American Association for Cancer Research
Browse
00085472can152383-sup-154577_1_supp_3221814_nxm0j5.xlsx (294.27 kB)

Supplementary Data 2 from HER2 Signaling Drives DNA Anabolism and Proliferation through SRC-3 Phosphorylation and E2F1-Regulated Genes

Download (294.27 kB)
dataset
posted on 2023-03-30, 23:40 authored by Bryan C. Nikolai, Rainer B. Lanz, Brian York, Subhamoy Dasgupta, Nicholas Mitsiades, Chad J. Creighton, Anna Tsimelzon, Susan G. Hilsenbeck, David M. Lonard, Carolyn L. Smith, Bert W. O'Malley

Spreadsheet containing Gene ID and ontology analysis for all transcripts downregulated with HER2 depletion.

Funding

NIH/NCI

NIH

Endocrine Society and Welch Foundation

Cancer Prevention and Research Institute of Texas

History

ARTICLE 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. Cancer Res; 76(6); 1463–75. ©2016 AACR.