Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
J Mol Cell Cardiol ; 187: 65-79, 2024 02.
Article in English | MEDLINE | ID: mdl-38181546

ABSTRACT

BACKGROUND: Vascular calcification (VC) is a prevalent independent risk factor for adverse cardiovascular events and is associated with diabetes, hypertension, chronic kidney disease, and atherosclerosis. However, the mechanisms regulating the osteogenic differentiation of vascular smooth muscle cells (VSMC) are not fully understood. METHODS: Using hydrogels of tuneable stiffness and lysyl oxidase-mediated stiffening of human saphenous vein ex vivo, we investigated the role of substrate stiffness in the regulation of VSMC calcification. RESULTS: We demonstrate that increased substrate stiffness enhances VSMC osteogenic differentiation and VSMC calcification. We show that the effects of substrate stiffness are mediated via a reduction in the level of actin monomer within the nucleus. We show that in cells interacting with soft substrate, elevated levels of nuclear actin monomer repress osteogenic differentiation and calcification by repressing YAP-mediated activation of both TEA Domain transcription factor (TEAD) and RUNX Family Transcription factor 2 (RUNX2). CONCLUSION: This work highlights for the first time the role of nuclear actin in mediating substrate stiffness-dependent VSMC calcification and the dual role of YAP-TEAD and YAP-RUNX2 transcriptional complexes.


Subject(s)
Actins , Vascular Calcification , Humans , Core Binding Factor Alpha 1 Subunit/genetics , Muscle, Smooth, Vascular , Osteogenesis , Cells, Cultured , Myocytes, Smooth Muscle
2.
Int J Obes (Lond) ; 41(2): 348-351, 2017 02.
Article in English | MEDLINE | ID: mdl-27899808

ABSTRACT

Adaptive immunity has been implicated in adipose tissue inflammation, obesity and its adverse metabolic consequences. No obesity-related autoantigen has yet been identified, although heat shock protein 60 (HSP60) has been implicated in other autoimmune diseases. We investigated whether feeding a high-fat diet to C57BL/6J mice would cause autoimmunity to HSP60 and whether immunomodulation with peptides from HSP60 would reverse the resulting obesity or metabolic dysfunction. Obese mice had higher circulating levels of HSP60 associated with increased T-lymphocyte proliferation responses and the emergence of circulating IgG1 and IgG2c antibody levels against HSP60. Treatment with escalating doses of a mixture of three proven immunomodulatory HSP60 peptides did not reduce weight but completely reversed the increase in VLDL/LDL levels and partially reversed the glucose intolerance in obese mice. Obese mice mount an autoimmune response to HSP60, which partly underlies the resulting metabolic disturbances.


Subject(s)
Autoimmunity/immunology , Chaperonin 60/immunology , Diet, High-Fat/adverse effects , Mitochondrial Proteins/immunology , Obesity/immunology , Adipose Tissue/immunology , Animals , Autoimmunity/drug effects , Chaperonin 60/blood , Chaperonin 60/pharmacology , Disease Models, Animal , Dose-Response Relationship, Immunologic , Immunohistochemistry , Inflammation/immunology , Lymphocyte Activation/drug effects , Metabolic Diseases/immunology , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/blood , Mitochondrial Proteins/pharmacology , Obesity/etiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Br J Pharmacol ; 155(6): 847-56, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18660830

ABSTRACT

BACKGROUND AND PURPOSE: To determine whether there is an association between vascular NADPH oxidase (NOX), superoxide, the small GTPase Rac(1) and PDE type 5 (PDE5) in human vascular smooth muscle cell (hVSMCs). EXPERIMENTAL APPROACH: hVSMCs were incubated with xanthine-xanthine oxidase (X-XO; a superoxide generating system) or the thromboxane A(2) analogue, U46619 (+/-superoxide dismutase (SOD) or apocynin) for 16 h. The expression of PDE5 and NOX-1 was assessed using Western blotting and superoxide measured. The role of Rac(1) in superoxide generation was assessed by overexpressing either the dominant-negative or constitutively active Rac isoforms. The effects of iloprost, DETA-NONOate and the Rho-kinase inhibitor, Y27632, on PDE5 and NOX-1 expression were also studied. KEY RESULTS: Following 16 h incubation, U46619 and X-XO promoted the expression of PDE5 and NOX-1, an effect blocked by SOD or apocynin when co-incubated over the same time course. X-XO and U46619 both promoted the formation of superoxide. Overexpression of dominant-negative Rac(1) or addition of iloprost, DETA-NONOate or Y27632 completely blocked both superoxide release and PDE5 protein expression and activity. CONCLUSIONS AND IMPLICATIONS: These data demonstrate that superoxide derived from NOX upregulates the expression of PDE5 in human VSMCs. As PDE5 hydrolyses cyclic GMP, this effect may blunt the vasculoprotective actions of NO.


Subject(s)
Iloprost/pharmacology , Muscle, Smooth, Vascular , NADPH Oxidases/metabolism , Nitroso Compounds/pharmacology , Phosphodiesterase 5 Inhibitors , Superoxides/metabolism , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Donors/pharmacology , Saphenous Vein/cytology , Superoxides/pharmacology , Up-Regulation , Vasodilator Agents/pharmacology
4.
Article in English | MEDLINE | ID: mdl-18420399

ABSTRACT

BACKGROUND: The over-production of superoxide (O(2)(-)) derived from NADPH oxidase (NOX) plays a central role in cardiovascular diseases. By contrast, nitric oxide (NO) and prostacyclin (PGI(2)) are vasculoprotective. The effect of the NO donor, NONOate and iloprost on O(2)(-) formation, p47(phox) and Rac(1) activation in human vascular smooth muscle cells (hVSMCs) was investigated. METHODS: hVSMCs were incubated with 10nM thromboxane A(2) analogue, U46619 for 16h, and then with apocynin (a NOX inhibitor), NONOate or iloprost for 1h and O(2)(-) measured spectrophometrically. The role of cyclic AMP and cyclic GMP was examined by co-incubation of drugs with protein kinase (PK) A and G inhibitors listed above. Rac(1) was studied using pull-down assays. RESULTS: NONOate and iloprost inhibited O(2)(-) formation, acutely, effects blocked by inhibition of PKG and PKA, respectively. Rac(1) and p47(phox) activation and translocation to the plasma membrane was completely inhibited by NONOate and iloprost, effects again reversed by co-incubation with PKG or PKA inhibitors. CONCLUSIONS: NO and PGI(2) block the acute activity of NOX in hVSMCs via the cGMP-PKG axis (for NO) and by the cAMP-PKA axis (for iloprost) through inhibition of Rac(1) and p47(phox) translocation. These findings have implications in the pathophysiology and treatment of CVD.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Iloprost/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/pharmacology , Superoxides/metabolism , rac1 GTP-Binding Protein/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Humans , Protein Kinase Inhibitors/pharmacology , Thromboxane A2/analogs & derivatives , Time Factors
5.
Cell Death Differ ; 15(2): 299-311, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17992191

ABSTRACT

Neurotrophins (NTs) control neuron survival and regeneration. Recent research showed that NTs possess cardiovascular actions. In this study, we investigated the hypothesis that the NT nerve growth factor (NGF) prevents cardiomyocyte apoptosis. We demonstrated that cultured rat neonatal cardiomyocytes (RNCMs) produce NGF and express its trkA (tropomyosin-related receptor A (NGF high-affinity receptor)) receptor. RNCMs given a neutralizing antibody for NGF or the trkA inhibitor K252a underwent apoptosis, thus suggesting that NGF is an endogenous prosurvival factor for cardiomyocytes. Adenovirus (Ad)-mediated NGF overexpression protected RNCMs from apoptosis induced by either hypoxia/reoxygenation or angiotensin II (AngII). Similarly, recombinant NGF inhibited AngII-induced apoptosis in isolated rat adult cardiomyocytes. Finally, in a rat model of myocardial infarction, NGF gene transfer promoted cardiomyocyte survival. In RNCMs, recombinant NGF induced trkA phosphorylation, followed by Ser473 phosphorylation and nuclear translocation of phospho-protein kinase B (Akt). In response to Akt activation, Forkhead transcription factors Foxo-3a and Foxo-1 were phosphorylated and excluded from the nucleus. The prosurvival effect of adenoviral vector carrying the human NGF gene was inhibited in vitro by K252a, LY294002 (a pan-phosphatidyl inositol 3-kinase - PI3K - inhibitor), an Akt small interfering RNA, and adenoviruses carrying a dominant negative mutant form of Akt (Ad.DN.Akt) or an Akt-resistant Foxo-3a (Ad.AAA-Foxo-3a). These results newly demonstrate the cardiac prosurvival action of NGF and provide mechanistic information on the signaling pathway, which encompasses trkA, PI3K-Akt, and Foxo.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Receptor, trkA/metabolism , Angiotensin II/metabolism , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Cell Survival , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Indole Alkaloids/pharmacology , Morpholines/pharmacology , Myocytes, Cardiac/drug effects , Nerve Growth Factor/genetics , Nerve Tissue Proteins/metabolism , PC12 Cells , Phosphorylation , Rats , Signal Transduction , Transfection
6.
Gene Ther ; 15(5): 340-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17989704

ABSTRACT

Increasing the level and duration of transgene expression and restricting expression to vascular cells are important goals for clinically useful gene therapy vectors. We evaluated several promoters, enhancers and introns in endothelial, smooth muscle and liver cells in tissue culture and in vivo, comparing local delivery to the carotid artery with intravenous delivery to the liver. A 1800-bp fragment of the oxidized LDL receptor (LOX-1) promoter showed highest in vivo activity in the carotid artery, achieving 39% the activity of the reference cytomegalovirus promoter, with 188-fold greater specificity for carotid artery over liver. An enhancer from the Tie2 gene in combination with the intracellular adhesion molecule-2 promoter improved endothelial specificity of plasmid vectors, increased the expression from adenoviral vectors in cultured endothelial cells and doubled the specificity for carotid artery over liver in vivo. Adding a short intron to expression cassettes increased expression in both endothelial and smooth muscle cells in vitro; however, the eNOS enhancer failed to consistently increase the expression or endothelial specificity of the vector. In conclusion, elements from the LOX-1 promoter and Tie2 enhancer together with an intron can be used to improve vectors for vascular gene transfer.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Muscle, Smooth, Vascular/metabolism , Vascular Diseases/therapy , Adenoviridae/genetics , Chi-Square Distribution , Endothelial Cells/metabolism , Enhancer Elements, Genetic , Gene Expression , Genetic Engineering , Humans , Introns , Linear Models , Liver/metabolism , Luciferases/genetics , Promoter Regions, Genetic , Receptor, TIE-2/genetics , Scavenger Receptors, Class E/genetics , Transduction, Genetic/methods
7.
Br J Ophthalmol ; 90(10): 1310-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16837541

ABSTRACT

BACKGROUND: Sorsby's fundus dystrophy (SFD) is caused by mutations in tissue inhibitor of metalloproteinase (TIMP)-3 and, with the exception of early onset, is similar to age-related macular degeneration. The pathological features of this condition relate to the accumulation of TIMP-3 in Bruch's membrane. AIMS: To compare the extracellular membrane-binding characteristics of wild-type and four SFD-mutant TIMP-3s. METHODS: COS-7 cells were transfected with wild-type, Ser-181, Gly-167, Ser-156 and Tyr-168 SFD-mutant TIMP-3 cDNA. The TIMP-3 proteins subsequently synthesised were harvested, analysed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis, semiquantified by ELISA and used in binding assays on the basis of the retention of the wild-type and SFD-mutant TIMP-3 proteins by components of Bruch's membrane. RESULTS: SFD-mutant TIMP-3s could not be distinguished from wild-type TIMP-3 by the extents to which they aggregated or adhered to type-I collagen, type-IV collagen, fibronectin, laminin, elastin, chondroitin sulphates A, B and C, and heparin sulphate. Of these macromolecules, the wild-type and SFD-mutant TIMP-3s exhibited greatest affinity for elastin and laminin. CONCLUSION: The similarity in the physical and extracellular membrane-binding characteristics of wild-type and SFD-mutant TIMP-3s indicates that these properties are not responsible for the difference in timing of onset of SFD and age-related macular degeneration.


Subject(s)
Bruch Membrane/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Animals , COS Cells , Chlorocebus aethiops , Corneal Dystrophies, Hereditary/genetics , Elastin/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Laminin/metabolism , Point Mutation , Tissue Inhibitor of Metalloproteinase-3/biosynthesis , Tissue Inhibitor of Metalloproteinase-3/genetics , Transfection
8.
Biochim Biophys Acta ; 1521(1-3): 12-8, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11690631

ABSTRACT

Adenosine production catalysed by cytosolic 5'-nucleotidase (cN-I) regulates diverse physiological processes. We report here a mouse cN-I (mcN-I) cloned from heart and testis. The open reading frame contains several potential translation initiation sites, which yield similarly active 5'-nucleotidases. Using overexpression in COS-7 cells we showed that mcN-I, like the previously cloned pigeon cN-I, is activated by ADP and catalyses adenosine formation during ATP breakdown. The N- and C-termini of mcN-I and pcN-I are divergent. Deletion of the 12 C-terminal amino acids or the first 19 N-terminal amino acids of pcN-I does not diminish activity, although deletion of the first 31 N-terminal amino acids reduces activity by 70%. Overall mcN-I is only 66% identical to pcN-I or the recently cloned human cN-I (hcN-I), while hcN-I and pcN-I are 85% identical. We report here a partial hcN-I sequence that is only 70% identical with the published hcN-I amino acid sequence but is 87% identical with mcN-I. Both hcN-I sequences have perfect matches to distinct human genome sequences. Our data imply the existence of at least two genes for cN-I, cN-I(A), previously cloned from pigeon and human, and cN-I(B) that we report here from mouse and partially from human.


Subject(s)
5'-Nucleotidase/genetics , Cytosol/enzymology , 5'-Nucleotidase/chemistry , 5'-Nucleotidase/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cloning, Molecular , Consensus Sequence , Humans , Male , Mice , Molecular Sequence Data , Nucleosides/genetics , Phosphotransferases/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Testis/enzymology
9.
Arterioscler Thromb Vasc Biol ; 21(8): 1275-80, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498453

ABSTRACT

Recently, we established a new mouse model of vein graft arteriosclerosis by grafting vena cava to carotid arteries. In many respects, the morphological features of this murine vascular graft model resemble those of human venous bypass graft disease. Using this model, we studied the effects of local gene transfer of tissue inhibitor of metalloproteinase-2 (TIMP-2) on vein graft remodeling. Mouse isogeneic vessels of the vena caval veins were grafted end to end into carotid arteries, then enveloped with the replication-defective recombinant adenoviruses overexpressing human TIMP-2 (RAdTIMP-2) or beta-galactosidase (RAdLacZ) at 1x10(10) plaque-forming units/mL in a total volume of 50 microL, and incubated at room temperature for 20 minutes. In the untreated group, vessel wall thickening was observed as early as 1 week after surgery and progressed to 4- to 10-fold the original thickness in grafted veins at 4 and 8 weeks, respectively. RAdLacZ vector treatment significantly enhanced neointimal lesions at 8 weeks, which was completely blocked by RAdTIMP-2 gene overexpression. Interestingly, RAdTIMP-2 gene transfer resulted in a reduction in vessel diameter of grafted veins compared with ungrafted veins (819+/-96 versus 624+/-67 microm, respectively; P<0.05). Maximal beta-galactosidase activity was found at 2 weeks and was detectable until 4 weeks after gene transfer. Double immunofluorescence studies demonstrated that cells overexpressing TIMP-2 were mostly localized in the adventitia and were MAC-1-positive monocytes/macrophages but not smooth muscle cells. Furthermore, the activity of matrix metalloproteinases was markedly decreased in the vessel walls treated with RAdTIMP-2 compared with that in the untreated control group and the RAdLacZ-treated group. Thus, this mouse model has been proven to be useful in gene transfer studies. Our findings demonstrate that local TIMP-2 gene transfer significantly reduces vein graft diameter, ie, remodeling to an artery-like vessel via inhibition of matrix metalloproteinase activity.


Subject(s)
Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Gene Transfer Techniques , Graft Occlusion, Vascular/enzymology , Graft Occlusion, Vascular/pathology , Models, Animal , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , Adenoviridae , Animals , Carotid Arteries , Fluorescent Antibody Technique , Genetic Vectors , Hyperplasia , Macrophages/enzymology , Matrix Metalloproteinases/metabolism , Mice , Transplantation, Autologous , Tunica Intima/enzymology , Tunica Intima/pathology , Venae Cavae/transplantation , beta-Galactosidase/biosynthesis
10.
Cardiovasc Res ; 50(3): 556-65, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11376631

ABSTRACT

OBJECTIVE: Matrix metalloproteinases (MMPs) contribute to the destruction of the extracellular matrix at the shoulder regions of atherosclerotic plaques that leads to plaque destabilisation and triggers clinical cardiovascular disease. There is therefore considerable interest in establishing the mechanisms responsible for increased MMP production. MMPs-1, -3 and -9 are upregulated by inflammatory cytokines and growth factors that are produced by plaque resident macrophages and smooth muscle cells. Our present studies focused on NF-kappaB, which regulates numerous inflammatory genes, and is activated in plaque smooth muscle cells. Moreover, an NF-kappaB binding site is present in the promoter of the MMP-9 gene and an NF-kappaB-like element in the promoter of the MMP-1 gene. METHODS: We used adenovirus mediated overexpression of its inhibitor, I kappaBalpha to investigate the role of NF-kappaB in regulation of MMP-1, -3 and -9 by isolated, cytokine stimulated rabbit aortic and human saphenous vein VSMC. RESULTS: IL-1alpha potently activated NF-kappa B in VSMCs and acted synergistically with growth factors to upregulate expression of MMP-1, -3 and -9. Overexpression of I kappaBalpha, almost completely inhibited expression of MMP-1, -3 and -9 in response to IL-1alpha alone or in combination with bFGF and PDGF. CONCLUSION: NF-kappaB is required for cytokine upregulation of MMP-1, -3 and -9 in VSMCs, which suggests that NF-kappaB inhibition may promote plaque stabilisation.


Subject(s)
I-kappa B Proteins , Matrix Metalloproteinases/biosynthesis , Muscle, Smooth, Vascular/enzymology , NF-kappa B/antagonists & inhibitors , Animals , Aorta/enzymology , Cell Culture Techniques , Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Growth Substances/pharmacology , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , NF-KappaB Inhibitor alpha , NF-kappa B/physiology , Rabbits , Saphenous Vein/enzymology , Up-Regulation/drug effects
11.
Atherosclerosis ; 155(2): 329-36, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254903

ABSTRACT

BACKGROUND: placing external non-restrictive macro-porous stents around porcine vein grafts prevents neointima formation and medial thickening in both the short and long term. Whether the porosity of the stent material influences this effect, however, has not been determined. Therefore, the effect on neointimal and medial thickening of external macro-porous (polyester) and micro-porous (polytetrafluorethylene) stents of equal diameter were compared. The effect on expression of platelet-derived growth factor (PDGF), a potent mediator of vascular smooth muscle cell migration and proliferation and its receptors was also investigated. METHODS AND RESULTS: saphenous vein-carotid artery interposition grafting was performed in Landrace pigs with external placement of 8 mm diameter macro- and micro-porous stents contralaterally. One month after surgery, graft wall dimensions, PDGF and PDGF receptor expression and cell proliferation using proliferating cell nuclear antigen (PCNA) were measured on histological sections. Macro-porous stents significantly reduced neointimal and medial thickening compared with micro-porous stents (0.1+/-0.02 vs. 0.25+/-0.03 mm, P<0.002, and 0.10+/-0.02 vs. 0.17+/-0.02 mm, P<0.014, respectively). Macro-porous stents significantly reduced the percentage of cells expressing PDGF and PCNA, compared with micro-porous stents (36+/-9 vs. 80+/-7, P < 0.002, and 11+/-3 vs. 21+/-2, P < 0.02, respectively). The percentage of cells expressing PDGF receptors was similar with both the stent types. Adventitial microvessel formation occurred across macro-porous stents but was markedly suppressed by micro-porous stents. CONCLUSIONS: porosity is crucial to the efficacy of external stents in reducing neointima formation in porcine vein grafts. Decreases in PDGF expression and cell proliferation accompany the reduction in neointima formation. In addition, macro-porous stents allow adventitial microvessels to connect with the vasculature outside the stent, thereby potentially improving oxygenation. Although external stenting is highly effective in reducing neointima formation after vein grafting, the properties of the stent material necessary for this effect have not been defined. This study establishes that macro-porosity is one essential feature required to reduce PDGF expression cell proliferation and neointima formation.


Subject(s)
Carotid Arteries/surgery , Graft Occlusion, Vascular/prevention & control , Polyesters , Polytetrafluoroethylene , Saphenous Vein/surgery , Stents , Tunica Intima/pathology , Tunica Media/pathology , Animals , Cell Division , Gene Expression Regulation , Hyperplasia , Materials Testing , Neovascularization, Physiologic , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Porosity , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/genetics , Receptors, Platelet-Derived Growth Factor/biosynthesis , Receptors, Platelet-Derived Growth Factor/genetics , Surface Properties , Swine
12.
Arthritis Rheum ; 44(12): 2888-98, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11762950

ABSTRACT

OBJECTIVE: Rheumatoid arthritis is a prototype of a destructive inflammatory disease. Inflammation triggered by the overexpression of tumor necrosis factor alpha (TNFalpha) is a driving force of this disorder and mediates tissue destruction. Since matrix metalloproteinases (MMPs) are among the molecules activated by TNFalpha, we hypothesized that overexpression of their natural inhibitor, tissue inhibitor of metalloproteinases 1 (TIMP-1), in TNFalpha transgenic mice could inhibit the development of destructive arthritis. METHODS: Systemic treatment was carried out by replication-defective adenoviral vectors for TIMP-1, beta-galactosidase, or phosphate buffered saline (PBS), which were applied once at the onset of arthritis. Clinical, serologic, radiologic, and histologic outcomes were assessed 18 days after the treatment. RESULTS: The AdTIMP-1 group showed significantly reduced paw swelling and increased grip strength compared with the 2 control groups, whereas total body weight, TNFalpha, and interleukin-6 levels were similar in all 3 groups. Radiographic assessment revealed a significant reduction of joint destruction in the AdTIMP-1 group; this was confirmed by histologic analyses showing reduced formation of pannus and erosions in the AdTIMP-1 group compared with the AdLacZ and PBS control groups. The formation of arthritis-specific autoantibodies to heterogeneous nuclear RNP A2 was not observed in the AdTIMP-1 group but was present in the 2 control groups. CONCLUSION: These results indicate a central role of MMPs in TNFalpha-mediated tissue damage in vivo and a promising therapeutic role for TIMP-1.


Subject(s)
Adenoviridae/genetics , Arthritis/pathology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Antigens, CD/blood , Arthritis/genetics , Arthritis/immunology , Arthrography , Autoantibodies/blood , Gene Expression Regulation, Viral/immunology , Gene Transfer Techniques , Humans , Interleukin-6/blood , Joints/immunology , Joints/pathology , Lac Operon , Matrix Metalloproteinase 3/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor, Type I , Tissue Inhibitor of Metalloproteinase-1/blood , Tumor Necrosis Factor-alpha/metabolism
13.
Methods Mol Med ; 52: 233-43, 2001.
Article in English | MEDLINE | ID: mdl-21340947

ABSTRACT

The development of an effective and safe gene therapy for prevention of vein graft failure, either acute or chronic, relies on the use of applicable and reproducible models of vein graft failure. Evaluation of potential new therapies usually involves assessment of beneficial phenotypic changes to vascular cells in isolated cells, in more complex organ cultures, and in in vivo models. We have extensively utilized gene transfer in isolated human vein vascular smooth muscle cells (SMCs) and endothelial cells (ECs) (1). Furthermore, we have extended our initial studies and evaluated potential gene therapies in a human saphenous vein organ culture model (2,3). The use of human saphenous vein, in this context, is advantageous as it is the most commonly used conduit for bypass grafting in the clinic. However, the organ culture model is static, and additionally studies need to be carried out in an in vivo model. In this chapter we describe a porcine model of vein graft neointima formation. Pig arteriovenous bypass grafting provides a reproducible model of intimal thickening, which is associated with late vein graft failure, and the effect of potential gene therapies can be assessed over long periods (up to 6 mo) (4; see Fig. 1). Furthermore, the saphenous vein is removed prior to grafting, and ex vivo genetic manipulation can be performed, providing a simple, controllable and safe method of gene transfer prior to grafting in vivo. Fig. 1. Neointima formation in porcine saphenous vein grafts. Removal of grafts at 7 d (A) and 28 d (B) reveals extensive and progressive neointima formation. The intimal/ medial border is indicated with arrows in each case. A, adventitia; M, media; I, intima.

15.
J Biol Chem ; 275(52): 41358-63, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11007798

ABSTRACT

The tissue inhibitors of metalloproteinases (TIMPs) are a family of four secreted inhibitors of matrix metalloproteinases (MMPs). Recently, additional functions have been attributed to the TIMPs, including cell growth and inhibition of angiogenesis. In particular, we demonstrated that TIMP-3 overexpression using gene transfer induces apoptosis in a variety of cell types and can inhibit vascular neointima formation in vivo. However, little is know about the mechanisms underlying TIMP-3-mediated apoptosis. Here, using both purified recombinant proteins and novel adenoviral vectors we demonstrate that the prodeath domain of TIMP-3 is located within the N-terminal three loops of TIMP-3. Although both wild type and N-terminal TIMP-3 proteins promoted apoptosis, a T-2/T-3 chimera, in which the N-terminal three loops of TIMP-3 are replaced by those of TIMP-2, failed to induce cell death. Furthermore, a point mutation at residue 1 of TIMP-3 totally abolished MMP-inhibitory activity of TIMP-3 and also failed to promote apoptosis. This study demonstrates, using multiple apoptosis assays, that the prodeath function of TIMP-3 is located within the N-terminal three loops and the presence of functional metalloproteinase-inhibitory activity is associated with the induction of apoptosis.


Subject(s)
Apoptosis , Metalloendopeptidases/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-3/chemistry , Animals , Cell Cycle , HeLa Cells , Humans , Rats , Tissue Inhibitor of Metalloproteinase-3/physiology
16.
Circ Res ; 87(5): 418-25, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969041

ABSTRACT

Chronic hypoxic pulmonary hypertension (PH) results from persistent vasoconstriction, excess muscularization, and extracellular matrix remodeling of pulmonary arteries. The matrix metalloproteinases (MMPs) are a family of proteinases implicated in extracellular matrix turnover and hence in smooth muscle and endothelial cell migration and proliferation. Because MMP expression and activity are increased in PH, we designed the present study to investigate whether inhibition of lung MMPs in rats subjected to chronic hypoxia (CH) contributes to or protects against vascular remodeling and PH. To achieve lung MMP inhibition, rats exposed to 10% O(2) for 15 days were treated with either doxycycline (20 mg/kg per day by gavage starting 2 days before and continuing throughout the CH period) or a single dose of recombinant adenovirus (Ad) for the human tissue inhibitors of metalloproteinases-1 (hTIMP-1) gene (Ad.hTIMP-1, 10(8) plaque-forming units given intratracheally 2 days before CH initiation). Control groups either received no treatment or were treated with an adenovirus containing no gene in the expression cassette (Ad.Null). Efficacy of hTIMP-1 gene transfer was assessed both by ELISA on bronchoalveolar lavages and by hTIMP-1 immunofluorescence on lung sections. MMP inhibition in lungs was evaluated by in situ zymography and gelatinolytic activity assessment using [(3)H]gelatin. Rats treated with either doxycycline or Ad.hTIMP-1 had higher pulmonary artery pressure and right heart ventricular hypertrophy more severe than their respective controls. Worsening of PH was associated with increased muscularization and periadventitial collagen accumulation in distal arteries. In conclusion, our study provides compelling evidence that MMPs play a pivotal role in protecting against pulmonary artery remodeling.


Subject(s)
Genetic Therapy/methods , Hypertension, Pulmonary/drug therapy , Lung/enzymology , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Tissue Inhibitor of Metalloproteinases/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Blood Pressure/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Doxycycline/therapeutic use , Gelatinases/metabolism , Gene Transfer Techniques , Genetic Vectors , Hypertension, Pulmonary/etiology , Hypoxia , Immunohistochemistry , Lung/blood supply , Lung/chemistry , Matrix Metalloproteinases/biosynthesis , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Rats , Rats, Wistar , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics
17.
J Biol Chem ; 275(16): 11666-71, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10766785

ABSTRACT

Catabolism of AMP during ATP breakdown produces adenosine, which restores energy balance. Catabolism of IMP may be a key step regulating purine nucleotide pools. Two, cloned cytosolic 5'-nucleotidases (cN-I and cN-II) have been implicated in AMP and IMP breakdown. To evaluate their roles directly, we expressed recombinant pigeon cN-I or human cN-II at similar activities in COS-7 or H9c2 cells. During rapid (more than 90% in 10 min) or slower (30-40% in 10 min) ATP catabolism, cN-I-transfected COS-7 and H9c2 cells produced significantly more adenosine than cN-II-transfected cells, which were similar to control-transfected cells. Inosine and hypoxanthine concentrations increased only during slower ATP catabolism. In COS-7 cells, 5'-nucleotidase activity was not rate-limiting for inosine and hypoxanthine production, which was therefore unaffected by cN-II- and actually reduced by cN-I- overexpression. In H9c2 cells, in which 5'-nucleotidase activity was rate-limiting, only cN-II overexpression accelerated inosine and hypoxanthine formation. Guanosine formation from GMP was also increased by cN-II. Our results imply distinct roles for cN-I and cN-II. Under the conditions tested in these cells, only cN-I plays a significant role in AMP breakdown to adenosine, whereas only cN-II breaks down IMP to inosine and GMP to guanosine.


Subject(s)
5'-Nucleotidase/physiology , Adenosine Monophosphate/metabolism , Inosine Monophosphate/metabolism , Isoenzymes/physiology , Muscles/metabolism , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , COS Cells , Cell Line , Columbidae , Cytosol/enzymology , Energy Metabolism , Heart/embryology , Humans , Hypoxanthine/metabolism , Isoenzymes/genetics , Kinetics , Rats , Recombinant Proteins/metabolism , Transfection
18.
Atherosclerosis ; 150(1): 33-41, 2000 May.
Article in English | MEDLINE | ID: mdl-10781633

ABSTRACT

Atherosclerosis causes occlusions in as many as 50% of human saphenous vein coronary artery bypass grafts. Monocyte infiltration is an early step in saphenous vein-graft atherosclerosis, however, comparatively little is known of its underlying mechanisms. As a first approach, we sought to define the occurrence, location and regulation of leukocyte adhesion molecules in human saphenous vein before and after surgical preparation for grafting, during neointima formation in culture and on stimulation with inflammatory cytokines. We compared the distribution of intercellular adhesion molecule (ICAM-1), vascular cell adhesion molecule (VCAM-1) and platelet endothelial cell adhesion molecule (PECAM-1 or CD-31) in endothelial cells and smooth muscle cells (SMCs), using immunocytochemistry. ICAM-1 was expressed on endothelial cells before culture and on both endothelial cells and medial or neointimal SMCs after culturing vein for 14 days in 30% foetal bovine serum or after culturing for 24 h with TNF-alpha. Relative tissue levels of ICAM-1 measured by Western blotting were significantly elevated by culturing freshly-isolated (0.02+/-0.01 to 0.18+/-0.03) and surgically-prepared (0.02+/-0.01 to 0.14+/-0.03; n=6) veins or following TNF-alpha treatment of surgically-prepared veins (0.04+/-0.01 to 0.32+/-0.11, n=7). VCAM-1 was undetectable before or after culturing but was strongly upregulated on endothelial cells by incubation with the cytokines TNF-alpha, IL-1alpha or interferon-gamma. PECAM-1 was expressed constitutively on endothelial cells. We conclude that human saphenous vein expresses several adhesion molecules capable of mediating monocyte migration. The increased expression of ICAM-1 in SMC after culturing or cytokine treatment and of VCAM-1 in endothelial cells suggests that interactions with beta1 and beta2 integrins are important pathways for stimulated monocyte ingress into human saphenous vein grafts.


Subject(s)
Cell Adhesion Molecules/metabolism , Cytokines/pharmacology , Inflammation Mediators/pharmacology , Saphenous Vein/metabolism , Blotting, Western , Cells, Cultured , Culture Techniques , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Muscle, Smooth, Vascular/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Saphenous Vein/cytology , Tumor Necrosis Factor-alpha/pharmacology , Tunica Intima/cytology , Vascular Cell Adhesion Molecule-1/metabolism
19.
Toxicol Lett ; 112-113: 519-29, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10720775

ABSTRACT

The innermost layer of a normal blood vessel wall, the tunica intima, consists of a simple monolayer of endothelial cells seated on a basement membrane. Expansion of the intima is a common characteristic of atherosclerosis, restenosis after angioplasty, late closure of saphenous vein grafts and transplant vascular disease. The intima becomes a complex connective tissue containing vascular smooth muscle cells that have invaded from the underlying tunica media and inflammatory cells that have invaded from the circulation. This brief review will concentrate on the molecular events underlying the generation of this neointima 'in response to injury' and its consequences for disease. It will also consider the implications for the consequences and early detection of vascular drug toxicity.


Subject(s)
Arteriosclerosis/etiology , Muscle, Smooth, Vascular , Animals , Arteriosclerosis/complications , Humans , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/anatomy & histology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/metabolism , Myocardial Infarction/etiology , Tunica Intima/pathology
20.
J Pathol ; 190(3): 300-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685064

ABSTRACT

Intimal hyperplasia is the process by which the cell population increases within the innermost layer of the arterial wall, such as occurs physiologically during closure of the ductus arteriosus and during involution of the uterus. It also occurs pathologically in pulmonary hypertension, atherosclerosis, after angioplasty, in transplanted organs, and in vein grafts. The underlying causes of intimal hyperplasia are migration and proliferation of vascular smooth muscle cells provoked by injury, inflammation, and stretch. This review discusses, at a molecular level, both the final common pathways leading to smooth muscle migration and proliferation and their (patho)-physiological triggers. It emphasizes the key roles played by growth factors and extracellular matrix-degrading metalloproteinases, which act in concert to remodel the extracellular matrix and permit cell migration and proliferation.


Subject(s)
Cell Movement/physiology , Muscle, Smooth, Vascular/pathology , Tunica Intima/pathology , Cell Division/physiology , Endothelial Growth Factors/physiology , Humans , Hyperplasia/etiology , Hyperplasia/pathology , Metalloendopeptidases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...