Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
2.
J Am Coll Cardiol ; 63(24): 2734-41, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24681145

ABSTRACT

OBJECTIVES: This study sought to investigate the effect of endothelial dysfunction on the development of cardiac hypertrophy and fibrosis. BACKGROUND: Endothelial dysfunction accompanies cardiac hypertrophy and fibrosis, but its contribution to these conditions is unclear. Increased nicotinamide adenine dinucleotide phosphate oxidase-2 (NOX2) activation causes endothelial dysfunction. METHODS: Transgenic mice with endothelial-specific NOX2 overexpression (TG mice) and wild-type littermates received long-term angiotensin II (AngII) infusion (1.1 mg/kg/day, 2 weeks) to induce hypertrophy and fibrosis. RESULTS: TG mice had systolic hypertension and hypertrophy similar to those seen in wild-type mice but developed greater cardiac fibrosis and evidence of isolated left ventricular diastolic dysfunction (p < 0.05). TG myocardium had more inflammatory cells and VCAM-1-positive vessels than did wild-type myocardium after AngII treatment (both p < 0.05). TG microvascular endothelial cells (ECs) treated with AngII recruited 2-fold more leukocytes than did wild-type ECs in an in vitro adhesion assay (p < 0.05). However, inflammatory cell NOX2 per se was not essential for the profibrotic effects of AngII. TG showed a higher level of endothelial-mesenchymal transition (EMT) than did wild-type mice after AngII infusion. In cultured ECs treated with AngII, NOX2 enhanced EMT as assessed by the relative expression of fibroblast versus endothelial-specific markers. CONCLUSIONS: AngII-induced endothelial NOX2 activation has profound profibrotic effects in the heart in vivo that lead to a diastolic dysfunction phenotype. Endothelial NOX2 enhances EMT and has proinflammatory effects. This may be an important mechanism underlying cardiac fibrosis and diastolic dysfunction during increased renin-angiotensin activation.


Subject(s)
Cardiomegaly/enzymology , Endothelium, Vascular/enzymology , Inflammation Mediators/physiology , Membrane Glycoproteins/physiology , Mesenchymal Stem Cells/enzymology , NADPH Oxidases/physiology , Ventricular Dysfunction, Left/enzymology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Endothelium, Vascular/pathology , Fibrosis/enzymology , Fibrosis/genetics , Fibrosis/pathology , Heart Failure, Diastolic/enzymology , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/pathology , Humans , Male , Membrane Glycoproteins/genetics , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
3.
Cardiovasc Res ; 103(2): 268-80, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24692173

ABSTRACT

AIMS: Tissue inhibitor of metalloproteinases (TIMPs) can mediate myocardial remodelling, hypertrophy, and fibrosis in heart disease. We investigated the impact of TIMP2 vs. TIMP3 deficiency in angiotensin II (Ang II)-induced myocardial remodelling and cardiac dysfunction. METHODS AND RESULTS: TIMP2(-/-), TIMP3(-/-), and wild-type (WT) mice received Ang II/saline (Alzet pump) for 2 weeks. Ang II infusion resulted in enhanced myocardial hypertrophy and lack of fibrosis in TIMP2(-/-), and conversely, excess fibrosis without hypertrophy in TIMP3(-/-) mice. Echocardiographic imaging revealed preserved ejection fraction in all groups; however, exacerbated left ventricular (LV) diastolic dysfunction was detected in Ang II-infused TIMP2(-/-) and TIMP3(-/-) mice, despite the suppressed Ang II-induced hypertension in TIMP3(-/-) mice. Enhanced hypertrophy in TIMP2(-/-) mice impaired active relaxation, while excess fibrosis in TIMP3(-/-) mice increased LV passive stiffness. Adult WT cardiomyocytes, only when co-cultured with cardiac fibroblasts, exhibited Ang II-induced hypertrophy which was suppressed in TIMP3(-/-) cardiomyocytes. In vitro studies on adult cardiofibroblasts (quiescent and cyclically stretched), and in vivo analyses, revealed that the increased fibrosis in TIMP3(-/-)-Ang II hearts is due to post-translational stabilization and deposition of collagen by matricellular proteins [osteopontin and Secreted Protein Acidic and Rich in Cysteine (SPARC)], which correlated with increased inflammation, rather than increased de novo synthesis. Reduced cross-linking enzymes, LOX and PLOD1, could underlie suppressed collagen deposition in TIMP2(-/-)-Ang II hearts. CONCLUSION: TIMP2 and TIMP3 play fundamental and differential roles in mediating pathological remodelling, independent from their MMP-inhibitory function. TIMP2(-/-) and TIMP3(-/-) mice provide a unique opportunity to study myocardial hypertrophy and fibrosis independently, and their impact on cardiac dysfunction.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/enzymology , Heart Failure, Diastolic/enzymology , Hypertrophy, Left Ventricular/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiomyopathies/genetics , Fibrosis/genetics , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Ventricular Remodeling/genetics
4.
Biochim Biophys Acta ; 1841(6): 880-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631848

ABSTRACT

In mice with temporally-induced cardiac-specific deficiency of acyl-CoA synthetase-1 (Acsl1(H-/-)), the heart is unable to oxidize long-chain fatty acids and relies primarily on glucose for energy. These metabolic changes result in the development of both a spontaneous cardiac hypertrophy and increased phosphorylated S6 kinase (S6K), a substrate of the mechanistic target of rapamycin, mTOR. Doppler echocardiography revealed evidence of significant diastolic dysfunction, indicated by a reduced E/A ratio and increased mean performance index, although the deceleration time and the expression of sarco/endoplasmic reticulum calcium ATPase and phospholamban showed no difference between genotypes. To determine the role of mTOR in the development of cardiac hypertrophy, we treated Acsl1(H-/-) mice with rapamycin. Six to eight week old Acsl1(H-/-) mice and their littermate controls were given i.p. tamoxifen to eliminate cardiac Acsl1, then concomitantly treated for 10weeks with i.p. rapamycin or vehicle alone. Rapamycin completely blocked the enhanced ventricular S6K phosphorylation and cardiac hypertrophy and attenuated the expression of hypertrophy-associated fetal genes, including α-skeletal actin and B-type natriuretic peptide. mTOR activation of the related Acsl3 gene, usually associated with pathologic hypertrophy, was also attenuated in the Acsl1(H-/-) hearts, indicating that alternative pathways of fatty acid activation did not compensate for the loss of Acsl1. Compared to controls, Acsl1(H-/-) hearts exhibited an 8-fold higher uptake of 2-deoxy[1-(14)C]glucose and a 35% lower uptake of the fatty acid analog 2-bromo[1-(14)C]palmitate. These data indicate that Acsl1-deficiency causes diastolic dysfunction and that mTOR activation is linked to the development of cardiac hypertrophy in Acsl1(H-/-) mice.


Subject(s)
Cardiomegaly/enzymology , Coenzyme A Ligases/genetics , Heart Failure, Diastolic/enzymology , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/genetics , Animals , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Coenzyme A Ligases/deficiency , Coenzyme A Ligases/metabolism , Endoplasmic Reticulum/metabolism , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/pathology , Humans , Lipid Metabolism/genetics , Mice , Oxidation-Reduction , Tamoxifen/pharmacology
5.
J UOEH ; 35(2): 147-58, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23774658

ABSTRACT

Nitric oxide (NO) is synthesized by three distinct NO synthase (NOS) isoforms (neuronal, inducible, and endothelial NOS), all of which are expressed in the human heart. The roles of NOSs in the pathogenesis of heart failure have been described in pharmacological studies with NOS inhibitors. Recently, genetically engineered animals have been used. We have generated mice in which all 3 NOS isoforms are completely disrupted (triple n/i/eNOS(-/-) mice). Morphological, echocardiographic, and hemodynamic analysis were performed in wild-type, singly nNOS(-/-), iNOS(-/-), eNOS(-/-), and triple n/i/eNOS(-/-) mice. Importantly, significant left ventricular (LV) hypertrophy and diastolic dysfunction was noted only in n/i/eNOS(-/-) mice, and those pathology was similar to diastolic heart failure in humans. Finally, treatment with an angiotensin II type 1 (AT1) receptor blocker, significantly prevented those abnormalities. These results provide the evidence that AT1 receptor pathway plays a center role in the pathogenesis of cardiac disorders in the n/i/eNOS(-/-) mice. Our studies with triple n/i/eNOS(-/-) mice provide pivotal insights into an understanding of the pathophysiology of NOSs in human heart failure.


Subject(s)
Heart Failure/enzymology , Nitric Oxide Synthase/physiology , Animals , Disease Models, Animal , Heart Failure, Diastolic/enzymology , Mice , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I/physiology , Nitric Oxide Synthase Type II/physiology , Nitric Oxide Synthase Type III/physiology
7.
Basic Res Cardiol ; 106(2): 263-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21174213

ABSTRACT

Transgenic (TG) Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) δ(C) mice develop systolic heart failure (HF). CaMKII regulates intracellular Ca(2+) handling proteins as well as sarcolemmal Na(+) channels. We hypothesized that CaMKII also contributes to diastolic dysfunction and arrhythmias via augmentation of the late Na(+) current (late I(Na)) in early HF (8-week-old TG mice). Echocardiography revealed severe diastolic dysfunction in addition to decreased systolic ejection fraction. Premature arrhythmogenic contractions (PACs) in isolated isometrically twitching papillary muscles only occurred in TG preparations (5 vs. 0, P < 0.05) which could be completely terminated when treated with the late I(Na) inhibitor ranolazine (Ran, 5 µmol/L). Force-frequency relationships revealed significantly reduced twitch force amplitudes in TG papillary muscles. Most importantly, diastolic tension increased with raising frequencies to a greater extent in TG papillary muscles compared to WT specimen (at 10 Hz: 3.7 ± 0.4 vs. 2.5 ± 0.3 mN/mm²; P < 0.05). Addition of Ran improved diastolic dysfunction to 2.1 ± 0.2 mN/mm² (at 10 Hz; P < 0.05) without negative inotropic effects. Mechanistically, the late I(Na) was markedly elevated in myocytes isolated from TG mice and could be completely reversed by Ran. In conclusion, our results show for the first time that TG CaMKIIδ(C) overexpression induces diastolic dysfunction and arrhythmogenic triggers possibly via an enhanced late I(Na). Inhibition of elevated late I(Na) had beneficial effects on arrhythmias as well as diastolic function in papillary muscles from CaMKIIδ(C) TG mice. Thus, late I(Na) inhibition appears to be a promising option for diastolic dysfunction and arrhythmias in HF where CaMKII is found to be increased.


Subject(s)
Arrhythmias, Cardiac/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure, Diastolic/enzymology , Sodium/metabolism , Animals , Calcium/metabolism , Heart Failure, Diastolic/pathology , Heart Failure, Diastolic/physiopathology , Mice , Mice, Transgenic , Myocardial Contraction , Myocardium/pathology , Papillary Muscles/physiopathology , Phenotype , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism
8.
Heart Fail Rev ; 15(5): 431-45, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20419345

ABSTRACT

Hypertension, atherosclerosis, and resultant chronic heart failure (HF) reach epidemic proportions among older persons, and the clinical manifestations and the prognoses of these worsen with increasing age. Thus, age per se is the major risk factor for cardiovascular disease. Changes in cardiac cell phenotype that occur with normal aging, as well as in HF associated with aging, include deficits in ss-adrenergic receptor (ss-AR) signaling, increased generation of reactive oxygen species (ROS), and altered excitation-contraction (EC) coupling that involves prolongation of the action potential (AP), intracellular Ca(2+) (Ca(i)(2+)) transient and contraction, and blunted force- and relaxation-frequency responses. Evidence suggests that altered sarcoplasmic reticulum (SR) Ca(2+) uptake, storage, and release play central role in these changes, which also involve sarcolemmal L-type Ca(2+) channel (LCC), Na(+)-Ca(2+) exchanger (NCX), and K(+) channels. We review the age-associated changes in the expression and function of Ca(2+) transporting proteins, and functional consequences of these changes at the cardiac myocyte and organ levels. We also review sexual dimorphism and self-renewal of the heart in the context of cardiac aging and HF.


Subject(s)
Aging , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/physiopathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/enzymology , Adaptation, Physiological , Age Factors , Aged , Aged, 80 and over , Disease Progression , Female , Heart Failure, Diastolic/enzymology , Heart Failure, Diastolic/epidemiology , Heart Failure, Systolic/enzymology , Heart Failure, Systolic/epidemiology , Humans , Male , Muscle Cells/pathology , Prognosis , Receptors, Adrenergic, beta/biosynthesis , Risk Factors , Sarcoplasmic Reticulum/pathology , Sex Factors , Signal Transduction , Stress, Physiological , United States/epidemiology
9.
Cardiovasc Toxicol ; 9(3): 142-50, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19644660

ABSTRACT

HIV infection in patients is associated with a surprisingly high frequency of diastolic dysfunction followed by the development of a dilated cardiomyopathy. Potential mechanisms include direct effects of HIV proteins, including gp120. We have previously reported direct inotropic and p38 MAP kinase signaling effects of HIV gp120 on isolated cardiac myocytes in vitro. We now report effects of a single injection of HIV gp120 on cardiac hemodynamics in vivo. HIV gp120 (50 microg/kg) was injected intravenously and hemodynamics assessed at 1, 24, 48 and 72 h in freely ambulatory, awake rats. Rats injected with gp120 demonstrated a blunted diastolic response to increasing intravenous (IV) injections of the beta-adrenergic agonist, isoproterenol (ISO), compared with similarly instrumented controls at 48 h (gp120 vs. control, P < 0.01; n = 6, for each). Pre-treatment with the p38 MAP kinase inhibitor, SB 203580, prevented the diastolic dysfunction (gp120 + SB vs. control, P = NS; n = 6, for each). Hemodynamic changes correlated with inhibition of both p38 MAP kinase and troponin I phosphorylation by SB in vivo. There were no differences in ISO dose-response curves between gp120-treated and control rats at 1, 24 or 72 h (P = NS; n = 6, for each). Thus, evidence of diastolic dysfunction is apparent in vivo in rats following a single dose of HIV gp120. To our knowledge, this is the first report of a hemodynamic effect of an HIV protein in vivo. These findings lend further support to the hypothesis that the HIV virus, itself, may contribute to myocardial dysfunction in patients infected with HIV via a p38 MAP kinase mechanism.


Subject(s)
Cardiomyopathies/enzymology , HIV Envelope Protein gp120/toxicity , Heart Failure, Diastolic/enzymology , Imidazoles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Dose-Response Relationship, Drug , HIV Envelope Protein gp120/administration & dosage , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/prevention & control , Imidazoles/pharmacology , Male , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/physiology
10.
Basic Res Cardiol ; 104(5): 499-509, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19255799

ABSTRACT

BACKGROUND: Diastolic heart failure is a rising problem with a high incidence and similar mortality and morbidity compared to patients with systolic heart failure. Nevertheless, the underlying pathophysiology is still debated. AIM: We investigated the effect of pharmacological enhancement of endothelial nitric oxide synthase (eNOS) on experimental diastolic heart failure (DHF). METHODS: DHF was induced in 60 DAHL salt-sensitive rats by salt diet in 8-week-old animals. 30 were treated with the eNOS enhancer AVE3085 (DHFeNOS) and 30 with placebo (DHF). Rats with normal salt intake served as controls. RESULTS AND CONCLUSION: Diastolic dysfunction with increased diastolic stiffness constant and increased left ventricular (LV) pressure was analyzed by invasive pressure-volume loop measurements in the DHF group compared to controls. Cardiac hypertrophy as indicated by LV mass measurements by echocardiography, and increased cardiac collagen content as measured by immunohistochemistry were associated with an increased activation state of calcineurin, AKT, ERK(1/2), but not JNK and p38 kinases. Titin isoforms were not altered in this model of DHF. Treatment with AVE3085 significantly increased eNOS mRNA and protein levels in the cardiac tissue and decreases NAD(P)H oxidase subunits p22phox and gp91phox. Diastolic dysfunction was attenuated and cardiac hypertrophy and fibrosis were improved in comparison with untreated DHF animals. This was associated with a normalized activation state of calcineurin, AKT and ERK(1/2). Therefore, we suggest that targeting the NO system might yield a future therapeutic aim for the treatment of DHF.


Subject(s)
Benzodioxoles/pharmacology , Cardiotonic Agents/pharmacology , Heart Failure, Diastolic/prevention & control , Hypertension/drug therapy , Indans/pharmacology , Myocardium/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Ventricular Function, Left/drug effects , Animals , Blood Pressure/drug effects , Calcineurin/metabolism , Cardiomegaly/enzymology , Cardiomegaly/etiology , Cardiomegaly/prevention & control , Collagen/metabolism , Disease Models, Animal , Fibrosis , Heart Failure, Diastolic/enzymology , Heart Failure, Diastolic/etiology , Heart Failure, Diastolic/physiopathology , Hypertension/complications , Hypertension/enzymology , Hypertension/etiology , Hypertension/physiopathology , Male , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/pathology , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/genetics , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Signal Transduction/drug effects , Sodium Chloride , Time Factors , Transcription, Genetic/drug effects , Up-Regulation , Ventricular Pressure/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...