Cancer-associated mutations in the p85α N-terminal SH2 domain activate a spectrum of receptor tyrosine kinases

التفاصيل البيبلوغرافية
العنوان: Cancer-associated mutations in the p85α N-terminal SH2 domain activate a spectrum of receptor tyrosine kinases
المؤلفون: Lydia W.T. Cheung, Angel S N Ng, Abdullah Aldehaiman, Amy Y T Lau, Patrick Kwok Shing Ng, Stefan T. Arold, Yuan Zhou, Xinran Li
المصدر: Proceedings of the National Academy of Sciences of the United States of America
سنة النشر: 2021
مصطلحات موضوعية: Receptor, ErbB-3, Class I Phosphatidylinositol 3-Kinases, Allosteric regulation, P110α, SH2 domain, medicine.disease_cause, Receptor tyrosine kinase, src Homology Domains, Transactivation, Phosphatidylinositol 3-Kinases, Protein Domains, Catalytic Domain, Cell Line, Tumor, Neoplasms, medicine, Humans, skin and connective tissue diseases, Protein kinase B, PI3K/AKT/mTOR pathway, Mutation, Multidisciplinary, biology, receptor tyrosine kinases, Chemistry, Receptor Protein-Tyrosine Kinases, p85α, Cell Biology, Biological Sciences, HCT116 Cells, Class Ia Phosphatidylinositol 3-Kinase, biology.protein, Cancer research, Proto-Oncogene Proteins c-akt, Signal Transduction
الوصف: Significance Phosphoinositide 3-kinase activation typically occurs following stimulation by upstream receptor tyrosine kinases (RTKs), which alleviate p110α inhibition by p85α. p85α and p110α driver mutations have been reported to activate p110α by disrupting the inhibitory interface between p85α and p110α. This study revealed that driver mutations in the p85α N-terminal SH2 domain can enhance p110α activity by inducing the activation of multiple RTKs. Furthermore, combination treatment with RTK and AKT inhibitors provides synergistic therapeutic efficacy. This previously uncharacterized oncogenic mechanism presents the exploitable vulnerability of a class of p85α mutant tumors.
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.
تدمد: 1091-6490
URL الوصول: https://explore.openaire.eu/search/publication?articleId=doi_dedup___::508557af3f0465bb61afc746140561dc
https://pubmed.ncbi.nlm.nih.gov/34507989
حقوق: OPEN
رقم الأكسشن: edsair.doi.dedup.....508557af3f0465bb61afc746140561dc
قاعدة البيانات: OpenAIRE