Oncotarget

Research Papers:

Histone modifications affect differential regulation of TGFβ- induced NADPH oxidase 4 (NOX4) by wild-type and mutant p53

Howard E. Boudreau, Wei Feng Ma, Agnieszka Korzeniowska, Jonathan J. Park, Medha A. Bhagwat and Thomas L. Leto _

PDF  |  HTML  |  How to cite

Oncotarget. 2017; 8:44379-44397. https://doi.org/10.18632/oncotarget.17892

Metrics: PDF 1930 views  |   HTML 2336 views  |   ?  


Abstract

Howard E. Boudreau1, Wei Feng Ma1, Agnieszka Korzeniowska1, Jonathan J. Park1, Medha A. Bhagwat2 and Thomas L. Leto1

1Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA

2Bioinformatics Support Program, National Institutes of Health Library, National Institutes of Health, Bethesda, Maryland, USA

Correspondence to:

Thomas L. Leto, email: [email protected]

Keywords: mutant p53, NOX4, p300, histone modification, cell migration

Received: December 16, 2016     Accepted: April 26, 2017     Published: May 16, 2017

ABSTRACT

Previously, we showed wild-type (WT) and mutant (mut) p53 differentially regulate reactive oxygen species (ROS) generation by NADPH oxidase-4 (NOX4): p53-WT suppresses TGFβ-induced NOX4, ROS and cell migration, whereas tumor-associated mut-p53 proteins enhance NOX4 expression and cell migration. Here, we extended our findings on the effects of p53 on NOX4 in several tumors and examined the basis of NOX4 transcriptional regulation by p53 and SMAD3. Statistical analysis of expression data from primary tumors available from The Cancer Genome Atlas (TCGA) detected correlations between mut-p53 and increased NOX4 expression. Furthermore, by altering p53 levels in cell culture models we showed several common tumor-associated mutant forms support TGFβ/SMAD3-dependent NOX4 expression. Deletion analysis revealed two critical SMAD3 binding elements (SBE) required for mut-p53-dependent NOX4 induction, whereas p53-WT caused dose-dependent suppression of NOX4 transcription. ChIP analysis revealed SMAD3 and p53-WT or mut-p53 associate with SBEs and p53 response elements in a TGFβ-dependent manner. Interestingly, the repressive effects of p53-WT on NOX4 were relieved by mutation of its transactivation domain or histone deacetylase (HDAC) inhibitor treatment. Overexpression of p300, a transcriptional co-regulator and histone acetyltransferase (HAT), enhanced p53-mediated NOX4 induction, whereas HAT-inactive p300 reduced NOX4 expression. Mut-p53 augmented TGFβ-stimulated histone acetylation within the NOX4 promoter. Finally, wound assays demonstrated NOX4 and p300 promote TGFβ/mut-p53-mediated cell migration. Our studies provide new insight into TGFβ/SMAD3 and mut-p53-mediated NOX4 induction involving epigenetic control of NOX4 in tumor cell migration, suggesting NOX4 is a potential therapeutic target to combat tumor progression and metastasis.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 17892