Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 17982, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504250

ABSTRACT

We recently highlighted a novel potential protective paracrine role of cardiac myeloid CD11b/c cells improving resistance of adult hypertrophied cardiomyocytes to oxidative stress and potentially delaying evolution towards heart failure (HF) in response to early ß-adrenergic stimulation. Here we characterized macrophages (Mφ) in hearts early infused with isoproterenol as compared to control and failing hearts and evaluated the role of upregulated CX3CL1 in cardiac remodeling. Flow cytometry, immunohistology and Mφ-depletion experiments evidenced a transient increase in Mφ number in isoproterenol-infused hearts, proportional to early concentric hypertrophy (ECH) remodeling and limiting HF. Combining transcriptomic and secretomic approaches we characterized Mφ-enriched CD45+ cells from ECH hearts as CX3CL1- and TNFα-secreting cells. In-vivo experiments, using intramyocardial injection in ECH hearts of either Cx3cl1 or Cx3cr1 siRNA, or Cx3cr1-/- knockout mice, identified the CX3CL1/CX3CR1 axis as a protective pathway delaying transition to HF. In-vitro results showed that CX3CL1 not only enhanced ECH Mφ proliferation and expansion but also supported adult cardiomyocyte hypertrophy via a synergistic action with TNFα. Our data underscore the in-vivo transient protective role of the CX3CL1/CX3CR1 axis in ECH remodeling and suggest the participation of CX3CL1-secreting Mφ and their crosstalk with CX3CR1-expressing cardiomyocytes to delay HF.


Subject(s)
Adrenergic beta-Agonists/adverse effects , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Heart Failure/chemically induced , Heart Failure/metabolism , Isoproterenol/adverse effects , Macrophages/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction/genetics , Animals , CX3C Chemokine Receptor 1/genetics , Cell Communication/genetics , Cell Proliferation/genetics , Cells, Cultured , Chemokine CX3CL1/genetics , Disease Models, Animal , Heart Failure/genetics , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/metabolism , Ventricular Remodeling/genetics
2.
J Physiol Pharmacol ; 66(3): 355-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26084217

ABSTRACT

Extracellular matrix metalloproteinase inducer (EMMPRIN), known for its ability to induce matrix metalloproteinase (MMP) expression, was proposed to play a role in the adverse cardiac extracellular matrix remodeling. After observing an age-associated increase in cardiac EMMPRIN expression in both mice and rats, the role and mechanism of action of EMMPRIN was investigated in the myocardial age-associated changes using 3, 12 and 24 month old EMMPRIN knock-out (KO) vs. wild-type (WT) mice, by cardiac echocardiography, Western blots, immunohistochemistry, ELISA and histology. Adilated cardiomyopathy characterized by a decreased ejection fraction and an enlargement of left ventricular chamber (LV) associated with LV hypertrophy, occurred in KO mice as soon as 12 month old. The increase in interstitial collagen deposition during aging in WT mice could not be detected in KO mice. This may be related to the reduced activation (48% reduction; P < 0.05) and signaling (smad2/3 nuclear translocation) of TGF-ß in the 12 month old KO mice which paralleled with a greater reduction in the TGF-ß known activating enzymes such as MT1-MMP and MMP-1 (33% and 37% reduction respectively, between 3 and 12 month old in KO mice; P < 0.05) as well as uPA. These findings demonstrate that EMMPRIN gene silencing is associated with an aberrant extracellular matrix remodeling, characterized by the absence of a detected age-associated fibrosis and consequently to dilated cardiopathy, indicating that a fine regulation of EMMPRIN is essential for the coordinated ECM remodeling during aging.


Subject(s)
Aging/physiology , Basigin/metabolism , Extracellular Matrix/metabolism , Ventricular Remodeling/physiology , Animals , Basigin/genetics , Collagen/metabolism , Female , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Rats , Rats, Wistar
3.
Gene Ther ; 20(9): 901-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23535897

ABSTRACT

Coronary artery disease represents the leading cause of mortality in the developed world. Percutaneous coronary intervention involving stent placement remains disadvantaged by restenosis or thrombosis. Vascular gene therapy-based methods may be approached, but lack a vascular gene delivery vector. We report a safe and efficient long-term transduction of rat carotid vessels after balloon injury intervention with a translational optimized AAV2.5 vector. Compared with other known adeno-associated virus (AAV) serotypes, AAV2.5 demonstrated the highest transduction efficiency of human coronary artery vascular smooth muscle cells (VSMCs) in vitro. Local delivery of AAV2.5-driven transgenes in injured carotid arteries resulted in transduction as soon as day 2 after surgery and persisted for at least 30 days. In contrast to adenovirus 5 vector, inflammation was not detected in AAV2.5-transduced vessels. The functional effects of AAV2.5-mediated gene transfer on neointimal thickening were assessed using the sarco/endoplasmic reticulum Ca(2+) ATPase isoform 2a (SERCA2a) human gene, known to inhibit VSMC proliferation. At 30 days, human SERCA2a messenger RNA was detected in transduced arteries. Morphometric analysis revealed a significant decrease in neointimal hyperplasia in AAV2.5-SERCA2a-transduced arteries: 28.36±11.30 (n=8) vs 77.96±24.60 (n=10) µm(2), in AAV2.5-green fluorescent protein-infected, P<0.05. In conclusion, AAV2.5 vector can be considered as a promising safe and effective vector for vascular gene therapy.


Subject(s)
Coronary Restenosis/therapy , Dependovirus/genetics , Genetic Therapy , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Actins/genetics , Actins/metabolism , Animals , Carotid Arteries/cytology , Cells, Cultured , Coronary Vessels/cytology , Dependovirus/physiology , Disease Models, Animal , Genetic Vectors , Humans , Male , Muscle, Smooth, Vascular/pathology , Neointima/physiopathology , Rats , Rats, Sprague-Dawley , Transduction, Genetic
4.
Gene Ther ; 20(3): 248-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22456325

ABSTRACT

Targeting diseased cells is a challenging issue in both pharmacological and biological therapeutics. Gene therapy is emerging as a novel approach for treating rare diseases and for illnesses for which there is no other alternative. An important limitation of gene therapy has been the off-target effects and therefore efforts have been focused on increasing the specificity of gene transfer to the targeted organ. Here, we describe a promoter containing six nuclear factor of activated T cells (NFAT) consensus sequences, which is as efficient as the cytomegalovirus (CMV) promoter to drive expression in vascular smooth muscle cells both in vitro and in vivo. In contrast to the CMV promoter it is activated in a Ca(2+)-dependent manner after endoplasmic reticulum depletion and allows the transgene expression only in proliferative/diseased cells. Overexpression of sarco/endoplasmic reticulum (SR/ER) Ca(2+) ATPase 2a under the control of this NFAT promoter inhibits restenosis after angioplasty in rats. In conclusion, this promoter may be useful for gene therapy in vascular proliferative diseases and other diseases involving upregulation of the NFAT pathway.


Subject(s)
Calcium/metabolism , Genetic Therapy/methods , Myocytes, Smooth Muscle/metabolism , NFATC Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Adenoviridae/genetics , Animals , Carotid Artery Injuries/genetics , Carotid Artery Injuries/therapy , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Culture Media/pharmacology , Cytomegalovirus/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Serum
5.
J Cardiovasc Pharmacol ; 35(5): 806-13, 2000 May.
Article in English | MEDLINE | ID: mdl-10813385

ABSTRACT

Mechanisms determining the benefit of beta blockade in patients with heart failure remain incompletely understood but are assumed consequent to prevention of deleterious effects of catecholamines. Recent studies have demonstrated that oxidative stress in congestive heart failure may be related to increased catecholamine levels. The aim of this study was to examine effects of long-term treatment with propranolol on progression of left ventricular (LV) dysfunction, remodeling and oxidative stress on an experimental model of chronic heart failure. Six weeks after myocardial infarction by coronary ligation, Wistar rats were randomized to two groups: 10 weeks of therapy with propranolol (50 mg/kg/day in drinking water) and no treatment (infarcted controls). A third group was sham-operated rats without treatment. Animals were anesthetized for hemodynamic measurements, and hearts were then removed for histologic analysis, papillary muscle contractility study, and oxidative stress measurements using thiobarbituric acid reactive substance (TBARS) determination. Control infarcted rats demonstrated significant alterations of hemodynamic parameters and remodeling with increase of heart weight/body weight, of right ventricular lateral wall thickness, of LV circumference, LV septal area/body weight, and LV papillary muscle weight/body weight as compared with sham. In propranolol-treated rats, hypertrophy of the LV septum, papillary muscle, and right ventricle were similar to those of the infarcted control. Myocardial oxidative stress was significantly increased in control infarcted rats compared with sham, and propranolol prevented such oxidative stress increase. Papillary muscle isometric tension parameters were not significantly different among groups. Propranolol treatment prevented isoprenaline-induced spontaneous papillary muscle activity in vitro. Oxidative stress is increased in the rat model of heart failure secondary to coronary ligation. Long-term treatment with propranolol in vivo does not modify the compensatory process of hypertrophy but completely abolishes the oxidative stress increase and reduces the increased cardiac sensitivity to catecholamine-induced arrhythmias observed in this experimental model of heart failure.


Subject(s)
Myocardial Infarction/drug therapy , Oxidative Stress/drug effects , Propranolol/pharmacology , Ventricular Remodeling/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Heart Failure/complications , Male , Muscle Contraction/drug effects , Myocardial Infarction/physiopathology , Papillary Muscles/drug effects , Papillary Muscles/physiopathology , Propranolol/therapeutic use , Rats , Rats, Wistar
6.
J Cardiovasc Pharmacol ; 32(6): 927-34, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869498

ABSTRACT

Cardiac fibrosis is linked to aldosterone-induced hypertension, but the effects on in vivo left ventricular (LV) function are not established. We studied the relations between in vivo LV function and aldosterone/salt cardiac fibrosis. Adult guinea pigs (GPs) were treated for 3 months with an aldosterone infusion and high-salt diet. This treatment induced arterial hypertension (+35%) and moderate LV hypertrophy (LVH; +60%) without right ventricular (RV) hypertrophy. Echo-Doppler LV assessment demonstrated unaltered cardiac output, stroke volume, or LV relaxation. Type I collagen messenger RNA (mRNA) was significantly increased in both ventricles (LV, +48%; RV, +77%) and accompanied by a significant increase in total collagen deposition (LV, from 0.52% in controls to 4.4% in treated GPs; RV, from 0.82 to 5.5% in treated GPs). Plasma norepinephrine levels increased 2.6-fold (p < 0.01) and correlated with the increase in collagen deposition in both ventricles. Collagen content was not correlated with hypertension or LVH. We conclude that aldosterone administration induces cardiac collagen accumulation and a sympathetic stimulation, which might preserve systolic and diastolic function.


Subject(s)
Collagen/biosynthesis , Hypertension/physiopathology , Myocardium/metabolism , Ventricular Function, Left , Aldosterone , Animals , Collagen/metabolism , Electrolytes/metabolism , Fibrosis/etiology , Guinea Pigs , Hormones/blood , Hypertension/chemically induced , Hypertension/complications , Hypertension/metabolism , Hypertrophy, Left Ventricular , Hypertrophy, Right Ventricular , Male , RNA, Messenger/metabolism , Salts
7.
Cardiovasc Res ; 38(2): 451-62, 1998 May.
Article in English | MEDLINE | ID: mdl-9709406

ABSTRACT

OBJECTIVE: The aim of this study was to determine whether changes in cardiac Na+,K(+)-ATPase subunits and Na+/Ca2+ exchanger expression are regulated in aldosterone-salt hypertensive guinea pigs. METHODS: Guinea pigs (GP) were unilaterally nephrectomized and randomized into three groups (aldosterone-salt; control-salt; control). After 90 days of treatment, echocardiographic M-mode assessment and right carotid arterial catheterization were performed in vivo, and plasma hormones and electrolytes were measured. mRNA and protein levels were studied by Northern and Western blot analysis. RESULTS: Aldosterone-salt treatment induced, (1) arterial hypertension (+40%) and LV hypertrophy (+60%) without altering LV-fractional shortening, (2) an increase in plasma norepinephrine levels (+262%) and suppression of renin activity. Northern blot analysis showed the presence of the mRNA encoding the three alpha isoforms and the beta 1 subunit of Na+,K(+)-ATPase in GP myocardium. In the aldosterone-salt group, levels of alpha 1 and beta 1 mRNAs were unchanged. alpha 2 mRNA was increased in both ventricles, whereas alpha 3 mRNA was increased in hypertrophied LV only. Furthermore, levels of the Na+/Ca2+ exchanger mRNA were decreased in both ventricles. At protein level, the two major transcripts (alpha 1 and alpha 2) were detected but alpha 3 isoform was not. Parallel changes in protein and mRNA accumulation of alpha 1 and alpha 2 isoforms were observed in hypertrophied LV. CONCLUSION: These results show that alpha 1 and alpha 2 isoforms are expressed in GP heart and that they are independently regulated in aldosterone-salt hypertension. Like the alpha 1 isoform in renal tissue, alpha 2 isoform is the main target of aldosterone-salt. Reciprocal expression of the Na+/Ca2+ exchanger and Na+,K(+)-ATPase suggests an adaptational mechanism which maintains an appropriate sodium gradient and calcium concentration in hypertensive myocardium.


Subject(s)
Hypertension/metabolism , Isoenzymes/metabolism , Myocardium/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Aldosterone , Animals , Blotting, Northern , Blotting, Western , Echocardiography , Guinea Pigs , Hypertension/diagnostic imaging , Hypertension/enzymology , Hypertrophy, Left Ventricular/metabolism , Isoenzymes/genetics , Male , Myocardium/enzymology , RNA, Messenger/analysis , Random Allocation , Sodium Chloride , Sodium-Potassium-Exchanging ATPase/genetics
8.
Atherosclerosis ; 133(2): 183-92, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9298678

ABSTRACT

Although it is established that highly oxidized LDL modify both vasodilator and vasoconstrictor responses in normal and atherosclerotic arterial tissue, there is a paucity of data on the relationship between the degree of the oxidative modification of LDL and vasomotor response. We therefore compared the impact of native LDL (Nat-LDL), and of partially (P-oxLDL), of moderately (M-oxLDL) and of highly oxidized LDL (H-oxLDL) on the vasomotor response of isolated human internal mammary artery and of rat thoracic aorta. Copper-mediated oxidative modification for up to 24 h at 37 degrees C was characterised by a progressive increase in the net negative electrical charge of LDL, and in the content of oxysterols; by contrast, lipid hydroperoxide and TBARS content peaked in M-oxLDL at 6 h. Neither basal vascular tone nor vasoconstriction induced by KCl (100 mmol/l) were modified significantly in arterial segments in relation to the degree of LDL oxidation. While Nat-LDL did not modify the contractile response of rat aorta to norepinephrine, increase in the degree of oxidative modification of LDL progressively and significantly shifted the norepinephrine response curve to the right (EC50 values for Nat-LDL, M-oxLDL and H-oxLDL: 1.2+/-0.5x10(-8), 3.5+/-1x10(-7), 1.3+/-0.4x10(-6) mol/l respectively) with reduction in the maximal effect (74.5+/-12.2 and 100.1+/-6.2% for H-oxLDL and M-oxLDL respectively, P < 0.05 versus controls). Similar findings were made in human arteries treated with H-oxLDL (P < 0.05 for EC50 and maximal response versus controls). The acetylcholine-induced, endothelial-dependent relaxation of rat aortic segments was significantly and progressively impaired with increase in the degree of LDL oxidation, maximal relaxation with H-oxLDL being 3-fold less (P < 0.05) than Nat-LDL at the same protein concentration (100 microg/ml). Acetylated LDL was without effect. Our data indicate that the increase in the degree of copper-mediated, oxidative modification of LDL parallels progressive reduction in the vasomotor response of the arterial wall to norepinephrine-induced contraction and to acetylcholine-induced relaxation subsequent to precontraction. Our data are consistent with the hypothesis that the major oxysterols (7-ketocholesterol, 7beta-hydroxycholesterol) present in Ox-LDL underlie such effects.


Subject(s)
Arteries/drug effects , Lipoproteins, LDL/pharmacology , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Arteries/innervation , Arteries/metabolism , Basal Metabolism/drug effects , Breast/blood supply , Breast/drug effects , Cholesterol/metabolism , Copper/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Lipid Peroxides/metabolism , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Norepinephrine/pharmacology , Oxidation-Reduction , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Vasomotor System/physiology
9.
J Cardiovasc Pharmacol ; 28(1): 11-8, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8797130

ABSTRACT

Therapeutic response to angiotensin-converting enzyme (ACE) inhibitors was reported to be better related to tissular than to circulating levels of ACE inhibition, especially during chronic therapy. We studied the relations between plasma concentrations of angiotensin I (AI), plasma renin activity (PRA), angiotensin II (AII), and aldosterone (by radioimmunoassay, RIA) and levels of serum and tissue ACE activities during acute and chronic quinapril administration in rats. Forty-eight male Wistar rats received quinapril by gavage for either 1 day (n = 24) or 15 days (n = 24) at different doses (control, 0.1, 1, and 10 mg/kg/day; 6 rats at each dose). Plasma hormonal parameters, serum, and tissue (lung, heart, and aorta) ACE activities were measured 3 h after the last gavage. Significant dose-dependent inhibitions of serum and lung ACE during acute and chronic treatments were observed (p < 0.05). Degrees of serum and heart ACE inhibition (at 0.1 mg/kg/day) were significantly lower with chronic than with acute treatment (p < 0.05). Degree of inhibition in lung, which represents the main source of total ACE, was similar during acute and chronic treatments. Among plasma hormonal parameters, plasma AI was correlated to PRA and showed the best correlation with ACE inhibition. After logarithmic transformation, log AI was significantly correlated to ACE activity in lung during chronic treatment (r = -0.85, p < 0.05). This parameter may provide a useful index for ACE inhibitor dosage adjustment during chronic therapy.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensins/blood , Isoquinolines/pharmacology , Peptidyl-Dipeptidase A/metabolism , Tetrahydroisoquinolines , Aldosterone/blood , Animals , Aorta/drug effects , Aorta/enzymology , Heart/drug effects , Lung/drug effects , Lung/enzymology , Male , Myocardium/enzymology , Quinapril , Rats , Rats, Wistar , Renin/blood , Time Factors
10.
Cryobiology ; 32(4): 327-33, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7656567

ABSTRACT

The ability of human internal mammary artery smooth muscle cells to maintain histoenzymatic activity and contractile response after various times of cold anoxia prior to and following cryostorage was evaluated. The results showed that the enzyme histochemical status of human mammary arteries was largely unchanged after both cold anoxia and cryopreservation. Neither in fresh nor in cryopreserved mammary arteries did cold anoxia for up to 24 h change maximal contractile responses to potassium depolarization and norepinephrine. However, compared to unfrozen controls, the contractile responses were significantly reduced in cryopreserved mammary arteries. In conclusion, after cryopreservation of human mammary arteries, the enzyme activities were globally maintained, whereas the contractile responses were reduced. For up to 24 h after harvesting cold anoxia at 4 degrees C is well tolerated and allows preservation of metabolic and functional properties of these arteries.


Subject(s)
Cryopreservation/methods , Mammary Arteries/physiology , Cell Hypoxia , Cold Temperature , Histocytochemistry , Humans , Hydrolases/metabolism , In Vitro Techniques , Mammary Arteries/cytology , Mammary Arteries/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocardial Revascularization , Norepinephrine/pharmacology , Oxidoreductases/metabolism , Potassium Chloride/pharmacology , Time Factors , Vasoconstriction/drug effects
11.
Cell Mol Biol (Noisy-le-grand) ; 40(6): 803-10, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7812188

ABSTRACT

Use of cryopreserved small-caliber arterial allografts for arterial bypass procedures has been suggested. In addition to immunological tolerance, long term in vivo success of these grafts may be dependent on the viability of arterial cells after cryopreservation. Metabolic and functional capabilities of arterial smooth muscle cells were evaluated by studying the enzyme histochemical expressions of sheep carotid arteries after various time of cryopreservation (7 days, 90 to 150 days) and their contractile responses after freezing. The results indicated that cryopreserved arteries exhibited 4 features: (a) the enzyme activities were globally maintained, (b) the spontaneous endogenous oxido-reduction (NitroBT test) was reduced, (c) contractile responses against KCl and norepinephrine were abolished, (d) metabolic status of frozen arteries was independent of the duration of cryopreservation. These data suggest that cryopreserved arterial muscle cells may be depleted in electron donors and/or energy-rich nucleotides substrates. This defect is present after short or long time of cryopreservation.


Subject(s)
Carotid Arteries , Cryopreservation , Hydrolases/analysis , Muscle, Smooth, Vascular , Oxidoreductases/analysis , Animals , Biomarkers , Carotid Arteries/enzymology , Carotid Arteries/physiology , Female , Histocytochemistry , Muscle Contraction , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Reference Values , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...