Your browser doesn't support javascript.
loading
Sex- and mutation-specific p53 gain-of-function activity in gliomagenesis.
Rockwell, Nathan C; Yang, Wei; Warrington, Nicole M; Staller, Max V; Griffith, Malachi; Griffith, Obi L; Gurnett, Christina A; Cohen, Barak A; Baldridge, Dustin; Rubin, Joshua B.
Affiliation
  • Rockwell NC; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
  • Yang W; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
  • Warrington NM; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
  • Staller MV; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
  • Griffith M; Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720.
  • Griffith OL; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
  • Gurnett CA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA.
  • Cohen BA; Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, 63110, MO, USA.
  • Baldridge D; Siteman Cancer Center, Washington University School of Medicine, St. Louis, 63110, MO, USA.
  • Rubin JB; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
Cancer Res Commun ; 1(3): 148-163, 2021 12.
Article in En | MEDLINE | ID: mdl-34957471
ABSTRACT
In cancer, missense mutations in the DNA-binding domain of TP53 are common. They abrogate canonical p53 activity and frequently confer gain-of-oncogenic function (GOF) through localization of transcriptionally active mutant p53 to non-canonical genes. We found that several recurring p53 mutations exhibit a sex difference in frequency in patients with glioblastoma (GBM). In vitro and in vivo analysis of three mutations, p53R172H, p53Y202C, and p53Y217C revealed unique interactions between cellular sex and p53 GOF mutations that determined each mutation's ability to transform male versus female primary mouse astrocytes. These phenotypic differences were correlated with sex- and p53 mutation- specific patterns of genomic localization to the transcriptional start sites of upregulated genes belonging to core cancer pathways. The promoter regions of these genes exhibited a sex difference in enrichment for different transcription factor DNA-binding motifs. Together, our data establish a novel mechanism for sex specific mutant p53 GOF activity in GBM with implications for all cancer.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Tumor Suppressor Protein p53 / Glioblastoma Limits: Animals Language: En Journal: Cancer Res Commun Year: 2021 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Tumor Suppressor Protein p53 / Glioblastoma Limits: Animals Language: En Journal: Cancer Res Commun Year: 2021 Document type: Article Affiliation country: United States