Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Circ Heart Fail ; 4(5): 651-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21685249

ABSTRACT

BACKGROUND: Extensive evidence implicates aldosterone excess in the development and progression of cardiovascular disease states including hypertension, metabolic syndrome, cardiac hypertrophy, heart failure, and cardiorenal fibrosis. Recent studies show that activation of inflammatory cascade may play a specific role in the sequelae of mineralocorticoid activation, although the linking mechanism remains unclear. We tested the possibility that secondary stimulation of the stromal-derived factor 1/CXC chemokine receptor 4 (SDF-1/CXCR4) pathway plays a contributory role. METHODS AND RESULTS: We investigated the effect of the highly selective CXCR4 antagonist AMD3465 (6 mg/kg per day for 6 weeks through minipump) in dexoycorticosterone acetate (DOCA)-treated, uninephrectomized mice. CXCR4 antagonism significantly attenuated the induction of cardiac fibrosis, renal fibrosis, hypertension, and left ventricular hypertrophy by DOCA. Mineralocorticoid excess also stimulated the accumulation of T-lymphocytes in the heart and kidney and this was significantly blunted by CXCR4 inhibition. CONCLUSIONS: Taken together, these data strongly implicate the SDF-1/CXCR4 axis in the pathogenesis of mineralocorticoid excess induced hypertension, inflammation, and cardiorenal fibrosis. This insight provides a new potential therapeutic approach for the treatment of specific aspects of mineralocorticoid mediated cardiovascular disease.


Subject(s)
Desoxycorticosterone/adverse effects , Hypertension/prevention & control , Hypertrophy, Left Ventricular/prevention & control , Kidney/pathology , Mineralocorticoids/adverse effects , Myocardium/pathology , Receptors, CXCR4/antagonists & inhibitors , Animals , Chemokine CXCL12/metabolism , Desoxycorticosterone/pharmacology , Disease Models, Animal , Fibrosis , Heart/drug effects , Hypertension/etiology , Hypertension/metabolism , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mineralocorticoids/pharmacology , Myocardium/metabolism , Pyridines/pharmacology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
2.
Am J Pathol ; 176(4): 1735-42, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20150435

ABSTRACT

Cardiac fibrosis contributes significantly to the phenotype of the chronically failing heart. It is not clear whether in this setting the fibrosis is contributed by native cardiac fibroblasts or alternatively by recruitment of cells arising from the bone marrow. We aimed to determine the contribution of bone marrow-derived cells to cardiac fibrosis in the failing heart and to investigate potentially contributing cytokines. Bone marrow-derived fibrocyte recruitment to the failing heart was studied in a transgenic (Mst1 mice) model of dilated cardiomyopathy. In conjunction, we examined the role of stromal-derived factor-1 (SDF-1), a key chemoattractant, by assessing myocardial expression and secretion by cardiomyocytes and in clinical samples. Bone marrow-derived cells were recruited in significantly greater numbers in Mst1 versus control mice (P < 0.001), contributing 17 +/- 4% of the total fibroblast load in heart failure. Patients with heart failure had higher plasma levels of SDF-1 than healthy control subjects (P < 0.01). We found that cardiomyocytes constitutively secrete SDF-1, which is significantly up-regulated by angiotensin II. SDF-1 was shown to increases cardiac fibroblast migration by 59% (P < 0.05). Taken together, our data suggest that recruitment of bone marrow-derived cells under the influence of factors, including SDF-1, may play an important role in the pathogenesis of cardiac fibrosis in heart failure.


Subject(s)
Bone Marrow Cells/cytology , Fibroblasts/cytology , Fibrosis/pathology , Heart Failure/pathology , Animals , Cardiomyopathy, Dilated/pathology , Cell Transplantation , Chemokine CXCL12/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Rats , Rats, Sprague-Dawley
3.
J Cell Biochem ; 108(1): 156-68, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19544385

ABSTRACT

Myocardial injury due to ischemia-reperfusion (I-R) damage remains a major clinical challenge. Its pathogenesis is complex including endothelial dysfunction and heightened oxidative stress although the key driving mechanism remains uncertain. In this study we tested the hypothesis that the I-R process induces a state of insufficient L-arginine availability for NO biosynthesis, and that this is pivotal in the development of myocardial I-R damage. In neonatal rat ventricular cardiomyocytes (NVCM), hypoxia-reoxygenation significantly decreased L-arginine uptake and NO production (42 +/- 2% and 71 +/- 4%, respectively, both P < 0.01), maximal after 2 h reoxygenation. In parallel, mitochondrial membrane potential significantly decreased and ROS production increased (both P < 0.01). NVCMs infected with adenovirus expressing the L-arginine transporter, CAT1, and NVCMs supplemented with L-arginine both exhibited significant (all P < 0.05) improvements in NO generation and mitochondrial membrane potentials, with a concomitant significant fall in ROS production and lactate dehydrogenase release during hypoxia-reoxygenation. In contrast, L-arginine deprived NVCM had significantly worsened responses to hypoxia-reoxygenation. In isolated perfused mouse hearts, L-arginine infusion during reperfusion significantly improved left ventricular function after I-R. These improved contractile responses were not dependent on coronary flow but were associated with a significant decrease in nitrotyrosine formation and increases in phosphorylation of both Akt and troponin I. Collectively, these data strongly implicate reduced L-arginine availability as a key factor in the pathogenesis of I-R injury. Increasing L-arginine availability via increased CAT1 expression or by supplementation improves myocardial responses to I-R. Restoration of L-arginine availability may therefore be a valuable strategy to ameliorate I-R injury.


Subject(s)
Arginine/metabolism , Myocardial Reperfusion Injury/etiology , Animals , Mice , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Oxygen Consumption , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transfection
4.
J Mol Cell Cardiol ; 46(1): 86-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18977358

ABSTRACT

The endothelium plays a vital role in the maintenance of vascular tone and structural vascular integrity, principally mediated via the actions of nitric oxide (NO). L-arginine is the immediate substrate for NO synthesis, and the availability of extracellular L-arginine is critical for the production of NO. Activation of protein kinase C (PKC) dependent signalling pathways are a feature of a number of cardiovascular disease states, and in this study we aimed to systematically evaluate the mechanism by which PKC regulates L-arginine transport in endothelial cells. In response to PKC activation (PMA 100 nM, 30 min), [(3)H]L-arginine uptake by bovine aortic endothelial cells (BAEC) was reduced to 45+4% of control (p<0.05). This resulted from a 53% reduction in the Vmax (p<0.05), with no change in the K(m) for L-arginine. Western blot analysis and confocal microscopy revealed no change in the expression or membrane distribution of CAT-1, the principal BAEC L-arginine transporter. Moreover in (32)P-labeling studies, PMA exposure did not result in CAT-1 phosphorylation. We therefore explored the possibility that PKC altered and interaction with MARCKS protein, a candidate membrane associated protein. By co-immunoprecipitation we show that CAT-1 interacts with, a membrane associated protein, that was significantly inhibited by PKC activation (p<0.05). Moreover antisense inhibition of MARCKS abolished the PMA effect on L-arginine transport. PKC dependent mechanisms regulate the transport of L-arginine, mediated via process involving MARCKS.


Subject(s)
Arginine/metabolism , Cationic Amino Acid Transporter 1/metabolism , Endothelium/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Protein Kinase C/metabolism , Animals , Arginine/chemistry , Biological Transport , Cattle , Cell Membrane/metabolism , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Membrane Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Nitric Oxide Synthase Type III/metabolism , Oligonucleotides, Antisense/chemistry , Phosphorylation
5.
J Am Coll Cardiol ; 50(3): 253-60, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17631218

ABSTRACT

OBJECTIVES: The purpose of this study was to develop a clinically applicable high-efficiency percutaneous means of therapeutic gene delivery to the failing heart. BACKGROUND: Substantial advances in the understanding of the cellular and molecular basis of heart failure (HF) have recently fostered interest in the potential utility of gene and cell therapy as novel therapeutic approaches. However, successful clinical translation is currently limited by the lack of safe, efficient, and selective delivery systems. METHODS: We developed a novel percutaneous closed-loop recirculatory system that provides homogeneous myocardial delivery for gene transfer in the failing large animal heart. After 4 weeks' rapid pacing in adult sheep to induce HF, the animals were randomly allocated to receive either adenovirus expressing a pseudophosphorylated mutant (AdS16E) of phospholamban (PLN) or Ad-beta-galactosidase (AdLacZ). RESULTS: Two weeks after gene delivery, in the presence of continued pacing, left ventricular (LV) ejection fraction had significantly improved in the AdS16E-treated animals (27 +/- 3% to 50 +/- 4%; p < 0.001), whereas a further decline occurred in the AdLacZ group (34 +/- 4% to 27 +/- 3%; p < 0.05). In conjunction, AdS16E delivery resulted in significant reductions in LV filling pressures and end-diastolic diameter (both p < 0.05). In conjunction, AdS16E-treated animals showed significant improvement in the expression of PLN and Ca2+-adenosine triphosphatase activity. In separate animals, recirculating AdLacZ delivery was shown to achieve superior myocardial gene expression in contrast to intracoronary delivery and was associated with lower systemic expression. CONCLUSIONS: We report the development of a novel closed-loop system for cardiac gene therapy. Using this approach delivery of AdS16E reversed HF progression in a large animal HF model.


Subject(s)
Calcium-Binding Proteins/pharmacology , Gene Transfer Techniques , Genetic Therapy/methods , Heart Failure/therapy , Myocardial Revascularization/methods , Animals , Disease Models, Animal , Genetic Vectors , Heart Failure/physiopathology , Probability , Random Allocation , Risk Assessment , Sensitivity and Specificity , Sheep , Statistics, Nonparametric
6.
Cardiovasc Res ; 72(1): 112-23, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16890211

ABSTRACT

OBJECTIVE: Reactive oxygen species (ROS) such as superoxide have been linked to the hypertrophic response of the heart to stimuli including angiotensin II (AngII), mechanical stretch, and pressure overload. We have previously demonstrated that cGMP and protein kinase G mediate the antihypertrophic actions of the natriuretic peptides in rat cardiomyocytes and isolated whole hearts. The impact of natriuretic peptides on cardiac ROS generation, however, has not been investigated. We tested the hypothesis that reduced superoxide accumulation contributes to the antihypertrophic action of atrial natriuretic peptide (ANP). METHODS: Neonatal rat cardiomyocytes were cultured in serum-free medium with and without AngII (1 micromol/L) or endothelin-1 (ET(1), 60 nmol/L) in the presence and absence of ANP (1 micromol/L) or tempol (100 micromol/L). Hypertrophic responses, cardiomyocyte superoxide generation, and cardiomyocyte expression of NADPH oxidase were determined. RESULTS: AngII induced increases in cardiomyocyte size (to 176 +/- 9% n = 8 p < 0.001, at 48 h), beta-myosin heavy chain expression (to 4.0 +/- 1.6-fold n = 6 p < 0.05, at 48 h), c-fos expression (to 1.9 +/- 0.5-fold n = 7 p < 0.01, at 6 h), superoxide generation (to 181+/-21% n = 8 p < 0.005, at 24 h), and expression of the gp91phox subunit of NADPH oxidase (to 2.4 +/- 0.5-fold n = 7 p < 0.05, at 48 h). These effects were all significantly inhibited by ANP: cardiomyocyte size, beta-myosin heavy chain expression, c-fos expression, superoxide generation and gp91phox expression were reduced to 107 +/- 5% (n = 5 p < 0.05), 1.2 +/- 0.2-fold (n = 6 p < 0.05), 0.9 +/- 0.2-fold (n = 7 p < 0.05), 141 +/- 21% (n = 8 p < 0.05), and to 1.0 +/- 0.5-fold (n = 7 p < 0.05), respectively. These effects were mimicked by tempol. ANP and tempol also significantly inhibited ET1-induced increases in cardiomyocyte size and superoxide generation, but had no effect on markers of hypertrophy when studied alone. CONCLUSION: This data indicates that the antihypertrophic actions of ANP are accompanied by reduced levels of superoxide, suggesting an antioxidant action contributes to the antihypertrophic actions of ANP.


Subject(s)
Antioxidants/pharmacology , Atrial Natriuretic Factor/pharmacology , Myocytes, Cardiac/metabolism , Angiotensin II/pharmacology , Animals , Animals, Newborn , Cell Enlargement/drug effects , Cells, Cultured , Cyclic N-Oxides/pharmacology , Endothelin-1/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Genes, fos , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , NADPH Oxidases/genetics , Rats , Rats, Sprague-Dawley , Spin Labels , Superoxides/analysis
7.
Pacing Clin Electrophysiol ; 29(8): 821-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16922997

ABSTRACT

OBJECTIVE: In this study, we aimed to compare the level of atrial sympathetic innervation in human atrial fibrillation (AF) to that in sinus rhythm (SR). BACKGROUND: Histological studies of atrial tissue obtained from animals with experimentally induced AF indicate that sympathetic hyperinnervation could play a role in the pathogenesis of AF. METHODS: In 24 patients (12 in SR and 12 in AF) undergoing bypass surgery, we collected right atrial appendage tissue. In AF patients, left atrial appendage tissue was also acquired. The degree of sympathetic innervation was quantified by measuring the amount of staining for tyrosine hydroxylase (TH) and tissue norepinephrine (NE). In conjunction, nerve growth factor (NGF) mRNA expression was assessed by real-time polymerase chain reaction (PCR). Growth-associated protein 43 (GAP43) immunostaining was used to assess degree of new neural growth. RESULTS: When corrected for differences in tissue fibrosis, the expression of both TH (AF 0.45 +/- 0.1%, SR 0.09 +/- 0.03%, P = 0.02) and tissue NE (AF 358 +/- 49 pg/mg, SR 225 +/- 39 pg/mg, P = 0.04) was greater in atrial tissue of the AF cohort. The degree of atrial TH staining (P = 0.01) and NE content (P < 0.001) was also significantly greater in the right compared with left atrial samples in the AF cohort. There were no differences in NGF mRNA expression or GAP43 staining. CONCLUSION: This study provides evidence for the presence of heightened atrial sympathetic innervation in patients with persistent AF, suggesting autonomic remodeling may be part of atrial substrate for AF.


Subject(s)
Atrial Fibrillation/pathology , Heart Atria/innervation , Heart Atria/pathology , Heart Conduction System/pathology , Sympathetic Nervous System/pathology , Adaptation, Physiological , Aged , Evidence-Based Medicine , Female , Humans , Male , Middle Aged
8.
Am J Physiol Heart Circ Physiol ; 289(4): H1359-65, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15894570

ABSTRACT

Cardiac-specific overexpression of nerve growth factor (NGF), a neurotrophin, leads to sympathetic hyperinnervation of heart. As a consequence, adverse functional changes that occur after chronically enhanced sympathoadrenergic stimulation of heart might develop in this model. However, NGF also facilitates synaptic transmission and norepinephrine uptake, effects that would be expected to restrain such deleterious outcomes. To test this, we examined 5- to 6-mo-old transgenic (TG) mice that overexpress NGF in heart and their wild-type (WT) littermates using echocardiography, invasive catheterization, histology, and catecholamine assays. In TG mice, hypertrophy of the right ventricle was evident (+67%), but the left ventricle was only mildly affected (+17%). Left ventricular (LV) fractional shortening and fractional area change values as indicated by echocardiography were similar between the two groups. Catheterization experiments revealed that LV +/-dP/dt values were comparable between TG and WT mice and responded similarly upon isoproterenol stimulation, which indicates lack of beta-adrenergic receptor dysfunction. Although norepinephrine levels in TG LV tissue were approximately twofold those of WT tissue, TG plasma levels of the neuronal norepinephrine metabolite dihydroxyphenylglycol were fivefold those of WT plasma. A greater neuronal uptake activity was also observed in TG LV tissue. In conclusion, overexpression of NGF in heart leads to sympathetic hyperinnervation that is not associated with detrimental effects on LV performance and is likely due to concomitantly enhanced norepinephrine neuronal uptake.


Subject(s)
Heart/innervation , Hypertrophy, Right Ventricular/physiopathology , Nerve Growth Factor/genetics , Sympathetic Nervous System/pathology , Ventricular Function, Left , Adrenergic beta-Agonists/pharmacology , Animals , Echocardiography , Female , Gene Expression , Heart/physiology , Hypertrophy, Right Ventricular/diagnostic imaging , Hypertrophy, Right Ventricular/genetics , Isoproterenol/pharmacology , Male , Mice , Mice, Transgenic , Myocardium/pathology , Norepinephrine/blood , Norepinephrine/pharmacokinetics , Sympathetic Nervous System/physiology , Tritium
9.
Circulation ; 110(24): 3680-6, 2004 Dec 14.
Article in English | MEDLINE | ID: mdl-15569830

ABSTRACT

BACKGROUND: Impaired endothelium-dependent NO-mediated vasodilation is a key feature of essential hypertension and may precede the increase in blood pressure. We investigated whether transport of the NO precursor L-arginine is related to decreased endothelial function. METHODS AND RESULTS: Radiotracer kinetics ([3H]L-arginine) were used to measure forearm and peripheral blood mononuclear cell arginine uptake in hypertensive subjects (n=12) and in 2 groups of healthy volunteers with (n=15) and without (n=15) a family history of hypertension. In conjunction, forearm blood flow responses to acetylcholine and sodium nitroprusside were measured before and after a supplemental intra-arterial infusion of L-arginine. In vivo and in vitro measures of L-arginine transport were substantially reduced in the essential hypertension and positive family history groups compared with the negative family history group; however, no difference was detected in peripheral blood mononuclear cell mRNA or protein expression levels for the cationic amino acid transporter CAT-1. Plasma concentrations of L-arginine and N(G),N(G')-dimethylarginine (ADMA) did not differ between groups. L-arginine supplementation improved the response to acetylcholine only in subjects with essential hypertension and positive family history. CONCLUSIONS: Similar to their hypertensive counterparts, normotensive individuals at high risk for the development of hypertension are characterized by impaired L-arginine transport, which may represent the link between a defective L-arginine/NO pathway and the onset of essential hypertension. The observed transport defect is not due to apparent alterations in CAT-1 expression or elevated endogenous ADMA.


Subject(s)
Arginine/metabolism , Endothelium, Vascular/physiopathology , Hypertension/metabolism , Hypertension/physiopathology , Acetylcholine/pharmacology , Adolescent , Adult , Arginine/blood , Arginine/pharmacokinetics , Biological Transport , Cationic Amino Acid Transporter 1/blood , Cationic Amino Acid Transporter 1/genetics , Cell Membrane/metabolism , Forearm , Genetic Predisposition to Disease , Humans , Hypertension/genetics , In Vitro Techniques , Male , Nitric Oxide/biosynthesis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase Type III , Nitroprusside/pharmacology , RNA, Messenger/blood , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...