Your browser doesn't support javascript.
loading
Cancer-associated mutations in the p85α N-terminal SH2 domain activate a spectrum of receptor tyrosine kinases.
Li, Xinran; Lau, Amy Y T; Ng, Angel S N; Aldehaiman, Abdullah; Zhou, Yuan; Ng, Patrick K S; Arold, Stefan T; Cheung, Lydia W T.
Afiliação
  • Li X; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
  • Lau AYT; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
  • Ng ASN; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
  • Aldehaiman A; Biological and Environmental Science and Engineering, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia.
  • Zhou Y; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
  • Ng PKS; Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.
  • Arold ST; Biological and Environmental Science and Engineering, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia; stefan.arold@kaust.edu.sa lydiacwt@hku.hk.
  • Cheung LWT; Centre de Biologie Structurale, INSERM, Université de Montpellier, Montpellier 34090, France.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article em En | MEDLINE | ID: mdl-34507989
ABSTRACT
The phosphoinositide 3-kinase regulatory subunit p85α is a key regulator of kinase signaling and is frequently mutated in cancers. In the present study, we showed that in addition to weakening the inhibitory interaction between p85α and p110α, a group of driver mutations in the p85α N-terminal SH2 domain activated EGFR, HER2, HER3, c-Met, and IGF-1R in a p110α-independent manner. Cancer cells expressing these mutations exhibited the activation of p110α and the AKT pathway. Interestingly, the activation of EGFR, HER2, and c-Met was attributed to the ability of driver mutations to inhibit HER3 ubiquitination and degradation. The resulting increase in HER3 protein levels promoted its heterodimerization with EGFR, HER2, and c-Met, as well as the allosteric activation of these dimerized partners; however, HER3 silencing abolished this transactivation. Accordingly, inhibitors of either AKT or the HER family reduced the oncogenicity of driver mutations. The combination of these inhibitors resulted in marked synergy. Taken together, our findings provide mechanistic insights and suggest therapeutic strategies targeting a class of recurrent p85α mutations.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Classe I de Fosfatidilinositol 3-Quinases / Classe Ia de Fosfatidilinositol 3-Quinase Tipo de estudo: Risk_factors_studies Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Classe I de Fosfatidilinositol 3-Quinases / Classe Ia de Fosfatidilinositol 3-Quinase Tipo de estudo: Risk_factors_studies Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article