Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
3.
Endocrinology ; 156(2): 707-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406019

ABSTRACT

Iodine deficiency (ID) induces microvascular changes in the thyroid gland via a TSH-independent reactive oxygen species-hypoxia inducible factor (HIF)-1α-vascular endothelial growth factor (VEGF) pathway. The involvement of nitric oxide (NO) in this pathway and the role of calcium (Ca(2+)) and of ryanodine receptors (RYRs) in NO synthase 3 (NOS3) activation were investigated in a murine model of goitrogenesis and in 3 in vitro models of ID, including primary cultures of human thyrocytes. ID activated NOS3 and the production of NO in thyrocytes in vitro and increased the thyroid blood flow in vivo. Using bevacizumab (a blocking antibody against VEGF-A) in mice, it appeared that NOS3 is activated upstream of VEGF-A. L-nitroarginine methyl ester (a NOS inhibitor) blocked the ID-induced increase in thyroid blood flow in vivo and NO production in vitro, as well as ID-induced VEGF-A mRNA and HIF-1α expression in vitro, whereas S-nitroso-acetyl-penicillamine (a NO donor) did the opposite. Ca(2+) is involved in this pathway as intracellular Ca(2+) flux increased after ID, and thapsigargin activated NOS3 and increased VEGF-A mRNA expression. Two of the 3 known mammalian RYR isoforms (RYR1 and RYR2) were shown to be expressed in thyrocytes. RYR inhibition using ryanodine at 10µM decreased ID-induced NOS3 activation, HIF-1α, and VEGF-A expression, whereas RYR activation with ryanodine at 1nM increased NOS3 activation and VEGF-A mRNA expression. In conclusion, during the early phase of TSH-independent ID-induced microvascular activation, ID sequentially activates RYRs and NOS3, thereby supporting ID-induced activation of the NO/HIF-1α/VEGF-A pathway in thyrocytes.


Subject(s)
Iodine/deficiency , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Thyroid Gland/blood supply , Animals , Calcium/metabolism , Cell Line , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Rats , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
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
5.
Atherosclerosis ; 218(2): 272-80, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21762914

ABSTRACT

UNLABELLED: Since heterozygous familial hypercholesterolemia (HeFH) is a disease that exposes the individual from birth onwards to severe hypercholesterolemia with the development of early cardiovascular disease, a clear consensus on the management of this disease in young patients is necessary. In Belgium, a panel of paediatricians, specialists in (adult) lipid management, general practitioners and representatives of the FH patient organization agreed on the following common recommendations. 1. Screening for HeFH should be performed only in children older than 2 years when HeFH has been identified or is suspected (based on a genetic test or clinical criteria) in one parent.2. The diagnostic procedure includes, as a first step, the establishment of a clear diagnosis of HeFH in one of the parents. If this precondition is satisfied, a low-density-lipoprotein cholesterol (LDL-C) levelabove 3.5 mmol/L (135 mg/dL) in the suspected child is predictive for differentiating affected from non-affected children. 3. A low saturated fat and low cholesterol diet should be started after 2 years, under the supervision of a dietician or nutritionist.4. The pharmacological treatment, using statins as first line drugs, should usually be started after 10 years if LDL-C levels remain above 5 mmol/L (190 mg/dL), or above 4 mmol/L (160 mg/dL) in the presence of a causative mutation, a family history of early cardiovascular disease or severe risk factors. The objective is to reduce LDL-C by at least 30% between 10 and 14 years and, thereafter, to reach LDL-C levels of less than 3.4 mmol/L (130 mg/dL). CONCLUSION: The aim of this consensus statement is to achieve more consistent management in the identification and treatment of children with HeFH in Belgium.


Subject(s)
Hyperlipoproteinemia Type II/therapy , Adult , Cardiology/methods , Child , Consensus Development Conferences as Topic , Decision Making , Female , Gastroenterology/methods , General Practice/methods , Guidelines as Topic , Heterozygote , Humans , Hyperlipoproteinemia Type II/diet therapy , Hyperlipoproteinemia Type II/genetics , Lipids/chemistry , Male , Nutritional Sciences , Pediatrics/methods , Young Adult
6.
Physiol Rev ; 89(2): 481-534, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19342613

ABSTRACT

Nitric oxide production in response to flow-dependent shear forces applied on the surface of endothelial cells is a fundamental mechanism of regulation of vascular tone, peripheral resistance, and tissue perfusion. This implicates the concerted action of multiple upstream "mechanosensing" molecules reversibly assembled in signalosomes recruiting endothelial nitric oxide synthase (eNOS) in specific subcellular locales, e.g., plasmalemmal caveolae. Subsequent short- and long-term increases in activity and expression of eNOS translate this mechanical stimulus into enhanced NO production and bioactivity through a complex transcriptional and posttranslational regulation of the enzyme, including by shear-stress responsive transcription factors, oxidant stress-dependent regulation of transcript stability, eNOS regulatory phosphorylations, and protein-protein interactions. Notably, eNOS expressed in cardiac myocytes is amenable to a similar regulation in response to stretching of cardiac muscle cells and in part mediates the length-dependent increase in cardiac contraction force. In addition to short-term regulation of contractile tone, eNOS mediates key aspects of cardiac and vascular remodeling, e.g., by orchestrating the mobilization, recruitment, migration, and differentiation of cardiac and vascular progenitor cells, in part by regulating the stabilization and transcriptional activity of hypoxia inducible factor in normoxia and hypoxia. The continuum of the influence of eNOS in cardiovascular biology explains its growing implication in mechanosensitive aspects of integrated physiology, such as the control of blood pressure variability or the modulation of cardiac remodeling in situations of hemodynamic overload.


Subject(s)
Cardiovascular Physiological Phenomena , Mechanotransduction, Cellular/physiology , Nitric Oxide Synthase Type III/physiology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Myocardial Contraction/physiology , Myocardium/cytology , Vasoconstriction/physiology
7.
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
8.
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
9.
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
10.
Acta Clin Belg ; 61(6): 326-34, 2006.
Article in English | MEDLINE | ID: mdl-17323842

ABSTRACT

Nitric oxide produced by three different isoforms of nitric oxide synthase (NOS) widely expressed in virtually all vascular cell types is mostly produced by the endothelial isoform (eNOS) in endothelial cells where it plays a crucial role in vascular tone and structure regulation. It also exerts an anti-inflammatory influence, inhibits platelets adhesion and aggregation, and prevents smooth muscle cells proliferation and migration. Several lines of evidence link endothelial dysfunction, characterized by decreased bioavailability of nitric oxide, with the development of many pathological conditions such as heart failure, hypertension, diabetes and atherosclerosis. This review focuses on nitric oxide-dependent endothelial dysfunction in cardiovascular diseases, its clinical detection and relevance, potential pathogenic mechanisms and possible therapies.


Subject(s)
Cardiovascular Diseases/physiopathology , Endothelium, Vascular/physiopathology , Nitric Oxide/physiology , Down-Regulation/physiology , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type III/physiology , PPAR gamma/metabolism , Phosphorylation , Signal Transduction/physiology
12.
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
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.
Article in English | MEDLINE | ID: mdl-15985381

ABSTRACT

The mammalian heart expresses all three isoforms of nitric oxide synthases (NOS) in diverse cell types of the myocardium. Despite their apparent promiscuity, the NOS isoforms support specific signaling because of their subcellular compartmentation with colocalized effectors and limited diffusibility of NO in muscle cells. eNOS and nNOS sustain normal EC coupling and contribute to the early and late phases of the Frank-Starling mechanism of the heart. They also attenuate the beta1-/beta2-adrenergic increase in inotropy and chronotropy, and reinforce the pre- and post-synaptic vagal control of cardiac contraction. By doing so, the NOS protect the heart against excessive stimulation by catecholamines, just as an "endogenous beta-blocker". In the ischemic and failing myocardium, induced iNOS further reinforces this effect, as does eNOS coupled to overexpressed beta3-adrenoceptors. nNOS expression also increases in the aging and infarcted heart, but its role (compensatory or deleterious) is less clear. In addition to their direct regulation of contractility, the NOS modulate oxygen consumption, substrate utilization, sensitivity to apoptosis, hypertrophy and regenerative potential, all of which illustrate the pleiotropic effects of this radical on the cardiac cell biology.


Subject(s)
Heart/physiology , Nitric Oxide Synthase/chemistry , Nitric Oxide/metabolism , Animals , Catecholamines/metabolism , Diastole , Humans , Models, Biological , Muscles/metabolism , Myocardial Contraction , Myocardial Infarction/metabolism , Myocardium/metabolism , Nitric Oxide Synthase/metabolism , Protein Isoforms , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Systole
15.
Arch Mal Coeur Vaiss ; 98(3): 242-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15816328

ABSTRACT

Despite the apparent redundancy of NOS isoforms in the myocardium, subcellular compartmentation dictates specific NO signaling from each isoform to colocalized effectors in response to physical (e.g. stretch) or receptor-mediated stimuli. Genetic deletion or overexpression experiments helped to characterize each isoform's respective role in the normal or diseased heart. eNOS and nNOS both contribute to sustain normal EC coupling and contribute to the early and late phases of the Frank-Starling mechanism of the heart. They also negatively modulate the beta1-/beta2-adrenergic increase in inotropy and chronotropy, and reinforce the (pre- and post-synaptic) vagal control of cardiac contraction, thereby protecting the heart against excessive stimulation by catecholamines. In the ischemic and failing myocardium, iNOS expression is induced and further contributes to attenuate the inotropic effect of catecholamines, as does eNOS coupled to overexpressed beta3-adrenoceptors. nNOS expression also increases in the aging and ischemic heart, but its role (compensatory or deleterious) remains to be defined. Many drugs currently used for the treatment of ischemic or failing cardiac diseases also activate and/or upregulate eNOS in the myocardium, which supports its proposed protective role, e.g. as "endogenous beta-blocker". Future pharmacologic modulation of the cardiac NOS will have to take into account their specific modulation of the various aspects of cardiac function, if one hopes to deliver more targeted and efficient therapy than currently achieved with exogenous NO donors.


Subject(s)
Myocardium/metabolism , Nitric Oxide Synthase/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Heart Failure/physiopathology , Humans , Myocardial Contraction/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Signal Transduction
16.
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
17.
Rev Med Interne ; 25(5): 386-9, 2004 May.
Article in French | MEDLINE | ID: mdl-15110957

ABSTRACT

INTRODUCTION: Amiodarone can induced hepatic dysfunction. OBSERVATIONS: We describe two patients who developed hepatotoxicity presenting suggestive histological features. The outcome was fatal in one case but lesions were reversible in the second case upon treatment interruption. CONCLUSION: These observations show the necessity of hepatic monitoring of patients treated with amiodarone in order to detect potentially severe hepatotoxicity.


Subject(s)
Amiodarone/adverse effects , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/therapeutic use , Chemical and Drug Induced Liver Injury/pathology , Aged , Fatal Outcome , Female , Humans , Male , Phospholipids/blood
18.
Semin Vasc Med ; 4(4): 367-75, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15861317

ABSTRACT

Hypertension and dyslipidemia are frequently associated as risk factors for cardiovascular diseases. Statins are among the most potent drugs to correct hypercholesterolemia, and their use across a wide range of cardiovascular risk levels significantly reduced morbidity and mortality in large intervention trials. Aside from (or in addition to) reducing plasma cholesterol, statins also reduce blood pressure, another effect associated with cardiovascular risk reduction by other antihypertensive drugs. This review examines the proposition that a part of the statins' beneficial effect in cardiovascular diseases may result from direct effects on blood pressure regulation, perhaps independent of lipid lowering. Potential molecular mechanisms are considered (e.g., "pleiotropic" effects on endothelial vasoactive mediators, oxidant stress, or inflammation), all of which may affect the central or peripheral control of blood pressure homeostasis, as well as modulate target organ damage. In particular, potential effects of statins on blood pressure and heart rate variability open new perspectives for a better tailoring of drug treatment in high-cardiovascular risk patients.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertension/drug therapy , Animals , Blood Pressure/drug effects , Humans , Hyperlipidemias/blood , Hyperlipidemias/complications , Hyperlipidemias/drug therapy , Hypertension/etiology , Hypertension/physiopathology , Lipids/blood , Risk Factors
19.
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
20.
J Physiol ; 546(Pt 1): 63-75, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12509479

ABSTRACT

The modulatory role of endothelial nitric oxide synthase (eNOS) on heart contraction, relaxation and rate is examined in light of recent studies using genetic deletion or overexpression in mice under specific conditions. Unstressed eNOS-/- hearts in basal conditions exhibit a normal inotropic and lusitropic function, with either decreased or unchanged heart rate. Under stimulation with catecholamines, eNOS-/- mice predominantly show a potentiation in their beta-adrenergic inotropic and lusitropic responsiveness. A similar phenotype is observed in beta 3-adrenoceptor deficient mice, pointing to a key role of this receptor subtype for eNOS coupling. The effect of eNOS on the muscarinic cholinergic modulation of cardiac function probably operates in conjunction with other NO-independent mechanisms, the persistence of which may explain the apparent dispensability of this isoform for the effect of acetylcholine in some eNOS-/- mouse strains. eNOS-/- hearts submitted to short term ischaemia-reperfusion exhibit variable alterations in systolic and diastolic function and infarct size, while those submitted to myocardial infarction present a worsened ventricular remodelling, increased 1 month mortality and loss of benefit from ACE inhibitor or angiotensin II type I receptor antagonist therapy. Although non-conditional eNOS gene deletion may engender phenotypic adaptations (e.g. ventricular hypertrophy resulting from chronic hypertension, or upregulation of the other NOS isoforms) potentially confounding the interpretation of comparative studies, the use of eNOS-/- mice has undoubtedly advanced (and will probably continue to improve) our understanding of the complex role of eNOS (in conjunction with the other NOSs) in the regulation of cardiac function. The challenge is now to confirm the emerging paradigms in human cardiac physiology and hopefully translate them into therapy.


Subject(s)
Heart Rate/physiology , Myocardial Contraction/physiology , Nitric Oxide Synthase/physiology , Animals , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III
SELECTION OF CITATIONS
SEARCH DETAIL
...