Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Am Heart Assoc ; 11(11): e025295, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35656993

ABSTRACT

Background cMyBP-C (Cardiac myosin binding protein-C) regulates cardiac contraction and relaxation. Previously, we demonstrated that elevated myocardial S-glutathionylation of cMyBP-C correlates with diastolic dysfunction (DD) in animal models. In this study, we tested whether circulating S-glutathionylated cMyBP-C would be a biomarker for DD. Methods and Results Humans, African Green monkeys, and mice had DD determined by echocardiography. Blood samples were acquired and analyzed for S-glutathionylated cMyBP-C by immunoprecipitation. Circulating S-glutathionylated cMyBP-C in human participants with DD (n=24) was elevated (1.46±0.13-fold, P=0.014) when compared with the non-DD controls (n=13). Similarly, circulating S-glutathionylated cMyBP-C was upregulated by 2.13±0.47-fold (P=0.047) in DD monkeys (n=6), and by 1.49 (1.22-2.06)-fold (P=0.031) in DD mice (n=5) compared with the respective non-DD controls. Circulating S-glutathionylated cMyBP-C was positively correlated with DD in humans. Conclusions Circulating S-glutathionylated cMyBP-C was elevated in humans, monkeys, and mice with DD. S-glutathionylated cMyBP-C may represent a novel biomarker for the presence of DD.


Subject(s)
Carrier Proteins/analysis , Heart Diseases , Animals , Biomarkers , Carrier Proteins/metabolism , Chlorocebus aethiops , Diastole/physiology , Heart Diseases/metabolism , Humans , Mice , Myocardial Contraction , Myocardium/metabolism , Phosphorylation
2.
Cardiovasc Res ; 118(16): 3211-3224, 2022 12 29.
Article in English | MEDLINE | ID: mdl-35018410

ABSTRACT

AIMS: Pulmonary arterial hypertension (PAH) is a fatal disease without a cure. Previously, we found that transcription factor RUNX1-dependent haematopoietic transformation of endothelial progenitor cells may contribute to the pathogenesis of PAH. However, the therapeutic potential of RUNX1 inhibition to reverse established PAH remains unknown. In the current study, we aimed to determine whether RUNX1 inhibition was sufficient to reverse Sugen/hypoxia (SuHx)-induced pulmonary hypertension (PH) in rats. We also aimed to demonstrate possible mechanisms involved. METHODS AND RESULTS: We administered a small molecule specific RUNX1 inhibitor Ro5-3335 before, during, and after the development of SuHx-PH in rats to investigate its therapeutic potential. We quantified lung macrophage recruitment and activation in vivo and in vitro in the presence or absence of the RUNX1 inhibitor. We generated conditional VE-cadherin-CreERT2; ZsGreen mice for labelling adult endothelium and lineage tracing in the SuHx-PH model. We also generated conditional Cdh5-CreERT2; Runx1(flox/flox) mice to delete Runx1 gene in adult endothelium and LysM-Cre; Runx1(flox/flox) mice to delete Runx1 gene in cells of myeloid lineage, and then subjected these mice to SuHx-PH induction. RUNX1 inhibition in vivo effectively prevented the development, blocked the progression, and reversed established SuHx-induced PH in rats. RUNX1 inhibition significantly dampened lung macrophage recruitment and activation. Furthermore, lineage tracing with the inducible VE-cadherin-CreERT2; ZsGreen mice demonstrated that a RUNX1-dependent endothelial to haematopoietic transformation occurred during the development of SuHx-PH. Finally, tissue-specific deletion of Runx1 gene either in adult endothelium or in cells of myeloid lineage prevented the mice from developing SuHx-PH, suggesting that RUNX1 is required for the development of PH. CONCLUSION: By blocking RUNX1-dependent endothelial to haematopoietic transformation and pulmonary macrophage recruitment and activation, targeting RUNX1 may be as a novel treatment modality for pulmonary arterial hypertension.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Rats , Mice , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Familial Primary Pulmonary Hypertension , Hypoxia/complications , Pulmonary Artery , Disease Models, Animal
3.
Aging (Albany NY) ; 12(24): 25939-25955, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33378745

ABSTRACT

Adult hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) ensuring homeostasis of blood production and immune response throughout life. Sex differences in immunocompetence and mortality are well-documented in humans. However, whether HSPCs behave dimorphically between sexes during aging remains unknown. Here, we show that a significant expansion of BM-derived HSPCs occurs in the middle age of female but in the old age of male mice. We then show that a decline of HSPCs in male mice, as indicated by the expression levels of select hematopoietic genes, occurs much earlier in the aging process than that in female mice. Sex-mismatched heterochronic BM transplantations indicate that the middle-aged female BM microenvironment plays a pivotal role in sustaining hematopoietic gene expression during aging. Furthermore, a higher concentration of the pituitary sex hormone follicle-stimulating hormone (FSH) in the serum and a concomitant higher expression of its receptor on HSPCs in the middle-aged and old female mice than age-matched male mice, suggests that FSH may contribute to the sexual dimorphism in aging hematopoiesis. Our study reveals that HSPCs in the BM niches are possibly regulated in a sex-specific manner and influenced differently by sex hormones during aging hematopoiesis.


Subject(s)
Aging/physiology , Follicle Stimulating Hormone/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Receptors, FSH/metabolism , Sex Characteristics , Animals , Antigens, Ly/metabolism , Bone Marrow , Bone Marrow Transplantation , Cell Lineage , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Follicle Stimulating Hormone/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Male , Membrane Proteins/metabolism , Mice , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Stem Cell Niche
4.
JCI Insight ; 4(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30626750

ABSTRACT

In heart failure and type 2 diabetes mellitus (DM), the majority of patients have hypomagnesemia, and magnesium (Mg) supplementation has improved cardiac function and insulin resistance. Recently, we have shown that DM can cause cardiac diastolic dysfunction (DD). Therefore, we hypothesized that Mg supplementation would improve diastolic function in DM. High-fat diet-induced diabetic mouse hearts showed increased cardiac DD and hypertrophy. Mice with DM showed a significantly increased E/e' ratio (the ratio of transmitral Doppler early filling velocity [E] to tissue Doppler early diastolic mitral annular velocity [e']) in the echocardiogram, left ventricular end diastolic volume (LVEDV), incidence of DD, left ventricular posterior wall thickness in diastole (PWTd), and ratio of heart weight to tibia length (HW/TL) when compared with controls. DM mice also had hypomagnesemia. Ventricular cardiomyocytes isolated from DM mice exhibited decreased mitochondrial ATP production, a 1.7- ± 0.2-fold increase of mitochondrial ROS, depolarization of the mitochondrial membrane potential, and mitochondrial Ca2+ overload. Dietary Mg administration (50 mg/ml in the drinking water) for 6 weeks increased plasma Mg concentration and improved cardiac function. At the cellular level, Mg improved mitochondrial function with increased ATP, decreased mitochondrial ROS and Ca2+ overload, and repolarized mitochondrial membrane potential. In conclusion, Mg supplementation improved mitochondrial function, reduced oxidative stress, and prevented DD in DM.

5.
J Am Heart Assoc ; 7(9)2018 04 20.
Article in English | MEDLINE | ID: mdl-29678826

ABSTRACT

BACKGROUND: Although transcription is the initial process of gene expression, posttranscriptional gene expression regulation has also played a critical role for fine-tuning gene expression in a fast, precise, and cost-effective manner. Although the regulation of sodium channel α-subunit (SCN5A) mRNA expression has been studied at both transcriptional and pre-mRNA splicing levels, the molecular mechanisms governing SCN5A mRNA expression are far from clear. METHODS AND RESULTS: Herein, we show that, as evidenced by ribonucleoprotein immunoprecipitation assay, RNA binding protein Hu antigen R/ELAV like RNA binding protein 1 (HuR/ELAVL1) and myocyte enhancer factor-2C (MEF2C) transcription factor mRNA are associated. HuR positively regulated transcription factor MEF2C mRNA expression by protecting its mRNA from degradation. As demonstrated by both chromatin immunoprecipitation-quantitative polymerase chain reaction assay and an electrophoretic mobility shift assay, MEF2C enhanced SCN5A transcription by binding to a putative MEF2C binding site within SCN5A promoter region. Overexpression of HuR increased the expression of SCN5A mRNA, and this effect was attenuated by the presence of MEF2C small interfering RNA in cardiomyocytes. CONCLUSIONS: In conclusion, our results suggested that HuR participates in a combined network at the DNA and RNA levels that regulates SCN5A mRNA expression. HuR upregulates MEF2C mRNA expression by protecting MEF2C mRNA from degradation, and consequently, the elevated MEF2C enhances SCN5A mRNA transcription.


Subject(s)
ELAV-Like Protein 1/metabolism , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , RNA Stability , RNA, Messenger/metabolism , Binding Sites , Cell Line , ELAV-Like Protein 1/genetics , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcriptional Activation , Up-Regulation
6.
Heart Rhythm ; 15(7): 1072-1080, 2018 07.
Article in English | MEDLINE | ID: mdl-29454929

ABSTRACT

BACKGROUND: Downregulated sodium currents in heart failure (HF) have been linked to increased arrhythmic risk. Reduced expression of the messenger RNA (mRNA)-stabilizing protein HuR (also known as ELAVL1) may be responsible for the downregulation of sodium channel gene SCN5A mRNA. OBJECTIVE: The purpose of this article was to investigate whether HuR regulates SCN5A mRNA expression and whether manipulation of HuR benefits arrhythmia control in HF. METHODS: Quantitative real-time reverse-transcriptase polymerase chain reaction was used to investigate the expression of SCN5A. Optical mapping of the intact heart was adopted to study the effects of HuR on the conduction velocity and action potential upstroke in mice with myocardial infarct and HF after injection of AAV9 viral particles carrying HuR. RESULTS: HuR was associated with SCN5A mRNA in cardiomyocytes, and expression of HuR was downregulated in failing hearts. The association of HuR and SCN5A mRNA protected SCN5A mRNA from decay. Injection of AAV9 viral particles carrying HuR increased SCN5A expression in mouse heart tissues after MI. Optical mapping of the intact heart demonstrated that overexpression of HuR improved action potential upstroke and conduction velocity in the infarct border zone, which reduced the risk of reentrant arrhythmia after MI. CONCLUSION: Our data indicate that HuR is an important RNA-binding protein in maintaining SCN5A mRNA abundance in cardiomyocytes. Reduced expression of HuR may be at least partially responsible for the downregulation of SCN5A mRNA expression in ischemic HF. Overexpression of HuR may rescue decreased SCN5A expression and reduce arrhythmic risk in HF. Increasing mRNA stability to increase ion channel currents may correct a fundamental defect in HF and represent a new paradigm in antiarrhythmic therapy.


Subject(s)
Arrhythmias, Cardiac/genetics , ELAV-Like Protein 1/genetics , Gene Expression Regulation , Heart Failure/genetics , Myocardium/pathology , NAV1.5 Voltage-Gated Sodium Channel/genetics , RNA, Messenger/genetics , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/pathology , Cells, Cultured , ELAV-Like Protein 1/biosynthesis , Heart Failure/complications , Heart Failure/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L748-L759, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28258105

ABSTRACT

Right ventricular (RV) dysfunction is associated with numerous smoking-related illnesses, including chronic obstructive pulmonary disease (COPD), in which it is present even in the absence of pulmonary hypertension. It is unknown whether exposure to cigarette smoke (CS) has direct effects on RV function and cardiac fibroblast (CF) proliferation or collagen synthesis. In this study, we evaluated cardiac function and fibrosis in mice exposed to CS and determined mechanisms of smoke-induced changes in CF signaling and fibrosis. AKR mice were exposed to CS for 6 wk followed by echocardiography and evaluation of cardiac hypertrophy, collagen content, and pulmonary muscularization. Proliferation and collagen content were evaluated in primary isolated rat CFs exposed to CS extract (CSE) or nicotine. Markers of cell proliferation, fibrosis, and proliferative signaling were determined by immunoblot or Sircol collagen assay. Mice exposed to CS had significantly decreased RV function, as determined by tricuspid annular plane systolic excursion. There were no changes in left ventricular parameters. RV collagen content was significantly elevated, but there was no change in RV hypertrophy or pulmonary vascular muscularization. CSE directly increased CF proliferation and collagen content in CF. Nicotine alone reproduced these effects. CSE and nicotine-induced fibroblast proliferation and collagen content were mediated through α7 nicotinic acetylcholine receptors and were dependent on PKC-α, PKC-δ, and reduced p38-MAPK phosphorylation. CS and nicotine have direct effects on CFs to induce proliferation and fibrosis, which may negatively affect right heart function.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Heart Ventricles/pathology , Myocardium/pathology , Smoking/adverse effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Fibroblasts/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/diagnostic imaging , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred AKR , Nicotine/pharmacology , Phosphorylation/drug effects , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Rats, Sprague-Dawley , Vascular Remodeling/drug effects , Ventricular Dysfunction, Right/complications , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/pathology , Ventricular Dysfunction, Right/physiopathology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Am Heart Assoc ; 5(5)2016 05 05.
Article in English | MEDLINE | ID: mdl-27151515

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is associated with mitochondrial oxidative stress. We have shown that myocardial oxidative stress leads to diastolic dysfunction in a hypertensive mouse model. Therefore, we hypothesized that diabetes mellitus could cause diastolic dysfunction through mitochondrial oxidative stress and that a mitochondria-targeted antioxidant (MitoTEMPO) could prevent diastolic dysfunction in a diabetic mouse model. METHODS AND RESULTS: C57BL/6J mice were fed either 60 kcal % fat diet (high-fat diet [HFD]) or normal chow (control) for 8 weeks with or without concurrent MitoTEMPO administration, followed by in vivo assessment of diastolic function and ex vivo studies. HFD mice developed impaired glucose tolerance compared with the control (serum glucose=495±45 mg/dL versus 236±30 mg/dL at 60 minutes after intraperitoneal glucose injection, P<0.05). Myocardial tagged cardiac magnetic resonance imaging showed significantly reduced diastolic circumferential strain (Ecc) rate in the HFD mice compared with controls (5.0±0.3 1/s versus 7.4±0.5 1/s, P<0.05), indicating diastolic dysfunction in the HFD mice. Systolic function was comparable in both groups (left ventricular ejection fraction=66.4±1.4% versus 66.7±1.2%, P>0.05). MitoTEMPO-treated HFD mice showed significant reduction in mitochondria reactive oxygen species, S-glutathionylation of cardiac myosin binding protein C, and diastolic dysfunction, comparable to the control. The fasting insulin levels of MitoTEMPO-treated HFD mice were also comparable to the controls (P>0.05). CONCLUSIONS: MitoTEMPO treatment prevented insulin resistance and diastolic dysfunction, suggesting that mitochondrial oxidative stress may be involved in the pathophysiology of both conditions.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/metabolism , Glucose/metabolism , Heart Failure, Diastolic/metabolism , Insulin Resistance , Mitochondria, Heart/metabolism , Oxidative Stress , Animals , Biomechanical Phenomena , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Diastole , Diet, High-Fat , Disease Models, Animal , Heart Failure, Diastolic/diagnostic imaging , Hemodynamics , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Reactive Oxygen Species/metabolism
9.
Biomed Res Int ; 2015: 825027, 2015.
Article in English | MEDLINE | ID: mdl-26504834

ABSTRACT

Animal models have suggested a role of renin-angiotensin system (RAS) activation and subsequent cardiac oxidation in heart failure with preserved ejection fraction (HFpEF). Nevertheless, RAS blockade has failed to show efficacy in treatment of HFpEF. We evaluated the role of RAS activation and subsequent systemic oxidation in HFpEF. Oxidative stress markers were compared in 50 subjects with and without early HFpEF. Derivatives of reactive oxidative metabolites (DROMs), F2-isoprostanes (IsoPs), and ratios of oxidized to reduced glutathione (E h GSH) and cysteine (E h CyS) were measured. Angiotensin converting enzyme (ACE) levels and activity were measured. On univariate analysis, HFpEF was associated with male sex (p = 0.04), higher body mass index (BMI) (p = 0.003), less oxidized E h CyS (p = 0.001), lower DROMs (p = 0.02), and lower IsoP (p = 0.03). Higher BMI (OR: 1.3; 95% CI: 1.1-1.6) and less oxidized E h CyS (OR: 1.2; 95% CI: 1.1-1.4) maintained associations with HFpEF on multivariate analysis. Though ACE levels were higher in early HFpEF (OR: 1.09; 95% CI: 1.01-1.05), ACE activity was similar to that in controls. HFpEF is not associated with significant systemic RAS activation or oxidative stress. This may explain the failure of RAS inhibitors to alter outcomes in HFpEF.


Subject(s)
Heart Failure/physiopathology , Oxidative Stress/physiology , Renin-Angiotensin System/physiology , Stroke Volume/physiology , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Female , Heart Failure/epidemiology , Humans , Male , Middle Aged
10.
Circ J ; 79(3): 470-7, 2015.
Article in English | MEDLINE | ID: mdl-25746522

ABSTRACT

Despite the growing number of patients affected, the understanding of diastolic dysfunction and heart failure with preserved ejection fraction (HFpEF) is still poor. Clinical trials, largely based on successful treatments for systolic heart failure, have been disappointing, suggesting that HFpEF has a different pathology to that of systolic dysfunction. In this review, general concepts, epidemiology, diagnosis, and treatment of diastolic dysfunction are summarized, with an emphasis on new experiments suggesting that oxidative stress plays a crucial role in the pathogenesis of at least some forms of the disease. This observation has lead to potential new diagnostics and therapeutics for diastolic dysfunction and heart failure caused by diastolic dysfunction.


Subject(s)
Diastole , Heart Failure , Oxidative Stress , Stroke Volume , Animals , Clinical Trials as Topic , Heart Failure/diagnosis , Heart Failure/physiopathology , Heart Failure/therapy , Humans
11.
R I Med J (2013) ; 97(2): 35-7, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24494212

ABSTRACT

Despite the fact that up to half of all heart failure occurs in patients without evidence of systolic cardiac dysfunction, there are no universally accepted diagnostic markers and no approved therapies for heart failure with preserved ejection fraction (HFpEF). HFpEF, otherwise known as diastolic heart failure, has nearly the same grim prognosis as systolic heart failure, and diastolic heart failure is increasing in incidence and prevalence. Major trials have shown that many of the treatments that are salutary in systolic heart failure have no beneficial effects in diastolic heart failure, suggesting different underlying mechanisms for these two disorders. Even criteria for diagnosis of HFpEF are still debated, and there is still no gold standard marker to detect diastolic dysfunction. Here, we will review some promising new insights into the pathogenesis of diastolic dysfunction that may lead to new diagnostic and therapeutic tools.


Subject(s)
Heart Failure, Diastolic/diagnosis , Heart Failure, Diastolic/therapy , Humans
12.
Circ Arrhythm Electrophysiol ; 6(5): 1018-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24036084

ABSTRACT

BACKGROUND: Human heart failure (HF) increases alternative mRNA splicing of the type V, voltage-gated cardiac Na+ channel α-subunit (SCN5A), generating variants encoding truncated, nonfunctional channels that are trapped in the endoplasmic reticulum. In this work, we tested whether truncated Na+ channels activate the unfolded protein response (UPR), contributing to SCN5A electric remodeling in HF. METHODS AND RESULTS: UPR and SCN5A were analyzed in human ventricular systolic HF tissue samples and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Cells were exposed to angiotensin II (AngII) and hypoxia, known activators of abnormal SCN5A mRNA splicing, or were induced to overexpress SCN5A variants. UPR effectors, protein kinase R-like ER kinase (PERK), calreticulin, and CHOP, were increased in human HF tissues. Induction of SCN5A variants with AngII or hypoxia or the expression of exogenous variants induced the UPR with concomitant downregulation of Na+ current. PERK activation destabilized SCN5A and, surprisingly, Kv4.3 channel mRNAs but not transient receptor potential cation channel M7 (TRPM7) channel mRNA. PERK inhibition prevented the loss of full-length SCN5A and Kv4.3 mRNA levels resulting from expressing Na+ channel mRNA splice variants. CONCLUSIONS: UPR can be initiated by Na+ channel mRNA splice variants and is involved in the reduction of cardiac Na+ current during human HF. Because the effect is not entirely specific to the SCN5A transcript, the UPR may play an important role in downregulation of multiple cardiac genes in HF.


Subject(s)
Heart Failure, Systolic/metabolism , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Unfolded Protein Response/physiology , Angiotensin II/pharmacology , Blotting, Western , CCAAT-Enhancer-Binding Proteins/metabolism , Calreticulin/metabolism , Electrophysiologic Techniques, Cardiac , Endoplasmic Reticulum/metabolism , Heart Failure, Systolic/physiopathology , Humans , NAV1.5 Voltage-Gated Sodium Channel/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transfection , eIF-2 Kinase/metabolism
13.
Circ Arrhythm Electrophysiol ; 6(3): 623-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559673

ABSTRACT

BACKGROUND: Previously, we showed that a mouse model (ACE8/8) of cardiac renin-angiotensin system activation has a high rate of spontaneous ventricular tachycardia and sudden cardiac death secondary to a reduction in connexin43 level. Angiotensin-II activation increases reactive oxygen species (ROS) production, and ACE8/8 mice show increased cardiac ROS. We sought to determine the source of ROS and whether ROS played a role in the arrhythmogenesis. METHODS AND RESULTS: Wild-type and ACE8/8 mice with and without 2 weeks of treatment with L-NIO (NO synthase inhibitor), sepiapterin (precursor of tetrahydrobiopterin), MitoTEMPO (mitochondria-targeted antioxidant), TEMPOL (a general antioxidant), apocynin (nicotinamide adenine dinucleotide phosphate oxidase inhibitor), allopurinol (xanthine oxidase inhibitor), and ACE8/8 crossed with P67 dominant negative mice to inhibit the nicotinamide adenine dinucleotide phosphate oxidase were studied. Western blotting, detection of mitochondrial ROS by MitoSOX Red, electron microscopy, immunohistochemistry, fluorescent dye diffusion technique for functional assessment of connexin43, telemetry monitoring, and in vivo electrophysiology studies were performed. Treatment with MitoTEMPO reduced sudden cardiac death in ACE8/8 mice (from 74% to 18%; P<0.005), decreased spontaneous ventricular premature beats, decreased ventricular tachycardia inducibility (from 90% to 17%; P<0.05), diminished elevated mitochondrial ROS to the control level, prevented structural damage to mitochondria, resulted in 2.6-fold increase in connexin43 level at the gap junctions, and corrected gap junction conduction. None of the other antioxidant therapies prevented ventricular tachycardia and sudden cardiac death in ACE8/8 mice. CONCLUSIONS: Mitochondrial oxidative stress plays a central role in angiotensin II-induced gap junction remodeling and arrhythmia. Mitochondria-targeted antioxidants may be effective antiarrhythmic drugs in cases of renin-angiotensin system activation.


Subject(s)
Antioxidants/pharmacology , Connexin 43/metabolism , Death, Sudden, Cardiac/etiology , Mitochondria/metabolism , Oxidative Stress/physiology , Tachycardia, Ventricular/drug therapy , Acetophenones/pharmacology , Animals , Connexin 43/drug effects , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Mice , Mice, Inbred Strains , NADPH Oxidases/pharmacology , Random Allocation , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/drug effects , Risk Factors , Sensitivity and Specificity , Spin Labels , Tachycardia, Ventricular/physiopathology
14.
J Mol Cell Cardiol ; 54: 25-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123323

ABSTRACT

Cardiomyopathy is associated with cardiac Na(+) channel downregulation that may contribute to arrhythmias. Previously, we have shown that elevated intracellular NADH causes a decrease in cardiac Na(+) current (I(Na)) signaled by an increase in mitochondrial reactive oxygen species (ROS). In this study, we tested whether the NADH-mitochondria ROS pathway was involved in the reduction of I(Na) in a nonischemic cardiomyopathic model and correlated the findings with myopathic human hearts. Nonischemic cardiomyopathy was induced in C57BL/6 mice by hypertension after unilateral nephrectomy, deoxycorticosterone acetate (DOCA) pellet implantation, and salt water substitution. Sham operated mice were used as controls. After six weeks, heart tissue and ventricular myocytes isolated from mice were utilized for whole cell patch clamp recording, NADH/NAD(+) level measurements, and mitochondrial ROS monitoring with confocal microscopy. Human explanted hearts were studied using optical mapping. Compared to the sham mice, the arterial blood pressure was higher, the left ventricular volume was significantly enlarged (104.7±3.9 vs. 87.9±6.1 µL, P<0.05), and the ejection fraction was reduced (37.1±1.8% vs. 49.4±3.7%, P<0.05) in DOCA mice. Both the whole cell and cytosolic NADH level were increased (279±70% and 123±2% of sham, respectively, P<0.01), I(Na) was decreased (60±10% of sham, P<0.01), and mitochondrial ROS overproduction was observed (2.9±0.3-fold of sham, P<0.01) in heart tissue and myocytes of myopathic mice vs. sham. Treatment of myocytes with NAD(+) (500 µM), mitoTEMPO (10 µM), chelerythrine (50 µM), or forskolin (5 µM) restored I(Na) back to the level of sham. Injection of NAD(+) (100mg/kg) or mitoTEMPO (0.7 mg/kg) twice (at 24h and 1h before myocyte isolation) to animals also restored I(Na). All treatments simultaneously reduced mitochondrial ROS levels to that of controls. CD38 was found to transduce the extracellular NAD(+) signal. Correlating with the mouse model, failing human hearts showed a reduction in conduction velocity that improved with NAD(+). Nonischemic cardiomyopathy was associated with elevated NADH level, PKC activation, mitochondrial ROS overproduction, and a concomitant decrease in I(Na). Reducing mitochondrial ROS by application of NAD(+), mitoTEMPO, PKC inhibitors, or PKA activators, restored I(Na). NAD(+) improved conduction velocity in human myopathic hearts.


Subject(s)
Cardiomyopathies/metabolism , Mitochondria, Heart/physiology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , ADP-ribosyl Cyclase 1/metabolism , Action Potentials/drug effects , Animals , Benzophenanthridines/pharmacology , Colforsin/pharmacology , Down-Regulation , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Humans , In Vitro Techniques , Membrane Glycoproteins/metabolism , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , NAD/metabolism , NAD/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Organophosphorus Compounds/pharmacology , Oxidative Stress , Patch-Clamp Techniques , Piperidines/pharmacology , Reactive Oxygen Species/metabolism
15.
J Mol Cell Cardiol ; 56: 44-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23247392

ABSTRACT

Despite the increasing prevalence of heart failure with preserved left ventricular function, there are no specific treatments, partially because the mechanism of impaired relaxation is incompletely understood. Evidence indicates that cardiac relaxation may depend on nitric oxide (NO), generated by NO synthase (NOS) requiring the co-factor tetrahydrobiopterin (BH(4)). Recently, we reported that hypertension-induced diastolic dysfunction was accompanied by cardiac BH(4) depletion, NOS uncoupling, a depression in myofilament cross-bridge kinetics, and S-glutathionylation of myosin binding protein C (MyBP-C). We hypothesized that the mechanism by which BH(4) ameliorates diastolic dysfunction is by preventing glutathionylation of MyBP-C and thus reversing changes of myofilament properties that occur during diastolic dysfunction. We used the deoxycorticosterone acetate (DOCA)-salt mouse model, which demonstrates mild hypertension, myocardial oxidative stress, and diastolic dysfunction. Mice were divided into two groups that received control diet and two groups that received BH(4) supplement for 7days after developing diastolic dysfunction at post-operative day 11. Mice were assessed by echocardiography. Left ventricular papillary detergent-extracted fiber bundles were isolated for simultaneous determination of force and ATPase activity. Sarcomeric protein glutathionylation was assessed by immunoblotting. DOCA-salt mice exhibited diastolic dysfunction that was reversed after BH(4) treatment. Diastolic sarcomere length (DOCA-salt 1.70±0.01 vs. DOCA-salt+BH(4) 1.77±0.01µm, P<0.001) and relengthening (relaxation constant, τ, DOCA-salt 0.28±0.02 vs. DOCA-salt+BH(4) 0.08±0.01, P<0.001) were also restored to control by BH(4) treatment. pCa(50) for tension increased in DOCA-salt compared to sham but reverted to sham levels after BH(4) treatment. Maximum ATPase rate and tension cost (ΔATPase/ΔTension) decreased in DOCA-salt compared to sham, but increased after BH(4) treatment. Cardiac MyBP-C glutathionylation increased in DOCA-salt compared to sham, but decreased with BH(4) treatment. MyBP-C glutathionylation correlated with the presence of diastolic dysfunction. Our results suggest that by depressing S-glutathionylation of MyBP-C, BH(4) ameliorates diastolic dysfunction by reversing a decrease in cross-bridge turnover kinetics. These data provide evidence for modulation of cardiac relaxation by post-translational modification of myofilament proteins.


Subject(s)
Biopterins/analogs & derivatives , Cardiovascular Agents/administration & dosage , Heart Failure, Diastolic/drug therapy , Myofibrils/physiology , Adenosine Triphosphatases/metabolism , Administration, Oral , Animals , Biopterins/administration & dosage , Carrier Proteins/metabolism , Cells, Cultured , Desoxycorticosterone/pharmacology , Diastole/drug effects , Dietary Supplements , Glutathione/metabolism , Heart Failure, Diastolic/diagnostic imaging , Heart Failure, Diastolic/physiopathology , Mice , Myofibrils/drug effects , Myofibrils/enzymology , Oxidative Stress , Protein Processing, Post-Translational , Stroke Volume/drug effects , Ultrasonography
16.
Congest Heart Fail ; 18(4): 187-91, 2012.
Article in English | MEDLINE | ID: mdl-22809257

ABSTRACT

Diastolic dysfunction (DD) with preserved left ventricular (LV) ejection fraction (EF) has been linked to obesity. Adiponectin is a cytokine related to obesity and obesity-linked cardiovascular complications. The authors aimed to determine the independent association of DD with adiponectin. Fifty patients with impaired relaxation DD and a normal EF and age-matched normal controls were recruited. Plasma levels of total and high molecular weight (HMW) adiponectin were measured. Mid and low molecular weight (MMW+LMW) fractions of adiponectin were calculated by subtracting HMW fraction from total adiponectin. The DD group had significantly lower total (median, 4.4 vs 12.7 µg/mL; P=.001), HMW fraction (median, 1.3 vs 3.4 µg/mL; P=.02), and MMW+LMW fraction of adiponectin (median, 3.8 vs 7.2 µg/mL; P=.01). Body mass index (BMI) negatively correlated with total (r:-0.46, P=.003), HMW (r:-0.32, P=.038), and MMW+LMW (r:-0.40, P=.006) fractions of adiponectin. DD had an independent association with both BMI (P<.05) and total adiponectin (P<.001) in linear regression model using sex, BMI, blood pressure, and total adiponectin as covariates. DD was associated with BMI (P=.02), HMW fraction (P=.03), and MMW+LMW fraction (P=.004) in similar linear regression analyses. Adiponectin deficiency may be one explanation for the adiposity-related cardiac oxidation known to be involved in the pathogenesis of DD.


Subject(s)
Adiponectin/blood , Heart Failure, Diastolic/blood , Obesity/blood , Ventricular Dysfunction, Left/blood , Aged , Body Mass Index , Case-Control Studies , Cross-Sectional Studies , Diastole , Disease Progression , Female , Heart Failure, Diastolic/pathology , Humans , Male , Middle Aged , Multivariate Analysis , Obesity/pathology , Risk Assessment , Ventricular Dysfunction, Left/pathology
17.
Circ Res ; 110(6): 841-50, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22343711

ABSTRACT

RATIONALE: Previously, we demonstrated that a deoxycorticosterone acetate (DOCA)-salt hypertensive mouse model produces cardiac oxidative stress and diastolic dysfunction with preserved systolic function. Oxidative stress has been shown to increase late inward sodium current (I(Na)), reducing the net cytosolic Ca(2+) efflux. OBJECTIVE: Oxidative stress in the DOCA-salt model may increase late I(Na), resulting in diastolic dysfunction amenable to treatment with ranolazine. METHODS AND RESULTS: Echocardiography detected evidence of diastolic dysfunction in hypertensive mice that improved after treatment with ranolazine (E/E':sham, 31.9 ± 2.8, sham+ranolazine, 30.2 ± 1.9, DOCA-salt, 41.8 ± 2.6, and DOCA-salt+ranolazine, 31.9 ± 2.6; P=0.018). The end-diastolic pressure-volume relationship slope was elevated in DOCA-salt mice, improving to sham levels with treatment (sham, 0.16 ± 0.01 versus sham+ranolazine, 0.18 ± 0.01 versus DOCA-salt, 0.23 ± 0.2 versus DOCA-salt+ranolazine, 0.17 ± 0.0 1 mm Hg/L; P<0.005). DOCA-salt myocytes demonstrated impaired relaxation, τ, improving with ranolazine (DOCA-salt, 0.18 ± 0.02, DOCA-salt+ranolazine, 0.13 ± 0.01, sham, 0.11 ± 0.01, sham+ranolazine, 0.09 ± 0.02 seconds; P=0.0004). Neither late I(Na) nor the Ca(2+) transients were different from sham myocytes. Detergent extracted fiber bundles from DOCA-salt hearts demonstrated increased myofilament response to Ca(2+) with glutathionylation of myosin binding protein C. Treatment with ranolazine ameliorated the Ca(2+) response and cross-bridge kinetics. CONCLUSIONS: Diastolic dysfunction could be reversed by ranolazine, probably resulting from a direct effect on myofilaments, indicating that cardiac oxidative stress may mediate diastolic dysfunction through altering the contractile apparatus.


Subject(s)
Acetanilides/pharmacology , Calcium/metabolism , Diastole/drug effects , Heart Failure, Diastolic/drug therapy , Myocytes, Cardiac/drug effects , Myofibrils/drug effects , Piperazines/pharmacology , Acetanilides/blood , Animals , Desoxycorticosterone/toxicity , Diastole/physiology , Disease Models, Animal , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/physiopathology , Mice , Mineralocorticoids/toxicity , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Myofibrils/metabolism , Oxidative Stress/physiology , Piperazines/blood , Ranolazine , Sodium/metabolism , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/physiopathology
18.
J Mol Cell Cardiol ; 52(2): 454-63, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21978629

ABSTRACT

Cardiac arrhythmias can cause sudden cardiac death (SCD) and add to the current heart failure (HF) health crisis. Nevertheless, the pathological processes underlying arrhythmias are unclear. Arrhythmic conditions are associated with systemic and cardiac oxidative stress caused by reactive oxygen species (ROS). In excitable cardiac cells, ROS regulate both cellular metabolism and ion homeostasis. Increasing evidence suggests that elevated cellular ROS can cause alterations of the cardiac sodium channel (Na(v)1.5), abnormal Ca(2+) handling, changes of mitochondrial function, and gap junction remodeling, leading to arrhythmogenesis. This review summarizes our knowledge of the mechanisms by which ROS may cause arrhythmias and discusses potential therapeutic strategies to prevent arrhythmias by targeting ROS and its consequences. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Subject(s)
Arrhythmias, Cardiac/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Calcium/metabolism , Humans , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Oxidation-Reduction/drug effects , Potassium Channels/metabolism
19.
Circulation ; 124(10): 1124-31, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21859973

ABSTRACT

BACKGROUND: Human heart failure is associated with decreased cardiac voltage-gated Na+ channel current (encoded by SCN5A), and the changes have been implicated in the increased risk of sudden death in heart failure. Nevertheless, the mechanism of SCN5A downregulation is unclear. A number of human diseases are associated with alternative mRNA splicing, which has received comparatively little attention in the study of cardiac disease. Splicing factor expression profiles during human heart failure and a specific splicing pathway for SCN5A regulation were explored in this study. METHODS AND RESULTS: Gene array comparisons between normal human and heart failure tissues demonstrated that 17 splicing factors, associated with all major spliceosome components, were upregulated. Two of these splicing factors, RBM25 and LUC7L3, were elevated in human heart failure tissue and mediated truncation of SCN5A mRNA in both Jurkat cells and human embryonic stem cell-derived cardiomyocytes. RBM25/LUC7L3-mediated abnormal SCN5A mRNA splicing reduced Na+ channel current 91.1±9.3% to a range known to cause sudden death. Overexpression of either splicing factor resulted in an increase in truncated mRNA and a concomitant decrease in the full-length SCN5A transcript. CONCLUSIONS: Of the 17 mRNA splicing factors upregulated in heart failure, RBM25 and LUC7L3 were sufficient to explain the increase in truncated forms and the reduction in full-length Na+ channel transcript. Because the reduction in channels was in the range known to be associated with sudden death, interruption of this abnormal mRNA processing may reduce arrhythmic risk in heart failure.


Subject(s)
Heart Failure/genetics , RNA Splicing , RNA-Binding Proteins/metabolism , Sodium Channels/genetics , Adult , Aged , Cells, Cultured , Down-Regulation , Embryonic Stem Cells/metabolism , Female , Gene Expression Profiling , Humans , Jurkat Cells , Male , Middle Aged , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Nuclear Proteins , Spliceosomes/metabolism , Up-Regulation , Young Adult
20.
Am J Physiol Heart Circ Physiol ; 301(3): H824-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21724869

ABSTRACT

Diastolic heart failure is a major cause of mortality in the elderly population. It is often preceded by diastolic dysfunction, which is characterized by impaired active relaxation and increased stiffness. We tested the hypothesis that senescence-prone (SAMP8) mice would develop diastolic dysfunction compared with senescence-resistant controls (SAMR1). Pulsed-wave Doppler imaging of the ratio of blood flow velocity through the mitral valve during early (E) vs. late (A) diastole was reduced from 1.3 ± 0.03 in SAMR1 mice to 1.2 ± 0.03 in SAMP8 mice (P < 0.05). Tissue Doppler imaging of the early (E') and late (A') diastolic mitral annulus velocities found E' reduced from 25.7 ± 0.9 mm/s in SAMR1 to 21.1 ± 0.8 mm/s in SAMP8 mice and E'/A' similarly reduced from 1.1 ± 0.02 to 0.8 ± 0.03 in SAMR1 vs. SAMP8 mice, respectively (P < 0.05). Invasive hemodynamics revealed an increased slope of the end-diastolic pressure-volume relationship (0.5 ± 0.05 vs. 0.8 ± 0.14; P < 0.05), indicating increased left ventricular chamber stiffness. There were no differences in systolic function or mean arterial pressure; however, diastolic dysfunction was accompanied by increased fibrosis in the hearts of SAMP8 mice. In SAMR1 vs. SAMP8 mice, interstitial collagen area increased from 0.3 ± 0.04 to 0.8 ± 0.09% and perivascular collagen area increased from 1.0 ± 0.11 to 1.6 ± 0.14%. Transforming growth factor-ß and connective tissue growth factor gene expression were increased in the hearts of SAMP8 mice (P < 0.05 for all data). In summary, SAMP8 mice show increased fibrosis and diastolic dysfunction similar to those seen in humans with aging and may represent a suitable model for future mechanistic studies.


Subject(s)
Aging, Premature/complications , Aging , Heart Failure/etiology , Myocardium/pathology , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left , Age Factors , Aging/genetics , Aging, Premature/genetics , Analysis of Variance , Animals , Cardiac Catheterization , Collagen/genetics , Connective Tissue Growth Factor/genetics , Diastole , Disease Models, Animal , Echocardiography, Doppler, Pulsed , Elasticity , Fibrosis , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics , Mice , Mice, Transgenic , Myocardium/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/genetics , Ventricular Pressure
SELECTION OF CITATIONS
SEARCH DETAIL
...