Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Lipid Res ; 63(11): 100274, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36115595

RESUMEN

Lipid accumulation in nonadipose tissues can cause lipotoxicity, leading to cell death and severe organ dysfunction. Adipose triglyceride lipase (ATGL) deficiency causes human neutral lipid storage disease and leads to cardiomyopathy; ATGL deficiency has no current treatment. One possible approach to alleviate this disorder has been to alter the diet and reduce the supply of dietary lipids and, hence, myocardial lipid uptake. However, in this study, when we supplied cardiac Atgl KO mice a low- or high-fat diet, we found that heart lipid accumulation, heart dysfunction, and death were not altered. We next deleted lipid uptake pathways in the ATGL-deficient mice through the generation of double KO mice also deficient in either cardiac lipoprotein lipase or cluster of differentiation 36, which is involved in an lipoprotein lipase-independent pathway for FA uptake in the heart. We show that neither deletion ameliorated ATGL-deficient heart dysfunction. Similarly, we determined that non-lipid-containing media did not prevent lipid accumulation by cultured myocytes; rather, the cells switched to increased de novo FA synthesis. Thus, we conclude that pathological storage of lipids in ATGL deficiency cannot be corrected by reducing heart lipid uptake.


Asunto(s)
Aciltransferasas , Cardiomiopatías , Lipoproteína Lipasa , Animales , Humanos , Ratones , Tejido Adiposo/metabolismo , Cardiomiopatías/metabolismo , Lipasa/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones Noqueados , Miocardio/metabolismo , Triglicéridos/metabolismo , Aciltransferasas/deficiencia , Aciltransferasas/genética
2.
Am J Transl Res ; 12(12): 7737-7761, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33437358

RESUMEN

Endothelial cells (ECs) maintain vascular integrity and mediate vascular repair and angiogenesis, by which new blood vessels are formed from pre-existing blood vessels. Hyperglycemia has been shown to increase EC angiogenic potential. However, few studies have investigated effects of fatty acids (FAs) on EC angiogenesis. Cluster of differentiation 36 (CD36) is a FA transporter expressed by ECs, but its role in EC proliferation, migration, and angiogenesis is unknown. We sought to determine if circulating FAs regulate angiogenic function in a CD36-dependent manner. CD36-dependent effects of FAs on EC proliferation and migration of mouse heart ECs (MHECs) and lung ECs (MLECs) were studied. We used both silencing RNA and antisense oligonucleotides to reduce CD36 expression. Oleic acid (OA) did not affect EC proliferation, but significantly increased migration of ECs in wound healing experiments. CD36 knockdown prevented OA-induced increases in wound healing potential. In EC transwell migration experiments, OA increased recruitment and migration of ECs, an effect abolished by CD36 knockdown. Phospho-AMP-activated protein kinase (AMPK) increased in MHECs exposed to OA in a CD36-dependent manner. To test whether in vivo CD36 affects angiogenesis, we studied 21-day recovery in post-hindlimb ischemia. EC-specific CD36 knockout mice had reduced blood flow recovery as assessed by laser Doppler imaging. EC content in post-ischemic muscle, assessed from CD31 expression, increased in ischemic muscle of control mice. However, mice with EC-specific CD36 deletion lacked the increase in CD31 and matrix metalloprotease 9 expression observed in controls. EC expression of CD36 and its function in FA uptake modulate angiogenic function and response to ischemia, likely due to reduced activation of the AMPK pathway.

4.
J Diabetes Complications ; 32(11): 1027-1034, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30197161

RESUMEN

OBJECTIVE: Systemic inflammation contributes to cardiovascular disease in patients with type 2 diabetes, and elevated white blood cell (WBC) counts are an established risk factor. Our goal is to describe changes in WBCs and inflammatory markers after glycemic reductions in diabetes. RESEARCH DESIGN AND METHODS: This study enrolled 63 subjects with poorly controlled diabetes, defined as hemoglobin A1c (HbA1c) ≥8% [64 mmol/mol]. Circulating granulocytes and mononuclear cells were separated by histopaque double-density protocol. Inflammatory markers from these isolated WBCs were assessed at baseline and after 3 months of medical management. RESULTS: After 3 months, significant glycemic reduction, defined as a decrease in HbA1c ≥ 1.5%, occurred in 42 subjects. Fasting plasma glucose decreased by 47% (165.6 mg/dL), and HbA1c decreased from 10.2 ±â€¯1.8 to 6.8 ±â€¯0.9. Glycemic reductions were associated with a 9.4% decrease in total WBC counts, 10.96% decrease in neutrophils, and 21.74% decrease in monocytes. The mRNA levels of inflammatory markers from granulocytes and mononuclear cells decreased, including receptor for advanced glycation endproducts; S100 calcium binding proteins A8, A9, A12; krüppel-like factor 5; and IL-1. Also, circulating levels of IL-1ß and C-reactive protein decreased. Insulin dose was a mediator between HbA1c and both total WBC and neutrophil counts, but not changes in WBC inflammatory markers. In contrast, the 17 subjects without significant glycemic reductions showed no significant differences in their WBC counts and proteins of inflammatory genes. CONCLUSION: Significant glycemic reduction in subjects with poorly controlled diabetes led to reduced circulating WBC counts and inflammatory gene expression.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Inflamación/genética , Recuento de Leucocitos , Adulto , Biomarcadores/sangre , Estudios de Cohortes , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Regulación hacia Abajo/genética , Femenino , Expresión Génica , Hemoglobina Glucada/análisis , Hemoglobina Glucada/metabolismo , Humanos , Inflamación/sangre , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad
5.
J Clin Invest ; 128(10): 4329-4342, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30047927

RESUMEN

Movement of circulating fatty acids (FAs) to parenchymal cells requires their transfer across the endothelial cell (EC) barrier. The multiligand receptor cluster of differentiation 36 (CD36) facilitates tissue FA uptake and is expressed in ECs and parenchymal cells such as myocytes and adipocytes. Whether tissue uptake of FAs is dependent on EC or parenchymal cell CD36, or both, is unknown. Using a cell-specific deletion approach, we show that EC, but not parenchymal cell, CD36 deletion increased fasting plasma FAs and postprandial triglycerides. EC-Cd36-KO mice had reduced uptake of radiolabeled long-chain FAs into heart, skeletal muscle, and brown adipose tissue; these uptake studies were replicated using [11C]palmitate PET scans. High-fat diet-fed EC-CD36-deficient mice had improved glucose tolerance and insulin sensitivity. Both EC and cardiomyocyte (CM) deletion of CD36 reduced heart lipid droplet accumulation after fasting, but CM deletion did not affect heart glucose or FA uptake. Expression in the heart of several genes modulating glucose metabolism and insulin action increased with EC-CD36 deletion but decreased with CM deletion. In conclusion, EC CD36 acts as a gatekeeper for parenchymal cell FA uptake, with important downstream effects on glucose utilization and insulin action.


Asunto(s)
Antígenos CD36/metabolismo , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Transporte Biológico Activo/genética , Antígenos CD36/genética , Células Endoteliales/patología , Ácidos Grasos/genética , Glucosa/genética , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Ratones , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/patología , Especificidad de Órganos
6.
Diabetes ; 67(7): 1272-1284, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29748289

RESUMEN

During reduced energy intake, skeletal muscle maintains homeostasis by rapidly suppressing insulin-stimulated glucose utilization. Loss of this adaptation is observed with deficiency of the fatty acid transporter CD36. A similar loss is also characteristic of the insulin-resistant state where CD36 is dysfunctional. To elucidate what links CD36 to muscle glucose utilization, we examined whether CD36 signaling might influence insulin action. First, we show that CD36 deletion specific to skeletal muscle reduces expression of insulin signaling and glucose metabolism genes. It decreases muscle ceramides but impairs glucose disposal during a meal. Second, depletion of CD36 suppresses insulin signaling in primary-derived human myotubes, and the mechanism is shown to involve functional CD36 interaction with the insulin receptor (IR). CD36 promotes tyrosine phosphorylation of IR by the Fyn kinase and enhances IR recruitment of P85 and downstream signaling. Third, pretreatment for 15 min with saturated fatty acids suppresses CD36-Fyn enhancement of IR phosphorylation, whereas unsaturated fatty acids are neutral or stimulatory. These findings define mechanisms important for muscle glucose metabolism and optimal insulin responsiveness. Potential human relevance is suggested by genome-wide analysis and RNA sequencing data that associate genetically determined low muscle CD36 expression to incidence of type 2 diabetes.


Asunto(s)
Antígenos CD36/fisiología , Glucosa/metabolismo , Insulina/metabolismo , Músculo Esquelético/metabolismo , Receptor de Insulina/metabolismo , Animales , Antígenos CD36/genética , Células CHO , Metabolismo de los Hidratos de Carbono/genética , Células Cultivadas , Cricetinae , Cricetulus , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Femenino , Humanos , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética
7.
J Lipid Res ; 58(6): 1132-1142, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28404638

RESUMEN

Lipid accumulation is a pathological feature of every type of kidney injury. Despite this striking histological feature, physiological accumulation of lipids in the kidney is poorly understood. We studied whether the accumulation of lipids in the fasted kidney are derived from lipoproteins or NEFAs. With overnight fasting, kidneys accumulated triglyceride, but had reduced levels of ceramide and glycosphingolipid species. Fasting led to a nearly 5-fold increase in kidney uptake of plasma [14C]oleic acid. Increasing circulating NEFAs using a ß adrenergic receptor agonist caused a 15-fold greater accumulation of lipid in the kidney, while mice with reduced NEFAs due to adipose tissue deficiency of adipose triglyceride lipase had reduced triglycerides. Cluster of differentiation (Cd)36 mRNA increased 2-fold, and angiopoietin-like 4 (Angptl4), an LPL inhibitor, increased 10-fold. Fasting-induced kidney lipid accumulation was not affected by inhibition of LPL with poloxamer 407 or by use of mice with induced genetic LPL deletion. Despite the increase in CD36 expression with fasting, genetic loss of CD36 did not alter fatty acid uptake or triglyceride accumulation. Our data demonstrate that fasting-induced triglyceride accumulation in the kidney correlates with the plasma concentrations of NEFAs, but is not due to uptake of lipoprotein lipids and does not involve the fatty acid transporter, CD36.


Asunto(s)
Ayuno/sangre , Ayuno/metabolismo , Ácidos Grasos no Esterificados/sangre , Riñón/metabolismo , Triglicéridos/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
8.
Cell Mol Gastroenterol Hepatol ; 3(1): 82-98, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28066800

RESUMEN

BACKGROUND & AIMS: CD36 has immuno-metabolic actions and is abundant in the small intestine on epithelial, endothelial and immune cells. We examined the role of CD36 in gut homeostasis using mice null for CD36 (CD36KO) and with CD36 deletion specific to enterocytes (Ent-CD36KO) or endothelial cells (EC-CD36KO). METHODS: Intestinal morphology was evaluated using immunohistochemistry and electron microscopy (EM). Intestinal inflammation was determined from neutrophil infiltration and expression of cytokines, toll-like receptors and COX-2. Barrier integrity was assessed from circulating lipopolysaccharide (LPS) and dextran administered intragastrically. Epithelial permeability to luminal dextran was visualized using two photon microscopy. RESULTS: The small intestines of CD36KO mice fed a chow diet showed several abnormalities including extracellular matrix (ECM) accumulation with increased expression of ECM proteins, evidence of neutrophil infiltration, inflammation and compromised barrier function. EM showed shortened desmosomes with decreased desmocollin 2 expression. Systemically, leukocytosis and neutrophilia were present together with 80% reduction of anti-inflammatory Ly6Clow monocytes. Bone marrow transplants supported the primary contribution of non-hematopoietic cells to the inflammatory phenotype. Specific deletion of endothelial but not of enterocyte CD36 reproduced many of the gut phenotypes of germline CD36KO mice including fibronectin deposition, increased interleukin 6, neutrophil infiltration, desmosome shortening and impaired epithelial barrier function. CONCLUSIONS: CD36 loss results in chronic neutrophil infiltration of the gut, impairs barrier integrity and systemically causes subclinical inflammation. Endothelial cell CD36 deletion reproduces the major intestinal phenotypes. The findings suggest an important role of the endothelium in etiology of gut inflammation and loss of epithelial barrier integrity.

9.
J Clin Invest ; 126(8): 2855-66, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27400128

RESUMEN

Hypertriglyceridemia is an independent risk factor for cardiovascular disease, and plasma triglycerides (TGs) correlate strongly with plasma apolipoprotein C-III (ApoC-III) levels. Antisense oligonucleotides (ASOs) for ApoC-III reduce plasma TGs in primates and mice, but the underlying mechanism of action remains controversial. We determined that a murine-specific ApoC-III-targeting ASO reduces fasting TG levels through a mechanism that is dependent on low-density lipoprotein receptors (LDLRs) and LDLR-related protein 1 (LRP1). ApoC-III ASO treatment lowered plasma TGs in mice lacking lipoprotein lipase (LPL), hepatic heparan sulfate proteoglycan (HSPG) receptors, LDLR, or LRP1 and in animals with combined deletion of the genes encoding HSPG receptors and LDLRs or LRP1. However, the ApoC-III ASO did not lower TG levels in mice lacking both LDLR and LRP1. LDLR and LRP1 were also required for ApoC-III ASO-induced reduction of plasma TGs in mice fed a high-fat diet, in postprandial clearance studies, and when ApoC-III-rich or ApoC-III-depleted lipoproteins were injected into mice. ASO reduction of ApoC-III had no effect on VLDL secretion, heparin-induced TG reduction, or uptake of lipids into heart and skeletal muscle. Our data indicate that ApoC-III inhibits turnover of TG-rich lipoproteins primarily through a hepatic clearance mechanism mediated by the LDLR/LRP1 axis.


Asunto(s)
Apolipoproteína C-III/sangre , Lipoproteínas/sangre , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptores de LDL/metabolismo , Triglicéridos/sangre , Proteínas Supresoras de Tumor/metabolismo , Animales , Femenino , Genotipo , Heparina/farmacología , Hepatocitos/metabolismo , Cetonas/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Factores de Riesgo
10.
J Biol Chem ; 289(43): 29881-91, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25157099

RESUMEN

Diacylglycerol acyltransferase 1 (DGAT1) catalyzes the final step in triglyceride synthesis, the conversion of diacylglycerol (DAG) to triglyceride. Dgat1(-/-) mice exhibit a number of beneficial metabolic effects including reduced obesity and improved insulin sensitivity and no known cardiac dysfunction. In contrast, failing human hearts have severely reduced DGAT1 expression associated with accumulation of DAGs and ceramides. To test whether DGAT1 loss alone affects heart function, we created cardiomyocyte-specific DGAT1 knock-out (hDgat1(-/-)) mice. hDgat1(-/-) mouse hearts had 95% increased DAG and 85% increased ceramides compared with floxed controls. 50% of these mice died by 9 months of age. The heart failure marker brain natriuretic peptide increased 5-fold in hDgat1(-/-) hearts, and fractional shortening (FS) was reduced. This was associated with increased expression of peroxisome proliferator-activated receptor α and cluster of differentiation 36. We crossed hDgat1(-/-) mice with previously described enterocyte-specific Dgat1 knock-out mice (hiDgat1(-/-)). This corrected the early mortality, improved FS, and reduced cardiac ceramide and DAG content. Treatment of hDgat1(-/-) mice with the glucagon-like peptide 1 receptor agonist exenatide also improved FS and reduced heart DAG and ceramide content. Increased fatty acid uptake into hDgat1(-/-) hearts was normalized by exenatide. Reduced activation of protein kinase Cα (PKCα), which is increased by DAG and ceramides, paralleled the reductions in these lipids. Our mouse studies show that loss of DGAT1 reproduces the lipid abnormalities seen in severe human heart failure.


Asunto(s)
Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/enzimología , Lípidos/sangre , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Envejecimiento/patología , Animales , Glucemia/metabolismo , Colesterol/sangre , Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Diacilglicerol O-Acetiltransferasa/metabolismo , Inhibidores Enzimáticos/farmacología , Exenatida , Ácidos Grasos/sangre , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/genética , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Especificidad de Órganos , Péptidos/farmacología , Fenotipo , Proteína Quinasa C/metabolismo , Triglicéridos/sangre , Ponzoñas/farmacología
11.
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
12.
J Biol Chem ; 288(20): 14046-14058, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23542081

RESUMEN

Adipose fat storage is thought to require uptake of circulating triglyceride (TG)-derived fatty acids via lipoprotein lipase (LpL). To determine how LpL affects the biology of adipose tissue, we created adipose-specific LpL knock-out (ATLO) mice, and we compared them with whole body LpL knock-out mice rescued with muscle LpL expression (MCK/L0) and wild type (WT) mice. ATLO LpL mRNA and activity were reduced, respectively, 75 and 70% in gonadal adipose tissue (GAT), 90 and 80% in subcutaneous tissue, and 84 and 85% in brown adipose tissue (BAT). ATLO mice had increased plasma TG levels associated with reduced chylomicron TG uptake into BAT and lung. ATLO BAT, but not GAT, had altered TG composition. GAT from MCK/L0 was smaller and contained less polyunsaturated fatty acids in TG, although GAT from ATLO was normal unless LpL was overexpressed in muscle. High fat diet feeding led to less adipose in MCK/L0 mice but TG acyl composition in subcutaneous tissue and BAT reverted to that of WT. Therefore, adipocyte LpL in BAT modulates plasma lipoprotein clearance, and the greater metabolic activity of this depot makes its lipid composition more dependent on LpL-mediated uptake. Loss of adipose LpL reduces fat accumulation only if accompanied by greater LpL activity in muscle. These data support the role of LpL as the "gatekeeper" for tissue lipid distribution.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo/metabolismo , Lipoproteína Lipasa/deficiencia , Lipoproteína Lipasa/genética , Adipocitos/citología , Animales , Trasplante de Médula Ósea , Quilomicrones/farmacocinética , Lípidos/química , Lipólisis , Macrófagos/citología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Triglicéridos/sangre , Triglicéridos/metabolismo
13.
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
14.
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
15.
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
16.
J Clin Invest ; 120(10): 3443-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20852389

RESUMEN

Excess lipid accumulation in the heart is associated with decreased cardiac function in humans and in animal models. The reasons are unclear, but this is generally believed to result from either toxic effects of intracellular lipids or excessive fatty acid oxidation (FAO). PPARγ expression is increased in the hearts of humans with metabolic syndrome, and use of PPARγ agonists is associated with heart failure. Here, mice with dilated cardiomyopathy due to cardiomyocyte PPARγ overexpression were crossed with PPARα-deficient mice. Surprisingly, this cross led to enhanced expression of several PPAR-regulated genes that mediate fatty acid (FA) uptake/oxidation and triacylglycerol (TAG) synthesis. Although FA oxidation and TAG droplet size were increased, heart function was preserved and survival improved. There was no marked decrease in cardiac levels of triglyceride or the potentially toxic lipids diacylglycerol (DAG) and ceramide. However, long-chain FA coenzyme A (LCCoA) levels were increased, and acylcarnitine content was decreased. Activation of PKCα and PKCδ, apoptosis, ROS levels, and evidence of endoplasmic reticulum stress were also reduced. Thus, partitioning of lipid to storage and oxidation can reverse cardiolipotoxicity despite increased DAG and ceramide levels, suggesting a role for other toxic intermediates such as acylcarnitines in the toxic effects of lipid accumulation in the heart.


Asunto(s)
Ácidos Grasos/metabolismo , Lípidos/toxicidad , Miocardio/metabolismo , PPAR alfa/fisiología , PPAR gamma/fisiología , Animales , Apoptosis , Ácidos Grasos no Esterificados/sangre , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Miocitos Cardíacos/ultraestructura , Oxidación-Reducción , PPAR alfa/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/biosíntesis
17.
J Clin Invest ; 117(10): 2791-801, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17823655

RESUMEN

Three forms of PPARs are expressed in the heart. In animal models, PPARgamma agonist treatment improves lipotoxic cardiomyopathy; however, PPARgamma agonist treatment of humans is associated with peripheral edema and increased heart failure. To directly assess effects of increased PPARgamma on heart function, we created transgenic mice expressing PPARgamma1 in the heart via the cardiac alpha-myosin heavy chain (alpha-MHC) promoter. PPARgamma1-transgenic mice had increased cardiac expression of fatty acid oxidation genes and increased lipoprotein triglyceride (TG) uptake. Unlike in cardiac PPARalpha-transgenic mice, heart glucose transporter 4 (GLUT4) mRNA expression and glucose uptake were not decreased. PPARgamma1-transgenic mice developed a dilated cardiomyopathy associated with increased lipid and glycogen stores, distorted architecture of the mitochondrial inner matrix, and disrupted cristae. Thus, while PPARgamma agonists appear to have multiple beneficial effects, their direct actions on the myocardium have the potential to lead to deterioration in heart function.


Asunto(s)
Cardiomiopatía Dilatada/genética , Metabolismo de los Lípidos , PPAR gamma/metabolismo , Envejecimiento/genética , Animales , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Ácidos Grasos/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Glucógeno/metabolismo , Corazón/fisiopatología , Metabolismo de los Lípidos/genética , Ratones , Ratones Transgénicos , PPAR gamma/agonistas , PPAR gamma/genética , Regiones Promotoras Genéticas/genética , Rosiglitazona , Tiazolidinedionas/farmacología , Miosinas Ventriculares/genética
18.
J Lipid Res ; 48(3): 646-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17189607

RESUMEN

Regulation of cholesterol metabolism in cultured cells and in the liver is dependent on actions of the LDL receptor. However, nonhepatic tissues have multiple pathways of cholesterol uptake. One possible pathway is mediated by LPL, an enzyme that primarily hydrolyzes plasma triglyceride into fatty acids. In this study, LDL uptake and tissue cholesterol levels in heart and skeletal muscle of wild-type and transgenic mice with alterations in LPL expression were assessed. Overexpression of a myocyte-anchored form of LPL in heart muscle led to increased uptake of LDL and greater heart cholesterol levels. Loss of LDL receptors did not alter LDL uptake into heart or skeletal muscle. To induce LDL receptors, mice were treated with simvastatin. Statin treatment increased LDL receptor expression and LDL uptake by liver and skeletal muscle but not heart muscle. Plasma creatinine phosphokinase as well as muscle mitochondria, cholesterol, and lipid droplet levels were increased in statin-treated mice overexpressing LPL in skeletal muscle. Thus, pathways affecting cholesterol balance in heart and skeletal muscle differ.


Asunto(s)
Colesterol/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lipoproteína Lipasa/metabolismo , Músculo Esquelético/efectos de los fármacos , Miocardio/metabolismo , Animales , Northern Blotting , Colesterol/farmacocinética , LDL-Colesterol/metabolismo , LDL-Colesterol/farmacocinética , Creatina Quinasa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteína Lipasa/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Miocardio/ultraestructura , Receptores de Lipoproteína/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simvastatina/farmacología
19.
Transplantation ; 78(5): 668-74, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371666

RESUMEN

BACKGROUND: Nitric oxide synthase (NOS)-2 is expressed during acute cardiac allograft rejection in association with death of heart muscle cells. The nuclear enzyme poly(adenosine diphosphate [ADP]-ribose) synthase (PARS) is activated by agonists such as NO and peroxynitrite, which cause single-strand DNA breaks; PARS, in turn can promote both necrosis and apoptosis. To investigate the hypothesis that NO produced by NOS-2 in cardiomyocytes activates PARS and contributes to heart muscle cell death by apoptosis, experiments were performed using a heterotopic rat abdominal heart transplant model and cytokine-stimulated heart muscle cells in tissue culture. METHODS: Cardiac allografts were treated after transplantation with either the PARS inhibitor 5-aminoisoquinolinone at 3 mg/kg subcutaneously daily or with vehicle. Isolated purified adult rat cardiomyocytes incubated with cytokines to induce NOS-2 were treated in vitro with another PARS inhibitor, 3-aminobenzamide (3AB). RESULTS: PARS inhibition increased cardiac-allograft survival from 6 +/- 2 to 10 +/- 3 days (n=6, n=6, P<0.05). The inflammatory infiltrate, NOS-2-positive macrophages, myocyte apoptosis, and myocyte content of nitrotyrosine and poly(ADP-ribose) were significantly decreased in PARS inhibited allografts at day 5 posttransplantation. Similarly, apoptosis and PARS activity were diminished in cytokine-stimulated adult rat cardiomyocytes when either 3AB or L-NMMA were applied. CONCLUSIONS: The data indicate that PARS activation occurs during acute cardiac-allograft rejection and contributes significantly to the inflammatory response and to the death of cardiac muscle cells by apoptosis. They suggest that PARS inhibition combined with immunosuppression might enhance salvage of heart-muscle cells during acute cardiac-allograft rejection.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Isoquinolinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Enfermedad Aguda , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Rechazo de Injerto/patología , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón/patología , Ratas , Ratas Endogámicas Lew , Ratas Endogámicas WF , Trasplante Homólogo
20.
Circulation ; 109(13): 1668-73, 2004 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-15051645

RESUMEN

BACKGROUND: Drugs that selectively block nitric oxide synthase (NOS) 2 enzyme activity by inhibiting dimerization of NOS2 monomers have recently been developed. METHODS AND RESULTS: To investigate whether selective inhibition of NOS2 is cardioprotective, rats were pretreated for 2 days with BBS2, an inhibitor of NOS2 dimerization, at 15 mg/kg SC. Isolated buffer-perfused hearts from treated (n=9) and control (n=7) hearts were subjected to 20 minutes of ischemia followed by 60 minutes of reperfusion. NOS2 protein was upregulated in all hearts at the end of ischemia and of reperfusion; NOS2 enzyme activity was 60% lower in hearts from the treated animals. In the treated hearts, the increase in end-diastolic pressure was significantly attenuated at the end of ischemia, and the return of developed pressure at reperfusion was greater (P<0.05). Creatine kinase release at reperfusion was lower in treated hearts than in controls (P=0.02). At the end of ischemia and of reperfusion, myocardial ATP levels were significantly higher in the treated hearts than in controls (P<0.05). In the treated hearts under ischemic conditions, lactate content was higher and the lactate/pyruvate ratio was lower than in controls (P<0.05); GAPDH activity was higher; and G-3-P and aldose reductase activity were lower. At reperfusion, in the treated hearts, there was less histological damage and less apoptosis of cardiac muscle cells. CONCLUSIONS: Pretreatment with BBS2, a selective inhibitor of NOS2, improves contractile performance, preserves myocardial ATP, and reduces damage and death of cardiac myocytes during ischemia and reperfusion of isolated buffer-perfused rat hearts.


Asunto(s)
Cardiotónicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Imidazoles/uso terapéutico , Isquemia Miocárdica/enzimología , Daño por Reperfusión Miocárdica/prevención & control , Óxido Nítrico Sintasa/antagonistas & inhibidores , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Cardiotónicos/farmacología , Creatina Quinasa/metabolismo , Forma MM de la Creatina-Quinasa , Dimerización , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Isoenzimas/metabolismo , Masculino , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico Sintasa de Tipo II , Piperazinas/farmacología , Premedicación , Pirimidinas/farmacología , Ratas , Ratas Endogámicas WF , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Función Ventricular Izquierda/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...