Oncotarget

Research Papers:

EPRS is a critical regulator of cell proliferation and estrogen signaling in ER+ breast cancer

Igor Katsyv, Minghui Wang, Won Min Song, Xianxiao Zhou, Yongzhong Zhao, Sun Park, Jun Zhu, Bin Zhang _ and Hanna Y. Irie

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:69592-69605. https://doi.org/10.18632/oncotarget.11870

Metrics: PDF 2240 views  |   HTML 2920 views  |   ?  


Abstract

Igor Katsyv1,2,3, Minghui Wang2,3, Won Min Song2,3, Xianxiao Zhou2,3, Yongzhong Zhao2,3, Sun Park4, Jun Zhu2,3, Bin Zhang2,3,5, Hanna Y. Irie4,5

1Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

2Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

3Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

4Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

5Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

Correspondence to:

Bin Zhang, email: [email protected]

Hanna Y. Irie, email: [email protected]

Keywords: breast cancer, ER+, EPRS, gene networks

Received: May 04, 2016     Accepted: August 25, 2016     Published: September 06, 2016

ABSTRACT

Aminoacyl tRNA synthetases (ARSs) are a class of enzymes with well-conserved housekeeping functions in cellular translation. Recent evidence suggests that ARS genes may participate in a wide array of cellular processes, and may contribute to the pathology of autoimmune disease, cancer, and other diseases. Several studies have suggested a role for the glutamyl prolyl tRNA synthetase (EPRS) in breast cancers, although none has identified any underlying mechanism about how EPRS contributes to carcinogenesis. In this study, we identified EPRS as upregulated in estrogen receptor positive (ER+) human breast tumors in the TCGA and METABRIC cohorts, with copy number gains in nearly 50% of samples in both datasets. EPRS expression is associated with reduced overall survival in patients with ER+ tumors in TCGA and METABRIC datasets. EPRS expression was also associated with reduced distant relapse-free survival in patients treated with adjuvant tamoxifen monotherapy for five years, and EPRS-correlated genes were highly enriched for genes predictive of a poor response to tamoxifen. We demonstrated the necessity of EPRS for proliferation of tamoxifen-resistant ER+ breast cancer, but not ER- breast cancer cells. Transcriptomic profiling showed that EPRS regulated cell cycle and estrogen response genes. Finally, we constructed a causal gene network based on over 2500 ER+ breast tumor samples to build up an EPRS-estrogen signaling pathway. EPRS and its regulated estrogenic gene network may offer a promising alternative approach to target ER+ breast cancers that are refractory to current anti-estrogens.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11870