Your browser doesn't support javascript.
loading
Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways.
Jia, Dongya; Lu, Mingyang; Jung, Kwang Hwa; Park, Jun Hyoung; Yu, Linglin; Onuchic, José N; Kaipparettu, Benny Abraham; Levine, Herbert.
Afiliação
  • Jia D; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005.
  • Lu M; Systems, Synthetic and Physical Biology Program, Rice University, Houston, TX 77005.
  • Jung KH; The Jackson Laboratory, Bar Harbor, ME 04609.
  • Park JH; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.
  • Yu L; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.
  • Onuchic JN; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005.
  • Kaipparettu BA; Applied Physics Program, Rice University, Houston, TX 77005.
  • Levine H; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005; jonuchic@rice.edu kaippare@bcm.edu herbert.levine@rice.edu.
Proc Natl Acad Sci U S A ; 116(9): 3909-3918, 2019 02 26.
Article em En | MEDLINE | ID: mdl-30733294
ABSTRACT
Metabolic plasticity enables cancer cells to switch their metabolism phenotypes between glycolysis and oxidative phosphorylation (OXPHOS) during tumorigenesis and metastasis. However, it is still largely unknown how cancer cells orchestrate gene regulation to balance their glycolysis and OXPHOS activities. Previously, by modeling the gene regulation of cancer metabolism we have reported that cancer cells can acquire a stable hybrid metabolic state in which both glycolysis and OXPHOS can be used. Here, to comprehensively characterize cancer metabolic activity, we establish a theoretical framework by coupling gene regulation with metabolic pathways. Our modeling results demonstrate a direct association between the activities of AMPK and HIF-1, master regulators of OXPHOS and glycolysis, respectively, with the activities of three major metabolic pathways glucose oxidation, glycolysis, and fatty acid oxidation. Our model further characterizes the hybrid metabolic state and a metabolically inactive state where cells have low activity of both glycolysis and OXPHOS. We verify the model prediction using metabolomics and transcriptomics data from paired tumor and adjacent benign tissue samples from a cohort of breast cancer patients and RNA-sequencing data from The Cancer Genome Atlas. We further validate the model prediction by in vitro studies of aggressive triple-negative breast cancer (TNBC) cells. The experimental results confirm that TNBC cells can maintain a hybrid metabolic phenotype and targeting both glycolysis and OXPHOS is necessary to eliminate their metabolic plasticity. In summary, our work serves as a platform to symmetrically study how tuning gene activity modulates metabolic pathway activity, and vice versa.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Subunidade alfa do Fator 1 Induzível por Hipóxia / Redes e Vias Metabólicas / Proteínas Quinases Ativadas por AMP / Neoplasias de Mama Triplo Negativas Tipo de estudo: Prognostic_studies Limite: Female / Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Subunidade alfa do Fator 1 Induzível por Hipóxia / Redes e Vias Metabólicas / Proteínas Quinases Ativadas por AMP / Neoplasias de Mama Triplo Negativas Tipo de estudo: Prognostic_studies Limite: Female / Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article