Your browser doesn't support javascript.
loading
Non-Canonical Role of PDK1 as a Negative Regulator of Apoptosis through Macromolecular Complexes Assembly at the ER-Mitochondria Interface in Oncogene-Driven NSCLC.
De Rosa, Viviana; Iommelli, Francesca; Terlizzi, Cristina; Leggiero, Eleonora; Camerlingo, Rosa; Altobelli, Giovanna G; Fonti, Rosa; Pastore, Lucio; Del Vecchio, Silvana.
Afiliação
  • De Rosa V; Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy.
  • Iommelli F; Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy.
  • Terlizzi C; Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
  • Leggiero E; CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy.
  • Camerlingo R; Department of Cell Biology and Biotherapy, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
  • Altobelli GG; Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
  • Fonti R; Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy.
  • Pastore L; CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy.
  • Del Vecchio S; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131 Naples, Italy.
Cancers (Basel) ; 13(16)2021 Aug 17.
Article em En | MEDLINE | ID: mdl-34439291
ABSTRACT
Here, we tested whether co-targeting of glucose metabolism and oncogene drivers may enhance tumor response to tyrosine kinase inhibitors (TKIs) in NSCLC. To this end, pyruvate dehydrogenase kinase 1 (PDK1) was stably downregulated in oncogene-driven NSCLC cell lines exposed or not to TKIs. H1993 and H1975 cells were stably transfected with scrambled (shCTRL) or PDK1-targeted (shPDK1) shRNA and then treated with MET inhibitor crizotinib (1 µM), double mutant EGFRL858R/T790M inhibitor WZ4002 (1 µM) or vehicle for 48 h. The effects of PDK1 knockdown on glucose metabolism and apoptosis were evaluated in untreated and TKI-treated cells. PDK1 knockdown alone did not cause significant changes in glycolytic cascade, ATP production and glucose consumption, but it enhanced maximal respiration in shPDK1 cells when compared to controls. When combined with TKI treatment, PDK1 downregulation caused a strong enhancement of OXPHOS and a marked reduction in key glycolytic enzymes. Furthermore, increased levels of apoptotic markers were found in shPDK1 cells as compared to shCTRL cells after treatment with TKIs. Co-immunoprecipitation studies showed that PDK1 interacts with PKM2, Bcl-2 and Bcl-xL, forming macromolecular complexes at the ER-mitochondria interface. Our findings showed that downregulation of PDK1 is able to potentiate the effects of TKIs through the disruption of macromolecular complexes involving PKM2, Bcl-2 and Bcl-xL.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Itália

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Itália