Your browser doesn't support javascript.
loading
Nitric Oxide Prevents Glioblastoma Stem Cells' Expansion and Induces Temozolomide Sensitization.
Salvatori, Luisa; Malatesta, Silvia; Illi, Barbara; Somma, Maria Patrizia; Fionda, Cinzia; Stabile, Helena; Fontanella, Rosaria Anna; Gaetano, Carlo.
Afiliación
  • Salvatori L; Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy.
  • Malatesta S; Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy.
  • Illi B; Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy.
  • Somma MP; Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy.
  • Fionda C; Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy.
  • Stabile H; Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy.
  • Fontanella RA; Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
  • Gaetano C; Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
Int J Mol Sci ; 24(14)2023 Jul 10.
Article en En | MEDLINE | ID: mdl-37511047
ABSTRACT
Glioblastoma multiforme (GBM) has high mortality and recurrence rates. Malignancy resilience is ascribed to Glioblastoma Stem Cells (GSCs), which are resistant to Temozolomide (TMZ), the gold standard for GBM post-surgical treatment. However, Nitric Oxide (NO) has demonstrated anti-cancer efficacy in GBM cells, but its potential impact on GSCs remains unexplored. Accordingly, we investigated the effects of NO, both alone and in combination with TMZ, on patient-derived GSCs. Experimentally selected concentrations of diethylenetriamine/NO adduct and TMZ were used through a time course up to 21 days of treatment, to evaluate GSC proliferation and death, functional recovery, and apoptosis. Immunofluorescence and Western blot analyses revealed treatment-induced effects in cell cycle and DNA damage occurrence and repair. Our results showed that NO impairs self-renewal, disrupts cell-cycle progression, and expands the quiescent cells' population. Consistently, NO triggered a significant but tolerated level of DNA damage, but not apoptosis. Interestingly, NO/TMZ cotreatment further inhibited cell cycle progression, augmented G0 cells, induced cell death, but also enhanced DNA damage repair activity. These findings suggest that, although NO administration does not eliminate GSCs, it stunts their proliferation, and makes cells susceptible to TMZ. The resulting cytostatic effect may potentially allow long-term control over the GSCs' subpopulation.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Contexto en salud: 6_ODS3_enfermedades_notrasmisibles Problema de salud: 6_brain_nervous_system_cancer Asunto principal: Neoplasias Encefálicas / Glioblastoma Límite: Humans Idioma: En Revista: Int J Mol Sci Año: 2023 Tipo del documento: Article País de afiliación: Italia

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Contexto en salud: 6_ODS3_enfermedades_notrasmisibles Problema de salud: 6_brain_nervous_system_cancer Asunto principal: Neoplasias Encefálicas / Glioblastoma Límite: Humans Idioma: En Revista: Int J Mol Sci Año: 2023 Tipo del documento: Article País de afiliación: Italia
...