HER2 Signaling Drives DNA Anabolism and Proliferation through SRC-3 Phosphorylation and E2F1-Regulated Genes

Cancer Res. 2016 Mar 15;76(6):1463-75. doi: 10.1158/0008-5472.CAN-15-2383. Epub 2016 Feb 1.

Abstract

Approximately 20% of early-stage breast cancers display amplification or overexpression of the ErbB2/HER2 oncogene, conferring poor prognosis and resistance to endocrine therapy. Targeting HER2(+) tumors with trastuzumab or the receptor tyrosine kinase (RTK) inhibitor lapatinib significantly improves survival, yet tumor resistance and progression of metastatic disease still develop over time. Although the mechanisms of cytosolic HER2 signaling are well studied, nuclear signaling components and gene regulatory networks that bestow therapeutic resistance and limitless proliferative potential are incompletely understood. Here, we use biochemical and bioinformatic approaches to identify effectors and targets of HER2 transcriptional signaling in human breast cancer. Phosphorylation and activity of the Steroid Receptor Coactivator-3 (SRC-3) is reduced upon HER2 inhibition, and recruitment of SRC-3 to regulatory elements of endogenous genes is impaired. Transcripts regulated by HER2 signaling are highly enriched with E2F1 binding sites and define a gene signature associated with proliferative breast tumor subtypes, cell-cycle progression, and DNA replication. We show that HER2 signaling promotes breast cancer cell proliferation through regulation of E2F1-driven DNA metabolism and replication genes together with phosphorylation and activity of the transcriptional coactivator SRC-3. Furthermore, our analyses identified a cyclin-dependent kinase (CDK) signaling node that, when targeted using the CDK4/6 inhibitor palbociclib, defines overlap and divergence of adjuvant pharmacologic targeting. Importantly, lapatinib and palbociclib strictly block de novo synthesis of DNA, mostly through disruption of E2F1 and its target genes. These results have implications for rational discovery of pharmacologic combinations in preclinical models of adjuvant treatment and therapeutic resistance.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Antineoplastic Agents / pharmacology
  • Binding Sites / drug effects
  • Binding Sites / genetics
  • Breast Neoplasms / drug therapy
  • Breast Neoplasms / genetics
  • Cell Cycle / drug effects
  • Cell Cycle / genetics
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Cell Proliferation / genetics*
  • Cyclin-Dependent Kinases / genetics
  • DNA / genetics*
  • DNA Replication / drug effects
  • DNA Replication / genetics
  • Drug Resistance, Neoplasm / drug effects
  • Drug Resistance, Neoplasm / genetics
  • E2F1 Transcription Factor / genetics*
  • Female
  • Humans
  • Nuclear Receptor Coactivator 3 / genetics*
  • Phosphorylation / drug effects
  • Phosphorylation / genetics*
  • Receptor, ErbB-2 / genetics*
  • Signal Transduction / drug effects
  • Signal Transduction / genetics
  • Transcriptional Activation / drug effects
  • Transcriptional Activation / genetics

Substances

  • Antineoplastic Agents
  • E2F1 Transcription Factor
  • E2F1 protein, human
  • DNA
  • NCOA3 protein, human
  • Nuclear Receptor Coactivator 3
  • ERBB2 protein, human
  • Receptor, ErbB-2
  • Cyclin-Dependent Kinases