Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Circ Heart Fail ; 4(4): 474-82, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558447

ABSTRACT

BACKGROUND: Currently, there are no specific therapies available to treat cardiac dysfunction caused by sepsis and other chronic inflammatory conditions. Activation of toll-like receptor 4 (TLR4) by lipopolysaccharide (LPS) is an early event in Gram-negative bacterial sepsis, triggering a robust inflammatory response and changes in metabolism. Peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) α and ß serve as critical physiological regulators of energy metabolic gene expression in heart. METHODS AND RESULTS: Injection of mice with LPS triggered a myocardial fuel switch similar to that of the failing heart: reduced mitochondrial substrate flux and myocyte lipid accumulation. The LPS-induced metabolic changes were associated with diminished ventricular function and suppression of the genes encoding PGC-1α and ß, known transcriptional regulators of mitochondrial function. This cascade of events required TLR4 and nuclear factor-κB activation. Restoration of PGC-1ß expression in cardiac myocytes in culture and in vivo in mice reversed the gene regulatory, metabolic, and functional derangements triggered by LPS. Interestingly, the effects of PGC-1ß overexpression were independent of the upstream inflammatory response, highlighting the potential utility of modulating downstream metabolic derangements in cardiac myocytes as a novel strategy to prevent or treat sepsis-induced heart failure. CONCLUSIONS: LPS triggers cardiac energy metabolic reprogramming through suppression of PGC-1 coactivators in the cardiac myocyte. Reactivation of PGC-1ß expression can reverse the metabolic and functional derangements caused by LPS-TLR4 activation, identifying the PGC-1 axis as a candidate therapeutic target for sepsis-induced heart failure.


Subject(s)
Energy Metabolism/physiology , Heart Failure/metabolism , Myocardium/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Transcription Factors/antagonists & inhibitors , Animals , Cells, Cultured , Disease Models, Animal , Fatty Acids/metabolism , Heart Failure/chemically induced , Heart Failure/physiopathology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Transcription Factors/metabolism
2.
J Biol Chem ; 286(13): 11155-62, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21321124

ABSTRACT

Diabetic cardiac dysfunction is associated with decreased rates of myocardial glucose oxidation (GO) and increased fatty acid oxidation (FAO), a fuel shift that has been shown to sensitize the heart to ischemic insult and ventricular dysfunction. We sought to evaluate the metabolic and functional consequences of chronic suppression of GO in heart as modeled by transgenic mice with cardiac-specific overexpression of pyruvate dehydrogenase kinase 4 (myosin heavy chain (MHC)-PDK4 mice), an inhibitor of pyruvate dehydrogenase. Hearts of MHC-PDK4 mice were shown to exhibit an insulin-resistant substrate utilization profile, characterized by low GO rates and high FAO flux. Surprisingly, MHC-PDK4 mice were not sensitized to cardiac ischemia-reperfusion injury despite a fuel utilization pattern that phenocopied the diabetic heart. In addition, MHC-PDK4 mice were protected against high fat diet-induced myocyte lipid accumulation, likely related to increased capacity for FAO. The high rates of mitochondrial FAO in the MHC-PDK4 heart were related to heightened activity of the AMP-activated protein kinase, reduced levels of malonyl-CoA, and increased capacity for mitochondrial uncoupled respiration. The expression of the known AMP-activated protein kinase target, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a master regulator of mitochondrial function and biogenesis, was also activated in the MHC-PDK4 heart. These results demonstrate that chronic activation of PDK4 triggers transcriptional and post-transcriptional mechanisms that re-program the heart for chronic high rates of FAO without the expected deleterious functional or metabolic consequences.


Subject(s)
Diabetic Cardiomyopathies/enzymology , Glucose/metabolism , Mitochondria, Heart/enzymology , Myocardium/enzymology , Protein Serine-Threonine Kinases/biosynthesis , Animals , Diabetic Cardiomyopathies/genetics , Disease Models, Animal , Glucose/genetics , Heart Rate/genetics , Insulin Resistance/genetics , Mice , Mice, Transgenic , Mitochondria, Heart/genetics , Myocardial Ischemia/enzymology , Myocardial Ischemia/genetics , Myocardium/pathology , Myosin Heavy Chains/genetics , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Ventricular Dysfunction/enzymology , Ventricular Dysfunction/genetics
3.
J Biol Chem ; 285(32): 24447-56, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20529848

ABSTRACT

Heart muscle is metabolically versatile, converting energy stored in fatty acids, glucose, lactate, amino acids, and ketone bodies. Here, we use mouse models in ketotic nutritional states (24 h of fasting and a very low carbohydrate ketogenic diet) to demonstrate that heart muscle engages a metabolic response that limits ketone body utilization. Pathway reconstruction from microarray data sets, gene expression analysis, protein immunoblotting, and immunohistochemical analysis of myocardial tissue from nutritionally modified mouse models reveal that ketotic states promote transcriptional suppression of the key ketolytic enzyme, succinyl-CoA:3-oxoacid CoA transferase (SCOT; encoded by Oxct1), as well as peroxisome proliferator-activated receptor alpha-dependent induction of the key ketogenic enzyme HMGCS2. Consistent with reduction of SCOT, NMR profiling demonstrates that maintenance on a ketogenic diet causes a 25% reduction of myocardial (13)C enrichment of glutamate when (13)C-labeled ketone bodies are delivered in vivo or ex vivo, indicating reduced procession of ketones through oxidative metabolism. Accordingly, unmetabolized substrate concentrations are higher within the hearts of ketogenic diet-fed mice challenged with ketones compared with those of chow-fed controls. Furthermore, reduced ketone body oxidation correlates with failure of ketone bodies to inhibit fatty acid oxidation. These results indicate that ketotic nutrient environments engage mechanisms that curtail ketolytic capacity, controlling the utilization of ketone bodies in ketotic states.


Subject(s)
Myocardium/metabolism , Animals , Carbon Isotopes/chemistry , Coenzyme A-Transferases/metabolism , Immunohistochemistry/methods , Ketone Bodies/chemistry , Ketones/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Myocytes, Cardiac/cytology , Peroxisome Proliferator-Activated Receptors/metabolism , Rats
4.
Circ Res ; 106(9): 1516-23, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20360254

ABSTRACT

RATIONALE: Tribbles (TRB)3 is an intracellular pseudokinase that modulates the activity of several signal transduction cascades. TRB3 has been reported to inhibit the activity of Akt protein kinases. TRB3 gene expression is highly regulated in many cell types, and amino acid starvation, hypoxia, or endoplasmic reticulum (ER) stress promotes TRB3 expression in noncardiac cells. OBJECTIVE: The objective of this work was to examine TRB3 expression and function in cultured cardiac myocytes and in mouse heart. METHODS AND RESULTS: Agents that induced ER stress increased TRB3 expression in cultured cardiac myocytes while blocking insulin-stimulated Akt activation in these cells. Knockdown of TRB3 in cultured cardiac myocytes reversed the effects of ER stress on insulin signaling. Experimental myocardial infarction led to increased TRB3 expression in murine heart tissue in the infarct border zone suggesting that ER stress may play a role in pathological cardiac remodeling. Transgenic mice with cardiac-specific overexpression of TRB3 were generated and they exhibited normal contractile function but altered cardiac signal transduction and metabolism with reduced cardiac glucose oxidation rates. Transgenic TRB3 mice were also sensitized to infarct expansion and cardiac myocyte apoptosis in the infarct border zone after myocardial infarction. CONCLUSIONS: These results demonstrate that TRB3 induction is a significant aspect of the ER stress response in cardiac myocytes and that TRB3 antagonizes cardiac glucose metabolism and cardiac myocyte survival.


Subject(s)
Endoplasmic Reticulum/metabolism , Myocytes, Cardiac/metabolism , TRPM Cation Channels/metabolism , Animals , Cells, Cultured , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Physiological , TRPM Cation Channels/genetics
5.
Circulation ; 121(3): 426-35, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20065164

ABSTRACT

BACKGROUND: Emerging evidence in obesity and diabetes mellitus demonstrates that excessive myocardial fatty acid uptake and oxidation contribute to cardiac dysfunction. Transgenic mice with cardiac-specific overexpression of the fatty acid-activated nuclear receptor peroxisome proliferator-activated receptor-alpha (myosin heavy chain [MHC]-PPARalpha mice) exhibit phenotypic features of the diabetic heart, which are rescued by deletion of CD36, a fatty acid transporter, despite persistent activation of PPARalpha gene targets involved in fatty acid oxidation. METHODS AND RESULTS: To further define the source of fatty acid that leads to cardiomyopathy associated with lipid excess, we crossed MHC-PPARalpha mice with mice deficient for cardiac lipoprotein lipase (hsLpLko). MHC-PPARalpha/hsLpLko mice exhibit improved cardiac function and reduced myocardial triglyceride content compared with MHC-PPARalpha mice. Surprisingly, in contrast to MHC-PPARalpha/CD36ko mice, the activity of the cardiac PPARalpha gene regulatory pathway is normalized in MHC-PPARalpha/hsLpLko mice, suggesting that PPARalpha ligand activity exists in the lipoprotein particle. Indeed, LpL mediated hydrolysis of very-low-density lipoprotein activated PPARalpha in cardiac myocytes in culture. The rescue of cardiac function in both models was associated with improved mitochondrial ultrastructure and reactivation of transcriptional regulators of mitochondrial function. CONCLUSIONS: MHC-PPARalpha mouse hearts acquire excess lipoprotein-derived lipids. LpL deficiency rescues myocyte triglyceride accumulation, mitochondrial gene regulatory derangements, and contractile function in MHC-PPARalpha mice. Finally, LpL serves as a source of activating ligand for PPARalpha in the cardiomyocyte.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Lipoprotein Lipase/genetics , Myocardium/metabolism , PPAR alpha/genetics , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Cholesterol, VLDL/pharmacokinetics , Fatty Acids/pharmacokinetics , Female , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Knockout , Mitochondria/physiology , Myocardium/cytology , Myosin Heavy Chains/genetics , PPAR alpha/metabolism , Phenotype , Triglycerides/pharmacokinetics
6.
Proc Natl Acad Sci U S A ; 106(27): 11276-81, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19549860

ABSTRACT

Studies in mice indicate that the gut microbiota promotes energy harvest and storage from components of the diet when these components are plentiful. Here we examine how the microbiota shapes host metabolic and physiologic adaptations to periods of nutrient deprivation. Germ-free (GF) mice and mice who had received a gut microbiota transplant from conventionally raised donors were compared in the fed and fasted states by using functional genomic, biochemical, and physiologic assays. A 24-h fast produces a marked change in gut microbial ecology. Short-chain fatty acids generated from microbial fermentation of available glycans are maintained at higher levels compared with GF controls. During fasting, a microbiota-dependent, Ppar alpha-regulated increase in hepatic ketogenesis occurs, and myocardial metabolism is directed to ketone body utilization. Analyses of heart rate, hydraulic work, and output, mitochondrial morphology, number, and respiration, plus ketone body, fatty acid, and glucose oxidation in isolated perfused working hearts from GF and colonized animals (combined with in vivo assessments of myocardial physiology) revealed that the fasted GF heart is able to sustain its performance by increasing glucose utilization, but heart weight, measured echocardiographically or as wet mass and normalized to tibial length or lean body weight, is significantly reduced in both fasted and fed mice. This myocardial-mass phenotype is completely reversed in GF mice by consumption of a ketogenic diet. Together, these results illustrate benefits provided by the gut microbiota during periods of nutrient deprivation, and emphasize the importance of further exploring the relationship between gut microbes and cardiovascular health.


Subject(s)
Fasting/physiology , Gastrointestinal Tract/microbiology , Ketone Bodies/metabolism , Metagenome , Myocardium/metabolism , Animals , Food , Genomics , Germ-Free Life , Glucose/metabolism , Hypertrophy , In Vitro Techniques , Liver/metabolism , Mice , Myocardium/pathology , Organ Size , Oxidation-Reduction , Perfusion , Physical Endurance
7.
J Biol Chem ; 283(41): 27410-27417, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18667418

ABSTRACT

Loss of the transcription factor E2F1 elicits a complex metabolic phenotype in mice underscored by reduced adiposity and protection from high fat diet-induced diabetes. Here, we demonstrate that E2F1 directly regulates the gene encoding PDK4 (pyruvate dehydrogenase kinase 4), a key nutrient sensor and modulator of glucose homeostasis that is chronically elevated in obesity and diabetes and acutely induced under the metabolic stress of starvation or fasting. We show that loss of E2F1 in vivo blunts PDK4 expression and improves myocardial glucose oxidation. The absence of E2F1 also corresponds to lower blood glucose levels, improved plasma lipid profile, and increased sensitivity to insulin stimulation. Consistently, enforced E2F1 expression up-regulates PDK4 levels and suppresses glucose oxidation in C(2)C(12) myoblasts. Furthermore, inactivation of Rb, the repressor of E2F-dependent transcription, markedly induces PDK4 and triggers the enrichment of E2F1 occupancy onto the PDK4 promoter as detected by chromatin immunoprecipitation analysis. Two overlapping E2F binding sites were identified on this promoter. Transactivation assays later verified E2F1 responsiveness of this promoter element in C(2)C(12) myoblasts and IMR90 fibroblasts, an effect that was completely abrogated following mutation of the E2F sites. Taken together, our data illustrate how the E2F1 mitogen directly regulates PDK4 levels and influences cellular bioenergetics, namely mitochondrial glucose oxidation. These results are relevant to the pathophysiology of chronic diseases like obesity and diabetes, where PDK4 is dysregulated and could have implications pertinent to the etiology of tumor metabolism, especially in cancers with Rb pathway defects.


Subject(s)
E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glucose/metabolism , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Retinoblastoma Protein/metabolism , Adiposity/physiology , Animals , Diabetes Mellitus/chemically induced , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Dietary Fats/metabolism , E2F1 Transcription Factor/genetics , Fasting/metabolism , Female , Fibroblasts/enzymology , Homeostasis/physiology , Male , Mice , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondria, Heart/genetics , Multiprotein Complexes/genetics , Mutation , Myoblasts/enzymology , Myocardium/enzymology , Neoplasms/enzymology , Neoplasms/genetics , Oxidation-Reduction , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Retinoblastoma Protein/genetics , Starvation/enzymology , Starvation/genetics , Up-Regulation/physiology
8.
Am J Physiol Heart Circ Physiol ; 295(1): H185-96, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18487436

ABSTRACT

High-capacity mitochondrial ATP production is essential for normal function of the adult heart, and evidence is emerging that mitochondrial derangements occur in common myocardial diseases. Previous overexpression studies have shown that the inducible transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha is capable of activating postnatal cardiac myocyte mitochondrial biogenesis. Recently, we generated mice deficient in PGC-1alpha (PGC-1alpha(-/-) mice), which survive with modestly blunted postnatal cardiac growth. To determine if PGC-1alpha is essential for normal cardiac energy metabolic capacity, mitochondrial function experiments were performed on saponin-permeabilized myocardial fibers from PGC-1alpha(-/-) mice. These experiments demonstrated reduced maximal (state 3) palmitoyl-l-carnitine respiration and increased maximal (state 3) pyruvate respiration in PGC-1alpha(-/-) mice compared with PGC-1alpha(+/+) controls. ATP synthesis rates obtained during maximal (state 3) respiration in permeabilized myocardial fibers were reduced for PGC-1alpha(-/-) mice, whereas ATP produced per oxygen consumed (ATP/O), a measure of metabolic efficiency, was decreased by 58% for PGC-1alpha(-/-) fibers. Ex vivo isolated working heart experiments demonstrated that PGC-1alpha(-/-) mice exhibited lower cardiac power, reduced palmitate oxidation, and increased reliance on glucose oxidation, with the latter likely a compensatory response. (13)C NMR revealed that hearts from PGC-1alpha(-/-) mice exhibited a limited capacity to recruit triglyceride as a source for lipid oxidation during beta-adrenergic challenge. Consistent with reduced mitochondrial fatty acid oxidative enzyme gene expression, the total triglyceride content was greater in hearts of PGC-1alpha(-/-) mice relative to PGC-1alpha(+/+) following a fast. Overall, these results demonstrate that PGC-1alpha is essential for the maintenance of maximal, efficient cardiac mitochondrial fatty acid oxidation, ATP synthesis, and myocardial lipid homeostasis.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism , Fatty Acids/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Trans-Activators/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Glucose/metabolism , Homeostasis , In Vitro Techniques , Isoproterenol/pharmacology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Knockout , Mitochondria, Heart/drug effects , Myocardial Contraction , Oxidation-Reduction , Oxidative Phosphorylation , Oxygen Consumption , Palmitoylcarnitine/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pyruvic Acid/metabolism , Trans-Activators/genetics , Transcription Factors , Triglycerides/metabolism
9.
J Clin Invest ; 117(12): 3930-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18037994

ABSTRACT

In the diabetic heart, chronic activation of the PPARalpha pathway drives excessive fatty acid (FA) oxidation, lipid accumulation, reduced glucose utilization, and cardiomyopathy. The related nuclear receptor, PPARbeta/delta, is also highly expressed in the heart, yet its function has not been fully delineated. To address its role in myocardial metabolism, we generated transgenic mice with cardiac-specific expression of PPARbeta/delta, driven by the myosin heavy chain (MHC-PPARbeta/delta mice). In striking contrast to MHC-PPARalpha mice, MHC-PPARbeta/delta mice had increased myocardial glucose utilization, did not accumulate myocardial lipid, and had normal cardiac function. Consistent with these observed metabolic phenotypes, we found that expression of genes involved in cellular FA transport were activated by PPARalpha but not by PPARbeta/delta. Conversely, cardiac glucose transport and glycolytic genes were activated in MHC-PPARbeta/delta mice, but repressed in MHC-PPARalpha mice. In reporter assays, we showed that PPARbeta/delta and PPARalpha exerted differential transcriptional control of the GLUT4 promoter, which may explain the observed isotype-specific effects on glucose uptake. Furthermore, myocardial injury due to ischemia/reperfusion injury was significantly reduced in the MHC-PPARbeta/delta mice compared with control or MHC-PPARalpha mice, consistent with an increased capacity for myocardial glucose utilization. These results demonstrate that PPARalpha and PPARbeta/delta drive distinct cardiac metabolic regulatory programs and identify PPARbeta/delta as a potential target for metabolic modulation therapy aimed at cardiac dysfunction caused by diabetes and ischemia.


Subject(s)
Cardiomyopathies/metabolism , Diabetes Mellitus/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , PPAR alpha/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Animals , Biological Transport/genetics , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Fatty Acids/genetics , Fatty Acids/metabolism , Glucose/metabolism , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Mice , Mice, Transgenic , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oxidation-Reduction , PPAR alpha/genetics , PPAR delta/genetics , PPAR-beta/genetics , Promoter Regions, Genetic/genetics
10.
Circ Res ; 100(8): 1208-17, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17363697

ABSTRACT

Obesity-related diabetes mellitus leads to increased myocardial uptake of fatty acids (FAs), resulting in a form of cardiac dysfunction referred to as lipotoxic cardiomyopathy. We have shown previously that chronic activation of the FA-activated nuclear receptor, peroxisome proliferator-activated receptor alpha (PPARalpha), is sufficient to drive the metabolic and functional abnormalities of the diabetic heart. Mice with cardiac-restricted overexpression of PPARalpha (myosin heavy chain [MHC]-PPARalpha) exhibit myocyte lipid accumulation and cardiac dysfunction. We sought to define the role of the long-chain FA transporter CD36 in the pathophysiology of lipotoxic forms of cardiomyopathy. MHC-PPARalpha mice were crossed with CD36-deficient mice (MHC-PPARalpha/CD36-/- mice). The absence of CD36 prevented myocyte triacylglyceride accumulation and cardiac dysfunction in the MHC-PPARalpha mice under basal conditions and following administration of high-fat diet. Surprisingly, the rescue of the MHC-PPARalpha phenotype by CD36 deficiency was associated with increased glucose uptake and oxidation rather than changes in FA utilization. As predicted by the metabolic changes, the activation of PPARalpha target genes involved in myocardial FA-oxidation pathways in the hearts of the MHC-PPARalpha mice was unchanged in the CD36-deficient background. However, PPARalpha-mediated suppression of genes involved in glucose uptake and oxidation was reversed in the MHC-PPARalpha/ CD36-/- mice. We conclude that CD36 is necessary for the development of lipotoxic cardiomyopathy in MHC-PPARalpha mice and that novel therapeutic strategies aimed at reducing CD36-mediated FA uptake show promise for the prevention or treatment of cardiac dysfunction related to obesity and diabetes.


Subject(s)
CD36 Antigens/genetics , Diabetic Angiopathies/genetics , Diabetic Angiopathies/prevention & control , Fatty Acids/metabolism , Fatty Acids/toxicity , Adiposity/genetics , Animals , CD36 Antigens/biosynthesis , CD36 Antigens/physiology , Diabetic Angiopathies/metabolism , Fatty Acids/biosynthesis , Glucose/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Oxidation-Reduction , PPAR alpha/deficiency , PPAR alpha/genetics , Phenotype
11.
J Biol Chem ; 282(12): 9216-27, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17213206

ABSTRACT

Previously, we identified calcium-independent phospholipase A2gamma (iPLA2gamma) with multiple translation initiation sites and dual mitochondrial and peroxisomal localization motifs. To determine the role of iPLA2gamma in integrating lipid and energy metabolism, we generated transgenic mice containing the alpha-myosin heavy chain promoter (alphaMHC) placed proximally to the human iPLA2gamma coding sequence that resulted in cardiac myocyte-restricted expression of iPLA2gamma (TGiPLA2gamma). TGiPLA2gamma mice possessed multiple phenotypes including: 1) a dramatic approximately 35% reduction in myocardial phospholipid mass in both the fed and mildly fasted states; 2) a marked accumulation of triglycerides during brief caloric restriction that represented 50% of total myocardial lipid mass; and 3) acute fasting-induced hemodynamic dysfunction. Biochemical characterization of the TGiPLA2gamma protein expressed in cardiac myocytes demonstrated over 25 distinct isoforms by two-dimensional SDS-PAGE Western analysis. Immunohistochemistry identified iPLA2gamma in the peroxisomal and mitochondrial compartments in both wild type and transgenic myocardium. Electron microscopy revealed the presence of loosely packed and disorganized mitochondrial cristae in TGiPLA2gamma mice that were accompanied by defects in mitochondrial function. Moreover, markedly elevated levels of 1-hydroxyl-2-arachidonoyl-sn-glycero-3-phosphocholine and 1-hydroxyl-2-docosahexaenoyl-sn-glycero-3-phosphocholine were prominent in the TGiPLA2gamma myocardium identifying the production of signaling metabolites by this enzyme in vivo. Collectively, these results identified the participation of iPLA2gamma in the remarkable lipid plasticity of myocardium, its role in generating signaling metabolites, and its prominent effects in modulating energy storage and utilization in myocardium in different metabolic contexts.


Subject(s)
Calcium/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Gene Expression Regulation , Myocardium/metabolism , Phospholipases A/genetics , Triglycerides/chemistry , Animals , Caloric Restriction , Group IV Phospholipases A2 , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Oxygen Consumption , Phospholipases A/biosynthesis , Spectrometry, Mass, Electrospray Ionization , Triglycerides/metabolism
12.
J Biol Chem ; 281(43): 32841-51, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16950770

ABSTRACT

The Akt family of serine-threonine kinases participates in diverse cellular processes, including the promotion of cell survival, glucose metabolism, and cellular protein synthesis. All three known Akt family members, Akt1, Akt2 and Akt3, are expressed in the myocardium, although Akt1 and Akt2 are most abundant. Previous studies demonstrated that Akt1 and Akt3 overexpression results in enhanced myocardial size and function. Yet, little is known about the role of Akt2 in modulating cardiac metabolism, survival, and growth. Here, we utilize murine models with targeted disruption of the akt2 or the akt1 genes to demonstrate that Akt2, but not Akt1, is required for insulin-stimulated 2-[(3)H]deoxyglucose uptake and metabolism. In contrast, akt2(-/-) mice displayed normal cardiac growth responses to provocative stimulation, including ligand stimulation of cultured cardiomyocytes, pressure overload by transverse aortic constriction, and myocardial infarction. However, akt2(-/-) mice were found to be sensitized to cardiomyocyte apoptosis in response to ischemic injury, and apoptosis was significantly increased in the peri-infarct zone of akt2(-/-) hearts 7 days after occlusion of the left coronary artery. These results implicate Akt2 in the regulation of cardiomyocyte metabolism and survival.


Subject(s)
Myocardium/enzymology , Myocardium/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Cell Survival/physiology , Cells, Cultured , Deoxyglucose/metabolism , Deoxyglucose/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Male , Mice , Mice, Knockout , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Time Factors
13.
Am J Physiol Heart Circ Physiol ; 290(1): H87-95, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16155108

ABSTRACT

High fatty acid oxidation (FAO) rates contribute to ischemia-reperfusion injury of the myocardium. Because peroxisome proliferator-activated receptor (PPAR)alpha regulates transcription of several FAO enzymes in the heart, we examined the response of mice with cardiac-restricted overexpression of PPARalpha (MHC-PPARalpha) or whole body PPARalpha deletion including the heart (PPARalpha-/-) to myocardial ischemia-reperfusion injury. Isolated working hearts from MHC-PPARalpha and nontransgenic (NTG) littermates were subjected to no-flow global ischemia followed by reperfusion. MHC-PPARalpha hearts had significantly higher FAO rates during aerobic and postischemic reperfusion (aerobic 1,479 +/- 171 vs. 699 +/- 117, reperfusion 1,062 +/- 214 vs. 601 +/- 70 nmol x g dry wt(-1) x min(-1); P < 0.05) and significantly lower glucose oxidation rates compared with NTG hearts (aerobic 225 +/- 36 vs. 1,563 +/- 165, reperfusion 402 +/- 54 vs. 1,758 +/- 165 nmol x g dry wt(-1) x min(-1); P < 0.05). In hearts from PPARalpha-/- mice, FAO was significantly lower during aerobic and reperfusion (aerobic 235 +/- 36 vs. 442 +/- 75, reperfusion 205 +/- 25 vs. 346 +/- 38 nmol x g dry wt(-1) x min(-1); P < 0.05) whereas glucose oxidation was significantly higher compared with wild-type (WT) hearts (aerobic 2,491 +/- 631 vs. 901 +/- 119, reperfusion 2,690 +/- 562 vs. 1,315 +/- 172 nmol x g dry wt(-1) x min(-1); P < 0.05). Increased FAO rates in MHC-PPARalpha hearts were associated with a markedly lower recovery of cardiac power (45 +/- 9% vs. 71 +/- 6% of preischemic levels in NTG hearts; P < 0.05). In contrast, the percent recovery of cardiac power of PPARalpha-/- hearts was not significantly different from that of WT hearts (80 +/- 8% vs. 75 +/- 9%). This study demonstrates that chronic activation of PPARalpha is detrimental to the cardiac recovery during reperfusion after ischemia.


Subject(s)
Fatty Acids/metabolism , Myocardial Ischemia/physiopathology , Myocardium/metabolism , PPAR alpha/metabolism , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/metabolism , Animals , Glucose/metabolism , Glycolysis , Heart Function Tests , Male , Mice , Mice, Transgenic , Multienzyme Complexes/metabolism , Myocardial Reperfusion Injury/physiopathology , PPAR alpha/deficiency , Palmitic Acid/metabolism , Protein Serine-Threonine Kinases/metabolism
14.
Can J Physiol Pharmacol ; 84(11): 1215-22, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17218986

ABSTRACT

The concentration of fatty acids in the blood or perfusate is a major determinant of the extent of myocardial fatty acid oxidation. Increasing fatty acid supply in adult rat increases myocardial fatty acid oxidation. Plasma levels of fatty acids increase post-surgery in infants undergoing cardiac bypass operation to correct congenital heart defects. How a newborn heart responds to increased fatty acid supply remains to be determined. In this study, we examined whether the tissue levels of malonyl CoA decrease to relieve the inhibition on carnitine palmitoyltransferase (CPT) I when the myocardium is exposed to higher concentrations of long-chain fatty acids in newborn rabbit heart. We then tested the contribution of the enzymes that regulate tissue levels of malonyl CoA, acetyl CoA carboxylase (ACC), and malonyl CoA decarboxylase (MCD). Our results showed that increasing fatty acid supply from 0.4 mmol/L (physiological) to 1.2 mmol/L (pathological) resulted in an increase in cardiac fatty acid oxidation rates and this was accompanied by a decrease in tissue malonyl CoA levels. The decrease in malonyl CoA was not related to any alterations in total and phosphorylated acetyl CoA carboxylase protein or the activities of acetyl CoA carboxylase and malonyl CoA decarboxylase. Our results suggest that the regulatory role of malonyl CoA remained when the hearts were exposed to high levels of fatty acids.


Subject(s)
Malonyl Coenzyme A/metabolism , Myocardium/metabolism , Palmitic Acid/metabolism , AMP-Activated Protein Kinases , Acetyl Coenzyme A/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Animals, Newborn , Carboxy-Lyases/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Citric Acid Cycle , Dose-Response Relationship, Drug , Glucose/metabolism , In Vitro Techniques , Multienzyme Complexes/metabolism , Myocardium/enzymology , Oxidation-Reduction , Palmitic Acid/pharmacology , Protein Serine-Threonine Kinases/metabolism , Rabbits
15.
Cell Metab ; 1(2): 133-44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16054054

ABSTRACT

The role of the peroxisome proliferator-activated receptor-alpha (PPARalpha) in the development of insulin-resistant diabetes was evaluated using gain- and loss-of-function approaches. Transgenic mice overexpressing PPARalpha in muscle (MCK-PPARalpha mice) developed glucose intolerance despite being protected from diet-induced obesity. Conversely, PPARalpha null mice were protected from diet-induced insulin resistance in the context of obesity. In skeletal muscle, MCK-PPARalpha mice exhibited increased fatty acid oxidation rates, diminished AMP-activated protein kinase activity, and reduced insulin-stimulated glucose uptake without alterations in the phosphorylation status of key insulin-signaling proteins. These effects on muscle glucose uptake involved transcriptional repression of the GLUT4 gene. Pharmacologic inhibition of fatty acid oxidation or mitochondrial respiratory coupling prevented the effects of PPARalpha on GLUT4 expression and glucose homeostasis. These results identify PPARalpha-driven alterations in muscle fatty acid oxidation and energetics as a potential link between obesity and the development of glucose intolerance and insulin resistance.


Subject(s)
Diabetes Mellitus/metabolism , Muscles/metabolism , Obesity/metabolism , PPAR alpha/metabolism , Animals , Blotting, Northern , Blotting, Western , DNA, Complementary/metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacokinetics , Insulin/metabolism , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscles/cytology , Oligonucleotide Array Sequence Analysis , Oxygen/metabolism , Phenotype , Phosphatidylinositol 3-Kinases , Phosphorylation , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcription, Genetic
16.
PLoS Biol ; 3(4): e101, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15760270

ABSTRACT

The gene encoding the transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) was targeted in mice. PGC-1alpha null (PGC-1alpha(-/-)) mice were viable. However, extensive phenotyping revealed multi-system abnormalities indicative of an abnormal energy metabolic phenotype. The postnatal growth of heart and slow-twitch skeletal muscle, organs with high mitochondrial energy demands, is blunted in PGC-1alpha(-/-) mice. With age, the PGC-1alpha(-/-) mice develop abnormally increased body fat, a phenotype that is more severe in females. Mitochondrial number and respiratory capacity is diminished in slow-twitch skeletal muscle of PGC-1alpha(-/-) mice, leading to reduced muscle performance and exercise capacity. PGC-1alpha(-/-) mice exhibit a modest diminution in cardiac function related largely to abnormal control of heart rate. The PGC-1alpha(-/-) mice were unable to maintain core body temperature following exposure to cold, consistent with an altered thermogenic response. Following short-term starvation, PGC-1alpha(-/-) mice develop hepatic steatosis due to a combination of reduced mitochondrial respiratory capacity and an increased expression of lipogenic genes. Surprisingly, PGC-1alpha(-/-) mice were less susceptible to diet-induced insulin resistance than wild-type controls. Lastly, vacuolar lesions were detected in the central nervous system of PGC-1alpha(-/-) mice. These results demonstrate that PGC-1alpha is necessary for appropriate adaptation to the metabolic and physiologic stressors of postnatal life.


Subject(s)
Fatty Liver/genetics , Muscular Diseases/genetics , Obesity/genetics , Trans-Activators/deficiency , Trans-Activators/genetics , Animals , Body Weight/genetics , Cerebrovascular Disorders/genetics , Exons , Fatty Liver/enzymology , Female , Insulin Resistance/genetics , Male , Mice , Mice, Knockout , Molecular Sequence Data , Muscular Diseases/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors , Transcription, Genetic
17.
Circ Res ; 96(2): 225-33, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15618539

ABSTRACT

Evidence is emerging that systemic metabolic disturbances contribute to cardiac myocyte dysfunction and clinically apparent heart failure, independent of associated coronary artery disease. To test the hypothesis that perturbation of lipid homeostasis in cardiomyocytes contributes to cardiac dysfunction, we engineered transgenic mice with cardiac-specific overexpression of fatty acid transport protein 1 (FATP1) using the alpha-myosin heavy chain gene promoter. Two independent transgenic lines demonstrate 4-fold increased myocardial free fatty acid (FFA) uptake that is consistent with the known function of FATP1. Increased FFA uptake in this model likely contributes to early cardiomyocyte FFA accumulation (2-fold increased) and subsequent increased cardiac FFA metabolism (2-fold). By 3 months of age, transgenic mice have echocardiographic evidence of impaired left ventricular filling and biatrial enlargement, but preserved systolic function. Doppler tissue imaging and hemodynamic studies confirm that these mice have predominantly diastolic dysfunction. Furthermore, ambulatory ECG monitoring reveals prolonged QT(c) intervals, reflecting reductions in the densities of repolarizing, voltage-gated K+ currents in ventricular myocytes. Our results show that in the absence of systemic metabolic disturbances, such as diabetes or hyperlipidemia, perturbation of cardiomyocyte lipid homeostasis leads to cardiac dysfunction with pathophysiological findings similar to those in diabetic cardiomyopathy. Moreover, the MHC-FATP model supports a role for FATPs in FFA import into the heart in vivo.


Subject(s)
Cardiomyopathies/metabolism , Fatty Acids, Nonesterified/metabolism , Membrane Transport Proteins/physiology , Myocytes, Cardiac/metabolism , Animals , Biological Transport , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cell Size , Diastole , Electrocardiography , Fatty Acid Transport Proteins , Gene Expression , Glucose/metabolism , Hypertrophy , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Myocardium/metabolism , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Positron-Emission Tomography , Potassium Channels, Voltage-Gated/metabolism , Recombinant Fusion Proteins/physiology
18.
Eur J Biochem ; 271(13): 2831-40, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15206948

ABSTRACT

Malonyl-CoA, a potent inhibitor of carnitine pamitoyl transferase-I (CPT-I), plays a pivotal role in fuel selection in cardiac muscle. Malonyl-CoA decarboxylase (MCD) catalyzes the degradation of malonyl-CoA, removes a potent allosteric inhibition on CPT-I and thereby increases fatty acid oxidation in the heart. Although MCD has several Ser/Thr phosphorylation sites, whether it is regulated by AMP-activated protein kinase (AMPK) has been controversial. We therefore overexpressed MCD (Ad.MCD) and constitutively active AMPK (Ad.CA-AMPK) in H9c2 cells, using an adenoviral gene delivery approach in order to examine if MCD is regulated by AMPK. Cells infected with Ad.CA-AMPK demonstrated a fourfold increase in AMPK activity as compared with control cells expressing green fluorescent protein (Ad.GFP). MCD activity increased 40- to 50-fold in Ad.MCD + Ad.GFP cells when compared with Ad.GFP control. Co-expressing AMPK with MCD further augmented MCD expression and activity in Ad.MCD + Ad.CA-AMPK cells compared with the Ad.MCD + Ad.GFP control. Subcellular fractionation further revealed that 54.7 kDa isoform of MCD expression was significantly higher in cytosolic fractions of Ad.MCD + Ad.CA-AMPK cells than of the Ad.MCD +Ad.GFP control. However, the MCD activities in cytosolic fractions were not different between the two groups. Interestingly, in the mitochondrial fractions, MCD activity significantly increased in Ad.MCD + Ad.CA-AMPK cells when compared with Ad.MCD + Ad.GFP cells. Using phosphoserine and phosphothreonine antibodies, no phosphorylation of MCD by AMPK was observed. The increase in MCD activity in mitochondria-rich fractions of Ad.MCD + Ad.CA-AMPK cells was accompanied by an increase in the level of the 50.7 kDa isoform of MCD protein in the mitochondria. This differential regulation of MCD expression and activity in the mitochondria by AMPK may potentially regulate malonyl-CoA levels at sites nearby CPT-I on the mitochondria.


Subject(s)
Adenoviridae/genetics , Adenylate Kinase/metabolism , Carboxy-Lyases/metabolism , Gene Transfer Techniques , Subcellular Fractions/enzymology , Animals , Cell Line , Phosphorylation , Rats
19.
Prog Lipid Res ; 42(3): 238-56, 2003 May.
Article in English | MEDLINE | ID: mdl-12689619

ABSTRACT

Myocardial ischemia is the leading cause of all cardiovascular deaths in North America. Myocardial ischemia is accompanied by profound changes in metabolism including alterations in glucose and fatty acid metabolism, increased uncoupling of glucose oxidation from glycolysis and accumulation of protons within the myocardium. These changes can contribute to a poor functional recovery of the heart. One key player in the ischemia-induced alteration in fatty acid and glucose metabolism is 5'AMP-activated protein kinase (AMPK). Accumulating evidence suggest that activation of AMPK during myocardial ischemia both increases glucose uptake and glycolysis while also increasing fatty acid oxidation during reperfusion. Gain-of-function mutations of AMPK in cardiac muscle may also be causally related to the development of hypertrophic cardiomyopathies. Therefore, a better understanding of role of AMPK in cardiac metabolism is necessary to appropriately modulate its activity as a potential therapeutic target in treating ischemia reperfusion injuries. This review attempts to update some of the recent findings that delineate various pathways through which AMPK regulates glucose and fatty acid metabolism in the ischemic myocardium.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Multienzyme Complexes/physiology , Myocardial Ischemia/metabolism , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases , Energy Metabolism , Glycolysis , Humans , Multienzyme Complexes/genetics , Mutation , Myocardial Reperfusion Injury/metabolism , Oxidation-Reduction , Protein Serine-Threonine Kinases/genetics
20.
Heart Fail Rev ; 7(2): 161-73, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11988640

ABSTRACT

In response to a prolonged pressure- or volume-overload, alterations occur in myocardial fatty acid, glucose, and glycogen metabolism. Oxidation of long chain fatty acids has been found to be reduced in hypertrophied hearts compared to non-hypertrophied hearts. However, this observation depends upon the degree of cardiac hypertrophy, the severity of carnitine deficiency, the concentration of fatty acid in blood or perfusate, and the myocardial workload. Glycolysis of exogenous glucose is accelerated in hypertrophied hearts. Despite the acceleration of glycolysis, glucose oxidation is not correspondingly increased leading to lower coupling between glycolysis and glucose oxidation and greater H(+) production than in non-hypertrophied hearts. Although glycogen metabolism does not differ in the absence of ischemia, synthesis and degradation of glycogen are accelerated in severely ischemic hypertrophied hearts. These alterations in carbohydrate metabolism may contribute to the increased susceptibility of hypertrophied hearts to injury during ischemia and reperfusion by causing disturbances in ion homeostasis that reduce contractile function and efficiency to a greater extent than normal. As in non-hypertrophied hearts, pharmacologic enhancement of coupling between glycolysis and glucose oxidation (e.g., by directly stimulating glucose oxidation) improves recovery of function of hypertrophied hearts after ischemia. This observation provides strong support for the concept that modulation of energy metabolism in the hypertrophied heart is a useful approach to improve function of the hypertrophied heart during ischemia and reperfusion. Future investigations are necessary to determine if alternative approaches, such as glucose-insulin-potassium infusion and inhibitors of fatty acid oxidation (e.g., ranolazine, trimetazidine), also produce beneficial effects in ischemic and reperfused hypertrophied hearts.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Energy Metabolism/physiology , Cardiomegaly/etiology , Glucose/metabolism , Humans , Myocardial Contraction/physiology , Myocardial Ischemia/etiology , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Severity of Illness Index , Stroke Volume/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...