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










Publication year range
1.
Front Genet ; 15: 1375467, 2024.
Article in English | MEDLINE | ID: mdl-38706791

ABSTRACT

Leigh syndrome French Canadian type (LSFC) is a recessive neurodegenerative disease characterized by tissue-specific deficiency in cytochrome c oxidase (COX), the fourth complex in the oxidative phosphorylation system. LSFC is caused by mutations in the leucine rich pentatricopeptide repeat containing gene (LRPPRC). Most LSFC patients in Quebec are homozygous for an A354V substitution that causes a decrease in the expression of the LRPPRC protein. While LRPPRC is ubiquitously expressed and is involved in multiple cellular functions, tissue-specific expression of LRPPRC and COX activity is correlated with clinical features. In this proof-of-principle study, we developed human induced pluripotent stem cell (hiPSC)-based models from fibroblasts taken from a patient with LSFC, homozygous for the LRPPRC*354V allele, and from a control, homozygous for the LRPPRC*A354 allele. Specifically, for both of these fibroblast lines we generated hiPSC, hiPSC-derived cardiomyocytes (hiPSC-CMs) and hepatocyte-like cell (hiPSC-HLCs) lines, as well as the three germ layers. We observed that LRPPRC protein expression is reduced in all cell lines/layers derived from LSFC patient compared to control cells, with a reduction ranging from ∼70% in hiPSC-CMs to undetectable levels in hiPSC-HLC, reflecting tissue heterogeneity observed in patient tissues. We next performed exploratory analyses of these cell lines and observed that COX protein expression was reduced in all cell lines derived from LSFC patient compared to control cells. We also observed that mutant LRPPRC was associated with altered expression of key markers of endoplasmic reticulum stress response in hiPSC-HLCs but not in other cell types that were tested. While this demonstrates feasibility of the approach to experimentally study genotype-based differences that have tissue-specific impacts, this study will need to be extended to a larger number of patients and controls to not only validate the current observations but also to delve more deeply in the pathogenic mechanisms of LSFC.

2.
Biochim Biophys Acta Mol Basis Dis ; 1869(8): 166843, 2023 12.
Article in English | MEDLINE | ID: mdl-37558007

ABSTRACT

Very-long chain acyl-CoA dehydrogenase (VLCAD) catalyzes the initial step of mitochondrial long chain (LC) fatty acid ß-oxidation (FAO). Inherited VLCAD deficiency (VLCADD) predisposes to neonatal arrhythmias whose pathophysiology is still not understood. We hypothesized that VLCADD results in global disruption of cardiac complex lipid homeostasis, which may set conditions predisposing to arrhythmia. To test this, we assessed the cardiac lipidome and related molecular markers in seven-month-old VLCAD-/- mice, which mimic to some extent the human cardiac phenotype. Mice were sacrificed in the fed or fasted state after receiving for two weeks a chow or a high-fat diet (HFD), the latter condition being known to worsen symptoms in human VLCADD. Compared to their littermate counterparts, HFD/fasted VLCAD-/- mouse hearts displayed the following lipid alterations: (1) Lower LC, but higher VLC-acylcarnitines accumulation, (2) higher levels of arachidonic acid (AA) and lower docosahexaenoic acid (DHA) contents in glycerophospholipids (GPLs), as well as (3) corresponding changes in pro-arrhythmogenic AA-derived isoprostanes and thromboxane B2 (higher), and anti-arrythmogenic DHA-derived neuroprostanes (lower). These changes were associated with remodeling in the expression of gene or protein markers of (1) GPLs remodeling: higher calcium-dependent phospholipase A2 and lysophosphatidylcholine-acyltransferase 2, (2) calcium handling perturbations, and (3) endoplasmic reticulum stress. Altogether, these results highlight global lipid dyshomeostasis beyond FAO in VLCAD-/- mouse hearts, which may set conditions predisposing the hearts to calcium mishandling and endoplasmic reticulum stress and thereby may contribute to the pathogenesis of arrhythmias in VLCADD in mice as well as in humans.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain , Mitochondrial Diseases , Mice , Humans , Animals , Infant , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Calcium , Mitochondrial Diseases/metabolism , Fatty Acids/metabolism , Fatty Acids, Unsaturated , Arrhythmias, Cardiac
3.
ESC Heart Fail ; 7(3): 920-932, 2020 06.
Article in English | MEDLINE | ID: mdl-32154661

ABSTRACT

AIMS: The abundance of beta 3-adrenergic receptors (ß3-ARs) is upregulated in diseased human myocardium. We previously showed that cardiac-specific expression of ß3-AR inhibits the hypertrophic response to neurohormonal stimulation. Here, we further analysed signalling pathways involved in the anti-hypertrophic effect of ß3-AR. METHODS AND RESULTS: In vitro hypertrophic responses to phenylephrine (PE) were analysed in neonatal rat ventricular myocytes (NRVM) infected with a recombinant adenovirus expressing the human ß3-AR (AdVhß3). We confirmed results in mice with cardiomyocyte-specific moderate expression of human ß3-AR (ß3-TG) and wild-type (WT) littermates submitted to thoracic transverse aortic constriction (TAC) for 9 weeks. We observed a colocalization of ß3-AR with the AMP-activated protein kinase (AMPK) both in neonatal rat and in adult mouse cardiomyocytes. Treatment of NRVM with PE induced hypertrophy and a decrease in phosphorylation of Thr172-AMPK (/2, P = 0.0487) and phosphorylation of Ser79-acetyl-CoA carboxylase (ACC) (/2.6, P = 0.0317), inducing an increase in phosphorylated Ser235/236 S6 protein (×2.5, P = 0.0367) known to be involved in protein synthesis. These effects were reproduced by TAC in WT mice but restored to basal levels in ß3-AR expressing cells/mice. siRNA targeting of AMPK partly abrogated the anti-hypertrophic effect of ß3-AR in response to PE in NRVM. Concomitant with hypertrophy, autophagy was decreased by PE, as measured by microtubule-associated protein 1 light chain 3 (LC3)-II/LC3-I ratio (/2.6, P = 0.0010) and p62 abundance (×3, P = 0.0016) in NRVM or by TAC in WT mice (LC3-II/LC3-I ratio: /5.4, P = 0.0159), but preserved in human ß3-AR expressing cells and mice, together with reduced hypertrophy. CONCLUSIONS: Cardiac-specific moderate expression of ß3-AR inhibits the hypertrophic response in part through AMPK activation followed by inhibition of protein synthesis and preservation of autophagy. Activation of the cardiac ß3-AR pathway may provide future therapeutic avenues for the modulation of hypertrophic remodelling.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Animals , Hypertrophy , Mice , Myocardium , Myocytes, Cardiac , Rats
4.
Can J Cardiol ; 35(9): 1256.e1-1256.e2, 2019 09.
Article in English | MEDLINE | ID: mdl-31472821

ABSTRACT

We report a case of sudden unexplained death in a young asymptomatic woman in whom postmortem genetic testing after a negative autopsy identified a homozygous pathogenic mutation in SLC22A5 which leads clinically to primary carnitine deficiency (PCD). Her brother was subsequently diagnosed clinically with short QT syndrome, received an implantable defibrillator, and was then found to carry the same pathogenic homozygous mutation and critically low levels of carnitine. His QT interval improved with the use of carnitine supplementation, highlighting the close relationship between electrophysiology and biochemistry, and the importance of postmortem genetic testing in the clinical management of surviving relatives.


Subject(s)
Cardiomyopathies/genetics , Carnitine/deficiency , Death, Sudden, Cardiac/etiology , Genetic Testing/methods , Hyperammonemia/genetics , Long QT Syndrome/genetics , Muscular Diseases/genetics , Mutation , Solute Carrier Family 22 Member 5/genetics , Adult , Autopsy , Cardiomyopathies/complications , Cardiomyopathies/metabolism , Carnitine/genetics , Carnitine/metabolism , DNA/genetics , Fatal Outcome , Female , Genetic Predisposition to Disease , Humans , Hyperammonemia/complications , Hyperammonemia/metabolism , Long QT Syndrome/etiology , Muscular Diseases/complications , Muscular Diseases/metabolism , Solute Carrier Family 22 Member 5/metabolism
5.
Article in English | MEDLINE | ID: mdl-30271380

ABSTRACT

The AMP-activated protein kinase (AMPK) is an important cellular energy sensor. Its activation under energetic stress is known to activate energy-producing pathways and to inactivate energy-consuming pathways, promoting ATP preservation and cell survival. AMPK has been shown to play protective role in many pathophysiological processes including cardiovascular diseases, diabetes, and cancer. Its action is multi-faceted and comprises short-term regulation of enzymes by direct phosphorylation as well as long-term adaptation via control of transcription factors and cellular events such as autophagy. During the last decade, several studies underline the particular importance of the interaction between AMPK and the post-translational modification called O-GlcNAcylation. O-GlcNAcylation means the O-linked attachment of a single N-acetylglucosamine moiety on serine or threonine residues. O-GlcNAcylation plays a role in multiple physiological cellular processes but is also associated with the development of various diseases. The first goal of the present review is to present the tight molecular relationship between AMPK and enzymes regulating O-GlcNAcylation. We then draw the attention of the reader on the putative importance of this interaction in different pathophysiological events.

6.
Nat Commun ; 9(1): 374, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371602

ABSTRACT

AMP-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy. Here, we show that submaximal AMPK activation blocks cardiomyocyte hypertrophy without affecting downstream targets previously suggested to be involved, such as p70 ribosomal S6 protein kinase, calcineurin/nuclear factor of activated T cells (NFAT) and extracellular signal-regulated kinases. Instead, cardiomyocyte hypertrophy is accompanied by increased protein O-GlcNAcylation, which is reversed by AMPK activation. Decreasing O-GlcNAcylation by inhibitors of the glutamine:fructose-6-phosphate aminotransferase (GFAT), blocks cardiomyocyte hypertrophy, mimicking AMPK activation. Conversely, O-GlcNAcylation-inducing agents counteract the anti-hypertrophic effect of AMPK. In vivo, AMPK activation prevents myocardial hypertrophy and the concomitant rise of O-GlcNAcylation in wild-type but not in AMPKα2-deficient mice. Treatment of wild-type mice with O-GlcNAcylation-inducing agents reverses AMPK action. Finally, we demonstrate that AMPK inhibits O-GlcNAcylation by mainly controlling GFAT phosphorylation, thereby reducing O-GlcNAcylation of proteins such as troponin T. We conclude that AMPK activation prevents cardiac hypertrophy predominantly by inhibiting O-GlcNAcylation.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acetylglucosamine/metabolism , Cardiomegaly/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nitrogenous Group Transferases/genetics , AMP-Activated Protein Kinases/deficiency , Acetylglucosamine/pharmacology , Acylation/drug effects , Animals , Animals, Newborn , Azaserine/pharmacology , Azo Compounds/pharmacology , Biphenyl Compounds , Cardiomegaly/metabolism , Cardiomegaly/pathology , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Gene Expression Regulation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Glycosylation/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nitrogenous Group Transferases/antagonists & inhibitors , Nitrogenous Group Transferases/metabolism , Norleucine/analogs & derivatives , Norleucine/pharmacology , Phosphorylation/drug effects , Primary Cell Culture , Pyrones/pharmacology , Rats , Rats, Wistar , Signal Transduction , Thiophenes/pharmacology , Troponin T/genetics , Troponin T/metabolism
7.
Circ Cardiovasc Genet ; 10(5)2017 Oct.
Article in English | MEDLINE | ID: mdl-29021306

ABSTRACT

BACKGROUND: Long-QT syndrome is a potentially fatal condition for which 30% of patients are without a genetically confirmed diagnosis. Rapid identification of causal mutations is thus a priority to avoid at-risk situations that can lead to fatal cardiac events. Massively parallel sequencing technologies are useful for the identification of sequence variants; however, electrophysiological testing of newly identified variants is crucial to demonstrate causality. Long-QT syndrome could, therefore, benefit from having a standardized platform for functional characterization of candidate variants in the physiological context of human cardiomyocytes. METHODS AND RESULTS: Using a variant in Kir2.1 (Gly52Val) revealed by whole-exome sequencing in a patient presenting with symptoms of long-QT syndrome as a proof of principle, we demonstrated that commercially available human induced pluripotent stem cell-derived cardiomyocytes are a powerful model for screening variants involved in genetic cardiac diseases. Immunohistochemistry experiments and whole-cell current recordings in human embryonic kidney cells expressing the wild-type or the mutant Kir2.1 demonstrated that Kir2.1-52V alters channel cellular trafficking and fails to form a functional channel. Using human induced pluripotent stem cell-derived cardiomyocytes, we not only confirmed these results but also further demonstrated that Kir2.1-52V is associated with a dramatic prolongation of action potential duration with evidence of arrhythmic activity, parameters which could not have been studied using human embryonic kidney cells. CONCLUSIONS: Our study confirms the pathogenicity of Kir2.1-52V in 1 patient with long-QT syndrome and also supports the use of isogenic human induced pluripotent stem cell-derived cardiomyocytes as a physiologically relevant model for the screening of variants of unknown function.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome , Models, Biological , Mutation, Missense , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying , Adult , Amino Acid Substitution , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/pathology , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Long QT Syndrome/pathology , Myocytes, Cardiac/pathology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism
8.
Am J Physiol Heart Circ Physiol ; 313(2): H432-H445, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28646031

ABSTRACT

High plasma leucine levels strongly correlate with type 2 diabetes. Studies of muscle cells have suggested that leucine alters the insulin response for glucose transport by activating an insulin-negative feedback loop driven by the mammalian target of rapamycin/p70 ribosomal S6 kinase (mTOR/p70S6K) pathway. Here, we examined the molecular mechanism involved in leucine's action on cardiac glucose uptake. Leucine was indeed able to curb glucose uptake after insulin stimulation in both cultured cardiomyocytes and perfused hearts. Although leucine activated mTOR/p70S6K, the mTOR inhibitor rapamycin did not prevent leucine's inhibitory action on glucose uptake, ruling out the contribution of the insulin-negative feedback loop. α-Ketoisocaproate, the first metabolite of leucine catabolism, mimicked leucine's effect on glucose uptake. Incubation of cardiomyocytes with [13C]leucine ascertained its metabolism to ketone bodies (KBs), which had a similar negative impact on insulin-stimulated glucose transport. Both leucine and KBs reduced glucose uptake by affecting translocation of glucose transporter 4 (GLUT4) to the plasma membrane. Finally, we found that leucine elevated the global protein acetylation level. Pharmacological inhibition of lysine acetyltransferases counteracted this increase in protein acetylation and prevented leucine's inhibitory action on both glucose uptake and GLUT4 translocation. Taken together, these results indicate that leucine metabolism into KBs contributes to inhibition of cardiac glucose uptake by hampering the translocation of GLUT4-containing vesicles via acetylation. They offer new insights into the establishment of insulin resistance in the heart.NEW & NOTEWORTHY Catabolism of the branched-chain amino acid leucine into ketone bodies efficiently inhibits cardiac glucose uptake through decreased translocation of glucose transporter 4 to the plasma membrane. Leucine increases protein acetylation. Pharmacological inhibition of acetylation reverses leucine's action, suggesting acetylation involvement in this phenomenon.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/leucine-metabolism-inhibits-cardiac-glucose-uptake/.


Subject(s)
Energy Metabolism/drug effects , Glucose/metabolism , Keto Acids/pharmacology , Ketone Bodies/pharmacology , Leucine/pharmacology , Myocytes, Cardiac/drug effects , Acetylation , Animals , Biological Transport , Cells, Cultured , Dose-Response Relationship, Drug , Glucose Transporter Type 4/metabolism , Insulin Resistance , Isolated Heart Preparation , Keto Acids/metabolism , Ketone Bodies/metabolism , Leucine/metabolism , Male , Myocytes, Cardiac/metabolism , Protein Transport , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors
9.
EMBO Mol Med ; 8(12): 1390-1408, 2016 12.
Article in English | MEDLINE | ID: mdl-27861123

ABSTRACT

Genetic causes of many familial arrhythmia syndromes remain elusive. In this study, whole-exome sequencing (WES) was carried out on patients from three different families that presented with life-threatening arrhythmias and high risk of sudden cardiac death (SCD). Two French Canadian probands carried identical homozygous rare variant in TECRL gene (p.Arg196Gln), which encodes the trans-2,3-enoyl-CoA reductase-like protein. Both patients had cardiac arrest, stress-induced atrial and ventricular tachycardia, and QT prolongation on adrenergic stimulation. A third patient from a consanguineous Sudanese family diagnosed with catecholaminergic polymorphic ventricular tachycardia (CPVT) had a homozygous splice site mutation (c.331+1G>A) in TECRL Analysis of intracellular calcium ([Ca2+]i) dynamics in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) generated from this individual (TECRLHom-hiPSCs), his heterozygous but clinically asymptomatic father (TECRLHet-hiPSCs), and a healthy individual (CTRL-hiPSCs) from the same Sudanese family, revealed smaller [Ca2+]i transient amplitudes as well as elevated diastolic [Ca2+]i in TECRLHom-hiPSC-CMs compared with CTRL-hiPSC-CMs. The [Ca2+]i transient also rose markedly slower and contained lower sarcoplasmic reticulum (SR) calcium stores, evidenced by the decreased magnitude of caffeine-induced [Ca2+]i transients. In addition, the decay phase of the [Ca2+]i transient was slower in TECRLHom-hiPSC-CMs due to decreased SERCA and NCX activities. Furthermore, TECRLHom-hiPSC-CMs showed prolonged action potentials (APs) compared with CTRL-hiPSC-CMs. TECRL knockdown in control human embryonic stem cell-derived CMs (hESC-CMs) also resulted in significantly longer APs. Moreover, stimulation by noradrenaline (NA) significantly increased the propensity for triggered activity based on delayed afterdepolarizations (DADs) in TECRLHom-hiPSC-CMs and treatment with flecainide, a class Ic antiarrhythmic drug, significantly reduced the triggered activity in these cells. In summary, we report that mutations in TECRL are associated with inherited arrhythmias characterized by clinical features of both LQTS and CPVT Patient-specific hiPSC-CMs recapitulated salient features of the clinical phenotype and provide a platform for drug screening evidenced by initial identification of flecainide as a potential therapeutic. These findings have implications for diagnosis and treatment of inherited cardiac arrhythmias.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Genetic Predisposition to Disease , Mutation , Oxidoreductases/genetics , Adolescent , Adult , Cells, Cultured , Exome , Female , Genome, Human , Humans , Male , Sequence Analysis, DNA , Young Adult
10.
Biochim Biophys Acta ; 1862(12): 2232-2243, 2016 12.
Article in English | MEDLINE | ID: mdl-27544701

ABSTRACT

O-linked attachment of the monosaccharide ß-N-acetyl-glucosamine (O-GlcNAcylation) is a post-translational modification occurring on serine and threonine residues, which is evolving as an important mechanism for the regulation of various cellular processes. The present review will, first, provide a general background on the molecular regulation of protein O-GlcNAcylation and will summarize the role of this post-translational modification in various acute cardiac pathologies including ischemia-reperfusion. Then, we will focus on research studies examining protein O-GlcNAcylation in the context of cardiac hypertrophy. A particular emphasis will be laid on the convergent but also divergent actions of O-GlcNAcylation according to the type of hypertrophy investigated, including physiological, pressure overload-induced and diabetes-linked cardiac hypertrophy. In an attempt to distinguish whether O-GlcNAcylation is detrimental or beneficial, this review will present the different O-GlcNAcylated targets involved in hypertrophy development. We will finally argue on potential interest to target O-GlcNAc processes to treat cardiac hypertrophy. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.


Subject(s)
Cardiomegaly/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational , Acylation , Animals , Cardiomegaly/pathology , Humans , Myocardium/pathology
11.
Heart Rhythm ; 13(6): 1346-54, 2016 06.
Article in English | MEDLINE | ID: mdl-26921763

ABSTRACT

BACKGROUND: Ventricular arrhythmias and sudden cardiac deaths are among the leading causes of mortality in patients with heart failure, and the underlying mechanisms remain incompletely understood. Chronic elevation of angiotensin II (ANGII) is known to be one of the main contributors to heart failure. OBJECTIVE: We tested whether ANGII can alter ventricular conduction and Na(+) current using transgenic mice with cardiomyocyte-restricted overexpression of ANGII type 1 receptor (AT1R). METHODS: We used surface electrocardiograms along with current- and voltage-clamp techniques to characterize the electrophysiological properties of AT1R mice while the underlying regulatory mechanisms were explored using reverse transcription/quantitative polymerase chain reaction, Western blots, and immunofluorescence techniques. RESULTS: Electrophysiological data indicated that chronic AT1R activation in ventricular myocytes caused a 60% reduction in Na(+) current density that slowed the maximal velocity of the action potential upstroke, leading to a prolongation of the QRS complex. These changes occur independently of cardiac hypertrophy, suggesting a direct role for ANGII/AT1R in slowing ventricular conduction. Western blots demonstrated a selective increase in sarcolemmal protein kinase Cα (PKCα) in AT1R mice, indicating PKCα activation. Furthermore, immunofluorescence analysis showed reorganization of PKCα expression to sarcolemma and colocalization with NaV1.5 in AT1R myocytes. The involvement of PKCα in regulating Na(+) current was subsequently demonstrated in human-induced pluripotent stem cell-derived cardiomyocytes where ANGII treatment reduced Na(+) current density. Concomitant treatment with αV5-3, a PKCα translocation inhibitor peptide, blocked the ANGII effect. CONCLUSION: Overall, this study suggests that in mouse and human cardiomyocytes, PKCα is an important mediator of the ANGII-induced reduction in Na(+) current and may contribute to ventricular arrhythmias.


Subject(s)
Angiotensin II/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C-alpha/metabolism , Sodium Channels/physiology , Action Potentials , Animals , Heart Conduction System/physiopathology , Humans , Induced Pluripotent Stem Cells/physiology , Mice
12.
Methods Enzymol ; 561: 107-47, 2015.
Article in English | MEDLINE | ID: mdl-26358903

ABSTRACT

There has been a resurgence of interest for the field of cardiac metabolism catalyzed by evidence demonstrating a role of metabolic dysregulation in the pathogenesis of heart disease as well as the increased need for new therapeutic targets for patients with these diseases. In this regard, measuring substrate fluxes is critical in providing insight into the dynamics of cellular metabolism and in delineating the regulation of metabolite production and utilization. This chapter provides a comprehensive description of concepts, guidelines, and tips to assess metabolic fluxes relevant to energy substrate metabolism using (13)C-labeled substrates and (13)C-isotopomer analysis by gas chromatography-mass spectrometry (GC-MS), and the ex vivo working heart as study model. The focus will be on the mouse and on flux parameters, which are commonly assessed in the field, namely, those relevant to substrate selection for energy metabolism, specifically the relative contribution of carbohydrate (glucose, lactate, and pyruvate) and fatty acid oxidation to acetyl-CoA formation for citrate synthesis, glycolysis, as well as anaplerosis. We provide detailed procedures for the heart isolation and perfusion in the working mode as well as for sample processing for metabolite extraction and analysis by GC-MS and subsequent data processing for calculation of metabolic flux parameters. Finally, we address practical considerations and discuss additional applications and future challenges.


Subject(s)
Energy Metabolism , Gas Chromatography-Mass Spectrometry/methods , Isolated Heart Preparation , Isotope Labeling/methods , Myocardium/metabolism , Animals , Carbon Isotopes , Mice , Pyruvic Acid
13.
Am J Physiol Heart Circ Physiol ; 306(12): H1619-30, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24748590

ABSTRACT

AMP-activated protein kinase (AMPK), a key cellular sensor of energy, regulates metabolic homeostasis and plays a protective role in the ischemic or diabetic heart. Stimulation of cardiac glucose uptake contributes to this AMPK-mediated protection. The small-molecule AMPK activator A-769662, which binds and directly activates AMPK, has recently been characterized. A-769662-dependent AMPK activation protects the heart against an ischemia-reperfusion episode but is unable to stimulate skeletal muscle glucose uptake. Here, we tried to reconcile these conflicting findings by investigating the impact of A-769662 on cardiac AMPK signaling and glucose uptake. We showed that A-769662 promoted AMPK activation, resulting in the phosphorylation of several downstream targets, but was incapable of stimulating glucose uptake in cultured cardiomyocytes and the perfused heart. The lack of glucose uptake stimulation can be explained by A-769662's narrow specificity, since it selectively activates cardiac AMPK heterotrimeric complexes containing α2/ß1-subunits, the others being presumably required for this metabolic outcome. However, when combined with classical AMPK activators, such as metformin, phenformin, oligomycin, or hypoxia, which impact AMPK heterotrimers more broadly via elevation of cellular AMP levels, A-769662 induced more profound AMPK phosphorylation and subsequent glucose uptake stimulation. The synergistic effect of A-769662 under such ischemia-mimetic conditions protected cardiomyocytes against ROS production and cell death. In conclusion, despite the fact that A-769662 activates AMPK, it alone does not significantly stimulate glucose uptake. However, strikingly, its ability of potentiating the action on other AMPK activators makes it a potentially useful participant in the protective role of AMPK in the heart.


Subject(s)
AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pyrones/pharmacology , Thiophenes/pharmacology , Adenosine Monophosphate/metabolism , Animals , Biphenyl Compounds , Cells, Cultured , Insulin/pharmacology , Male , Models, Animal , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Phenformin/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
14.
Am J Physiol Heart Circ Physiol ; 306(1): H78-87, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24186097

ABSTRACT

In mice, genetic background is known to influence various parameters, including cardiac function. Its impact on cardiac energy substrate metabolism-a factor known to be closely related to function and contributes to disease development-is, however, unclear. This was examined in this study. In commonly used control mouse substrains SJL/JCrNTac, 129S6/SvEvTac, C57Bl/6J, and C57Bl/6NCrl, we assessed the functional and metabolic phenotypes of 3-mo-old working mouse hearts perfused ex vivo with physiological concentrations of (13)C-labeled carbohydrates (CHO) and a fatty acid (FA). Marked variations in various functional and metabolic flux parameters were observed among all mouse substrains, although the pattern observed differed for these parameters. For example, among all strains, C57Bl/6NCrl hearts had a greater cardiac output (+1.7-fold vs. SJL/JCrNTac and C57Bl/6J; P < 0.05), whereas at the metabolic level, 129S6/SvEvTac hearts stood out by displaying (vs. all 3 strains) a striking shift from exogenous FA (~-3.5-fold) to CHO oxidation as well as increased glycolysis (+1.7-fold) and FA incorporation into triglycerides (+2-fold). Correlation analyses revealed, however, specific linkages between 1) glycolysis, FA oxidation, and pyruvate metabolism and 2) cardiac work, oxygen consumption with heart rate, respectively. This implies that any genetically determined factors affecting a given metabolic flux parameter may impact on the associated functional parameters. Our results emphasize the importance of selecting the appropriate control strain for cardiac metabolic studies using transgenic mice, a factor that has often been neglected. Understanding the molecular mechanisms underlying the diversity of strain-specific cardiac metabolic and functional profiles, particularly the 129S6/SvEvTac, may ultimately disclose new specific metabolic targets for interventions in heart disease.


Subject(s)
Basal Metabolism/genetics , Cardiac Output/genetics , Heart/physiology , Mice, Inbred Strains/physiology , Myocardium/metabolism , Animals , Carbohydrate Metabolism , Fatty Acids/metabolism , Glycolysis , Lipid Peroxidation , Mice , Mice, Inbred Strains/genetics , Mice, Inbred Strains/metabolism , Oxygen Consumption , Pyruvic Acid/metabolism , Species Specificity , Triglycerides/metabolism
15.
J Mol Cell Cardiol ; 55: 92-100, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23201305

ABSTRACT

Glutamine, the most abundant amino acid in plasma, has attracted considerable interest for its cardioprotective properties. The primary effect of glutamine in the heart is commonly believed to be mediated via its anaplerotic metabolism to citric acid cycle (CAC) intermediates; however, there is little direct evidence to support this concept. Another potential candidate is the hexosamine biosynthetic pathway (HBP), which has recently been shown to modulate cardiomyocyte function and metabolism. Therefore, the goal of this study was to evaluate the contribution of anaplerosis and the HBP to the acute metabolic effects of glutamine in the heart. Normoxic ex vivo working rat hearts were perfused with (13)C-labeled substrates to assess relevant metabolic fluxes either with a physiological mixture of carbohydrates and a fatty acid (control) or under conditions of restricted pyruvate anaplerosis. Addition of a physiological concentration of glutamine (0.5mM) had no effect on contractile function of hearts perfused under the control condition, but improved that of hearts perfused under restricted pyruvate anaplerosis. Changes in CAC intermediate concentrations as well as (13)C-enrichment from [U-(13)C]glutamine did not support a major role of glutamine anaplerosis under any conditions. Under the control condition, however, glutamine significantly increased the contribution of exogenous oleate to ß-oxidation, 1.6-fold, and triglyceride formation, 2.8-fold. Glutamine had no effect on malonyl-CoA or AMP kinase activity levels; however, it resulted in a higher plasma membrane level of the fatty acid transporter CD36. These metabolic effects of glutamine were reversed by azaserine, which inhibits glucose entry into the HPB. Our results reveal a metabolic role of physiological concentration of glutamine in the healthy working heart beyond anaplerosis. This role appears to involve the HBP and regulation of fatty acid entry and metabolism via CD36. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".


Subject(s)
Glutamine/metabolism , Heart/physiology , Myocardium/metabolism , Animals , Biosynthetic Pathways , Energy Metabolism , Fatty Acids/metabolism , Glutamine/pharmacology , Heart/drug effects , Hexosamines/biosynthesis , In Vitro Techniques , Male , Oxidation-Reduction , Pyruvic Acid/metabolism , Rats
16.
Am J Physiol Heart Circ Physiol ; 301(3): H813-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21685264

ABSTRACT

Patients with very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency frequently present cardiomyopathy and heartbeat disorders. However, the underlying factors, which may be of cardiac or extra cardiac origins, remain to be elucidated. In this study, we tested for metabolic and functional alterations in the heart from 3- and 7-mo-old VLCAD null mice and their littermate counterparts, using validated experimental paradigms, namely, 1) ex vivo perfusion in working mode, with concomitant evaluation of myocardial contractility and metabolic fluxes using (13)C-labeled substrates under various conditions; as well as 2) in vivo targeted lipidomics, gene expression analysis as well as electrocardiogram monitoring by telemetry in mice fed various diets. Unexpectedly, when perfused ex vivo, working VLCAD null mouse hearts maintained values similar to those of the controls for functional parameters and for the contribution of exogenous palmitate to ß-oxidation (energy production), even at high palmitate concentration (1 mM) and increased energy demand (with 1 µM epinephrine) or after fasting. However, in vivo, these hearts displayed a prolonged rate-corrected QT (QTc) interval under all conditions examined, as well as the following lipid alterations: 1) age- and condition-dependent accumulation of triglycerides, and 2) 20% lower docosahexaenoic acid (an omega-3 polyunsaturated fatty acid) in membrane phospholipids. The latter was independent of liver but affected by feeding a diet enriched in saturated fat (exacerbated) or fish oil (attenuated). Our finding of a longer QTc interval in VLCAD null mice appears to be most relevant given that such condition increases the risk of sudden cardiac death.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Heart Conduction System/physiopathology , Lipid Metabolism/genetics , Long QT Syndrome/enzymology , Metabolism, Inborn Errors/enzymology , Mitochondrial Diseases/enzymology , Muscular Diseases/enzymology , Myocardium/enzymology , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Age Factors , Aging , Analysis of Variance , Animals , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Electrocardiography, Ambulatory , Fish Oils/administration & dosage , Fish Oils/metabolism , Lipid Metabolism, Inborn Errors , Liver/metabolism , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Long QT Syndrome/prevention & control , Male , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Muscular Diseases/complications , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Myocardial Contraction , Oxidation-Reduction , Palmitic Acid/metabolism , Perfusion , Telemetry , Triglycerides/metabolism
17.
Am J Physiol Heart Circ Physiol ; 300(3): H845-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21257916

ABSTRACT

Heart rate reduction (HRR) is an important target in the management of patients with chronic stable angina. Most available drugs for HRR, such as ß-blockers, have adverse effects, including on cardiac energy substrate metabolism, a well-recognized determinant of cardiac homeostasis. This study aimed at 1) testing whether HRR by ivabradine (IVA) alters substrate metabolism in the healthy normoxic working heart and 2) comparing the effect of IVA with that of the ß-blocker metoprolol (METO). This was assessed using our well-established model of ex vivo mouse heart perfusion in the working mode, which enables concomitant evaluation of myocardial contractility and metabolic fluxes using (13)C-labeled substrates. Hearts were perfused in the absence (controls; n = 10) or presence of IVA (n = 10, 3 µM) with or without atrial pacing to abolish HRR in the IVA group. IVA significantly reduced HR (35 ± 5%) and increased stroke volume (39 ± 9%) while maintaining similar cardiac output, contractility, power, and efficiency. Effects of IVA on HR and stroke volume were reversed by atrial pacing. At the metabolic level, IVA did not impact on substrate selection to citrate formation, rates of glycolysis, or tissue levels of high-energy phosphates. In contrast, METO, at concentrations up to 40 µM, decreased markedly cardiac function (flow: 25 ± 6%; stroke volume: 30 ± 10%; contractility: 31 ± 9%) as well as glycolysis (2.9-fold) but marginally affected HR. Collectively, these results demonstrate that IVA selectively reduces HR while preserving energy substrate metabolism of normoxic healthy working mouse hearts perfused ex vivo, a model that mimics to some extent the denervated transplanted heart. Our results provide the impetus for testing selective HRR by IVA on cardiac substrate metabolism in pathological models.


Subject(s)
Benzazepines/pharmacology , Heart Rate/drug effects , Heart/drug effects , Myocardium/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Cardiac Output/drug effects , Energy Metabolism/drug effects , Ivabradine , Male , Metoprolol/pharmacology , Mice , Mice, Inbred C57BL , Oxygen Consumption/drug effects
18.
Am J Physiol Endocrinol Metab ; 298(5): E1049-57, 2010 May.
Article in English | MEDLINE | ID: mdl-20159856

ABSTRACT

The purpose of this study was to determine in vivo myocardial energy metabolism and function in a nutritional model of type 2 diabetes. Wistar rats rendered insulin-resistant and mildly hyperglycemic, hyperinsulinemic, and hypertriglyceridemic with a high-fructose/high-fat diet over a 6-wk period with injection of a small dose of streptozotocin (HFHFS) and control rats were studied using micro-PET (microPET) without or with a euglycemic hyperinsulinemic clamp. During glucose clamp, myocardial metabolic rate of glucose measured with [(18)F]fluorodeoxyglucose ([(18)F]FDG) was reduced by approximately 81% (P < 0.05), whereas myocardial plasma nonesterified fatty acid (NEFA) uptake as determined by [(18)F]fluorothia-6-heptadecanoic acid ([(18)F]FTHA) was not significantly changed in HFHFS vs. control rats. Myocardial oxidative metabolism as assessed by [(11)C]acetate and myocardial perfusion index as assessed by [(13)N]ammonia were similar in both groups, whereas left ventricular ejection fraction as assessed by microPET was reduced by 26% in HFHFS rats (P < 0.05). Without glucose clamp, NEFA uptake was approximately 40% lower in HFHFS rats (P < 0.05). However, myocardial uptake of [(18)F]FTHA administered by gastric gavage was significantly higher in HFHFS rats (P < 0.05). These abnormalities were associated with reduced Glut4 mRNA expression and increased Cd36 mRNA expression and mitochondrial carnitine palmitoyltransferase 1 activity (P < 0.05). HFHFS rats display type 2 diabetes complicated by left ventricular contractile dysfunction with profound reduction in myocardial glucose utilization, activation of fatty acid metabolic pathways, and preserved myocardial oxidative metabolism, suggesting reduced myocardial metabolic efficiency. In this model, increased myocardial fatty acid exposure likely occurs from circulating triglyceride, but not from circulating plasma NEFA.


Subject(s)
Cardiomyopathies/metabolism , Diabetes Mellitus, Experimental/metabolism , Energy Metabolism/physiology , Glucose/metabolism , Myocardium/metabolism , Analysis of Variance , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/diagnostic imaging , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Nonesterified/blood , Glucose Clamp Technique , Heart/diagnostic imaging , Heart Failure/diagnostic imaging , Heart Failure/etiology , Heart Failure/metabolism , Insulin/blood , Male , Radionuclide Imaging , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
19.
J Mol Cell Cardiol ; 45(2): 230-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18590915

ABSTRACT

While the balance between carbohydrates and fatty acids for energy production appears to be crucial for cardiac homeostasis, much remains to be learned about the molecular mechanisms underlying this relationship. Given the reported benefits of cGMP signaling on the myocardium, we investigated the impact of its chronic activation on cardiac energy metabolism using mice overexpressing a constitutively active cytoplasmic guanylate cyclase (GC(+/0)) in cardiomyocytes. Ex vivo working GC(+/0) heart perfusions with (13)C-labeled substrates revealed an altered pattern of exogenous substrate fuel selection compared to controls, namely a 38+/-9% lower contribution of exogenous fatty acids to acetyl-CoA formation, while that of carbohydrates remains unchanged despite a two-fold increase in glycolysis. The lower contribution of exogenous fatty acids to energy production is not associated with changes in energy demand or supply (contractile function, oxygen consumption, tissue acetyl-CoA or CoA levels, citric acid cycle flux rate) or in the regulation of beta-oxidation (acetyl-CoA carboxylase activity, tissue malonyl-CoA levels). However, GC(+/0) hearts show a two-fold increase in the incorporation of exogenous oleate into triglycerides. Furthermore, the following molecular data are consistent with a concomitant increase in triglyceride hydrolysis: (i) increased abundance of hormone sensitive lipase (HSL) protein (24+/-11%) and mRNA (22+/-4%) as well as (ii) several phosphorylation events related to HSL inhibitory (AMPK) and activation (ERK 1/2) sites, which should contribute to enhance its activity. These changes in exogenous fatty acid trafficking in GC(+/0) hearts appear to be functionally relevant, as demonstrated by their resistance to fasting-induced triglyceride accumulation. While the documented metabolic profile of GC(+/0) mouse hearts is partly reminiscent of hypertrophied hearts, the observed changes in lipid trafficking have not been previously documented, and may be part of the molecular mechanism underlying the benefits of cGMP signaling on the myocardium.


Subject(s)
Cyclic GMP/physiology , Fatty Acids/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Triglycerides/antagonists & inhibitors , Triglycerides/metabolism , Acetyl Coenzyme A/metabolism , Animals , Biological Transport, Active/physiology , Glycolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/physiology , Oleic Acid/metabolism
20.
Cardiovasc Drugs Ther ; 22(2): 97-106, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18253821

ABSTRACT

INTRODUCTION: Medium-chain fatty acids (MCFAs) have physical and metabolic properties that are distinct from those of long-chain fatty acids, which make them a readily available cellular energy source. These properties have been used advantageously in the clinics for more than 50 years for treating lipid absorption disorders, undernourished patients, and more recently subjects with long-chain fatty acid oxidation defects. In these latter subjects, nutritional interventions with MCFA-containing triglycerides have been shown to improve clinical symptoms, particularly cardiomyopathies. POTENTIAL BENEFITS OF MCFA METABOLISM IN CARDIAC DISEASES: There is, however, only a limited number of studies that have considered the potential use of MCFAs as metabolic therapy for cardiac diseases in general. Nevertheless, current experimental evidence does support the notion that the diseased heart is energy deficient and that alterations in myocardial energy substrate metabolism contribute to contractile dysfunction and cardiac disease development and progression. Hence, this article will review current literature on MCFAs with a specific emphasis on their metabolism and potential benefits for the heart. It will include practical considerations about the potential clinical application of MCFA therapy for the management of patients with cardiac diseases.


Subject(s)
Fatty Acids/metabolism , Fatty Acids/therapeutic use , Heart Diseases/drug therapy , Animals , Heart/drug effects , Heart Diseases/metabolism , Humans , Models, Biological , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...