Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Mol Cell Biochem ; 432(1-2): 7-24, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28303408

ABSTRACT

Diabetic cardiomyopathy is preceded by mitochondrial alterations, and progresses to heart failure. We studied whether treatment with methylene blue (MB), a compound that was reported to serve as an alternate electron carrier within the mitochondrial electron transport chain (ETC), improves mitochondrial metabolism and cardiac function in type 1 diabetes. MB was administered at 10 mg/kg/day to control and diabetic rats. Both echocardiography and hemodynamic studies were performed to assess cardiac function. Mitochondrial studies comprised the measurement of oxidative phosphorylation and specific activities of fatty acid oxidation enzymes. Proteomic studies were employed to compare the level of lysine acetylation on cardiac mitochondrial proteins between the experimental groups. We found that MB facilitates NADH oxidation, increases NAD+, and the activity of deacetylase Sirtuin 3, and reduces protein lysine acetylation in diabetic cardiac mitochondria. We identified that lysine acetylation on 83 sites in 34 proteins is lower in the MB-treated diabetic group compared to the same sites in the untreated diabetic group. These changes occur across critical mitochondrial metabolic pathways including fatty acid transport and oxidation, amino acid metabolism, tricarboxylic acid cycle, ETC, transport, and regulatory proteins. While the MB treatment has no effect on the activities of acyl-CoA dehydrogenases, it decreases 3-hydroxyacyl-CoA dehydrogenase activity and long-chain fatty acid oxidation, and improves cardiac function. Providing an alternative route for mitochondrial electron transport is a novel therapeutic approach to decrease lysine acetylation, alleviate cardiac metabolic inflexibility, and improve cardiac function in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Lysine/metabolism , Methylene Blue/pharmacology , Mitochondria, Heart/metabolism , Acetylation/drug effects , Animals , Male , Rats , Rats, Inbred Lew
2.
J Am Soc Nephrol ; 27(9): 2616-21, 2016 09.
Article in English | MEDLINE | ID: mdl-26839367

ABSTRACT

Regulation of blood pH-critical for virtually every facet of life-requires that the renal proximal tubule (PT) adjust its rate of H(+) secretion (nearly the same as the rate of HCO3 (-) reabsorption, JHCO3 ) in response to changes in blood [CO2] and [HCO3 (-)]. Yet CO2/HCO3 (-) sensing mechanisms remain poorly characterized. Because receptor tyrosine kinase inhibitors render JHCO3 in the PT insensitive to changes in CO2 concentration, we hypothesized that the structural features of receptor protein tyrosine phosphatase-γ (RPTPγ) that are consistent with binding of extracellular CO2 or HCO3 (-) facilitate monitoring of blood CO2/HCO3 (-) concentrations. We now report that PTs express RPTPγ on blood-facing membranes. Moreover, RPTPγ deletion in mice eliminated the CO2 and HCO3 (-) sensitivities of JHCO3 as well as the normal defense of blood pH during whole-body acidosis. Thus, RPTPγ appears to be a novel extracellular CO2/HCO3 (-) sensor critical for pH homeostasis.


Subject(s)
Bicarbonates/metabolism , Carbon Dioxide/metabolism , Extracellular Fluid/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/physiology , Animals , Kidney Tubules, Proximal/metabolism , Mice
3.
Cardiovasc Res ; 107(4): 453-65, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26101264

ABSTRACT

AIMS: Cardiomyopathy is a major complication of diabetes. Our study was aimed to identify the sites of mitochondrial dysfunction and delineate its consequences on mitochondrial metabolism in a model of type 1 diabetes. METHODS AND RESULTS: Diabetes was induced by streptozotocin injection to male Lewis rats. We found a decrease in mitochondrial biogenesis pathway and electron transport chain complex assembly that targets Complex I. Oxidation of Complex II and long-chain fatty acid substrates support the electron leak and superoxide production. Mitochondrial defects do not limit fatty acid oxidation as the heart's preferred energy source indicating that the diabetic heart has a significant reserve in Complex I- and II-supported ATP production. Both mitochondrial fatty acid oxidation and Complex I defect are responsible for increased protein lysine acetylation despite an unchanged amount of the NAD(+)-dependent mitochondrial deacetylase sirt3. We quantitatively analysed mitochondrial lysine acetylation post-translational modifications and identified that the extent of lysine acetylation on 54 sites in 22 mitochondrial proteins is higher in diabetes compared with the same sites in the control. The increased lysine acetylation of the mitochondrial trifunctional protein subunit α may be responsible for the increased fatty acid oxidation in the diabetic heart. CONCLUSION: We identified the specific defective sites in the electron transport chain responsible for the decreased mitochondrial oxidative phosphorylation in the diabetic heart. Mitochondrial protein lysine acetylation is the common consequence of both increased fatty acid oxidation and mitochondrial Complex I defect, and may be responsible for the metabolic inflexibility of the diabetic heart.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Fatty Acids/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Acetylation , Animals , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Electron Transport Complex I/metabolism , Heart/physiopathology , Lipid Metabolism/physiology , Lysine/metabolism , Male , Oxidation-Reduction , Rats, Inbred Lew
4.
Cell Transplant ; 24(6): 971-83, 2015.
Article in English | MEDLINE | ID: mdl-24332083

ABSTRACT

Cellular therapy for myocardial repair has been one of the most intensely investigated interventional strategies for acute myocardial infarction. Although the therapeutic potential of stem cells has been demonstrated in various studies, the underlying mechanisms for such improvements are poorly understood. In the present study, we investigated the long-term effects of stem cell therapy on both myocardial fiber organization and regional contractile function using a rat model of postinfarct remodeling. Human nonhematopoietic umbilical cord blood stem cells (nh-UCBSCs) were administered via tail vein to rats 2 days after infarct surgery. Animals were maintained without immunosuppressive therapy. In vivo and ex vivo MR imaging was performed on infarct hearts 10 months after cell transplantation. Compared to the age-matched rats exposed to the identical surgery, both global and regional cardiac functions of the nh-UCBSC-treated hearts, such as ejection fraction, ventricular strain, and torsion, were significantly improved. More importantly, the treated hearts exhibited preserved fiber orientation and water diffusivities that were similar to those in sham-operated control hearts. These data provide the first evidence that nh-UCBSC treatment may prevent/delay untoward structural remodeling in postinfarct hearts, which supports the improved LV function observed in vivo in the absence of immunosuppression, suggesting a beneficial paracrine effect occurred with the cellular therapy.


Subject(s)
Cord Blood Stem Cell Transplantation , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/pathology , Ventricular Remodeling , Animals , Body Weight , Diastole , Fluorescent Antibody Technique , Heart Function Tests , Hemodynamics , Humans , Magnetic Resonance Imaging, Cine , Male , Myocardial Infarction/pathology , Proto-Oncogene Proteins c-kit/metabolism , Rats, Inbred Lew , Stem Cells/cytology
5.
Physiol Rep ; 2(5)2014 May 01.
Article in English | MEDLINE | ID: mdl-24844640

ABSTRACT

We have previously shown that prolonged high-saturated fat feeding (SAT) for 8 weeks after myocardial infarction (MI) improves ventricular function and prevents the metabolic remodeling commonly observed in heart failure. The current study was designed to delineate the interplay between markers of energy metabolism and indices of cardiac remodeling with 2 and 4 weeks of post-MI SAT in male Wistar rats. By 2 weeks, less remodeling was noted in MI-SAT evidenced by diminished chamber dilation and greater ejection fraction assessed by echocardiography and hemodynamic measures. In addition, gene expression of energy metabolism targets involved in FA uptake, oxidation, and glucose oxidation regulation was increased in MI-SAT with respect to MI alone, although no change in PDH phosphorylation was observed. The regulatory kinase, phosphoinositide 3 kinase (Pi3k), was strongly induced by 2 weeks in the MI-SAT group, although AKT protein content (a primary downstream target of PI3K that affects metabolism) was decreased by both MI and SAT alone, indicating early involvement of cellular signaling pathways in lipid-mediated cardioprotection. Our results demonstrate that cardioprotection occurs acutely with SAT following MI, with improvement in indices of both cardiac function and fatty acid oxidation, suggesting a mechanistic role for energy metabolism in the beneficial effects of high dietary fat following cardiac injury.

6.
Cardiovasc Res ; 99(1): 74-82, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23585471

ABSTRACT

AIMS: The transcription factor hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) regulates myocardial vascularization and growth during cardiogenesis. Our aim was to determine whether HEXIM1 also has a beneficial role in modulating vascularization, myocardial growth, and function within the adult heart. METHODS AND RESULTS: To achieve our objective, we created and investigated a mouse line wherein HEXIM1 was re-expressed in adult cardiomyocytes to levels found in the foetal heart. Our findings support a beneficial role for HEXIM1 through increased vascularization, myocardial growth, and increased ejection fraction within the adult heart. HEXIM1 re-expression induces angiogenesis, that is, essential for physiological hypertrophy and maintenance of cardiac function. The ability of HEXIM1 to co-ordinate processes associated with physiological hypertrophy may be attributed to HEXIM1 regulation of other transcription factors (HIF-1-α, c-Myc, GATA4, and PPAR-α) that, in turn, control many genes involved in myocardial vascularization, growth, and metabolism. Moreover, the mechanism for HEXIM1-induced physiological hypertrophy appears to be distinct from that involving the PI3K/AKT pathway. CONCLUSION: HEXIM1 re-expression results in the induction of angiogenesis that allows for the co-ordination of tissue growth and angiogenesis during physiological hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , Transcription Factors/metabolism , Animals , Cardiomegaly/diagnosis , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cells, Cultured , Echocardiography , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Genotype , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , PPAR alpha/metabolism , Phenotype , Physical Endurance , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins , Stroke Volume , Transcription Factors/genetics , Transfection
7.
J Mol Cell Cardiol ; 55: 147-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23032157

ABSTRACT

Prolonged high fat feeding is associated with myocardial contractile dysfunction in rodents. However, epidemiological data do not necessarily support the concept that fat-enriched diets adversely affect cardiac function in humans. When fed in an ad libitum manner, laboratory rodents consume chow throughout the day. In contrast, humans typically consume food only during the awake phase. Discrepancies between rodent and human feeding behaviors led us to hypothesize that the time of day at which dietary lipids are consumed significantly influences myocardial adaptation. In order to better mimic feeding behavior in humans, mice were fed (either a control or high fat diet) only during the 12-hour dark phase (i.e., no food was provided during the light phase). We report that compared to dark phase restricted control diet fed mice, mice fed a high fat diet during the dark phase exhibit: 1) essentially normal body weight gain and energy balance; 2) increased fatty acid oxidation at whole body, as well as skeletal and cardiac muscle (in the presence of insulin and/or at high workloads) levels; 3) induction of fatty acid responsive genes, including genes promoting triglyceride turnover in the heart; 4) no evidence of cardiac hypertrophy; and 5) persistence/improvement of myocardial contractile function, as assessed ex vivo. These data are consistent with the hypothesis that ingestion of dietary fat only during the more active/awake period allows adequate metabolic adaptation, thereby preserving myocardial contractile function. This article is part of a Special Issue entitled "Focus on cardiac metabolism".


Subject(s)
Adaptation, Physiological , Diet, High-Fat/adverse effects , Heart/physiopathology , Myocardium/metabolism , Animals , Eating , Energy Metabolism , Fatty Acids/metabolism , In Vitro Techniques , Male , Mice , Muscle, Skeletal/metabolism , Myocardial Contraction , Oxidation-Reduction , Transcription, Genetic
8.
Circ Heart Fail ; 5(5): 635-44, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855556

ABSTRACT

BACKGROUND: Decreased expression of cardiac myosin binding protein C (cMyBPC) in the heart has been implicated as a consequence of mutations in cMyBPC that lead to abnormal contractile function at the myofilament level, thereby contributing to the development of hypertrophic cardiomyopathy in humans. It has not been established whether increasing the levels of cMyBPC in the intact heart can improve myofilament and in vivo contractile function and attenuate maladaptive remodeling processes because of reduced levels of cMyBPC. METHODS AND RESULTS: We performed in vivo gene transfer of cMyBPC by direct injection into the myocardium of cMyBPC-deficient (cMyBPC(-/-)) mice, and mechanical experiments were conducted on skinned myocardium isolated from cMyBPC(-/-) hearts 21 days and 20 weeks after gene transfer. Cross-bridge kinetics in skinned myocardium isolated from cMyBPC(-/-) hearts after cMyBPC gene transfer were significantly slower compared with untreated cMyBPC(-/-) myocardium and were comparable to wild-type myocardium and cMyBPC(-/-) myocardium that was reconstituted with recombinant cMyBPC in vitro. cMyBPC content in cMyBPC(-/-) skinned myocardium after in vivo cMyBPC gene transfer or in vitro cMyBPC reconstitution was similar to wild-type levels. In vivo echocardiography studies of cMyBPC(-/-) hearts after cMyBPC gene transfer revealed improved systolic and diastolic contractile function and reductions in left ventricular wall thickness. CONCLUSIONS: This proof-of-concept study demonstrates that gene therapy designed to increase expression of cMyBPC in the cMyBPC-deficient myocardium can improve myofilament and in vivo contractile function, suggesting that cMyBPC gene therapy may be a viable approach for treatment of cardiomyopathies because of mutations in cMyBPC.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/therapy , Carrier Proteins/metabolism , Gene Transfer Techniques , Genetic Therapy , Myocardial Contraction , Myocardium/metabolism , Animals , Cardiomyopathy, Hypertrophic, Familial/diagnostic imaging , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Carrier Proteins/genetics , Disease Models, Animal , Feasibility Studies , Gene Expression Regulation , Kinetics , Male , Mice , Mice, 129 Strain , Mice, Knockout , Myocardial Contraction/genetics , Phosphorylation , Recombinant Proteins/metabolism , Recovery of Function , Ultrasonography , Ventricular Function, Left
9.
J Mol Cell Cardiol ; 53(1): 125-33, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22542451

ABSTRACT

The normal heart relies primarily on the oxidation of fatty acids (FA) for ATP production, whereas during heart failure (HF) glucose utilization increases, implying pathological changes to cardiac energy metabolism. Despite the noted lipotoxic effects of elevating FA, our work has demonstrated a cardioprotective effect of a high fat diet (SAT) when fed after myocardial infarction (MI), as compared to normal chow (NC) fed cohorts. This data has suggested a mechanistic link to energy metabolism. The goal of this study was to determine the impact of SAT on the metabolic phenotype of the heart after MI. Male Wistar rats underwent coronary ligation surgery (MI) and were evaluated after 8 weeks of SAT. Induction of MI was verified by echocardiography. LV function assessed by in vivo hemodynamic measurements revealed improvements in the MI-SAT group as compared to MI-NC. Perfused working hearts revealed a decrease in cardiac work in MI-NC that was improved in MI-SAT. Glucose oxidation was increased and FA oxidation decreased in MI-NC compared to shams suggesting an alteration in the metabolic profile that was ameliorated by SAT. (31)P NMR analysis of Langendorff perfused hearts revealed no differences in PCr:ATP indicating no overt energy deficit in MI groups. Phospho-PDH and PDK(4) were increased in MI-SAT, consistent with a shift towards fatty acid oxidation (FAO). Overall, these results support the hypothesis that SAT post-infarction promotes a normal metabolic phenotype that may serve a cardioprotective role in the injured heart.


Subject(s)
Diet, High-Fat , Metabolome , Myocardial Infarction/metabolism , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Echocardiography , Energy Metabolism , Hemodynamics , Male , Myocardial Infarction/physiopathology , Myocardium/metabolism , Phenotype , Phosphocreatine/metabolism , Phosphorylation , Protein Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats
10.
Am J Physiol Heart Circ Physiol ; 299(6): H1917-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20852054

ABSTRACT

Previous studies have reported that high fat feeding in mild to moderate heart failure (HF) results in the preservation of contractile function. Recent evidence has suggested that preventing the switch from fatty acid to glucose metabolism in HF may ameliorate dysfunction, and insulin resistance is one potential mechanism for regulating substrate utilization. This study was designed to determine whether peripheral and myocardial insulin resistance exists with HF and/or a high-fat diet and whether myocardial insulin signaling was altered accordingly. Rats underwent coronary artery ligation (HF) or sham surgery and were randomized to normal chow (NC; 14% kcal from fat) or a high-fat diet (SAT; 60% kcal from fat) for 8 wk. HF + SAT animals showed preserved systolic (+dP/dt and stroke work) and diastolic (-dP/dt and time constant of relaxation) function compared with HF + NC animals. Glucose tolerance tests revealed peripheral insulin resistance in sham + SAT, HF + NC, and HF + SAT animals compared with sham + NC animals. PET imaging confirmed myocardial insulin resistance only in HF + SAT animals, with an uptake ratio of 2.3 ± 0.3 versus 4.6 ± 0.7, 4.3 ± 0.4, and 4.2 ± 0.6 in sham + NC, sham + SAT, and HF + NC animals, respectively; the myocardial glucose utilization rate was similarly decreased in HF + SAT animals only. Western blot analysis of insulin signaling protein expression was indicative of cardiac insulin resistance in HF + SAT animals. Specifically, alterations in Akt and glycogen synthase kinase-3ß protein expression in HF + SAT animals compared with HF + NC animals may be involved in mediating myocardial insulin resistance. In conclusion, HF animals fed a high-saturated fat exhibited preserved myocardial contractile function, peripheral and myocardial insulin resistance, decreased myocardial glucose utilization rates, and alterations in cardiac insulin signaling. These results suggest that myocardial insulin resistance may serve a cardioprotective function with high fat feeding in mild to moderate HF.


Subject(s)
Dietary Fats/metabolism , Energy Metabolism , Heart Failure/physiopathology , Insulin Resistance , Insulin/metabolism , Myocardial Contraction , Myocardium/metabolism , Ventricular Function, Left , Animals , Blood Glucose/metabolism , Blotting, Western , Dietary Fats/administration & dosage , Dietary Fats/blood , Disease Models, Animal , Echocardiography, Doppler , Glucose Tolerance Test , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heart Failure/diagnosis , Heart Failure/metabolism , Male , Phosphorylation , Positron-Emission Tomography , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Time Factors , Ventricular Pressure
11.
Am J Physiol Heart Circ Physiol ; 299(2): H410-21, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20511406

ABSTRACT

Impaired myocardial contractile function is a hallmark of heart failure (HF), which may present under resting conditions and/or during physiological stress. Previous studies have reported that high fat feeding in mild to moderate HF/left ventricular (LV) dysfunction is associated with improved contractile function at baseline. The goal of this study was to determine whether myocardial function is compromised in response to physiological stress and to evaluate the global gene expression profile of rats fed high dietary fat after infarction. Male Wistar rats underwent ligation or sham surgery and were fed normal chow (NC; 10% kcal fat; Sham + NC and HF + NC groups) or high-fat chow (SAT; 60% kcal saturated fat; Sham + SAT and HF + SAT groups) for 8 wk. Myocardial contractile function was assessed using a Millar pressure-volume conductance catheter at baseline and during inferior vena caval occlusions and dobutamine stress. Steady-state indexes of systolic function, LV +dP/dt(max), stroke work, and maximal power were increased in the HF + SAT group versus the HF + NC group and reduced in the HF + NC group versus the Sham + NC group. Preload recruitable measures of contractility were decreased in HF + NC group but not in the HF + SAT group. beta-Adrenergic responsiveness [change in LV +dP/dt(max) and change in cardiac output with dobutamine (0-10 microg x kg(-1) x min(-1))] was reduced in HF, but high fat feeding did not further impact the contractile reserve in HF. The contractile reserve was reduced by the high-fat diet in the Sham + SAT group. Microarray gene expression analysis revealed that the majority of significantly altered pathways identified contained multiple gene targets correspond to cell signaling pathways and energy metabolism. These findings suggest that high saturated fat improves myocardial function at rest and during physiological stress in infarcted hearts but may negatively impact the contractile reserve under nonpathological conditions. Furthermore, high fat feeding-induced alterations in gene expression related to energy metabolism and specific signaling pathways revealed promising targets through which high saturated fat potentially mediates cardioprotection in mild to moderate HF/LV dysfunction.


Subject(s)
Dietary Fats/administration & dosage , Fatty Acids/administration & dosage , Heart Failure/physiopathology , Myocardial Contraction , Myocardial Infarction/physiopathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Adrenergic beta-Agonists/pharmacology , Animals , Cardiac Output , Dietary Fats/blood , Disease Models, Animal , Dobutamine/pharmacology , Energy Metabolism , Fatty Acids/blood , Gene Expression Profiling/methods , Gene Expression Regulation , Heart Failure/diagnostic imaging , Heart Failure/genetics , Heart Failure/metabolism , Male , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar , Recovery of Function , Signal Transduction , Stress, Physiological , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/genetics , Ventricular Function, Left/drug effects , Ventricular Function, Left/genetics , Ventricular Pressure
12.
J Biol Chem ; 285(5): 2918-29, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19940111

ABSTRACT

Maintenance of circadian alignment between an organism and its environment is essential to ensure metabolic homeostasis. Synchrony is achieved by cell autonomous circadian clocks. Despite a growing appreciation of the integral relation between clocks and metabolism, little is known regarding the direct influence of a peripheral clock on cellular responses to fatty acids. To address this important issue, we utilized a genetic model of disrupted clock function specifically in cardiomyocytes in vivo (termed cardiomyocyte clock mutant (CCM)). CCM mice exhibited altered myocardial response to chronic high fat feeding at the levels of the transcriptome and lipidome as well as metabolic fluxes, providing evidence that the cardiomyocyte clock regulates myocardial triglyceride metabolism. Time-of-day-dependent oscillations in myocardial triglyceride levels, net triglyceride synthesis, and lipolysis were markedly attenuated in CCM hearts. Analysis of key proteins influencing triglyceride turnover suggest that the cardiomyocyte clock inactivates hormone-sensitive lipase during the active/awake phase both at transcriptional and post-translational (via AMP-activated protein kinase) levels. Consistent with increased net triglyceride synthesis during the end of the active/awake phase, high fat feeding at this time resulted in marked cardiac steatosis. These data provide evidence for direct regulation of triglyceride turnover by a peripheral clock and reveal a potential mechanistic explanation for accelerated metabolic pathologies after prevalent circadian misalignment in Western society.


Subject(s)
Gene Expression Regulation , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Triglycerides/metabolism , Animals , Circadian Rhythm , Fatty Acids , Gene Expression Profiling , Heart , Male , Mice , Perfusion , Protein Processing, Post-Translational , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
13.
J Mol Cell Cardiol ; 46(6): 883-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19265702

ABSTRACT

Previous studies have reported that elevated myocardial lipids in a model of mild-to-moderate heart failure increased mitochondrial function, but did not alter left ventricular function. Whether more prolonged exposure to high dietary lipids would promote a lipotoxic phenotype in mitochondrial and myocardial contractile function has not been determined. We tested the hypothesis that prolonged exposure to high dietary lipids, following coronary artery ligation, would preserve myocardial and mitochondrial function in heart failure. Rats underwent ligation or sham surgery and were fed normal (10% kcal fat) (SHAM, HF) or high fat diet (60% kcal saturated fat) (SHAM+FAT, HF+FAT) for sixteen weeks. Although high dietary fat was accompanied by myocardial tissue triglyceride accumulation (SHAM 1.47+/-0.14; SHAM+FAT 2.32+/-0.14; HF 1.34+/-0.14; HF+FAT 2.21+/-0.20 micromol/gww), fractional shortening was increased 16% in SHAM+FAT and 28% in HF+FAT compared to SHAM and HF, respectively. Despite increased medium-chain acyl-CoA dehydrogenase (MCAD) activity in interfibrillar mitochondria (IFM) of both SHAM+FAT and HF+FAT, dietary lipids also were associated with decreased state 3 respiration using palmitoylcarnitine (SHAM 369+/-14; SHAM+FAT 307+/-23; HF 354+/-13; HF+FAT 366+/-18 nAO min(-1) mg(-1)) in SHAM+FAT compared to SHAM and HF+FAT. State 3 respiration in IFM also was decreased in SHAM+FAT relative to SHAM using succinate and DHQ. In conclusion, high dietary lipids promoted myocardial lipid accumulation, but were not accompanied by alterations in myocardial contractile function typically associated with lipotoxicity. In normal animals, high dietary fat decreased mitochondrial respiration, but also increased MCAD activity. These studies support the concept that high fat feeding can modify multiple cellular pathways that differentially affect mitochondrial function under normal and pathological conditions.


Subject(s)
Dietary Fats/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain , Adiponectin/blood , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Butyryl-CoA Dehydrogenase , Dietary Fats/administration & dosage , Echocardiography , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Nonesterified/blood , Hemodynamics/drug effects , Insulin/blood , Leptin/blood , Male , Mitochondrial Proteins/metabolism , Organ Size/drug effects , Rats , Rats, Wistar
14.
Cardiovasc Res ; 80(1): 30-9, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18710878

ABSTRACT

AIMS: Mitochondrial dysfunction is a major factor in heart failure (HF). A pronounced variability of mitochondrial electron transport chain (ETC) defects is reported to occur in severe acquired cardiomyopathies without a consistent trend for depressed activity or expression. The aim of this study was to define the defect in the integrative function of cardiac mitochondria in coronary microembolization-induced HF. METHODS AND RESULTS: Studies were performed in the canine coronary microembolization-induced HF model of moderate severity. Oxidative phosphorylation was assessed as the integrative function of mitochondria, using a comprehensive variety of substrates in order to investigate mitochondrial membrane transport, dehydrogenase activity and electron-transport coupled to ATP synthesis. The supramolecular organization of the mitochondrial ETC also was investigated by native gel electrophoresis. We found a dramatic decrease in ADP-stimulated respiration that was not relieved by an uncoupler. Moreover, the ADP/O ratio was normal, indicating no defect in the phosphorylation apparatus. The data point to a defect in oxidative phosphorylation within the ETC. However, the individual activities of ETC complexes were normal. The amount of the supercomplex consisting of complex I/complex III dimer/complex IV, the major form of respirasome considered essential for oxidative phosphorylation, was decreased. CONCLUSIONS: We propose that the mitochondrial defect lies in the supermolecular assembly rather than in the individual components of the ETC.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/biosynthesis , Animals , Cell Respiration , Dogs , Electron Transport , Hemodynamics , Membrane Transport Proteins/metabolism , Mitochondrial Myopathies/metabolism , Oxidoreductases/metabolism
15.
Cardiovasc Res ; 79(2): 331-40, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18339649

ABSTRACT

AIMS: Heart failure is associated with decreased myocardial fatty acid oxidation capacity and has been likened to energy starvation. Increased fatty acid availability results in an induction of genes promoting fatty acid oxidation. The aim of the present study was to investigate possible mechanisms by which high fat feeding improved mitochondrial and contractile function in heart failure. METHODS AND RESULTS: Male Wistar rats underwent coronary artery ligation (HF) or sham surgery and were immediately fed either a normal (14% kcal fat) (SHAM, HF) or high-fat diet (60% kcal saturated fat) (SHAM+FAT, HF+FAT) for 8 weeks. Mitochondrial respiration and gene expression and enzyme activities of fatty acid-regulated mitochondrial genes and proteins were assessed. Subsarcolemmal (SSM) and interfibrillar mitochondria were isolated from the left ventricle. State 3 respiration using lipid substrates octanoylcarnitine and palmitoylcarnitine increased in the SSM of HF+FAT compared with SHAM+FAT and HF, respectively (242 +/- 21, 246 +/- 21 vs. 183 +/- 8, 181 +/- 6 and 193 +/- 17, 185 +/- 16 nAO min(-1) mg(-1)). Despite decreased medium-chain acyl-CoA dehydrogenase (MCAD) mRNA in HF and HF+FAT, MCAD protein was not altered, and MCAD activity increased in HF+FAT (HF, 65.1 +/- 2.7 vs. HF+FAT, 81.5 +/- 5.4 nmoles min(-1) mg(-1)). Activities of short- and long-chain acyl-CoA dehydrogenase also were elevated and correlated to increased state 3 respiration. This was associated with an improvement in myocardial contractility as assessed by left ventricular +dP/dt max. CONCLUSION: Administration of a high-fat diet increased state 3 respiration and acyl-CoA dehydrogenase activities, but did not normalize mRNA or protein levels of acyl-CoA dehydrogenases in coronary artery ligation-induced heart failure rats.


Subject(s)
Acyl-CoA Dehydrogenase/metabolism , Heart Failure/enzymology , Heart Failure/physiopathology , Mitochondria, Heart/metabolism , Myocardial Contraction/physiology , Adiponectin/metabolism , Animals , Blood Glucose/metabolism , Carnitine/analogs & derivatives , Carnitine/metabolism , Dietary Fats/pharmacology , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Fatty Acids, Nonesterified/metabolism , Insulin/metabolism , Leptin/metabolism , Male , Myocardial Contraction/drug effects , Rats , Rats, Wistar , Triglycerides/metabolism
16.
Am J Physiol Heart Circ Physiol ; 294(2): H1036-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18156197

ABSTRACT

Virtually every mammalian cell, including cardiomyocytes, possesses an intrinsic circadian clock. The role of this transcriptionally based molecular mechanism in cardiovascular biology is poorly understood. We hypothesized that the circadian clock within the cardiomyocyte influences diurnal variations in myocardial biology. We, therefore, generated a cardiomyocyte-specific circadian clock mutant (CCM) mouse to test this hypothesis. At 12 wk of age, CCM mice exhibit normal myocardial contractile function in vivo, as assessed by echocardiography. Radiotelemetry studies reveal attenuation of heart rate diurnal variations and bradycardia in CCM mice (in the absence of conduction system abnormalities). Reduced heart rate persisted in CCM hearts perfused ex vivo in the working mode, highlighting the intrinsic nature of this phenotype. Wild-type, but not CCM, hearts exhibited a marked diurnal variation in responsiveness to an elevation in workload (80 mmHg plus 1 microM epinephrine) ex vivo, with a greater increase in cardiac power and efficiency during the dark (active) phase vs. the light (inactive) phase. Moreover, myocardial oxygen consumption and fatty acid oxidation rates were increased, whereas cardiac efficiency was decreased, in CCM hearts. These observations were associated with no alterations in mitochondrial content or structure and modest mitochondrial dysfunction in CCM hearts. Gene expression microarray analysis identified 548 and 176 genes in atria and ventricles, respectively, whose normal diurnal expression patterns were altered in CCM mice. These studies suggest that the cardiomyocyte circadian clock influences myocardial contractile function, metabolism, and gene expression.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Gene Expression/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Myocytes, Cardiac/physiology , Animals , DNA/biosynthesis , DNA/genetics , Echocardiography , Electrocardiography , Heart Rate/physiology , In Vitro Techniques , Mice , Mitochondria, Heart/physiology , Muscle Proteins/metabolism , Myocardial Contraction/genetics , Oligonucleotide Array Sequence Analysis , Perfusion , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Telemetry
17.
Am J Physiol Heart Circ Physiol ; 293(4): H2385-93, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17616739

ABSTRACT

Multiple extracardiac stimuli, such as workload and circulating nutrients (e.g., fatty acids), known to influence myocardial metabolism and contractile function exhibit marked circadian rhythms. The aim of the present study was to investigate whether the rat heart exhibits circadian rhythms in its responsiveness to changes in workload and/or fatty acid (oleate) availability. Thus, hearts were isolated from male Wistar rats (housed during a 12:12-h light-dark cycle: lights on at 9 AM) at 9 AM, 3 PM, 9 PM, and 3 AM and perfused in the working mode ex vivo with 5 mM glucose plus either 0.4 or 0.8 mM oleate. Following 20-min perfusion at normal workload (i.e., 100 cm H(2)O afterload), hearts were challenged with increased workload (140 cm H(2)O afterload plus 1 microM epinephrine). In the presence of 0.4 mM oleate, myocardial metabolism exhibited a marked circadian rhythm, with decreased rates of glucose oxidation, increased rates of lactate release, decreased glycogenolysis capacity, and increased channeling of oleate into nonoxidative pathways during the light phase. Rat hearts also exhibited a modest circadian rhythm in responsiveness to the workload challenge when perfused in the presence of 0.4 mM oleate, with increased myocardial oxygen consumption at the dark-to-light phase transition. However, rat hearts perfused in the presence of 0.8 mM oleate exhibited a markedly blunted contractile function response to the workload challenge during the light phase. In conclusion, these studies expose marked circadian rhythmicities in myocardial oxidative and nonoxidative metabolism as well as responsiveness of the rat heart to changes in workload and fatty acid availability.


Subject(s)
Circadian Rhythm , Heart/physiology , Myocardial Contraction , Myocardium/metabolism , Oleic Acid/metabolism , Animals , Glucose/metabolism , Glycogenolysis , Heart/drug effects , Lactic Acid/metabolism , Male , Myocardial Contraction/drug effects , Myocardium/enzymology , Oleic Acid/pharmacology , Oxidation-Reduction , Oxygen Consumption , Perfusion , Rats , Rats, Wistar , Research Design , Time Factors
18.
Am J Physiol Heart Circ Physiol ; 293(3): H1609-16, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17545473

ABSTRACT

Clinical studies have shown a greater incidence of myocardial infarction in diabetic patients, and following an infarction, diabetes is associated with an increased risk for the development of left ventricular (LV) dysfunction and heart failure. The goal of this study was to determine if the progression of heart failure following myocardial infarction in type 2 diabetic (T2D) rats is accelerated compared with nondiabetic rats. Male nondiabetic Wistar-Kyoto (WKY) and T2D Goto-Kakizaki (GK) rats underwent coronary artery ligation or sham surgery to induce heart failure. Postligation (8 and 20 wk), two-dimensional echocardiography and LV pressure measurements were made. Heart failure progression, as assessed by enhanced LV remodeling and contractile dysfunction, was accelerated 8 wk postligation in the T2D animals. LV remodeling was evident from increased end-diastolic and end-systolic diameters and areas in the GK compared with the WKY infarcted group. Furthermore, enhanced LV contractile dysfunction was evident from a greater deterioration in fractional shortening and enhanced myocardial performance index (an index of global LV dysfunction) in the GK infarcted group. This accelerated progression was accompanied by greater increases in atrial natriuretic factor and skeletal alpha-actin (gene markers of heart failure and hypertrophy) mRNA levels in GK infarcted hearts. Despite similar decreases in metabolic gene expression (i.e., peroxisome proliferator-activated receptor-alpha-regulated genes associated with fatty acid oxidation) between infarcted WKY and GK rat hearts, myocardial triglyceride levels were elevated in the GK hearts only. These results, demonstrating enhanced remodeling and LV dysfunction 8 wk postligation provide evidence of an accelerated progression of heart failure in T2D rats.


Subject(s)
Cardiac Output, Low/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Myocardial Infarction/physiopathology , Actins/genetics , Actins/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blood Glucose/metabolism , Cardiac Output, Low/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Disease Progression , Fatty Acids, Nonesterified/blood , Heart Rate/physiology , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Rats, Inbred WKY , Ventricular Dysfunction, Left , Ventricular Remodeling
19.
Am J Hypertens ; 20(4): 403-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17386347

ABSTRACT

BACKGROUND: Chronic hypertension leads to cardiac hypertrophy, heart failure, and premature death. Little is known about the impact of dietary macronutrient composition on hypertension-induced cardiac hypertrophy and mortality. We investigated the effects of consuming either a high complex carbohydrate diet, a high simple sugar diet, or a high fat diet on cardiac hypertrophy and mortality in hypertensive Dahl salt-sensitive (DSS) rats. METHODS: Rats were assigned to four diets: complex carbohydrate (CC; 70% starch, 10% fat, 20% protein by energy), high fat (FAT; 20% carbohydrates, 60% fat, 20% protein), high fructose (FRU; 70% fructose, 10% fat, 20% protein), and "western" (WES; 35% fructose, 45% fat, 20% protein). Hypertension was initiated by adding 6% NaCl (+S) to the chow of half the animals within each diet (n = 10 to 13/group). Tail cuff blood pressure measurements were assessed after 5 and 11 weeks of treatment, and echocardiography were assessed after 12 weeks of treatment. RESULTS: All rats fed a high salt diet had similar levels of hypertension (CC+S 220 +/-2 mm Hg, FAT+S 221 +/- 3 mm Hg, FRU+S 221 +/- 1 mm Hg, WES+S 226 +/- 3 mm Hg). Echocardiography results show that the addition of salt to FRU resulted in increased regional wall thickness that was not observed in other dietary groups. All rats fed a low salt diet (CC, FAT, FRU, WES) and the FAT+S group survived 90 days. On the other hand, there was 90-day mortality in the WES+S group (18% mortality) and the CC+S group (30% mortality). In addition, FRU+S rats started dying after 45 days of salt feeding, and only 15% survived the full 90 days. CONCLUSIONS: These results demonstrate that a high fructose diet consumed during hypertension increases mortality and left ventricular (LV) wall thickness compared to either a high fat, high starch, or a "western" diet.


Subject(s)
Dietary Carbohydrates/adverse effects , Dietary Fats/adverse effects , Dietary Sucrose/adverse effects , Fructose/adverse effects , Hypertension/mortality , Animals , Blood Glucose/metabolism , Blood Pressure/physiology , Body Mass Index , Cardiomegaly/etiology , Cardiomegaly/pathology , Electrocardiography , Hypertension/complications , Hypertrophy, Left Ventricular/etiology , Male , Myocardium/pathology , Rats , Rats, Inbred Dahl , Triglycerides/blood , Ventricular Dysfunction, Left/physiopathology
20.
Clin Exp Pharmacol Physiol ; 34(1-2): 77-80, 2007.
Article in English | MEDLINE | ID: mdl-17201739

ABSTRACT

1. Alterations in myocardial energy metabolism accompany pressure overload-induced hypertrophy. We previously described a novel model of catecholamine-induced hypertrophy in which A/J mice exhibit more robust cardiac hypertrophy than B6 mice. Accordingly, we assessed the influence of mouse strain on the activities of key myocardial metabolic enzymes and whether there are strain-related metabolic adaptations to short-term, high-dose isoproterenol (ISO) administration. 2. Thirty-nine male mice (19 A/J mice, 20 B6 mice), aged 12-15 weeks, were randomly assigned to receive either ISO (100 mg/kg, s.c.) or vehicle (sterile water) daily for 5 days. On Day 6, all hearts were excised, weighed, freeze clamped and assayed for pyruvate dehydrogenase (PDH), medium chain acyl-CoA dehydrogenase, carnitine palmitoyl transferase I and citrate synthase activities. Plasma fatty acids (FA) were also measured. 3. The ISO-treated A/J mice demonstrated greater percentage increases in gravimetric heart weight/bodyweight ratio than ISO-treated B6 mice (24 vs 3%, respectively; P < 0.001). All enzyme activities were significantly greater in vehicle-treated B6 mice than in A/J mice, illustrating a greater capacity for aerobic metabolism in B6 mice. Administration of ISO reduced PDHa (active form) activity in B6 mice by 47% (P < 0.001), with no significant change seen in A/J mice. Free FA levels were not significantly different between groups; thus, the differences in PDHa were not due to changes in FA. 4. The basal activity of myocardial metabolic enzymes is greater in B6 mice than in A/J mice and ISO alters myocardial PDH activity in a mouse strain-dependent manner. Compared with A/J mice, B6 mice demonstrate less ISO-induced cardiac hypertrophy, but greater activity of key enzymes regulating FA and carbohydrate oxidation, which may protect against the development of hypertrophy. The metabolic adaptations associated with ISO-induced hypertrophy differ from those reported with pressure overload hypertrophy.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Myocardium/enzymology , Acyl-CoA Dehydrogenase/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Citrate (si)-Synthase/metabolism , Hypertrophy, Left Ventricular/chemically induced , Isoproterenol , Ketone Oxidoreductases/metabolism , Male , Mice , Mice, Inbred Strains , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...