Your browser doesn't support javascript.
loading
Distinct functions of wild-type and R273H mutant Δ133p53α differentially regulate glioblastoma aggressiveness and therapy-induced senescence.
Joruiz, Sebastien M; Von Muhlinen, Natalia; Horikawa, Izumi; Gilbert, Mark R; Harris, Curtis C.
Afiliação
  • Joruiz SM; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.
  • Von Muhlinen N; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.
  • Horikawa I; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.
  • Gilbert MR; Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Harris CC; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA. harrisc@mail.nih.gov.
Cell Death Dis ; 15(6): 454, 2024 Jun 27.
Article em En | MEDLINE | ID: mdl-38937431
ABSTRACT
Despite being mutated in 92% of TP53 mutant cancers, how mutations on p53 isoforms affect their activities remain largely unknown. Therefore, exploring the effect of mutations on p53 isoforms activities is a critical, albeit unexplored area in the p53 field. In this article, we report for the first time a mutant Δ133p53α-specific pathway which increases IL4I1 and IDO1 expression and activates AHR, a tumor-promoting mechanism. Accordingly, while WT Δ133p53α reduces apoptosis to promote DNA repair, mutant R273H also reduces apoptosis but fails to maintain genomic stability, increasing the risks of accumulation of mutations and tumor's deriving towards a more aggressive phenotype. Furthermore, using 2D and 3D spheroids culture, we show that WT Δ133p53α reduces cell proliferation, EMT, and invasion, while the mutant Δ133p53α R273H enhances all three processes, confirming its oncogenic potential and strongly suggesting a similar in vivo activity. Importantly, the effects on cell growth and invasion are independent of mutant full-length p53α, indicating that these activities are actively carried by mutant Δ133p53α R273H. Furthermore, both WT and mutant Δ133p53α reduce cellular senescence in a senescence inducer-dependent manner (temozolomide or radiation) because they regulate different senescence-associated target genes. Hence, WT Δ133p53α rescues temozolomide-induced but not radiation-induced senescence, while mutant Δ133p53α R273H rescues radiation-induced but not temozolomide-induced senescence. Lastly, we determined that IL4I1, IDO1, and AHR are significantly higher in GBMs compared to low-grade gliomas. Importantly, high expression of all three genes in LGG and IL4I1 in GBM is significantly associated with poorer patients' survival, confirming the clinical relevance of this pathway in glioblastomas. These data show that, compared to WT Δ133p53α, R273H mutation reorientates its activities toward carcinogenesis and activates the oncogenic IL4I1/IDO1/AHR pathway, a potential prognostic marker and therapeutic target in GBM by combining drugs specifically modulating Δ133p53α expression and IDO1/Il4I1/AHR inhibitors.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteína Supressora de Tumor p53 / Senescência Celular / Glioblastoma / Proliferação de Células / Mutação Limite: Humans Idioma: En Revista: Cell Death Dis Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteína Supressora de Tumor p53 / Senescência Celular / Glioblastoma / Proliferação de Células / Mutação Limite: Humans Idioma: En Revista: Cell Death Dis Ano de publicação: 2024 Tipo de documento: Article