Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Metab ; 2(11): 1248-1264, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33106689

RESUMEN

In addition to fatty acids, glucose and lactate are important myocardial substrates under physiologic and stress conditions. They are metabolized to pyruvate, which enters mitochondria via the mitochondrial pyruvate carrier (MPC) for citric acid cycle metabolism. In the present study, we show that MPC-mediated mitochondrial pyruvate utilization is essential for the partitioning of glucose-derived cytosolic metabolic intermediates, which modulate myocardial stress adaptation. Mice with cardiomyocyte-restricted deletion of subunit 1 of MPC (cMPC1-/-) developed age-dependent pathologic cardiac hypertrophy, transitioning to a dilated cardiomyopathy and premature death. Hypertrophied hearts accumulated lactate, pyruvate and glycogen, and displayed increased protein O-linked N-acetylglucosamine, which was prevented by increasing availability of non-glucose substrates in vivo by a ketogenic diet (KD) or a high-fat diet, which reversed the structural, metabolic and functional remodelling of non-stressed cMPC1-/- hearts. Although concurrent short-term KDs did not rescue cMPC1-/- hearts from rapid decompensation and early mortality after pressure overload, 3 weeks of a KD before transverse aortic constriction was sufficient to rescue this phenotype. Together, our results highlight the centrality of pyruvate metabolism to myocardial metabolism and function.


Asunto(s)
Adaptación Fisiológica/fisiología , Proteínas de Transporte de Anión/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Miocardio/metabolismo , Estrés Fisiológico/fisiología , Adaptación Fisiológica/genética , Animales , Proteínas de Transporte de Anión/genética , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Constricción Patológica , Citosol/metabolismo , Dieta Alta en Grasa , Dieta Cetogénica , Ecocardiografía , Técnicas In Vitro , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Miocitos Cardíacos/metabolismo , Ácido Pirúvico/metabolismo , Estrés Fisiológico/genética
3.
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
4.
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
5.
JCI Insight ; 52019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31145700

RESUMEN

The Mitochondrial Pyruvate Carrier (MPC) occupies a central metabolic node by transporting cytosolic pyruvate into the mitochondrial matrix and linking glycolysis with mitochondrial metabolism. Two reported human MPC1 mutations cause developmental abnormalities, neurological problems, metabolic deficits, and for one patient, early death. We aimed to understand biochemical mechanisms by which the human patient C289T and T236A MPC1 alleles disrupt MPC function. MPC1 C289T encodes two protein variants, a mis-spliced, truncation mutant (A58G) and a full length point mutant (R97W). MPC1 T236A encodes a full length point mutant (L79H). Using human patient fibroblasts and complementation of CRISPR-deleted, MPC1 null mouse C2C12 cells, we investigated how MPC1 mutations cause MPC deficiency. Truncated MPC1 A58G protein was intrinsically unstable and failed to form MPC complexes. The MPC1 R97W protein was less stable but when overexpressed formed complexes with MPC2 that retained pyruvate transport activity. Conversely, MPC1 L79H protein formed stable complexes with MPC2, but these complexes failed to transport pyruvate. These findings inform MPC structure-function relationships and delineate three distinct biochemical pathologies resulting from two human patient MPC1 mutations. They also illustrate an efficient gene pass-through system for mechanistically investigating human inborn errors in pyruvate metabolism.


Asunto(s)
Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Adolescente , Alelos , Animales , Línea Celular , Niño , Femenino , Fibroblastos , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/metabolismo , Piel , Relación Estructura-Actividad , Adulto Joven
6.
Mol Metab ; 6(11): 1468-1479, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29107293

RESUMEN

OBJECTIVE: Excessive hepatic gluconeogenesis is a defining feature of type 2 diabetes (T2D). Most gluconeogenic flux is routed through mitochondria. The mitochondrial pyruvate carrier (MPC) transports pyruvate from the cytosol into the mitochondrial matrix, thereby gating pyruvate-driven gluconeogenesis. Disruption of the hepatocyte MPC attenuates hyperglycemia in mice during high fat diet (HFD)-induced obesity but exerts minimal effects on glycemia in normal chow diet (NCD)-fed conditions. The goal of this investigation was to test whether hepatocyte MPC disruption provides sustained protection from hyperglycemia during long-term HFD and the differential effects of hepatocyte MPC disruption on TCA cycle metabolism in NCD versus HFD conditions. METHOD: We utilized long-term high fat feeding, serial measurements of postabsorptive blood glucose and metabolomic profiling and 13C-lactate/13C-pyruvate tracing to investigate the contribution of the MPC to hyperglycemia and altered hepatic TCA cycle metabolism during HFD-induced obesity. RESULTS: Hepatocyte MPC disruption resulted in long-term attenuation of hyperglycemia induced by HFD. HFD increased hepatic mitochondrial pyruvate utilization and TCA cycle capacity in an MPC-dependent manner. Furthermore, MPC disruption decreased progression of fibrosis and levels of transcript markers of inflammation. CONCLUSIONS: By contributing to chronic hyperglycemia, fibrosis, and TCA cycle expansion, the hepatocyte MPC is a key mediator of the pathophysiology induced in the HFD model of T2D.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Ciclo del Ácido Cítrico/fisiología , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Animales , Glucemia/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Hepatocitos/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Transportadores de Ácidos Monocarboxílicos , Obesidad/metabolismo , Ácido Pirúvico/metabolismo
7.
J Biol Chem ; 291(14): 7409-17, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26823462

RESUMEN

The discovery that theMPC1andMPC2genes encode the protein components of the mitochondrial pyruvate carrier (MPC) has invigorated studies of mitochondrial pyruvate transport and its regulation in normal and disease states. Indeed, recent reports have demonstrated MPC involvement in the control of cell fate in cancer and gluconeogenesis in models of type 2 diabetes. Biochemical measurements of MPC activity are foundational for understanding the role of pyruvate transport in health and disease. We developed a 96-well scaled method of [(14)C]pyruvate uptake that markedly decreases sample requirements and increases throughput relative to previous techniques. This method was applied to determine the mouse liver MPCKm(28.0 ± 3.9 µm) andVmax(1.08 ± 0.05 nmol/min/mg), which have not previously been reported.KmandVmaxof the rat liver MPC were found to be 71.2 ± 17 µmand 1.42 ± 0.14 nmol/min/mg, respectively. Additionally, we performed parallel pyruvate uptake and oxidation experiments with the same biological samples and show differential results in response to fasting, demonstrating the continued importance of a direct MPC activity assay. We expect this method will be of value for understanding the contribution of the MPC activity to health and disease states where pyruvate metabolism is expected to play a prominent role.


Asunto(s)
Proteínas de Transporte de Anión , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial , Ácido Pirúvico/metabolismo , Animales , Proteínas de Transporte de Anión/análisis , Proteínas de Transporte de Anión/metabolismo , Ratones , Proteínas de Transporte de Membrana Mitocondrial/análisis , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos , Ácido Pirúvico/química , Ratas , Ratas Sprague-Dawley
8.
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
9.
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
10.
Biochemistry ; 50(40): 8656-63, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21877689

RESUMEN

Helicobacter pylori's unique ability to colonize and survive in the acidic environment of the stomach is critically dependent on uptake of urea through the urea channel, HpUreI. Hence, HpUreI may represent a promising target for the development of specific drugs against this human pathogen. To obtain insight into the structure-function relationship of this channel, we developed conditions for the high-yield expression and purification of stable recombinant HpUreI. Detergent-solubilized HpUreI forms a homotrimer, as determined by chemical cross-linking. Urea dissociation kinetics of purified HpUreI were determined by means of the scintillation proximity assay, whereas urea efflux was measured in HpUreI-containing proteoliposomes using stopped-flow spectrometry to determine the kinetics and selectivity of the urea channel. The kinetic analyses revealed that urea conduction in HpUreI is pH-sensitive and saturable with a half-saturation concentration (or K(0.5)) of ~163 mM. The extent of binding of urea by HpUreI was increased at lower pH; however, the apparent affinity of urea binding (~150 mM) was not significantly pH-dependent. The solute selectivity analysis indicated that HpUreI is highly selective for urea and hydroxyurea. Removing either amino group of urea molecules diminishes their permeability through HpUreI. Similar to urea conduction, diffusion of water through HpUreI is pH-dependent with low water permeability at neutral pH.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Urea/metabolismo , Proteínas Bacterianas/genética , Transporte Biológico , Helicobacter pylori/química , Helicobacter pylori/genética , Concentración de Iones de Hidrógeno , Cinética , Proteínas de Transporte de Membrana/genética , Unión Proteica , Especificidad por Sustrato , Urea/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...