Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pflugers Arch ; 466(2): 237-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23873354

ABSTRACT

The water channels, aquaporins (AQPs) are key mediators of transcellular fluid transport. However, their expression and role in cardiac tissue is poorly characterized. Particularly, AQP1 was suggested to transport other molecules (nitric oxide (NO), hydrogen peroxide (H2O2)) with potential major bearing on cardiovascular physiology. We therefore examined the expression of all AQPs and the phenotype of AQP1 knockout mice (vs. wild-type littermates) under implanted telemetry in vivo, as well as endothelium-dependent relaxation in isolated aortas and resistance vessels ex vivo. Four aquaporins were expressed in wild-type heart tissue (AQP1, AQP7, AQP4, AQP8) and two aquaporins in aortic and mesenteric vessels (AQP1-AQP7). AQP1 was expressed in endothelial as well as cardiac and vascular muscle cells and co-segregated with caveolin-1. AQP1 knockout (KO) mice exhibited a prominent microcardia and decreased myocyte transverse dimensions despite no change in capillary density. Both male and female AQP1 KO mice had lower mean BP, which was not attributable to altered water balance or autonomic dysfunction (from baroreflex and frequency analysis of BP and HR variability). NO-dependent BP variability was unperturbed. Accordingly, endothelium-derived hyperpolarizing factor (EDH(F)) or NO-dependent relaxation were unchanged in aorta or resistance vessels ex vivo. However, AQP1 KO mesenteric vessels exhibited an increase in endothelial prostanoids-dependent relaxation, together with increased expression of COX-2. This enhanced relaxation was abrogated by COX inhibition. We conclude that AQP1 does not regulate the endothelial EDH or NO-dependent relaxation ex vivo or in vivo, but its deletion decreases baseline BP together with increased prostanoids-dependent relaxation in resistance vessels. Strikingly, this was associated with microcardia, unrelated to perturbed angiogenesis. This may raise interest for new inhibitors of AQP1 and their use to treat hypertrophic cardiac remodeling.


Subject(s)
Aquaporin 1/deficiency , Blood Pressure/physiology , Animals , Aquaporin 1/physiology , Biological Factors/physiology , Female , Heart Defects, Congenital/pathology , Hypotension/physiopathology , Male , Mice , Mice, Knockout , Myocardial Contraction/physiology , Nitric Oxide/physiology
4.
Nat Cell Biol ; 3(10): 867-73, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11584267

ABSTRACT

Stretching of cardiac muscle modulates contraction through the enhancement of the Ca2+ transient, but how this occurs is still not known. We found that stretching of myocytes modulates the elementary Ca2+ release process from ryanodine-receptor Ca2+-release channels (RyRCs), Ca2+ sparks and the electrically stimulated Ca2+ transient. Stretching induces PtdIns-3-OH kinase (PI(3)K)-dependent phosphorylation of both Akt and the endothelial isoform of nitric oxide synthase (NOS), nitric oxide (NO) production, and a proportionate increase in Ca2+-spark frequency that is abolished by inhibiting NOS and PI(3)K. Exogenously generated NO reversibly increases Ca2+-spark frequency without cell stretching. We propose that myocyte NO produced by activation of the PI(3)K-Akt-endothelial NOS axis acts as a second messenger of stretch by enhancing RyRC activity, contributing to myocardial contractile activation.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases , Animals , Electric Stimulation , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/metabolism , Male , Myocardial Contraction/drug effects , Myocardium/cytology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , S-Nitroso-N-Acetylpenicillamine/pharmacology , Spectrometry, Fluorescence
5.
Circulation ; 117(8): 1065-74, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18268148

ABSTRACT

BACKGROUND: In endothelial cells, caveolin-1, the structural protein of caveolae, acts as a scaffolding protein to cluster lipids and signaling molecules within caveolae and, in some instances, regulates the activity of proteins targeted to caveolae. Specifically, different putative mediators of the endothelium-derived hyperpolarizing factor (EDHF)-mediated relaxation are located in caveolae and/or regulated by the structural protein caveolin-1, such as potassium channels, calcium regulatory proteins, and connexin 43, a molecular component of gap junctions. METHODS AND RESULTS: Comparing relaxation in vessels from caveolin-1 knockout mice and their wild-type littermates, we observed a complete absence of EDHF-mediated vasodilation in isolated mesenteric arteries from caveolin-1 knockout mice. The absence of caveolin-1 is associated with an impairment of calcium homeostasis in endothelial cells, notably, a decreased activity of Ca2+-permeable TRPV4 cation channels that participate in nitric oxide- and EDHF-mediated relaxation. Moreover, morphological characterization of caveolin-1 knockout and wild-type arteries showed fewer gap junctions in vessels from knockout animals associated with a lower expression of connexins 37, 40, and 43 and altered myoendothelial communication. Finally, we showed that TRPV4 channels and connexins colocalize with caveolin-1 in the caveolar compartment of the plasma membrane. CONCLUSIONS: We demonstrated that expression of caveolin-1 is required for EDHF-related relaxation by modulating membrane location and activity of TRPV4 channels and connexins, which are both implicated at different steps in the EDHF-signaling pathway.


Subject(s)
Biological Factors/metabolism , Calcium Signaling/physiology , Caveolin 1/metabolism , Cell Compartmentation/physiology , Endothelial Cells/metabolism , Vasodilation/physiology , Animals , Calcium/metabolism , Caveolae/metabolism , Caveolin 1/genetics , Connexins/metabolism , Endothelial Cells/ultrastructure , Gap Junctions/metabolism , Mice , Mice, Knockout , Microcirculation , Nitric Oxide/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
6.
J Clin Invest ; 97(8): 1908-15, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8621775

ABSTRACT

Cardiac myocytes express the nitric oxide synthase isoform originally identified in constitutive nitric oxide synthase cells (NOS3), which mediates the attenuation by muscarinic cholinergic agonists of beta-adrenergic stimulation of L-type calcium current and contractility in these cells. However, calcium current and contractility in these cells. However, the reciprocal regulation of NOS3 activity in myocytes by agents that elevate cAMP has not been reported. In this study, we show that NOS3 and mRNA and protein levels in cardiac myocytes are reduced both in vitro after treatment with cAMP elevating drugs, and in vivo after 3 d of treatment with milrinone, a type III cAMP phosphodiesterase inhibitor. This effect on NOS3 activity by cAMP is cell type specific because treatment of cardiac microvascular endothelial cells in vitro or in vivo did not decrease NOS3 mRNA or protein in these cells. NOS3 downregulation in myocytes appeared to be at the level of transcription since there was no modification of NOS3 mRNA half-life by agents that increase intracellular cAMP. To determine the functional effects of NOS3 downregulation, we examined the contractile responsiveness of isolated electrically paced ventricular myocytes, isolated from animals that had been treated in vivo with milrinone, to the beta-adrenergic agonist isoproterenol and the muscarinic cholinergic agonist carbamylcholine. There was no difference in baseline contractile function in cells that had been pretreated with cAMP elevating agents compared to controls, but cells exposed to milrinone in vivo exhibited an accentuation in their contractile responsiveness to isoproterenol compared to controls and a loss of responsiveness to carbamylcholine. Downregulation of myocyte NOS3 by sustained elevation of cAMP may have important implications for the regulation of myocardial contractile state by the autonomic nervous system.


Subject(s)
Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Cyclic AMP/physiology , Gene Expression Regulation, Enzymologic , Heart/physiology , Isoproterenol/pharmacology , Myocardial Contraction , Myocardium/enzymology , Nitric Oxide Synthase/biosynthesis , Phosphodiesterase Inhibitors/pharmacology , Pyridones/pharmacology , Transcription, Genetic , 1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Colforsin/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Heart/drug effects , Kinetics , Male , Milrinone , Myocardial Contraction/drug effects , Myocardium/cytology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
7.
J Clin Invest ; 103(6): 897-905, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079111

ABSTRACT

Hypercholesterolemia is a central pathogenic factor of endothelial dysfunction caused in part by an impairment of endothelial nitric oxide (NO) production through mechanisms that remain poorly characterized. The activity of the endothelial isoform of NO synthase (eNOS) was recently shown to be modulated by its reciprocal interactions with the stimulatory Ca2+-calmodulin complex and the inhibitory protein caveolin. We examined whether hypercholesterolemia may reduce NO production through alteration of this regulatory equilibrium. Bovine aortic endothelial cells were cultured in the presence of serum obtained from normocholesterolemic (NC) or hypercholesterolemic (HC) human volunteers. Exposure of endothelial cells to the HC serum upregulated caveolin abundance without any measurable effect on eNOS protein levels. This effect of HC serum was associated with an impairment of basal NO release paralleled by an increase in inhibitory caveolin-eNOS complex formation. Similar treatment with HC serum significantly attenuated the NO production stimulated by the calcium ionophore A23187. Accordingly, higher calmodulin levels were required to disrupt the enhanced caveolin-eNOS heterocomplex from HC serum-treated cells. Finally, cell exposure to the low-density lipoprotein (LDL) fraction alone dose-dependently reproduced the inhibition of basal and stimulated NO release, as well as the upregulation of caveolin expression and its heterocomplex formation with eNOS, which were unaffected by cotreatment with antioxidants. Together, our data establish a new mechanism for the cholesterol-induced impairment of NO production through the modulation of caveolin abundance in endothelial cells, a mechanism that may participate in the pathogenesis of endothelial dysfunction and the proatherogenic effects of hypercholesterolemia.


Subject(s)
Caveolins , Endothelium, Vascular/metabolism , Hypercholesterolemia/metabolism , Membrane Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Allosteric Regulation , Animals , Calcimycin/pharmacology , Calcium/pharmacology , Calmodulin/pharmacology , Cattle , Caveolin 1 , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Ion-Selective Electrodes , Lipoproteins, LDL/metabolism , Nitric Oxide Synthase Type III , Precipitin Tests , Protein Binding/drug effects
8.
J Clin Invest ; 95(1): 360-6, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7529262

ABSTRACT

In vitro studies indicate that muscarinic cholinergic inhibition of beta-adrenergic cardiac responses may be modulated in part by nitric oxide (NO). To evaluate the role of NO in parasympathetic inhibition of the beta-adrenergic contractile response in vivo, we assessed the inotropic response to dobutamine before and during bilateral vagus nerve stimulation in closed-chest dogs. Dobutamine administration and vagal stimulation were repeated during intracoronary infusion of the NO synthase inhibitor NG-monomethyl-L-arginine (L-NMMA, 10 mumol/min) and again following infusion of L-arginine (100 mg/kg). In eight dogs, intracoronary dobutamine infusion at rates of 25 and 50 micrograms/min increased peak +dP/dt by 131 +/- 24 and 168 +/- 22%, respectively (P < 0.0001). Vagal stimulation (2.5 Hz) attenuated the responses to dobutamine (25 and 50 micrograms/min) by 23 +/- 4 and 21 +/- 4%, respectively (P < 0.001). L-NMMA reduced (by 44-62%; P < 0.001) and L-arginine restored vagal inhibition of the dobutamine-stimulated inotropic response. In a second group of nine dogs, dobutamine was administered systemically to assure a constant concentration in the coronary circulation. Vagal stimulation (2.5 Hz) attenuated the dobutamine-stimulated inotropic response (2.5 and 5.0 micrograms/kg per min) by 40 +/- 12% and 57 +/- 8%, respectively (P < 0.004). As with intracoronary dobutamine, L-NMMA diminished and L-arginine restored vagal inhibition of the inotropic response to dobutamine. Intracoronary infusion of atropine (12 micrograms/min) abolished the vagal inhibitory effect, and intracoronary infusion of 8-bromo-cyclic GMP (1 and 10 mM) caused a dose-dependent attenuation of the dobutamine-stimulated increase in +dP/dt. These data suggest that NO mediates, at least in part, vagal inhibition of the inotropic response to beta-adrenergic stimulation by dobutamine, and thus may play a role in normal physiologic regulation of myocardial autonomic responses.


Subject(s)
Dobutamine/pharmacology , Myocardial Contraction/physiology , Nitric Oxide/metabolism , Receptors, Adrenergic, beta/physiology , Vagus Nerve/physiology , Amino Acid Oxidoreductases/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cyclic AMP/pharmacology , Dogs , Electric Stimulation , Heart/innervation , Heart Rate , Muscarinic Antagonists , Myocardial Contraction/drug effects , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase , omega-N-Methylarginine
9.
J Clin Invest ; 102(7): 1377-84, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9769330

ABSTRACT

Beta1- and beta2-adrenoceptors in heart muscle cells mediate the catecholamine-induced increase in the force and frequency of cardiac contraction. Recently, in addition, we demonstrated the functional expression of beta3-adrenoceptors in the human heart. Their stimulation, in marked contrast with that of beta1- and beta2-adrenoceptors, induces a decrease in contractility through presently unknown mechanisms. In the present study, we examined the role of a nitric oxide (NO) synthase pathway in mediating the beta3-adrenoceptor effect on the contractility of human endomyocardial biopsies. The negative inotropic effects of a beta3-adrenoceptor agonist, BRL 37344, and also of norepinephrine in the presence of alpha- and beta1-2-blockade were inhibited both by a nonspecific blocker of NO, methylene blue, and two NO synthase (NOS) inhibitors, L-N-monomethyl-arginine and L-nitroarginine-methyl ester. The effect of the NOS inhibitors was reversed by an excess of L-arginine, the natural substrate of NOS, but not by D-arginine. Moreover, the effects of the beta3-adrenoceptor agonist on contractility were associated with parallel increases in the production of NO and intracellular cGMP, which were also inhibited by NOS inhibitors. Immunohistochemical staining of human ventricular biopsies showed the expression of the endothelial constitutive (eNOS), but not the inducible (iNOS) isoform of NOS in both ventricular myocytes and endothelial cells. These results demonstrate that beta3-adrenoceptor stimulation decreases cardiac contractility through activation of an NOS pathway. Changes in the expression of this pathway may alter the balance between positive and negative inotropic effects of catecholamines on the heart potentially leading to myocardial dysfunction.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Ethanolamines/pharmacology , Heart/physiology , Myocardial Contraction/physiology , Myocardium/enzymology , Nitric Oxide Synthase/biosynthesis , Nitroarginine/pharmacology , Receptors, Adrenergic, beta/physiology , Arginine/pharmacology , Cyclic GMP/metabolism , Female , Heart/drug effects , Heart Ventricles , Humans , Isoproterenol/pharmacology , Kinetics , Male , Middle Aged , Myocardial Contraction/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Norepinephrine/pharmacology , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta-3 , omega-N-Methylarginine/pharmacology
10.
J Clin Invest ; 91(5): 2314-9, 1993 May.
Article in English | MEDLINE | ID: mdl-8486792

ABSTRACT

The mechanism by which soluble mediators of immune cell origin depress myocardial contractility, either globally as in systemic sepsis, or regionally in areas of inflammatory myocardial infiltrates, remains unclear. When freshly isolated ventricular myocytes from adult rat hearts were preincubated for at least 24 h in medium conditioned by endotoxin (LPS)-activated rat alveolar macrophages, their subsequent inotropic response to the beta-adrenergic agonist isoproterenol was reduced from 225 +/- 19% to 155 +/- 10% of the baseline amplitude of shortening (mean +/- SEM, P < 0.05). Neither baseline contractile function nor the contractile response to high extracellular calcium were affected. To determine whether an endogenous nitric-oxide (NO)-signaling pathway within ventricular myocytes was responsible for their decreased responsiveness to isoproterenol, the L-arginine analogue L-NMMA was added to the preincubation medium. While L-NMMA did not affect baseline contractile function or the response of control myocytes to isoproterenol, it completely restored the positive inotropic response to isoproterenol in myocytes preincubated in LPS-activated macrophage medium. Release of NO by ventricular myocytes following exposure to activated macrophage medium was detected as an increase in cGMP content in a reporter-cell (RFL-6) bioassay and also as increased nitrite content in myocyte-conditioned medium. Thus, the depressed contractile response of adult rat ventricular myocytes to beta-adrenergic agonists by a 24-h exposure to soluble inflammatory mediators is mediated at least in party by induction of an autocrine NO signaling pathway.


Subject(s)
Heart/physiology , Macrophage Activation , Macrophages, Alveolar/physiology , Myocardial Contraction , Myocardium/metabolism , Nitric Oxide/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cell Line , Cells, Cultured , Culture Media, Conditioned , Heart/drug effects , Heart Ventricles , Kinetics , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/cytology , Male , Rats , Rats, Sprague-Dawley , omega-N-Methylarginine
11.
Br J Pharmacol ; 151(3): 347-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17384667

ABSTRACT

BACKGROUND AND PURPOSE: Our goal was to elucidate mechanisms of the inhibitory effect of rosuvastatin on the accumulation of plaque oxidized low density lipoproteins (oxLDL) and on plaque volume, without lowering cholesterol, in mice with combined leptin and LDL-receptor deficiency (DKO). EXPERIMENTAL APPROACH: Twelve-week old DKO mice were treated with rosuvastatin (10 mg kg(-1) day(-1), s.c.) or placebo or no treatment for 12 weeks. The effect on blood variables, aortic plaque volume and composition and gene expression in the aorta and in THP-1 cells was assessed. KEY RESULTS: Rosuvastatin lowered free fatty acids (FFA), triglycerides, and increased insulin sensitivity, without affecting cholesterol. Rosuvastatin lowered the plaque volume, inhibited macrophage, lipid and oxLDL accumulation, and decreased the oxLDL-to-LDL ratio of plaques in the aortic arch. It increased superoxide dismutase 1 (SOD1), CD36, LXR-alpha, ABCA-1 and PPAR-gamma RNA expression in aortic extracts. SOD1 was the strongest inverse correlate of oxLDL. In THP-1 macrophages and foam cells, expression of SOD1 was lower than in THP-1 monocytes. Rosuvastatin restored expression of SOD1 in THP-1 macrophages and foam cells. CONCLUSIONS AND IMPLICATIONS: Rosuvastatin restored SOD1 expression in THP-1 macrophages and foam cells in vitro and in the aorta of DKO mice. The latter was associated with less oxLDL accumulation within atherosclerotic plaques and inhibition of plaque progression. This effect was obtained at a dose not affecting cholesterol levels but improving insulin sensitivity. SOD1 is a potentially important mediator of the prevention of oxLDL accumulation within atherosclerotic plaques.


Subject(s)
Aorta/drug effects , Fluorobenzenes/pharmacology , Lipoproteins, LDL/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Superoxide Dismutase/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Blood Glucose/metabolism , Body Weight , Cell Line , Dyslipidemias/blood , Dyslipidemias/genetics , Dyslipidemias/physiopathology , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Insulin/blood , Leptin/deficiency , Leptin/genetics , Lipids/blood , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/blood , Obesity/genetics , Obesity/physiopathology , PPAR gamma/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium , Superoxide Dismutase/genetics
12.
Eur J Heart Fail ; 9(12): 1163-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17999941

ABSTRACT

OBJECTIVE: To analyze the implication of the beta3-adrenoceptor (beta3-AR) pathway in human septic myocardium and a murine model of sepsis, a condition associated with myocardial depression. METHODS AND RESULTS: beta3-AR and eNOS protein abundance were increased (332+/-66.4% and 218+/-39.3; P<0.05) in hearts from septic patients. The effect of BRL37344, a beta3-AR-preferential agonist, was analyzed by videomicroscopy on the contractility of neonatal mouse ventricular myocytes (NMVM) incubated with conditioned medium from LPS-stimulated cultured macrophages (Mc-LPS+ medium). Stimulation of untreated NMVM with BRL37344 dose-dependently decreased the amplitude of contractile shortening (P<0.05). This response was abolished by L-NAME (NOS inhibitor). Incubation in Mc-LPS+ medium potentiated the depressing effect of BRL37344 (P<0.05) as well as of SR58611A (P<0.05) in wild-type myocytes. Importantly, the contractile depression was abrogated in cardiomyocytes from beta3-AR KO mice. CONCLUSIONS: beta3-AR are upregulated during sepsis in the human myocardium and by cytokines in murine cardiomyocytes, where they mediate an increased negative inotropic response to beta3 agonists. Activation of the beta3-AR pathway by catecholamines may contribute to the myocardial dysfunction in sepsis.


Subject(s)
Myocytes, Cardiac/metabolism , RNA/genetics , Receptors, Adrenergic, beta-3/genetics , Sepsis/metabolism , Up-Regulation , Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Adult , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Ethanolamines/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , RNA/biosynthesis , Rats , Rats, Wistar , Receptors, Adrenergic, beta-3/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/pathology , Sepsis/physiopathology , Stereoisomerism
13.
Pharmacol Ther ; 108(3): 225-56, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15949847

ABSTRACT

Nitric oxide synthases (NOS) are the enzymes responsible for nitric oxide (NO) generation. To date, 3 distinct NOS isoforms have been identified: neuronal NOS (NOS1), inducible NOS (NOS2), and endothelial NOS (NOS3). Biochemically, NOS consists of a flavin-containing reductase domain, a heme-containing oxygenase domain, and regulatory sites. NOS catalyse an overall 5-electron oxidation of one Nomega-atom of the guanidino group of L-arginine to form NO and L-citrulline. NO exerts a plethora of biological effects in the cardiovascular system. The basal formation of NO in mitochondria by a mitochondrial NOS seems to be one of the main regulators of cellular respiration, mitochondrial transmembrane potential, and transmembrane proton gradient. This review focuses on recent advances in the understanding of the role of enzyme and enzyme-independent NO formation, regulation of NO bioactivity, new aspects of NO on cardiac function and morphology, and the clinical impact and perspectives of these recent advances in our knowledge on NO-related pathways.


Subject(s)
Cardiovascular System/metabolism , Nitric Oxide/metabolism , Animals , Atherosclerosis/etiology , Heart/physiology , Homeostasis , Humans , Nitric Oxide Synthase/metabolism
14.
Circ Res ; 93(5): 388-98, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12958142

ABSTRACT

Nitric oxide (NO) is produced from virtually all cell types composing the myocardium and regulates cardiac function through both vascular-dependent and -independent effects. The former include regulation of coronary vessel tone, thrombogenicity, and proliferative and inflammatory properties as well as cellular cross-talk supporting angiogenesis. The latter comprise the direct effects of NO on several aspects of cardiomyocyte contractility, from the fine regulation of excitation-contraction coupling to modulation of (presynaptic and postsynaptic) autonomic signaling and mitochondrial respiration. This multifaceted involvement of NO in cardiac physiology is supported by a tight molecular regulation of the three NO synthases, from cellular spatial confinement to posttranslational allosteric modulation by specific interacting proteins, acting in concert to restrict the influence of NO to a particular intracellular target in a stimulus-specific manner. Loss of this specificity, such as produced on excessive NO delivery from inflammatory cells (or cytokine-stimulated cardiomyocytes themselves), may result in profound cellular disturbances leading to heart failure. Future therapeutic manipulations of cardiac NO synthesis will necessarily draw on additional characterization of the cellular and molecular determinants for the net effect of this versatile radical on the cardiomyocyte biology.


Subject(s)
Heart/physiology , Nitric Oxide/metabolism , Animals , Heart/physiopathology , Heart Diseases/physiopathology , Humans , Myocardial Contraction/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I
15.
Circ Res ; 89(10): 866-73, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11701613

ABSTRACT

3-Hydroxy-3-methylglutaryl (HMG)-coenzyme A reductase inhibitors or statins exert direct beneficial effects on the endothelium in part through an increase in nitric oxide (NO) production. Here, we examined whether posttranslational modifications of the endothelial NO synthase (eNOS) could account for the proangiogenic effects of statins. We used endothelial cells (ECs) isolated from cardiac microvasculature, aorta, and umbilical veins, as well as dissected microvessels and aortic rings, that were cultured on reconstituted basement membrane matrix (Matrigel). Tube or precapillary formation was evaluated after statin treatment, in parallel with immunoblotting and immunoprecipitation experiments. Atorvastatin stimulated NO-dependent angiogenesis from both isolated and outgrowing (vessel-derived) ECs, independently of changes in eNOS expression. We found that in macro- but not microvascular ECs, atorvastatin stabilized tube formation through a decrease in caveolin abundance and its inhibitory interaction with eNOS. We also identified the chaperone protein hsp90 as a key target for the proangiogenic effects of statins. Using geldanamycin, an inhibitor of hsp90 function, and overexpression of recombinant hsp90, we documented that the statin-induced phosphorylation of eNOS on Ser1177 was directly dependent on the ability of hsp90 to recruit Akt in the eNOS complex. Finally, we showed that statin promoted the tyrosine phosphorylation of hsp90 and the direct interaction of hsp90 with Akt, which further potentiated the NO-dependent angiogenic processes. Our study provides new mechanistic insights into the NO-mediated angiogenic effects of statins and underscores the potential of these drugs and other modulators of hsp90 and caveolin abundance to promote neovascularization in disease states associated or not with atherosclerosis.


Subject(s)
Caveolins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases , Animals , Atorvastatin , Benzoquinones , Biological Assay/methods , Cattle , Caveolin 1 , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heptanoic Acids/pharmacology , Humans , Immunoblotting , Lactams, Macrocyclic , Mice , Models, Cardiovascular , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyrroles/pharmacology , Quinones/pharmacology , Rats , Transfection
16.
Circulation ; 103(1): 113-8, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11136695

ABSTRACT

BACKGROUND: Hypercholesterolemia is causally associated with defects of endothelial nitric oxide (NO)-dependent vasodilation. Increased uptake of cholesterol by endothelial cells (ECs) upregulates the abundance of the structural protein caveolin-1 and impairs NO release through the stabilization of the inhibitory heterocomplex between caveolin-1 and endothelial NO synthase (eNOS). Therefore, we examined whether the hydroxy-methylglutaryl-coenzyme A reductase inhibitor atorvastatin modulates caveolin abundance, eNOS activity, and NO release through a reduction in endogenous cholesterol levels. METHODS AND RESULTS: ECs were incubated with increasing doses of atorvastatin in the absence or in the presence of human LDL cholesterol (LDL-Chol) fractions in the presence of antioxidants. Our results show that atorvastatin (10 nmol/L to 1 micromol/L) reduced caveolin-1 abundance in the absence (-75%) and in the presence (-20% to 70%) of LDL-Chol. This was paralleled by a decreased inhibitory interaction between caveolin-1 and eNOS and a restoration and/or potentiation of the basal (+45%) and agonist-stimulated (+107%) eNOS activity. These effects were observed in the absence of changes in eNOS abundance and were reversed with mevalonate. In the presence of LDL-Chol, atorvastatin also promoted the agonist-induced association of eNOS and the chaperone Hsp90, resulting in the potentiation of eNOS activation. CONCLUSIONS: We provide biochemical and functional evidence that atorvastatin promotes NO production by decreasing caveolin-1 expression in ECs, regardless of the level of extracellular LDL-Chol. These findings highlight the therapeutic potential of inhibiting cholesterol synthesis in peripheral cells to correct NO-dependent endothelial dysfunction associated with hypercholesterolemia and possibly other diseases.


Subject(s)
Caveolins/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Nitric Oxide Synthase/metabolism , Transcription Factors , Animals , Atorvastatin , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Cattle , Caveolin 1 , Cells, Cultured , Cholesterol/biosynthesis , Cholesterol, LDL/metabolism , Cholesterol, LDL/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , HSP90 Heat-Shock Proteins/metabolism , Heptanoic Acids/pharmacology , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Leupeptins/pharmacology , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Pyrroles/pharmacology , Sterol Regulatory Element Binding Protein 1 , Up-Regulation/drug effects
17.
Circulation ; 110(8): 948-54, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15302798

ABSTRACT

BACKGROUND: Coronary vessel tone is modulated in part by beta-adrenergic relaxation. However, the implication of specific beta-adrenoceptor subtypes and their downstream vasorelaxing mechanism(s) in human coronary resistance arteries is poorly defined. beta3-Adrenoceptors were recently shown to vasodilate animal vessels and are expressed in human hearts. METHODS AND RESULTS: We examined the expression and functional role of beta3-adrenoceptors in human coronary microarteries and their coupling to vasodilating nitric oxide (NO) and/or hyperpolarization mechanisms. The expression of beta3-adrenoceptor mRNA and protein was demonstrated in extracts of human coronary microarteries. Immunohistochemical analysis revealed their exclusive localization in the endothelium, with no staining of vascular smooth muscle. In contractility experiments in which videomicroscopy was used, the nonspecific beta-agonist isoproterenol and the beta3-preferential agonist BRL37344 evoked an approximately 50% relaxation of endothelin-1-preconstricted human coronary microarteries. Relaxations were blocked by the beta1/beta2/beta3-adrenoceptor antagonist bupranolol but were insensitive to the beta1/beta2-adrenoceptor antagonist nadolol, confirming a beta3-adrenoceptor-mediated pathway. Relaxation in response to BRL37344 was absent in human coronary microarteries devoid of functional endothelium. When human coronary microarteries were precontracted with KCl (thereby preventing vessel hyperpolarization), the relaxation to BRL37344 was reduced to 15.5% and totally abrogated by the NO synthase inhibitor L-omega-nitroarginine, confirming the participation of a NO synthase-mediated relaxation. The NO synthase-independent relaxation was completely inhibited by the Ca2+-activated K+ channel inhibitors apamin and charybdotoxin, consistent with an additional endothelium-derived hyperpolarizing factor-like response. Accordingly, membrane potential recordings demonstrated vessel hyperpolarization in response to beta3-adrenoceptor stimulation. CONCLUSIONS: Beta3-adrenoceptors are expressed in the endothelium of human coronary resistance arteries and mediate adrenergic vasodilatation through both NO and vessel hyperpolarization.


Subject(s)
Coronary Vessels/physiology , Endothelium, Vascular/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Receptors, Adrenergic, beta-3/physiology , Vasodilation/physiology , Adolescent , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Adult , Aged , Aged, 80 and over , Apamin/pharmacology , Arterioles/drug effects , Arterioles/physiology , Bupranolol/pharmacology , Charybdotoxin/pharmacology , Child , Coronary Vessels/drug effects , Endothelium, Vascular/drug effects , Ethanolamines/pharmacology , Female , Humans , Isoproterenol/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microcirculation/drug effects , Microcirculation/physiology , Microscopy, Video , Middle Aged , Nadolol/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Nitroarginine/pharmacology , Potassium Channel Blockers/pharmacology , RNA, Messenger/biosynthesis , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-3/biosynthesis , Receptors, Adrenergic, beta-3/drug effects , Receptors, Adrenergic, beta-3/genetics , Vasodilation/drug effects
18.
Circulation ; 103(12): 1649-55, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11273992

ABSTRACT

BACKGROUND: Contrary to beta(1)- and beta(2)-adrenoceptors, beta(3)-adrenoceptors mediate a negative inotropic effect in human ventricular muscle. To assess their functional role in heart failure, our purpose was to compare the expression and contractile effect of beta(3)-adrenoceptors in nonfailing and failing human hearts. METHODS AND RESULTS: We analyzed left ventricular samples from 29 failing (16 ischemic and 13 dilated cardiomyopathic) hearts (ejection fraction 18.6+/-2%) and 25 nonfailing (including 12 innervated) explanted hearts (ejection fraction 64.2+/-3%). beta(3)-Adrenoceptor proteins were identified by immunohistochemistry in ventricular cardiomyocytes from nonfailing and failing hearts. Contrary to beta(1)-adrenoceptor mRNA, Western blot analysis of beta(3)-adrenoceptor proteins showed a 2- to 3-fold increase in failing compared with nonfailing hearts. A similar increase was observed for Galpha(i-2) proteins that couple beta(3)-adrenoceptors to their negative inotropic effect. Contractile tension was measured in electrically stimulated myocardial samples ex vivo. In failing hearts, the positive inotropic effect of the nonspecific amine isoprenaline was reduced by 75% compared with that observed in nonfailing hearts. By contrast, the negative inotropic effect of beta(3)-preferential agonists was only mildly reduced. CONCLUSIONS: Opposite changes occur in beta(1)- and beta(3)-adrenoceptor abundance in the failing left ventricle, with an imbalance between their inotropic influences that may underlie the functional degradation of the human failing heart.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiotonic Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go , Isoproterenol/pharmacology , Myocardial Contraction/drug effects , Myocardial Ischemia/metabolism , Proto-Oncogene Proteins , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Adult , Biopsy , Blotting, Western , Cardiomyopathy, Dilated/pathology , Contraindications , Female , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Immunohistochemistry , In Vitro Techniques , Male , Middle Aged , Myocardial Ischemia/pathology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Proto-Oncogene Proteins/metabolism , Up-Regulation/drug effects
19.
Trends Pharmacol Sci ; 21(11): 426-31, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11121573

ABSTRACT

beta-Adrenoceptors of the beta1 and beta2 subtypes classically mediate the effects of catecholamines on the contractility of cardiac muscle and the relaxation of vascular smooth muscle. Since the molecular characterization of the beta3-adrenoceptor in 1989, most studies of this adrenoceptor subtype have focused on its control of lipolysis in adipose tissues. However, more recent studies have investigated the involvement of beta3-adrenoceptors in the physiological control of cardiac and vascular contractility. In this article, the pharmacological and molecular evidence that supports the functional role of beta3-adrenoceptors in cardiovasculartissues of various species, including humans, will be discussed. These data might provide new insights into our understanding of the abnormal responsiveness of the cardiovascular system to catecholamines in heart failure and its treatment with beta3-adrenoceptor antagonists.


Subject(s)
Cardiovascular Physiological Phenomena , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Antagonists , Adrenergic beta-Antagonists/pharmacology , Animals , Cardiovascular Physiological Phenomena/drug effects , Humans , Receptors, Adrenergic, beta-3/biosynthesis
20.
Ann N Y Acad Sci ; 1047: 173-82, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16093495

ABSTRACT

The role of nitric oxide (NO) as a regulator of cardiac contraction was suggested in the early nineties, but a consensual view of its main functions in cardiac physiology has only recently emerged with the help of experiments using genetic deletion or overexpression of the three nitric oxide synthase (NOS) isoforms in cardiomyocytes. Contrary to the effects of exogenous, pharmacologic NO donors, signaling by endogenous NO is restricted to intracellular effectors co-localized with NOS in specific subcellular compartments. This both ensures coordinate signaling by the three NOS isoforms on different aspects of the cardiomyocyte function and helps to reconcile previous apparently contradictory observations based on the use of non-isoform-specific NOS inhibitors. This review will emphasize the role of NOS on excitation-contraction coupling in the normal and diseased heart. Endothelial NOS and neuronal NOS contribute to maintain an adequate balance between adrenergic and vagal input to the myocardium and participate in the early and late phases of the Frank-Starling adaptation of the heart. At the early phases of cardiac diseases, inducible NOS reinforces these effects, which may become maladaptive as disease progresses.


Subject(s)
Myocardium/chemistry , Nitric Oxide Synthase/analysis , Nitric Oxide/physiology , Electrophysiology , Heart Diseases/enzymology , Heart Diseases/etiology , Humans , Models, Biological , Myocardium/enzymology , Nitric Oxide Synthase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL