Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 71(14): 2577-604, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24363178

RESUMEN

Pyruvate is a keystone molecule critical for numerous aspects of eukaryotic and human metabolism. Pyruvate is the end-product of glycolysis, is derived from additional sources in the cellular cytoplasm, and is ultimately destined for transport into mitochondria as a master fuel input undergirding citric acid cycle carbon flux. In mitochondria, pyruvate drives ATP production by oxidative phosphorylation and multiple biosynthetic pathways intersecting the citric acid cycle. Mitochondrial pyruvate metabolism is regulated by many enzymes, including the recently discovered mitochondria pyruvate carrier, pyruvate dehydrogenase, and pyruvate carboxylase, to modulate overall pyruvate carbon flux. Mutations in any of the genes encoding for proteins regulating pyruvate metabolism may lead to disease. Numerous cases have been described. Aberrant pyruvate metabolism plays an especially prominent role in cancer, heart failure, and neurodegeneration. Because most major diseases involve aberrant metabolism, understanding and exploiting pyruvate carbon flux may yield novel treatments that enhance human health.


Asunto(s)
Errores Innatos del Metabolismo del Piruvato/metabolismo , Ácido Pirúvico/metabolismo , Ciclo del Ácido Cítrico , Citosol/metabolismo , Cardiopatías/metabolismo , Humanos , Mitocondrias/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Ácido Pirúvico/química
2.
Cell Rep ; 28(10): 2608-2619.e6, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31484072

RESUMEN

Hepatocellular carcinoma (HCC) is a devastating cancer increasingly caused by non-alcoholic fatty liver disease (NAFLD). Disrupting the liver Mitochondrial Pyruvate Carrier (MPC) in mice attenuates NAFLD. Thus, we considered whether liver MPC disruption also prevents HCC. Here, we use the N-nitrosodiethylamine plus carbon tetrachloride model of HCC development to test how liver-specific MPC knock out affects hepatocellular tumorigenesis. Our data show that liver MPC ablation markedly decreases tumorigenesis and that MPC-deficient tumors transcriptomically downregulate glutathione metabolism. We observe that MPC disruption and glutathione depletion in cultured hepatomas are synthetically lethal. Stable isotope tracing shows that hepatocyte MPC disruption reroutes glutamine from glutathione synthesis into the tricarboxylic acid (TCA) cycle. These results support a model where inducing metabolic competition for glutamine by MPC disruption impairs hepatocellular tumorigenesis by limiting glutathione synthesis. These findings raise the possibility that combining MPC disruption and glutathione stress may be therapeutically useful in HCC and additional cancers.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Hepatocelular/metabolismo , Ciclo del Ácido Cítrico , Glutamina/metabolismo , Glutatión/biosíntesis , Neoplasias Hepáticas/metabolismo , Mitocondrias/metabolismo , Ácido Pirúvico/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/genética , Ratones Endogámicos C57BL , Proteínas de Neoplasias/metabolismo , Especificidad de Órganos , Transcriptoma/genética
3.
Elife ; 82019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31305240

RESUMEN

Metabolic cycles are a fundamental element of cellular and organismal function. Among the most critical in higher organisms is the Cori Cycle, the systemic cycling between lactate and glucose. Here, skeletal muscle-specific Mitochondrial Pyruvate Carrier (MPC) deletion in mice diverted pyruvate into circulating lactate. This switch disinhibited muscle fatty acid oxidation and drove Cori Cycling that contributed to increased energy expenditure. Loss of muscle MPC activity led to strikingly decreased adiposity with complete muscle mass and strength retention. Notably, despite decreasing muscle glucose oxidation, muscle MPC disruption increased muscle glucose uptake and whole-body insulin sensitivity. Furthermore, chronic and acute muscle MPC deletion accelerated fat mass loss on a normal diet after high fat diet-induced obesity. Our results illuminate the role of the skeletal muscle MPC as a whole-body carbon flux control point. They highlight the potential utility of modulating muscle pyruvate utilization to ameliorate obesity and type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Redes y Vías Metabólicas , Mitocondrias Musculares/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Ácido Pirúvico/metabolismo , Delgadez , Adiposidad , Animales , Proteínas de Transporte de Anión/deficiencia , Eliminación de Gen , Lactatos/metabolismo , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial/deficiencia , Transportadores de Ácidos Monocarboxílicos/deficiencia , Fuerza Muscular
4.
Cell Metab ; 22(4): 669-81, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26344103

RESUMEN

Gluconeogenesis is critical for maintenance of euglycemia during fasting. Elevated gluconeogenesis during type 2 diabetes (T2D) contributes to chronic hyperglycemia. Pyruvate is a major gluconeogenic substrate and requires import into the mitochondrial matrix for channeling into gluconeogenesis. Here, we demonstrate that the mitochondrial pyruvate carrier (MPC) comprising the Mpc1 and Mpc2 proteins is required for efficient regulation of hepatic gluconeogenesis. Liver-specific deletion of Mpc1 abolished hepatic MPC activity and markedly decreased pyruvate-driven gluconeogenesis and TCA cycle flux. Loss of MPC activity induced adaptive utilization of glutamine and increased urea cycle activity. Diet-induced obesity increased hepatic MPC expression and activity. Constitutive Mpc1 deletion attenuated the development of hyperglycemia induced by a high-fat diet. Acute, virally mediated Mpc1 deletion after diet-induced obesity decreased hyperglycemia and improved glucose tolerance. We conclude that the MPC is required for efficient regulation of gluconeogenesis and that the MPC contributes to the elevated gluconeogenesis and hyperglycemia in T2D.


Asunto(s)
Glucosa/metabolismo , Mitocondrias Hepáticas/enzimología , Proproteína Convertasa 1/metabolismo , Acrilatos/farmacología , Animales , Células Cultivadas , Ciclo del Ácido Cítrico/efectos de los fármacos , Dieta Alta en Grasa , Gluconeogénesis/efectos de los fármacos , Glutamina/metabolismo , Glucógeno/análisis , Hepatocitos/citología , Hepatocitos/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Proproteína Convertasa 1/deficiencia , Proproteína Convertasa 1/genética , Proproteína Convertasa 2/antagonistas & inhibidores , Proproteína Convertasa 2/genética , Proproteína Convertasa 2/metabolismo , Ácido Pirúvico/metabolismo , Triglicéridos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA