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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Circulation ; 133(8): 698-705, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26819376

RESUMO

BACKGROUND: Significant evidence indicates that the failing heart is energy starved. During the development of heart failure, the capacity of the heart to utilize fatty acids, the chief fuel, is diminished. Identification of alternate pathways for myocardial fuel oxidation could unveil novel strategies to treat heart failure. METHODS AND RESULTS: Quantitative mitochondrial proteomics was used to identify energy metabolic derangements that occur during the development of cardiac hypertrophy and heart failure in well-defined mouse models. As expected, the amounts of proteins involved in fatty acid utilization were downregulated in myocardial samples from the failing heart. Conversely, expression of ß-hydroxybutyrate dehydrogenase 1, a key enzyme in the ketone oxidation pathway, was increased in the heart failure samples. Studies of relative oxidation in an isolated heart preparation using ex vivo nuclear magnetic resonance combined with targeted quantitative myocardial metabolomic profiling using mass spectrometry revealed that the hypertrophied and failing heart shifts to oxidizing ketone bodies as a fuel source in the context of reduced capacity to oxidize fatty acids. Distinct myocardial metabolomic signatures of ketone oxidation were identified. CONCLUSIONS: These results indicate that the hypertrophied and failing heart shifts to ketone bodies as a significant fuel source for oxidative ATP production. Specific metabolite biosignatures of in vivo cardiac ketone utilization were identified. Future studies aimed at determining whether this fuel shift is adaptive or maladaptive could unveil new therapeutic strategies for heart failure.


Assuntos
Dieta Cetogênica/métodos , Ácidos Graxos/metabolismo , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Corpos Cetônicos/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica/métodos , Insuficiência Cardíaca/dietoterapia , Camundongos , Camundongos Endogâmicos C57BL
2.
Circ Res ; 114(4): 626-36, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24366168

RESUMO

RATIONALE: Increasing evidence has shown that proper control of mitochondrial dynamics (fusion and fission) is required for high-capacity ATP production in the heart. Transcriptional coactivators, peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1) α and PGC-1ß, have been shown to regulate mitochondrial biogenesis in the heart at the time of birth. The function of PGC-1 coactivators in the heart after birth has been incompletely understood. OBJECTIVE: Our aim was to assess the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts in mice. METHODS AND RESULTS: Conditional gene targeting was used in mice to explore the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts. Marked mitochondrial structural derangements were observed in hearts of PGC-1α/ß-deficient mice during postnatal growth, including fragmentation and elongation, associated with the development of a lethal cardiomyopathy. The expression of genes involved in mitochondrial fusion (Mfn1, Opa1) and fission (Drp1, Fis1) was altered in the hearts of PGC-1α/ß-deficient mice. PGC-lα was shown to directly regulate Mfn1 gene transcription by coactivating the estrogen-related receptor α on a conserved DNA element. Surprisingly, PGC-1α/ß deficiency in the adult heart did not result in evidence of abnormal mitochondrial dynamics or heart failure. However, transcriptional profiling demonstrated that PGC-1 coactivators are required for high-level expression of nuclear- and mitochondrial-encoded genes involved in mitochondrial dynamics and energy transduction in the adult heart. CONCLUSIONS: These results reveal distinct developmental stage-specific programs involved in cardiac mitochondrial dynamics.


Assuntos
Cardiomiopatias/metabolismo , Coração/crescimento & desenvolvimento , Mitocôndrias Cardíacas/metabolismo , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Cardiomiopatias/genética , Progressão da Doença , Metabolismo Energético/fisiologia , Receptor alfa de Estrogênio/metabolismo , Feminino , GTP Fosfo-Hidrolases/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética
3.
J Biol Chem ; 289(4): 2250-9, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24337569

RESUMO

The energy demands of the adult mammalian heart are met largely by ATP generated via oxidation of fatty acids in a high capacity mitochondrial system. Peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1)-α and -ß serve as inducible transcriptional coregulators of genes involved in mitochondrial biogenesis and metabolism. Whether PGC-1 plays a role in the regulation of mitochondrial structure is unknown. In this study, mice with combined deficiency of PGC-1α and PGC-1ß (PGC-1αß(-/-)) in adult heart were analyzed. PGC-1αß(-/-) hearts exhibited a distinctive mitochondrial cristae-stacking abnormality suggestive of a phospholipid abnormality as has been described in humans with genetic defects in cardiolipin (CL) synthesis (Barth syndrome). A subset of molecular species, containing n-3 polyunsaturated species in the CL, phosphatidylcholine, and phosphatidylethanolamine profiles, was reduced in PGC-1αß-deficient hearts. Gene expression profiling of PGC-1αß(-/-) hearts revealed reduced expression of the gene encoding CDP-diacylglycerol synthase 1 (Cds1), an enzyme that catalyzes the proximal step in CL biosynthesis. Cds1 gene promoter-reporter cotransfection experiments and chromatin immunoprecipitation studies demonstrated that PGC-1α coregulates estrogen-related receptors to activate the transcription of the Cds1 gene. We conclude that the PGC-1/estrogen-related receptor axis coordinately regulates metabolic and membrane structural programs relevant to the maintenance of high capacity mitochondrial function in heart.


Assuntos
Diacilglicerol Colinofosfotransferase/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Fosfatidilcolinas/biossíntese , Fosfatidiletanolaminas/biossíntese , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Animais , Síndrome de Barth/genética , Síndrome de Barth/metabolismo , Síndrome de Barth/patologia , Linhagem Celular , Diacilglicerol Colinofosfotransferase/genética , Feminino , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfatidilcolinas/genética , Fosfatidiletanolaminas/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Fatores de Transcrição/genética
4.
J Lipid Res ; 55(4): 635-44, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24395925

RESUMO

Carnitine acetyltransferase (CrAT) is a mitochondrial matrix enzyme that catalyzes the interconversion of acetyl-CoA and acetylcarnitine. Emerging evidence suggests that this enzyme functions as a positive regulator of total body glucose tolerance and muscle activity of pyruvate dehydrogenase (PDH), a mitochondrial enzyme complex that promotes glucose oxidation and is feedback inhibited by acetyl-CoA. Here, we used tandem mass spectrometry-based metabolic profiling to identify a negative relationship between CrAT activity and muscle content of lipid intermediates. CrAT specific activity was diminished in muscles from obese and diabetic rodents despite increased protein abundance. This reduction in enzyme activity was accompanied by muscle accumulation of long-chain acylcarnitines (LCACs) and acyl-CoAs and a decline in the acetylcarnitine/acetyl-CoA ratio. In vitro assays demonstrated that palmitoyl-CoA acts as a direct mixed-model inhibitor of CrAT. Similarly, in primary human myocytes grown in culture, nutritional and genetic manipulations that promoted mitochondrial influx of fatty acids resulted in accumulation of LCACs but a pronounced decrease of CrAT-derived short-chain acylcarnitines. These results suggest that lipid-induced antagonism of CrAT might contribute to decreased PDH activity and glucose disposal in the context of obesity and diabetes.


Assuntos
Carnitina O-Acetiltransferase/metabolismo , Obesidade/enzimologia , Acetilcoenzima A/metabolismo , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , Diabetes Mellitus/enzimologia , Humanos , Metabolismo dos Lipídeos , Masculino , Fibras Musculares Esqueléticas/enzimologia , Complexo Piruvato Desidrogenase/metabolismo , Ratos Wistar , Ratos Zucker
5.
Methods ; 46(4): 295-303, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18952177

RESUMO

Mitochondrial morphology and length change during fission and fusion and mitochondrial movement varies dependent upon the cell type and the physiological conditions. Here, we describe fundamental wide-field fluorescence microscopy and 3D imaging techniques to assess mitochondrial shape, number and length in various cell types including cancer cell lines, motor neurons and astrocytes. Furthermore, we illustrate how to assess mitochondrial fission and fusion events by 3D time-lapse imaging and to calculate mitochondrial length and numbers as a function of time. These imaging methods provide useful tools to investigate mitochondrial dynamics in health, aging and disease.


Assuntos
Imageamento Tridimensional/métodos , Microscopia de Fluorescência/métodos , Mitocôndrias/ultraestrutura , Animais , Astrócitos/ultraestrutura , Células HeLa , Humanos , Processamento de Imagem Assistida por Computador/métodos , Mitocôndrias/fisiologia , Tamanho Mitocondrial , Neurônios Motores/ultraestrutura , Ratos
6.
JCI Insight ; 2(1)2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26998524

RESUMO

Myocardial fuel and energy metabolic derangements contribute to the pathogenesis of heart failure. Recent evidence implicates posttranslational mechanisms in the energy metabolic disturbances that contribute to the pathogenesis of heart failure. We hypothesized that accumulation of metabolite intermediates of fuel oxidation pathways drives posttranslational modifications of mitochondrial proteins during the development of heart failure. Myocardial acetylproteomics demonstrated extensive mitochondrial protein lysine hyperacetylation in the early stages of heart failure in well-defined mouse models and the in end-stage failing human heart. To determine the functional impact of increased mitochondrial protein acetylation, we focused on succinate dehydrogenase A (SDHA), a critical component of both the tricarboxylic acid (TCA) cycle and respiratory complex II. An acetyl-mimetic mutation targeting an SDHA lysine residue shown to be hyperacetylated in the failing human heart reduced catalytic function and reduced complex II-driven respiration. These results identify alterations in mitochondrial acetyl-CoA homeostasis as a potential driver of the development of energy metabolic derangements that contribute to heart failure.

7.
Circ Heart Fail ; 7(6): 1022-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25236884

RESUMO

BACKGROUND: An unbiased systems approach was used to define energy metabolic events that occur during the pathological cardiac remodeling en route to heart failure (HF). METHODS AND RESULTS: Combined myocardial transcriptomic and metabolomic profiling were conducted in a well-defined mouse model of HF that allows comparative assessment of compensated and decompensated (HF) forms of cardiac hypertrophy because of pressure overload. The pressure overload data sets were also compared with the myocardial transcriptome and metabolome for an adaptive (physiological) form of cardiac hypertrophy because of endurance exercise training. Comparative analysis of the data sets led to the following conclusions: (1) expression of most genes involved in mitochondrial energy transduction were not significantly changed in the hypertrophied or failing heart, with the notable exception of a progressive downregulation of transcripts encoding proteins and enzymes involved in myocyte fatty acid transport and oxidation during the development of HF; (2) tissue metabolite profiles were more broadly regulated than corresponding metabolic gene regulatory changes, suggesting significant regulation at the post-transcriptional level; (3) metabolomic signatures distinguished pathological and physiological forms of cardiac hypertrophy and served as robust markers for the onset of HF; and (4) the pattern of metabolite derangements in the failing heart suggests bottlenecks of carbon substrate flux into the Krebs cycle. CONCLUSIONS: Mitochondrial energy metabolic derangements that occur during the early development of pressure overload-induced HF involve both transcriptional and post-transcriptional events. A subset of the myocardial metabolomic profile robustly distinguished pathological and physiological cardiac remodeling.


Assuntos
Cardiomegalia/fisiopatologia , Metabolismo Energético/fisiologia , Insuficiência Cardíaca/fisiopatologia , Remodelação Ventricular/fisiologia , Aminoácidos/metabolismo , Animais , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Metabolismo Energético/genética , Perfilação da Expressão Gênica , Insuficiência Cardíaca/metabolismo , Metabolismo dos Lipídeos/genética , Metaboloma , Camundongos , Regulação para Cima/fisiologia
8.
J Clin Endocrinol Metab ; 95(7): 3400-10, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20427507

RESUMO

CONTEXT: Intracellular lipid partitioning toward storage and the incomplete oxidation of fatty acids (FA) have been linked to insulin resistance. OBJECTIVE: To gain insight into how intracellular lipid metabolism is related to insulin signal transduction, we examined the effects of severe obesity, excess FA, and overexpression of the FA transporter, FA translocase (FAT)/CD36, in primary human skeletal myocytes. DESIGN, SETTING, AND PATIENTS: Insulin signal transduction, FA oxidation, and metabolism were measured in skeletal muscle cells harvested from lean and severely obese women. To emulate the obesity phenotype in our cell culture system, we incubated cells from lean individuals with excess FA or overexpressed FAT/CD36 using recombinant adenoviral technology. RESULTS: Complete oxidation of FA was significantly reduced, whereas total lipid accumulation, FA esterification into lipid intermediates, and incomplete oxidation were up-regulated in the muscle cells of severely obese subjects. Insulin signal transduction was reduced in the muscle cells from severely obese subjects compared to lean controls. Incubation of muscle cells from lean subjects with lipids reduced insulin signal transduction and increased lipid storage and incomplete FA oxidation. CD36 overexpression increased FA transport capacity, but did not impair complete FA oxidation and insulin signal transduction in muscle cells from lean subjects. CONCLUSIONS: Cultured myocytes from severely obese women express perturbations in FA metabolism and insulin signaling reminiscent of those observed in vivo. The obesity phenotype can be recapitulated in muscle cells from lean subjects via exposure to excess lipid, but not by overexpressing the FAT/CD36 FA transporter.


Assuntos
Ácidos Graxos/metabolismo , Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Obesidade Mórbida/metabolismo , Adulto , Análise de Variância , Western Blotting , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Proteínas de Transporte de Ácido Graxo/genética , Proteínas de Transporte de Ácido Graxo/metabolismo , Feminino , Humanos , Resistência à Insulina , Obesidade Mórbida/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
9.
Inorg Chem ; 47(9): 3584-93, 2008 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-18399627

RESUMO

In view of the biological and commercial interest in models for Oxalate Decarboxylases (OxDC) and Oxalate Oxidases (OxOx), we have synthesized and characterized three new Mn (II) complexes ( 1- 3) employing N3O-donor amino-carboxylate ligands (TCMA, 1,4,7-triazacyclononane- N-acetic acid; K (i) Pr 2TCMA, potassium 1,4-diisopropyl-1,4,7-triazacyclononane- N-acetate; and KBPZG, potassium N,N-bis(3,5-dimethylpyrazolyl methyl)glycinate). These complexes were characterized by several techniques including X-ray crystallographic analysis, X-band electron paramagnetic resonance (EPR), electrospray ionization mass spectrometry (ESI-MS), and cyclic voltammetry. The crystal structures of 1 and 3 revealed that both form infinite polymeric chains of Mn (II) complexes linked by the pendant carboxylate arms of the TCMA (-) and the BPZG (-) ligands in a syn-antipattern. Complex 2 crystallizes as a mononuclear Mn (II) cation, six-coordinate in a distorted octahedral geometry. Although complexes 1 and 3 crystallize as polymeric chains, all compounds present the same N3O-donor set atoms around the metal center as observed in the crystallographically characterized OxDC and OxOx. Moreover, complex 2 also contains two water molecules coordinated to the Mn center as observed in the active site of OxDC and OxOx. ESI-MS spectrometry, combined with EPR, were useful techniques to establish that complexes 1- 3 are present as mononuclear Mn (II) species in solution. Finally, complexes 1- 3 are able to model the resting state active sites, with special attention focused on complex 2 which provides the first exact first coordination sphere ligand structural model for the resting states of both OxDC and OxOx.


Assuntos
Carboxiliases/química , Oxirredutases/química , Bacillus subtilis/enzimologia , Sítios de Ligação , Cristalografia por Raios X , Eletroquímica , Espectroscopia de Ressonância de Spin Eletrônica , Compostos Heterocíclicos/química , Hordeum/enzimologia , Manganês/química , Modelos Moleculares , Compostos Organometálicos/química , Espectrometria de Massas por Ionização por Electrospray , Thermotoga maritima/enzimologia
10.
J Biol Chem ; 281(1): 484-90, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16269413

RESUMO

The GLUT4 glucose transporter is predominantly retained inside basal fat and muscle cells, and it is rapidly recruited to the plasma membrane with insulin stimulation. There is controversy regarding the mechanism of basal GLUT4 retention. One model is that GLUT4 retention is dynamic, based on slow exocytosis and rapid internalization of the entire pool of GLUT4 (Karylowski, O., Zeigerer, A., Cohen, A., and McGraw, T. E. (2004) Mol. Biol. Cell 15, 870-882). In this model, insulin increases GLUT4 in the plasma membrane by modulating GLUT4 exocytosis and endocytosis. The second model is that GLUT4 retention is static, with approximately 90% of GLUT4 stored in compartments that are not in equilibrium with the cell surface in basal conditions (Govers, R., Coster, A. C., and James, D. E. (2004) Mol. Cell Biol. 24, 6456-6466). In this model, insulin increases GLUT4 in the plasma membrane by releasing it from the static storage compartment. Here we show that under all experimental conditions examined, basal GLUT4 retention is by a bipartite dynamic mechanism involving slow efflux and rapid internalization. To establish that the dynamic model developed in studies of the extreme conditions of >100 nm insulin and no insulin also describes GLUT4 behavior at more physiological insulin concentrations, we characterized GLUT4 trafficking in 0.5 nm insulin. This submaximal insulin concentration promotes an intermediate effect on both GLUT4 exocytosis and endocytosis, resulting in an intermediate degree of redistribution to the plasma membrane. These data establish that changes in the steady-state surface/total distributions of GLUT4 are the result of gradated, insulin-induced changes in GLUT4 exocytosis and endocytosis rates.


Assuntos
Adipócitos/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Transporte Proteico/efeitos dos fármacos , Células 3T3-L1 , Adipócitos/citologia , Animais , Membrana Celular/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Camundongos , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA