Abstract

Simple SummaryOverexpressed ErbB2/HER2 receptor drives up to a quarter of breast cancers. One aspect of ErbB2 biology that is poorly understood is how it reaches the cell surface following biosynthesis in the endoplasmic reticulum (ER). Here, the authors show that the CHIP (C-terminus of HSC70-Interacting protein)/STUB1 (STIP1-homologous U-Box containing protein 1) protein targets the newly synthesized ErbB2 for ubiquitin/proteasome-dependent degradation in the ER and Golgi, identifying a novel mechanism that negatively regulates cell surface expression of ErbB2. These findings provide one explanation for frequent loss of CHIP expression is ErbB2-overexpressing breast cancers. The authors further show that ErbB2-overexpressing breast cancer cells with low CHIP expression exhibit higher ER stress inducibility, and ER stress-inducing anticancer drug Bortezomib synergizes with ErbB2-targeted humanized antibody Trastuzumab to inhibit cancer cell proliferation. These new insights suggest that reduced CHIP expression may specify ErbB2-overexpressing breast cancers suitable for combined treatment with Trastuzumab and ER stress inducing agents.Overexpression of the epidermal growth factor receptor (EGFR) family member ErbB2 (HER2) drives oncogenesis in up to 25% of invasive breast cancers. ErbB2 expression at the cell surface is required for oncogenesis but mechanisms that ensure the optimal cell surface display of overexpressed ErbB2 following its biosynthesis in the endoplasmic reticulum are poorly understood. ErbB2 is dependent on continuous association with HSP90 molecular chaperone for its stability and function as an oncogenic driver. Here, we use knockdown and overexpression studies to show that the HSP90/HSC70-interacting negative co-chaperone CHIP (C-terminus of HSC70-Interacting protein)/STUB1 (STIP1-homologous U-Box containing protein 1) targets the newly synthesized, HSP90/HSC70-associated, ErbB2 for ubiquitin/proteasome-dependent degradation in the endoplasmic reticulum and Golgi, thus identifying a novel mechanism that negatively regulates cell surface ErbB2 levels in breast cancer cells, consistent with frequent loss of CHIP expression previously reported in ErbB2-overexpressing breast cancers. ErbB2-overexpressing breast cancer cells with low CHIP expression exhibited higher endoplasmic reticulum stress inducibility. Accordingly, the endoplasmic reticulum stress-inducing anticancer drug Bortezomib combined with ErbB2-targeted humanized antibody Trastuzumab showed synergistic inhibition of ErbB2-overexpressing breast cancer cell proliferation. Our findings reveal new insights into mechanisms that control the surface expression of overexpressed ErbB2 and suggest that reduced CHIP expression may specify ErbB2-overexpressing breast cancers suitable for combined treatment with Trastuzumab and ER stress inducing agents.

Highlights

  • The ErbB family (ErbB1-4) of transmembrane receptor tyrosine kinases (RTKs) plays critical physiological roles [1,2,3]

  • FACS analyses showed that cell surface ErbB2 levels increased in both SKBR3 and 21MT1 CHIP KD cells compared with control cells

  • Display of overexpressed ErbB2 at the cell surface following its biosynthesis in the endoplasmic reticulum is required for oncogenesis, but mechanisms that regulate how this process is optimized in tumor cells are poorly understood

Read more

Summary

Introduction

The ErbB family (ErbB1-4) of transmembrane receptor tyrosine kinases (RTKs) plays critical physiological roles [1,2,3]. ErbB1 (EGFR) and ErbB2 (HER2/Neu) drive oncogenesis in many human malignancies; ErbB2 overexpression, due to gene amplification and/or increased transcription, drives oncogenesis in up to a quarter of human breast cancer patients and specifies poor overall patient survival [4,5]. ErbB2 overexpression has been successfully exploited for therapeutic targeting with humanized monoclonal antibodies (e.g., Trastuzumab, Pertuzumab) and more recently with small molecule kinase inhibitors (e.g., Lapatinib), resulting in significant improvement of the treatment outcomes when added to conventional chemo-radiotherapy [6,7]. Newer insights into ErbB2 biology are required to open avenues to promote more effective and durable responses to targeted therapy of ErbB2-driven breast and other cancers. Increase in the abundance of misfolded proteins triggers an unfolded protein response, one arm of which carries out the dislocation of misfolded/unassembled

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call