Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Mol Cell Cardiol ; 136: 95-101, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31536744

RESUMEN

TRIM72 is a membrane repair protein that protects against ischemia reperfusion (I/R) injury. We previously identified Cys144 (C144) on TRIM72 as a site of S-nitrosylation. To study the importance of C144, we generated a knock-in mouse with C144 mutated to a serine (TRIM72 C144S). We subjected ex vivo perfused mouse hearts to 20 min of ischemia followed by 90 min of reperfusion and observed less injury in TRIM72 C144S compared to WT hearts. Infarct size was smaller (54 vs 27% infarct size) and cardiac functional recovery (37 vs 62% RPP) was higher for the TRIM72 C144S mouse hearts. We also demonstrated that TRIM72 C144S hearts were protected against I/R injury using an in vivo LAD occlusion model. As TRIM72 has been reported to be released from muscle we tested whether C144 is involved in TRIM72 release. After I/R there was significantly less TRIM72 in the perfusate normalized to total released protein from the TRIM72 C144S compared to WT hearts, suggesting that C144 of TRIM72 regulates myocardial TRIM72 release during I/R injury. In addition to TRIM72's protective role in I/R injury, TRIM72 has also been implicated in cardiac hypertrophy and insulin resistance, and secreted TRIM72 has recently been shown to impair insulin sensitivity. However, insulin sensitivity (measured by glucose and insulin tolerance) of TRIM72 C144S mice was not impaired. Further, whole body metabolism, as measured using metabolic cages, was not different in WT vs TRIM72 C144S mice and we did not observe enhanced cardiac hypertrophy in the TRIM72 C144S mice. In agreement, protein levels of the TRIM72 ubiquitination targets insulin receptor ß, IRS1, and focal adhesion kinase were similar between WT and TRIM72 C144S hearts. Overall, these data indicate that mutation of TRIM72 C144 is protective during I/R and reduces myocardial TRIM72 release without impairing insulin sensitivity or enhancing the development of hypertrophy.


Asunto(s)
Cisteína/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Angiotensina II/farmacología , Animales , Cardiomegalia/genética , Enfermedad de la Arteria Coronaria , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Resistencia a la Insulina/genética , Ratones Endogámicos C57BL , Ratones Mutantes , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocardio/patología
2.
Biochem J ; 475(5): 959-976, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29438065

RESUMEN

The role of carnitine acetyltransferase (CrAT) in regulating cardiac energy metabolism is poorly understood. CrAT modulates mitochondrial acetyl-CoA/CoA (coenzyme A) ratios, thus regulating pyruvate dehydrogenase activity and glucose oxidation. Here, we propose that cardiac CrAT also provides cytosolic acetyl-CoA for the production of malonyl-CoA, a potent inhibitor of fatty acid oxidation. We show that in the murine cardiomyocyte cytosol, reverse CrAT activity (RCrAT, producing acetyl-CoA) is higher compared with the liver, which primarily uses ATP-citrate lyase to produce cytosolic acetyl-CoA for lipogenesis. The heart displayed a lower RCrAT Km for CoA compared with the liver. Furthermore, cytosolic RCrAT accounted for 4.6 ± 0.7% of total activity in heart tissue and 12.7 ± 0.2% in H9C2 cells, while highly purified heart cytosolic fractions showed significant CrAT protein levels. To investigate the relationship between CrAT and acetyl-CoA carboxylase (ACC), the cytosolic enzyme catalyzing malonyl-CoA production from acetyl-CoA, we studied ACC2-knockout mouse hearts which showed decreased CrAT protein levels and activity, associated with increased palmitate oxidation and acetyl-CoA/CoA ratio compared with controls. Conversely, feeding mice a high-fat diet for 10 weeks increased cardiac CrAT protein levels and activity, associated with a reduced acetyl-CoA/CoA ratio and glucose oxidation. These data support the presence of a cytosolic CrAT with a low Km for CoA, favoring the formation of cytosolic acetyl-CoA, providing an additional source to the classical ATP-citrate lyase pathway, and that there is an inverse relation between CrAT and the ratio of acetyl-CoA/CoA as evident in conditions affecting the regulation of cardiac energy metabolism.


Asunto(s)
Acetilcoenzima A/metabolismo , Carnitina O-Acetiltransferasa/fisiología , Citosol/metabolismo , Metabolismo Energético/genética , Miocardio/metabolismo , Animales , Carnitina O-Acetiltransferasa/genética , Carnitina O-Acetiltransferasa/metabolismo , Células Cultivadas , Dieta Alta en Grasa , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Oxidación-Reducción
3.
Am J Physiol Endocrinol Metab ; 315(5): E1046-E1052, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30106622

RESUMEN

Recent studies have proposed that elevated branched-chain amino acids (BCAAs) may induce insulin resistance (IR) in muscle secondary to increased BCAA oxidation inhibiting glucose oxidation (GO) and fatty acid oxidation (FAO). However, BCAA oxidation rates have not been assessed in muscle IR, and cardiac FAO rates are actually already elevated in obesity-associated IR. We therefore directly examined cardiac BCAA oxidation in mice fed a high-fat diet (HFD) to induce insulin resistance to better understand the role of cardiac BCAA oxidation in cardiac IR. BCAA oxidation, GO, FAO, and glycolysis were measured in isolated working hearts from mice fed either a low-fat diet (LFD) or HFD for 10 wk. Insulin stimulation of cardiac GO and inhibition of FAO were blunted in HFD mice, resulting in a marked increase in FAO contribution to ATP production compared with LFD mice hearts (71.2% vs. 37.1%, respectively). Surprisingly, cardiac BCAA oxidation rate was reduced in HFD compared with LFD mice (33.5 ± 3.4 vs. 56.7 ± 7.1 nmol·min-1·g dry wt-1, respectively, P < 0.05, n = 9/group). In addition, BCAA oxidation contributed ~1% of the ATP production of the heart, and, as a result, alterations in BCAA oxidation could not significantly impact either GO or FAO rates. However, the decrease in BCAA oxidation was accompanied by an increase in BCAA concentration and impaired insulin signaling. These results suggest that cardiac IR is not due to an increase in BCAA oxidation and subsequent inhibition of GO and FAO. Rather, we propose that an inhibition of BCAA oxidation rate contributes to IR by leading to increased BCAA concentration, which negatively impacts insulin signaling.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Miocardio/metabolismo , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Glucólisis/fisiología , Corazón/efectos de los fármacos , Insulina/farmacología , Ratones , Oxidación-Reducción , Transducción de Señal/efectos de los fármacos
4.
Mol Med ; 24(1): 3, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30134787

RESUMEN

BACKGROUND: Alterations in cardiac energy metabolism contribute to the development and severity of heart failure (HF). In severe HF, overall mitochondrial oxidative metabolism is significantly decreased resulting in a reduced energy reserve. However, despite the high prevalence of HF with preserved ejection fraction (HFpEF) in our society, it is not clear what changes in cardiac energy metabolism occur in HFpEF, and whether alterations in energy metabolism contribute to the development of contractile dysfunction. METHODS: We directly assessed overall energy metabolism during the development of HFpEF in Dahl salt-sensitive rats fed a high salt diet (HSD) for 3, 6 and 9 weeks. RESULTS: Over the course of 9 weeks, the HSD caused a progressive decrease in diastolic function (assessed by echocardiography assessment of E'/A'). This was accompanied by a progressive increase in cardiac glycolysis rates (assessed in isolated working hearts obtained at 3, 6, and 9 weeks of HSD). In contrast, the subsequent oxidation of pyruvate from glycolysis (glucose oxidation) was not altered, resulting in an uncoupling of glucose metabolism and a significant increase in proton production. Increased glucose transporter (GLUT)1 expression accompanied this elevation in glycolysis. Decreases in cardiac fatty acid oxidation and overall adenosine triphosphate (ATP) production rates were not observed in early HF, but both significantly decreased as HF progressed to HF with reduced EF (i.e. 9 weeks of HSD). CONCLUSIONS: Overall, we show that increased glycolysis is the earliest energy metabolic change that occurs during HFpEF development. The resultant increased proton production from uncoupling of glycolysis and glucose oxidation may contribute to the development of HFpEF.


Asunto(s)
Glucosa/metabolismo , Glucólisis , Insuficiencia Cardíaca/metabolismo , Animales , Cardiomegalia/fisiopatología , Corazón/fisiología , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocardio/metabolismo , Oxidación-Reducción , Ratas Endogámicas Dahl , Cloruro de Sodio Dietético/administración & dosificación
5.
Exp Physiol ; 100(6): 730-41, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25786668

RESUMEN

NEW FINDINGS: What is the central question of this study? The aim was to determine whether the accumulation of ceramide contributes to skeletal muscle insulin resistance in the JCR obese rat. What is the main finding and its importance? Our main new finding is that ceramides accumulate only in slow-twitch skeletal muscle in the JCR obese rat and that reducing ceramide content in this muscle type by inhibition of serine palmitoyl transferase-1 halts the progression of insulin resistance in this rat model predisposed to early development of type 2 diabetes. Our findings highlight the importance of assessing insulin signalling/sensitivity and lipid intermediate accumulation in different muscle fibre types. It has been postulated that insulin resistance results from the accumulation of cytosolic lipid metabolites (i.e. diacylglycerol/ceramide) that impede insulin signalling and impair glucose homeostasis. De novo ceramide synthesis is catalysed by serine palmitoyl transferase-1. Our aim was to determine whether de novo ceramide synthesis plays a role during development of insulin resistance in the JCR:LA-cp obese rat. Ten-week-old JCR:LA-cp obese rats were supplemented with either vehicle or the serine palmitoyl transferase-1 inhibitor l-cycloserine (360 mg l(-1) ) in their drinking water for a 2 week period, and glycaemia was assessed by meal tolerance testing. Treatment of JCR:LA-cp obese rats with l-cycloserine improved their plasma glucose and insulin levels during a meal tolerance test. Examination of muscle lipid metabolites and protein phosphorylation patterns revealed differential signatures in slow-twitch (soleus) versus fast-twitch muscle (gastrocnemius), in that ceramide levels were increased in soleus but not gastrocnemius muscles of JCR:LA-cp obese rats. Likewise, improved glycaemia in l-cycloserine-treated JCR:LA-cp obese rats was associated with enhanced Akt and pyruvate dehydrogenase signalling in soleus but not gastrocnemius muscles, probably as a result of l-cycloserine reducing elevated ceramides in this muscle type. Potential mechanisms of ceramide-mediated insulin resistance involve activation of atypical protein kinase Cζ/λ and protein phosphatase 2A; however, neither of these was altered in muscles of JCR:LA-cp obese rats. Our results suggest a key role for ceramide in the development of insulin resistance in the JCR:LA-cp obese rat, while supporting serine palmitoyl transferase-1 inhibition as a novel target for treatment of obesity-associated insulin resistance.


Asunto(s)
Ceramidas/metabolismo , Resistencia a la Insulina , Fibras Musculares de Contracción Lenta/metabolismo , Obesidad/metabolismo , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Cicloserina/farmacología , Modelos Animales de Enfermedad , Metabolismo Energético , Inhibidores Enzimáticos/farmacología , Insulina/sangre , Isoenzimas/metabolismo , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Obesidad/sangre , Obesidad/fisiopatología , Fosforilación , Proteína Quinasa C/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Ratas , Serina C-Palmitoiltransferasa/antagonistas & inhibidores , Serina C-Palmitoiltransferasa/metabolismo , Transducción de Señal , Factores de Tiempo
6.
Biochim Biophys Acta ; 1833(4): 857-65, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22960640

RESUMEN

Heart failure is a major cause of morbidity and mortality in the world. Cardiac energy metabolism, specifically fatty acid and glucose metabolism, is altered in heart failure and has been implicated as a contributing factor in the impaired heart function observed in heart failure patients. There is emerging evidence demonstrating that correcting these changes in energy metabolism by modulating mitochondrial oxidative metabolism may be an effective treatment for heart failure. Promising strategies include the downregulation of fatty acid oxidation and an increased coupling of glycolysis to glucose oxidation. Carnitine palmitoyl transferase I (CPT1), fatty acid ß-oxidation enzymes, and pyruvate dehydrogenase kinase (PDK) are examples of metabolic targets for the treatment of heart failure. While targeting mitochondrial oxidative metabolism is a promising strategy to treat heart failure, further studies are needed to confirm the potential beneficial effect of modulating these metabolic targets as an approach to treating heart failure. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Corazón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Antagonistas Adrenérgicos beta/farmacología , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/antagonistas & inhibidores , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Corazón/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Mitocondrias/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación Oxidativa/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo
7.
J Pharmacol Exp Ther ; 349(3): 487-96, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700885

RESUMEN

There is a growing need to understand the underlying mechanisms involved in the progression of cardiovascular disease during obesity and diabetes. Although inhibition of fatty acid oxidation has been proposed as a novel approach to treat ischemic heart disease and heart failure, reduced muscle fatty acid oxidation rates may contribute to the development of obesity-associated insulin resistance. Our aim was to determine whether treatment with the antianginal agent trimetazidine, which inhibits fatty acid oxidation in the heart secondary to inhibition of 3-ketoacyl-CoA thiolase (3-KAT), may have off-target effects on glycemic control in obesity. We fed C57BL/6NCrl mice a high-fat diet (HFD) for 10 weeks before a 22-day treatment with the 3-KAT inhibitor trimetazidine (15 mg/kg per day). Insulin resistance was assessed via glucose/insulin tolerance testing, and lipid metabolite content was assessed in gastrocnemius muscle. Trimetazidine-treatment led to a mild shift in substrate preference toward carbohydrates as an oxidative fuel source in obese mice, evidenced by an increase in the respiratory exchange ratio. This shift in metabolism was accompanied by an accumulation of long-chain acyl-CoA and a trend to an increase in triacylglycerol content in gastrocnemius muscle, but did not exacerbate HFD-induced insulin resistance compared with control-treated mice. It is noteworthy that trimetazidine treatment reduced palmitate oxidation rates in the isolated working mouse heart and neonatal cardiomyocytes but not C2C12 skeletal myotubes. Our findings demonstrate that trimetazidine therapy does not adversely affect HFD-induced insulin resistance, suggesting that treatment with trimetazidine would not worsen glycemic control in obese patients with angina.


Asunto(s)
Acetil-CoA C-Aciltransferasa/antagonistas & inhibidores , Angina de Pecho/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Trimetazidina/efectos adversos , Vasodilatadores/efectos adversos , Angina de Pecho/tratamiento farmacológico , Angina de Pecho/enzimología , Angina de Pecho/etiología , Animales , Células Cultivadas , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Obesidad/complicaciones , Obesidad/enzimología , Oxidación-Reducción , Ratas , Trimetazidina/administración & dosificación , Trimetazidina/uso terapéutico , Vasodilatadores/administración & dosificación , Vasodilatadores/uso terapéutico
8.
IUBMB Life ; 66(3): 139-146, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24591219

RESUMEN

Alterations in cardiac energy metabolism are an important contributor to the high incidence and severity of heart disease in the world. These alterations can include an impairment of the production of ATP necessary to meet the high energy demands of the heart, as well as adverse switches in energy substrate preference by the heart. With regard to this latter point, evidence suggests that a decrease in cardiac efficiency, caused by a rise in cardiac fatty acid oxidation and/or an increase in the uncoupling of glycolysis from glucose oxidation, impairs cardiac function and is a contributing factor to cardiac disease. In support of this concept, therapeutic strategies that modulate these metabolic pathways and increase cardiac efficiency produce beneficial results in the setting of heart disease. One such strategy is to increase cardiac malonyl CoA levels, an important inhibitor of mitochondrial fatty acid uptake. This includes malonyl CoA decarboxylase (MCD) inhibition that results in increased cardiac malonyl CoA levels, decreased cardiac fatty acid oxidation rates, and improved cardiac efficiency. Preclinical studies have shown that MCD inhibition can improve cardiac function in various forms of heart disease. Here, we focus on the importance of malonyl CoA in the regulation of cardiac energy metabolism and function in the normal and diseased heart and discuss the evidence that suggests that inhibition of fatty acid oxidation especially via regulation of malonyl CoA, through MCD inhibition, is a promising strategy to treat cardiac disease. © 2014 IUBMB Life, 66(3):139-146, 2014.

9.
Am J Physiol Endocrinol Metab ; 303(12): E1459-68, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23074239

RESUMEN

We previously showed that genetic inactivation of malonyl-CoA decarboxylase (MCD), which regulates fatty acid oxidation, protects mice against high-fat diet-induced insulin resistance. Development of insulin resistance has been associated with activation of the inflammatory response. Therefore, we hypothesized that the protective effect of MCD inhibition might be caused by a favorable effect on the inflammatory response. We examined if pharmacological inhibition of MCD protects neonatal cardiomyocytes and peritoneal macrophages against inflammatory-induced metabolic perturbations. Cardiomyocytes and macrophages were treated with LPS to induce an inflammatory response, in the presence or absence of an MCD inhibitor (CBM-301106, 10 µM). Inhibition of MCD attenuated the LPS-induced inflammatory response in cardiomyocytes and macrophages. MCD inhibition also prevented LPS impairment of insulin-stimulated glucose uptake in cardiomyocytes and increased phosphorylation of Akt. Additionally, inhibition of MCD strongly diminished LPS-induced activation of palmitate oxidation. We also found that treatment with an MCD inhibitor prevented LPS-induced collapse of total cellular antioxidant capacity. Interestingly, treatment with LPS or an MCD inhibitor did not alter intracellular triacylglycerol content. Furthermore, inhibition of MCD prevented LPS-induced increases in the level of ceramide in cardiomyocytes and macrophages while also ameliorating LPS-initiated decreases in PPAR binding. This suggests that the anti-inflammatory effect of MCD inhibition is mediated via accumulation of long-chain acyl-CoA, which in turn stimulates PPAR binding. Our results also demonstrate that pharmacological inhibition of MCD is a novel and promising approach to treat insulin resistance and its associated metabolic complications.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Carboxiliasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Resistencia a la Insulina , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Transporte Biológico/efectos de los fármacos , Carboxiliasas/metabolismo , Cardiotónicos/farmacología , Células Cultivadas , Ceramidas/metabolismo , Glucosa/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Compuestos de Fenilurea/farmacología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
10.
PLoS One ; 17(3): e0265007, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35259201

RESUMEN

While fatty acid metabolism is altered under physiological conditions, alterations can also be maladaptive in diseases such as diabetes and heart failure. Peroxisome Proliferator Activated Receptor α (PPARα) is a transcription factor that regulates fat metabolism but its role in regulating lipid storage in the heart is unclear. The aim of this study is to improve our understanding of how cardiac PPARα regulates cardiac health and lipid accumulation. To study the role of cardiac PPARα, tamoxifen inducible cardiac-specific PPARα knockout mouse (cPPAR-/-) were treated for 5 days with tamoxifen and then studied after 1-2 months. Under baseline conditions, cPPAR-/- mice appear healthy with normal body weight and mortality is not altered. Importantly, cardiac hypertrophy or reduced cardiac function was also not observed at baseline. Mice were fasted to elevate circulating fatty acids and induce cardiac lipid accumulation. After fasting, cPPAR-/- mice had dramatically lower cardiac triglyceride levels than control mice. Interestingly, cPPAR-/- hearts also had reduced Plin2, a key protein involved in lipid accumulation and lipid droplet regulation, which may contribute to the reduction in cardiac lipid accumulation. Overall, this suggests that a decline in cardiac PPARα may blunt cardiac lipid accumulation by decreasing Plin2 and that independent of differences in systemic metabolism a decline in cardiac PPARα does not seem to drive pathological changes in the heart.


Asunto(s)
Ayuno , PPAR alfa , Animales , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Ratones , Ratones Noqueados , PPAR alfa/genética , PPAR alfa/metabolismo , Perilipina-2/metabolismo , Tamoxifeno/metabolismo
12.
J Am Heart Assoc ; 8(24): e013465, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31818212

RESUMEN

Background Heart failure is one of the leading causes of death in Western countries, and there is a need for new therapeutic approaches. Relaxin-2 is a peptide hormone that mediates pleiotropic cardiovascular effects, including antifibrotic, angiogenic, vasodilatory, antiapoptotic, and anti-inflammatory effects in vitro and in vivo. Methods and Results We developed RELAX10, a fusion protein composed of human relaxin-2 hormone and the Fc of a human antibody, to test the hypothesis that extended exposure of the relaxin-2 peptide could reduce cardiac hypertrophy and fibrosis. RELAX10 demonstrated the same specificity and similar in vitro activity as the relaxin-2 peptide. The terminal half-life of RELAX10 was 7 days in mouse and 3.75 days in rat after subcutaneous administration. We evaluated whether treatment with RELAX10 could prevent and reverse isoproterenol-induced cardiac hypertrophy and fibrosis in mice. Isoproterenol administration in mice resulted in increased cardiac hypertrophy and fibrosis compared with vehicle. Coadministration with RELAX10 significantly attenuated the cardiac hypertrophy and fibrosis compared with untreated animals. Isoproterenol administration significantly increased transforming growth factor ß1 (TGF-ß1)-induced fibrotic signaling, which was attenuated by RELAX10. We found that RELAX10 also significantly increased protein kinase B/endothelial NO synthase signaling and protein S-nitrosylation. In the reversal study, RELAX10-treated animals showed significantly reduced cardiac hypertrophy and collagen levels. Conclusions These findings support a potential role for RELAX10 in the treatment of heart failure.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Miocardio/patología , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/prevención & control , Fibrosis/inducido químicamente , Fibrosis/tratamiento farmacológico , Fibrosis/prevención & control , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL
13.
JACC Basic Transl Sci ; 4(3): 385-400, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31312761

RESUMEN

Alterations in cardiac energy metabolism after a myocardial infarction contribute to the severity of heart failure (HF). Although fatty acid oxidation can be impaired in HF, it is unclear if stimulating fatty acid oxidation is a desirable approach to treat HF. Both immediate and chronic malonyl coenzyme A decarboxylase inhibition, which decreases fatty acid oxidation, improved cardiac function through enhancing cardiac efficiency in a post-myocardial infarction rat that underwent permanent left anterior descending coronary artery ligation. The beneficial effects of MCD inhibition were attributed to a decrease in proton production due to an improved coupling between glycolysis and glucose oxidation.

14.
Cardiovasc Res ; 113(4): 370-377, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28158412

RESUMEN

In contrast to ischemic cardiomyopathies which are more common in men, women are over-represented in diabetic cardiomyopathies. Diabetes is a risk factor for cardiovascular disease; however, there is a sexual dimorphism in this risk factor: heart disease is five times more common in diabetic women but only two-times more common in diabetic men. Heart failure with preserved ejection fraction, which is associated with metabolic syndrome, is also more prevalent in women. This review will examine potential mechanisms for the sex differences in metabolic cardiomyopathies. Sex differences in metabolism, calcium handling, nitric oxide, and structural proteins will be evaluated. Nitric oxide synthase and PPARα exhibit sex differences and have also been proposed to mediate the development of hypertrophy and heart failure. We focused on a role for these signalling pathways in regulating sex differences in metabolic cardiomyopathies.

15.
J Am Heart Assoc ; 6(8)2017 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-28862954

RESUMEN

BACKGROUND: Heart failure preceded by hypertrophy is a leading cause of death, and sex differences in hypertrophy are well known, although the basis for these sex differences is poorly understood. METHODS AND RESULTS: This study used a systems biology approach to investigate mechanisms underlying sex differences in cardiac hypertrophy. Male and female mice were treated for 2 and 3 weeks with angiotensin II to induce hypertrophy. Sex differences in cardiac hypertrophy were apparent after 3 weeks of treatment. RNA sequencing was performed on hearts, and sex differences in mRNA expression at baseline and following hypertrophy were observed, as well as within-sex differences between baseline and hypertrophy. Sex differences in mRNA were substantial at baseline and reduced somewhat with hypertrophy, as the mRNA differences induced by hypertrophy tended to overwhelm the sex differences. We performed an integrative analysis to identify mRNA networks that were differentially regulated in the 2 sexes by hypertrophy and obtained a network centered on PPARα (peroxisome proliferator-activated receptor α). Mouse experiments further showed that acute inhibition of PPARα blocked sex differences in the development of hypertrophy. CONCLUSIONS: The data in this study suggest that PPARα is involved in the sex-dimorphic regulation of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Miocardio/metabolismo , PPAR alfa/metabolismo , Biología de Sistemas/métodos , Angiotensina II , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Miocardio/patología , Oxazoles/farmacología , PPAR alfa/antagonistas & inhibidores , PPAR alfa/genética , Mapas de Interacción de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Caracteres Sexuales , Factores Sexuales , Transducción de Señal , Factores de Tiempo , Tirosina/análogos & derivados , Tirosina/farmacología
16.
Diabetes ; 65(7): 1883-91, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207536

RESUMEN

Aging is associated with the development of chronic diseases such as insulin resistance and type 2 diabetes. A reduction in mitochondrial fat oxidation is postulated to be a key factor contributing to the progression of these diseases. Our aim was to investigate the contribution of impaired mitochondrial fat oxidation toward age-related disease. Mice deficient for malonyl CoA decarboxylase (MCD(-/-)), a mouse model of reduced fat oxidation, were allowed to age while life span and a number of physiological parameters (glucose tolerance, insulin tolerance, indirect calorimetry) were assessed. Decreased fat oxidation in MCD(-/-) mice resulted in the accumulation of lipid intermediates in peripheral tissues, but this was not associated with a worsening of age-associated insulin resistance and, conversely, improved longevity. This improvement was associated with reduced oxidative stress and reduced acetylation of the antioxidant enzyme superoxide dismutase 2 in muscle but not the liver of MCD(-/-) mice. These findings were recapitulated in aged mice treated with an MCD inhibitor (CBM-3001106), and these mice also demonstrated improvements in glucose and insulin tolerance. Therefore, our results demonstrate that in addition to decreasing fat oxidation, MCD inhibition also has novel effects on protein acetylation. These combined effects protect against age-related metabolic dysfunction, demonstrating that MCD inhibitors may have utility in the battle against chronic disease in the elderly.


Asunto(s)
Envejecimiento/metabolismo , Carboxiliasas/genética , Intolerancia a la Glucosa/genética , Resistencia a la Insulina/genética , Estrés Oxidativo/genética , Consumo de Oxígeno/genética , Envejecimiento/genética , Animales , Calorimetría Indirecta , Carboxiliasas/antagonistas & inhibidores , Diglicéridos/metabolismo , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Longevidad/genética , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Estrés Oxidativo/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Triglicéridos/metabolismo
17.
PLoS One ; 10(4): e0123318, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25884410

RESUMEN

Enhanced expression and activity of the Na+/H+ exchanger isoform 1 (NHE1) has been implicated in cardiomyocyte hypertrophy in various experimental models. The upregulation of NHE1 was correlated with an increase in osteopontin (OPN) expression in models of cardiac hypertrophy (CH), and the mechanism for this remains to be delineated. To determine whether the expression of active NHE1-induces OPN and contributes to the hypertrophic response in vitro, cardiomyocytes were infected with the active form of the NHE1 adenovirus or transfected with OPN silencing RNA (siRNA-OPN) and characterized for cardiomyocyte hypertrophy. Expression of NHE1 in cardiomyocytes resulted in a significant increase in cardiomyocyte hypertrophy markers: cell surface area, protein content, ANP mRNA and expression of phosphorylated-GATA4. NHE1 activity was also significantly increased in cardiomyocytes expressing active NHE1. Interestingly, transfection of cardiomyocytes with siRNA-OPN significantly abolished the NHE1-induced cardiomyocyte hypertrophy. siRNA-OPN also significantly reduced the activity of NHE1 in cardiomyocytes expressing NHE1 (68.5±0.24%; P<0.05), confirming the role of OPN in the NHE1-induced hypertrophic response. The hypertrophic response facilitated by NHE1-induced OPN occurred independent of the extracellular-signal-regulated kinases and Akt, but required p90-ribosomal S6 kinase (RSK). The ability of OPN to facilitate the NHE1-induced hypertrophic response identifies OPN as a potential therapeutic target to reverse the hypertrophic effect induced by the expression of active NHE1.


Asunto(s)
Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Osteopontina/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Aumento de la Célula , Células Cultivadas , Miocitos Cardíacos/patología , Osteopontina/genética , Fosforilación , ARN Interferente Pequeño , Ratas , Transducción de Señal/fisiología , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética , Transfección
18.
PLoS One ; 10(3): e0120257, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25768019

RESUMEN

Successful stem cell therapy requires the optimal proliferation, engraftment, and differentiation of stem cells into the desired cell lineage of tissues. However, stem cell therapy clinical trials to date have had limited success, suggesting that a better understanding of stem cell biology is needed. This includes a better understanding of stem cell energy metabolism because of the importance of energy metabolism in stem cell proliferation and differentiation. We report here the first direct evidence that human bone marrow mesenchymal stem cell (BMMSC) energy metabolism is highly glycolytic with low rates of mitochondrial oxidative metabolism. The contribution of glycolysis to ATP production is greater than 97% in undifferentiated BMMSCs, while glucose and fatty acid oxidation combined only contribute 3% of ATP production. We also assessed the effect of physiological levels of fatty acids on human BMMSC survival and energy metabolism. We found that the saturated fatty acid palmitate induces BMMSC apoptosis and decreases proliferation, an effect prevented by the unsaturated fatty acid oleate. Interestingly, chronic exposure of human BMMSCs to physiological levels of palmitate (for 24 hr) reduces palmitate oxidation rates. This decrease in palmitate oxidation is prevented by chronic exposure of the BMMSCs to oleate. These results suggest that reducing saturated fatty acid oxidation can decrease human BMMSC proliferation and cause cell death. These results also suggest that saturated fatty acids may be involved in the long-term impairment of BMMSC survival in vivo.


Asunto(s)
Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Células Madre Mesenquimatosas/fisiología , Análisis de Varianza , Western Blotting , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Técnica del Anticuerpo Fluorescente , Glucólisis/fisiología , Humanos , Mitocondrias/fisiología , Oxidación-Reducción/efectos de los fármacos , Palmitatos/farmacología , Sales de Tetrazolio , Tiazoles
19.
Can J Cardiol ; 30(8): 940-4, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25064584

RESUMEN

Obesity is a significant risk factor for the development of cardiovascular disease. Inhibiting fatty acid oxidation has emerged as a novel approach for the treatment of ischemic heart disease. Our aim was to determine whether pharmacologic inhibition of 3-ketoacyl-coenzyme A thiolase (3-KAT), which catalyzes the final step of fatty acid oxidation, could improve obesity-induced cardiomyopathy. A 3-week treatment with the 3-KAT inhibitor trimetazidine prevented obesity-induced reduction in both systolic and diastolic function. Therefore, targeting cardiac fatty acid oxidation may be a novel therapeutic approach to alleviate the growing burden of obesity-related cardiomyopathy.


Asunto(s)
Cardiomiopatías/prevención & control , Obesidad/complicaciones , Trimetazidina/farmacología , Vasodilatadores/farmacología , Acetil-CoA C-Aciltransferasa/antagonistas & inhibidores , Animales , Cardiomiopatías/etiología , Ecocardiografía , Ventrículos Cardíacos/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Palmitatos/metabolismo
20.
Cardiovasc Res ; 103(4): 485-97, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24966184

RESUMEN

AIMS: Lysine acetylation is a novel post-translational pathway that regulates the activities of enzymes involved in both fatty acid and glucose metabolism. We examined whether lysine acetylation controls heart glucose and fatty acid oxidation in high-fat diet (HFD) obese and SIRT3 knockout (KO) mice. METHODS AND RESULTS: C57BL/6 mice were placed on either a HFD (60% fat) or a low-fat diet (LFD; 4% fat) for 16 or 18 weeks. Cardiac fatty acid oxidation rates were significantly increased in HFD vs. LFD mice (845 ± 76 vs. 551 ± 87 nmol/g dry wt min, P < 0.05). Activities of the fatty acid oxidation enzymes, long-chain acyl-CoA dehydrogenase (LCAD), and ß-hydroxyacyl-CoA dehydrogenase (ß-HAD) were increased in hearts from HFD vs. LFD mice, and were associated with LCAD and ß-HAD hyperacetylation. Cardiac protein hyperacetylation in HFD-fed mice was associated with a decrease in SIRT3 expression, while expression of the mitochondrial acetylase, general control of amino acid synthesis 5 (GCN5)-like 1 (GCN5L1), did not change. Interestingly, SIRT3 deletion in mice also led to an increase in cardiac fatty acid oxidation compared with wild-type (WT) mice (422 ± 29 vs. 291 ± 17 nmol/g dry wt min, P < 0.05). Cardiac lysine acetylation was increased in SIRT3 KO mice compared with WT mice, including increased acetylation and activity of LCAD and ß-HAD. Although the HFD and SIRT3 deletion decreased glucose oxidation, pyruvate dehydrogenase acetylation was unaltered. However, the HFD did increase Akt acetylation, while decreasing its phosphorylation and activity. CONCLUSION: We conclude that increased cardiac fatty acid oxidation in response to high-fat feeding is controlled, in part, via the down-regulation of SIRT3 and concomitant increased acetylation of mitochondrial ß-oxidation enzymes.


Asunto(s)
Ácidos Grasos/metabolismo , Insulina/metabolismo , Miocardio/metabolismo , Obesidad/metabolismo , Transducción de Señal , Sirtuina 3/genética , Acetilación , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Animales , Corazón/fisiología , Lisina/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Transducción de Señal/fisiología , Sirtuina 3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA