Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Wilderness Environ Med ; 26(4 Suppl): S15-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26617374

RESUMEN

A thorough medical history is perhaps the most important aspect when evaluating an athlete before wilderness adventure. A physical examination should follow focusing on conditions that may be affected by changes in atmospheric pressure, extremes of temperature, or altitude. This information can then be used to make safety recommendations ensuring that adventurers are able to safely enjoy participation in the wilderness pursuit of their choice.


Asunto(s)
Anamnesis/métodos , Examen Físico/métodos , Medicina Deportiva/métodos , Vida Silvestre , Atletas , Humanos , Pediatría , Medición de Riesgo , Seguridad , Sociedades Médicas , Deportes
2.
Clin J Sport Med ; 25(5): 392-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26340730

RESUMEN

A thorough medical history is perhaps the most important aspect when evaluating an athlete before wilderness adventure. A physical examination should follow focusing on conditions that may be affected by changes in atmospheric pressure, extremes of temperature, or altitude. This information can then be used to make safety recommendations ensuring that adventurers are able to safely enjoy participation in the wilderness pursuit of their choice.


Asunto(s)
Examen Físico/métodos , Seguridad , Deportes , Medicina Silvestre , Atletas , Humanos , Medición de Riesgo
3.
Mol Cell Biol ; 35(5): 831-46, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25535334

RESUMEN

Sustained Akt activation induces cardiac hypertrophy (LVH), which may lead to heart failure. This study tested the hypothesis that Akt activation contributes to mitochondrial dysfunction in pathological LVH. Akt activation induced LVH and progressive repression of mitochondrial fatty acid oxidation (FAO) pathways. Preventing LVH by inhibiting mTOR failed to prevent the decline in mitochondrial function, but glucose utilization was maintained. Akt activation represses expression of mitochondrial regulatory, FAO, and oxidative phosphorylation genes in vivo that correlate with the duration of Akt activation in part by reducing FOXO-mediated transcriptional activation of mitochondrion-targeted nuclear genes in concert with reduced signaling via peroxisome proliferator-activated receptor α (PPARα)/PGC-1α and other transcriptional regulators. In cultured myocytes, Akt activation disrupted mitochondrial bioenergetics, which could be partially reversed by maintaining nuclear FOXO but not by increasing PGC-1α. Thus, although short-term Akt activation may be cardioprotective during ischemia by reducing mitochondrial metabolism and increasing glycolysis, long-term Akt activation in the adult heart contributes to pathological LVH in part by reducing mitochondrial oxidative capacity.


Asunto(s)
Cardiomegalia/metabolismo , Núcleo Celular/metabolismo , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Ácidos Grasos/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucólisis , Corazón/fisiología , Hemodinámica , Hipertrofia , Masculino , Ratones , Células Musculares/citología , Oxígeno/metabolismo , PPAR alfa/metabolismo , Proteómica , Transducción de Señal , Transcripción Genética , Transgenes
4.
Circ Res ; 109(7): 783-93, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21799152

RESUMEN

RATIONALE: Pressure overload cardiac hypertrophy, a risk factor for heart failure, is associated with reduced mitochondrial fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS) proteins that correlate in rodents with reduced PGC-1α expression. OBJECTIVE: To determine the role of PGC-1ß in maintaining mitochondrial energy metabolism and contractile function in pressure overload hypertrophy. METHODS AND RESULTS: PGC-1ß deficient (KO) mice and wildtype (WT) controls were subjected to transverse aortic constriction (TAC). Although LV function was modestly reduced in young KO hearts, there was no further decline with age so that LV function was similar between KO and WT when TAC was performed. WT-TAC mice developed relatively compensated LVH, despite reduced mitochondrial function and repression of OXPHOS and FAO genes. In nonstressed KO hearts, OXPHOS gene expression and palmitoyl-carnitine-supported mitochondrial function were reduced to the same extent as banded WT, but FAO gene expression was normal. Following TAC, KO mice progressed more rapidly to heart failure and developed more severe mitochondrial dysfunction, despite a similar overall pattern of repression of OXPHOS and FAO genes as WT-TAC. However, in relation to WT-TAC, PGC-1ß deficient mice exhibited greater degrees of oxidative stress, decreased cardiac efficiency, lower rates of glucose metabolism, and repression of hexokinase II protein. CONCLUSIONS: PGC-1ß plays an important role in maintaining baseline mitochondrial function and cardiac contractile function following pressure overload hypertrophy by preserving glucose metabolism and preventing oxidative stress.


Asunto(s)
Presión Sanguínea , Metabolismo Energético , Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Izquierda/complicaciones , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Transactivadores/deficiencia , Disfunción Ventricular Izquierda/etiología , Envejecimiento , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo Energético/genética , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Hexoquinasa/metabolismo , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones , Ratones Noqueados , Contracción Miocárdica , Oxidación-Reducción , Fosforilación Oxidativa , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/genética , Factores de Transcripción , Ultrasonografía , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda
5.
Cardiovasc Res ; 82(2): 351-60, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19147655

RESUMEN

AIMS: Diet-induced obesity is associated with increased myocardial fatty acid (FA) utilization, insulin resistance, and cardiac dysfunction. The study was designed to test the hypothesis that impaired glucose utilization accounts for initial changes in FA metabolism. METHODS AND RESULTS: Ten-week-old C57BL6J mice were fed a high-fat diet (HFD, 45% calories from fat) or normal chow (4% calories from fat). Cardiac function and substrate metabolism in isolated working hearts, glucose uptake in isolated cardiomyocytes, mitochondrial function, insulin-stimulated protein kinase B (Akt/PKB) and Akt substrate (AS-160) phosphorylation, glucose transporter 4 (GLUT4) translocation, pyruvate dehydrogenase (PDH) activity, and mRNA levels for metabolic genes were determined after 2 or 5 weeks of HFD. Two weeks of HFD reduced basal rates of glycolysis and glucose oxidation and prevented insulin stimulation of glycolysis in hearts and reduced insulin-stimulated glucose uptake in cardiomyocytes. Insulin-stimulated Akt/PKB and AS-160 phosphorylation were preserved, and PDH activity was unchanged. GLUT4 content was reduced by 55% and GLUT4 translocation was significantly attenuated. HFD increased FA oxidation rates and myocardial oxygen consumption (MVO2), which could not be accounted for by mitochondrial uncoupling or by increased expression of peroxisome proliferator activated receptor-alpha (PPAR-alpha) target genes, which increased only after 5 weeks of HFD. CONCLUSION: Rates of myocardial glucose utilization are altered early in the course of HFD because of reduced GLUT4 content and GLUT4 translocation despite normal insulin signalling to Akt/PKB and AS-160. The reciprocal increase in FA utilization is not due to PPAR-alpha-mediated signalling or mitochondrial uncoupling. Thus, the initial increase in myocardial FA utilization in response to HFD likely results from impaired glucose transport that precedes impaired insulin signalling.


Asunto(s)
Grasas de la Dieta/farmacología , Ácidos Grasos/metabolismo , Corazón/efectos de los fármacos , Miocardio/metabolismo , Animales , Modelos Animales de Enfermedad , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Glucólisis/efectos de los fármacos , Insulina/farmacología , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Complejo Piruvato Deshidrogenasa/metabolismo
6.
Cell Cycle ; 7(16): 2509-18, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18719368

RESUMEN

Long-chain fatty acids (FA) are the primary energy source utilized by the adult heart. However, during pathological cardiac hypertrophy the fetal gene program is reactivated and glucose becomes the major fuel source metabolized by the heart. Herein we describe the metabolic phenotype associated with caveolin-1(Cav1) gene ablation (Cav1ko) in cardiac fibroblasts. Cav1, the primary protein component of caveolae in non-muscle cells co-localizes with a number of proteins involved in substrate metabolism, including, FA translocase (CD36) and the insulin receptor. We demonstrate that Cav1ko hearts develop cardiac hypertrophy and contractile dysfunction at 5-6mos of age. Surprisingly, we observed an increase in the uptake of Intralipid triglyceride and albumin bound FA by 25% and 47%, respectively, in Cav1ko hearts. Isolated perfused heart studies revealed no significant difference in glucose oxidation and glycolysis, however, we observed a trend toward increased FA oxidation in Cav1ko hearts. Real-time PCR analysis revealed no significant changes in the expression of genes involved in FA and glucose metabolism. We also report myocardial triglyceride, fatty acid and cholesterol levels are significantly reduced in Cav1ko hearts. Microarray gene expression analysis revealed changes in genes that regulate calcium ion and lipid transport as well as a number of genes not previously linked to cardiac hypertrophy. We observed a 4-fold increase in tetraspanin-2 gene expression, a transmembrane protein implicated in regulating intracellular trafficking. Oxysterol binding protein related protein-3, which has been implicated in intracellular lipid synthesis and transport, was increased 3.6-fold. In addition, sarcoplasmic reticulum Ca(2+)-ATPase 3, and calcyclin gene transcripts were significantly increased in Cav1ko hearts. In summary, targeted loss of Cav1 produces a unique model of cardiac hypertrophy with normal substrate utilization and expression of genes involved in energy metabolism.


Asunto(s)
Antígenos CD36/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Caveolina 1/genética , Miocardio/metabolismo , Animales , Cardiomegalia/patología , AMP Cíclico/metabolismo , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Glucosa/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Noqueados , Miocardio/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Proteínas ras/metabolismo
7.
J Lipid Res ; 49(10): 2101-12, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18515784

RESUMEN

Ceramide is among a number of potential lipotoxic molecules that are thought to modulate cellular energy metabolism. The heart is one of the tissues thought to become dysfunctional due to excess lipid accumulation. Dilated lipotoxic cardiomyopathy, thought to be the result of diabetes and severe obesity, has been modeled in several genetically altered mice, including animals with cardiac-specific overexpression of glycosylphosphatidylinositol (GPI)-anchored human lipoprotein lipase (LpL(GPI)). To test whether excess ceramide was implicated in cardiac lipotoxicity, de novo ceramide biosynthesis was inhibited pharmacologically by myriocin and genetically by heterozygous deletion of LCB1, a subunit of serine palmitoyltransferase (SPT). Inhibition of SPT, a rate-limiting enzyme in ceramide biosynthesis, reduced fatty acid and increased glucose oxidation in isolated perfused LpL(GPI) hearts, improved systolic function, and prolonged survival rates. Our results suggest a critical role for ceramide accumulation in the pathogenesis of lipotoxic cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cardiotoxinas/metabolismo , Ceramidas/metabolismo , Animales , Biomarcadores/metabolismo , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Cardiotoxinas/antagonistas & inhibidores , Bovinos , Ceramidas/antagonistas & inhibidores , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Glicosilfosfatidilinositoles/metabolismo , Corazón/efectos de los fármacos , Corazón/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Serina C-Palmitoiltransferasa/antagonistas & inhibidores , Serina C-Palmitoiltransferasa/genética , Serina C-Palmitoiltransferasa/metabolismo , Tasa de Supervivencia
8.
Am J Physiol Heart Circ Physiol ; 295(2): H657-66, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18552160

RESUMEN

Caveolin-3 (Cav3), the primary protein component of caveolae in muscle cells, regulates numerous signaling pathways including insulin receptor signaling and facilitates free fatty acid (FA) uptake by interacting with several FA transport proteins. We previously reported that Cav3 knockout mice (Cav3KO) develop cardiac hypertrophy with diminished contractile function; however, the effects of Cav3 gene ablation on cardiac substrate utilization are unknown. The present study revealed that the uptake and oxidation of FAs and glucose were normal in hypertrophic Cav3KO hearts. Real-time PCR analysis revealed normal expression of lipid metabolism genes including FA translocase (CD36) and carnitine palmitoyl transferase-1 in Cav3KO hearts. Interestingly, myocardial cAMP content was significantly increased by 42%; however, this had no effect on PKA activity in Cav3KO hearts. Microarray expression analysis revealed a marked increase in the expression of genes involved in receptor trafficking to the plasma membrane, including Rab4a and the expression of WD repeat/FYVE domain containing proteins. We observed a fourfold increase in the expression of cellular retinol binding protein-III and a 3.5-fold increase in 17beta-hydroxysteroid dehydrogenase type 11, a member of the short-chain dehydrogenase/reductase family involved in the biosynthesis and inactivation of steroid hormones. In summary, a loss of Cav3 in the heart leads to cardiac hypertrophy with normal substrate utilization. Moreover, a loss of Cav3 mRNA altered the expression of several genes not previously linked to cardiac growth and function. Thus we have identified a number of new target genes associated with the pathogenesis of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Caveolina 3/metabolismo , Metabolismo Energético , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Miocardio/metabolismo , Animales , Transporte Biológico/genética , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Caveolina 3/deficiencia , Caveolina 3/genética , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endocitosis/genética , Metabolismo Energético/genética , Fibrosis , Perfilación de la Expresión Génica/métodos , Inmunidad Innata/genética , Antígenos Comunes de Leucocito/análisis , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/enzimología , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Ultrasonografía , Vitamina A/metabolismo
9.
Endocrinology ; 149(8): 4043-50, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18450963

RESUMEN

The goal of this study was to determine whether inhibiting the renin-angiotensin system would restore insulin signaling and normalize substrate use in hearts from obese ob/ob mice. Mice were treated for 4 wk with Captopril (4 mg/kg x d). Circulating levels of free fatty acids, triglycerides, and insulin were measured and glucose tolerance tests performed. Rates of palmitate oxidation and glycolysis, oxygen consumption, and cardiac power were determined in isolated working hearts in the presence and absence of insulin, along with levels of phosphorylation of Akt and AMP-activated protein kinase (AMPK). Captopril treatment did not correct the hyperinsulinemia or impaired glucose tolerance in ob/ob mice. Rates of fatty acid oxidation were increased and glycolysis decreased in ob/ob hearts, and insulin did not modulate substrate use in hearts of ob/ob mice and did not increase Akt phosphorylation. Captopril restored the ability of insulin to regulate fatty acid oxidation and glycolysis in hearts of ob/ob mice, possibly by increasing Akt phosphorylation. Moreover, AMPK phosphorylation, which was increased in hearts of ob/ob mice, was normalized by Captopril treatment, suggesting that in addition to restoring insulin sensitivity, Captopril treatment improved myocardial energetics. Thus, angiotensin-converting enzyme inhibitors restore the responsiveness of ob/ob mouse hearts to insulin and normalizes AMPK activity independently of effects on systemic metabolic homeostasis.


Asunto(s)
Captopril/farmacología , Glucosa/metabolismo , Corazón/efectos de los fármacos , Insulina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Miocardio/metabolismo , Obesidad/metabolismo , Angiotensina II/sangre , Animales , Antihipertensivos/farmacología , Peso Corporal/efectos de los fármacos , Captopril/uso terapéutico , Enfermedades Cardiovasculares/prevención & control , Evaluación Preclínica de Medicamentos , Corazón/anatomía & histología , Insulina/farmacología , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Técnicas de Cultivo de Órganos , Tamaño de los Órganos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Cell Metab ; 6(4): 294-306, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17908558

RESUMEN

Physiological cardiac hypertrophy is associated with mitochondrial adaptations that are characterized by activation of PGC-1alpha and increased fatty acid oxidative (FAO) capacity. It is widely accepted that phosphatidylinositol 3-kinase (PI3K) signaling to Akt1 is required for physiological cardiac growth. However, the signaling pathways that coordinate physiological hypertrophy and metabolic remodeling are incompletely understood. We show here that activation of PI3K is sufficient to increase myocardial FAO capacity and that inhibition of PI3K signaling prevents mitochondrial adaptations in response to physiological hypertrophic stimuli despite increased expression of PGC-1alpha. We also show that activation of the downstream kinase Akt is not required for the mitochondrial adaptations that are secondary to PI3K activation. Thus, in physiological cardiac growth, PI3K is an integrator of cellular growth and metabolic remodeling. Although PI3K signaling to Akt1 is required for cellular growth, Akt-independent pathways mediate the accompanying mitochondrial adaptations.


Asunto(s)
Cardiomegalia/enzimología , Mitocondrias/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Condicionamiento Físico Animal , Proteínas Proto-Oncogénicas c-akt/fisiología , Adaptación Fisiológica , Animales , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Mutantes , Miocitos Cardíacos/enzimología , Oxidación-Reducción , Fosfatidilinositol 3-Quinasas/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
11.
J Biol Chem ; 281(13): 8716-23, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16410253

RESUMEN

Long-chain fatty acids (FAs) are the predominant energy substrate utilized by the adult heart. The heart can utilize unesterified FA bound to albumin or FA obtained from lipolysis of lipoprotein-bound triglyceride (TG). We used heart-specific lipoprotein lipase knock-out mice (hLpL0) to test whether these two sources of FA are interchangeable and necessary for optimal heart function. Hearts unable to obtain FA from lipoprotein TG were able to compensate by increasing glucose uptake, glycolysis, and glucose oxidation. HLpL0 hearts had decreased expression of pyruvate dehydrogenase kinase 4 and increased cardiomyocyte expression of glucose transporter 4. Conversely, FA oxidation rates were reduced in isolated perfused hLpL0 hearts. Following abdominal aortic constriction expression levels of genes regulating FA and glucose metabolism were acutely up-regulated in control and hLpL0 mice, yet all hLpL0 mice died within 48 h of abdominal aortic constriction. Older hLpL0 mice developed cardiac dysfunction characterized by decreased fractional shortening and interstitial and perivascular fibrosis. HLpL0 hearts had increased expression of several genes associated with transforming growth factor-beta signaling. Thus, long term reduction of lipoprotein FA uptake is associated with impaired cardiac function despite a compensatory increase in glucose utilization.


Asunto(s)
Cardiomiopatías/metabolismo , Glucosa/metabolismo , Lipoproteína Lipasa/deficiencia , Lipoproteína Lipasa/fisiología , Miocardio/enzimología , Adenosina Trifosfato/análisis , Animales , Glucemia/análisis , Radioisótopos de Carbono , Cardiomiopatías/etiología , Colesterol/sangre , Colesterol/metabolismo , Ecocardiografía , Ayuno , Ácidos Grasos/metabolismo , Femenino , Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Glucólisis , Cinética , Lipoproteína Lipasa/análisis , Lipoproteína Lipasa/genética , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosfocreatina/análogos & derivados , Fosfocreatina/análisis , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Triglicéridos/sangre
12.
Endocrinology ; 146(12): 5341-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16141388

RESUMEN

Hyperglycemia is associated with altered myocardial substrate use, a condition that has been hypothesized to contribute to impaired cardiac performance. The goals of this study were to determine whether changes in cardiac metabolism, gene expression, and function precede or follow the onset of hyperglycemia in two mouse models of obesity, insulin resistance, and diabetes (ob/ob and db/db mice). Ob/ob and db/db mice were studied at 4, 8, and 15 wk of age. Four-week-old mice of both strains were normoglycemic but hyperinsulinemic. Hyperglycemia develops in db/db mice between 4 and 8 wk of age and in ob/ob mice between 8 and 15 wk. In isolated working hearts, rates of glucose oxidation were reduced by 28-37% at 4 wk and declined no further at 15 wk in both strains. Fatty acid oxidation rates and myocardial oxygen consumption were increased in 4-wk-old mice of both strains. Fatty acid oxidation rates progressively increased in db/db mice in parallel with the earlier onset and greater duration of hyperglycemia. In vivo, cardiac catheterization revealed significantly increased left ventricular contractility and relaxation (positive and negative dP/dt) in both strains at 4 wk of age. dP/dt declined over time in db/db mice but remained elevated in ob/ob mice at 15 wk of age. Increased beta-myosin heavy chain isoform expression was present in 4-wk-old mice and persisted in 15-wk-old mice. Increased expression of peroxisomal proliferator-activated receptor-alpha regulated genes was observed only at 15 wk in both strains. These data indicate that altered myocardial substrate use and reduced myocardial efficiency are early abnormalities in the hearts of obese mice and precede the onset of hyperglycemia. Obesity per se does not cause contractile dysfunction in vivo, but loss of the hypercontractile phenotype of obesity and up-regulation of peroxisomal proliferator-activated receptor-alpha regulated genes occur later and are most pronounced in the presence of longstanding hyperglycemia.


Asunto(s)
Corazón/fisiopatología , Hiperglucemia/etiología , Resistencia a la Insulina , Contracción Miocárdica , Miocardio/metabolismo , Obesidad/complicaciones , Obesidad/fisiopatología , Animales , Complicaciones de la Diabetes , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Expresión Génica , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Cadenas Pesadas de Miosina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Consumo de Oxígeno , PPAR alfa/metabolismo
13.
Diabetes ; 53(9): 2366-74, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331547

RESUMEN

Diabetes alters cardiac substrate metabolism. The cardiac phenotype in insulin-resistant states has not been comprehensively characterized. The goal of these studies was to determine whether the hearts of leptin-deficient 8-week-old ob/ob mice were able to modulate cardiac substrate utilization in response to insulin or to changes in fatty acid delivery. Ob/ob mice were insulin resistant and glucose intolerant. Insulin signal transduction and insulin-stimulated glucose uptake were markedly impaired in ob/ob cardiomyocytes. Insulin-stimulated rates of glycolysis and glucose oxidation were 1.5- and 1.8-fold higher in wild-type hearts, respectively, versus ob/ob, and glucose metabolism in ob/ob hearts was unresponsive to insulin. Increasing concentrations of palmitate from 0.4 mmol/l (low) to 1.2 mmol/l (high) led to a decline in glucose oxidation in wild-type hearts, whereas glucose oxidation remained depressed and did not change in ob/ob mouse hearts. In contrast, fatty acid utilization in ob/ob hearts was 1.5- to 2-fold greater in the absence or presence of 1 nmol/l insulin and rose with increasing palmitate concentrations. Moreover, the ability of insulin to reduce palmitate oxidation rates was blunted in the hearts of ob/ob mice. Under low-palmitate and insulin-free conditions, cardiac performance was significantly greater in wild-type hearts. However, in the presence of high palmitate and 1 nmol/l insulin, cardiac performance in ob/ob mouse hearts was relatively preserved, whereas function in wild-type mouse hearts declined substantially. Under all perfusion conditions, myocardial oxygen consumption was higher in ob/ob hearts, ranging from 30% higher in low-palmitate conditions to greater than twofold higher under high-palmitate conditions. These data indicate that although the hearts of glucose-intolerant ob/ob mice are capable of maintaining their function under conditions of increased fatty acid supply and hyperinsulinemia, they are insulin-resistant, metabolically inefficient, and unable to modulate substrate utilization in response to changes in insulin and fatty acid supply.


Asunto(s)
Resistencia a la Insulina/fisiología , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Palmitatos/metabolismo , Animales , Glucosa/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Miocardio/citología , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/fisiología , Palmitatos/farmacología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA