Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Biol Chem ; 287(22): 18429-39, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22493506

RESUMEN

The role of serine palmitoyltransferase (SPT) and de novo ceramide biosynthesis in cardiac ceramide and sphingomyelin metabolism is unclear. To determine whether the de novo synthetic pathways, rather than ceramide uptake from circulating lipoproteins, is important for heart ceramide levels, we created cardiomyocyte-specific deficiency of Sptlc2, a subunit of SPT. Heart-specific Sptlc2-deficient (hSptlc2 KO) mice had a >35% reduction in ceramide, which was limited to C18:0 and very long chain ceramides. Sphingomyelinase expression, and levels of sphingomyelin and diacylglycerol were unchanged. But surprisingly phospholipids and acyl CoAs contained increased saturated long chain fatty acids. hSptlc2 KO mice had decreased fractional shortening and thinning of the cardiac wall. While the genes regulating glucose and fatty acid metabolism were not changed, expression of cardiac failure markers and the genes involved in the formation of extracellular matrices were up-regulated in hSptlc2 KO hearts. In addition, ER-stress markers were up-regulated leading to increased apoptosis. These results suggest that Sptlc2-mediated de novo ceramide synthesis is an essential source of C18:0 and very long chain, but not of shorter chain, ceramides in the heart. Changes in heart lipids other than ceramide levels lead to cardiac toxicity.


Asunto(s)
Ceramidas/metabolismo , Corazón/fisiopatología , Miocardio/enzimología , Serina C-Palmitoiltransferasa/metabolismo , Animales , Glucemia/metabolismo , Western Blotting , Células Cultivadas , Etiquetado Corte-Fin in Situ , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Serina C-Palmitoiltransferasa/genética
2.
Circulation ; 125(23): 2844-53, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22586279

RESUMEN

BACKGROUND: Heart failure is associated with impaired myocardial metabolism with a shift from fatty acids to glucose use for ATP generation. We hypothesized that cardiac accumulation of toxic lipid intermediates inhibits insulin signaling in advanced heart failure and that mechanical unloading of the failing myocardium corrects impaired cardiac metabolism. METHODS AND RESULTS: We analyzed the myocardium and serum of 61 patients with heart failure (body mass index, 26.5±5.1 kg/m(2); age, 51±12 years) obtained during left ventricular assist device implantation and at explantation (mean duration, 185±156 days) and from 9 control subjects. Systemic insulin resistance in heart failure was accompanied by decreased myocardial triglyceride and overall fatty acid content but increased toxic lipid intermediates, diacylglycerol, and ceramide. Increased membrane localization of protein kinase C isoforms, inhibitors of insulin signaling, and decreased activity of insulin signaling molecules Akt and Foxo were detectable in heart failure compared with control subjects. Left ventricular assist device implantation improved whole-body insulin resistance (homeostatic model of analysis-insulin resistance, 4.5±0.6-3.2±0.5; P<0.05) and decreased myocardial levels of diacylglycerol and ceramide, whereas triglyceride and fatty acid content remained unchanged. Improved activation of the insulin/phosphatidylinositol-3 kinase/Akt signaling cascade after left ventricular assist device implantation was confirmed by increased phosphorylation of Akt and Foxo, which was accompanied by decreased membrane localization of protein kinase C isoforms after left ventricular assist device implantation. CONCLUSIONS: Mechanical unloading after left ventricular assist device implantation corrects systemic and local metabolic derangements in advanced heart failure, leading to reduced myocardial levels of toxic lipid intermediates and improved cardiac insulin signaling.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/cirugía , Corazón Auxiliar , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/fisiología , Miocardio/metabolismo , Adulto , Anciano , Línea Celular , Ceramidas/metabolismo , Diglicéridos/metabolismo , Ácidos Grasos/metabolismo , Femenino , Insuficiencia Cardíaca/diagnóstico por imagen , Humanos , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/metabolismo , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Transducción de Señal/fisiología , Triglicéridos/metabolismo , Ultrasonografía
3.
Am J Physiol Endocrinol Metab ; 305(11): E1339-47, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24085031

RESUMEN

Hearts utilize fatty acids as a primary source of energy. The sources of those lipids include free fatty acids and lipoprotein triglycerides. Deletion of the primary triglyceride-hydrolyzing enzyme lipoprotein lipase (LPL) leads to cardiac dysfunction. Whether heart LPL-knockout (hLPL0) mice are compromised due a deficiency in energetic substrates is unknown. To test whether alternative sources of energy will prevent cardiac dysfunction in hLPL0 mice, two different models were used to supply nonlipid energy. 1) hLPL0 mice were crossed with mice transgenically expressing GLUT1 in cardiomyocytes to increase glucose uptake into the heart; this cross-corrected cardiac dysfunction, reduced cardiac hypertrophy, and increased myocardial ATP. 2) Mice were randomly assigned to a sedentary or training group (swimming) at 3 mo of age, which leads to increased skeletal muscle production of lactate. hLPL0 mice had greater expression of the lactate transporter monocarboxylate transporter-1 (MCT-1) and increased cardiac lactate uptake. Compared with hearts from sedentary hLPL0 mice, hearts from trained hLPL0 mice had adaptive hypertrophy and improved cardiac function. We conclude that defective energy intake and not the reduced uptake of fat-soluble vitamins or cholesterol is responsible for cardiac dysfunction in hLPL0 mice. In addition, our studies suggest that adaptations in cardiac metabolism contribute to the beneficial effects of exercise on the myocardium of patients with heart failure.


Asunto(s)
Metabolismo Energético/genética , Corazón/fisiología , Lipoproteína Lipasa/genética , Miocardio/metabolismo , Triglicéridos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/prevención & control , Ecocardiografía , Transportador de Glucosa de Tipo 1/genética , Lipoproteína Lipasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , Especificidad de Órganos/genética
4.
J Cardiovasc Pharmacol ; 61(4): 345-54, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23567901

RESUMEN

Fish oil (FO) supplementation may improve cardiac function in some patients with heart failure, especially those with diabetes. To determine why this occurs, we studied the effects of FO in mice with heart failure either due to transgenic expression of the lipid uptake protein acyl CoA synthetase 1 (ACS1) or overexpression of the transcription factor peroxisomal proliferator-activated receptor (PPAR) γ via the cardiac-specific myosin heavy chain (MHC) promoter. ACS1 mice and control littermates were fed 3 diets containing low-dose or high-dose FO or nonpurified diet (NPD) for 6 weeks. MHC-PPARγ mice were fed low-dose FO or NPD. Compared with control mice fed with NPD, ACS1, and MHC-PPARγ, mice fed with NPD had reduced cardiac function and survival with cardiac fibrosis. In contrast, ACS1 mice fed with high-dose FO had better cardiac function, survival, and less myocardial fibrosis. FO increased eicosapentaenoic and docosahexaenoic acids and reduced saturated fatty acids in cardiac diacylglycerols. This was associated with reduced protein kinase C alpha and beta activation. In contrast, low-dose FO reduced MHC-PPARγ mice survival with no change in protein kinase C activation or cardiac function. Thus, dietary FO reverses fibrosis and improves cardiac function and survival of ACS1 mice but does not benefit all forms of lipid-mediated cardiomyopathy.


Asunto(s)
Coenzima A Ligasas/genética , Aceites de Pescado/farmacología , Cadenas Pesadas de Miosina/genética , PPAR gamma/genética , Animales , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/patología , Femenino , Fibrosis , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Proteína Quinasa C-alfa/metabolismo , Tasa de Supervivencia
5.
J Lipid Res ; 53(8): 1482-92, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22628613

RESUMEN

Accumulation of excess lipids is associated with heart failure. The effects of transgenic expression of diacylglycerol acyl transferase 1 (DGAT1) in cardiomyocytes is controversial. We explored whether mice expressing DGAT1 via the myosin heavy chain (MHC) promoter develop heart dysfunction with aging or after crossing with mice over expressing peroxisome proliferator-activated receptor γ (PPARγ) in the heart. MHC-DGAT1 transgenic mice had increased heart triglyceride but no evidence of heart dysfunction, even up to age 12 months. The MHC-DGAT1 transgene improved heart dysfunction and survival of MHC-PPARγ-expressing transgenic mice. Both diacylglycerol and ceramide levels in the heart were reduced by this cross, as were the levels of several mRNAs of genes involved in lipid metabolism. There were fewer large lipid droplets in MHC-DGAT1×MHC-PPARγ mice compared with MHC-PPARγ, but total lipid content was not changed. Therefore, overexpression of DGAT1 is not toxic to the heart but reduces levels of toxic lipids and improves lipotoxic cardiomyopathy. Moreover, the beneficial effects of DGAT1 illustrate the interrelationship of several lipid metabolic pathways and the difficulty of assigning benefit to an isolated change in one potentially toxic lipid species.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/genética , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , PPAR gamma/genética , Animales , Transporte Biológico/genética , Femenino , Expresión Génica , Corazón/fisiología , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/citología , Cadenas Pesadas de Miosina/genética , Proteína Quinasa C/metabolismo , Transducción de Señal/genética , Análisis de Supervivencia
6.
J Biol Chem ; 286(42): 36331-9, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21873422

RESUMEN

Septic shock results from bacterial infection and is associated with multi-organ failure, high mortality, and cardiac dysfunction. Sepsis causes both myocardial inflammation and energy depletion. We hypothesized that reduced cardiac energy production is a primary cause of ventricular dysfunction in sepsis. The JNK pathway is activated in sepsis and has also been implicated in impaired fatty acid oxidation in several tissues. Therefore, we tested whether JNK activation inhibits cardiac fatty acid oxidation and whether blocking JNK would restore fatty acid oxidation during LPS treatment. LPS treatment of C57BL/6 mice and adenovirus-mediated activation of the JNK pathway in cardiomyocytes inhibited peroxisome proliferator-activated receptor α expression and fatty acid oxidation. Surprisingly, none of the adaptive responses that have been described in other types of heart failure, such as increased glucose utilization, reduced αMHC:ßMHC ratio or induction of certain microRNAs, occurred in LPS-treated mice. Treatment of C57BL/6 mice with a general JNK inhibitor (SP600125) increased fatty acid oxidation in mice and a cardiomyocyte-derived cell line. JNK inhibition also prevented LPS-mediated reduction in fatty acid oxidation and cardiac dysfunction. Inflammation was not alleviated in LPS-treated mice that received the JNK inhibitor. We conclude that activation of JNK signaling reduces fatty acid oxidation and prevents the peroxisome proliferator-activated receptor α down-regulation that occurs with LPS.


Asunto(s)
Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Cardiopatías/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/toxicidad , Proteínas Musculares/biosíntesis , Miocardio/metabolismo , PPAR alfa/biosíntesis , Animales , Antracenos/farmacología , Línea Celular , Ácidos Grasos/genética , Cardiopatías/inducido químicamente , Cardiopatías/genética , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Ratones Noqueados , Proteínas Musculares/antagonistas & inhibidores , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción/efectos de los fármacos , PPAR alfa/genética
7.
Circulation ; 124(25): 2812-21, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22124376

RESUMEN

BACKGROUND: Diabetes mellitus and obesity, which confer an increased risk of sudden cardiac death, are associated with cardiomyocyte lipid accumulation and altered cardiac electric properties, manifested by prolongation of the QRS duration and QT interval. It is difficult to distinguish the contribution of cardiomyocyte lipid accumulation from the contribution of global metabolic defects to the increased incidence of sudden death and electric abnormalities. METHODS AND RESULTS: In order to study the effects of metabolic abnormalities on arrhythmias without the complex systemic effects of diabetes mellitus and obesity, we studied transgenic mice with cardiac-specific overexpression of peroxisome proliferator-activated receptor γ 1 (PPARγ1) via the cardiac α-myosin heavy-chain promoter. The PPARγ transgenic mice develop abnormal accumulation of intracellular lipids and die as young adults before any significant reduction in systolic function. Using implantable ECG telemeters, we found that these mice have prolongation of the QRS and QT intervals and spontaneous ventricular arrhythmias, including polymorphic ventricular tachycardia and ventricular fibrillation. Isolated cardiomyocytes demonstrated prolonged action potential duration caused by reduced expression and function of the potassium channels responsible for repolarization. Short-term exposure to pioglitazone, a PPARγ agonist, had no effect on mortality or rhythm in WT mice but further exacerbated the arrhythmic phenotype and increased the mortality in the PPARγ transgenic mice. CONCLUSIONS: Our findings support an important link between PPARγ activation, cardiomyocyte lipid accumulation, ion channel remodeling, and increased cardiac mortality.


Asunto(s)
PPAR gamma/genética , Periodo Refractario Electrofisiológico/fisiología , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/fisiopatología , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Muerte Súbita Cardíaca/epidemiología , Modelos Animales de Enfermedad , Electrocardiografía , Hipoglucemiantes/farmacología , Incidencia , Lípido A/metabolismo , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , PPAR gamma/fisiología , Fenotipo , Pioglitazona , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología , Periodo Refractario Electrofisiológico/efectos de los fármacos , Sodio/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/mortalidad , Tiazolidinedionas/farmacología , Fibrilación Ventricular/genética , Fibrilación Ventricular/mortalidad , Remodelación Ventricular/fisiología
8.
Hum Mol Genet ; 19(1): 170-80, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19837698

RESUMEN

Mutations in SCO2, a protein required for the proper assembly and functioning of cytochrome c oxidase (COX; complex IV of the mitochondrial respiratory chain), cause a fatal infantile cardioencephalomyopathy with COX deficiency. We have generated mice harboring a Sco2 knock-out (KO) allele and a Sco2 knock-in (KI) allele expressing an E-->K mutation at position 129 (E129K), corresponding to the E140K mutation found in almost all human SCO2-mutated patients. Whereas homozygous KO mice were embryonic lethals, homozygous KI and compound heterozygous KI/KO mice were viable, but had muscle weakness; biochemically, they had respiratory chain deficiencies as well as complex IV assembly defects in multiple tissues. There was a concomitant reduction in mitochondrial copper content, but the total amount of copper in examined tissues was not reduced. These mouse models should be of use in further studies of Sco2 function, as well as in testing therapeutic approaches to treat the human disorder.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/enzimología , Deficiencia de Citocromo-c Oxidasa/genética , Complejo IV de Transporte de Electrones/genética , Mutación/genética , Animales , Western Blotting , Cobre/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Pruebas de Enzimas , Inmunohistoquímica , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Chaperonas Moleculares , Músculos/enzimología , Músculos/patología , Especificidad de Órganos
9.
Circ J ; 76(9): 2148-52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22785609

RESUMEN

BACKGROUND: Chronic heart failure is associated with higher risk for developing diabetes mellitus. Secretory products from adipocytes may contribute to the deterioration in glycemic control and increased insulin resistance (IR). Retinol binding protein 4 (RBP4) is an adipose tissue-derived protein with pro-diabetogenic effects. The aim of the present study was to investigate the relationship of RBP4 in patients with heart failure. METHODS AND RESULTS: Serum levels of RBP4, insulin, and fasting glucose were assessed in 58 patients with severe heart failure at the time of left ventricular assist device (LVAD) implantation and in 44 patients at the time of explantation, as well as in 10 normal control subjects. Serum RBP4 levels were measured by specific enzyme-linked immunosorbent assay, and IR was assessed using the homeostatic model of IR (HOMA-IR). Fasting glucose, insulin and HOMA-IR were significantly higher in patients at the time of LVAD implantation compared to controls (all P<0.01). RBP-4 and HOMA-IR significantly decreased after LVAD implantation (21.7 ± 8.8 mg/dl to 16.0 ± 3.8 mg/dl, P<0.05; 4.2 ± 2.7 to 2.5 ± 2.0, P<0.01). CONCLUSIONS: Patients with advanced heart failure have increased levels of RBP4, and LVAD implantation reduces RBP4. These findings implicate RBP4 in the cascade of reversible metabolic derangements in advanced heart failure.


Asunto(s)
Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Hemodinámica , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Adulto , Anciano , Glucemia/metabolismo , Ensayo de Inmunoadsorción Enzimática , Ayuno/sangre , Femenino , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad
10.
J Lipid Res ; 52(4): 732-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21205704

RESUMEN

Diacylglycerol (DAG) acyl transferase 1 (Dgat1) knockout ((-/-)) mice are resistant to high-fat-induced obesity and insulin resistance, but the reasons are unclear. Dgat1(-/-) mice had reduced mRNA levels of all three Ppar genes and genes involved in fatty acid oxidation in the myocardium of Dgat1(-/-) mice. Although DGAT1 converts DAG to triglyceride (TG), tissue levels of DAG were not increased in Dgat1(-/-) mice. Hearts of chow-diet Dgat1(-/-) mice were larger than those of wild-type (WT) mice, but cardiac function was normal. Skeletal muscles from Dgat1(-/-) mice were also larger. Muscle hypertrophy factors phospho-AKT and phospho-mTOR were increased in Dgat1(-/-) cardiac and skeletal muscle. In contrast to muscle, liver from Dgat1(-/-) mice had no reduction in mRNA levels of genes mediating fatty acid oxidation. Glucose uptake was increased in cardiac and skeletal muscle in Dgat1(-/-) mice. Treatment with an inhibitor specific for DGAT1 led to similarly striking reductions in mRNA levels of genes mediating fatty acid oxidation in cardiac and skeletal muscle. These changes were reproduced in cultured myocytes with the DGAT1 inhibitor, which also blocked the increase in mRNA levels of Ppar genes and their targets induced by palmitic acid. Thus, loss of DGAT1 activity in muscles decreases mRNA levels of genes involved in lipid uptake and oxidation.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Receptores Activados del Proliferador del Peroxisoma/genética , Absorciometría de Fotón , Animales , Western Blotting , Línea Celular , Ceramidas/metabolismo , Diacilglicerol O-Acetiltransferasa/deficiencia , Diacilglicerol O-Acetiltransferasa/genética , Diglicéridos/metabolismo , Ecocardiografía , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Glucosa/metabolismo , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Tamaño de los Órganos/genética , Ácido Palmítico/farmacología , Reacción en Cadena de la Polimerasa
11.
Am J Physiol Endocrinol Metab ; 300(3): E489-99, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21139071

RESUMEN

Normal hearts have increased contractility in response to catecholamines. Because several lipids activate PKCs, we hypothesized that excess cellular lipids would inhibit cardiomyocyte responsiveness to adrenergic stimuli. Cardiomyocytes treated with saturated free fatty acids, ceramide, and diacylglycerol had reduced cellular cAMP response to isoproterenol. This was associated with increased PKC activation and reduction of ß-adrenergic receptor (ß-AR) density. Pharmacological and genetic PKC inhibition prevented both palmitate-induced ß-AR insensitivity and the accompanying reduction in cell surface ß-ARs. Mice with excess lipid uptake due to either cardiac-specific overexpression of anchored lipoprotein lipase, PPARγ, or acyl-CoA synthetase-1 or high-fat diet showed reduced inotropic responsiveness to dobutamine. This was associated with activation of protein kinase C (PKC)α or PKCδ. Thus, several lipids that are increased in the setting of lipotoxicity can produce abnormalities in ß-AR responsiveness. This can be attributed to PKC activation and reduced ß-AR levels.


Asunto(s)
Lípidos/fisiología , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/fisiología , Receptores Adrenérgicos beta/fisiología , Animales , Western Blotting , Ceramidas/metabolismo , AMP Cíclico/metabolismo , Dieta , Grasas de la Dieta/farmacología , Diglicéridos/metabolismo , Ecocardiografía , Activación Enzimática/fisiología , Cromatografía de Gases y Espectrometría de Masas , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Miocitos Cardíacos/enzimología , ARN/genética , ARN/aislamiento & purificación , ARN Interferente Pequeño/genética
12.
Curr Opin Clin Nutr Metab Care ; 13(2): 145-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20010095

RESUMEN

PURPOSE OF REVIEW: Diseases associated with ectopic disposition of lipids are becoming an increasingly important medical problem as the incidence of type 2 diabetes and obesity increases. One of the organs affected by lipotoxicity is the heart and this review presents an update on human and animal studies of this problem. RECENT FINDINGS: Human studies have clearly correlated heart dysfunction with the content of triglyceride. More recently human heart samples have been used to assess gene changes associated with altered lipid accumulation. Genetically altered mice have been created that develop lipotoxic cardiomyopathies and newer investigations are attempting to delineate curative therapies. SUMMARY: Human studies will confirm the metabolic changes associated with lipotoxic cardiomyopathy and, hopefully, animal studies will guide treatment options.


Asunto(s)
Cardiomiopatías/etiología , Miocardio/metabolismo , Triglicéridos/metabolismo , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Expresión Génica , Humanos , Triglicéridos/genética
14.
Sci Rep ; 4: 4257, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24604065

RESUMEN

There is a great need for the development of therapeutic strategies that can target biomolecules to damaged myocardium. Necrosis of myocardium during a myocardial infarction (MI) is characterized by extracellular release of DNA, which can serve as a potential target for ischemic tissue. Hoechst, a histological stain that binds to double-stranded DNA can be conjugated to a variety of molecules. Insulin-like growth factor-1 (IGF-1), a small protein/polypeptide with a short circulating-half life is cardioprotective following MI but its clinical use is limited by poor delivery, as intra-myocardial injections have poor retention and chronic systemic presence has adverse side effects. Here, we present a novel delivery vehicle for IGF-1, via its conjugation to Hoechst for targeting infarcted tissue. Using a mouse model of ischemia-reperfusion, we demonstrate that intravenous delivery of Hoechst-IGF-1 results in activation of Akt, a downstream target of IGF-1 and protects from cardiac fibrosis and dysfunction following MI.


Asunto(s)
ADN/metabolismo , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Espacio Extracelular/metabolismo , Fibrosis , Humanos , Factor I del Crecimiento Similar a la Insulina/química , Macrófagos/metabolismo , Masculino , Ratones , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Unión Proteica , Transporte de Proteínas
15.
Biomaterials ; 34(31): 7790-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23856052

RESUMEN

Myocardial infarction (MI) is the most common cause of heart failure (HF), the leading cause of death in the developed world. Oxidative stress due to excessive production of reactive oxygen species (ROS) plays a key role in the pathogenesis of cardiac remodeling leading to HF. NADPH oxidase with Nox2 as the catalytic subunit is a major source for cardiac ROS production. Nox2-NADPH expression is significantly increased in the infarcted myocardium, primarily in neutrophils, macrophages and myocytes. Moreover, mice lacking the Nox2 gene are protected from ischemic injury, implicating Nox2 as a potential therapeutic target. RNAi-mediated gene silencing holds great promise as a therapeutic owing to its high specificity and potency. However, in vivo delivery hurdles have limited its effective clinical use. Here, we demonstrate acid-degradable polyketal particles as delivery vehicles for Nox2-siRNA to the post-MI heart. In vitro, Nox2-siRNA particles are effectively taken up by macrophages and significantly knockdown Nox2 expression and activity. Following in vivo intramyocardial injection in experimental mice models of MI, Nox2-siRNA particles prevent upregulation of Nox2 and significantly recovered cardiac function. This study highlights the potential of polyketals as siRNA delivery vehicles to the MI heart and represents a viable therapeutic approach for targeting oxidative stress.


Asunto(s)
Glicoproteínas de Membrana/genética , Infarto del Miocardio/terapia , NADPH Oxidasas/genética , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/genética , Animales , Línea Celular , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , Nanopartículas/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química
16.
Circ Heart Fail ; 6(3): 550-62, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23572494

RESUMEN

BACKGROUND: Cardiac dysfunction with sepsis is associated with both inflammation and reduced fatty acid oxidation. We hypothesized that energy deprivation accounts for sepsis-related cardiac dysfunction. METHODS AND RESULTS: Escherichia coli lipopolysaccharide (LPS) administered to C57BL/6 mice (wild type) induced cardiac dysfunction and reduced fatty acid oxidation and mRNA levels of peroxisome proliferator-activated receptor (PPAR)-α and its downstream targets within 6-8 hours. Transgenic mice in which cardiomyocyte-specific expression of PPARγ is driven by the α-myosin heavy chain promoter (αMHC-PPARγ) were protected from LPS-induced cardiac dysfunction. Despite a reduction in PPARα, fatty acid oxidation and associated genes were not decreased in hearts of LPS-treated αMHC-PPARγ mice. LPS treatment, however, continued to induce inflammation-related genes, such as interleukin-1α, interleukin-1ß, interleukin-6, and tumor necrosis factor-α in hearts of αMHC-PPARγ mice. Treatment of wild-type mice with LPS and the PPARγ agonist, rosiglitazone, but not the PPARα agonist (WY-14643), increased fatty acid oxidation, prevented LPS-mediated reduction of mitochondria, and treated cardiac dysfunction, as well as it improved survival, despite continued increases in the expression of cardiac inflammatory markers. CONCLUSIONS: Activation of PPARγ in LPS-treated mice prevented cardiac dysfunction and mortality, despite development of cardiac inflammation and PPARα downregulation.


Asunto(s)
Cardiopatías/fisiopatología , PPAR gamma/metabolismo , Sepsis/complicaciones , Animales , Ácidos Grasos/metabolismo , Cardiopatías/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , PPAR gamma/agonistas , Rosiglitazona , Sepsis/fisiopatología , Sepsis/terapia , Tiazolidinedionas/farmacología , Disfunción Ventricular Izquierda/fisiopatología
17.
PLoS One ; 7(9): e46549, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029549

RESUMEN

Aldose reductase (AR), an enzyme mediating the first step in the polyol pathway of glucose metabolism, is associated with complications of diabetes mellitus and increased cardiac ischemic injury. We investigated whether deleterious effects of AR are due to its actions specifically in cardiomyocytes. We created mice with cardiac specific expression of human AR (hAR) using the α-myosin heavy chain (MHC) promoter and studied these animals during aging and with reduced fatty acid (FA) oxidation. hAR transgenic expression did not alter cardiac function or glucose and FA oxidation gene expression in young mice. However, cardiac overexpression of hAR caused cardiac dysfunction in older mice. We then assessed whether hAR altered heart function during ischemia reperfusion. hAR transgenic mice had greater infarct area and reduced functional recovery than non-transgenic littermates. When the hAR transgene was crossed onto the PPAR alpha knockout background, another example of greater heart glucose oxidation, hAR expressing mice had increased heart fructose content, cardiac fibrosis, ROS, and apoptosis. In conclusion, overexpression of hAR in cardiomyocytes leads to cardiac dysfunction with aging and in the setting of reduced FA and increased glucose metabolism. These results suggest that pharmacological inhibition of AR will be beneficial during ischemia and in some forms of heart failure.


Asunto(s)
Aldehído Reductasa/fisiología , Insuficiencia Cardíaca/enzimología , Isquemia Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Aldehído Reductasa/biosíntesis , Aldehído Reductasa/genética , Animales , Apoptosis , Ceramidas/metabolismo , Ácidos Grasos/metabolismo , Fibrosis/enzimología , Fructosa/metabolismo , Glucosa/metabolismo , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Humanos , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/fisiopatología , Miocardio/enzimología , Miocardio/metabolismo , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Oxidación-Reducción , PPAR alfa/genética , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Recuperación de la Función , Daño por Reperfusión/enzimología , Daño por Reperfusión/fisiopatología , Ultrasonografía
18.
Circ Heart Fail ; 5(3): 340-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22379072

RESUMEN

BACKGROUND: Heart failure (HF) is characterized by inflammation, insulin resistance, and progressive catabolism. We hypothesized that patients with advanced HF also develop adipose tissue inflammation associated with impaired adipokine signaling and that hemodynamic correction through implantation of ventricular assist devices (VADs) would reverse adipocyte activation and correct adipokine signaling in advanced HF. METHODS AND RESULTS: Circulating insulin, adiponectin, leptin, and resistin levels were measured in 36 patients with advanced HF before and after VAD implantation and 10 healthy control subjects. Serum adiponectin was higher in HF patients before VAD implantation compared with control subjects (13.3±4.9 versus 6.4±2.1 µg/mL, P=0.02). VAD implantation (mean, 129±99 days) reduced serum adiponectin (7.4±3.4 µg/mL, P<0.05) and improved insulin resistance (Homeostasis Assessment Model of insulin resistance: 7.6±7.7-4.5±3.6; P=0.012). [corrected] Adiponectin expression in adipose tissue decreased after VAD implantation (-65%; P<0.03). Adiponectin receptor expression was suppressed in the failing myocardium compared with control subjects and increased after mechanical unloading. Histomorphometric analysis of adipose tissue specimens revealed reduced adipocyte size in patients with advanced HF compared with control subjects (2105±585 µm(2) [corrected] versus 5583±142 µm(2) in control subjects; P<0.05), which increased after VAD placement. Of note, macrophage infiltration in adipose tissue was higher in advanced HF patients compared with control subjects (+25%; P<0.01), which normalized after VAD implantation. CONCLUSIONS: Adipose tissue inflammation and adiponectin resistance develop in advanced HF. Mechanical unloading of the failing myocardium reverses adipose tissue macrophage infiltration, inflammation, and adiponectin resistance in patients with advanced HF.


Asunto(s)
Adiponectina/sangre , Tejido Adiposo/patología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Inflamación/patología , Índice de Severidad de la Enfermedad , Adulto , Anciano , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/sangre , Hemodinámica/fisiología , Humanos , Insulina/sangre , Resistencia a la Insulina/fisiología , Leptina/sangre , Macrófagos/patología , Masculino , Persona de Mediana Edad , Resistina/sangre , Estudios Retrospectivos , Transducción de Señal/fisiología
19.
J Invasive Cardiol ; 19(12): E378-80, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18180533

RESUMEN

Left ventricular apical ballooning is an increasingly reported phenomenon with an onset that is usually triggered by severe and often acute emotional incidents. We report a rare case of acute left ventricular apical ballooning syndrome, mimicking acute ST-elevation myocardial infarction, in a post menopausal woman whose only predisposing factor was an all-night cocaine binge.


Asunto(s)
Trastornos Relacionados con Cocaína/complicaciones , Disfunción Ventricular Izquierda/inducido químicamente , Angiografía Coronaria , Diagnóstico Diferencial , Ecocardiografía , Electrocardiografía , Femenino , Estudios de Seguimiento , Imagen de Acumulación Sanguínea de Compuerta , Humanos , Persona de Mediana Edad , Recuperación de la Función , Volumen Sistólico , Disfunción Ventricular Izquierda/diagnóstico , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda