Abstract

The commonality between most phospho-tyrosine signaling networks is their shared use of adaptor proteins to transduce mitogenic signals. ShcA (SHC1) is one such adaptor protein that employs two phospho-tyrosine binding domains (PTB and SH2) and key phospho-tyrosine residues to promote mammary tumorigenesis. Receptor tyrosine kinases (RTK), such as ErbB2, bind the ShcA PTB domain to promote breast tumorigenesis by engaging Grb2 downstream of the ShcA tyrosine phosphorylation sites to activate AKT/mTOR signaling. However, breast tumors also rely on the ShcA PTB domain to bind numerous negative regulators that limit activation of secondary mitogenic signaling networks. This study examines the role of PTB-independent ShcA pools in controlling breast tumor growth and resistance to tyrosine kinase inhibitors. We demonstrate that PTB-independent ShcA complexes predominately rely on the ShcA SH2 domain to activate multiple Src family kinases (SFK), including Src and Fyn, in ErbB2-positive breast cancers. Using genetic and pharmacologic approaches, we show that PTB-independent ShcA complexes augment mammary tumorigenesis by increasing the activity of the Src and Fyn tyrosine kinases in an SH2-dependent manner. This bifurcation of signaling complexes from distinct ShcA pools transduces non-redundant signals that integrate the AKT/mTOR and SFK pathways to cooperatively increase breast tumor growth and resistance to tyrosine kinase inhibitors, including lapatinib and PP2. This study mechanistically dissects how the interplay between diverse intracellular ShcA complexes impacts the tyrosine kinome to affect breast tumorigenesis.Implications: The ShcA adaptor, within distinct signaling complexes, impacts tyrosine kinase signaling, breast tumor growth, and resistance to tyrosine kinase inhibitors. Mol Cancer Res; 16(5); 894-908. ©2018 AACR.

Highlights

  • Aberrant phospho-tyrosine signaling networks are essential drivers of breast cancer progression [1]

  • NMuMG-NeuNT cell lines were stably transfected with a pMSCV/ Hygromycin expression vector (Clontech catalog no. 634401) subcloned with cDNAs expressing: (i) wild-type ShcA (ShcAWT), (ii) a ShcA mutant harboring an arginine to glutamine substitution at amino acid 175 in the phospho-tyrosine binding pocket of the PTB domain of ShcA (PTBMUT), (iii) a PTB mutant (PTBMUT) harboring tyrosine to phenylalanine substitution at amino acids 239, 240, and 313 (PTBMUT/3F), or (iv) a PTBMUT with an arginine to lysine substitution at amino acid 397 in the phospho-tyrosine binding pocket of the SH2 domain (PTB/SH2MUT)

  • NIC/SrcÀ/À cell lines were established from mammary tumor virus (MMTV)/Neu-internal ribosome entry site (IRES)-Cre (NIC) mammary tumors, in which both c-Src alleles have been deleted by Cre-mediated excision [25,26,27]

Read more

Summary

Introduction

Aberrant phospho-tyrosine signaling networks are essential drivers of breast cancer progression [1]. Numerous tyrosine kinases, including Met, EGFR, and Lyn [4,5,6], contribute to the emergence of basal-like breast cancer that has confounded the identification of effective targeted therapies against this poor outcome subtype. In this regard, HER2þ breast cancer cells reprogram their tyrosine kinome through the aberrant activation of numerous receptor and nonreceptor tyrosine kinases, including but not limited to ErbB3, Met, and Src, to confer trastuzumab resistance [7,8,9,10]. Tyrosine kinases serve an essential role in establishing breast cancer heterogeneity and therapeutic sensitivity

Methods
Results
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