Your browser doesn't support javascript.
loading
p85ß alters response to EGFR inhibitor in ovarian cancer through p38 MAPK-mediated regulation of DNA repair.
Mak, Victor Cy; Li, Xinran; Rao, Ling; Zhou, Yuan; Tsao, Sai-Wah; Cheung, Lydia Wt.
Afiliación
  • Mak VC; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
  • Li X; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
  • Rao L; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
  • Zhou Y; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
  • Tsao SW; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
  • Cheung LW; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China. Electronic address: lydiacwt@hku.hk.
Neoplasia ; 23(7): 718-730, 2021 07.
Article en En | MEDLINE | ID: mdl-34144267
ABSTRACT
EGFR signaling promotes ovarian cancer tumorigenesis, and high EGFR expression correlates with poor prognosis. However, EGFR inhibitors alone have demonstrated limited clinical benefit for ovarian cancer patients, owing partly to tumor resistance and the lack of predictive biomarkers. Cotargeting EGFR and the PI3K pathway has been previously shown to yield synergistic antitumor effects in ovarian cancer. Therefore, we reasoned that PI3K may affect cellular response to EGFR inhibition. In this study, we revealed PI3K isoform-specific effects on the sensitivity of ovarian cancer cells to the EGFR inhibitor erlotinib. Gene silencing of PIK3CA (p110α) and PIK3CB (p110ß) rendered cells more susceptible to erlotinib. In contrast, low expression of PIK3R2 (p85ß) was associated with erlotinib resistance. Depletion of PIK3R2, but not PIK3CA or PIK3CB, led to increased DNA damage and reduced level of the nonhomologous end joining DNA repair protein BRD4. Intriguingly, these defects in DNA repair were reversed upon erlotinib treatment, which caused activation and nuclear import of p38 MAPK to promote DNA repair with increased protein levels of 53BP1 and BRD4 and foci formation of 53BP1. Remarkably, inhibition of p38 MAPK or BRD4 re-sensitized PIK3R2-depleted cells to erlotinib. Collectively, these data suggest that p38 MAPK activation and the subsequent DNA repair serve as a resistance mechanism to EGFR inhibitor. Combined inhibition of EGFR and p38 MAPK or DNA repair may maximize the therapeutic potential of EGFR inhibitor in ovarian cancer.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias Ováricas / Proteínas Quinasas p38 Activadas por Mitógenos / Inhibidores de Proteínas Quinasas / Reparación del ADN / Fosfatidilinositol 3-Quinasa Clase Ia Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: Neoplasia Asunto de la revista: NEOPLASIAS Año: 2021 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias Ováricas / Proteínas Quinasas p38 Activadas por Mitógenos / Inhibidores de Proteínas Quinasas / Reparación del ADN / Fosfatidilinositol 3-Quinasa Clase Ia Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: Neoplasia Asunto de la revista: NEOPLASIAS Año: 2021 Tipo del documento: Article País de afiliación: China