Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 104(1): 147-59, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25139748

ABSTRACT

AIMS: Human thoracic aneurysm of the ascending aorta (TAA) is a chronic disease characterized by dilatation of the aortic wall, which can progress to vessel dissection and rupture. TAA has several aetiologies, but all forms present common features, including tissue remodelling. Here, we determined and characterized the angiogenic process associated with TAA and its relation with wall remodelling. METHODS AND RESULTS: Immunostaining for blood vessels showed an increased density of microvessels originating from the adventitia in the external medial layer of TAA compared with healthy aortas. Proteomic array analysis of 55 angiogenic factors in medial and adventitial layers showed different expression profiles in both tissue compartments between aneurysmal and healthy aortas. Quantification by ELISA confirmed that all forms of TAA contained higher levels of several pro- and anti-angiogenic factors, including angiopoietin-1 and -2, fibroblast growth factor-acidic, and thrombospondin-1, than that of healthy aortas. However, all groups showed comparable levels of vascular endothelial growth factor-A. Quantitative RT-PCR demonstrated that angiopoietins were overexpressed in TAA media. Immunostaining and electron microscopy revealed that neovessels had defective endothelial junctions and poor mural cell coverage. This incomplete structure was associated with the accumulation of plasminogen and albumin in the media of TAA. CONCLUSION: We describe, for the first time, leaky neovessel formation in TAA media in association with an imbalance of angiogenic factor levels. Although the initiating mechanisms of neo-angiogenesis in TAA and the potential aetiology-related differences remain to be determined, our results suggest that neo-angiogenesis could participate in TAA wall remodelling and weakening through deposition of blood-borne zymogens.


Subject(s)
Aorta, Thoracic/physiopathology , Aortic Aneurysm, Thoracic/physiopathology , Microvessels/physiopathology , Neovascularization, Pathologic , Vascular Remodeling , Adult , Aged , Aged, 80 and over , Angiogenic Proteins/analysis , Angiogenic Proteins/genetics , Aorta, Thoracic/chemistry , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Capillary Permeability , Case-Control Studies , Cell Differentiation , Dilatation, Pathologic , Female , Humans , Male , Microvessels/chemistry , Microvessels/pathology , Middle Aged , Phenotype , Proteomics/methods , Vasoconstriction
2.
Circ Res ; 113(7): 881-90, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23825360

ABSTRACT

RATIONALE: The activation of the Smad2 signaling pathway is thought to play an important role in human aneurysmal diseases as described by an important body of research. We previously showed that constitutive Smad2 activation is associated with Smad2 mRNA overexpression in aneurysmal vascular smooth muscle cells (VSMCs), which is dependent on epigenetic regulation of the SMAD2 promoter involving histone modifications. However, the underlying molecular mechanisms controlling Smad2 overexpression are currently unknown. OBJECTIVE: The aim of the present study is to understand the mechanisms regulating the constitutive Smad2 overexpression in VSMCs by identification of the histone-modifying enzymes, transcription factors, and cofactors responsible for Smad2 promoter activation in aneurysmal disease. METHODS AND RESULTS: This study was performed on medial tissue extracts and primary cultures of VSMCs of human thoracic aneurysms (n=17) and normal thoracic aortas (n=10). Here, we demonstrate that the activation of SMAD2 promoter is driven by the recruitment of a multipartner complex, including the transcription factor p53 and histone acetyltransferases. Remarkably, the transcriptional regulatory network of the SMAD2 promoter is dramatically altered in human aneurysmal VSMCs in vitro and in situ with a switch from Myc-dependent repression of SMAD2 in normal vessel to a p53-dependent activation of SMAD2 in aneurysms. Furthermore, histone acetyltransferases p300 and P300/CBP-associated protein play a major role in SMAD2 promoter activation by acting on histone acetylation, p53 recruitment, and acetylation. CONCLUSIONS: These results provide evidence for a major role of p53 and the complex composed of p300 and p300/CBP-associated protein in Smad2 activation in human aneurysmal VSMCs.


Subject(s)
Aortic Aneurysm/metabolism , Chromatin/metabolism , Myocytes, Smooth Muscle/metabolism , Smad2 Protein/metabolism , Adult , Aged , Aged, 80 and over , Aorta/pathology , Aortic Aneurysm/pathology , Chromatin Assembly and Disassembly , Female , Gene Regulatory Networks , Histone Acetyltransferases/metabolism , Humans , Male , Middle Aged , Promoter Regions, Genetic , RNA, Messenger/metabolism , Smad2 Protein/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/metabolism
3.
Arterioscler Thromb Vasc Biol ; 33(9): 2222-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23814118

ABSTRACT

OBJECTIVE: Tissue activation of proteolysis is involved in acute intramural rupture (dissections, acute ascending aortic dissection) and in progressive dilation (aneurysms, thoracic aneurysm of the ascending aorta) of human ascending aorta. The translational aim of this study was to characterize the regulation of antiproteolytic serpin expression in normal, aneurysmal, and dissecting aorta. APPROACH AND RESULTS: We explored expression of protease nexin-1 (PN-1) and plasminogen activator inhibitor-1 and their regulation by the Smad2 signaling pathway in human tissue and cultured vascular smooth muscle cells (VSMCs) of aneurysms (thoracic aneurysm of the ascending aorta; n=46) and acute dissections (acute ascending aortic dissection; n=10) of the ascending aorta compared with healthy aortas (n=10). Both PN-1 and plasminogen activator inhibitor-1 mRNA and proteins were overexpressed in medial tissue extracts and primary VSMC cultures from thoracic aneurysm of the ascending aorta compared with acute ascending aortic dissection and controls. Transforming growth factor-ß induced increased PN-1 expression in control but not in aneurysmal VSMCs. PN-1 and plasminogen activator inhibitor-1 overexpression by aneurysmal VSMCs was associated with increased Smad2 binding on their promoters and, functionally, resulted in VSMC self-protection from plasmin-induced detachment and death. This phenomenon was restricted to aneurysms and not observed in acute dissections. CONCLUSIONS: These results demonstrate that epigenetically regulated PN-1 overexpression promotes development of an antiproteolytic VSMC phenotype and might favor progressive aneurysmal dilation, whereas absence of this counter-regulation in dissections would lead to acute wall rupture.


Subject(s)
Aortic Aneurysm, Thoracic/metabolism , Aortic Dissection/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Serpin E2/metabolism , Smad2 Protein/metabolism , Acute Disease , Adult , Aged , Aged, 80 and over , Aortic Dissection/etiology , Aortic Dissection/genetics , Aortic Dissection/pathology , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Aortic Valve/abnormalities , Bicuspid Aortic Valve Disease , Binding Sites , Biomarkers/metabolism , Cells, Cultured , Chronic Disease , Female , Fibrillins , Genetic Predisposition to Disease , Genotype , Heart Valve Diseases/complications , Humans , Male , Marfan Syndrome/complications , Marfan Syndrome/genetics , Microfilament Proteins/genetics , Middle Aged , Muscle, Smooth, Vascular/pathology , Mutation , Myocytes, Smooth Muscle/pathology , Phenotype , Plasminogen Activator Inhibitor 1/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Risk Factors , Serpin E2/genetics , Time Factors , Transforming Growth Factor beta1/metabolism , Up-Regulation
4.
Circ Res ; 109(9): 1003-14, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21885829

ABSTRACT

RATIONALE: Neovascularization favors intraplaque hemorrhage and plaque rupture. Development of therapeutic strategies against atheromatous angiogenesis requires elucidation of its initiating factors. OBJECTIVE: We investigated the contribution of smooth muscle cells (SMCs) and atheroma-derived lipids to the initiation of atheroma-associated neoangiogenesis. METHODS AND RESULTS: Forty human aortic segments, each harvested from a different donor, were classified as healthy or as bearing early atheromatous lesions, including fatty streaks and fibrolipidic atheroma, according to their histological features. Immunostaining for blood vessels and vascular endothelial growth factor-A (VEGF-A), as well as measurement of VEGF-A protein and mRNA levels by ELISA and real-time PCR, revealed that angiogenesis and VEGF-A production were enhanced in the medial layer of atheromatous aortas. The intramedial vessel density and invasiveness and the production of VEGF-A by medial SMCs were indeed increased in atheromatous aortas compared with healthy aortas. Furthermore, intimal layers of atheromatous aortas were enriched in soluble lipid mediators capable of inducing a sustained increase in VEGF-A production by medial SMCs, turning these cells into potent inducers of angiogenesis when incorporated into mouse Matrigel implants. Both effects were inhibited by the peroxisome proliferator-activated receptor-γ inhibitor GW9662 and mimicked by its agonist, rosiglitazone. CONCLUSIONS: We show that VEGF-A production is upregulated in medial SMCs of human atheromatous aortas and that peroxisome proliferator-activated receptor-γ agonists derived from early intimal lesions are likely to contribute to this phenotypic change. Our findings suggest that medial SMCs are central organizers of an angiogenic response initiated by the subendothelial accumulation of atherogenic lipids.


Subject(s)
Aorta/physiopathology , Muscle, Smooth, Vascular/physiopathology , Neovascularization, Pathologic/physiopathology , PPAR gamma/physiology , Plaque, Atherosclerotic/physiopathology , Anilides/pharmacology , Aorta/metabolism , Cells, Cultured , Humans , Lipid Metabolism/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Plaque, Atherosclerotic/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology , Vascular Endothelial Growth Factor A/metabolism
5.
FEBS J ; 278(15): 2655-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21615688

ABSTRACT

Human disc-large homolog (hDlg), also known as synapse-associated protein 97, is a scaffold protein, a member of the membrane-associated guanylate kinase family, implicated in neuronal synapses and epithelial-epithelial cell junctions whose expression and function remains poorly characterized in most tissues, particularly in the vasculature. In human vascular tissues, hDlg is highly expressed in smooth muscle cells (VSMCs). Using the yeast two-hybrid system to screen a human aorta cDNA library, we identified mitogen-activated protein/extracellular signal-responsive kinase (ERK) kinase (MEK)2, a member of the ERK cascade, as an hDlg binding partner. Site-directed mutagenesis showed a major involvement of the PSD-95, disc-large, ZO-1 domain-2 of hDlg and the C-terminal sequence RTAV of MEK2 in this interaction. Coimmunoprecipitation assays in both human VSMCs and human embryonic kidney 293 cells, demonstrated that endogenous hDlg physically interacts with MEK2 but not with MEK1. Confocal microscopy suggested a colocalization of the two proteins at the inner layer of the plasma membrane of confluent human embryonic kidney 293 cells, and in a perinuclear area in human VSMCs. Additionally, hDlg also associates with the endoplasmic reticulum and microtubules in these latter cells. Taken together, these findings allow us to hypothesize that hDlg acts as a MEK2-specific scaffold protein for the ERK signaling pathway, and may improve our understanding of how scaffold proteins, such as hDlg, differentially tune MEK1/MEK2 signaling and cell responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MAP Kinase Kinase 2/metabolism , Membrane Proteins/metabolism , Cell Membrane/enzymology , Discs Large Homolog 1 Protein , Endoplasmic Reticulum/enzymology , HEK293 Cells , Humans , MAP Kinase Kinase 1/metabolism , Microtubules/enzymology , Muscle, Smooth, Vascular/cytology , Signal Transduction/genetics , Two-Hybrid System Techniques
6.
Cardiovasc Res ; 89(2): 446-56, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20829218

ABSTRACT

AIMS: Human thoracic aortic aneurysms (TAAs) are characterized by extracellular matrix breakdown associated with progressive smooth muscle cell (SMC) rarefaction. These features are present in all types of TAA: monogenic forms [mainly Marfan syndrome (MFS)], forms associated with bicuspid aortic valve (BAV), and degenerative forms. Initially described in a mouse model of MFS, the transforming growth factor-ß1 (TGF-ß1)/Smad2 signalling pathway is now assumed to play a role in TAA of various aetiologies. However, the relation between the aetiological diversity and the common cell phenotype with respect to TGF-ß signalling remains unexplained. METHODS AND RESULTS: This study was performed on human aortic samples, including TAA [MFS, n = 14; BAV, n = 15; and degenerative, n = 19] and normal aortas (n = 10) from which tissue extracts and human SMCs and fibroblasts were obtained. We show that all types of TAA share a complex dysregulation of Smad2 signalling, independent of TGF-ß1 in TAA-derived SMCs (pharmacological study, qPCR). The Smad2 dysregulation is characterized by an SMC-specific, heritable activation and overexpression of Smad2, compared with normal aortas. The cell specificity and heritability of this overexpression strongly suggest the implication of epigenetic control of Smad2 expression. By chromatin immunoprecipitation, we demonstrate that the increases in H3K9/14 acetylation and H3K4 methylation are involved in Smad2 overexpression in TAA, in a cell-specific and transcription start site-specific manner. CONCLUSION: Our results demonstrate the heritability, the cell specificity, and the independence with regard to TGF-ß1 and genetic backgrounds of the Smad2 dysregulation in human thoracic aneurysms and the involvement of epigenetic mechanisms regulating histone marks in this process.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Epigenesis, Genetic , Marfan Syndrome/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Acetylation , Adult , Aged , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Case-Control Studies , Cells, Cultured , Chromatin Assembly and Disassembly , DNA Methylation , Genetic Predisposition to Disease , Heredity , Histones/metabolism , Humans , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation , Young Adult
7.
Histopathology ; 57(6): 917-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21166705

ABSTRACT

AIMS: Thoracic ascending aortic aneurysms (TAA) are characterized by elastic fibre breakdown and cystic medial degeneration within the aortic media, associated with progressive smooth muscle cell (SMC) rarefaction. The transforming growth factor (TGF)-ß/Smad2 signalling pathway is involved in this process. Because the pericellular fibrinolytic system activation is able to degrade adhesive proteins, activate matrix metalloproteinase (MMP), induce SMC disappearance and increase the bioavailability of TGF-ß, the aim was to investigate the plasminergic system in TAA. METHODS AND RESULTS: Ascending aortas [21 controls and 19 TAAs (of three different aetiologies)] were analysed. Immunohistochemistry showed accumulation of t-PA, u-PA and plasmin in TAAs, associated with residual SMCs. Overexpression of t-PA and u-PA was confirmed by reverse transcription-polymerase chain reaction (RT-PCR), immunoblotting and zymography on TAA extracts and culture medium conditioned by TAA. Plasminogen was present on the SMC surface and inside cytoplasmic vesicles, but plasminogen mRNA was undetectable in the TAA medial layer. Plasmin-antiplasmin complexes were detected in TAA-conditioned medium and activation of the fibrinolytic system was associated with increased fibronectin turnover. Fibronectin-related material was detected immunohistochemically in dense clumps around SMCs and colocalized with latent TGF-ß binding protein-1. CONCLUSIONS: The fibrinolytic pathway could play a critical role in TAA progression, via direct or indirect impact on ECM and consecutive modulation of TGF-ß bioavailability.


Subject(s)
Aorta/metabolism , Aortic Aneurysm, Thoracic/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Adult , Aged , Aortic Aneurysm, Thoracic/genetics , Blotting, Western , Female , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Tissue Plasminogen Activator/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Urokinase-Type Plasminogen Activator/genetics
8.
Biomacromolecules ; 10(11): 3074-80, 2009 Nov 09.
Article in English | MEDLINE | ID: mdl-19761207

ABSTRACT

In patients receiving drug eluting stents, there is a growing concern about both the long-term toxicity/degradability of the polymers used for the coating, and the nature of the therapeutic agents. We hypothesized that the use of a functionalized biocompatible polymer for a stent coating could be appropriate for local arterial therapy. A cationized pullulan hydrogel was thus prepared to cover bare metal stents that could be further loaded with small interfering RNA (siRNA) targeted at MMP2 for gene silencing in vascular cells. The efficient coverage of the stent struts by a smooth polymeric layer, which can withstand the crimping of the stent on a balloon-catheter and its deployment, was demonstrated by fluorescence microscopy, scanning electron microscopy, and atomic force microscopy. The release of siRNA from the stents was modulated by the presence of the cationic groups, as compared to noncationized pullulan hydrogel. In vivo implantation of coated stents was successful and cationized pullulan-based hydrogels loaded with siRNA in rabbit balloon-injured carotid arteries induced an uptake of siRNA into the arterial wall and a decrease of pro-MMP2 activity. These results suggest that cationized pullulan-based hydrogel could be used as a new biocompatible and biodegradable stent coating for local gene therapy in the arterial wall.


Subject(s)
Carotid Arteries/surgery , Coated Materials, Biocompatible/administration & dosage , Coated Materials, Biocompatible/chemical synthesis , Drug-Eluting Stents , Gene Transfer Techniques , RNA, Small Interfering/administration & dosage , Animals , Carotid Arteries/physiology , Drug Discovery/methods , Genetic Therapy/methods , Polymers/administration & dosage , Polymers/chemistry , RNA, Small Interfering/genetics , Rabbits
9.
J Pathol ; 218(1): 131-42, 2009 May.
Article in English | MEDLINE | ID: mdl-19224541

ABSTRACT

Common features such as elastic fibre destruction, mucoid accumulation, and smooth muscle cell apoptosis are co-localized in aneurysms of the ascending aorta of various aetiologies. Recent experimental studies reported an activation of TGF-beta in aneurysms related to Marfan (and Loeys-Dietz) syndrome. Here we investigate TGF-beta signalling in normal and pathological human ascending aortic wall in syndromic and non-syndromic aneurysmal disease. Aneurysmal ascending aortic specimens, classified according to aetiology: syndromic MFS (n = 15, including two mutations in TGFBR2), associated with BAV (n = 15) or degenerative forms (n = 19), were examined. We show that the amounts of TGF-beta1 protein retained within and released by aneurysmal tissue were greater than for control aortic tissue, whatever the aetiology, contrasting with an unchanged TGF-beta1 mRNA level. The increase in stored TGF-beta1 was associated with enhanced LTBP-1 protein and mRNA levels. These dysregulations of the extracellular ligand are associated with higher phosphorylated Smad2 and Smad2 mRNA levels in the ascending aortic wall from all types of aneurysm. This activation correlated with the degree of elastic fibre fragmentation. Surprisingly, there was no consistent association between the nuclear location of pSmad2 and extracellular TGF-beta1 and LTBP-1 staining and between their respective mRNA expressions. In parallel, decorin was focally increased in aneurysmal media, whereas biglycan was globally decreased in aneurysmal aortas. In conclusion, this study highlights independent dysregulations of TGF-beta retention and Smad2 signalling in syndromic and non-syndromic aneurysms of the ascending aorta.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/metabolism , Marfan Syndrome/metabolism , Signal Transduction/physiology , Smad2 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Aged , Aged, 80 and over , Aorta/pathology , Aortic Aneurysm/complications , Aortic Aneurysm/genetics , Biomarkers/analysis , Case-Control Studies , Cell Differentiation , Gene Expression , Humans , Immunoblotting/methods , Immunohistochemistry , Latent TGF-beta Binding Proteins/analysis , Latent TGF-beta Binding Proteins/genetics , Marfan Syndrome/complications , Marfan Syndrome/genetics , Middle Aged , Muscle, Smooth, Vascular/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/analysis , Smad2 Protein/genetics , Statistics, Nonparametric , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/genetics
10.
J Gene Med ; 11(1): 92-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19040207

ABSTRACT

BACKGROUND: Small interfering RNA (siRNA) delivery is a promising approach for the treatment of cardiovascular diseases. Matrix metalloproteinase (MMP) 2 over-expression in the arterial wall has been implicated in restenosis after percutaneous coronary intervention, as well as in spontaneous atherosclerotic plaque rupture. We hypothesized that in vivo local delivery of siRNA targeted at MMP2 (MMP2-siRNA) in the balloon-injured carotid artery of hypercholesterolemic rabbits may lead to inhibition of MMP2 expression. METHODS: Two weeks after balloon injury, 5 micromol/l of Tamra-tagged MMP2-siRNA, scramble siRNA or saline was locally injected in the carotid artery and incubated for 1 h. RESULTS: Fluorescent microscopy studies showed the circumferential uptake of siRNA in the superficial layers of neointimal cells. MMP2 mRNA levels, measured by the real-time reverse transcriptase-polymerase chain reaction, was decreased by 79 +/- 25% in MMP2-siRNA- versus scramble siRNA-transfected arteries (p < 0.05). MMP2 activity, measured by gelatin zymography performed on the conditioned media of MMP2-siRNA versus scramble siRNA transfected arteries, decreased by 53 +/- 29%, 50 +/- 24% and 46 +/- 14% at 24, 48 and 72 h, respectively (p < 0.005 for all). No effect was observed on MMP9, pro-MMP9 and TIMP-2 levels. CONCLUSIONS: The results obtained in the present study suggest that significant inhibition of gene expression can be achieved with local delivery of siRNA in the arterial wall in vivo.


Subject(s)
Carotid Arteries/enzymology , Carotid Arteries/pathology , Carotid Artery Injuries/therapy , Matrix Metalloproteinase Inhibitors , RNA Interference , RNA, Small Interfering/administration & dosage , Angioplasty, Balloon/adverse effects , Animals , Carotid Artery Injuries/genetics , Disease Models, Animal , Immunohistochemistry , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rabbits , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transfection
11.
Rejuvenation Res ; 11(5): 883-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18803461

ABSTRACT

The extracellular matrix (ECM) plays an important role in vascular tissue structure, maintenance, and function. Lysyl oxidases catalyze a key step in the posttranslational cross-linking of elastin and collagens in the ECM. Gene knockout studies in mice suggested a role for lysyl oxidase-like (LOXL1) in adult elastin synthesis and a role for its isoform, lysyl oxidase (LOX), in the synthesis of both collagens and elastin during development. However, the relative expression of both isoforms as a function of age is not known and was therefore investigated here. LOX and LOXL1 immunohistochemistry and real-time RT-PCR were performed during development, growth and aging in the aorta of LOU and Brown-Norway (BN) rats, two inbred strains with different susceptibilities to arterial fragility. In addition, expression of genes encoding for elastic fiber proteins and type I collagen, together with elastin and collagen contents, was measured in adult and old rat aortas. High aortic LOX expression was observed early in the development (embryonic day 15), followed by a drastic reduction in adulthood, whereas LOXL1 was mainly detectable in the intima and media; its expression was maintained throughout life in the LOU rat. Expression of tropoelastin, type-I collagen, and LOXL1 genes was reduced in the aorta of 6-week-old BN rats. Aging is characterized by a decreased elastin/collagen ratio and a greatly decreased expression of LOX, tropoelastin, and type-I collagen. These findings indicate a different spatial and temporal expression of LOX and LOXL1 during growth and aging in the rat aorta and suggest specific roles for LOX and LOXL1 in the synthesis and remodeling of elastic and collagen fibers.


Subject(s)
Aging/genetics , Aging/metabolism , Aorta/metabolism , Collagen/metabolism , Elastin/metabolism , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Animals , Aorta/embryology , Aorta/growth & development , Base Sequence , Extracellular Matrix Proteins/genetics , Fibrillins , Gene Expression Regulation, Developmental , Male , Microfilament Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred Strains , Recombinant Proteins/genetics
12.
Am J Physiol Heart Circ Physiol ; 294(4): H1851-61, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18245566

ABSTRACT

Membrane- associated guanylate kinase proteins (MAGUKs) are important determinants of localization and organization of ion channels into specific plasma membrane domains. However, their exact role in channel function and cardiac excitability is not known. We examined the effect of synapse-associated protein 97 (SAP97), a MAGUK abundantly expressed in the heart, on the function and localization of Kv1.5 subunits in cardiac myocytes. Recombinant SAP97 or Kv1.5 subunits tagged with green fluorescent protein (GFP) were overexpressed in rat neonatal cardiac myocytes and in Chinese hamster ovary (CHO) cells from adenoviral or plasmidic vectors. Immunocytochemistry, fluorescence recovery after photobleaching, and patch-clamp techniques were used to study the effects of SAP97 on the localization, mobility, and function of Kv1.5 subunits. Adenovirus-mediated SAP97 overexpression in cardiac myocytes resulted in the clustering of endogenous Kv1.5 subunits at myocyte-myocyte contacts and an increase in both the maintained component of the outward K(+) current, I(Kur) (5.64 +/- 0.57 pA/pF in SAP97 myocytes vs. 3.23 +/- 0.43 pA/pF in controls) and the number of 4-aminopyridine-sensitive potassium channels in cell-attached membrane patches. In live myocytes, GFP-Kv1.5 subunits were mobile and organized in clusters at the basal plasma membrane, whereas SAP97 overexpression reduced their mobility. In CHO cells, Kv1.5 channels were diffusely distributed throughout the cell body and freely mobile. When coexpressed with SAP97, Kv subunits were organized in plaquelike clusters and poorly mobile. In conclusion, SAP97 regulates the K(+) current in cardiac myocytes by retaining and immobilizing Kv1.5 subunits in the plasma membrane. This new regulatory mechanism may contribute to the targeting of Kv channels in cardiac myocytes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Kv1.5 Potassium Channel/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Potassium/metabolism , 4-Aminopyridine/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adenoviridae/genetics , Animals , Animals, Newborn , CHO Cells , Cell Membrane/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Discs Large Homolog 1 Protein , Fluorescence Recovery After Photobleaching , Genetic Vectors , Immunohistochemistry , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/genetics , Membrane Potentials , Membrane Proteins/genetics , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Transport , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
13.
Rejuvenation Res ; 11(1): 97-112, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18173368

ABSTRACT

Elastin, the main component of elastic fibers, is synthesized only in early life and provides the blood vessels with their elastic properties. With aging, elastin is progressively degraded, leading to arterial enlargement, stiffening, and dysfunction. Also, elastin is a key regulator of vascular smooth muscle cell proliferation and migration during development since heterozygous mutations in its gene (Eln) are responsible for a severe obstructive vascular disease, supravalvular aortic stenosis, isolated or associated to Williams syndrome. Here, we have studied whether early elastin synthesis could also influence the aging processes, by comparing the structure and function of ascending aorta from 6- and 24-month-old Eln+/- and Eln+/+ mice. Eln+/- animals have high blood pressure and arteries with smaller diameters and more rigid walls containing additional although thinner elastic lamellas. Nevertheless, longevity of these animals is unaffected. In young adult Eln+/- mice, some features resemble vascular aging of wild-type animals: cardiac hypertrophy, loss of elasticity of the arterial wall through enhanced fragmentation of the elastic fibers, and extracellular matrix accumulation in the aortic wall, in particular in the intima. In Eln+/- animals, we also observed an age-dependent alteration of endothelial vasorelaxant function. On the contrary, Eln+/- mice were protected from several classical consequences of aging visible in aged Eln+/+ mice, such as arterial wall thickening and alteration of alpha(1)-adrenoceptor-mediated vasoconstriction. Our results suggest that early elastin expression and organization modify arterial aging through their impact on both vascular cell physiology and structure and mechanics of blood vessels.


Subject(s)
Aging/genetics , Aorta/physiology , Elastin/genetics , Loss of Heterozygosity/physiology , Aging/physiology , Animals , Aorta/cytology , Aorta/ultrastructure , Cardiovascular Physiological Phenomena , Desmosine/analysis , Elastin/chemistry , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Hydroxyproline/analysis , Male , Mice , Mice, Inbred C57BL
14.
Am J Physiol Heart Circ Physiol ; 293(6): H3593-601, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17890430

ABSTRACT

Matrix metalloproteinase-2 (MMP-2) is constitutively expressed in vascular smooth muscle cells (VSMCs). Using small interfering RNA (siRNA), we evaluated the effect of MMP-2 inhibition in VSMCs in vitro and ex vivo. Rabbit VSMCs were transfected in vitro with 50 nmol/l MMP-2 siRNA or scramble siRNA. Flow cytometry and confocal microscopy showed cellular uptake of siRNA in approximately 80% of VSMCs. MMP-2 mRNA levels evaluated by real-time RT-PCR, pro-MMP-2 activity from conditioned culture media evaluated by gelatin zymography, and VSMC migration were reduced by 44 +/- 19%, 43 +/- 14%, and 36 +/- 14%, respectively, in MMP-2 siRNA-transfected compared with scramble siRNA-transfected VSMCs (P < 0.005 for all). Ex vivo MMP-2 siRNA transfection was performed 2 wk after balloon injury of hypercholesterolemic rabbit carotid arteries. Fluorescence microscopy showed circumferential siRNA uptake in neointimal cells. Gelatin zymography of carotid artery culture medium demonstrated a significant decrease of pro-MMP-2 activity in MMP-2 siRNA-transfected compared with scramble siRNA-transfected arteries (P < 0.01). Overall, our results demonstrate that in vitro MMP-2 siRNA transfection in VSMCs markedly inhibits MMP-2 gene expression and VSMC migration and that ex vivo delivery of MMP-2 siRNA in balloon-injured arteries reduces pro-MMP-2 activity in neointimal cells, suggesting that siRNA could be used to modify arterial biology in vivo.


Subject(s)
Carotid Artery Injuries/therapy , Gene Expression Regulation, Enzymologic , Genetic Therapy/methods , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , RNA Interference , RNA, Small Interfering/metabolism , Angioplasty, Balloon/adverse effects , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cell Line , Cell Movement , Culture Media, Conditioned/metabolism , Disease Models, Animal , Enzyme Activation , Enzyme Precursors/metabolism , Feasibility Studies , Flow Cytometry , Matrix Metalloproteinase 2/genetics , Microscopy, Confocal , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , RNA, Messenger/metabolism , Rabbits , Time Factors , Transfection
15.
Cardiovasc Res ; 72(1): 69-79, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16839528

ABSTRACT

OBJECTIVES: The expression and distribution of connexins is abnormal in a number of cardiac diseases, including atrial fibrillation, and is believed to favor conduction slowing and arrhythmia. Here, we studied the role of atrial structural remodeling in the disorganization of gap junctions and whether redistributed connexins can form new functional junction channels. METHODS: Expression of connexin-43 (Cx43) was characterized by immunoblotting and immunohistochemistry in human right atrial specimens and in rat atria after myocardial infarction (MI). Gap junctions were studied by electron and 3-D microscopy, and myocyte-myocyte coupling was determined by Lucifer yellow dye transfer. RESULTS: In both chronically hemodynamically overloaded human atria in sinus rhythm and in dilated atria from MI-rats, Cx43 were dephosphorylated and redistributed from the intercalated disc to the lateral cell membranes as observed during atrial fibrillation. In MI-rats, the gap junctions at the intercalated disc were smaller (20% decrease) and contained very little Cx43 (0 or 1 gold particle vs. 42 to 98 in sham-operated rats). In the lateral membranes of myocytes, numerous connexon aggregates comprising non-phosphorylated Cx43 were observed. These connexon aggregates were in no case assembled into gap junction plaque-like structures. However, N-cadherin was well organized in the intercalated disc. There was very little myocyte-myocyte coupling in MI-rat atria and no myocyte-fibroblast coupling. Regression of the atrial remodeling was associated with the normalization of Cx43 localization. CONCLUSION: Structural alteration of the atrial myocardium is an important factor in the disorganization of connexins and gap junction. Moreover, redistributed Cx43 do not form junction channels.


Subject(s)
Atrial Fibrillation/pathology , Connexin 43/analysis , Gap Junctions/pathology , Heart Atria/ultrastructure , Animals , Atrial Fibrillation/metabolism , Cell Communication , Fibrosis , Freeze Fracturing , Gap Junctions/metabolism , Heart Atria/metabolism , Humans , Imaging, Three-Dimensional , Immunoblotting , Immunohistochemistry , Microscopy, Electron , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Rats
16.
Arterioscler Thromb Vasc Biol ; 26(6): 1337-43, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16574891

ABSTRACT

OBJECTIVE: Because culprit atherosclerotic plaques contain proteases, we hypothesized that the diminished heat shock protein 27 (HSP27) released by atherosclerotic plaques could be due to proteolysis. We assessed the role of HSP27 in human vascular smooth muscle cells (VSMCs) under proteolytic injury. METHODS AND RESULTS: Active plasmin is present in culprit atherosclerotic plaques. Recombinant HSP27 was cleaved by plasmin and this effect was prevented by different inhibitors. Fragments and aggregated forms of HSP27 appeared after incubation of mammary control endarteries with plasmin. Coincubation of atherosclerotic plaques with recombinant HSP27 or mammary endarteries led to HSP27 proteolysis. After incubation of VSMCs with plasmin, HSP27 was overexpressed, phosphorylated, aggregated, and redistributed from the cytoskeleton to the cytosol, nucleus, and cell membrane. Plasmin-induced VSMC apoptosis was significantly higher in VSMCs treated by HSP27 siRNA. Immunohistochemical analysis of atherosclerotic plaques showed that plasmin(ogen) and apoptotic cells are localized in the core/shoulder whereas HSP27 and VSMCs are mainly expressed in the cap/media. CONCLUSIONS: Extracellular HSP27 can be degraded by enzymes released from atherosclerotic plaques and may reflect a proteolytic imbalance. Intracellular HSP27 downregulation decreases VSMCs resistance to proteolytically-induced apoptosis. HSP27 might play a pivotal role in the prevention of plaque instability and rupture.


Subject(s)
Atherosclerosis/physiopathology , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Apoptosis , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Cells, Cultured , Fibrinolysin/metabolism , Fibrinolysin/pharmacology , HSP27 Heat-Shock Proteins , Humans , Molecular Chaperones , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Tissue Culture Techniques
17.
Nat Genet ; 38(3): 343-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16444274

ABSTRACT

We have recently described two kindreds presenting thoracic aortic aneurysm and/or aortic dissection (TAAD) and patent ductus arteriosus (PDA) and mapped the disease locus to 16p12.2-p13.13 (ref. 3). We now demonstrate that the disease is caused by mutations in the MYH11 gene affecting the C-terminal coiled-coil region of the smooth muscle myosin heavy chain, a specific contractile protein of smooth muscle cells (SMC). All individuals bearing the heterozygous mutations, even if asymptomatic, showed marked aortic stiffness. Examination of pathological aortas showed large areas of medial degeneration with very low SMC content. Abnormal immunological recognition of SM-MHC and the colocalization of wild-type and mutant rod proteins in SMC, in conjunction with differences in their coimmunoprecipitation capacities, strongly suggest a dominant-negative effect. Human MYH11 gene mutations provide the first example of a direct change in a specific SMC protein leading to an inherited arterial disease.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Aortic Dissection/genetics , Ductus Arteriosus, Patent/genetics , Mutation , Myosin Heavy Chains/genetics , Adult , Amino Acid Sequence , Base Sequence , Female , Humans , Male , Molecular Sequence Data , Pedigree , Protein Structure, Secondary
18.
FEBS J ; 272(13): 3387-400, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15978044

ABSTRACT

Plasmin(ogen) kringles 1 and 4 are involved in anchorage of plasmin(ogen) to fibrin and cells, an essential step in fibrinolysis and pericellular proteolysis. Their contribution to these processes was investigated by selective neutralization of their lysine-binding function. Blocking the kringle 1 lysine-binding site with monoclonal antibody 34D3 fully abolished binding and activation of Glu-plasminogen and prevented both fibrinolysis and plasmin-induced cell detachment-induced apoptosis. In contrast, blocking the kringle 4 lysine-binding site with monoclonal antibody A10.2 did not impair its activation although it partially inhibited plasmin(ogen) binding, fibrinolysis and cell detachment. This remarkable, biologically relevant, distinctive response was not observed for plasmin or Lys-plasminogen; each antibody inhibited their binding and activation of Lys-plasminogen to a limited extent, and full inhibition of fibrinolysis required simultaneous neutralization of both kringles. Thus, in Lys-plasminogen and plasmin, kringles 1 and 4 act as independent and complementary domains, both able to support binding and activation. We conclude that Glu-/Lys-plasminogen and plasmin conformations are associated with transitions in the lysine-binding function of kringles 1 and 4 that modulate fibrinolysis and pericellular proteolysis and may be of biological relevance during athero-thrombosis and inflammatory states. These findings constitute the first biological link between plasmin(ogen) transitions and functions.


Subject(s)
Apoptosis , Fibrinolysin/pharmacology , Fibrinolysis/drug effects , Kringles/physiology , Peptide Fragments/metabolism , Plasminogen/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antifibrinolytic Agents/chemistry , Binding Sites , CHO Cells , Cell Adhesion/drug effects , Cricetinae , Fibrin/metabolism , Glutamic Acid/chemistry , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Plasminogen/antagonists & inhibitors , Plasminogen/genetics , Protein Binding/drug effects , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
J Gene Med ; 6(10): 1112-24, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15386736

ABSTRACT

BACKGROUND: Syngeneic vascular cells are interesting tools for indirect gene therapy in the cardiovascular system. This study aims to optimize transfection conditions of primary cultures of vascular smooth muscle cells (VSMCs) using different non-viral vectors and zinc as an adjuvant and to implant these transfected cells in vivo. METHODS: Non-liposomal cationic vectors (FuGene 6), polyethylenimines (ExGen 500), and histidylated polylysine (HPL) were used as non-viral vectors in vitro with secreted alkaline phosphatase (SEAP) as reporter gene. Transfection efficiency was compared in cultured rat, rabbit and human VSMCs and fibroblasts. Zinc chloride (ZnCl2) was added to optimize transfection of rat VSMCs in vitro which were then seeded in vivo. RESULTS: Much higher SEAP levels were obtained in rabbit cells with FuGene 6 (p <0.0001) at day 2 than in equivalent rat and human cells. Rat VSMCs transfected in vitro with FuGene 6 and ExGen 500 expressed higher SEAP levels than with HPL. In rat VSMCs, SEAP secretion was more than doubled by addition of 250 microM ZnCl2 (p <0.0001) for all vectors. Seeding of syngeneic VSMCs transfected under optimized conditions (FuGene 6/pcDNA3-SEAP +250 microM ZnCl2) into healthy Lewis rats using various routes or into post-infarct myocardial scar resulted in a peak of SEAP expression at day 2 and detectable activity in the plasma for at least 8 days. CONCLUSIONS: FuGene 6 is an efficient non-viral transfection reagent for gene transfer in somatic smooth muscle cells in vitro and ZnCl2 enhances its efficiency. This increased expression of the transgene product is maintained after seeding in vivo.


Subject(s)
Genetic Therapy , Genetic Vectors , Alkaline Phosphatase/metabolism , Animals , Aorta/metabolism , Cations , Cell Survival , Cells, Cultured , Chlorides/chemistry , Chlorides/metabolism , Chlorides/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Genes, Reporter , Humans , Myocytes, Smooth Muscle/cytology , Plasmids/metabolism , Polyethyleneimine/metabolism , Polylysine/chemistry , Rabbits , Rats , Rats, Inbred Lew , Time Factors , Transfection , Zinc/chemistry , Zinc Compounds/chemistry , Zinc Compounds/metabolism , Zinc Compounds/pharmacology
20.
Am J Pathol ; 164(6): 2077-87, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161642

ABSTRACT

To explore possible mechanisms responsible for the absence of cell re-colonization of mural thrombi in aneurysms, we analyzed the release and storage of leukocyte proteases in the most luminal layer versus intermediate and abluminal layers of 10 mural thrombi of human abdominal aortic aneurysms. The luminal layer contained many polymorphonuclear leukocytes (PMNs), which released pro-matrix metalloproteinase (MMP)-9 and MMP-8. Leukocyte elastase was also stored and released by the luminal layer (immunohistochemistry, activity on synthetic substrates, and casein zymography). Acid buffer allowed extraction of leukocyte elastase from the luminal layer, which was inhibited by elastase inhibitors. Casein zymography of luminal extracts and conditioned medium from formyl-methionyl-leucyl-phenylalanine (fMLP)-stimulated PMNs exhibited a similar lysis pattern, corresponding to elastase activity. Smooth muscle cell (SMC) seeding resulted in colonization of the intermediate thrombus layer ex vivo but not of the luminal layer. Extracts of the luminal layer induced loss of anchorage of both cultured human smooth muscle cells and stromal cells of bone marrow origin (anoikis). This anoikis was prevented by preincubation of the extracts with serine protease inhibitors. Moreover, adhesion of human SMCs and stromal bone marrow cells on fibrin gels was strongly inhibited when the gel was preincubated with pure elastase, medium of fMLP-stimulated PMNs, or extracts of luminal layers of mural thrombi. This loss of cell anchorage was prevented by the preincubation of the medium or extracts with alpha(1)-antitrypsin, but not when alpha(1)-antitrypsin was added after binding of elastase to the fibrin gel. In conclusion, elastase released by PMNs trapped within the mural thrombus impairs the spontaneous anchorage of mesenchymal cells to a fibrin matrix. This phenomenon could be one mechanism by which cellular healing of the mural thrombus in aneurysms is prevented.


Subject(s)
Leukocyte Elastase/physiology , Thrombosis/enzymology , Aneurysm/enzymology , Aneurysm/pathology , Aneurysm/surgery , Anoikis/drug effects , Bone Marrow Cells/cytology , Cell Adhesion , Cell Culture Techniques , Fibronectins/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth, Vascular/cytology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/pathology , Neutrophils/physiology , Serine Proteinase Inhibitors/pharmacology , Stromal Cells/cytology , Stromal Cells/physiology , Thrombosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...