151
|
Affiliation(s)
- Anupam Agarwal
- Department of Medicine, Division of Nephrology, Hypertension and Transplantation, University of Florida College of Medicine, Gainesville, Florida, USA.
| | | |
Collapse
|
152
|
Hilker M, Längin T, Hake U, Schmid FX, Kuroczynski W, Lehr HA, Oelert H, Buerke M. Gene expression profiling of human stenotic aorto-coronary bypass grafts by cDNA array analysis. Eur J Cardiothorac Surg 2003; 23:620-5. [PMID: 12694787 DOI: 10.1016/s1010-7940(03)00017-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Aorto-coronary bypass graft disease with its increasing clinical signification represents an unsolved problem in cardiological and heart surgery practice. Late occlusion of autologous saphenous vein grafts is due to medial and neointimal thickening secondary to migration and proliferation of smooth muscle cells (SMCs) and the subsequent formation of atherosclerotic plaques. This study is aimed at identifying differentially expressed genes in human stenotic bypass grafts to detect unknown pathomechanism and to identify novel targets for prophylactic treatment options. METHODS Stenotic saphenous aorto-coronary bypass grafts (n=5) were retrieved during re-do aorto-coronary bypass surgery. Ungrafted saphenous vein segments (n=5) were taken from the same group of patients and served as internal controls. cDNA samples were prepared and hybridized to cDNA arrays. RESULTS Some of the differentially expressed genes complied with expected gene expression including upregulation of c-jun and CDK10. In addition, previously unidentified gene expression patterns were detected such as upregulation of HSP70, fibronectin1, erbB3 proto-oncogene and c-myc. To confirm the latter finding, upregulation of c-myc in neointimal and medial SMCs of stenotic graft segments was confirmed by in situ hybridization studies and by immunohistochemistry. CONCLUSION Gene expression patterns of human stenotic bypass grafts retrieved by re-do operations can be reliably analyzed by cDNA array technology. With this technique, new therapeutic targets in patients could be identified as shown by the findings regarding c-myc. c-myc is a proto-oncogene acting as a transcription factor and blocking c-myc has shown a reduction of neointima formation in animal models. Our study yields a rational for the use of antisense c-myc oligonucleotides to reduce neointima formation and to avoid stenosis in patients.
Collapse
Affiliation(s)
- Michael Hilker
- Department of Thoracic and Cardiovascular Surgery, Johannes Gutenberg University, Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Knoflach M, Mayrl B, Mayerl C, Sedivy R, Wick G. Atherosclerosis as a paradigmatic disease of the elderly: role of the immune system. Immunol Allergy Clin North Am 2003; 23:117-32. [PMID: 12645882 DOI: 10.1016/s0889-8561(02)00077-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
When a new hypothesis about the etiology and pathogenesis of a disease is developed, there is always the danger that it will be presented as the only acceptable explanation for the occurrence of a given pathologic condition. In view of the well-proven multifactoral pathogenesis of atherosclerosis, we would like to emphasize that we are not postulating that immunity to HSP60 is the only cause of atherogenesis, especially in the later stages where there are clinically-apparent sequelae, such as myocardial infarction, stroke, and other atherosclerosis-dependent symptoms. In this article, we summarized some of the experimental and clinical data that we and others have collected in support of the concept that atherosclerosis is a good example of pleotropic antagonism, and postulated that age-dependent diseases are the price we pay for genetic traits established by natural selection to assure maximum survival until the age of reproduction, the effects of which may, however, become deleterious later in life. In the present case, the cost we pay for protective immunity to microbial and altered autologous HSP60 is the risk of cross-reactivity with HSP60 expressed by arterial endothelial cells that are subjected to stress factors already known as classical atherosclerosis risk factors. We showed that the first inflammatory stage of atherosclerosis starts early in life, long before it becomes clinically apparent. More severe lesions that lead to atherosclerosis-dependent organ-specific or systemic symptoms will only occur if classical atherosclerosis risk factors, especially those involving the cholesterol metabolism, remain present.
Collapse
Affiliation(s)
- Michael Knoflach
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria
| | | | | | | | | |
Collapse
|
154
|
Henderson B, Tagwerker A, Mayrl C, Pfister G, Boeck G, Ulmer H, Dietrich H, Wick G. Progression of arteriovenous bypass restenosis in mice exposed to a 50 Hz magnetic field. Cell Stress Chaperones 2003; 8:373-80. [PMID: 15115289 PMCID: PMC514908 DOI: 10.1379/1466-1268(2003)008<0373:poabri>2.0.co;2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2003] [Accepted: 09/02/2003] [Indexed: 11/24/2022] Open
Abstract
The controversy over whether magnetic fields (MF) produced by electrical wiring and appliances contribute to diseases such as cancer has been debated in the literature for more than 2 decades. These extremely low frequency fields at 50 or 60 Hz are omnipresent in the industrialized world and have been linked to various forms of cancer by epidemiological studies. Little has been published investigating any possible role of MF and cardiovascular disease, and this is the first study looking specifically at the effect of exposure to high-intensity MF on the development and progression of restenosis. A mouse arteriovenous bypass model was used, and mice were exposed to MF for periods of 1, 2, or 3 weeks. Neointima formation, infiltration of mononuclear cells, and heat shock protein 60 expression were all studied at the conclusion of the exposure regimen. Animals exposed to the MF for 1 week showed significantly smaller neointima formation compared with control mice exposed to a null field, although this difference was not observed in mice exposed for 2 or 3 weeks. No difference was found between mice exposed to MF and controls in any of the other parameters investigated.
Collapse
Affiliation(s)
- Blair Henderson
- Institute of Pathophysiology, Medical School, University of Innsbruck, Fritz-Pregl Strasse 3, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Angelini GD, Lloyd C, Bush R, Johnson J, Newby AC. An external, oversized, porous polyester stent reduces vein graft neointima formation, cholesterol concentration, and vascular cell adhesion molecule 1 expression in cholesterol-fed pigs. J Thorac Cardiovasc Surg 2002; 124:950-6. [PMID: 12407378 DOI: 10.1067/mtc.2002.127004] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Reducing neointima formation and atherosclerosis are key goals in preventing late vein graft failure. Although pharmacologic and mechanical solutions have been proposed, the demonstration that these influence both aspects of vein graft pathology have been lacking. Supporting grafts externally with an oversized, highly porous polyester stent dramatically reduces neointima formation in normocholesterolemic pigs. However, its effects in the presence of hypercholesterolemia are unknown. METHODS We compared wall thickening, cholesterol concentration, foam-cell formation, and the expression of the leukocyte adhesion molecule vascular cell adhesion molecule 1 after 3 months in stented and unstented saphenous vein interposition grafts into the carotid arteries of pigs fed cholesterol to cause modest hypercholesterolemia (11.2 +/- 1.2 mmol/L). RESULTS Stenting reduced neointima formation from 5.6 +/- 0.4 to 1.2 +/- 0.2 mm(2) (n = 7; P <.00002, paired t test) and graft cholesterol concentration from 4.7 +/- 1.2 to 2.1 +/- 1.3 mg/g wet weight (P <.02). Foam cells were observed in unstented grafts (mean, 1.5% +/- 0.5% of all cells) but never in stented grafts. Vascular cell adhesion molecule 1 was strongly expressed in 53% +/- 8% of intimal and medial cells in unstented grafts but was weakly expressed in only 19% +/- 3% (n = 4, P <.05) of stented grafts. CONCLUSIONS We conclude that external stenting with polyester favorably influences both neointima formation and early atherosclerosis, both of which are key aspects of vein graft disease, and that decreased expression of vascular cell adhesion molecule 1 is part of the underlying mechanism.
Collapse
Affiliation(s)
- Gianni D Angelini
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, United Kingdom
| | | | | | | | | |
Collapse
|
156
|
Abstract
Gene therapy could improve human saphenous vein (HSV) coronary vein-graft patency by reducing early thrombosis, neointimal hyperplasia and atherosclerosis. Mouse and rabbit models use veins with much thinner walls than pig or HSVs but atherosclerosis can be more easily induced; none of these models shows early thrombosis. Prostacyclin synthase, tissue factor pathway inhibitor, and tissue plasminogen activator might decrease thrombus formation. Tissue inhibitors of metalloproteinases (TIMPs) reduce intimal migration of smooth muscle cells, while TIMP-3 and the p53 tumor suppressor protein promote apoptosis. Prostacyclin synthase and nitric oxide synthase, and cell cycle inhibitors, such as E2F decoy oligonucleotides (D-E2F), reduce neointima formation. This might be enough by itself to decrease later atherosclerosis. Alternatively, direct targeting with nitric oxide synthase, decoy adhesion molecules, or interleukin-10 might be possible.
Collapse
Affiliation(s)
- Stephen J White
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| | | |
Collapse
|
157
|
Lardenoye JHP, de Vries MR, Löwik CWGM, Xu Q, Dhore CR, Cleutjens JPM, van Hinsbergh VWM, van Bockel JH, Quax PHA. Accelerated atherosclerosis and calcification in vein grafts: a study in APOE*3 Leiden transgenic mice. Circ Res 2002; 91:577-84. [PMID: 12364385 DOI: 10.1161/01.res.0000036901.58329.d7] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vein grafts fail due to development of intimal hyperplasia and accelerated atherosclerosis. Many murine genetic models in which genes are overexpressed, deleted, or mutated have been introduced recently. Therefore, mouse models are very well suited to dissect the relative contribution of different genes in the development of accelerated atherosclerosis. In the present study, we evaluated whether accelerated atherosclerosis in human vein grafts could be mimicked in hypercholesterolemic APOE*3 Leiden transgenic mice. Venous bypass grafting was performed in the carotid artery in APOE*3 Leiden mice fed either a standard chow diet or a high cholesterol-rich diet for 4 weeks. At several time points (0 hour to 28 days), mice were euthanized and the morphology of the vein grafts was analyzed. In normocholesterolemic mice, vein graft thickening up to 10-fold original thickness, predominantly consisting of alpha-smooth muscle cell actin-positive cells, was observed after 28 days. In hypercholesterolemic mice, accelerated atherosclerosis with accumulation of lipid-loaded foam cells was observed within 7 days after surgery. This accelerated atherosclerosis progressed in time and resulted in significant increase in vein graft thickening up to 50 times original thickness with foam cell-rich lesions and calcification within 28 days after surgery. The atherosclerotic lesions observed in these murine grafts show high morphological resemblance with the atherosclerotic lesions observed in human vein grafts. This accelerated, diet-dependent induction of atherosclerotic-like lesions in murine vein grafts provides a valuable tool in evaluating the mechanisms of accelerated atherosclerosis and therapeutic interventions of vein graft disease.
Collapse
|
158
|
Hu Y, Mayr M, Metzler B, Erdel M, Davison F, Xu Q. Both donor and recipient origins of smooth muscle cells in vein graft atherosclerotic lesions. Circ Res 2002; 91:e13-20. [PMID: 12364395 DOI: 10.1161/01.res.0000037090.34760.ee] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Smooth muscle cell (SMC) accumulation in the inner layer of the vessel wall is a key event in the pathogenesis of atherosclerosis in vein grafts, but the origin of the cells in these lesions has yet to be shown. Herein, we use animal models of vein grafts in transgenic mice to clearly identify the sources of SMCs in atherosclerosis. Vena cava segments were isografted to carotid arteries between four types of transgenic mice, including SM-LacZ expressing beta-galactosidase (beta-gal) in vascular SMCs, SM-LacZ/apoE(-/-), ROSA26 expressing beta-gal in all tissues, and wild-type mice. beta-gal-positive cells were observed in neointimal and atherosclerotic lesions of all vein segments grafted between LacZ transgenic and wild-type mice. Double staining for beta-gal and cell nuclei revealed that about 40% of SMCs originated from hosts and 60% from the donor vessel. This was confirmed by double labeling of the Y-chromosome and alpha-actin in the lesions of sex-mismatched vein grafts. The possibility that bone marrow cells were the source of SMCs in grafts was eliminated by the absence of beta-gal staining in atherosclerotic lesions of chimeric mice. Furthermore, vein SMCs of SM-LacZ mice did not express beta-gal in situ, but did so when these cells appeared in atherosclerotic lesions in vivo, suggesting that hemodynamic forces may be crucial for SMC differentiation. Thus, we provide the first evidence of SMC origins in the atherosclerotic lesions of vein grafts, which will be essential for providing insight into new types of therapy for the disease. The full text of this article is available at http://www.circresaha.org.
Collapse
MESH Headings
- Actins/analysis
- Actins/immunology
- Animals
- Apolipoproteins E/genetics
- Arteriosclerosis/etiology
- Arteriosclerosis/pathology
- Cell Differentiation
- Cells, Cultured
- Chimera
- Hematopoietic Stem Cells/cytology
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle, Smooth, Vascular/chemistry
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/transplantation
- Tissue Donors
- Veins/chemistry
- Veins/transplantation
- beta-Galactosidase/genetics
Collapse
Affiliation(s)
- Yanhua Hu
- Institute for Pathophysiology, University of Innsbruck Medical School, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
159
|
Hilker M, Tellmann G, Buerke M, Gloger K, Moersig W, Oelert H, Hake U, Lehr HA. Proliferative activity in stenotic human aortocoronary bypass grafts. Cardiovasc Pathol 2002; 11:284-90. [PMID: 12361839 DOI: 10.1016/s1054-8807(02)00113-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Aortocoronary bypass graft disease is responsible for long-term failure of autologous vein grafts. The analyses of proliferation and cell type characterisation in human bypass grafts harvested during re-do surgery make it possible to investigate the cellular processes leading to bypass graft failure. METHODS 30 stenotic vein grafts and 25 control veins were explantated during re-do heart surgery procedures. The total area and cell count of the neointima, media and adventitia were calculated computer-assisted. Actively proliferating cells were identified using antibody to Ki-67 and positive cells were determined by double-label immunocytochemistry with SMC alpha-actin, CD 31 (endothelial cells), CD 68 (macrophages) and CD 45 (T-lymphocytes). RESULTS Active proliferation was detected in different cell types with an average proliferation index of 0.15%, 0.18% and 0.086% for neointima, media and adventitia. Only 9% of proliferating cells in the neointima were SMC (not identified cells 40%); correspondingly, 14% SMC (not identified cells 33%) were detected in the media. Endothelial cells turned out to be the predominant proliferating cell type in all sections of the vessel wall. CONCLUSION Proliferation in our series of stenotic vein grafts occurred at a low level, but was significantly higher compared to native control veins. While proliferation may play an important role in early lesions, our data clearly show low proliferation activity in advanced graft lesions. The identification of proliferating macrophages and T-lymphocytes implicate an additional inflammatory component in the development of human bypass graft disease. SUMMARY To clarify the role of cellular proliferation in human aortocoronary bypass grafts, we characterized the cellular composition and proliferation index in 30 stenotic saphenous vein grafts in comparison to 25 native veins. Proliferation in our series of stenotic vein grafts occurred at a low level, but was significantly higher compared to native control veins.
Collapse
Affiliation(s)
- Michael Hilker
- Department of Thoracic and Cardiovascular Surgery, Johannes Gutenberg University Mainz, Langenbeckstr. 1, D-55131 Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
160
|
Lardenoye JHP, De Vries MR, Grimbergen JM, Havekes LM, Knaapen MWM, Kockx MM, van Hinsbergh VWM, van Bockel JH, Quax PHA. Inhibition of accelerated atherosclerosis in vein grafts by placement of external stent in apoE*3-Leiden transgenic mice. Arterioscler Thromb Vasc Biol 2002; 22:1433-8. [PMID: 12231562 DOI: 10.1161/01.atv.0000030339.79524.6e] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vein grafts fail because of the development of intimal hyperplasia and accelerated atherosclerosis. Placement of an external stent around vein grafts resulted in an inhibition of intimal hyperplasia in several animal studies. Here, we assess the effects of external stenting on accelerated atherosclerosis in early vein grafts in carotid arteries in hypercholesterolemic apolipoprotein E*3-Leiden transgenic mice. METHODS AND RESULTS Venous interposition grafting was performed in apolipoprotein E*3-Leiden mice fed standard chow or a highly cholesterol-rich diet for 4 weeks. After engraftment, external stents with different inner diameters (0.4 or 0.8 mm) were placed. In unstented vein grafts in hypercholesterolemic mice, thickening up to 50 times the original thickness, with foam cell-rich lesions, calcification, and necrosis, was observed within 28 days. The atherosclerotic lesions observed show high morphological resemblance to atherosclerotic lesions observed in human vein grafts. In stented vein grafts in hypercholesterolemic mice, no foam cell accumulation or accelerated atherosclerosis was observed. Compared with unstented vein grafts, stenting of vein grafts in a hypercholesterolemic environment resulted in a 94% reduction of vessel wall thickening. These effects were independent of stent size. CONCLUSIONS Extravascular stent placement results in strong inhibition of accelerated vein graft atherosclerosis in hypercholesterolemic transgenic mice and thereby provides a perspective for therapeutic intervention in vein graft diseases.
Collapse
|
161
|
Chen AF, Ren J, Miao CY. Nitric oxide synthase gene therapy for cardiovascular disease. JAPANESE JOURNAL OF PHARMACOLOGY 2002; 89:327-36. [PMID: 12233810 DOI: 10.1254/jjp.89.327] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Gene therapy refers to the transfer of specific genes to the host tissue to intervene in a disease process, with resultant alleviation of the symptoms of a particular disease. Cardiovascular gene transfer is not only a powerful technique for studying the function of specific genes in cardiovascular biology and pathobiology, but also a novel and promising strategy for treating cardiovascular diseases. Since the mid-1990s, nitric oxide synthase (NOS), the enzyme that catalyzes the formation of nitric oxide (NO) from L-arginine, has received considerable attention as a potential candidate for cardiovascular gene therapy, because NO exerts critical and diverse functions in the cardiovascular system, and abnormalities in NO biology are apparent in a number of cardiovascular disease processes including cerebral vasospasm, atherosclerosis, postangioplasty restenosis, transplant vasculopathy, hypertension, diabetes mellitus, impotence and delayed wound healing. There are three NOS isoforms, i.e., endothelial (eNOS), neuronal (nNOS) and inducible (iNOS). All three NOS isoforms have been used in cardiovascular gene transfer studies with encouraging results. This review will discuss the rationale of NOS gene therapy in different cardiovascular disease settings and summarize the results of experimental NOS gene therapy from various animal models of cardiovascular disease to date.
Collapse
Affiliation(s)
- Alex F Chen
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing 48824-1317, USA.
| | | | | |
Collapse
|
162
|
Hilker M, Tellmann G, Buerke M, Moersig W, Oelert H, Lehr HA, Hake U. Expression of the proto-oncogene c-myc in human stenotic aortocoronary bypass grafts. Pathol Res Pract 2002; 197:811-6. [PMID: 11795828 DOI: 10.1078/0344-0338-00164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Proliferation and differentiation of vascular smooth muscle cells (VSMC) are central events in vascular pathobiology and play a major role in the development of stenotic and restenotic lesions. The proto-oncogene c-myc and other early cell cycle-regulating genes have been implicated in the induction of cell proliferation and differentiation under diverse pathophysiological conditions. In the present study we analyzed c-myc mRNA expression by indirect nonradioactive in situ hybridization technique (NISH) in human stenotic venous bypass grafts (n = 32) retrieved during re-do operations of coronary artery disease and compared the results with 28 native veins (vena saphena magna) from the same patients. Stenotic bypass grafts showed enhanced c-myc expression located predominantly in VSMC in the media and neointima (severity score: ++-+++, 32/32 stenotic veins). In native veins we observed only low levels of c-myc mRNA (severity score: +, 28/28 native veins), all signals were restricted to endothelial cells of either the innermost intimal layer or of the vasa vasorum. Our in situ hybridization studies demonstrate enhanced mRNA expression of the proto-oncogene c-myc in stenotic venous bypass grafts. These results suggest that--in analogy to other pathophysiological conditions--c-myc exerts essential regulatory functions in cellular events operative during the initiation and progression of venous bypass graft disease.
Collapse
Affiliation(s)
- M Hilker
- Klinik für Herz-, Thorax- und Gefässchirurgie, Universitätskliniken der Johannes-Gutenberg-Universität Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
163
|
Mayr U, Mayr M, Li C, Wernig F, Dietrich H, Hu Y, Xu Q. Loss of p53 accelerates neointimal lesions of vein bypass grafts in mice. Circ Res 2002; 90:197-204. [PMID: 11834713 DOI: 10.1161/hh0202.103715] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcription factor p53 is essentially involved in regulation of cell death and proliferation. Recently, we have established a mouse model for vein graft arteriosclerosis by grafting autologous jugular veins or vena cava to carotid arteries. Using this model, we studied the role of p53 in the development of vein graft arteriosclerosis in p53(-/-) mice. Four weeks after grafting, neointimal hyperplasia of vein grafts in p53(-/-) mice was increased 2-fold compared with that of wild-type controls. Cell component analysis revealed that neointimal lesions in p53(-/-) mice consisted mainly of alpha-actin positive smooth muscle cells (SMCs), whereas the majority of cells in wild-type mice were MAC-1 (CD11b/18)-positive at 4 weeks. Importantly, SMC apoptosis as determined by TUNEL assay was significantly reduced in p53(-/-) vein grafts. TUNEL positive cells in wild-type vein grafts markedly increased from 0.5% to 6.4% of total cells 4 weeks postoperatively, but remained virtually unchanged in p53(-/-) grafts (0.8%). Immunofluorescence analysis revealed that increased p53 expression in neointimal SMCs of wild-type, but not p53(-/-), mice coincided with oxidative DNA damage in vein grafts. Interestingly, SMCs of p53(-/-) mice showed increased apoptosis in response to TNFalpha and decreased apoptosis in response to sodium nitroprusside. Additionally, p53-deficient SMCs showed a higher rate of proliferation and migration and expressed higher levels of matrix metalloproteinases. Thus, p53 deficiency accelerates neointima formation by facilitating SMC proliferation as well as abrogating cell apoptosis.
Collapse
MESH Headings
- Actins/biosynthesis
- Animals
- Apoptosis/drug effects
- Carotid Arteries/surgery
- Cell Division/genetics
- Cell Movement/genetics
- DNA Damage/genetics
- Disease Progression
- Genetic Predisposition to Disease
- Graft Occlusion, Vascular/etiology
- Graft Occlusion, Vascular/genetics
- Graft Occlusion, Vascular/metabolism
- Graft Occlusion, Vascular/pathology
- Heterozygote
- In Situ Nick-End Labeling
- Jugular Veins/metabolism
- Jugular Veins/pathology
- Jugular Veins/transplantation
- Macrophage-1 Antigen/biosynthesis
- Mice
- Mice, Knockout
- Mice, Mutant Strains
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Nitroprusside/pharmacology
- Oxidation-Reduction
- Transplantation, Autologous/adverse effects
- Tumor Necrosis Factor-alpha/pharmacology
- Tumor Suppressor Protein p53/deficiency
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Vascular Surgical Procedures/adverse effects
- Veins/metabolism
- Veins/pathology
- Veins/transplantation
Collapse
Affiliation(s)
- Ursula Mayr
- Institute for Pathophysiology, University of Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
164
|
Wernig F, Xu Q. Mechanical stress-induced apoptosis in the cardiovascular system. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2002; 78:105-37. [PMID: 12429110 DOI: 10.1016/s0079-6107(02)00008-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
All tissues in the body are subjected to physical forces originating either from tension, created by cells themselves, or from the environment. Particularly, the cardiovascular system is continuously subjected to haemodynamic forces created by blood flow and blood pressure. While biomechanical force at physiological levels is essential to develop and maintain organic structure and function, elevated mechanical stress may result in cell death leading to pathological conditions. In recent years, however, it has been widely recognized that cell death, namely apoptosis, is not just the response to an injury but a highly regulated and controlled process. Therefore, physical stimuli must be sensed by cells and transmitted through intracellular signal transduction pathways to the nucleus, resulting in cell apoptosis. Disturbances in the regulatory mechanisms of apoptosis often precede the development of a disease. Exploration of the molecular signalling mechanisms leading to mechanical stress-induced apoptosis in cardiovascular disorders revealed the crucial role of apoptosis in the pathogenesis of these diseases. For instance, heart failure, hypertension and atherosclerosis are believed to be related to sustained mechanical overloading or stress. In this review we summarize the recent data focusing on molecular mechanisms of mechanical stress-induced apoptosis and highlight the role of apoptosis in the development of cardiovascular disorders, which may lead to new therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Florian Wernig
- Department of Cardiological Sciences, St George's Hospital Medical School, Cranmer Terrace, Tooting, London SW17 0RE, UK
| | | |
Collapse
|
165
|
Leitges M, Mayr M, Braun U, Mayr U, Li C, Pfister G, Ghaffari-Tabrizi N, Baier G, Hu Y, Xu Q. Exacerbated vein graft arteriosclerosis in protein kinase Cδ–null mice. J Clin Invest 2001. [DOI: 10.1172/jci200112902] [Citation(s) in RCA: 200] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
166
|
Leitges M, Mayr M, Braun U, Mayr U, Li C, Pfister G, Ghaffari-Tabrizi N, Baier G, Hu Y, Xu Q. Exacerbated vein graft arteriosclerosis in protein kinase Cdelta-null mice. J Clin Invest 2001; 108:1505-12. [PMID: 11714742 PMCID: PMC209416 DOI: 10.1172/jci12902] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Smooth muscle cell (SMC) accumulation is a key event in the development of atherosclerosis, including vein bypass graft arteriosclerosis. Because members of the protein kinase C (PKC) family signal cells to undergo proliferation, differentiation, or apoptosis, we generated PKCdelta knockout mice and performed vein bypass grafts on these animals. PKCdelta(-/-) mice developed normally and were fertile. Vein segments from PKCdelta(-/-) mice isografted to carotid arteries of recipient mice of either genotype led to a more severe arteriosclerosis than was seen with PKCdelta(+/+) vein grafts. Arteriosclerotic lesions in PKCdelta(-/-) mice showed a significantly higher number of SMCs than were found in wild-type animals; this was correlated with decreased SMC death in lesions of PKCdelta(-/-) mice. SMCs derived from PKCdelta(-/-) aortae were resistant to cell death induced by any of several stimuli, but they were similar to wild-type SMCs with respect to mitogen-stimulated cell proliferation in vitro. Furthermore, pro-apoptotic treatments led to diminished caspase-3 activation, poly(ADP-ribose) polymerase cleavage, and cytochrome c release in PKCdelta(-/-) relative to wild-type SMCs, suggesting that their apoptotic resistance involves the loss of free radical generation and mitochondrial dysfunction in response to stress stimuli. Our data indicate that PKCdelta maintains SMC homeostasis and that its function in the vessel wall per se is crucial in the development of vein graft arteriosclerosis.
Collapse
Affiliation(s)
- M Leitges
- Max Planck Institute for Experimental Endocrinology, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Hu Y, Baker AH, Zou Y, Newby AC, Xu Q. Local gene transfer of tissue inhibitor of metalloproteinase-2 influences vein graft remodeling in a mouse model. Arterioscler Thromb Vasc Biol 2001; 21:1275-80. [PMID: 11498453 DOI: 10.1161/hq0801.093658] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
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.
Collapse
Affiliation(s)
- Y Hu
- Institute for General and Experimental Pathology, University of Innsbruck Medical School, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
168
|
Abstract
Arteriosclerosis, a paradigmatic age-related disease, encompasses (spontaneous) atherosclerosis, restenosis after percutaneous transluminal coronary angioplasty, autologous arterial or vein graft arteriosclerosis and transplant arteriosclerosis. In all types of arteriosclerosis, vascular smooth muscle cell (SMC) accumulation in the intima is a key event, but abundant evidence also indicates the importance of SMC apoptosis in the development of arteriosclerosis. Because SMC proliferation and apoptosis coincide in arteriosclerotic lesions, the balance between these two processes could be a determinant during vessel remodeling and disease development. Various stimuli, including oxidized lipoproteins, altered hemodynamic stress and free radicals, can induce SMC apoptosis in vitro. As risk factors for arteriosclerosis, these stimuli may also lead to vascular cell apoptosis in vivo. The presence of apoptotic cells in atherosclerotic and restenotic lesions could have potential clinical implications for atherogenesis and contributes to the instability of the lesion. Based on the progress in this field, this review focuses on the mechanism and impact of SMC apoptosis in the pathogenesis of arteriosclerosis and highlights the role of biomechanical stress in SMC apoptosis.
Collapse
Affiliation(s)
- M Mayr
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria
| | | |
Collapse
|
169
|
Dietrich H, Hu Y, Zou Y, Huemer U, Metzler B, Li C, Mayr M, Xu Q. Rapid development of vein graft atheroma in ApoE-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:659-69. [PMID: 10934168 PMCID: PMC1850116 DOI: 10.1016/s0002-9440(10)64576-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Several animal models manifesting lesions resembling neointimal hyperplasia of human vein grafts have been developed, but no spontaneous atheromatous lesions in their vein grafts have been observed. We developed and here characterize a new animal model of vein graft atheroma, a maturated atherosclerotic plaque, in apoE-deficient mice. The lesion displayed classical complex morphological features and heterogeneous cellular compositions and consisted of a fibrous cap, infiltrated mononuclear cells, foam cells, cholesterol crystal structure, necrotic core with calcification, and neovasculature. Cell component analysis revealed smooth muscle cells (SMCs) localized in the cap region, macrophages which made up a large portion of the lesions, and CD4+ T cells scattered under the cap. Importantly, apoptotic/necrotic cells determined by TUNEL assay in vein grafts into apoE-/- mice were significantly higher than wild-type mice, although a similar number of proliferating cell nuclear antigen-positive cells in both types of lesions was found. Interestingly, vascular SMCs cultivated from aortas of apoE-deficient mice showed a high rate of spontaneous apoptosis/necrosis and a higher rate of cell death stimulated by a nitric oxide donor, sodium nitroprusside, H(2)O(2), and oxidized low density lipoprotein (LDL), although no difference in proliferation of both SMCs incubated with platelet-derived growth factor, angiotensin II, LDL, and oxidized LDL was seen. Thus, the pathogenic mechanisms of vein graft atheroma involve increased intimal cell death initiated by biomechanical stress and amplified by hypercholesterolemia, which leads to continuous recruitment of blood mononuclear cells to constitute atheromatous lesions. This mouse model resembling human vein graft disease has many advantages over other animal models.
Collapse
Affiliation(s)
- H Dietrich
- Institute for General and Experimental Pathology, University of Innsbruck Medical School, Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
170
|
Abstract
Mechanical force is an important modulator of cellular morphology and function in a variety of tissues, and is particularly important in cardiovascular systems. Vascular smooth muscle cell (VSMC) hypertrophy and proliferation contribute to the development of atherosclerosis, hypertension, and restenosis, where mechanical forces are largely disturbed. How VSMCs sense and transduce the extracellular mechanical signals into the cell nucleus resulting in quantitative and qualitative changes in gene expression is an interesting and important research field. Recently, it has been demonstrated that mechanical stress rapidly induced phosphorylation of platelet-derived growth factor (PDGF) receptor, activation of integrin receptor, stretch-activated cation channels, and G proteins, which might serve as mechanosensors. Once mechanical force is sensed, protein kinase C and mitogen-activated protein kinases (MAPKs) were activated, leading to increased c-fos and c-jun gene expression and enhanced transcription factor AP-1 DNA-binding activity. Interestingly, physical forces also rapidly resulted in expression of MAPK phosphatase-1 (MKP-1), which inactivates MAPKs. Thus, mechanical stresses can directly stretch the cell membrane and alter receptor or G protein conformation, thereby initiating signalling pathways, usually used by growth factors. These findings have significantly enhanced our knowledge of the pathogenesis of arteriosclerosis and provided promising information for therapeutic interventions for vascular diseases.
Collapse
Affiliation(s)
- C Li
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, A-6020, Innsbruck, Austria
| | | |
Collapse
|
171
|
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.
Collapse
Affiliation(s)
- A C Newby
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol, UK.
| |
Collapse
|
172
|
Zou Y, Hu Y, Mayr M, Dietrich H, Wick G, Xu Q. Reduced neointima hyperplasia of vein bypass grafts in intercellular adhesion molecule-1-deficient mice. Circ Res 2000; 86:434-40. [PMID: 10700448 DOI: 10.1161/01.res.86.4.434] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recently, we established a new mouse model of vein graft arteriosclerosis through the grafting of 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. With this model, we studied the role of intercellular adhesion molecule-1 (ICAM-1) in the development of vein graft arteriosclerosis in ICAM-1-deficient mice. Neointimal hyperplasia of vein grafts in ICAM-1 -/- mice was reduced 30% to 50% compared with that of wild-type control animals. Immmunofluorescent analysis revealed that increased ICAM-1 expression was observed on the endothelium and smooth muscle cells (SMCs) of the grafted veins in wild-type, but not ICAM-1 -/-, mice. MAC-1 (CD11b/18)-positive cells that adhered to the surface of vein grafts in ICAM-1 -/- mice were significantly less as identified with en face immunofluorescence, and these positive cells were more abundant in the intimal lesions of vein grafts in wild-type mice. Furthermore, aortic SMCs cultivated from wild-type mice exhibited high ICAM-1 expression in response to tumor necrosis factor-alpha. When tumor necrosis factor-alpha-stimulated SMCs were incubated with mouse spleen leukocytes, the number of cells that adhered to ICAM-1 -/- SMCs was significantly lower than the number that adhered to ICAM-1 +/+ SMCs, which was markedly blocked through pretreatment of leukocytes with the anti-MAC-1 antibody. Taken together, our findings demonstrate that ICAM-1 is critical in the development of venous bypass graft arteriosclerosis, which provides essential information for therapeutic intervention for vein graft disease in patients undergoing bypass surgery.
Collapse
Affiliation(s)
- Y Zou
- Institute for Biomedical Aging Research, Austrian Academy of Sciences Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
173
|
Abstract
By definition, diseases of aging become clinically manifested in elderly patients. However, their pathogenetic basis has to be sought earlier in life. The general thread of this presentation relies on the concept of an evolutionary-Darwinian view of the development of age-related diseases. In essence, this concept states that we may have to "pay" for genetic traits that play a beneficial role earlier in life by the later development of diseases since there is no post-reproductive selective pressure that may have eliminated the potential late onset detrimental effects of such genes. Examples for this kind of trade-off are taken from diseases involving the immune system (infections), the endocrine system (andropause), the nervous system (Alzheimer's disease), the locomoter system (osteoporosis), the cardio-vascular system (atherosclerosis) and cancer.
Collapse
Affiliation(s)
- G Wick
- Institute for Biomedical Aging Research of the Austrian Academy of Sciences, Rennweg 10, 6020, Innsbruck, Austria.
| | | | | | | | | |
Collapse
|
174
|
Mayr M, Li C, Zou Y, Huemer U, Hu Y, Xu Q. Biomechanical stress-induced apoptosis in vein grafts involves p38 mitogen-activated protein kinases. FASEB J 2000; 14:261-70. [PMID: 10660448 DOI: 10.1096/fasebj.14.2.261] [Citation(s) in RCA: 123] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The present study was designed to investigate whether apoptosis occurs in early-stage vein grafts and to determine the mechanisms by which mechanical stress contributes to apoptosis in vascular smooth muscle cells (SMCs). Apoptosis in vessel walls of mouse vein grafts was confirmed by morphological changes and by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL). TUNEL(+) cells in vein grafts 1, 4, and 8 wk postoperatively was 13%, 29%, and 21%, respectively, and apoptosis occurred mainly in veins grafted to arteries, remaining unchanged in vein-to-vein grafts. When mouse, rat, and human arterial SMCs were cultured on a flexible membrane and subjected to cyclic strain stress, apoptosis was observed in a time- and strength-dependent manner. All three types of SMCs showed apoptotic death as confirmed by TUNEL, propidium iodide, and annexin V staining. To further study the signal pathways leading to apoptosis, activities of p38, a subfamily of mitogen-activated protein kinases (MAPKs), were determined. Mechanical stress resulted in p38 MAPK activation, reaching high levels within 8 min. SB 202190, a specific inhibitor for p38 MAPKs, prevented SMC apoptosis in response to mechanical stress. SMC lines stably transfected with a dominant negative rac, an upstream signal transducer, or overexpressing MAPK phosphatase-1, a negative regulator for MAPKs, completely inhibited mechanical stress stimulated p38 activation and abolished mechanical stress-induced apoptosis. Thus, we provide solid evidence that one of the earliest events in venous bypass grafts is apoptosis, in which mechanical stress-induced p38-MAPK activation is responsible for transducing signals leading to apoptosis.-Mayr, M., Li, C., Zou, Y., Huemer, U., Hu, Y., Xu, Q. Biomechanical stress-induced apoptosis in vein grafts involves p38 mitogen-activated protein kinases.
Collapse
Affiliation(s)
- M Mayr
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
175
|
Dietrich H, Hu Y, Zou Y, Dirnhofer S, Kleindienst R, Wick G, Xu Q. Mouse model of transplant arteriosclerosis: role of intercellular adhesion molecule-1. Arterioscler Thromb Vasc Biol 2000; 20:343-52. [PMID: 10669629 DOI: 10.1161/01.atv.20.2.343] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transplant-accelerated arteriosclerosis in coronary arteries is the major limitation to long-term survival of patients with heart transplantation. The pathogenesis of this disease is not fully understood. Herein, we describe a simplified model of artery allografts in the mouse that allows us to take advantage of transgenic, knockout, or mutant animals. Common carotid arteries or aortic vessels were end-to-end allografted into carotid arteries between C57BL/6J and BALB/c mice. Neointimal lesions were observed as early as 2 weeks after surgery and had progressed at 4 and 6 weeks postoperatively. The lumen of grafted arteries was significantly narrowed due to neointima hyperplasia 4 weeks after transplantation. Using this model, we studied the role of intercellular adhesion molecule-1 (ICAM-1) in the development of transplant arteriosclerosis in ICAM-1-deficient mice. Neointimal lesions of artery grafts from ICAM-1 -/- C57BL/6J to BALB/c mice were reduced up to 60% compared with wild-type controls. MAC-1 (CD11b/18)-positive cells adhering to the surface of ICAM-1 -/- artery grafts were significantly less as identified by en face immunofluorescence, and these positive cells were more abundant in intimal lesions of artery grafts in wild-type mice. Furthermore, the major cell component of neointimal lesions 4 weeks after surgery was found to be alpha-actin-positive smooth muscle cells, which were significantly reduced in lesions of ICAM-1 -/- artery grafts. Thus, this model has been proven to be useful for understanding the mechanism of transplant arteriosclerosis. Our findings demonstrate that ICAM-1 is critical in the development of allograft arteriosclerosis via mediation of leukocyte adhesion to, and infiltration into, the vessel wall.
Collapse
Affiliation(s)
- H Dietrich
- Institute for General and Experimental Pathology, University of Innsbruck Medical School, Austria
| | | | | | | | | | | | | |
Collapse
|
176
|
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.
Collapse
Affiliation(s)
- A C Newby
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| | | |
Collapse
|
177
|
Affiliation(s)
- G Wick
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | |
Collapse
|
178
|
Abstract
Poor long-term patency and a lack of suitable systemic pharmacologic therapy for the prevention of vein graft failure have prompted the search for effective local gene therapy. Vein grafts are particularly well suited for gene transfer in the clinic because direct access to vein is available during surgical preparation for grafting. In this review, the available animal models are discussed and a new mouse model is highlighted. Recent advances in gene transfer technology are reviewed, including the use of adeno-associated virus and modified adenoviruses that can prolong in vivo transgene expression for months. Gene therapy is intended to reduce early thrombosis, reduce neointima formation, and prevent atherosclerosis in vein grafts. Promising antithrombotic targets include tissue plasminogen activator and thrombomodulin. Nitric oxide synthase, prostacyclin synthase, and tissue inhibitors of metalloproteinases have been used to reduce neointima formation, and vein graft atheroma remains a challenge for the future.
Collapse
Affiliation(s)
- A C Newby
- Bristol Heart Institute, University of Bristol, United Kingdom.
| | | |
Collapse
|
179
|
Abstract
Irrespective of their intended use, the best models of spontaneous aneurysms in humans are produced under conditions analogous to those occurring in humans and specific for the site. Even if appropriate for other purposes, models dependent on artificial conditions (physical and thermal trauma, chemical and enzymatic degradative processes), not in compliance with the above, bear no relationship to the etiology or pathology of the lesion or disease under investigation. Surgical models of poststenotic dilatation and aneurysm, arteriovenous shunts, and venous graft aneurysms are suitable for study of the prevailing hemodynamics and pathological effects of the associated stresses on the vessel wall which have bearing on degenerative aneurysms at other sites. The protracted course of atherosclerosis and constraints of time and research funds when reproducing the pathology and conditions prevailing in the human situation legitimize the use of models which accelerate development and complications. The limitations of any model are of paramount consideration. The value of some current models of aortic and cerebral aneurysms is discussed.
Collapse
Affiliation(s)
- W E Stehbens
- Department of Pathlogy, Wellington School of Medicine, Wellington, New Zealand
| |
Collapse
|
180
|
Hu Y, Zou Y, Dietrich H, Wick G, Xu Q. Inhibition of neointima hyperplasia of mouse vein grafts by locally applied suramin. Circulation 1999; 100:861-8. [PMID: 10458724 DOI: 10.1161/01.cir.100.8.861] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Saphenous vein grafts are widely used for aortocoronary bypass surgery as treatment for severe atherosclerosis and often are complicated by subsequent occlusion of the graft vessel. METHODS AND RESULTS We described a mouse model of venous bypass graft arteriosclerosis that can be effectively retarded by locally applied suramin, a growth factor receptor antagonist. Mouse isogeneic vessels of the vena cava veins pretreated with suramin were grafted end to end into the carotid arteries and enveloped with a mixture of suramin (1 mmol/L) and pluronic-127 gel. In the untreated group, vessel wall thickening was observed as early as 1 week after surgery and progressed to 4-fold and 10-fold the original thickness in grafted veins at 4 and 8 weeks, respectively. Pluronic-127 gel alone did not influence neointima formation. Suramin treatment reduced the neointima hyperplasia 50% to 70% compared with untreated controls. Immunohistochemical studies demonstrated that a significant proliferation of vascular smooth muscle cells (SMCs) constituted neointimal lesions between 4 and 8 weeks. The majority of SMCs expressed platelet-derived growth factor (PDGF) receptors-alpha and -beta, which were significantly reduced by suramin treatment. In vitro studies indicated that suramin completely blocked PDGF receptor activation or phosphorylation stimulated by PDGF-AB, inhibited activation of mitogen-activated protein kinase (ERK) kinases (MEK1/2) and ERK1/2, and abrogated transcription factor AP-1 DNA-binding activity. CONCLUSIONS Suramin inhibited SMC migration and proliferation in vivo and in vitro by blocking PDGF-initiated PDGF receptor and MAPK-AP-1 signaling. These findings indicate that locally applied suramin is effective in a mouse model of venous bypass graft arteriosclerosis.
Collapse
Affiliation(s)
- Y Hu
- Institute for General and Experimental Pathology, University of Innsbruck Medical School, Innsbruck, Austria
| | | | | | | | | |
Collapse
|