Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
FASEB J ; 30(6): 2123-34, 2016 06.
Article in English | MEDLINE | ID: mdl-26891734

ABSTRACT

Higher cardiovascular morbidity in patients with a wide range of autoimmune diseases highlights the importance of autoimmunity in promoting atherosclerosis. Our purpose was to investigate the mechanisms of accelerated atherosclerosis and identified vascular autoantigens targeted by autoimmunity. We created a mouse model of autoimmunity-associated atherosclerosis by transplanting bone marrow from FcγRIIB knockout (FcRIIB(-/-)) mice into LDL receptor knockout mice. We characterized the cellular and molecular mechanisms of atherogenesis and identified specific aortic autoantigens using serologic proteomic studies. En face lesion area analysis showed more aggressive atherosclerosis in autoimmune mice compared with control mice (0.64 ± 0.12 vs 0.32 ± 0.05 mm(2); P < 0.05, respectively). At the cellular level, FcRIIB(-/-) macrophages showed significant reduction (46-72%) in phagocytic capabilities. Proteomic analysis revealed circulating autoantibodies in autoimmune mice that targeted 25 atherosclerotic lesion proteins, including essential components of adhesion complex, cytoskeleton, and extracellular matrix, and proteins involved in critical functions and pathways. Microscopic examination of atherosclerotic plaques revealed essential colocalization of autoantibodies with endothelial cells, their adherence to basement membranes, the internal elastica lamina, and necrotic cores. The new vascular autoimmunosome may be a useful target for diagnostic and immunotherapeutic interventions in autoimmunity-associated diseases that have accelerated atherosclerosis.-Merched, A. J., Daret, D., Li, L., Franzl, N., Sauvage-Merched, M. Specific autoantigens in experimental autoimmunity-associated atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Autoantigens/physiology , Gene Expression Regulation/physiology , Receptors, IgG/metabolism , Receptors, LDL/metabolism , Animals , Antibody Specificity , Female , Macrophages/physiology , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , Proteomics , Receptors, IgG/genetics , Receptors, LDL/genetics
2.
Mol Ther ; 18(8): 1545-52, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20551912

ABSTRACT

Mesenchymal stem cells (MSC) are multipotent postnatal stem cells, involved in the treatment of ischemic vascular diseases. We investigate the ability of MSC, exposed to short-term hypoxic conditions, to participate in vascular and tissue regeneration in an in vivo model of hindlimb ischemia. Transplantation of hypoxic preconditioned murine MSC (HypMSC) enhanced skeletal muscle regeneration at day 7, improved blood flow and vascular formation compared to injected nonpreconditioned MSC (NormMSC). These observed effects were correlated with an increase in HypMSC engraftment and a putative role in necrotic skeletal muscle fiber clearance. Moreover, HypMSC transplantation resulted in a large increase in Wnt4 (wingless-related MMTV integration site 4) expression and we demonstrate its functional significance on MSC proliferation and migration, endothelial cell (EC) migration, as well as myoblast differentiation. Furthermore, suppression of Wnt4 expression in HypMSC, abrogated the hypoxia-induced vascular regenerative properties of these cells in the mouse hindlimb ischemia model. Our data suggest that hypoxic preconditioning plays a critical role in the functional capabilities of MSC, shifting MSC location in situ to enhance ischemic tissue recovery, facilitating vascular cell mobilization, and skeletal muscle fiber regeneration via a paracrine Wnt-dependent mechanism.


Subject(s)
Hindlimb/metabolism , Hindlimb/pathology , Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Wnt Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Wnt Proteins/genetics , Wnt4 Protein
3.
Circ Heart Fail ; 3(3): 431-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20200330

ABSTRACT

BACKGROUND: Inflammatory processes play a critical role in myocarditis, dilated cardiomyopathy, and heart failure. The expression of the inflammatory chemokine osteopontin (OPN) is dramatically increased in cardiomyocytes and inflammatory cells during myocarditis and heart failure in human and animals. However, its role in the development of heart diseases is not known. METHODS AND RESULTS: To understand whether OPN is involved in cardiomyopathies, we generated a transgenic mouse (MHC-OPN) that specifically overexpresses OPN in cardiomyocytes with cardiac-specific promoter-directed OPN expression. Young MHC-OPN mice were phenotypically indistinguishable from their control littermates, but most of them died prematurely with a half-life of 12 weeks of age. Electrocardiography revealed conduction defects. Echocardiography showed left ventricular dilation and systolic dysfunction. Histological analysis revealed cardiomyocyte loss, severe fibrosis, and inflammatory cell infiltration. Most of these inflammatory cells were activated T cells with Th1 polarization and cytotoxic activity. Autoantibodies against OPN, cardiac myosin, or troponin I, were not found in the serum of MHC-OPN mice. CONCLUSIONS: These data show that OPN expression in the heart induces in vivo T-cell recruitment and activation leading to chronic myocarditis, the consequence of which is myocyte destruction and hence, dilated cardiomyopathy. Thus, OPN might therefore constitute a potential therapeutic target to limit heart failure.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Heart Failure/metabolism , Heart Failure/pathology , Myocytes, Cardiac/metabolism , Osteopontin/metabolism , Animals , Cardiomyopathy, Dilated/etiology , Disease Models, Animal , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Heart Failure/etiology , Lymphocyte Activation , Mice , Mice, Transgenic , Myocarditis/etiology , Myocarditis/metabolism , Myocarditis/pathology , Neutrophil Infiltration
4.
Stem Cells ; 26(11): 2991-3001, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18757297

ABSTRACT

Mesenchymal stem cell (MSC) transplantation offers a great angiogenic opportunity in vascular regenerative medicine. The canonical Wnt/beta-catenin signaling pathway has been demonstrated to play an essential role in stem cell fate. Recently, genetic studies have implicated the Wnt/Frizzled (Fz) molecular pathway, namely Wnt7B and Fz4, in blood growth regulation. Here, we investigated whether MSC could be required in shaping a functional vasculature and whether secreted Frizzled-related protein-1 (sFRP1), a modulator of the Wnt/Fz pathway, could modify MSC capacities, endowing MSC to increase vessel maturation. In the engraftment model, we show that murine bone marrow-derived MSC induced a beneficial vascular effect through a direct cellular contribution to vascular cells. MSC quickly organized into primitive immature vessel tubes connected to host circulation; this organization preceded host endothelial cell (EC) and smooth muscle cell (SMC) recruitment to later form mature neovessel. MSC sustained neovessel organization and maturation. We report here that sFRP1 forced expression enhanced MSC surrounding neovessel, which was correlated with an increase in vessel maturation and functionality. In vitro, sFRP1 strongly increased platelet-derived growth factor-BB (PDGF-BB) expression in MSC and enhanced beta-catenin-dependent cell-cell contacts between MSC themselves and EC or SMC. In vivo, sFRP1 increased their functional integration around neovessels and vessel maturation through a glycogen synthase kinase 3 beta (GSK3beta)-dependent pathway. sFRP1-overexpressing MSC compared with control MSC were well elongated and in a closer contact with the vascular wall, conditions required to achieve an organized mature vessel wall. We propose that genetically modifying MSC to overexpress sFRP1 may be potentially effective in promoting therapeutic angiogenesis/arteriogenesis processes. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/physiology , Proteins/metabolism , Animals , Becaplermin , Cattle , Cell Adhesion/physiology , Cells, Cultured , Collagen , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/physiology , Intracellular Signaling Peptides and Proteins , Laminin , Lentivirus/genetics , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Platelet-Derived Growth Factor/metabolism , Proteins/genetics , Proteoglycans , Proto-Oncogene Proteins c-sis , Transplantation, Heterologous , beta Catenin/metabolism
5.
Arterioscler Thromb Vasc Biol ; 28(12): 2131-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18772499

ABSTRACT

OBJECTIVE: Estradiol (E(2)) is known to accelerate reendothelialization and thus prevent intimal thickening and in-stent restenosis after angioplasty. Transplantation experiments with ERalpha(-/-) mice have previously shown that E(2) acts through local and bone marrow cell compartments to enhance endothelial healing. However, the downstream mechanisms induced by E(2) to mediate endothelial repair are still poorly understood. METHODS AND RESULTS: We show here that after endovascular carotid artery injury, E(2)-enhanced endothelial repair is lost in osteopontin-deficient mice (OPN(-/-)). Transplantation of OPN(-/-) bone marrow into wild-type lethally irradiated mice, and vice versa, suggested that osteopontin plays a crucial role in both the local and the bone marrow actions of E(2). In the vascular compartment, using transgenic mice expressing doxycyclin regulatable-osteopontin, we show that endothelial cell specific osteopontin overexpression mimics E(2)-enhanced endothelial cell migration and proliferation in the regenerating endothelium. In the bone marrow cell compartment, we demonstrate that E(2) enhances bone marrow-derived mononuclear cell adhesion to regenerating endothelium in vivo, and that this effect is dependent on osteopontin. CONCLUSIONS: We demonstrate here that E(2) acceleration of the endothelial repair requires osteopontin, both for bone marrow-derived cell recruitment and for endothelial cell migration and proliferation.


Subject(s)
Carotid Artery Injuries/physiopathology , Endothelial Cells/cytology , Endothelial Cells/physiology , Estradiol/pharmacology , Osteopontin/physiology , Animals , Bone Marrow Transplantation , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Female , Mice , Mice, Knockout , Mice, Transgenic , Osteopontin/deficiency , Osteopontin/genetics , Regeneration/drug effects , Regeneration/physiology
6.
Eur J Cardiothorac Surg ; 34(2): 248-54, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18457957

ABSTRACT

OBJECTIVES: To assess, using an in vivo engraftment strategy combining bone marrow cell (BMC) transplantation and tissue cardiomyoplasty, the functional outcome of distinct vascular progenitor cell therapy (endothelial progenitor (EPC) and mesenchymal stem (MSC) cells) at distance of myocardium infarction (MI). The study was also designed to test whether scaffold mixing progenitors with unfractionated BMC could improve progenitor recruitment in the damaged myocardium. METHODS: To track engrafted progenitor cells in vivo, cultured murine MSC and EPC were transduced with eGFP lentiviruses. Thirty days after cryogenical induction of MI, C57BL/6J mice were randomized to receive muscle patch placement coated or not (control group), labeled EPC or MSC mixed to the ration of 1:10, or not with unfractionated BMC. Two weeks after transplantation, cardiac function was recorded and heart sections were examined to detect GFP-labeled progenitor cells and analyze cell differentiation. RESULTS: This study showed that either type of mono cell therapy improved angiogenesis and cell survival in the scar but only MSC exhibited the capacity to invade the scar. We found no evidence of myocardial or vascular regeneration from progenitor cells. Engraftment of the progenitors/unfractionated BMC mix increased repopulation and thickness of the scar. CONCLUSION: Combined therapy with unfractionated BMC and expanded MSC appeared thus promising for scar repopulation.


Subject(s)
Myocardial Infarction/therapy , Pericardium/pathology , Stem Cell Transplantation/methods , Abdominal Muscles/transplantation , Animals , Cell Differentiation , Cell Movement , Cell Survival , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Heart/physiology , Immunophenotyping , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth/physiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic , Regeneration
7.
J Biol Chem ; 281(52): 40135-43, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17068330

ABSTRACT

Our aim was to investigate by in vivo biopanning the lesions developed early in atherosclerosis and identify human antibodies that home to diseased regions. We have designed a two-step approach for a rapid isolation of human Monoclonal phage-display single-chain antibodies (MoPhabs) reactive with proteins found in lesions developed in an animal model of atherosclerosis. After a single round of in vivo biopanning, the MoPhabs were eluted from diseased sections of rabbit aorta identified by histology and NMR microscopy. MoPhabs expressed in situ were selected by subtractive colony filter screening for their capacity to recognize atherosclerotic but not normal aorta. MoPhabs selected by our method predominantly bind atherosclerotic lesions. Two of them, B3.3G and B3.GER, produced as scFv fragments, recognized an epitope present on the surface in early atherosclerotic lesions and within the intimal thickness in more complex plaques. These human MoPhabs homed to atherosclerotic lesions in ApoE(-/-) mice after in vivo injection. A protein of approximately 56 kDa recognized by B3.3G was affinity-purified and identified by mass spectrometry analysis as vitronectin. This is the first time that single round in vivo biopanning has been used to select human antibodies as candidates for diagnostic imaging and for obtaining insight into targets displayed in atherosclerotic plaques.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/pathology , Bacteriophage M13/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/isolation & purification , Peptide Library , Animals , Antibodies, Monoclonal/metabolism , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/diagnosis , Binding Sites, Antibody/genetics , Genetic Vectors , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/isolation & purification , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rabbits , Vitronectin/genetics , Vitronectin/immunology , Vitronectin/metabolism
8.
Clin Exp Pharmacol Physiol ; 33(1-2): 131-8, 2006.
Article in English | MEDLINE | ID: mdl-16445712

ABSTRACT

1. Smooth muscle cells (SMC) participate in both inflammatory and dedifferentiation processes during atherosclerosis, as well as during mechanical injury following angioplasty. In the latter, we studied medial SMC differentiation and inflammation processes implicated early after de-endothelialization in relation to mechanical stresses. We hypothesized that activation of a subpopulation of SMC within the media plays a crucial role in the early phase of neointimal formation. 2. For this purpose, we used a rabbit model of balloon injury to study activation and differentiation of medial SMC in the early time after denudation and just before neointima thickening. Inflammation was evaluated by the expression of vascular cell adhesion molecule (VCAM)-1, integrin alpha4beta1 and nuclear factor (NF)-kB. Myosin isoforms and 2P1A2 antigen, a membrane protein expressed by rabbit dedifferentiated SMC, were used as markers of differentiation. 3. On day 2 after de-endothelialization, VCAM-1, alpha4beta1 and NF-kB were coexpressed by a well-defined subpopulation of SMC of the internal part of the media, in the vicinity of the blood stream. At the same time, the majority of SMC throughout the media expressed non-muscle myosin heavy chain-B (nm-MHC-B) and 2P1A2 antigen. On day 7, when intimal thickening appeared, SMC of the media were no longer activated, whereas some intimal SMC expressed the activation markers. Thus, after de-endothelialization, early dedifferentiation occurs in most of the medial SMC, whereas activation concerned only a subpopulation of SMC located in the internal media. Using the T-type voltage-operated calcium channel blocker mibefradil (0.1-1 micromol/L) in SMC culture, we showed that this agent exhibited an antiproliferative effect in a dose-dependent manner only on undifferentiated cells. 4. In conclusion, the results suggest that the activated SMC represent cells that are potentially able to migrate and participate in the intimal thickening process. Thus, the medial SMC inflammatory process, without any contribution of inflammatory cells, may represent a major mechanism underlying the development of intimal thickening following mechanical stress. In humans, inhibition of T-type calcium channels may be a tool to prevent the early proliferation step leading to neointimal formation.


Subject(s)
Aorta/metabolism , Myocytes, Smooth Muscle/metabolism , Tunica Media/metabolism , Animals , Aorta/drug effects , Aorta/injuries , Catheterization/adverse effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Immunohistochemistry , In Situ Hybridization , Inflammation/metabolism , Inflammation/pathology , Integrin alpha4beta1/analysis , Mibefradil/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myosin Heavy Chains/analysis , Myosin Heavy Chains/genetics , NF-kappa B/analysis , Rabbits , Time Factors , Tunica Media/drug effects , Tunica Media/pathology , Vascular Cell Adhesion Molecule-1/analysis , Vasodilator Agents/pharmacology
9.
Can J Cardiol ; 21(7): 563-8, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15940353

ABSTRACT

BACKGROUND: Cellular cardiomyoplasty has created new possibilities in cardiac regeneration. Several cell types can be used in the procedure, such as skeletal myoblasts and bone marrow cells. Recent publications have suggested that bone marrow cells may be excellent candidates due to their pluripotency, but their actual role in cardiac regeneration is unknown. OBJECTIVE: To evaluate the exact physiological role of bone marrow cells in the healing process after myocardial infarction. METHODS: A mouse bone marrow cell transplantation model was used in which transplanted cells were easily detectable by immunohistochemistry. Chimeric mice were subjected to myocardial infarction by ligation of the left descending coronary artery. After one month, the mice were sacrificed and the scars were analyzed. RESULTS: Transplanted bone marrow cells were detected in the scars and these cells seemed able to transdifferentiate into endothelial cells, but no transdifferentiation into cardiomyocytes occurred. This mechanism of regeneration was dismissed because only 2% of the vessels in the scars were positive for transplanted cells. CONCLUSIONS: Bone marrow cells might be involved in myocardial healing, but this physiological mechanism is insufficient to allow correct regeneration.


Subject(s)
Bone Marrow Transplantation/methods , Coronary Vessels/physiology , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Regeneration/physiology , Analysis of Variance , Animals , Coronary Circulation/physiology , Disease Models, Animal , Female , Graft Rejection , Graft Survival , Heart Function Tests , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion/methods , Probability , Risk Factors , Sensitivity and Specificity , Vascular Patency
10.
Circ Res ; 96(12): 1299-306, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15920021

ABSTRACT

Phosphorylation and subsequent inactivation of glycogen synthase kinase (GSK)-3beta via the Akt/PI3-Kinase pathway during ischemic preconditioning (PC) has been shown to be cardioprotective. As FrzA/sFRP-1, a secreted antagonist of the Wnt/Frizzled pathway, is expressed in the heart and is able to decrease the phosphorylation of GSK-3beta in vitro on vascular cells, we examined its effect during PC using transgenic mouse overexpressing FrzA in cardiomyocytes (alpha-MHC promoter) under a conditional transgene expression approach (tet-off system). Overexpression of FrzA inhibited the increase in GSK-3beta phosphorylation as well as protein kinase C (PKC) epsilon activation in transgenic mice after PC as compared with littermates. Phospho-Akt (P-Akt), phospho-JNK, or the cytoplasmic beta-catenin levels were not modified, phospho-p38 (P-p38) was slightly increased in transgenic mice after PC as compared with littermates. FrzA transgenic mice displayed a larger infarct size and a greater worsening of cardiac function compared with littermates. All these differences were reversed by the addition of doxycycline. This study demonstrates for the first time that disruption of a beta-catenin independent Wnt/Frizzled pathway induces the activation of GSK-3beta and reverses the benefit of preconditioning.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Ischemic Preconditioning, Myocardial , Membrane Proteins/physiology , Signal Transduction/physiology , Animals , Cytoskeletal Proteins/analysis , Doxycycline/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Mice, Transgenic , Myocardial Infarction/therapy , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Trans-Activators/analysis , Wnt Proteins , beta Catenin , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Can J Cardiol ; 20(14): 1467-75, 2004 Dec.
Article in French | MEDLINE | ID: mdl-15614343

ABSTRACT

Myocardial infarction involves scar-formation mechanisms in which inflammation, proliferation, cell differentiation, apoptosis and angiogenesis all play a role. Better knowledge of the scar-formation process would be helpful in developing new therapies. The authors have generated a mouse model for infarction because its possible application in transgenic mice would allow the role of target genes in postinfarction scar-formation mechanisms to be studied. An infarction is caused by ligating the descending branch of the left coronary artery. At various times after ligation, the mice are sacrificed to determine the size of the infarction, left ventricular function and the overall myocardial scar-formation process. Early mortality was 10%. Between the fourth and sixth day postsurgery, 25% of mice died of a ruptured, infarcted left ventricle. The size of the infarctions diminished with time, while the surface of the left ventricle increased. In hemodynamics, 15 and 30 days after infarction, left ventricle telediastolic pressure was higher, telesystolic pressure was lower and contractility in indexes had collapsed. After an inflammatory phase in which polynuclear neutrophils colonized the scar, granulation tissue set in with a proliferation of myofibroblasts and growth of new blood vessels. These cells disappeared from the scar gradually, leaving behind a matrix rich in collagen and devoid of any contractile properties. The authors have characterized a murine model of myocardial infarction, with applications in transgenic mice and in view of establishing new agents in postmyocardial infarction repair.


Subject(s)
Cicatrix/pathology , Coronary Vessels/pathology , Heart Rupture, Post-Infarction/pathology , Myocardial Infarction/surgery , Ventricular Function, Left/physiology , Animals , Cicatrix/etiology , Disease Models, Animal , Female , Heart Rupture, Post-Infarction/mortality , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Postoperative Complications/pathology , Risk Factors , Sensitivity and Specificity , Survival Analysis , Ventricular Remodeling
12.
J Histochem Cytochem ; 52(11): 1459-66, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15505340

ABSTRACT

Lysyl oxidases (Lox), which are members of the amine oxidase family, are involved in the maturation of elastic lamellae and collagen fibers. Modifications of amine oxidases in idiopathic annulo-aortic ectasia disease (IAAED) have never been investigated. Our aim was to examine the expression of several proteins that might interfere with elastic fiber organization in control (n=10) and IAAED (n=18) aortic tissues obtained at surgery. Expression of amine oxidases and semicarbazide-sensitive amine oxidase (SSAO), and cellular phenotypic markers were examined by immunohistopathology and confocal microscopy. The expression of these proteins was assessed in relation to clinical and histomorphological features of the arterial wall. In control aorta, SSAO staining was expressed along elastic lamellae, whereas in aneurysmal areas of IAAED, SSAO was markedly decreased, in association with severe disorganization of elastic lamellae. Smooth muscle myosin heavy chain was also decreased in IAAED compared with controls, indicating smooth muscle cell dedifferentiation. Multiple regression analysis showed that elastic lamellar thickness (ELT) was correlated positively with the SSAO:elastin ratio and negatively with the Lox:elastin ratio, and that the clinical features of IAAED (aneurysm, thoracic aorta diameter, and aortic insufficiency) were positively correlated with ELT but not with SSAO. The relationship between SSAO expression and ELT suggests that this amine oxidase may be involved in elastic fiber organization. However, in advanced IAAED, the deficit in SSAO expression could be secondary to the decrease and fragmentation of elastic fibers and/or to vascular smooth muscle cell dedifferentiation.


Subject(s)
Amine Oxidase (Copper-Containing)/biosynthesis , Aortic Aneurysm, Thoracic/metabolism , Aortic Valve Insufficiency/metabolism , Elastin/biosynthesis , Extracellular Matrix/metabolism , Protein-Lysine 6-Oxidase/biosynthesis , Aorta, Thoracic/enzymology , Aorta, Thoracic/metabolism , Aorta, Thoracic/ultrastructure , Aortic Aneurysm, Thoracic/complications , Aortic Aneurysm, Thoracic/enzymology , Aortic Valve Insufficiency/complications , Aortic Valve Insufficiency/enzymology , Cell Differentiation , Extracellular Matrix/enzymology , Extracellular Matrix/ultrastructure , Female , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myosin Heavy Chains/biosynthesis , Regression Analysis
13.
Ann Thorac Surg ; 78(4): 1409-17, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15464506

ABSTRACT

BACKGROUND: Myocardial infarction results in irreversible myocyte loss. In a murine model, we tested the feasibility of a novel repair technique combining bone marrow cell (BMC) transplantation and cardiomyoplasty. METHODS: Myocardial infarction was induced cryogenically in backcrossed ROSA 26 transgenic x C57BL/6J mice (n = 75). Thirty days later, surviving mice (n = 69) were randomized to sham treatment (rethoracotomy only; n = 11), patch only treatment (n = 29), or patch + BMC treatment (n = 29). Abdominal muscle patches were harvested from donor littermates not expressing the beta-galactosidase reporter gene and sutured on the epicardium directly above the infarct zone. Patch only-treated mice received uncoated patches. Patch + BMC-treated mice received patches coated with 5 x 10(6) beta-galactosidase-expressing BMCs embedded in a collagen-rich three-dimensional matrix. RESULTS: Mortality rate was 52% after muscle patch implantation. Bone marrow cells were able to migrate from muscle patch into the infarct zone, as demonstrated by beta-galactosidase immunostaining, and ultimately constituted 8% of all cells in scar tissue (mean +/- standard deviation, 219 +/- 111/mm2). Angiogenesis and cell survival in the scar were improved by patch + BMC treatment. Left ventricular geometry and cardiac function were improved by patch treatment, with or without BMC, although the effects were stronger after patch + BMC treatment. CONCLUSIONS: Epicardial deposition of a BMC-coated muscle patch is a promising approach to restoring cardiac function after myocardial infarction.


Subject(s)
Abdominal Muscles/transplantation , Bone Marrow Transplantation/methods , Myocardial Infarction/surgery , Pericardium/surgery , Tissue Engineering , Transplantation, Heterotopic , Animals , Cell Movement , Cicatrix/pathology , Collagen , Crosses, Genetic , Feasibility Studies , Genes, Reporter , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/pathology , Neovascularization, Physiologic , Random Allocation , Wound Healing
14.
Cardiovasc Res ; 64(1): 144-53, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15364622

ABSTRACT

OBJECTIVE: 7-Ketocholesterol, a major oxysterol in oxidized low-density lipoproteins in advanced atherosclerotic plaques, induces vascular smooth muscle cell (SMC) death. We investigated whether cytochrome c release participated in SMC death induced by 7-ketocholesterol and whether the processes were reversible. METHODS: SMC cultures derived from the rabbit aorta were exposed to 25 microM 7-ketocholesterol. Cytochrome c and Bax were studied by means of immunofluorescence and immunoblotting, apoptosis by the TUNEL technique and mitochondrial structure by transmission electron microscopy. RESULTS: 7-Ketocholesterol induced rapid upregulation of the proapoptotic protein Bax and its translocation from cytosol into the mitochondria (4 h). This was followed by mitochondrial cytochrome c release (65% at 8 h) into the cytosol, which was almost complete at 16 h. The mitochondria became spherical and ultracondensed, without showing signs of lysis. They clustered around the nucleus and were wrapped by wide cisternae of the rough endoplasmic reticulum. Cytochrome c release was not blocked by the pan-caspase inhibitor zVAD-fmk, in contrast to DNA fragmentation and SMC loss. Interestingly, upon removal of 7-ketocholesterol after 16 h and re-exposure to serum for 24 h, the mitochondrial cytochrome c content, their transmembrane potential and TUNEL labelling normalised and SMC loss decreased. However, none of these cell death markers was rescued when the SMCs had been exposed to the oxysterol for 24 h. CONCLUSION: The results indicate that cytochrome c release during oxysterol-induced SMC apoptosis is not caspase-dependent and occurs as a result of a reversible mitochondrial conformational change rather than swelling and rupture of the outer membrane. The reversibility of these events suggests that the apoptotic cascade could be arrested before a point of no return.


Subject(s)
Cytochromes c/metabolism , Ketocholesterols/pharmacology , Muscle, Smooth, Vascular/enzymology , Animals , Aorta , Apoptosis , Biological Transport , Biomarkers/analysis , Blotting, Western/methods , Cells, Cultured , Cytochromes c/analysis , Cytosol/chemistry , Cytosol/metabolism , DNA Fragmentation , Immunohistochemistry/methods , In Situ Nick-End Labeling , Microscopy, Electron , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Swelling , Muscle, Smooth, Vascular/ultrastructure , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , bcl-2-Associated X Protein
15.
Cardiovasc Res ; 63(4): 731-8, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15306229

ABSTRACT

OBJECTIVE: FrzA, a member of the group of secreted frizzled related proteins (sFRP) that is expressed in the cardiovascular system, has been shown to antagonize the Wnt/frizzled signaling pathway. We have recently demonstrated its role in vascular cell growth control in vitro. In this study, we aimed to examine the mechanisms by which FrzA exerts its antiproliferative effect on vascular cells in vitro and its potential effect in vivo. METHODS AND RESULTS: On synchronized, growth-arrested endothelial cells (EC) and smooth muscle cells (SMC) treated with the recombinant purified FrzA protein, flow cytometry analysis showed that the recombinant FrzA protein delayed G1 phase and entry into S-phase. Western blot experiments demonstrated that the treatment of EC or SMC with FrzA was associated with a decrease in the level of the cyclins and cyclin-dependent kinases and an increase in cytosolic phospho-beta-catenin levels. The FrzA-induced cell cycle delay was resolved by 24 h. C57BL/6J mice underwent surgery to produce unilateral hindlimb ischemia and empty adenoviruses (AdE) or adenoviruses coding for FrzA (AdFrzA) were injected at the time of the surgery. In AdFrzA-treated mice in the 7 days following surgery, we showed a decrease in cell proliferation, capillary density, and blood flow recovery and a reduced expression of cyclin and cdk activity in the ischemic muscle compared to that in the AdE-treated ischemic muscle. To gain insight into the pathway activated by FrzA overexpression, we showed an increase in the level of cytosolic phospho-beta-catenin, a marker of beta-catenin degradation, in AdFrzA-treated ischemic muscle compared to that in control AdE-treated ischemic muscle. CONCLUSION: We provided the first evidence that an impairment of the Wnt-Frizzled pathway, via FrzA overexpression, controlled proliferation and neovascularization after muscle ischemia.


Subject(s)
Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction/physiology , Adenoviridae/genetics , Animals , Blotting, Western/methods , Cell Division , Cells, Cultured , Cyclin-Dependent Kinases , Cyclins , Cytoskeletal Proteins/metabolism , Flow Cytometry , Genetic Vectors/administration & dosage , Hindlimb , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Trans-Activators/metabolism , Transduction, Genetic/methods , beta Catenin
16.
Eur J Cardiothorac Surg ; 25(1): 76-83, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14690736

ABSTRACT

OBJECTIVE: Frizzled A is a very recent protein expressed in the cardiovascular hood by cardiomyocytes and by endothelial cells. This protein plays key roles in vitro in vascular cell proliferation and is able to induce an in vivo angiogenic response. Regarding these properties, we assess the hypothesis that Frizzled A could act in the healing process after myocardial infarction. METHODS: To investigate the role of Frizzled A, we established a transgenic mouse line overexpressing the protein and developed a model of myocardial infarction by coronary artery ligation. RESULTS: The incidence of cardiac rupture after myocardial infarction was reduced in transgenic mice (6.5 versus 26.4% in controls, n=165; P<0.01). Infarct sizes were smaller in transgenic mice (18% of left ventricle circumference versus 28.1% in control at day 30; P<0.001; n=6) and the cardiac function was improved (3800 +/- 370 versus 2800 +/- 840 mmHg/s dp/dtmax in controls, -2800 +/- 440 versus -1800 +/- 211 dp/dtmin in controls at day 15; P<0.001; n=6). Early leukocyte infiltration had decreased in transgenic mice during the first week (103 +/- 59 versus 730 +/- 463 cells/mm2 in controls at day 7; P<0.001; n=6) and the apoptotic index was decreased by 50% at day 7. Capillary density in the scar was higher in transgenic mice (290 +/- 103 versus 104 +/- 43 vessels/mm2 in control at day 15; P<0.001) and vessels were more muscularized and mean lumen area was 3-fold higher (952 +/- 902 versus 313 +/- 350 microm2 in control; P<0.001). CONCLUSION: Overexpression of Frizzled A reduced the infarct size, improved cardiac recovery, modified inflammatory response and amplified angiogenesis. For these reasons, this protein would be of interest for cardiac surgeons using angiogenic therapy (as gene or protein injection) in ischemic heart diseases in non-revascularizable patients.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Heart Rupture, Post-Infarction/prevention & control , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Myocardial Infarction/metabolism , Animals , Cicatrix/metabolism , Male , Mice , Mice, Transgenic , Neovascularization, Physiologic
17.
Circulation ; 106(24): 3097-103, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12473558

ABSTRACT

BACKGROUND: The secreted frizzled related proteins (sFRP) are soluble proteins thought to interfere with the Wnt signaling. Our group previously demonstrated that one of these members, sFRP-1/FrzA, is strongly expressed during early phases of the vascularization process in embryonic vasculature and in the endothelium of arteries and capillaries in adults and modulated vascular cell proliferation. METHODS AND RESULTS: Analysis of the expression of sFRP-1 during cyclic ovarian angiogenesis revealed that sFRP-1 is expressed during the formation of neovessels and becomes undetectable when the vasculature is fully maturated. We then studied the role of FrzA in several distinct angiogenic models. FrzA induced angiogenesis in a chick chorioallantoic membrane model. Moreover, gene transfer of AdFrzA in grafted mesenchymal and glioma cells increased vessel density and tumor growth. FrzA induced formation of vessels, which were enlarged, longer, and appeared to be more mature compared with vessels formed under control treatments. In vitro, FrzA increased migration and tube formation of endothelial cells and seemed to protect them from apoptosis. FrzA-angiogenic effect in vitro was independent of vascular endothelial growth factor, fibroblast growth factor-2, or angiopiotin-1 induction and Akt activation. In contrast, FrzA decreased glycogen synthase kinase-3 phosphorylation. CONCLUSIONS: These results showed that FrzA has proangiogenic effects and suggest that Wnt signaling may be involved in normal differentiation as well as in the pathological development of vasculature.


Subject(s)
Membrane Proteins , Neovascularization, Pathologic/chemically induced , Neovascularization, Physiologic/drug effects , Protein Serine-Threonine Kinases , Proteins/pharmacology , Allantois/blood supply , Allantois/cytology , Allantois/drug effects , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Chick Embryo , Chorion/blood supply , Chorion/cytology , Chorion/drug effects , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Glioma/genetics , Glioma/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Intracellular Signaling Peptides and Proteins , Lymphokines/metabolism , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Morphogenesis/drug effects , Neoplasm Transplantation , Ovary/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Cardiovasc Res ; 53(4): 952-62, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11922905

ABSTRACT

OBJECTIVE: Smooth muscle cell (SMC) migration involves interactions with extracellular matrix (ECM) and is an important process in response to arterial wall injury. We investigated the expression and the functional role of vitronectin (VN) in the response after vascular injury. METHODS: VN and alpha v beta 3/beta 5 integrin expressions were investigated after balloon carotid injury of Sprague-Dawley rats. Adventitial delivery of blocking antibodies to VN, alpha v beta 5 and beta 3 integrins were performed to assess their roles in neointima formation. In vitro, migration assays were carried out on human SMC. RESULTS: Immunohistochemistry and in situ hybridization for VN showed an upregulation of VN during the early time points of intima formation. alpha v beta 3/beta 5 integrins expression correlated with VN expression. After 7 days, blocking antibodies to VN, alpha v beta 5 and beta 3 induced a significant decrease on intimal area associated with a decrease in intimal cell counts. A slight decrease in intimal cell proliferation without any effect on apoptosis was observed after VN blockade. In vitro, migrating SMC strongly expressed VN after injury and neutralizing anti-VN antibody inhibited SMC migration. Blocking experiment with anti-alpha v beta 5 and -alpha v beta 3 integrin antibodies showed that not only VN-alpha v beta 3 but also VN-alpha v beta 5 interactions are required for SMC migration. CONCLUSION: This study characterizes the VN-ECM interaction in SMC and supports the role of VN in mediating SMC migration and neointimal formation in response to injury.


Subject(s)
Angioplasty, Balloon/adverse effects , Carotid Artery Injuries/metabolism , Carotid Stenosis/therapy , Up-Regulation , Vitronectin/metabolism , Adult , Animals , Carotid Artery Injuries/etiology , Cell Division , Cell Movement , Cells, Cultured , Disease Models, Animal , Extracellular Matrix/metabolism , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , Muscle, Smooth, Vascular/pathology , Rats , Rats, Sprague-Dawley , Receptors, Vitronectin/metabolism , Recurrence , Tunica Intima/metabolism , Tunica Intima/pathology , Vitronectin/antagonists & inhibitors
19.
Am J Physiol Heart Circ Physiol ; 282(3): H821-31, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11834475

ABSTRACT

The aim of this study was to investigate mitochondrial alterations in an animal model of chronic myocardial ischemia in rats obtained by surgical constriction of the left coronary artery. Resting coronary blood flow was measured using the fluorescent microsphere technique. Contractile function, defined by rate-pressure product, and myocardial oxygen consumption were measured in a Langendorff preparation. The mitochondrial function was evaluated on permeabilized skinned fibers. Three weeks after surgery, ischemic hearts showed a significant decrease in coronary blood flow compared with sham. Hemodynamic measurements showed a significant systolic and diastolic dysfunction. Alterations in mitochondrial function in ischemic hearts were mainly characterized by a significant decrease in the maximal velocity and apparent half-saturation constant for ADP, loss of the stimulatory effect of creatine, and a stimulatory effect of exogenous cytochrome c. These functional alterations were supported by structural alterations characterized by mitochondrial clustering and swelling associated with membrane rupture. We conclude that the alterations in systolic function after chronic ischemia are supported by severe modifications of mitochondrial structure and function.


Subject(s)
Hemodynamics/physiology , Mitochondria, Heart/metabolism , Myocardial Ischemia/physiopathology , Adenosine Diphosphate/metabolism , Animals , Calcium/metabolism , Coronary Circulation , Coronary Vessels/physiology , Creatine/metabolism , Cytochrome c Group/metabolism , Energy Metabolism , In Vitro Techniques , Mitochondria, Heart/ultrastructure , Models, Cardiovascular , Myocardial Contraction , Myocardium/ultrastructure , Oxygen Consumption , Permeability , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...