Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Diabetes ; 69(10): 2094-2111, 2020 10.
Article in English | MEDLINE | ID: mdl-32366681

ABSTRACT

Cardiac glucose uptake and oxidation are reduced in diabetes despite hyperglycemia. Mitochondrial dysfunction contributes to heart failure in diabetes. It is unclear whether these changes are adaptive or maladaptive. To directly evaluate the relationship between glucose delivery and mitochondrial dysfunction in diabetic cardiomyopathy, we generated transgenic mice with inducible cardiomyocyte-specific expression of the GLUT4. We examined mice rendered hyperglycemic following low-dose streptozotocin prior to increasing cardiomyocyte glucose uptake by transgene induction. Enhanced myocardial glucose in nondiabetic mice decreased mitochondrial ATP generation and was associated with echocardiographic evidence of diastolic dysfunction. Increasing myocardial glucose delivery after short-term diabetes onset exacerbated mitochondrial oxidative dysfunction. Transcriptomic analysis revealed that the largest changes, driven by glucose and diabetes, were in genes involved in mitochondrial function. This glucose-dependent transcriptional repression was in part mediated by O-GlcNAcylation of the transcription factor Sp1. Increased glucose uptake induced direct O-GlcNAcylation of many electron transport chain subunits and other mitochondrial proteins. These findings identify mitochondria as a major target of glucotoxicity. They also suggest that reduced glucose utilization in diabetic cardiomyopathy might defend against glucotoxicity and caution that restoring glucose delivery to the heart in the context of diabetes could accelerate mitochondrial dysfunction by disrupting protective metabolic adaptations.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Animals , Diabetic Cardiomyopathies/genetics , Fatty Acids , Glucose , Mice , Mitochondria , Myocardium
2.
Am J Physiol Heart Circ Physiol ; 313(6): H1098-H1108, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28822962

ABSTRACT

Pathological cardiac hypertrophy may be associated with reduced expression of glucose transporter 4 (GLUT4) in contrast to exercise-induced cardiac hypertrophy, where GLUT4 levels are increased. However, mice with cardiac-specific deletion of GLUT4 (G4H-/-) have normal cardiac function in the unstressed state. This study tested the hypothesis that cardiac GLUT4 is required for myocardial adaptations to hemodynamic demands. G4H-/- and control littermates were subjected to either a pathological model of left ventricular pressure overload [transverse aortic constriction (TAC)] or a physiological model of endurance exercise (swim training). As predicted after TAC, G4H-/- mice developed significantly greater hypertrophy and more severe contractile dysfunction. Somewhat surprisingly, after exercise training, G4H-/- mice developed increased fibrosis and apoptosis that was associated with dephosphorylation of the prosurvival kinase Akt in concert with an increase in protein levels of the upstream phosphatase protein phosphatase 2A (PP2A). Exercise has been shown to decrease levels of ceramide; G4H-/- hearts failed to decrease myocardial ceramide in response to exercise. Furthermore, G4H-/- hearts have reduced levels of the transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator-1, lower carnitine palmitoyl-transferase activity, and reduced hydroxyacyl-CoA dehydrogenase activity. These basal changes may also contribute to the impaired ability of G4H-/- hearts to adapt to hemodynamic stresses. In conclusion, GLUT4 is required for the maintenance of cardiac structure and function in response to physiological or pathological processes that increase energy demands, in part through secondary changes in mitochondrial metabolism and cellular stress survival pathways such as Akt.NEW & NOTEWORTHY Glucose transporter 4 (GLUT4) is required for myocardial adaptations to exercise, and its absence accelerates heart dysfunction after pressure overload. The requirement for GLUT4 may extend beyond glucose uptake to include defects in mitochondrial metabolism and survival signaling pathways that develop in its absence. Therefore, GLUT4 is critical for responses to hemodynamic stresses.


Subject(s)
Cardiomegaly, Exercise-Induced , Cardiomegaly/metabolism , Glucose Transporter Type 4/deficiency , Hemodynamics , Myocardium/metabolism , Ventricular Function, Left , Ventricular Remodeling , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Adaptation, Physiological , Animals , Aorta/physiopathology , Aorta/surgery , Cardiomegaly/etiology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Carnitine O-Palmitoyltransferase/metabolism , Constriction , Disease Models, Animal , Genetic Predisposition to Disease , Glucose Transporter Type 4/genetics , Mice, Knockout , Myocardial Contraction , Myocardium/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Physical Exertion , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism
3.
J Mol Cell Cardiol ; 85: 104-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26004364

ABSTRACT

Cardiac dysfunction in obesity is associated with mitochondrial dysfunction, oxidative stress and altered insulin sensitivity. Whether oxidative stress directly contributes to myocardial insulin resistance remains to be determined. This study tested the hypothesis that ROS scavenging will improve mitochondrial function and insulin sensitivity in the hearts of rodent models with varying degrees of insulin resistance and hyperglycemia. The catalytic antioxidant MnTBAP was administered to the uncoupling protein-diphtheria toxin A (UCP-DTA) mouse model of insulin resistance (IR) and obesity, at early and late time points in the evolution of IR, and to db/db mice with severe obesity and type-two diabetes. Mitochondrial function was measured in saponin-permeabilized cardiac fibers. Aconitase activity and hydrogen peroxide emission were measured in isolated mitochondria. Insulin-stimulated glucose oxidation, glycolysis and fatty acid oxidation rates were measured in isolated working hearts, and 2-deoxyglucose uptake was measured in isolated cardiomyocytes. Four weeks of MnTBAP attenuated glucose intolerance in 13-week-old UCP-DTA mice but was without effect in 24-week-old UCP-DTA mice and in db/db mice. Despite the absence of improvement in the systemic metabolic milieu, MnTBAP reversed cardiac mitochondrial oxidative stress and improved mitochondrial bioenergetics by increasing ATP generation and reducing mitochondrial uncoupling in all models. MnTBAP also improved myocardial insulin mediated glucose metabolism in 13 and 24-week-old UCP-DTA mice. Pharmacological ROS scavenging improves myocardial energy metabolism and insulin responsiveness in obesity and type 2 diabetes via direct effects that might be independent of changes in systemic metabolism.


Subject(s)
Antioxidants/pharmacology , Metabolic Syndrome/drug therapy , Metalloporphyrins/pharmacology , Mitochondria, Heart/metabolism , Animals , Antioxidants/therapeutic use , Drug Evaluation, Preclinical , Energy Metabolism , Fatty Acids/metabolism , Homeostasis , Insulin/blood , Insulin Resistance , Metabolic Syndrome/blood , Metalloporphyrins/therapeutic use , Mice, Inbred C57BL , Mice, Obese , Myocardium/metabolism , Oxidative Stress , Signal Transduction
4.
Mol Cell Biol ; 35(5): 831-46, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25535334

ABSTRACT

Sustained Akt activation induces cardiac hypertrophy (LVH), which may lead to heart failure. This study tested the hypothesis that Akt activation contributes to mitochondrial dysfunction in pathological LVH. Akt activation induced LVH and progressive repression of mitochondrial fatty acid oxidation (FAO) pathways. Preventing LVH by inhibiting mTOR failed to prevent the decline in mitochondrial function, but glucose utilization was maintained. Akt activation represses expression of mitochondrial regulatory, FAO, and oxidative phosphorylation genes in vivo that correlate with the duration of Akt activation in part by reducing FOXO-mediated transcriptional activation of mitochondrion-targeted nuclear genes in concert with reduced signaling via peroxisome proliferator-activated receptor α (PPARα)/PGC-1α and other transcriptional regulators. In cultured myocytes, Akt activation disrupted mitochondrial bioenergetics, which could be partially reversed by maintaining nuclear FOXO but not by increasing PGC-1α. Thus, although short-term Akt activation may be cardioprotective during ischemia by reducing mitochondrial metabolism and increasing glycolysis, long-term Akt activation in the adult heart contributes to pathological LVH in part by reducing mitochondrial oxidative capacity.


Subject(s)
Cardiomegaly/metabolism , Cell Nucleus/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adenosine Triphosphate/metabolism , Animals , Fatty Acids/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Glycolysis , Heart/physiology , Hemodynamics , Hypertrophy , Male , Mice , Muscle Cells/cytology , Oxygen/metabolism , PPAR alpha/metabolism , Proteomics , Signal Transduction , Transcription, Genetic , Transgenes
5.
Diabetes ; 61(12): 3260-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22912419

ABSTRACT

These studies investigate the role of uncoupling protein 3 (UCP3) in cardiac energy metabolism, cardiac O(2) consumption (MVO(2)), cardiac efficiency (CE), and mitochondrial uncoupling in high fat (HF)-fed or leptin-deficient mice. UCP3KO and wild-type (WT) mice were fed normal chow or HF diets for 10 weeks. Substrate utilization rates, MVO(2), CE, and mitochondrial uncoupling were measured in perfused working hearts and saponin-permeabilized cardiac fibers, respectively. Similar analyses were performed in hearts of ob/ob mice lacking UCP3 (U3OB mice). HF increased cardiac UCP3 protein. However, fatty acid (FA) oxidation rates were similarly increased by HF diet in WT and UCP3KO mice. By contrast, MVO(2) increased in WT, but not in UCP3KO with HF, leading to increased CE in UCP3KO mice. Consistent with increased CE, mitochondrial coupling was increased in the hearts of HF-fed UCP3KO mice. Unexpectedly, UCP3 deletion in ob/ob mice reduced FA oxidation but had no effect on MVO(2) or CE. In addition, FA-induced mitochondrial uncoupling was similarly enhanced in U3OB compared with ob/ob hearts and was associated with elevated mitochondrial thioesterase-1 protein content. These studies show that although UCP3 may mediate mitochondrial uncoupling and reduced CE after HF feeding, it does not mediate uncoupling in leptin-deficient states.


Subject(s)
Energy Metabolism/physiology , Ion Channels/metabolism , Leptin/deficiency , Mitochondrial Proteins/metabolism , Oxygen Consumption/physiology , Animals , Dietary Fats/adverse effects , Energy Metabolism/genetics , Ion Channels/genetics , Leptin/genetics , Male , Mice , Mice, Knockout , Mitochondria , Mitochondrial Proteins/genetics , Oxygen Consumption/genetics , Palmitoyl-CoA Hydrolase/metabolism , Uncoupling Protein 3
6.
J Mol Cell Cardiol ; 52(5): 1019-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22342406

ABSTRACT

AIMS: To determine the contribution of insulin signaling versus systemic metabolism to metabolic and mitochondrial alterations in type 1 diabetic hearts and test the hypothesis that antecedent mitochondrial dysfunction contributes to impaired cardiac efficiency (CE) in diabetes. METHODS AND RESULTS: Control mice (WT) and mice with cardiomyocyte-restricted deletion of insulin receptors (CIRKO) were rendered diabetic with streptozotocin (WT-STZ and CIRKO-STZ, respectively), non-diabetic controls received vehicle (citrate buffer). Cardiac function was determined by echocardiography; myocardial metabolism, oxygen consumption (MVO(2)) and CE were determined in isolated perfused hearts; mitochondrial function was determined in permeabilized cardiac fibers and mitochondrial proteomics by liquid chromatography mass spectrometry. Pyruvate supported respiration and ATP synthesis were equivalently reduced by diabetes and genotype, with synergistic impairment in ATP synthesis in CIRKO-STZ. In contrast, fatty acid delivery and utilization was increased by diabetes irrespective of genotype, but not in non-diabetic CIRKO. Diabetes and genotype synergistically increased MVO(2) in CIRKO-STZ, leading to reduced CE. Irrespective of diabetes, genotype impaired ATP/O ratios in mitochondria exposed to palmitoyl carnitine, consistent with mitochondrial uncoupling. Proteomics revealed reduced content of fatty acid oxidation proteins in CIRKO mitochondria, which were induced by diabetes, whereas tricarboxylic acid cycle and oxidative phosphorylation proteins were reduced both in CIRKO mitochondria and by diabetes. CONCLUSIONS: Deficient insulin signaling and diabetes mediate distinct effects on cardiac mitochondria. Antecedent loss of insulin signaling markedly impairs CE when diabetes is induced, via mechanisms that may be secondary to mitochondrial uncoupling and increased FA utilization.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Heart/physiopathology , Receptor, Insulin/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Gene Knockout Techniques , In Vitro Techniques , Insulin/physiology , Ion Channels/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/physiology , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Organelle Size , Oxidation-Reduction , Oxidative Stress , Oxygen Consumption , Proteome/metabolism , Reactive Oxygen Species/metabolism , Receptor, Insulin/deficiency , Uncoupling Protein 3
7.
Diabetes ; 60(5): 1424-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21441440

ABSTRACT

OBJECTIVE: ob/ob and db/db mice manifest myocardial hypertrophy, insulin resistance, altered substrate utilization, mitochondrial dysfunction, and lipid accumulation. This study was designed to determine the contribution of central and peripheral leptin signaling to myocardial metabolism and function in ob/ob and db/db mice in the absence of diabetes and morbid obesity. RESEARCH DESIGN AND METHODS: Male ob/ob mice (aged 4 weeks) were caloric restricted by pairfeeding to a leptin-treated ob/ob group. In addition to determining glucose tolerance and circulating lipid concentrations, myocardial substrate metabolism and mitochondrial function were determined in saponin-permeabilized cardiac fibers. Second, experiments were performed to determine whether leptin treatment by intraperitoneal injection or intracerebroventricular infusion could normalize myocardial palmitate oxidation in caloric-restricted ob/ob mouse hearts. RESULTS: Despite normalizing body weight and glucose tolerance, fat mass and circulating lipid levels remained increased in caloric-restricted ob/ob animals. Palmitate oxidation remained elevated in caloric-restricted ob/ob hearts and was normalized by intraperitoneal or intracerebroventricular leptin. Intraperitoneal and intracerebroventricular treatment also normalized circulating free fatty acid levels, myocardial fatty acid oxidation gene expression, and myocardial insulin sensitivity. CONCLUSIONS: These data suggest that impaired hypothalamic leptin signaling is sufficient to increase myocardial fatty acid oxidation by increasing delivery of free fatty acid substrates and peroxisome proliferator-activated receptor-α ligands to the heart.


Subject(s)
Caloric Restriction , Fatty Acids/metabolism , Heart/drug effects , Leptin/blood , Leptin/pharmacology , Myocardium/metabolism , Signal Transduction , Adiponectin/blood , Animals , Body Composition/drug effects , Fatty Acids/blood , Glucose Tolerance Test , Injections, Intraperitoneal , Leptin/administration & dosage , Male , Mice , Mice, Obese , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Myocardium/ultrastructure , Oxidation-Reduction/drug effects , Palmitic Acids/metabolism , Polymerase Chain Reaction , Triglycerides/blood , Triglycerides/metabolism
8.
J Clin Invest ; 120(10): 3443-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852389

ABSTRACT

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


Subject(s)
Fatty Acids/metabolism , Lipids/toxicity , Myocardium/metabolism , PPAR alpha/physiology , PPAR gamma/physiology , Animals , Apoptosis , Fatty Acids, Nonesterified/blood , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Myocytes, Cardiac/ultrastructure , Oxidation-Reduction , PPAR alpha/deficiency , Reactive Oxygen Species/metabolism , Triglycerides/biosynthesis
9.
Am J Physiol Endocrinol Metab ; 299(5): E721-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739511

ABSTRACT

Adiponectin promotes cardioprotection by various mechanisms, and this study used primary cardiomyocytes and the isolated working perfused heart to investigate cardiometabolic effects. We show in adult cardiomyocytes that adiponectin increased CD36 translocation and fatty acid uptake as well as insulin-stimulated glucose transport and Akt phosphorylation. Coimmunoprecipitation showed that adiponectin enhanced association of AdipoR1 with APPL1, subsequent binding of APPL1 with AMPKα2, which led to phosphorylation and inhibition of ACC and increased fatty acid oxidation. Using siRNA to effectively knockdown APPL1 in neonatal cardiomyocytes, we demonstrated an essential role for APPL1 in mediating increased fatty acid uptake and oxidation by adiponectin. Importantly, enhanced fatty acid oxidation in conjunction with AMPK and ACC phosphorylation was also observed in the isolated working heart. Despite increasing fatty acid oxidation and myocardial oxygen consumption, adiponectin increased hydraulic work and maintained cardiac efficiency. In summary, the present study documents several beneficial metabolic effects mediated by adiponectin in the heart and provides novel insight into the mechanisms behind these effects, in particular the importance of APPL1.


Subject(s)
Adenylate Kinase/metabolism , Adiponectin/metabolism , CD36 Antigens/metabolism , Carrier Proteins/metabolism , Myocardium/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Fatty Acids/metabolism , Immunoblotting , Immunohistochemistry , Immunoprecipitation , In Vitro Techniques , Male , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adiponectin/metabolism , Signal Transduction
10.
Diabetes ; 57(11): 2924-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18678617

ABSTRACT

OBJECTIVE: Fatty acid-induced mitochondrial uncoupling and oxidative stress have been proposed to reduce cardiac efficiency and contribute to cardiac dysfunction in type 2 diabetes. We hypothesized that mitochondrial uncoupling may also contribute to reduced cardiac efficiency and contractile dysfunction in the type 1 diabetic Akita mouse model (Akita). RESEARCH DESIGN AND METHODS: Cardiac function and substrate utilization were determined in isolated working hearts and in vivo function by echocardiography. Mitochondrial function and coupling were determined in saponin-permeabilized fibers, and proton leak kinetics was determined in isolated mitochondria. Hydrogen peroxide production and aconitase activity were measured in isolated mitochondria, and total reactive oxygen species (ROS) were measured in heart homogenates. RESULTS: Resting cardiac function was normal in Akita mice, and myocardial insulin sensitivity was preserved. Although Akita hearts oxidized more fatty acids, myocardial O(2) consumption was not increased, and cardiac efficiency was not reduced. ADP-stimulated mitochondrial oxygen consumption and ATP synthesis were decreased, and mitochondria showed grossly abnormal morphology in Akita. There was no evidence of oxidative stress, and despite a twofold increase in uncoupling protein 3 (UCP3) content, ATP-to-O ratios and proton leak kinetics were unchanged, even after perfusion of Akita hearts with 1 mmol/l palmitate. CONCLUSIONS: Insulin-deficient Akita hearts do not exhibit fatty acid-induced mitochondrial uncoupling, indicating important differences in the basis for mitochondrial dysfunction between insulin-responsive type 1 versus insulin-resistant type 2 diabetic hearts. Increased UCP3 levels do not automatically increase mitochondrial uncoupling in the heart, which supports the hypothesis that fatty acid-induced mitochondrial uncoupling as exists in type 2 diabetic hearts requires a concomitant increase in ROS generation.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin/metabolism , Ion Channels/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Animals , Blotting, Western , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Echocardiography , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria, Heart/ultrastructure , Mitochondrial Proteins/genetics , Myocardium/ultrastructure , Oxidative Stress , Oxygen Consumption , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uncoupling Protein 2 , Uncoupling Protein 3
11.
J Lipid Res ; 49(10): 2101-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18515784

ABSTRACT

Ceramide is among a number of potential lipotoxic molecules that are thought to modulate cellular energy metabolism. The heart is one of the tissues thought to become dysfunctional due to excess lipid accumulation. Dilated lipotoxic cardiomyopathy, thought to be the result of diabetes and severe obesity, has been modeled in several genetically altered mice, including animals with cardiac-specific overexpression of glycosylphosphatidylinositol (GPI)-anchored human lipoprotein lipase (LpL(GPI)). To test whether excess ceramide was implicated in cardiac lipotoxicity, de novo ceramide biosynthesis was inhibited pharmacologically by myriocin and genetically by heterozygous deletion of LCB1, a subunit of serine palmitoyltransferase (SPT). Inhibition of SPT, a rate-limiting enzyme in ceramide biosynthesis, reduced fatty acid and increased glucose oxidation in isolated perfused LpL(GPI) hearts, improved systolic function, and prolonged survival rates. Our results suggest a critical role for ceramide accumulation in the pathogenesis of lipotoxic cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiotoxins/metabolism , Ceramides/metabolism , Animals , Biomarkers/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cardiotoxins/antagonists & inhibitors , Cattle , Ceramides/antagonists & inhibitors , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/pharmacology , Gene Deletion , Gene Expression Regulation/drug effects , Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Glycosylphosphatidylinositols/metabolism , Heart/drug effects , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Lipoprotein Lipase/metabolism , Mice , Mice, Transgenic , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphorylation/drug effects , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Survival Rate
12.
Cell Metab ; 6(4): 294-306, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17908558

ABSTRACT

Physiological cardiac hypertrophy is associated with mitochondrial adaptations that are characterized by activation of PGC-1alpha and increased fatty acid oxidative (FAO) capacity. It is widely accepted that phosphatidylinositol 3-kinase (PI3K) signaling to Akt1 is required for physiological cardiac growth. However, the signaling pathways that coordinate physiological hypertrophy and metabolic remodeling are incompletely understood. We show here that activation of PI3K is sufficient to increase myocardial FAO capacity and that inhibition of PI3K signaling prevents mitochondrial adaptations in response to physiological hypertrophic stimuli despite increased expression of PGC-1alpha. We also show that activation of the downstream kinase Akt is not required for the mitochondrial adaptations that are secondary to PI3K activation. Thus, in physiological cardiac growth, PI3K is an integrator of cellular growth and metabolic remodeling. Although PI3K signaling to Akt1 is required for cellular growth, Akt-independent pathways mediate the accompanying mitochondrial adaptations.


Subject(s)
Cardiomegaly/enzymology , Mitochondria/physiology , Phosphatidylinositol 3-Kinases/physiology , Physical Conditioning, Animal , Proto-Oncogene Proteins c-akt/physiology , Adaptation, Physiological , Animals , Fatty Acids/metabolism , Gene Expression Regulation , Isoenzymes/antagonists & inhibitors , Mice , Mice, Mutant Strains , Myocytes, Cardiac/enzymology , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/genetics , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...