Blockage of conformational changes of heat shock protein gp96 on cell membrane by a α-helix peptide inhibits HER2 dimerization and signaling in breast cancer

PLoS One. 2015 Apr 21;10(4):e0124647. doi: 10.1371/journal.pone.0124647. eCollection 2015.

Abstract

Cell membrane translocation of heat shock protein gp96 from the endoplasmic reticulum has been observed in multiple tumors and is associated with tumor malignancy. However, the cancer-intrinsic function and the related mechanism of cell membrane gp96 as a pro-oncogenic chaperone remain further elucidated. In this study, we found that inhibition of gp96 intramolecular conformational changes by a single α-helix peptide p37 dramatically increased its binding to HER2, whereas decreased HER2 dimerization, phosphorylation and downstream signaling. Targeting cell membrane gp96 promoted HER2 ubiquitination and subsequent lysosomal degradation, which led to decreased cell growth and increased apoptosis, and inhibited tumor growth in vivo. We also demonstrate that gp96 inhibitory peptide p37 synergized with trastuzumab to suppress cell growth and induce apoptosis. Our work demonstrates that blocking gp96 conformational changes directs HER2 for cellular degradation, and represents a new therapeutic strategy for inhibiting HER2 signaling in cancer.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Amino Acid Sequence
  • Animals
  • Antigens, Neoplasm / genetics*
  • Antigens, Neoplasm / metabolism
  • Antineoplastic Agents / chemical synthesis
  • Antineoplastic Agents / pharmacology*
  • Apoptosis / drug effects
  • Breast Neoplasms / drug therapy*
  • Breast Neoplasms / genetics
  • Breast Neoplasms / metabolism
  • Breast Neoplasms / pathology
  • Cell Line, Tumor
  • Endoplasmic Reticulum / drug effects
  • Endoplasmic Reticulum / metabolism
  • Female
  • Gene Expression Regulation, Neoplastic*
  • Humans
  • Mice
  • Molecular Sequence Data
  • Peptides / chemical synthesis
  • Peptides / pharmacology*
  • Phosphorylation / drug effects
  • Protein Binding / drug effects
  • Protein Conformation / drug effects
  • Protein Multimerization / drug effects
  • Protein Transport / drug effects
  • Proteolysis / drug effects
  • Receptor, ErbB-2 / antagonists & inhibitors*
  • Receptor, ErbB-2 / genetics
  • Receptor, ErbB-2 / metabolism
  • Signal Transduction
  • Trastuzumab / pharmacology
  • Ubiquitination / drug effects
  • Xenograft Model Antitumor Assays

Substances

  • Antigens, Neoplasm
  • Antineoplastic Agents
  • Peptides
  • sarcoma glycoprotein gp96 rejection antigens
  • ERBB2 protein, human
  • Receptor, ErbB-2
  • Trastuzumab

Grants and funding

This work was supported by a grant from Major State Basic Research Development Program of China (973 Program, No.2014CB542602), grants from the National Natural Science Foundation of China (31230026, 81321063, 81102018, 81471960), and grants from Key Projects in the National Science & Technology Program (2013ZX10002001-003-003). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.