Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 88(2): 367-75, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20562095

ABSTRACT

AIMS: Matrix metalloproteinases (MMP) and plasminogen activator (PA)/plasmin-mediated proteolysis, especially at the cell surface, play important roles in matrix degeneration and smooth muscle cell migration, which largely contributes to vein graft failure. In this study, a novel hybrid protein was designed to inhibit both protease systems simultaneously. MMP and plasmin activity were inhibited at the cell surface by this hybrid protein, consisting of the receptor-binding amino-terminal fragment (ATF) of urokinase-type PA, linked to both the tissue inhibitor of metalloproteinases (TIMP-1) and bovine pancreas trypsin inhibitor (BPTI), a potent protease inhibitor. The effect of overexpression of this protein on vein graft disease was studied. METHODS AND RESULTS: A non-viral expression vector encoding the hybrid protein TIMP-1.ATF.BPTI was constructed and validated. Next, cultured segments of human veins were transfected with this vector. Expressing TIMP-1.ATF.BPTI in vein segments resulted in a mean 36 ± 14% reduction in neointima formation after 4 weeks. In vivo inhibition of vein graft disease by TIMP-1.ATF.BPTI is demonstrated in venous interpositions placed into carotid arteries of hypercholesterolaemic APOE*3Leiden mice. After 4 weeks, vein graft thickening was significantly inhibited in mice treated with the domains TIMP-1, ATF, or BPTI (36-49% reduction). In the TIMP-1.ATF.BPTI-treated mice, vein graft thickening was reduced by 67±4%, which was also significantly stronger when compared with the individual components. CONCLUSION: These data provide evidence that cell surface-bound inhibition of the PA and MMP system by the hybrid protein TIMP-1.ATF.BPTI, overexpressed in distant tissues after electroporation-mediated non-viral gene transfer, is a powerful approach to prevent vein graft disease.


Subject(s)
Cell Proliferation , Fibrinolysin/metabolism , Genetic Therapy , Graft Occlusion, Vascular/prevention & control , Matrix Metalloproteinases/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Saphenous Vein/metabolism , Venae Cavae/metabolism , Animals , Apolipoprotein E3/genetics , Aprotinin/biosynthesis , Aprotinin/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Carotid Arteries/surgery , Cattle , Cell Line , Disease Models, Animal , Electroporation , Fibrinolysin/antagonists & inhibitors , Genetic Therapy/methods , Graft Occlusion, Vascular/genetics , Graft Occlusion, Vascular/metabolism , Graft Occlusion, Vascular/pathology , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hyperplasia , Male , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Recombinant Fusion Proteins/biosynthesis , Saphenous Vein/pathology , Saphenous Vein/surgery , Time Factors , Tissue Culture Techniques , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Transfection , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/genetics , Venae Cavae/pathology , Venae Cavae/transplantation
2.
J Vasc Surg ; 51(2): 429-37, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20036101

ABSTRACT

BACKGROUND: Smooth muscle cell (SMC) migration and proliferation are important in the development of intimal hyperplasia, the major cause of vein graft failure. Proteases of the plasminogen activator (PA) system and of the matrix metalloproteinase (MMP) system are pivotal in extracellular matrix degradation and, by that, SMC migration. Previously, we demonstrated that inhibition of both protease systems simultaneously with viral gene delivery of the hybrid protein TIMP-1.ATF, consisting of the tissue inhibitor of metalloproteinase-1 (TIMP-1) and the receptor-binding amino terminal fragment (ATF) of urokinase, reduces SMC migration and neointima formation in an in vitro restenosis model using human saphenous vein cultures more efficiently than both protease systems separately. Because use of viral gene delivery is difficult in clinical application, this study used nonviral delivery of TIMP-1.ATF plasmid to reduce vein graft disease in a murine bypass model. Nonviral gene transfer by electroporation was used to avert major disadvantages of viral gene delivery, such as immune responses and short-term expression. METHODS: Plasmids encoding ATF, TIMP-1, TIMP-1.ATF, or luciferase, as a control, were injected and electroporated in both calf muscles of hypercholesterolemic apolipoprotein E3-Leiden (APOE*3Leiden) mice (n = 8). One day after electroporation, a venous interposition of a donor mouse was placed into the carotid artery of a recipient mouse. In this model, vein graft thickening develops with features of accelerated atherosclerosis. Vein grafts were harvested 4 weeks after electroporation and surgery, and histologic analysis of the vessel wall was performed. RESULTS: Electroporation-mediated overexpression of the plasmid vectors resulted in a prolonged expression of the transgenes and resulted in a significant reduction of vein graft thickening (ATF: 36% +/- 9%, TIMP-1: 49% +/- 5%, TIMP-1.ATF: 58% +/- 5%; P < .025). Although all constructs reduced vein graft thickening compared with the controls, the luminal area was best preserved in the TIMP-1.ATF-treated mice. CONCLUSION: Intramuscular electroporation of TIMP-1.ATF inhibits vein graft thickening in vein grafts in carotid arteries of hypercholesterolemic mice. Binding of TIMP-1.ATF hybrid protein to the u-PA receptor at the cell surface enhances the inhibitory effect of TIMP-1 on vein graft remodeling in vitro as well as in vivo and may be an effective strategy to prevent vein graft disease.


Subject(s)
Atherosclerosis/prevention & control , Electroporation , Gene Transfer Techniques , Genetic Therapy/methods , Graft Occlusion, Vascular/prevention & control , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Urokinase-Type Plasminogen Activator/biosynthesis , Venae Cavae/transplantation , Animals , Apolipoprotein E3/genetics , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Arteries/surgery , Disease Models, Animal , Graft Occlusion, Vascular/enzymology , Graft Occlusion, Vascular/genetics , Graft Occlusion, Vascular/pathology , Graft Survival , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Hypercholesterolemia/therapy , Hyperplasia , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Peptide Fragments/biosynthesis , Receptors, Urokinase Plasminogen Activator/metabolism , Recombinant Fusion Proteins/biosynthesis , Time Factors , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/genetics , Urokinase-Type Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/genetics , Venae Cavae/enzymology , Venae Cavae/pathology
3.
Hum Gene Ther ; 18(9): 861-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17850190

ABSTRACT

For the successful application of RNA interference in vivo, it is desired to achieve (local) delivery of small interfering RNAs (siRNAs) and long-term gene silencing. Nonviral electrodelivery is suitable to obtain local and prolonged expression of transgenes. By intramuscular electrodelivery of a plasmid in which two opposing human polymerase III promoters (H1 and U6) drive the expression of siRNA constructs that form functional double-stranded siRNAs, in combination with in vivo bioluminescence imaging, we were able to knock down exogenous delivered luciferase for at least 100 days in murine calf muscles. This effect was sequence specific, because scrambled siRNA had no effect. Moreover, we were able to demonstrate in vivo reduction of endogenous TLR4 expression for at least 1 week, using a similar vector expressing an siRNA for TLR4 in the muscle. In this study, we demonstrate that in vivo suppression of both endogenous (for at least 1 week) and introduced genes (>100 days) is feasible via plasmid-driven siRNA expression after electroporation-mediated intramuscular gene transfer. With this approach the short-term effect of oligonucleotides and the drawbacks of viral gene delivery, like immunological responses, could be circumvented. Therefore, this application of RNA interference is a useful tool with which to investigate gene function and might be promising as a therapeutic tool for locally acting diseases such as restenosis or tumors.


Subject(s)
Gene Silencing , Plasmids/genetics , RNA Interference , RNA, Small Interfering/genetics , Animals , Cattle , Cell Line, Transformed , Cell Transformation, Viral , Electroporation , Feasibility Studies , Genes, Reporter , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Luminescent Measurements , Male , Mice , Mice, Inbred Strains , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , NIH 3T3 Cells , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Time Factors , Toll-Like Receptor 4/metabolism
4.
Atherosclerosis ; 193(2): 335-42, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17087966

ABSTRACT

OBJECTIVE: Inflammatory factors are thought to play a regulatory role in restenosis. Interleukin-10 (IL10) is an important anti-inflammatory cytokine with anti-atherogenic potentials. The aim of this study was to assess the effects of IL10 modulation on cuff-induced neointima formation in hypercholesterolemic APOE*3-Leiden mice. METHODS: The involvement of IL10 in neointima formation was studied in a hypercholesterolemic mouse model of cuff-induced stenosis of the femoral artery by IL10 knocking-out or overexpression procedures. IL10(+/-) mice were crossbred with APOE*3-Leiden mice to generate hypercholesterolemic APOE*3-LeidenIL10(-/-) mice. To achieve IL10 overexpression in APOE*3-Leiden mice, a single intramuscular injection of a murine IL10 overexpression plasmid was performed followed by electroporation. RESULTS: Knocking-out IL10, in hypercholesterolemic APOE*3-Leiden mice, resulted in a significant 1.9-fold increase of neointima surface as compared to APOE*3-LeidenIL10(+/+) littermates (p=0.02). Conversely, a marked 45% inhibition on cuff-induced neointima formation was obtained after IL10 overexpression (p=0.02). Electrodelivery of IL10 vector leads to detectable IL10 serum levels, with a sustained expression over the experimental period of 3 weeks. IL10 overexpression reduced plasma cholesterol levels in APOE*3-Leiden mice, whereas IL10 deficiency in these mice did not lead to altered cholesterol levels as compared to the IL10(+/+) group. Finally, IL10 overexpression stimulated endogenous IL10 mRNA expression in the spleen and reduced the transcriptional responses of several pro-inflammatory cytokines. CONCLUSION: Here, we clearly demonstrate the role of IL10 in the development of neointima formation in hypercholesterolemic mice and the potential therapeutic effect of non-viral electrodelivery of IL10 cDNA to inhibit post-angioplasty restenosis.


Subject(s)
Hypercholesterolemia/immunology , Interleukin-10/immunology , Tunica Intima/immunology , Vascular Diseases/immunology , Animals , Disease Models, Animal , Hypercholesterolemia/genetics , Interleukin-10/biosynthesis , Mice , Mice, Knockout , Vascular Diseases/genetics
5.
Cardiovasc Res ; 68(1): 136-43, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-15963479

ABSTRACT

BACKGROUND: Furin-like proprotein convertases (PCs) are proteolytic activators of proproteins, like membrane type 1-matrix metalloproteinase (MT1-MMP) and transforming growth factor beta (TGF-beta), that are described in the arterial response to injury. However, the involvement of furin-like PCs in the arterial response to injury has not been studied yet. We studied furin, MT1-MMP, MMP levels and TGF-beta signaling after arterial injury. We also investigated the effect of an inhibitor of furin-like PCs, alpha1-antitrypsin Portland (alpha1-PDX), on arterial injury following balloon dilation. METHODS AND RESULTS: NZW rabbit femoral and iliac arteries (N=42) were balloon dilated unilaterally and harvested after 2, 7, 14, 28 or 42 days. Furin mRNA levels were increased after 2 and 7 days. MMP-2 and MT1-MMP levels were increased after day 7 and TGF-beta signaling, by phosphorylating Smad 1/5 and 2/3, was increased at all time points. Inhibition of furin-like PCs, by adenoviral over-expression of alpha1-PDX, blocked proTGF-beta activation and Smad phosphorylation, and reduced MT1-MMP and MMP-2 activation (N=5). In vivo adventitial inhibition of furin-like PCs (N=9) resulted in a reduction of 13.1+/-5.2% in advential and 23.6+/-7.9% in intimal areas (P<0.05), but had no effect on lumen size due to decreased vessel areas. CONCLUSIONS: This study demonstrates that furin-like PCs are involved in the arterial response to injury possibly through activation of the TGF-beta-Smad signaling pathway and identifies furin-like PCs as a possible target to inhibit intimal hyperplasia.


Subject(s)
Atherosclerosis/metabolism , Catheterization , Femoral Artery/injuries , Furin/physiology , Iliac Artery/injuries , Adenoviridae/genetics , Animals , Enzyme Activation , Femoral Artery/metabolism , Furin/antagonists & inhibitors , Genetic Vectors/administration & dosage , Iliac Artery/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases, Membrane-Associated , Models, Animal , Rabbits , Smad Proteins, Receptor-Regulated/metabolism , Transduction, Genetic/methods , Transforming Growth Factor beta/metabolism , alpha 1-Antitrypsin/genetics
6.
J Clin Endocrinol Metab ; 89(11): 5828-36, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15531549

ABSTRACT

In the endometrium, angiogenesis is a physiological process, whereas in most adult tissues neovascularization is initiated only during tissue repair or pathological conditions. Pericellular proteolysis plays an important role in angiogenesis being required for endothelial cell migration, invasion, and tube formation. We studied the expression of proteases by human endometrial microvascular endothelial cells (hEMVECs) and their involvement in the formation of capillary tubes and compared these requirements with those of foreskin MVECs (hFMVECs). Inhibition of urokinase and matrix metalloproteinase (MMP) both reduced tube formation in a fibrin or fibrin/collagen matrix. hEMVECs expressed various MMP mRNAs and proteins; in particular MMP-1, MMP-2, and membrane-type (MT)1-, MT3-, and MT4-MMPs. MT3- and MT4-MMP mRNA expressions were significantly higher in hEMVECs than in hFMVECs. Other MT-MMP mRNAs and MMP-9 were hardly detectable. Immunohistochemistry confirmed the presence of MT3-MMP in endothelial cells of endometrial tissue. Overexpression of tissue inhibitor of MMP (TIMP)-1 or TIMP-3 by adenoviral transduction of hEMVECs reduced tube formation to the same extent, whereas only TIMP-3 was able to inhibit tube formation by hFMVECs. Tube formation by hEMVECs was partly inhibited by the presence of anti-MT3-MMP IgG. Thus, in contrast to tube formation by hFMVECs, which largely depends on MT1-MMP, capillary-like tube formation by hEMVECs is, at least in part, regulated by MT3-MMP.


Subject(s)
Endometrium/blood supply , Endothelial Cells/physiology , Metalloendopeptidases/physiology , Neovascularization, Physiologic , Adenoviridae/genetics , Cells, Cultured , Collagen Type I/pharmacology , Female , Fibrinolysin/physiology , Humans , Matrix Metalloproteinase 16 , Matrix Metalloproteinases, Membrane-Associated , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Urokinase-Type Plasminogen Activator/physiology
7.
Circ Res ; 95(1): 58-66, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15155530

ABSTRACT

Therapeutic angiogenesis using vascular endothelial growth factor (VEGF) is considered a promising new therapy for patients with arterial obstructive disease. Clinical improvements observed consist of improved muscle function and regression of rest pain or angina. However, direct evidence for improved vascularization, as evaluated by angiography, is weak. In this study, we report an angiogenesis-independent effect of VEGF on ischemic skeletal muscle, ie, upregulation of myoglobin after VEGF treatment. Mice received intramuscular injection with adenoviral VEGF-A or either adenoviral LacZ or PBS as control, followed by surgical induction of acute hindlimb ischemia at day 3. At day 6, capillary density was increased in calf muscle of Ad.VEGF-treated versus control mice (P<0.01). However, angiographic score of collateral arteries was unchanged between Ad.VEGF-treated and control mice. More interestingly, an increase in myoglobin was observed in Ad.VEGF-treated mice. Active myoglobin was 1.5-fold increased in calf muscle of Ad.VEGF-treated mice (P< or =0.01). In addition, the number of myoglobin-stained myofibers was 2.6-fold increased in Ad.VEGF-treated mice (P=0.001). Furthermore, in ischemic muscle of 15 limb amputation patients, VEGF and myoglobin were coexpressed. Finally, in cultured C2C12 myotubes treated with rhVEGF, myoglobin mRNA was 2.8-fold raised as compared with PBS-treated cells (P=0.02). This effect could be blocked with the VEGF receptor tyrosine kinase inhibitor SU5416. In conclusion, we show that VEGF upregulates myoglobin in ischemic muscle both in vitro and in vivo. Increased myoglobin expression in VEGF-treated muscle implies an improved muscle oxygenation, which may, at least partly, explain observed clinical improvements in VEGF-treated patients, in the absence of improved vascularization.


Subject(s)
Ischemia/metabolism , Muscle, Skeletal/blood supply , Myoglobin/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Aged , Amputation, Surgical , Animals , Capillaries/growth & development , Female , Gene Expression , Genetic Therapy , Genetic Vectors , Humans , Ischemia/diagnostic imaging , Ischemia/therapy , Male , Mice , Middle Aged , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/metabolism , Myoglobin/genetics , Neovascularization, Physiologic , RNA, Messenger/biosynthesis , Radiography , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
8.
Arterioscler Thromb Vasc Biol ; 23(9): 1535-40, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12842839

ABSTRACT

OBJECTIVE: Endothelial cells play a pivotal role in vascular homeostasis. In this study, we investigated the function of the nerve growth factor-induced protein-B (NGFI-B) subfamily of nuclear receptors comprising the TR3 orphan receptor (TR3), mitogen-induced nuclear orphan receptor (MINOR), and nuclear orphan receptor of T cells (NOT) in endothelial cells. METHODS AND RESULTS: The mRNA expression of TR3, MINOR, and NOT in atherosclerotic lesions was assessed in human vascular specimens. Each of these factors is expressed in smooth muscle cells, as described before, and in subsets of endothelial cells, implicating that they might affect endothelial cell function. Adenoviral overexpression of TR3 in cultured endothelial cells resulted in decreased [3H]thymidine incorporation, whereas a dominant-negative TR3 variant that inhibits the activity of endogenous TR3-like factors enhanced DNA synthesis. TR3 interfered with progression of the cell cycle by upregulating p27Kip1 and downregulating cyclin A, whereas expression levels of a number of other cell cycle-associated proteins remained unchanged. CONCLUSIONS: These findings demonstrate that TR3 is a modulator of endothelial cell proliferation and arrests endothelial cells in the G1 phase of the cell cycle by influencing cell cycle protein levels. We hypothesize involvement of TR3 in the maintenance of integrity of the vascular endothelium.


Subject(s)
Cell Cycle/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Receptors, Steroid/biosynthesis , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/physiology , Adenoviridae/genetics , Cell Division/genetics , Cell Division/physiology , Cells, Cultured , DNA, Complementary/genetics , Endothelium, Vascular/virology , Gene Expression Regulation/genetics , Gene Transfer Techniques , Genetic Variation/genetics , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1 , Peptides/genetics , Peptides/physiology , Protein Structure, Tertiary/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics , Sequence Deletion/genetics , Stem Cells/chemistry , Stem Cells/metabolism , Stem Cells/virology , Transcriptional Activation/genetics , Umbilical Veins/cytology , Umbilical Veins/virology , Virus Replication/genetics
9.
Blood ; 101(10): 3933-9, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12522008

ABSTRACT

It has been established that low-density lipoprotein receptor-related protein (LRP) is involved in the cellular uptake and degradation of coagulation factor VIII (FVIII) in vitro. To address the physiologic role of LRP in regulating plasma FVIII in vivo, we used cre/loxP-mediated conditional LRP- deficient mice (MX1cre(+)LRP(flox/flox)). Upon inactivation of the LRP gene, MX1cre(+)LRP(flox/flox) mice had significantly higher plasma FVIII as compared with control LRP(flox/flox) mice (3.4 and 2.0 U/mL, respectively; P <.001). Elevated plasma FVIII levels in MX1cre(+)LRP(flox/flox) mice coincided with increased plasma von Willebrand factor (VWF) (2.0 and 1.6 U/mL for MX1cre(+)LRP(flox/flox) and control LRP(flox/flox) mice, respectively; P <.05). Elevation of plasma FVIII and VWF persisted for at least 6 weeks after inactivation of the LRP gene. Upon comparing plasma FVIII and VWF in individual mice, we observed an increase of the FVIII/VWF ratio in MX1cre(+)LRP(flox/flox) mice as compared with control LRP(flox/flox) mice. Administration of either a vasopressin analog or an endotoxin resulted in increased plasma VWF, but not FVIII. In clearance experiments, MX1cre(+)LRP(flox/flox) mice displayed a 1.5-fold prolongation of FVIII mean residence time. Adenovirus-mediated overexpression of the 39-kDa receptor-associated protein (RAP) in normal mice resulted in a 3.5-fold increase of plasma FVIII. These data confirm that the regulation of plasma FVIII in vivo involves a RAP-sensitive mechanism. Surprisingly, plasma FVIII in MX1cre(+)LRP(flox/flox) mice increased 2-fold after RAP gene transfer. We propose that RAP-sensitive determinants other than hepatic LRP contribute to the regulation of plasma FVIII in vivo.


Subject(s)
Factor VIII/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , von Willebrand Factor/metabolism , Adenoviridae/genetics , Animals , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Genotype , Humans , Kinetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout
10.
Circulation ; 106(12): 1530-5, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12234960

ABSTRACT

BACKGROUND: Smooth muscle cells (SMCs) play a key role in intimal thickening in atherosclerosis and restenosis. The precise signaling pathways by which the proliferation of SMCs is regulated are largely unknown. The TR3 orphan receptor, the mitogen-induced nuclear orphan receptor (MINOR), and the nuclear receptor of T cells (NOT) are a subfamily of transcription factors belonging to the nuclear receptor superfamily and are induced in activated SMCs. In this study, we investigated the role of these transcription factors in SMC proliferation in atherogenesis. METHODS AND RESULTS: Multiple human vascular specimens at distinct stages of atherosclerosis (lesion types II to V by American Heart Association classification) derived from 14 different individuals were studied for expression of these transcription factors. We observed expression of TR3, MINOR, and NOT in neointimal SMCs, whereas no expression was detected in medial SMCs. Adenovirus-mediated expression of a dominant-negative variant of TR3, which suppresses the transcriptional activity of each subfamily member, increases DNA synthesis and decreases p27(Kip1) protein expression in cultured SMCs. We generated transgenic mice that express this dominant-negative variant or full-length TR3 under control of a vascular SMC-specific promoter. Carotid artery ligation of transgenic mice that express the dominant-negative variant of TR3 in arterial SMCs, compared with lesions formed in wild-type mice, results in a 3-fold increase in neointimal formation, whereas neointimal formation is inhibited 5-fold in transgenic mice expressing full-length TR3. CONCLUSIONS: Our results reveal that TR3 and possibly other members of this transcription factor subfamily inhibit vascular lesion formation. These transcription factors could serve as novel targets in the treatment of vascular disease.


Subject(s)
Arteriosclerosis/etiology , DNA-Binding Proteins/physiology , Muscle, Smooth, Vascular/metabolism , Receptors, Steroid , Receptors, Thyroid Hormone , Transcription Factors/physiology , Adenoviridae/genetics , Animals , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cardiotonic Agents/metabolism , Carotid Arteries/surgery , DNA/biosynthesis , DNA-Binding Proteins/genetics , Genetic Vectors , Humans , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins , Nuclear Receptor Subfamily 4, Group A, Member 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
11.
Circ Res ; 90(10): 1128-34, 2002 May 31.
Article in English | MEDLINE | ID: mdl-12039804

ABSTRACT

Activin A alters the characteristics of human arterial smooth muscle cells (SMCs) toward a contractile, quiescent phenotype. We hypothesize that activin A may prevent SMC-rich neointimal hyperplasia. Here, we study the effect of adenovirus-mediated expression of activin A on neointima formation in vitro and in vivo. Human saphenous vein organ cultures, in which a neointima is formed spontaneously, were infected either with activin A- or lacZ-adenovirus. Activin A-overexpression reduces neointima formation by 78%, whereas no significant reduction was observed after control infection. In addition, the effect of activin A on neointima formation was assessed in vivo in mice with cuffed femoral arteries. In activin A adenovirus-infected mice (IV injection), neointimal hyperplasia is reduced by 77% compared with the SMC-rich neointima in mock-infected or in noninfected mice. Cultured human saphenous vein SMCs and murine aorta SMCs were incubated with activin A and an increased expression of SM22alpha and SM alpha-actin mRNA, and SM alpha-actin protein was demonstrated. Laser-capture microdissection on sections of cuffed murine arteries and subsequent real-time RT-PCR established in vivo induction of SM alpha-actin mRNA in the media of activin A-treated mice. In summary, activin A inhibits neointima formation in vitro and in vivo by preventing SMC dedifferentiation.


Subject(s)
Activins/genetics , Adenoviridae/genetics , Arterial Occlusive Diseases/prevention & control , Inhibin-beta Subunits/genetics , Muscle, Smooth, Vascular/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Cell Differentiation , Cell Line , Cells, Cultured , Femoral Artery/cytology , Femoral Artery/pathology , Genetic Vectors , Humans , Hyperplasia , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/pharmacology , Kinetics , Mice , Muscle Contraction , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Organ Culture Techniques , Phenotype , RNA, Messenger/biosynthesis , Saphenous Vein/anatomy & histology , Saphenous Vein/cytology , Tunica Intima/pathology
12.
Vascul Pharmacol ; 39(6): 293-301, 2002 Dec.
Article in English | MEDLINE | ID: mdl-14567067

ABSTRACT

Intimal hyperplasia resulting from vascular injury remains a major obstacle in the long-term success of coronary artery bypass grafts. Inhibition of smooth muscle cell (SMC) proliferation using adenoviral gene transfer of cell cycle inhibitors resulted in reduced neointima formation in various animal models. However, little is known about the effect on human SMCs and neointima formation. Here we report the effects of infection with an adenoviral vector encoding a constitutively active form of the retinoblastoma gene (Ad. delta Rb) on proliferation of human saphenous vein SMCs (HSVSMCs) and neointima formation in organ cultures of human saphenous vein. Proliferation of SMCs was inhibited dose-dependently after infection with Ad. delta Rb. A near-total inhibition was found at an Ad. delta Rb concentration of 10(8) pfu/ml. Organ cultures of human saphenous vein segments were used to evaluate the effect of Ad. delta Rb infection on neointima formation and vein graft disease. Segments cultured for 4 weeks develop a neointima that is morphologically highly similar to early initimal lesions found in pathological vein grafts in vivo. Infection of saphenous vein segments with 2 x 10(9) pfu/ml Ad. delta Rb resulted in a 59% reduction of neointimal area when compared to uninfected counterparts, whereas infection with control adenovirus, Ad.LacZ, had no significant effect. The results of this study show that Ad. delta Rb gene transfer might be an efficient approach to prevent neointima formation in human saphenous vein grafts.


Subject(s)
Adenoviridae/genetics , Coronary Artery Bypass , Graft Occlusion, Vascular/pathology , Muscle, Smooth, Vascular/physiology , Mutation/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Cell Division/physiology , Cells, Cultured , Graft Occlusion, Vascular/genetics , Heterozygote , Humans , Hyperplasia/pathology , Image Processing, Computer-Assisted , Immunohistochemistry , Muscle, Smooth, Vascular/cytology , Organ Culture Techniques , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein/cytology , Saphenous Vein/growth & development , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...