Oncotarget

Research Papers:

CXCR2 is a negative regulator of p21 in p53-dependent and independent manner via Akt-mediated Mdm2 in ovarian cancer

Rosa Mistica C. Ignacio, Yuan-Lin Dong, Syeda M. Kabir, Hyeongjwa Choi, Eun-Sook Lee, Andrew J. Wilson, Alicia Beeghly-Fadiel, Margaret M. Whalen and Deok-Soo Son _

PDF  |  HTML  |  How to cite

Oncotarget. 2018; 9:9751-9765. https://doi.org/10.18632/oncotarget.24231

Metrics: PDF 1952 views  |   HTML 2257 views  |   ?  


Abstract

Rosa Mistica C. Ignacio1, Yuan-Lin Dong1, Syeda M. Kabir1, Hyeongjwa Choi1, Eun-Sook Lee2, Andrew J. Wilson3,4, Alicia Beeghly-Fadiel4,5, Margaret M. Whalen6 and Deok-Soo Son1

1Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA

2Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA

3Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, USA

4Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA

5Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA

6Department of Chemistry, Tennessee State University, Nashville, TN 37209, USA

Correspondence to:

Deok-Soo Son, email: [email protected]

Keywords: Akt; CXCR2; Mdm2; ovarian cancer; p53

Received: October 30, 2017     Accepted: January 09, 2018     Published: January 15, 2018

ABSTRACT

Ovarian cancer (OC) has the highest rate of mortality among gynecological malignancy. Chemokine receptor CXCR2 in OC is associated with poor outcomes. However, the mechanisms by which CXCR2 regulates OC proliferation remain poorly understood. We generated CXCR2-positive cells from parental p53 wild-type (WT), mutant and null OC cells, and assessed the roles of CXCR2 on proliferation of OC cells in p53-dependent and independent manner. CXCR2 promoted cell growth rate: p53WT > mutant = null cells. Nutlin-3, a p53 stabilizer, inhibited cell proliferation in p53WT cells, but had little effect in p53-mutant or null cells, indicating p53-dependence of CXCR2-mediated proliferation. CXCR2 decreased p53 protein, a regulator of p21, and downregulated p21 promoter activity only in p53WT cells. The p53 responsive element (RE) of p21 promoter played a critical role in this CXCR2-mediated p21 downregulation. Moreover, CXCR2-positive cells activated more Akt than CXCR2-negative cells followed by enhanced murine double minute (Mdm2). Silencing Mdm2 or Akt1 upregulated p21 expression, whereas Akt1 overexpression downregulated p21 at the promoter and protein levels in p53WT cells. Cell cycle analysis revealed that CXCR2 decreased p21 gene in p53-null cells. Interestingly, romidepsin (histone deacetylase inhibitor)-induced p21 upregulation did not involve the p53 RE in the p21 promoter in p53-null cells. Romidepsin decreased the protein levels of Akt1 and Mdm2, leading to induction of p21 in p53-null cells. CXCR2 reduced romidepsin-induced p21 upregulation by activating Akt-induced Mdm2. Taken together, CXCR2 enhances cell proliferation by suppressing p21 through Akt-Mdm2 signaling in p53-dependent and independent manner.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 24231