Your browser doesn't support javascript.
loading
Histone modifications affect differential regulation of TGFß- induced NADPH oxidase 4 (NOX4) by wild-type and mutant p53.
Boudreau, Howard E; Ma, Wei Feng; Korzeniowska, Agnieszka; Park, Jonathan J; Bhagwat, Medha A; Leto, Thomas L.
Afiliación
  • Boudreau HE; Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
  • Ma WF; Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
  • Korzeniowska A; Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
  • Park JJ; Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
  • Bhagwat MA; Bioinformatics Support Program, National Institutes of Health Library, National Institutes of Health, Maryland, USA.
  • Leto TL; Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
Oncotarget ; 8(27): 44379-44397, 2017 Jul 04.
Article en En | MEDLINE | ID: mdl-28574838
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 TGFß-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 TGFß/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 TGFß-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 TGFß-stimulated histone acetylation within the NOX4 promoter. Finally, wound assays demonstrated NOX4 and p300 promote TGFß/mut-p53-mediated cell migration. Our studies provide new insight into TGFß/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.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Histonas / Regulación de la Expresión Génica / Proteína p53 Supresora de Tumor / Factor de Crecimiento Transformador beta / NADPH Oxidasa 4 Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: Oncotarget Año: 2017 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Histonas / Regulación de la Expresión Génica / Proteína p53 Supresora de Tumor / Factor de Crecimiento Transformador beta / NADPH Oxidasa 4 Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: Oncotarget Año: 2017 Tipo del documento: Article País de afiliación: Estados Unidos
...