Oncotarget

Research Papers:

p85α promotes nucleolin transcription and subsequently enhances EGFR mRNA stability and EGF-induced malignant cellular transformation

Qipeng Xie, Xirui Guo, Jiayan Gu, Liping Zhang, Honglei Jin, Haishan Huang, Jingxia Li and Chuanshu Huang _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:16636-16649. https://doi.org/10.18632/oncotarget.7674

Metrics: PDF 1685 views  |   HTML 2358 views  |   ?  


Abstract

Qipeng Xie1,*, Xirui Guo1,*, Jiayan Gu1, Liping Zhang1, Honglei Jin1,2, Haishan Huang1, Jingxia Li2, Chuanshu Huang1,2

1Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China

2Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA

*These authors contributed equally to this work

Correspondence to:

Chuanshu Huang, e-mail: [email protected]

Keywords: PI3K, p85α, EGFR, nucleolin, cell transformation

Received: December 24, 2015     Accepted: January 16, 2016     Published: February 24, 2016

ABSTRACT

p85α is a regulatory subunit of phosphatidylinositol 3-kinase (PI3K) that is a key lipid enzyme for generating phosphatidylinositol 3, 4, 5-trisphosphate, and subsequently activates signaling that ultimately regulates cell cycle progression, cell growth, cytoskeletal changes, and cell migration. In addition to form a complex with the p110 catalytic subunit, p85α also exists as a monomeric form due to that there is a greater abundance of p85α than p110 in many cell types. Our previous studies have demonstrated that monomeric p85α exerts a pro-apoptotic role in UV response through induction of TNF-α gene expression in PI3K-independent manner. In current studies, we identified a novel biological function of p85α as a positive regulator of epidermal growth factor receptor (EGFR) expression and cell malignant transformation via nucleolin-dependent mechanism. Our results showed that p85α was crucial for EGFR and nucleolin expression and subsequently resulted in an increase of malignant cellular transformation by using both specific knockdown and deletion of p85α in its normal expressed cells. Mechanistic studies revealed that p85α upregulated EGFR protein expression mainly through stabilizing its mRNA, whereas nucleolin (NCL) was able to bind to egfr mRNA and increase its mRNA stability. Consistently, overexpression of NCL in p85α−/− cells restored EGFR mRNA stabilization, protein expression and cell malignant transformation. Moreover, we discovered that p85α upregulated NCL gene transcription via enhancing C-Jun activation. Collectively, our studies demonstrate a novel function of p85α as a positive regulator of EGFR mRNA stability and cell malignant transformation, providing a significant insight into the understanding of biomedical nature of p85α protein in mammalian cells and further supporting that p85α might be a potential target for cancer prevention and therapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 7674