Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 158: 115-127, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34081952

RESUMEN

RATIONALE: The nutrient sensing mechanistic target of rapamycin complex 1 (mTORC1) and its primary inhibitor, tuberin (TSC2), are cues for the development of cardiac hypertrophy. The phenotype of mTORC1 induced hypertrophy is unknown. OBJECTIVE: To examine the impact of sustained mTORC1 activation on metabolism, function, and structure of the adult heart. METHODS AND RESULTS: We developed a mouse model of inducible, cardiac-specific sustained mTORC1 activation (mTORC1iSA) through deletion of Tsc2. Prior to hypertrophy, rates of glucose uptake and oxidation, as well as protein and enzymatic activity of glucose 6-phosphate isomerase (GPI) were decreased, while intracellular levels of glucose 6-phosphate (G6P) were increased. Subsequently, hypertrophy developed. Transcript levels of the fetal gene program and pathways of exercise-induced hypertrophy increased, while hypertrophy did not progress to heart failure. We therefore examined the hearts of wild-type mice subjected to voluntary physical activity and observed early changes in GPI, followed by hypertrophy. Rapamycin prevented these changes in both models. CONCLUSION: Activation of mTORC1 in the adult heart triggers the development of a non-specific form of hypertrophy which is preceded by changes in cardiac glucose metabolism.


Asunto(s)
Cardiomegalia/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Glucosa/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal/genética , Animales , Cardiomegalia/dietoterapia , Cardiomegalia/genética , Cardiomegalia/prevención & control , Células Cultivadas , Dieta/métodos , Modelos Animales de Enfermedad , Activación Enzimática/genética , Glucosa-6-Fosfatasa/metabolismo , Isomerasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosforilación/genética , Sirolimus/administración & dosificación , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
2.
FASEB J ; 27(10): 3966-78, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23825227

RESUMEN

Diabetic patients with acute myocardial infarction are more likely to die than nondiabetic patients. In the present study we examined the effect of insulin resistance on myocardial ischemia tolerance. Hearts of rats, rendered insulin resistant by high-sucrose feeding, were subjected to ischemia/reperfusion ex vivo. Cardiac power of control hearts from chow-fed rats recovered to 93%, while insulin-resistant hearts recovered only to 80% (P<0.001 vs. control). Unexpectedly, impaired contractile recovery did not result from an impairment of glucose oxidation (576±36 vs. 593±42 nmol/min/g dry weight; not significant), but from a failure to increase and to sustain oxidation of the long-chain fatty acid oleate on reperfusion (1878±56 vs. 2070±67 nmol/min/g dry weight; P<0.05). This phenomenon was due to a reduced ability to transport oleate into mitochondria and associated with a 38-58% decrease in the mitochondrial uncoupling protein 3 (UCP3) levels. Contractile function was rescued by replacing oleate with a medium-chain fatty acid or by restoring UCP3 levels with 24 h of food withdrawal. Lastly, the knockdown of UCP3 in rat L6 myocytes also decreased oleate oxidation by 13-18% following ischemia. Together the results expose UCP3 as a critical regulator of long-chain fatty acid oxidation in the stressed heart postischemia and identify octanoate as an intervention by which myocardial metabolism can be manipulated to improve function of the insulin-resistant heart.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Resistencia a la Insulina/fisiología , Daño por Reperfusión/metabolismo , Alimentación Animal , Animales , Caprilatos , Dieta , Carbohidratos de la Dieta , Regulación de la Expresión Génica , Glucosa/metabolismo , Insulina/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Sacarosa , Proteína Desacopladora 3
3.
FASEB J ; 26(8): 3118-26, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22611083

RESUMEN

Insulin resistance is a prominent feature in heart failure, while hyperglycemia impairs cardiac contraction. We propose that decreased insulin-mediated glucose uptake by the heart preserves cardiac function in response to metabolic and hemodynamic stress. To test this hypothesis, we fed rats a high-sucrose diet (HSD). Energy substrate metabolism and cardiac work were determined ex vivo in a sequential protocol simulating metabolic and hemodynamic stress. Compared to chow-fed, control rats, HSD impaired myocardial insulin responsiveness and induced profound metabolic changes in the heart, characterized by reduced rates of glucose uptake (7.91 ± 0.30 vs. 10.73 ± 0.67 µmol/min/g dry weight; P<0.001) but increased rates of glucose oxidation (2.38 ± 0.17 vs. 1.50 ± 0.15 µmol/min/g dry weight; P<0.001) and oleate oxidation (2.29 ± 0.11 vs. 1.96 ± 0.12 µmol/min/g dry weight; P<0.05). Tight coupling of glucose uptake and oxidation and improved cardiac efficiency were associated with a reduction in glucose 6-phosphate and oleoyl-CoA levels, as well as a reduction in the content of uncoupling protein 3. Our results suggest that insulin resistance lessens fuel toxicity in the stressed heart. This calls for a new exploration of the mechanisms regulating substrate uptake and oxidation in the insulin-resistant heart.


Asunto(s)
Corazón/fisiología , Resistencia a la Insulina/fisiología , Miocardio/metabolismo , Estrés Fisiológico/fisiología , Animales , Sacarosa en la Dieta/administración & dosificación , Sacarosa en la Dieta/farmacología , Regulación hacia Abajo , Glucosa/metabolismo , Técnicas In Vitro , Insulina/fisiología , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Contracción Miocárdica/efectos de los fármacos , Ácido Oléico/metabolismo , Perfusión , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína Desacopladora 3
4.
Am J Physiol Heart Circ Physiol ; 295(1): H256-65, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18487439

RESUMEN

The cardiac-enriched isoform of acetyl-CoA carboxylase (ACC2) is a key regulator of mitochondrial fatty acid (FA) uptake via carnitine palmitoyltransferase 1 (CPT1). To test the hypothesis that oxidative metabolism is upregulated in hearts from animals lacking ACC2 (employing a transgenic Acc2-mutant mouse), we assessed cardiac function in vivo and determined rates of myocardial substrate oxidation ex vivo. When examined by echocardiography, there was no difference in systolic function, but left ventricular mass of the Acc2-mutant (MUT) mouse was significantly reduced ( approximately 25%) compared with wild-types (WT). Reduced activation of the mammalian target of rapamycin (mTOR) and its downstream target p70S6K was found in MUT hearts. Exogenous oxidation rates of oleate were increased approximately 22%, and, unexpectedly, exogenous glucose oxidation rates were also increased in MUT hearts. Using a hyperinsulinemic-euglycemic clamp, we found that glucose uptake in MUT hearts was increased by approximately 83%. Myocardial triglyceride levels were significantly reduced in MUT vs. WT while glycogen content was the same. In parallel, transcript levels of PPARalpha and its target genes, pyruvate dehydrogenase kinase-4 (PDK-4), malonyl-CoA decarboxylase (MCD), and mCPT1, were downregulated in MUT mice. In summary, we report that 1) Acc2-mutant hearts exhibit a marked preference for the oxidation of both glucose and FAs coupled with greater utilization of endogenous fuel substrates (triglycerides), 2) attenuated mTOR signaling may result in reduced heart sizes observed in Acc2-mutant mice, and 3) Acc2-mutant hearts displayed normal functional parameters despite a significant decrease in size.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Metabolismo Energético , Mutación , Miocardio/enzimología , Acetil-CoA Carboxilasa/genética , Animales , Carboxiliasas/genética , Carboxiliasas/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Regulación hacia Abajo , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Glucógeno/metabolismo , Ventrículos Cardíacos/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/patología , Ácido Oléico/metabolismo , Tamaño de los Órganos , Oxidación-Reducción , PPAR gamma/genética , PPAR gamma/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR , Factores de Tiempo , Triglicéridos/metabolismo , Ultrasonografía
5.
Cardiovasc Res ; 76(1): 71-80, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17553476

RESUMEN

OBJECTIVE: Insulin regulates both glucose uptake and postnatal cardiac growth. The anabolic effects of insulin are mediated by the mammalian target of rapamycin (mTOR), an evolutionarily conserved kinase which is also a convergence point between nutrient sensing and cell growth. We postulated that mTOR signalling in the heart requires the metabolism of glucose. METHODS: We interrogated the insulin-mediated mTOR signalling pathway in response to different metabolic interventions regulating substrate metabolism in the isolated working rat heart and in isolated cardiomyocytes. RESULTS: Although insulin enhanced Akt activity, phosphorylation of mTOR and its downstream targets (p70S6K and 4EBP1) required the addition of glucose. Glucose-dependent p70S6K phosphorylation was independent of the hexosamine biosynthetic pathway, the AMP kinase pathway, and the pentose phosphate pathway. However, inhibition of glycolysis downstream of hexokinase markedly enhanced p70S6K phosphorylation. Furthermore, 2-deoxyglucose activated p70S6K suggesting that phosphorylation of glucose is required for carbohydrate-mediated mTOR signalling in the heart. Lastly, we also found enhanced p70S6K phosphorylation in the hearts of diabetic rats. CONCLUSION: Phosphorylation of glucose is necessary for insulin-dependent mTOR activity in the heart, suggesting a link between intermediary metabolism and cardiac growth.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Western Blotting/métodos , Proteínas de Ciclo Celular , Células Cultivadas , Glucólisis , Hexosaminas/biosíntesis , Masculino , Pentosafosfatos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR
6.
Gene Regul Syst Bio ; 1: 251-62, 2007 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19936093

RESUMEN

BACKGROUND: During pressure overload-induced hypertrophy, unloading-induced atrophy, and diabetes mellitus, the heart induces 'fetal' genes (e.g. myosin heavy chain beta; mhc beta). HYPOTHESIS: We propose that altered glucose homeostasis within the cardiomyocyte acts as a central mechanism for the regulation of gene expression in response to environmental stresses. The evidence is as follows. METHODS AND RESULTS: Forced glucose uptake both ex vivo and in vivo results in mhc isoform switching. Restricting dietary glucose prevents mhc isoform switching in hearts of both GLUT1-Tg mice and rats subjected to pressure overload-induced hypertrophy. Thus, glucose availability correlates with mhc isoform switching under all conditions investigated. A potential mechanism by which glucose affects gene expression is through O-linked glycosylation of specific transcription factors. Glutamine:fructose-6-phosphate amidotransferase (GFAT) catalyzes the flux generating step in UDP-N-acetylglucosamine biosynthesis, the rate determining metabolite in protein glycosylation. Ascending aortic constriction increased intracellular levels of UDP-N-acetylglucosamine, and the expression of gfat2, but not gfat1, in the rat heart. CONCLUSIONS: Collectively, the results strongly suggest glucose-regulated gene expression in the heart, and the involvement of glucose metabolites in isoform switching of sarcomeric proteins characteristic for the fetal gene program.

7.
Am J Physiol Endocrinol Metab ; 289(2): E328-36, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15797988

RESUMEN

It is suggested that insulin resistance and metabolic maladaptation of the heart are causes of contractile dysfunction. We tested the hypothesis whether systemic PPARgamma activation, by changing the metabolic profile in a model of insulin resistance and type 2 diabetes (the ZDF rat) in vivo, improves contractile function of the heart in vitro. Male Zucker diabetic fatty (ZDF) and Zucker lean (ZL) rats, at 53-56 days of age, were treated with either GI-262570 (a nonthiazolidinedione PPARgamma agonist; A) or vehicle (V) for 1 wk. Agonist treatment resulted in correction of hyperglycemia and dyslipidemia, as well as in reduced hyperinsulinemia. The accumulation of triacylglycerols in the myocardium, characteristic of the ZDF rat, disappeared with treatment. Cardiac power and rates of glucose oxidation in the isolated working heart were significantly reduced in ZDF-V rats, but both parameters increased to nondiabetic levels with agonist treatment. In ZDF-V hearts, transcript levels of PPARalpha-regulated genes and of myosin heavy chain-beta were upregulated, whereas GLUT4 was downregulated compared with ZL. Agonist treatment of ZDF rats reduced PPARalpha-regulated genes and increased transcripts of GLUT4 and GLUT1. In conclusion, by changing the metabolic profile, reducing myocardial lipid accumulation, and promoting the downregulation of PPARalpha-regulated genes, PPARgamma activation leads to an increased capacity of the myocardium to oxidize glucose and to a tighter coupling of oxidative metabolism and contraction in the setting of insulin resistance and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Contracción Miocárdica/fisiología , Miocardio/metabolismo , PPAR gamma/metabolismo , Adaptación Fisiológica , Animales , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Técnicas In Vitro , Resistencia a la Insulina/fisiología , Masculino , Contracción Miocárdica/efectos de los fármacos , Oxazoles/farmacología , PPAR gamma/agonistas , Ratas , Ratas Endogámicas , Ratas Zucker , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Triglicéridos/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacología
8.
Am J Physiol Endocrinol Metab ; 285(6): E1312-6, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12944323

RESUMEN

To what extent does glutamine turnover keep pace with oxidative metabolism in the rat heart? To address this question, the following groups of substrates were presented to the isolated, working rat heart: 1) glucose (5 mM), insulin (40 microU/ml), and [2-13C]acetate (5 mM; high workload, n = 5); 2) pyruvate (2.5 mM) and [2-13C]acetate (5 mM; normal workload, n = 5); or 3) propionate (1 mM) and [2-13C]acetate (2.5 mM; normal workload, n = 3). In a subset of these experiments, the exchange of glutamate and glutamine was quantified by separation with ion exchange chromatography and analysis by GC-MS. There was an apparent equilibration of mass isotopomers of glutamate and glutamine after 50 min of perfusion, although the extent of equilibration was not determined. The fractional enrichment in glutamine was 31% of the enrichment of glutamate with the three different perfusates. From high-resolution nuclear magnetic resonance spectra, we found a ratio of glutamine to glutamate content of 94.1, 53.4, and 96.9%, respectively, for each experimental group. In experiments for which l-[1-13C]glutamine (5 mM) was included in the perfusate of group 2, [1-13C]glutamine was detected in the heart, but transfer of 13C from glutamine to glutamate was not detected (n = 4). We conclude that, in the perfused working heart, production of glutamine by amidation of glutamate takes place and can be detected, whereas the reverse process, generation of glutamate from glutamine, remains undetected.


Asunto(s)
Ácido Glutámico/metabolismo , Glutamina/metabolismo , Corazón/fisiología , Miocardio/metabolismo , Adaptación Fisiológica , Animales , Gasto Cardíaco/fisiología , Glucosa/metabolismo , Técnicas In Vitro , Insulina/metabolismo , Masculino , Consumo de Oxígeno/fisiología , Propionatos/metabolismo , Ácido Pirúvico/metabolismo , Ratas , Ratas Sprague-Dawley , Acetato de Sodio/metabolismo , Ciclo del Sustrato/fisiología
9.
J Biol Chem ; 278(15): 13143-50, 2003 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-12522136

RESUMEN

The addition of glutamine as a major nutrient to cultured neonatal rat cardiomyocytes produced an increase in myocyte size and the organization of actin into myofibrillar arrays. The cellular response was associated with increased abundance of the mRNAs encoding the contractile proteins, alpha-myosin heavy chain and cardiac alpha-actin, and the metabolic enzymes, muscle carnitine palmitoyl transferase I and muscle adenylosuccinate synthetase (ADSS1). Adss1 gene expression was induced approximately 5-fold in glutamine-treated rat neonatal cardiac myocytes. The induction was mediated through the protein kinase A and mammalian target of rapamycin signaling pathways and required a cyclic AMP response element associated with the promoter region of the Adss1 gene. These results highlight glutamine as a major nutrient regulator of cardiac gene expression and identify protein kinase A and mammalian target of rapamycin signaling pathways as mediators of the cardiomyocyte transcriptional response.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glutamina/farmacología , Corazón/fisiología , Proteínas Quinasas/metabolismo , Transcripción Genética/fisiología , Adenilosuccinato Sintasa/genética , Angiotensina II/farmacología , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Corazón/efectos de los fármacos , Modelos Biológicos , Miocardio/citología , Cadenas Pesadas de Miosina/genética , ARN Mensajero/genética , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Transcripción Genética/efectos de los fármacos
10.
Diabetes ; 51(8): 2587-95, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12145175

RESUMEN

We investigated whether decreased responsiveness of the heart to physiological increases in fatty acid availability results in lipid accumulation and lipotoxic heart disease. Lean and obese Zucker rats were either fed ad libitum or fasted overnight. Fasting increased plasma nonesterified fatty acid levels in both lean and obese rats, although levels were greatest in obese rats regardless of nutritional status. Despite increased fatty acid availability, the mRNA transcript levels of peroxisome proliferator-activated receptor (PPAR)-alpha-regulated genes were similar in fed lean and fed obese rat hearts. Fasting increased expression of all PPAR-alpha -regulated genes in lean Zucker rat hearts, whereas, in obese Zucker rat hearts, muscle carnitine palmitoyltransferase and medium-chain acyl-CoA dehydrogenase were unaltered with fasting. Rates of oleate oxidation were similar for hearts from fed rats. However, fasting increased rates of oleate oxidation only in hearts from lean rats. Dramatic lipid deposition occurred within cardiomyocytes of obese, but not lean, Zucker rats upon fasting. Cardiac output was significantly depressed in hearts isolated from obese rats compared with lean rats, regardless of nutritional status. Fasting increased cardiac output in hearts of lean rats only. Thus, the heart's inability to increase fatty acid oxidation in proportion to increased fatty acid availability is associated with lipid accumulation and contractile dysfunction of the obese Zucker rat.


Asunto(s)
Ácidos Grasos no Esterificados/sangre , Ácidos Grasos/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación de la Expresión Génica , Músculo Esquelético/fisiopatología , Obesidad/fisiopatología , Acil-CoA Deshidrogenasa , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Animales , Carnitina O-Palmitoiltransferasa , Proteínas Portadoras/genética , Ayuno , Técnicas In Vitro , Canales Iónicos , Isoenzimas/genética , Metabolismo de los Lípidos , Masculino , Proteínas Mitocondriales , Músculo Esquelético/enzimología , Contracción Miocárdica , Miocardio/metabolismo , Obesidad/genética , Obesidad/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Proteínas Quinasas/genética , Ratas , Ratas Zucker , Receptores Citoplasmáticos y Nucleares/fisiología , Delgadez/genética , Factores de Transcripción/fisiología , Transcripción Genética , Proteína Desacopladora 3
11.
J Mol Cell Cardiol ; 34(2): 223-31, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11851361

RESUMEN

The heart, like other organs, possesses an internal circadian clock. These clocks provide the selective advantage of anticipation, enabling the organ to prepare for a given stimulus, thereby optimizing the appropriate response. The heart in diabetes is associated with alterations in morphology, gene expression, metabolism and contractile performance. The present study investigated whether diabetes also alters the circadian clock in the heart. Insulin-dependent diabetes mellitus was induced in rats by treatment with streptozotocin (STZ; 65 mg/kg). STZ increased humoral (glucose and non-esterified fatty acids) and heart gene expression (myosin heavy chain beta, pyruvate dehydrogenase kinase 4 and uncoupling protein 3) markers of diabetes. The circadian patterns of gene expression of seven components of the mammalian clock (bmal1, clock, cry1, cry2, per1, per2 and per3), as well as three clock output genes (dbp, hlf and tef), were compared in hearts isolated from control and STZ-induced diabetic rats. All components of the clock investigated possessed circadian rhythms of gene expression. In the hearts isolated from STZ-induced diabetic rats, the phases of these circadian rhythms were altered (approximately 3 h early) compared to those observed for control hearts. The clock in the heart has therefore lost normal synchronization with its environment during diabetes. Whether this loss of synchronization plays a role in the development of contractile dysfunction of the heart in diabetes remains to be determined.


Asunto(s)
Ritmo Circadiano/fisiología , Diabetes Mellitus Experimental/fisiopatología , Proteínas de Drosophila , Proteínas del Ojo , Corazón/fisiología , Células Fotorreceptoras de Invertebrados , Animales , Ritmo Circadiano/genética , Criptocromos , Flavoproteínas/fisiología , Regulación de la Expresión Génica/fisiología , Masculino , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...