Shc Signaling Adaptor Proteins / metabolism
  • Signal Transduction / genetics

Substances

  • Cadherins
  • GRB2 Adaptor Protein
  • GRB2 protein, human
  • Shc Signaling Adaptor Proteins
  • MET protein, human
  • Proto-Oncogene Proteins c-met