Your browser doesn't support javascript.
loading
MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma.
Zhang, Yiru; Nguyen, Trang T T; Shang, Enyuan; Mela, Angeliki; Humala, Nelson; Mahajan, Aayushi; Zhao, Junfei; Shu, Chang; Torrini, Consuelo; Sanchez-Quintero, Maria J; Kleiner, Giulio; Bianchetti, Elena; Westhoff, Mike-Andrew; Quinzii, Catarina M; Karpel-Massler, Georg; Bruce, Jeffrey N; Canoll, Peter; Siegelin, Markus D.
Afiliación
  • Zhang Y; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Nguyen TTT; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Shang E; Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, New York.
  • Mela A; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Humala N; Department of Neurological Surgery, Columbia University Medical Center, New York, New York.
  • Mahajan A; Department of Neurological Surgery, Columbia University Medical Center, New York, New York.
  • Zhao J; Department of Biomedical Informatics, Columbia University, New York, New York.
  • Shu C; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Torrini C; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Sanchez-Quintero MJ; Department of Neurology, Columbia University Medical Center, New York, New York.
  • Kleiner G; Department of Neurology, Columbia University Medical Center, New York, New York.
  • Bianchetti E; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Westhoff MA; Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.
  • Quinzii CM; Department of Neurology, Columbia University Medical Center, New York, New York.
  • Karpel-Massler G; Department of Neurosurgery, Ulm University Medical Center, Ulm, Germany.
  • Bruce JN; Department of Neurological Surgery, Columbia University Medical Center, New York, New York.
  • Canoll P; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
  • Siegelin MD; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York. ms4169@cumc.columbia.edu.
Cancer Res ; 80(1): 30-43, 2020 01 01.
Article en En | MEDLINE | ID: mdl-31694905
ABSTRACT
The receptor kinase c-MET has emerged as a target for glioblastoma therapy. However, treatment resistance emerges inevitably. Here, we performed global metabolite screening with metabolite set enrichment coupled with transcriptome and gene set enrichment analysis and proteomic screening, and identified substantial reprogramming of tumor metabolism involving oxidative phosphorylation and fatty acid oxidation (FAO) with substantial accumulation of acyl-carnitines accompanied by an increase of PGC1α in response to genetic (shRNA and CRISPR/Cas9) and pharmacologic (crizotinib) inhibition of c-MET. Extracellular flux and carbon tracing analyses (U-13C-glucose, U-13C-glutamine, and U-13C-palmitic acid) demonstrated enhanced oxidative metabolism, which was driven by FAO and supported by increased anaplerosis of glucose carbons. These findings were observed in concert with increased number and fusion of mitochondria and production of reactive oxygen species. Genetic interference with PGC1α rescued this oxidative phenotype driven by c-MET inhibition. Silencing and chromatin immunoprecipitation experiments demonstrated that cAMP response elements binding protein regulates the expression of PGC1α in the context of c-MET inhibition. Interference with both oxidative phosphorylation (metformin, oligomycin) and ß-oxidation of fatty acids (etomoxir) enhanced the antitumor efficacy of c-MET inhibition. Synergistic cell death was observed with c-MET inhibition and gamitrinib treatment. In patient-derived xenograft models, combination treatments of crizotinib and etomoxir, and crizotinib and gamitrinib were significantly more efficacious than single treatments and did not induce toxicity. Collectively, we have unraveled the mechanistic underpinnings of c-MET inhibition and identified novel combination therapies that may enhance its therapeutic efficacy.

SIGNIFICANCE:

c-MET inhibition causes profound metabolic reprogramming that can be targeted by drug combination therapies.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Encefálicas / Protocolos de Quimioterapia Combinada Antineoplásica / Glioblastoma / Proteínas Proto-Oncogénicas c-met / Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancer Res Año: 2020 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Encefálicas / Protocolos de Quimioterapia Combinada Antineoplásica / Glioblastoma / Proteínas Proto-Oncogénicas c-met / Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancer Res Año: 2020 Tipo del documento: Article