Your browser doesn't support javascript.
loading
Inhibitor of differentiation 1 transcription factor promotes metabolic reprogramming in hepatocellular carcinoma cells.
Sharma, Bal Krishan; Kolhe, Ravindra; Black, Stephen M; Keller, Jonathan R; Mivechi, Nahid F; Satyanarayana, Ande.
Afiliación
  • Sharma BK; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
  • Kolhe R; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
  • Black SM; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
  • Keller JR; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
  • Mivechi NF; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
  • Satyanarayana A; *Department of Biochemistry and Molecular Biology, Molecular Oncology and Biomarkers Program, Georgia Regents University Cancer Center, Department of Pathology, and Program in Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA; and Basic Science Pr
FASEB J ; 30(1): 262-75, 2016 Jan.
Article en En | MEDLINE | ID: mdl-26330493
ABSTRACT
Reprograming of metabolism is one of the central hallmarks of cancer. The majority of cancer cells depend on high rates of glycolysis and glutaminolysis for their growth and survival. A number of oncogenes and tumor suppressors have been connected to the regulation of altered glucose and glutamine metabolism in cancer cells. For example, the oncogene c-Myc plays vital roles in cancer cell metabolic adaptation by directly regulating various genes that participate in aerobic glycolysis and glutaminolysis. Inhibitor of differentiation 1 (Id1) is a helix-loop-helix transcription factor that plays important roles in cell proliferation, differentiation, and cell fate determination. Overexpression of Id1 causes intestinal adenomas and thymic lymphomas in mice, suggesting that Id1 could function as an oncogene. Despite it being an oncogene, whether Id1 plays any prominent role in cancer cell metabolic reprograming is unknown. Here, we demonstrate that Id1 is strongly expressed in human and mouse liver tumors and in hepatocellular carcinoma (HCC) cell lines, whereas its expression is very low or undetectable in normal liver tissues. In HCC cells, Id1 expression is regulated by the MAPK/ERK pathway at the transcriptional level. Knockdown of Id1 suppressed aerobic glycolysis and glutaminolysis, suggesting that Id1 promotes a metabolic shift toward aerobic glycolysis. At the molecular level, Id1 mediates its metabolic effects by regulating the expression levels of c-Myc. Knockdown of Id1 resulted in down-regulation (∼75%) of c-Myc, whereas overexpression of Id1 strongly induced (3-fold) c-Myc levels. Interestingly, knockdown of c-Myc resulted in down-regulation (∼60%) of Id1, suggesting a positive feedback-loop regulatory mechanism between Id1 and c-Myc. Under anaerobic conditions, both Id1 and c-Myc are down-regulated (50-70%), and overexpression of oxygen-insensitive hypoxia-inducible factor 1α (Hif1α) or its downstream target Mxi1 resulted in a significant reduction of c-Myc and Id1 (∼70%), suggesting that Hif1α suppresses Id1 and c-Myc under anaerobic conditions via Mxi1. Together, our findings indicate a prominent novel role for Id1 in liver cancer cell metabolic adaptation.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Oxígeno / Carcinoma Hepatocelular / Proteína 1 Inhibidora de la Diferenciación / Neoplasias Hepáticas Límite: Animals / Humans / Male Idioma: En Revista: FASEB J Asunto de la revista: BIOLOGIA / FISIOLOGIA Año: 2016 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Oxígeno / Carcinoma Hepatocelular / Proteína 1 Inhibidora de la Diferenciación / Neoplasias Hepáticas Límite: Animals / Humans / Male Idioma: En Revista: FASEB J Asunto de la revista: BIOLOGIA / FISIOLOGIA Año: 2016 Tipo del documento: Article