Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
FASEB J ; 38(11): e23709, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38809700

RESUMEN

Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of ß-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-ß1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.


Asunto(s)
Tejido Adiposo Pardo , Fibrosis , Proteína Desacopladora 1 , Animales , Tejido Adiposo Pardo/metabolismo , Ratones , Masculino , Proteína Desacopladora 1/metabolismo , Fibrosis/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Ratones Endogámicos C57BL , Cardiomegalia/metabolismo , Cardiomegalia/patología , Miocardio/metabolismo , Miocardio/patología , Estrés Fisiológico , Remodelación Ventricular/fisiología , Ratones Noqueados , Frío
2.
J Mol Cell Cardiol ; 182: 15-24, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37451081

RESUMEN

Cardiac function is a dynamic process that must adjust efficiently to the immediate demands of physical state and activity. So too, the metabolic support of cardiac function is a dynamic process that must respond, in time, to the demands of cardiac function and viability. Flux through metabolic pathways provides chemical energy and generates signaling molecules that regulate activity among intracellular compartments to meet these demands. Thus, flux through metabolic pathways provides a dynamic mode of support of cardiomyocytes during physiological and pathophysiological challenges. Any inability of metabolic flux to keep pace with the demands of the cardiomyocyte results in progressive dysfunction that contributes to cardiac disease. Thus, the priority in maintaining and regulating flux through metabolic pathways in the cardiomyocyte cannot be understated. Great potential exists in current efforts to elucidate metabolic mechanisms as therapeutic targets for the diseased heart. As a consequence, detecting metabolic flux in the functioning myocardium of the heart, under normal and diseased conditions, is essential in elucidating the metabolic basis of contractile dysfunction. As a companion to the 2022 ISHR Research Achievement Award lecture, this review examines the use and applications of stable isotope kinetics to quantify metabolic flux through intermediary pathways and the exchange and transport of intermediates across the mitochondrial membrane and sarcolemma of intact functioning hearts in determining how these intracellular events are coordinated to support cardiac function and health. Finally, this work reviews recently demonstrated metabolic defects in diseased hearts and the potential for metabolic alleviation of heart disease.


Asunto(s)
Cardiopatías , Miocardio , Humanos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Cardiopatías/etiología , Cardiopatías/metabolismo , Redes y Vías Metabólicas , Cinética , Metabolismo Energético/fisiología
3.
Circulation ; 143(18): 1797-1808, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33601938

RESUMEN

BACKGROUND: The failing heart is energy starved with impaired oxidation of long-chain fatty acids (LCFAs) at the level of reduced CPT1 (carnitine palmitoyltransferase 1) activity at the outer mitochondrial membrane. Recent work shows elevated ketone oxidation in failing hearts as an alternate carbon source for oxidative ATP generation. We hypothesized that another short-chain carbon source, short-chain fatty acids (SCFAs) that bypass carnitine palmitoyltransferase 1, could similarly support energy production in failing hearts. METHODS: Cardiac hypertrophy and dysfunction were induced in rats by transverse-aortic constriction (TAC). Fourteen weeks after TAC or sham operation, isolated hearts were perfused with either the 4 carbon, 13C-labeled ketone (D3-hydroxybutyrate) or the 4 carbon, 13C-labeled SCFA butyrate in the presence of glucose and the LCFA palmitate. Oxidation of ketone and SCFA was compared by in vitro 13C nuclear magnetic resonance spectroscopy, as was the capacity for short-chain carbon sources to compensate for impaired LCFA oxidation in the hypertrophic heart. Adaptive changes in enzyme expression and content for the distinct pathways of ketone and SCFA oxidation were examined in both failing rat and human hearts. RESULTS: TAC produced pathological hypertrophy and increased the fractional contributions of ketone to acetyl coenzyme-A production in the tricarboxylic acid cycle (0.60±0.02 sham ketone versus 0.70±0.02 TAC ketone; P<0.05). However, butyrate oxidation in failing hearts was 15% greater (0.803±0.020 TAC SCFA) than ketone oxidation. SCFA was also more readily oxidized than ketone in sham hearts by 15% (0.693±0.020 sham SCFA). Despite greater SFCA oxidation, TAC did not change short-chain acyl coenzyme-A dehydrogenase content. However, failing hearts of humans and the rat model both contain significant increases in acyl coenzyme-A synthetase medium-chain 3 enzyme gene expression and protein content. The increased oxidation of SCFA and ketones occurred at the expense of LCFA oxidation, with LCFA contributing less to acetyl coenzyme-A production in failing hearts perfused with SCFA (0.190±0.012 TAC SCFA versus 0.3163±0.0360 TAC ketone). CONCLUSIONS: SCFAs are more readily oxidized than ketones in failing hearts, despite both bypassing reduced CPT1 activity and represent an unexplored carbon source for energy production in failing hearts.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Insuficiencia Cardíaca/fisiopatología , Cetonas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
4.
Circulation ; 143(2): 145-159, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106031

RESUMEN

BACKGROUND: Brown adipose tissue (BAT) is an important tissue for thermogenesis, making it a potential target to decrease the risks of obesity, type 2 diabetes, and cardiovascular disease, and recent studies have also identified BAT as an endocrine organ. Although BAT has been implicated to be protective in cardiovascular disease, to this point there are no studies that identify a direct role for BAT to mediate cardiac function. METHODS: To determine the role of BAT on cardiac function, we utilized a model of BAT transplantation. We then performed lipidomics and identified an increase in the lipokine 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME). We utilized a mouse model with sustained overexpression of 12,13-diHOME and investigated the role of 12,13-diHOME in a nitric oxide synthase type 1 deficient (NOS1-/-) mouse and in isolated cardiomyocytes to determine effects on function and respiration. We also investigated 12,13-diHOME in a cohort of human patients with heart disease. RESULTS: Here, we determined that transplantation of BAT (+BAT) improves cardiac function via the release of the lipokine 12,13-diHOME. Sustained overexpression of 12,13-diHOME using tissue nanotransfection negated the deleterious effects of a high-fat diet on cardiac function and remodeling, and acute injection of 12,13-diHOME increased cardiac hemodynamics via direct effects on the cardiomyocyte. Furthermore, incubation of cardiomyocytes with 12,13-diHOME increased mitochondrial respiration. The effects of 12,13-diHOME were absent in NOS1-/- mice and cardiomyocytes. We also provide the first evidence that 12,13-diHOME is decreased in human patients with heart disease. CONCLUSIONS: Our results identify an endocrine role for BAT to enhance cardiac function that is mediated by regulation of calcium cycling via 12,13-diHOME and NOS1.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/trasplante , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , Lipidómica/métodos , Ácidos Oléicos/metabolismo , Anciano , Animales , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ácidos Oléicos/administración & dosificación , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/fisiología
5.
Circulation ; 139(24): 2765-2777, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30909726

RESUMEN

BACKGROUND: Metabolic remodeling in heart failure contributes to dysfunctional lipid trafficking and lipotoxicity. Acyl coenzyme A synthetase-1 (ACSL1) facilitates long-chain fatty acid (LCFA) uptake and activation with coenzyme A (CoA), mediating the fate of LCFA. The authors tested whether cardiac ACSL1 overexpression aids LCFA oxidation and reduces lipotoxicity under pathological stress of transverse aortic constriction (TAC). METHODS: Mice with cardiac restricted ACSL1 overexpression (MHC-ACSL1) underwent TAC or sham surgery followed by serial in vivo echocardiography for 14 weeks. At the decompensated stage of hypertrophy, isolated hearts were perfused with 13C LCFA during dynamic-mode 13C nuclear magnetic resonance followed by in vitro nuclear magnetic resonance and mass spectrometry analysis to assess intramyocardial lipid trafficking. In parallel, acyl CoA was measured in tissue obtained from heart failure patients pre- and postleft ventricular device implantation plus matched controls. RESULTS: TAC-induced cardiac hypertrophy and dysfunction was mitigated in MHC-ACSL1 hearts compared with nontransgenic hearts. At 14 weeks, TAC increased heart weight to tibia length by 46% in nontransgenic mice, but only 26% in MHC-ACSL1 mice, whereas ACSL1 mice retained greater ejection fraction (ACSL1 TAC: 65.8±7.5%; nontransgenic TAC: 45.9±7.3) and improvement in diastolic E/E'. Functional improvements were mediated by ACSL1 changes to cardiac LCFA trafficking. ACSL1 accelerated LCFA uptake, preventing C16 acyl CoA loss post-TAC. Long-chain acyl CoA was similarly reduced in human failing myocardium and restored to control levels by mechanical unloading. ACSL1 trafficked LCFA into ceramides without normalizing the reduced triglyceride storage in TAC. ACSL1 prevented de novo synthesis of cardiotoxic C16- and C24-, and C24:1 ceramides and increased potentially cardioprotective C20- and C22-ceramides post-TAC. ACLS1 overexpression activated AMP activated protein kinase at baseline, but during TAC, prevented the reduced LCFA oxidation in hypertrophic hearts and normalized energy state (phosphocreatine:ATP) and consequently, AMP activated protein kinase activation. CONCLUSIONS: This is the first demonstration of reduced acyl CoA in failing hearts of humans and mice, and suggests possible mechanisms for maintaining mitochondrial oxidative energy metabolism by restoring long-chain acyl CoA through ASCL1 activation and mechanical unloading. By mitigating cardiac lipotoxicity, via redirected LCFA trafficking to ceramides, and restoring acyl CoA, ACSL1 delayed progressive cardiac remodeling and failure.


Asunto(s)
Acilcoenzima A/metabolismo , Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Metabolismo de los Lípidos , Miocardio/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Proteínas Quinasas Activadas por AMP/metabolismo , Anciano , Animales , Transporte Biológico , Ceramidas/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Femenino , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Miocardio/patología , Oxidación-Reducción , Triglicéridos/metabolismo , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
6.
Circulation ; 140(14): 1205-1216, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769940

RESUMEN

Mitochondria have emerged as a central factor in the pathogenesis and progression of heart failure, and other cardiovascular diseases, as well, but no therapies are available to treat mitochondrial dysfunction. The National Heart, Lung, and Blood Institute convened a group of leading experts in heart failure, cardiovascular diseases, and mitochondria research in August 2018. These experts reviewed the current state of science and identified key gaps and opportunities in basic, translational, and clinical research focusing on the potential of mitochondria-based therapeutic strategies in heart failure. The workshop provided short- and long-term recommendations for moving the field toward clinical strategies for the prevention and treatment of heart failure and cardiovascular diseases by using mitochondria-based approaches.


Asunto(s)
Sistema Cardiovascular , Educación/métodos , Insuficiencia Cardíaca/terapia , Mitocondrias/fisiología , National Heart, Lung, and Blood Institute (U.S.) , Informe de Investigación , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Sistema Cardiovascular/patología , Educación/tendencias , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/epidemiología , Humanos , National Heart, Lung, and Blood Institute (U.S.)/tendencias , Informe de Investigación/tendencias , Investigación Biomédica Traslacional/métodos , Investigación Biomédica Traslacional/tendencias , Estados Unidos/epidemiología
7.
Circ Res ; 122(6): 836-845, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29386187

RESUMEN

RATIONALE: Metabolic remodeling in hypertrophic hearts includes inefficient glucose oxidation via increased anaplerosis fueled by pyruvate carboxylation. Pyruvate carboxylation to malate through elevated ME1 (malic enzyme 1) consumes NADPH necessary for reduction of glutathione and maintenance of intracellular redox state. OBJECTIVE: To elucidate upregulated ME1 as a potential maladaptive mechanism for inefficient glucose oxidation and compromised redox state in hypertrophied hearts. METHODS AND RESULTS: ME1 expression was selectively inhibited, in vivo, via non-native miR-ME1 (miRNA specific to ME1) in pressure-overloaded rat hearts. Rats subjected to transverse aortic constriction (TAC) or Sham surgery received either miR-ME1 or PBS. Effects of ME1 suppression on anaplerosis and reduced glutathione (GSH) content were studied in isolated hearts supplied 13C-enriched substrate: palmitate, glucose, and lactate. Human myocardium collected from failing and nonfailing hearts during surgery enabled RT-qPCR confirmation of elevated ME1 gene expression in clinical heart failure versus nonfailing human hearts (P<0.04). TAC induced elevated ME1 content, but ME1 was lowered in hearts infused with miR-ME1 versus PBS. Although Sham miR-ME1 hearts showed no further reduction of inherently low anaplerosis in normal heart, miR-ME1 reduced anaplerosis in TAC to baseline: TAC miR-ME1=0.034±0.004; TAC PBS=0.081±0.005 (P<0.01). Countering elevated anaplerosis in TAC shifted pyruvate toward oxidation in the tricarboxylic acid cycle. Importantly, via the link to NADPH consumption by pyruvate carboxylation, ME1 suppression in TAC restored GSH content, reduced lactate production, and ultimately improved contractility. CONCLUSIONS: A maladaptive increase in anaplerosis via ME1 in TAC is associated with reduced GSH content. Suppressing increased ME1 expression in hypertrophied rat hearts, which is also elevated in failing human hearts, reduced pyruvate carboxylation thereby normalizing anaplerosis, restoring GSH content, and reducing lactate accumulation. Reducing ME1 induced favorable metabolic shifts for carbohydrate oxidation, improving intracellular redox state and enhanced cardiac performance in pathological hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Glucosa/metabolismo , Malato Deshidrogenasa/metabolismo , Anciano , Animales , Glutatión/metabolismo , Humanos , Malato Deshidrogenasa/genética , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Miocardio/metabolismo , NADP/metabolismo , Oxidación-Reducción , Ácido Pirúvico/metabolismo , Ratas , Ratas Sprague-Dawley
8.
Circulation ; 138(3): 305-315, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30012703

RESUMEN

Lipid droplets (LDs) are distinct and dynamic organelles that affect the health of cells and organs. Much progress has been made in understanding how these structures are formed, how they interact with other cellular organelles, how they are used for storage of triacylglycerol in adipose tissue, and how they regulate lipolysis. Our understanding of the biology of LDs in the heart and vascular tissue is relatively primitive in comparison with LDs in adipose tissue and liver. The National Heart, Lung, and Blood Institute convened a working group to discuss how LDs affect cardiovascular diseases. The goal of the working group was to examine the current state of knowledge on the cell biology of LDs, including current methods to study them in cells and organs and reflect on how LDs influence the development and progression of cardiovascular diseases. This review summarizes the working group discussion and recommendations on research areas ripe for future investigation that will likely improve our understanding of atherosclerosis and heart function.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Gotas Lipídicas/metabolismo , Miocardio/metabolismo , Animales , Enfermedades Cardiovasculares/genética , Consensus Development Conferences, NIH as Topic , Modelos Animales de Enfermedad , Interacción Gen-Ambiente , Humanos , Metabolismo de los Lípidos , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos
9.
Circ Res ; 118(10): 1659-701, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-27012580

RESUMEN

In a complex system of interrelated reactions, the heart converts chemical energy to mechanical energy. Energy transfer is achieved through coordinated activation of enzymes, ion channels, and contractile elements, as well as structural and membrane proteins. The heart's needs for energy are difficult to overestimate. At a time when the cardiovascular research community is discovering a plethora of new molecular methods to assess cardiac metabolism, the methods remain scattered in the literature. The present statement on "Assessing Cardiac Metabolism" seeks to provide a collective and curated resource on methods and models used to investigate established and emerging aspects of cardiac metabolism. Some of those methods are refinements of classic biochemical tools, whereas most others are recent additions from the powerful tools of molecular biology. The aim of this statement is to be useful to many and to do justice to a dynamic field of great complexity.


Asunto(s)
American Heart Association , Técnicas de Imagen Cardíaca/métodos , Enfermedades Cardiovasculares/metabolismo , Biología Computacional/métodos , Miocardio/metabolismo , Animales , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/genética , Humanos , Estados Unidos
10.
Biochim Biophys Acta ; 1861(10): 1492-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26993578

RESUMEN

No longer regarded as physiologically inert the endogenous triacylglyceride (TAG) pool within the cardiomyocyte is now recognized to play a dynamic role in metabolic regulation. Beyond static measures of content, the relative rates of interconversion among acyl intermediates are more closely linked to dynamic processes of physiological function in normal and diseased hearts, with the potential for both adaptive and maladaptive contributions. Indeed, multiple inefficiencies in cardiac metabolism have been identified in the decompensated, hypertrophied and failing heart. Among the intracellular responses to physiological, metabolic and pathological stresses, TAG plays a central role in the balance of lipid handling and signaling mechanisms. TAG dynamics are profoundly altered from normal in both diabetic and pathologically stressed hearts. More than just expansion or contraction of the stored lipid pool, the turnover rates of TAG are sensitive to and compete against other enzymatic pathways, anabolic and catabolic, for reactive acyl-CoA units. The rates of TAG synthesis and lipolysis thusly affect multiple components of cardiomyocyte function, including energy metabolism, cell signaling, and enzyme activation, as well as the regulation of gene expression in both normal and diseased states. This review examines the multiple etiologies and metabolic consequences of the failing heart and the central role of lipid storage dynamics in the pathogenic process. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Triglicéridos/metabolismo , Animales , Ácidos Grasos/metabolismo , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción , Transducción de Señal
11.
Circulation ; 133(8): 698-705, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26819376

RESUMEN

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.


Asunto(s)
Dieta Cetogénica/métodos , Ácidos Grasos/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Cuerpos Cetónicos/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/dietoterapia , Ratones , Ratones Endogámicos C57BL
12.
Am J Physiol Heart Circ Physiol ; 312(4): H681-H690, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130336

RESUMEN

Although alterations in fatty acid (FA) metabolism have been shown to have a negative impact on contractility of the hypertrophied heart, the targets of action remain elusive. In this study we compared the function of skinned fiber bundles from transgenic (Tg) mice that overexpress a relatively low level of the peroxisome proliferator-activated receptor α (PPARα), and nontransgenic (NTg) littermates. The mice (NTg-T and Tg-T) were stressed by transverse aortic constriction (TAC) and compared with shams (NTg-S and Tg-S). There was an approximate 4-fold increase in PPARα expression in Tg-S compared with NTg-S, but Tg-T hearts showed the same PPARα expression as NTg-T. Expression of PPARα did not alter the hypertrophic response to TAC but did reduce ejection fraction (EF) in Tg-T hearts compared with other groups. The rate of actomyosin ATP hydrolysis was significantly higher in Tg-S skinned fiber bundles compared with all other groups. Tg-T hearts showed an increase in phosphorylation of specific sites on cardiac myosin binding protein-C (cMyBP-C) and ß-myosin heavy chain isoform. These results advance our understanding of potential signaling to the myofilaments induced by altered FA metabolism under normal and pathological states. We demonstrate that chronic and transient PPARα activation during pathological stress alters myofilament response to Ca2+ through a mechanism that is possibly mediated by MyBP-C phosphorylation and myosin heavy chain isoforms.NEW & NOTEWORTHY Data presented here demonstrate novel signaling to sarcomeric proteins by chronic alterations in fatty acid metabolism induced by PPARα. The mechanism involves modifications of key myofilament regulatory proteins modifying cross-bridge dynamics with differential effects in controls and hearts stressed by pressure overload.


Asunto(s)
Cardiomegalia/fisiopatología , Miofibrillas , PPAR alfa/biosíntesis , PPAR alfa/genética , Adenosina Trifosfatasas/metabolismo , Animales , Señalización del Calcio/genética , Cardiomegalia/etiología , Proteínas Portadoras/metabolismo , Ácidos Grasos/metabolismo , Corazón/fisiopatología , Hipertensión/complicaciones , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Miocardio/citología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fosforilación , Volumen Sistólico
13.
J Mol Cell Cardiol ; 94: 1-9, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26995156

RESUMEN

RATIONALE: Acyl CoA synthetase-1 (ACSL1) is localized at intracellular membranes, notably the mitochondrial membrane. ACSL1 and female sex are suggested to indirectly facilitate lipid availability to the heart and other organs. However, such mechanisms in intact, functioning myocardium remain unexplored, and roles of ACSL1 and sex in the uptake and trafficking of fats are poorly understood. OBJECTIVE: To determine the potential for ACSL1 and sex-dependent differences in metabolic trapping and trafficking effects of long-chain fatty acids (LCFA) within cardiomyocytes of intact hearts. METHODS AND RESULTS: (13)C NMR of intact, beating mouse hearts, supplied (13)C palmitate, revealed 44% faster trans-sarcolemmal uptake of LCFA in male hearts overexpressing ACSL1 (MHC-ACSL1) than in non-transgenic (NTG) males (p<0.05). Acyl CoA content was elevated by ACSL1 overexpression, 404% in males and 164% in female, relative to NTG. Despite similar ACSL1 content, NTG females displayed faster LCFA uptake kinetics compared to NTG males, which was reversed by ovariectomy. NTG female LCFA uptake rates were similar to those in ACSL1 males and ACSL1 females. ACSL1 and female sex hormones both accelerated LCFA uptake without affecting triglyceride content or turnover. ACSL1 hearts contained elevated ceramide, particularly C22 ceramide in both sexes and specifically, C24 in males. ACSL1 also induced lower content of fatty acid transporter-6 (FATP6) indicating cooperative regulation with ACSL1. Surprisingly, ACSL1 overexpression did not increase mitochondrial oxidation of exogenous palmitate, which actually dropped in female ACSL1 hearts. CONCLUSIONS: ACSL1-mediated metabolic trapping of exogenous LCFA accelerates LCFA uptake rates, albeit to a lesser extent in females, which distinctly affects LCFA trafficking to acyl intermediates but not triglyceride storage or mitochondrial oxidation and is affected by female sex hormones.


Asunto(s)
Coenzima A Ligasas/metabolismo , Metabolismo Energético , Ácidos Grasos/metabolismo , Miocardio/metabolismo , Animales , Transporte Biológico , Membrana Celular/metabolismo , Coenzima A Ligasas/genética , Femenino , Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Pruebas de Función Cardíaca , Hemodinámica , Metabolismo de los Lípidos , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Oxidación-Reducción , Sarcómeros/metabolismo , Factores Sexuales
15.
Magn Reson Med ; 76(6): 1879-1886, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26749052

RESUMEN

PURPOSE: Many cardiovascular diseases are associated with abnormal function of myocardial contractility or dilatability, which is related to elasticity changes of the myocardium over the cardiac cycle. The mouse is a common animal model in studies of the progression of various cardiomyopathies. We introduce a novel noninvasive approach using microscopic scale MR elastography (MRE) to measure the myocardium stiffness change during the cardiac cycle on a mouse model. METHODS: A harmonic mechanical wave of 400 Hz was introduced into the mouse body. An electrocardiograph-gated and respiratory-gated fractional encoding cine-MRE pulse sequence was applied to encode the resulting oscillatory motion on a short-axis slice of the heart. Five healthy mice (age range, 3-13.5 mo) were examined. The weighted summation effective stiffness of the left ventricle wall during the cardiac cycle was estimated. RESULTS: The ratio of stiffness at end diastole and end systole was 0.5-0.67. Additionally, variation in shear wave amplitude in the left ventricle wall throughout the cardiac cycle was measured and found to correlate with estimates of stiffness variation. CONCLUSION: This study demonstrates the feasibility of implementing cardiac MRE on a mouse model. Magn Reson Med 76:1879-1886, 2016. © 2016 International Society for Magnetic Resonance in Medicine.


Asunto(s)
Técnicas de Imagen Cardíaca/instrumentación , Técnicas de Imagen Cardíaca/veterinaria , Diagnóstico por Imagen de Elasticidad/instrumentación , Diagnóstico por Imagen de Elasticidad/veterinaria , Imagen por Resonancia Cinemagnética/instrumentación , Imagen por Resonancia Cinemagnética/veterinaria , Función Ventricular Izquierda/fisiología , Animales , Técnicas de Imagen Cardíaca/métodos , Módulo de Elasticidad/fisiología , Diagnóstico por Imagen de Elasticidad/métodos , Diseño de Equipo , Análisis de Falla de Equipo , Estudios de Factibilidad , Femenino , Ventrículos Cardíacos/anatomía & histología , Imagen por Resonancia Cinemagnética/métodos , Ratones , Ratones Endogámicos C57BL , Proyectos Piloto , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
16.
Circ Res ; 114(4): 717-29, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24526677

RESUMEN

Metabolic signaling mechanisms are increasingly recognized to mediate the cellular response to alterations in workload demand, as a consequence of physiological and pathophysiological challenges. Thus, an understanding of the metabolic mechanisms coordinating activity in the cytosol with the energy-providing pathways in the mitochondrial matrix becomes critical for deepening our insights into the pathogenic changes that occur in the stressed cardiomyocyte. Processes that exchange both metabolic intermediates and cations between the cytosol and mitochondria enable transduction of dynamic changes in contractile state to the mitochondrial compartment of the cell. Disruption of such metabolic transduction pathways has severe consequences for the energetic support of contractile function in the heart and is implicated in the pathogenesis of heart failure. Deficiencies in metabolic reserve and impaired metabolic transduction in the cardiomyocyte can result from inherent deficiencies in metabolic phenotype or maladaptive changes in metabolic enzyme expression and regulation in the response to pathogenic stress. This review examines both current and emerging concepts of the functional linkage between the cytosol and the mitochondrial matrix with a specific focus on metabolic reserve and energetic efficiency. These principles of exchange and transport mechanisms across the mitochondrial membrane are reviewed for the failing heart from the perspectives of chronic pressure overload and diabetes mellitus.


Asunto(s)
Metabolismo Energético/fisiología , Insuficiencia Cardíaca/metabolismo , Corazón/fisiología , Mitocondrias/metabolismo , Miocardio/metabolismo , Animales , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Mitocondrias/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
17.
J Mol Cell Cardiol ; 79: 13-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450611

RESUMEN

Diabetic hearts are subject to more extensive ischemia/reperfusion (ISC/REP) damage. This study examined the efficiency of citric acid cycle (CAC) flux and the transfer of cytosolic reducing equivalents into the mitochondria for oxidative support of cardiac work following ISC/REP in hearts of c57bl/6 (NORM) and type 2 diabetic, db/db mouse hearts. Flux through the CAC and malate-aspartate shuttle (MA) were monitored via dynamic (13)C NMR of isolated hearts perfused with (13)C palmitate+glucose. MA flux was lower in db/db than NORM. Oxoglutarate malate carrier (OMC) was elevated in the db/db heart, suggesting a compensatory response to low NADHc. Baseline CAC flux per unit work (rate-pressure-product, RPP) was similar between NORM and db/db, but ISC/REP reduced the efficiency of CAC flux/RPP by 20% in db/db. ISC/REP also increased UCP3 transcription, indicating potential for greater uncoupling. Therefore, ISC/REP induces inefficient carbon utilization through the CAC in hearts of diabetic mice due to the combined inefficiencies in NADHc transfer per OMC content and increased uncoupling via UCP3. Ischemia and reperfusion exacerbated pre-existing mitochondrial defects and metabolic limitations in the cytosol of diabetic hearts. These limitations and defects render diabetic hearts more susceptible to inefficient carbon fuel utilization for oxidative energy metabolism.


Asunto(s)
Ciclo del Ácido Cítrico , Citosol/metabolismo , Diabetes Mellitus Experimental/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Isquemia Miocárdica/metabolismo , NAD/metabolismo , Animales , Ácido Aspártico/metabolismo , Espectroscopía de Resonancia Magnética con Carbono-13 , Proteínas Portadoras/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Hemodinámica , Malatos/metabolismo , Masculino , Ratones Endogámicos C57BL , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Oxidación-Reducción , PPAR alfa/metabolismo , Perfusión , Proteína Desacopladora 2 , Proteína Desacopladora 3
18.
Circulation ; 130(20): 1790-9, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25266948

RESUMEN

BACKGROUND: Intramyocardial triglyceride (TG) turnover is reduced in pressure-overloaded, failing hearts, limiting the availability of this rich source of long-chain fatty acids for mitochondrial ß-oxidation and nuclear receptor activation. This study explored 2 major dietary fats, palmitate and oleate, in supporting endogenous TG dynamics and peroxisome proliferator-activated receptor-α activation in sham-operated (SHAM) and hypertrophied (transverse aortic constriction [TAC]) rat hearts. METHODS AND RESULTS: Isolated SHAM and TAC hearts were provided media containing carbohydrate with either (13)C-palmitate or (13)C-oleate for dynamic (13)C nuclear magnetic resonance spectroscopy and end point liquid chromatography/mass spectrometry of TG dynamics. With palmitate, TAC hearts contained 48% less TG versus SHAM (P=0.0003), whereas oleate maintained elevated TG in TAC, similar to SHAM. TG turnover in TAC was greatly reduced with palmitate (TAC, 46.7±12.2 nmol/g dry weight per min; SHAM, 84.3±4.9; P=0.0212), as was ß-oxidation of TG. Oleate elevated TG turnover in both TAC (140.4±11.2) and SHAM (143.9±15.6), restoring TG oxidation in TAC. Peroxisome proliferator-activated receptor-α target gene transcripts were reduced by 70% in TAC with palmitate, whereas oleate induced normal transcript levels. Additionally, mRNA levels for peroxisome proliferator-activated receptor-γ-coactivator-1α and peroxisome proliferator-activated receptor-γ-coactivator-1ß in TAC hearts were maintained by oleate. With these metabolic effects, oleate also supported a 25% improvement in contractility over palmitate with TAC (P=0.0202). CONCLUSIONS: The findings link reduced intracellular lipid storage dynamics to impaired peroxisome proliferator-activated receptor-α signaling and contractility in diseased hearts, consistent with a rate-dependent lipolytic activation of peroxisome proliferator-activated receptor-α. In decompensated hearts, oleate may serve as a beneficial energy substrate versus palmitate by upregulating TG dynamics and nuclear receptor signaling.


Asunto(s)
Grasas de la Dieta/farmacología , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Ácido Oléico/farmacología , PPAR alfa/fisiología , Palmitatos/farmacología , Triglicéridos/metabolismo , Animales , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/metabolismo , Núcleo Celular/metabolismo , Ceramidas/análisis , Ciclo del Ácido Cítrico , Grasas de la Dieta/farmacocinética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/dietoterapia , Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/metabolismo , Lipólisis , Masculino , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Resonancia Magnética Nuclear Biomolecular , Ácido Oléico/administración & dosificación , Ácido Oléico/farmacocinética , Oxidación-Reducción , Palmitatos/administración & dosificación , Palmitatos/farmacocinética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transcripción Genética
20.
Am J Physiol Heart Circ Physiol ; 308(11): H1414-22, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25795713

RESUMEN

Sudden cardiac arrest (SCA) is a leading cause of death in the United States. Despite return of spontaneous circulation, patients die due to post-SCA syndrome that includes myocardial dysfunction, brain injury, impaired metabolism, and inflammation. No medications improve SCA survival. Our prior work suggests that optimal Akt activation is critical for cooling protection and SCA recovery. Here, we investigate a small inhibitor of PTEN, an Akt-related phosphatase present in heart and brain, as a potential therapy in improving cardiac and neurological recovery after SCA. Anesthetized adult female wild-type C57BL/6 mice were randomized to pretreatment of VO-OHpic (VO) 30 min before SCA or vehicle control. Mice underwent 8 min of KCl-induced asystolic arrest followed by CPR. Resuscitated animals were hemodynamically monitored for 2 h and observed for 72 h. Outcomes included heart pressure-volume loops, energetics (phosphocreatine and ATP from (31)P NMR), protein phosphorylation of Akt, GSK3ß, pyruvate dehydrogenase (PDH) and phospholamban, circulating inflammatory cytokines, plasma lactate, and glucose as measures of systemic metabolic recovery. VO reduced deterioration of left ventricular maximum pressure, maximum rate of change in the left ventricular pressure, and Petco2 and improved 72 h neurological intact survival (50% vs. 10%; P < 0.05). It reduced plasma lactate, glucose, IL-1ß, and Pre-B cell colony enhancing factor, while increasing IL-10. VO increased phosphorylation of Akt and GSK3ß in both heart and brain, and cardiac phospholamban phosphorylation while reducing p-PDH. Moreover, VO improved cardiac bioenergetic recovery. We concluded that pharmacologic PTEN inhibition enhances Akt activation, improving metabolic, cardiovascular, and neurologic recovery with increased survival after SCA. PTEN inhibitors may be a novel pharmacologic strategy for treating SCA.


Asunto(s)
Metabolismo Energético , Inhibidores Enzimáticos/uso terapéutico , Paro Cardíaco/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Fosfohidrolasa PTEN/antagonistas & inhibidores , Animales , Citocinas/sangre , Femenino , Paro Cardíaco/metabolismo , Hemodinámica , Ratones , Ratones Endogámicos C57BL , Compuestos Organometálicos/farmacología , Resucitación/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA