Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
J Cardiol Cases ; 28(3): 105-108, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37671260

ABSTRACT

Cardiac amyloidosis is a restrictive cardiomyopathy for which diuretics are frequently used, but vasodilators have classically been relatively contraindicated due to side effects of hypotension. In the setting of decompensated heart failure, this may not be the case. We report a man with advanced cardiac amyloidosis who presented to the hospital with decompensated heart failure, in part, due to elevated systemic vascular resistance. Through the use of invasive hemodynamic testing, we were able to demonstrate an increase in cardiac output in response to a nitroprusside challenge. In turn, the patient had an improvement in his symptoms and was sent home on afterload reducing medications. This discerns a subpopulation of cardiac amyloidosis patients in decompensated heart failure who benefit from medications that reduce systemic vascular resistance, and can benefit from hemodynamic testing, especially when diuretics fail to control symptoms. Learning objective: Medications that cause peripheral vasodilation are standard therapy for patients with reduced ejection fraction, however, they are seldom used for patients with cardiac amyloidosis due to adverse effects. In some cases, there may be value in using hemodynamic measurements in patients with advanced cardiac amyloidosis to guide management as some patients may have hemodynamics that resemble those of systolic heart failure. This may offer a novel approach to symptomatic treatment of advanced cardiac amyloidosis.

2.
Biomed Pharmacother ; 160: 114310, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36731341

ABSTRACT

BACKGROUND: Elevated myocardial intracellular sodium ([Na+]i) was shown to decrease mitochondrial calcium ([Ca2+]MITO) via mitochondrial sodium/calcium exchanger (NCXMITO), resulting in decreased mitochondrial ATP synthesis. The sodium-glucose co-transporter 2 inhibitor (SGLT2i) ertugliflozin (ERTU) improved energetic deficit and contractile dysfunction in a mouse model of high fat, high sucrose (HFHS) diet-induced diabetic cardiomyopathy (DCMP). As SGLT2is were shown to lower [Na+]i in isolated cardiomyocytes, we hypothesized that energetic improvement in DCMP is at least partially mediated by a decrease in abnormally elevated myocardial [Na+]i. METHODS: Forty-two eight-week-old male C57BL/6J mice were fed a control or HFHS diet for six months. In the last month, a subgroup of HFHS-fed mice was treated with ERTU. At the end of the study, left ventricular contractile function and energetics were measured simultaneously in isolated beating hearts by 31P NMR (Nuclear Magnetic Resonance) spectroscopy. A subset of untreated HFHS hearts was perfused with vehicle vs. CGP 37157, an NCXMITO inhibitor. Myocardial [Na+]i was measured by 23Na NMR spectroscopy. RESULTS: HFHS hearts showed diastolic dysfunction, decreased contractile reserve, and impaired energetics as reflected by decreased phosphocreatine (PCr) and PCr/ATP ratio. Myocardial [Na+]i was elevated > 2-fold in HFHS (vs. control diet). ERTU reversed the impairments in HFHS hearts to levels similar to or better than control diet and decreased myocardial [Na+]i to control levels. CGP 37157 normalized the PCr/ATP ratio in HFHS hearts. CONCLUSIONS: Elevated myocardial [Na+]i contributes to mitochondrial and contractile dysfunction in DCMP. Targeting myocardial [Na+]i and/or NCXMITO may be an effective strategy in DCMP and other forms of heart disease associated with elevated myocardial [Na+]i.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Sodium-Glucose Transporter 2 Inhibitors , Mice , Male , Animals , Diabetic Cardiomyopathies/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium , Calcium , Deoxycytidine Monophosphate , Myocardial Contraction , Mice, Inbred C57BL , Myocardium , Adenosine Triphosphate
3.
J Am Heart Assoc ; 10(13): e019995, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34169737

ABSTRACT

Background Inhibitors of the sodium-glucose linked transporter 2 improve cardiovascular outcomes in patients with or without type 2 diabetes mellitus, but the effects on cardiac energetics and mitochondrial function are unknown. We assessed the effects of sodium-glucose linked transporter 2 inhibition on mitochondrial function, high-energy phosphates, and genes encoding mitochondrial proteins in hearts of mice with and without diet-induced diabetic cardiomyopathy. Methods and Results Mice fed a control diet or a high-fat, high-sucrose diet received ertugliflozin mixed with the diet (0.5 mg/g of diet) for 4 months. Isolated mitochondria were assessed for functional capacity. High-energy phosphates were assessed by 31P nuclear magnetic resonance spectroscopy concurrently with contractile performance in isolated beating hearts. The high-fat, high-sucrose diet caused myocardial hypertrophy, diastolic dysfunction, mitochondrial dysfunction, and impaired energetic response, all of which were prevented by ertugliflozin. With both diets, ertugliflozin caused supernormalization of contractile reserve, as measured by rate×pressure product at high work demand. Likewise, the myocardial gene sets most enriched by ertugliflozin were for oxidative phosphorylation and fatty acid metabolism, both of which were enriched independent of diet. Conclusions Ertugliflozin not only prevented high-fat, high-sucrose-induced pathological cardiac remodeling, but improved contractile reserve and induced the expression of oxidative phosphorylation and fatty acid metabolism gene sets independent of diabetic status. These effects of sodium-glucose linked transporter 2 inhibition on cardiac energetics and metabolism may contribute to improved structure and function in cardiac diseases associated with mitochondrial dysfunction, such as heart failure.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Energy Metabolism/drug effects , Hypertrophy, Left Ventricular/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Ventricular Dysfunction, Left/prevention & control , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Diet, High-Fat , Dietary Sucrose , Energy Metabolism/genetics , Gene Expression Regulation , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
4.
JACC Basic Transl Sci ; 5(9): 916-927, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33015414

ABSTRACT

Mice with obesity and metabolic heart disease (MHD) due to a high-fat, high-sucrose diet were treated with placebo, a clinically relevant dose of sacubitril (SAC)/valsartan (VAL), or an equivalent dose of VAL for 4 months. There were striking differences between SAC/VAL and VAL with regard to: 1) diastolic dysfunction; 2) interstitial fibrosis; and to a lesser degree; 3) oxidative stress-all of which were more favorably affected by SAC/VAL. SAC/VAL and VAL similarly attenuated myocardial hypertrophy and improved myocardial energetics. In mice with obesity-related MHD, neprilysin inhibition exerts favorable effects on diastolic function.

5.
Circulation ; 142(25): 2459-2469, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33076678

ABSTRACT

BACKGROUND: SERCA [sarco(endo)plasmic reticulum calcium ATPase] is regulated by oxidative posttranslational modifications at cysteine 674 (C674). Because sarcoplasmic reticulum (SR) calcium has been shown to play a critical role in mediating mitochondrial dysfunction in response to reactive oxygen species, we hypothesized that SERCA oxidation at C674 would modulate the effects of reactive oxygen species on mitochondrial calcium and mitochondria-dependent apoptosis in cardiac myocytes. METHODS: Adult rat ventricular myocytes expressing wild-type SERCA2b or a redox-insensitive mutant in which C674 is replaced by serine (C674S) were exposed to H2O2 (100 µmol/Lµ). Free mitochondrial calcium concentration was measured in adult rat ventricular myocytes with a genetically targeted fluorescent probe, and SR calcium content was assessed by measuring caffeine-stimulated release. Mice with heterozygous knock-in of the SERCA C674S mutation were subjected to chronic ascending aortic constriction. RESULTS: In adult rat ventricular myocytes expressing wild-type SERCA, H2O2 caused a 25% increase in mitochondrial calcium concentration that was associated with a 50% decrease in SR calcium content, both of which were prevented by the ryanodine receptor inhibitor tetracaine. In cells expressing the C674S mutant, basal SR calcium content was decreased by 31% and the H2O2-stimulated rise in mitochondrial calcium concentration was attenuated by 40%. In wild-type cells, H2O2 caused cytochrome c release and apoptosis, both of which were prevented in C674S-expressing cells. In myocytes from SERCA knock-in mice, basal SERCA activity and SR calcium content were decreased. To test the effect of C674 oxidation on apoptosis in vivo, SERCA knock-in mice were subjected to chronic ascending aortic constriction. In wild-type mice, ascending aortic constriction caused myocyte apoptosis, LV dilation, and systolic failure, all of which were inhibited in SERCA knock-in mice. CONCLUSIONS: Redox activation of SERCA C674 regulates basal SR calcium content, thereby mediating the pathologic reactive oxygen species-stimulated rise in mitochondrial calcium required for myocyte apoptosis and myocardial failure.


Subject(s)
Apoptosis , Calcium/metabolism , Heart Failure/enzymology , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Apoptosis/drug effects , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Hydrogen Peroxide/toxicity , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria, Heart/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidants/toxicity , Oxidation-Reduction , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Ventricular Function, Left , Ventricular Remodeling
6.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32948023

ABSTRACT

Cardiovascular diseases are the leading cause of death worldwide, and as rates continue to increase, discovering mechanisms and therapeutic targets become increasingly important. An underlying cause of most cardiovascular diseases is believed to be excess reactive oxygen or nitrogen species. Glutathione, the most abundant cellular antioxidant, plays an important role in the body's reaction to oxidative stress by forming reversible disulfide bridges with a variety of proteins, termed glutathionylation (GSylation). GSylation can alter the activity, function, and structure of proteins, making it a major regulator of cellular processes. Glutathione-protein mixed disulfide bonds are regulated by glutaredoxins (Glrxs), thioltransferase members of the thioredoxin family. Glrxs reduce GSylated proteins and make them available for another redox signaling cycle. Glrxs and GSylation play an important role in cardiovascular diseases, such as myocardial ischemia and reperfusion, cardiac hypertrophy, peripheral arterial disease, and atherosclerosis. This review primarily concerns the role of GSylation and Glrxs, particularly glutaredoxin-1 (Glrx), in cardiovascular diseases and the potential of Glrx as therapeutic agents.


Subject(s)
Cardiovascular Diseases/metabolism , Glutaredoxins/physiology , Glutathione/metabolism , Protein Processing, Post-Translational , Animals , Antioxidants/metabolism , Cardiovascular Diseases/drug therapy , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine/metabolism , Disulfides/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Glutaredoxins/deficiency , Glutaredoxins/therapeutic use , Homeostasis , Humans , Lipid Metabolism/physiology , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
7.
NMR Biomed ; 33(5): e4258, 2020 05.
Article in English | MEDLINE | ID: mdl-32066202

ABSTRACT

Metabolic heart disease (MHD), which is strongly associated with heart failure with preserved ejection fraction, is characterized by reduced mitochondrial energy production and contractile performance. In this study, we tested the hypothesis that an acute increase in ATP synthesis, via short chain fatty acid (butyrate) perfusion, restores contractile function in MHD. Isolated hearts of mice with MHD due to consumption of a high fat high sucrose (HFHS) diet or on a control diet (CD) for 4 months were studied using 31 P NMR spectroscopy to measure high energy phosphates and ATP synthesis rates during increased work demand. At baseline, HFHS hearts had increased ADP and decreased free energy of ATP hydrolysis (ΔG~ATP ), although contractile function was similar between the two groups. At high work demand, the ATP synthesis rate in HFHS hearts was reduced by over 50%. Unlike CD hearts, HFHS hearts did not increase contractile function at high work demand, indicating a lack of contractile reserve. However, acutely supplementing HFHS hearts with 4mM butyrate normalized ATP synthesis, ADP, ΔG~ATP and contractile reserve. Thus, acute reversal of depressed mitochondrial ATP production improves contractile dysfunction in MHD. These findings suggest that energy starvation may be a reversible cause of myocardial dysfunction in MHD, and opens new therapeutic opportunities.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Butyrates/pharmacology , Cardiovascular Diseases/metabolism , Metabolic Diseases/metabolism , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , Animals , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/physiopathology , Energy Metabolism/drug effects , Hemodynamics/drug effects , Hydrolysis , Magnetic Resonance Spectroscopy , Male , Metabolic Diseases/diagnostic imaging , Metabolic Diseases/physiopathology , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Thermodynamics
8.
Int J Biochem Cell Biol ; 114: 105569, 2019 09.
Article in English | MEDLINE | ID: mdl-31299273

ABSTRACT

Calcium (Ca2+), an important second messenger, regulates many cellular activities and varies spatiotemporally within the cell. Conventional methods to monitor Ca2+ changes, such as synthetic Ca2+ indicators, are not targetable, while genetically encoded Ca2+ indicators (GECI) can be precisely directed to cellular compartments. GECIs are chimeric proteins composed of calmodulin (or other proteins that change conformation on Ca2+ binding) coupled with two fluorescent proteins that come closer together after an increase in [Ca2+], and enhance Förster resonance energy transfer (FRET) that allows for ratiometric [Ca2+] assessment. Here, adult rat ventricular myocytes were transfected with specifically targeted calmodulin-based GECIs and Ca2+ responses to a physiological stimulus, norepinephrine (NE, 10 µM), were observed in a) sarcoplasmic reticulum (SR), b) mitochondria, c) the space between the mitochondria and SR, termed the Mitochondria Associated Membrane space (MAM) and d) cytosol for 10 min after stimulation. In SR and mitochondria, NE increased the [Ca2+] ratio by 17% and by 8%, respectively. In the MAM the [Ca2+] ratio decreased by 16%, while in cytosol [Ca2+] remained unchanged. In conclusion, adrenergic stimulation causes distinct responses in the cardiomyocyte SR, mitochondria and MAM. Additionally, our work provides a toolkit-update for targeted [Ca2+] measurements in multiple cellular compartments.


Subject(s)
Calcium Signaling , Calcium/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Myocytes, Cardiac/metabolism , Receptors, Adrenergic/metabolism , Animals , Male , Myocytes, Cardiac/cytology , Rats , Rats, Sprague-Dawley
9.
Antioxid Redox Signal ; 31(7): 539-549, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31088291

ABSTRACT

Aims: Metabolic syndrome is associated with metabolic heart disease (MHD) that is characterized by left ventricular (LV) hypertrophy, interstitial fibrosis, contractile dysfunction, and mitochondrial dysfunction. Overexpression of catalase in mitochondria (transgenic expression of catalase targeted to the mitochondria [mCAT]) prevents the structural and functional features of MHD caused by a high-fat, high-sucrose (HFHS) diet for ≥4 months. However, it is unclear whether the effect of mCAT is due to prevention of reactive oxygen species (ROS)-mediated cardiac remodeling, a direct effect on mitochondrial function, or both. To address this question, we measured myocardial function and energetics in mice, with or without mCAT, after 1 month of HFHS, before the development of cardiac structural remodeling. Results: HFHS diet for 1 month had no effect on body weight, heart weight, LV structure, myocyte size, or interstitial fibrosis. Isolated cardiac mitochondria from HFHS-fed mice produced 2.2- to 3.8-fold more H2O2, and 16%-29% less adenosine triphosphate (ATP). In isolated beating hearts from HFHS-fed mice, [phosphocreatine (PCr)] and the free energy available for ATP hydrolysis (ΔG∼ATP) were decreased, and they failed to increase with work demands. Overexpression of mCAT normalized ROS and ATP production in isolated mitochondria, and it corrected myocardial [PCr] and ΔG∼ATP in the beating heart. Innovation: This is the first demonstration that in MHD, mitochondrial ROS mediate energetic dysfunction that is sufficient to impair contractile function. Conclusion: ROS produced and acting in the mitochondria impair myocardial energetics, leading to slowed relaxation and decreased contractile reserve. These effects precede structural remodeling and are corrected by mCAT, indicating that ROS-mediated energetic impairment, per se, is sufficient to cause contractile dysfunction in MHD.


Subject(s)
Energy Metabolism , Heart Diseases/metabolism , Metabolic Diseases/metabolism , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Disease Susceptibility , Echocardiography , Fibrosis , Heart Diseases/diagnostic imaging , Heart Diseases/etiology , Heart Diseases/pathology , Hydrogen Peroxide/metabolism , Immunohistochemistry , Metabolic Diseases/etiology , Metabolic Diseases/pathology , Mice , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology
10.
J Mol Cell Cardiol ; 116: 106-114, 2018 03.
Article in English | MEDLINE | ID: mdl-29409987

ABSTRACT

Metabolic syndrome is a cluster of obesity-related metabolic abnormalities that lead to metabolic heart disease (MHD) with left ventricular pump dysfunction. Although MHD is thought to be associated with myocardial energetic deficiency, two key questions have not been answered. First, it is not known whether there is a sufficient energy deficit to contribute to pump dysfunction. Second, the basis for the energy deficit is not clear. To address these questions, mice were fed a high fat, high sucrose (HFHS) 'Western' diet to recapitulate the MHD phenotype. In isolated beating hearts, we used 31P NMR spectroscopy with magnetization transfer to determine a) the concentrations of high energy phosphates ([ATP], [ADP], [PCr]), b) the free energy of ATP hydrolysis (∆G~ATP), c) the rate of ATP production and d) flux through the creatine kinase (CK) reaction. At the lowest workload, the diastolic pressure-volume relationship was shifted upward in HFHS hearts, indicative of diastolic dysfunction, whereas systolic function was preserved. At this workload, the rate of ATP synthesis was decreased in HFHS hearts, and was associated with decreases in both [PCr] and ∆G~ATP. Higher work demands unmasked the inability of HFHS hearts to increase systolic function and led to a further decrease in ∆G~ATP to a level that is not sufficient to maintain normal function of sarcoplasmic Ca2+-ATPase (SERCA). While [ATP] was preserved at all work demands in HFHS hearts, the progressive increase in [ADP] led to a decrease in ∆G~ATP with increased work demands. Surprisingly, CK flux, CK activity and total creatine were normal in HFHS hearts. These findings differ from dilated cardiomyopathy, in which the energetic deficiency is associated with decreases in CK flux, CK activity and total creatine. Thus, in HFHS-fed mice with MHD there is a distinct metabolic phenotype of the heart characterized by a decrease in ATP production that leads to a functionally-important energetic deficiency and an elevation of [ADP], with preservation of CK flux.


Subject(s)
Adenosine Triphosphate/metabolism , Heart Diseases/metabolism , Heart Diseases/physiopathology , Myocardial Contraction , Animals , Body Weight , Creatine Kinase/metabolism , Diastole , Diet, High-Fat , Dietary Sucrose , Energy Metabolism , Hydrolysis , Magnetic Resonance Spectroscopy , Male , Mice, Inbred C57BL , Organ Size , Perfusion
11.
J Am Heart Assoc ; 5(1)2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26755553

ABSTRACT

BACKGROUND: Mitochondrial reactive oxygen species (ROS) are associated with metabolic heart disease (MHD). However, the mechanism by which ROS cause MHD is unknown. We tested the hypothesis that mitochondrial ROS are a key mediator of MHD. METHODS AND RESULTS: Mice fed a high-fat high-sucrose (HFHS) diet develop MHD with cardiac diastolic and mitochondrial dysfunction that is associated with oxidative posttranslational modifications of cardiac mitochondrial proteins. Transgenic mice that express catalase in mitochondria and wild-type mice were fed an HFHS or control diet for 4 months. Cardiac mitochondria from HFHS-fed wild-type mice had a 3-fold greater rate of H2O2 production (P=0.001 versus control diet fed), a 30% decrease in complex II substrate-driven oxygen consumption (P=0.006), 21% to 23% decreases in complex I and II substrate-driven ATP synthesis (P=0.01), and a 62% decrease in complex II activity (P=0.002). In transgenic mice that express catalase in mitochondria, all HFHS diet-induced mitochondrial abnormalities were ameliorated, as were left ventricular hypertrophy and diastolic dysfunction. In HFHS-fed wild-type mice complex II substrate-driven ATP synthesis and activity were restored ex vivo by dithiothreitol (5 mmol/L), suggesting a role for reversible cysteine oxidative posttranslational modifications. In vitro site-directed mutation of complex II subunit B Cys100 or Cys103 to redox-insensitive serines prevented complex II dysfunction induced by ROS or high glucose/high palmitate in the medium. CONCLUSION: Mitochondrial ROS are pathogenic in MHD and contribute to mitochondrial dysfunction, at least in part, by causing oxidative posttranslational modifications of complex I and II proteins including reversible oxidative posttranslational modifications of complex II subunit B Cys100 and Cys103.


Subject(s)
Diet, High-Fat , Dietary Sucrose , Hypertrophy, Left Ventricular/etiology , Mitochondria, Heart/metabolism , Mitochondrial Diseases/etiology , Oxidative Stress , Reactive Oxygen Species/metabolism , Ventricular Dysfunction, Left/etiology , Adenosine Triphosphate/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Electron Transport Complex I/metabolism , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Energy Metabolism , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Mitochondrial Diseases/prevention & control , Mutation , Oxidation-Reduction , Protein Processing, Post-Translational , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left
12.
J Mol Cell Cardiol ; 78: 165-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25109264

ABSTRACT

BACKGROUND: Diet-induced obesity leads to metabolic heart disease (MHD) characterized by increased oxidative stress that may cause oxidative post-translational modifications (OPTM) of cardiac mitochondrial proteins. The functional consequences of OPTM of cardiac mitochondrial proteins in MHD are unknown. Our objective was to determine whether cardiac mitochondrial dysfunction in MHD due to diet-induced obesity is associated with cysteine OPTM. METHODS AND RESULTS: Male C57BL/6J mice were fed either a high-fat, high-sucrose (HFHS) or control diet for 8months. Cardiac mitochondria from HFHS-fed mice (vs. control diet) had an increased rate of H2O2 production, a decreased GSH/GSSG ratio, a decreased rate of complex II substrate-driven ATP synthesis and decreased complex II activity. Complex II substrate-driven ATP synthesis and complex II activity were partially restored ex-vivo by reducing conditions. A biotin switch assay showed that HFHS feeding increased cysteine OPTM in complex II subunits A (SDHA) and B (SDHB). Using iodo-TMT multiplex tags we found that HFHS feeding is associated with reversible oxidation of cysteines 89 and 231 in SDHA, and 100, 103 and 115 in SDHB. CONCLUSIONS: MHD due to consumption of a HFHS "Western" diet causes increased H2O2 production and oxidative stress in cardiac mitochondria associated with decreased ATP synthesis and decreased complex II activity. Impaired complex II activity and ATP production are associated with reversible cysteine OPTM of complex II. Possible sites of reversible cysteine OPTM in SDHA and SDHB were identified by iodo-TMT tag labeling. Mitochondrial ROS may contribute to the pathophysiology of MHD by impairing the function of complex II. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".


Subject(s)
Diet, High-Fat/adverse effects , Electron Transport Complex II/metabolism , Mitochondria, Heart/metabolism , Protein Processing, Post-Translational , Adenosine Triphosphate/metabolism , Animals , Enzyme Activation , Glutathione/metabolism , Hydrogen Peroxide , Male , Mice , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
14.
Am J Physiol Heart Circ Physiol ; 306(10): H1453-63, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24633550

ABSTRACT

Oxidative stress in the myocardium plays an important role in the pathophysiology of hemodynamic overload. The mechanism by which reactive oxygen species (ROS) in the cardiac myocyte mediate myocardial failure in hemodynamic overload is not known. Accordingly, our goals were to test whether myocyte-specific overexpression of peroxisomal catalase (pCAT) that localizes in the sarcoplasm protects mice from hemodynamic overload-induced failure and prevents oxidation and inhibition of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA), an important sarcoplasmic protein. Chronic hemodynamic overload was caused by ascending aortic constriction (AAC) for 12 wk in mice with myocyte-specific transgenic expression of pCAT. AAC caused left ventricular hypertrophy and failure associated with a generalized increase in myocardial oxidative stress and specific oxidative modifications of SERCA at cysteine 674 and tyrosine 294/5. pCAT overexpression ameliorated myocardial hypertrophy and apoptosis, decreased pathological remodeling, and prevented the progression to heart failure. Likewise, pCAT prevented oxidative modifications of SERCA and increased SERCA activity without changing SERCA expression. Thus cardiac myocyte-restricted expression of pCAT effectively ameliorated the structural and functional consequences of chronic hemodynamic overload and increased SERCA activity via a post-translational mechanism, most likely by decreasing inhibitory oxidative modifications. In pressure overload-induced heart failure cardiac myocyte cytosolic ROS play a pivotal role in mediating key pathophysiologic events including hypertrophy, apoptosis, and decreased SERCA activity.


Subject(s)
Apoptosis/physiology , Cytosol/metabolism , Heart Failure/metabolism , Hydrogen Peroxide/metabolism , Hypertrophy, Left Ventricular/metabolism , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Disease Models, Animal , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics/physiology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Sarcoplasmic Reticulum/metabolism , Signal Transduction/physiology
15.
J Biol Chem ; 289(11): 7293-306, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24451382

ABSTRACT

Sirtuin-1 (SirT1), a member of the NAD(+)-dependent class III histone deacetylase family, is inactivated in vitro by oxidation of critical cysteine thiols. In a model of metabolic syndrome, SirT1 activation attenuated apoptosis of hepatocytes and improved liver function including lipid metabolism. We show in SirT1-overexpressing HepG2 cells that oxidants (nitrosocysteine and hydrogen peroxide) or metabolic stress (high palmitate and high glucose) inactivated SirT1 by reversible oxidative post-translational modifications (OPTMs) on three cysteines. Mutating these oxidation-sensitive cysteines to serine preserved SirT1 activity and abolished reversible OPTMs. Overexpressed mutant SirT1 maintained deacetylase activity and attenuated proapoptotic signaling, whereas overexpressed wild type SirT1 was less protective in metabolically or oxidant-stressed cells. To prove that OPTMs of SirT1 are glutathione (GSH) adducts, glutaredoxin-1 was overexpressed to remove this modification. Glutaredoxin-1 overexpression maintained endogenous SirT1 activity and prevented proapoptotic signaling in metabolically stressed HepG2 cells. The in vivo significance of oxidative inactivation of SirT1 was investigated in livers of high fat diet-fed C57/B6J mice. SirT1 deacetylase activity was decreased in the absence of changes in SirT1 expression and associated with a marked increase in OPTMs. These results indicate that glutathione adducts on specific SirT1 thiols may be responsible for dysfunctional SirT1 associated with liver disease in metabolic syndrome.


Subject(s)
Apoptosis , Liver/metabolism , Mutation , Oxidative Stress , Sirtuin 1/genetics , Amino Acid Sequence , Animals , Glutaredoxins/genetics , Glutathione/chemistry , HEK293 Cells , Hep G2 Cells , Humans , Liver Diseases/metabolism , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidants/chemistry , Oxidation-Reduction , Oxygen/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Sequence Homology, Amino Acid , Signal Transduction
16.
PLoS One ; 8(7): e68697, 2013.
Article in English | MEDLINE | ID: mdl-23894332

ABSTRACT

Diastolic heart failure (HF) i.e., "HF with preserved ejection fraction" (HF-preserved EF) accounts for up to 50% of all HF presentations; however there have been no therapeutic advances. This stems in part from an incomplete understanding about HF-preserved EF. Hypertension is the major cause of HF-preserved EF whilst HF-preserved EF is also highly associated with obesity. Similarly, excessive reactive oxygen species (ROS), i.e., oxidative stress occurs in hypertension and obesity, sensitizing the heart to the renin-angiotensin-aldosterone system, inducing autophagic type-II programmed cell death and accelerating the propensity to adverse cardiac remodeling, diastolic dysfunction and HF. Adiponectin (APN), an adipokine, mediates cardioprotective actions but it is unknown if APN modulates cardiomyocyte autophagy. We tested the hypothesis that APN ameliorates oxidative stress-induced autophagy in cardiomyocytes. Isolated adult rat ventricular myocytes were pretreated with recombinant APN (30 µg/mL) followed by 1mM hydrogen peroxide (H2O2) exposure. Wild type (WT) and APN-deficient (APN-KO) mice were infused with angiotensin (Ang)-II (3.2 mg/kg/d) for 14 days to induced oxidative stress. Autophagy-related proteins, mTOR, AMPK and ERK expression were measured. H2O2 induced LC3I to LC3II conversion by a factor of 3.4±1.0 which was abrogated by pre-treatment with APN by 44.5±10%. However, neither H2O2 nor APN affected ATG5, ATG7, or Beclin-1 expression. H2O2 increased phospho-AMPK by 49±6.0%, whilst pretreatment with APN decreased phospho-AMPK by 26±4%. H2O2 decreased phospho-mTOR by 36±13%, which was restored by APN. ERK inhibition demonstrated that the ERK-mTOR pathway is involved in H2O2-induced autophagy. Chronic Ang-II infusion significantly increased myocardial LC3II/I protein expression ratio in APN-KO vs. WT mice. These data suggest that excessive ROS caused cardiomyocyte autophagy which was ameliorated by APN by inhibiting an H2O2-induced AMPK/mTOR/ERK-dependent mechanism. These findings demonstrate the anti-oxidant potential of APN in oxidative stress-associated cardiovascular diseases, such as hypertension-induced HF-preserved EF.


Subject(s)
Adiponectin/genetics , Autophagy/genetics , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Animals , Autophagy/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Heat-Shock Proteins/genetics , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sequestosome-1 Protein , TOR Serine-Threonine Kinases/metabolism
17.
J Biol Chem ; 288(21): 15380-9, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23548900

ABSTRACT

We demonstrate for the first time that endomembrane-delimited H-Ras mediates VEGF-induced activation of endothelial nitric-oxide synthase (eNOS) and migratory response of human endothelial cells. Using thiol labeling strategies and immunofluorescent cell staining, we found that only 31% of total H-Ras is S-palmitoylated, tethering the small GTPase to the plasma membrane but leaving the function of the large majority of endomembrane-localized H-Ras unexplained. Knockdown of H-Ras blocked VEGF-induced PI3K-dependent Akt (Ser-473) and eNOS (Ser-1177) phosphorylation and nitric oxide-dependent cell migration, demonstrating the essential role of H-Ras. Activation of endogenous H-Ras led to recruitment and phosphorylation of eNOS at endomembranes. The loss of migratory response in cells lacking endogenous H-Ras was fully restored by modest overexpression of an endomembrane-delimited H-Ras palmitoylation mutant. These studies define a newly recognized role for endomembrane-localized H-Ras in mediating nitric oxide-dependent proangiogenic signaling.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Cells, Cultured , Endothelial Cells/cytology , Enzyme Induction/physiology , Humans , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Vascular Endothelial Growth Factor A/genetics
18.
Free Radic Biol Med ; 52(9): 1760-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22406435

ABSTRACT

Activation of thromboxane receptors (TPr) may promote atherosclerosis by enhancing oxidative stress and inflammation. This study examined the role of Nox1, an NADPH-oxidase subunit, in the enhancement of interleukin (IL)-1ß-induced monocyte adhesion by TPr. In cultured rat aortic vascular smooth muscle cells (VSMCs), U46619, a stable thromboxane A(2) mimetic, together with interleukin-1ß significantly enhanced Nox1 mRNA expression, as well as adhesion of THP-1 monocytes. Activation of TPr also enhanced IL-1ß-induced vascular cell adhesion molecule (VCAM)-1 expression, but inhibited inducible nitric oxide synthase (iNOS) expression. Silencing Nox1 expression by siRNA prevented the U46619 enhancement of IL-1ß-induced monocyte adhesion, but had no significant effect on VCAM-1 or iNOS expression. Furthermore, monocyte adhesion was inhibited by superoxide dismutase, enhanced by a specific iNOS inhibitor, l-N(6)-(1-iminoethyl)-lysine, but not influenced by catalase. U46619 inhibited IL-1ß-induced cyclic GMP production, and the inhibition was partially prevented by superoxide dismutase. In conclusion, activation of TPr enhances IL-1ß-induced Nox1 expression in VSMCs, which is responsible for the up-regulation of monocyte adhesion. The effect of Nox1 is independent of the changes in VCAM-1 and iNOS expression, but depends on the inactivation of nitric oxide via generation of superoxide anion.


Subject(s)
Cell Adhesion/physiology , Interleukin-1beta/physiology , NADH, NADPH Oxidoreductases/physiology , Receptors, Thromboxane/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Small Interfering , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Cell Adhesion Molecule-1/metabolism
19.
Proc Natl Acad Sci U S A ; 108(43): E899-906, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21987816

ABSTRACT

Factors secreted by the heart, referred to as "cardiokines," have diverse actions in the maintenance of cardiac homeostasis and remodeling. Follistatin-like 1 (Fstl1) is a secreted glycoprotein expressed in the adult heart and is induced in response to injurious conditions that promote myocardial hypertrophy and heart failure. The aim of this study was to investigate the role of cardiac Fstl1 in the remodeling response to pressure overload. Cardiac myocyte-specific Fstl1-KO mice were constructed and subjected to pressure overload induced by transverse aortic constriction (TAC). Although Fstl1-KO mice displayed no detectable baseline phenotype, TAC led to enhanced cardiac hypertrophic growth and a pronounced loss in ventricular performance by 4 wk compared with control mice. Conversely, mice that acutely or chronically overexpressed Fstl1 were resistant to pressure overload-induced hypertrophy and cardiac failure. Fstl1-deficient mice displayed a reduction in TAC-induced AMP-activated protein kinase (AMPK) activation in heart, whereas Fstl1 overexpression led to increased myocardial AMPK activation under these conditions. In cultured neonatal cardiomyocytes, administration of Fstl1 promoted AMPK activation and antagonized phenylephrine-induced hypertrophy. Inhibition of AMPK attenuated the antihypertrophic effect of Fstl1 treatment. These results document that cardiac Fstl1 functions as an autocrine/paracrine regulatory factor that antagonizes myocyte hypertrophic growth and the loss of ventricular performance in response to pressure overload, possibly through a mechanism involving the activation of the AMPK signaling axis.


Subject(s)
Cardiomegaly/metabolism , Follistatin-Related Proteins/metabolism , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Analysis of Variance , Animals , Blotting, Western , DNA Primers/genetics , Echocardiography , Follistatin-Related Proteins/genetics , Mice , Mice, Knockout , Pressure , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Statistics, Nonparametric , Ventricular Remodeling/genetics
20.
Circulation ; 124(7): 806-13, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21788586

ABSTRACT

BACKGROUND: Oxidative stress and mitochondrial dysfunction are central mediators of cardiac dysfunction after ischemia/reperfusion. ATP binding cassette mitochondrial erythroid (ABC-me; ABCB10; mABC2) is a mitochondrial transporter highly induced during erythroid differentiation and predominantly expressed in bone marrow, liver, and heart. Until now, ABC-me function in heart was unknown. Several lines of evidence demonstrate that the yeast ortholog of ABC-me protects against increased oxidative stress. Therefore, ABC-me is a potential modulator of the outcome of ischemia/reperfusion in the heart. METHODS AND RESULTS: Mice harboring 1 functional allele of ABC-me (ABC-me(+/-)) were generated by replacing ABC-me exons 2 and 3 with a neomycin resistance cassette. Cardiac function was assessed with Langendorff perfusion and echocardiography. Under basal conditions, ABC-me(+/-) mice had normal heart structure, hemodynamic function, mitochondrial respiration, and oxidative status. However, after ischemia/reperfusion, the recovery of hemodynamic function was reduced by 50% in ABC-me(+/-) hearts as a result of impairments in both systolic and diastolic function. This reduction was associated with impaired mitochondrial bioenergetic function and with oxidative damage to both mitochondrial lipids and sarcoplasmic reticulum calcium ATPase after reperfusion. Treatment of ABC-me(+/-) hearts with the superoxide dismutase/catalase mimetic EUK-207 prevented oxidative damage to mitochondria and sarcoplasmic reticulum calcium ATPase and restored mitochondrial and cardiac function to wild-type levels after reperfusion. CONCLUSIONS: Inactivation of 1 allele of ABC-me increases the susceptibility to oxidative stress induced by ischemia/reperfusion, leading to increased oxidative damage to mitochondria and sarcoplasmic reticulum calcium ATPase and to impaired functional recovery. Thus, ABC-me is a novel gene that determines the ability to tolerate cardiac ischemia/reperfusion.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Mitochondria/physiology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/genetics , Recovery of Function/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cardiac Volume/physiology , Catalase/metabolism , Female , Genetic Predisposition to Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria/drug effects , Mutagenesis, Insertional , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/drug therapy , Organometallic Compounds/pharmacology , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Ventricular Pressure/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...