51
|
Valente AJ, Yoshida T, Izadpanah R, Delafontaine P, Siebenlist U, Chandrasekar B. Interleukin-18 enhances IL-18R/Nox1 binding, and mediates TRAF3IP2-dependent smooth muscle cell migration. Inhibition by simvastatin. Cell Signal 2013; 25:1447-56. [PMID: 23541442 DOI: 10.1016/j.cellsig.2013.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/11/2013] [Indexed: 01/28/2023]
Abstract
We investigated the role of TRAF3 interacting protein 2 (TRAF3IP2), a redox-sensitive adapter protein and an upstream regulator of IKK and JNK in interleukin (IL)-18 induced smooth muscle cell migration, and the mechanism of its inhibition by simvastatin. The pleiotropic cytokine IL-18 induced human coronary artery SMC migration through the induction of TRAF3IP2. IL-18 induced Nox1-dependent ROS generation, TRAF3IP2 expression, and IKK/NF-κB and JNK/AP-1 activation. IL-18 induced its own expression and that of its receptor subunit IL-18Rα. Using co-IP/IB and GST pull-down assays, we show for the first time that the subunits of the IL-18R heterodimer physically associate with Nox1 under basal conditions, and IL-18 appears to enhance their binding. Importantly, the HMG-coA reductase inhibitor simvastatin attenuated IL-18-induced TRAF3IP2 induction. These inhibitory effects were reversed by mevalonate and geranylgeranylpyrophosphate (GGPP), but not by farnesylpyrophosphate (FPP). Interestingly, simvastatin, GGPP, FPP, or Rac1 inhibition did not modulate ectopically expressed TRAF3IP2. These results demonstrate that the promigratory effects of IL-18 are mediated through TRAF3IP2 in a redox-sensitive manner, and this may involve IL-18R/Nox1 physical association. Further, Simvastatin inhibits inducible, but not ectopically-xpressed TRAF3IP2. Targeting TRAF3IP2 may blunt progression of hyperplastic vascular diseases in vivo.
Collapse
Affiliation(s)
- Anthony J Valente
- Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX 78229, United States
| | | | | | | | | | | |
Collapse
|
52
|
Vascular miRNAs After Balloon Angioplasty. Trends Cardiovasc Med 2013; 23:9-14. [DOI: 10.1016/j.tcm.2012.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 11/21/2022]
|
53
|
Perek B, Malińska A, Nowicki M, Misterski M, Ostalska-Nowicka D, Jemielity M. Histological evaluation of age-related variations in saphenous vein grafts used for coronary artery bypass grafting. Arch Med Sci 2012; 8:1041-7. [PMID: 23319979 PMCID: PMC3542494 DOI: 10.5114/aoms.2012.32412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 02/02/2011] [Accepted: 02/12/2011] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Venous coronary artery bypass grafts (CABG) might undergo a process of arterialization resulting in neointimal formation and medial hypertrophy. It is often followed by critical occlusion of the graft lumen. The aim of the study was to assess histological representative features of saphenous vein reconstruction in aging as well as to establish optimal patients' age limits applicable for optimal selection of grafts. MATERIAL AND METHODS One hundred and ten patients undergoing venous CABG were divided into 4 age subgroups: (A) 50 years and less, (B) 51-60 years, (C) 61-70 years and (D) > 70 year-old subjects. Distal venous graft segments were saved for an adequate morphometric assay which was followed by suitable statistical analysis. RESULTS The entire venous wall thickness as well as its tunica media were found to become significantly thinner between subgroups A and D. The number of smooth muscle cell (SMC) nuclei within the tunica media did not differ between study subgroups. The majority of these nuclei in subgroup D were found, however, to be more elongated than in subgroup A (SMC length/width index in subgroup D was found to be significantly higher than in subgroup A). CONCLUSIONS Progressive, age-related thinning of the venous wall and tunica media as well as SMC nucleus elongation might suggest impairment of SMCs' migration and proliferation rate. Consequently, individuals aged 70 years and over may benefit clinically more from venous CABG than younger patients due to the lower risk of arterialization and occlusion of the graft lumen in the future.
Collapse
Affiliation(s)
- Bartłomiej Perek
- Department of Cardiac Surgery, Poznan University of Medical Sciences, Poland
| | - Agnieszka Malińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poland
| | - Marcin Misterski
- Department of Cardiac Surgery, Poznan University of Medical Sciences, Poland
| | - Danuta Ostalska-Nowicka
- Department of Paediatric Cardiology and Embryology, Poznan University of Medical Sciences, Poland
| | - Marek Jemielity
- Department of Cardiac Surgery, Poznan University of Medical Sciences, Poland
| |
Collapse
|
54
|
Wood ST, Dean BC, Dean D. A computational approach to understand phenotypic structure and constitutive mechanics relationships of single cells. Ann Biomed Eng 2012. [PMID: 23180027 DOI: 10.1007/s10439-012-0690-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The goal of this study is to construct a representative 3D finite element model (FEM) of individual cells based on their sub-cellular structures that predicts cell mechanical behavior. The FEM simulations replicate atomic force microscopy (AFM) nanoindentation experiments on live vascular smooth muscle cells. Individual cells are characterized mechanically with AFM and then imaged in 3D using a spinning disc confocal microscope. Using these images, geometries for the FEM are automatically generated via image segmentation and linear programming algorithms. The geometries consist of independent structures representing the nucleus, actin stress fiber network, and cytoplasm. These are imported into commercial software for mesh refinement and analysis. The FEM presented here is capable of predicting AFM results well for 500 nm indentations. The FEM results are relatively insensitive to both the exact number and diameter of fibers used. Despite the localized nature of AFM nanoindentation, the model predicts that stresses are distributed in an anisotropic manner throughout the cell body via the actin stress fibers. This pattern of stress distribution is likely a result of the geometric arrangement of the actin network.
Collapse
Affiliation(s)
- Scott T Wood
- Department of Bioengineering, Clemson University, 301 Rhodes Research Center, Clemson, SC 29634, USA
| | | | | |
Collapse
|
55
|
Trends in tissue engineering for blood vessels. J Biomed Biotechnol 2012; 2012:956345. [PMID: 23251085 PMCID: PMC3518873 DOI: 10.1155/2012/956345] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
Over the years, cardiovascular diseases continue to increase and affect not only human health but also the economic stability worldwide. The advancement in tissue engineering is contributing a lot in dealing with this immediate need of alleviating human health. Blood vessel diseases are considered as major cardiovascular health problems. Although blood vessel transplantation is the most convenient treatment, it has been delimited due to scarcity of donors and the patient's conditions. However, tissue-engineered blood vessels are promising alternatives as mode of treatment for blood vessel defects. The purpose of this paper is to show the importance of the advancement on biofabrication technology for treatment of soft tissue defects particularly for vascular tissues. This will also provide an overview and update on the current status of tissue reconstruction especially from autologous stem cells, scaffolds, and scaffold-free cellular transplantable constructs. The discussion of this paper will be focused on the historical view of cardiovascular tissue engineering and stem cell biology. The representative studies featured in this paper are limited within the last decade in order to trace the trend and evolution of techniques for blood vessel tissue engineering.
Collapse
|
56
|
Abstract
Receptors for basic (b) and acidic (a) fibroblast growth factor (FGF) are upregulated in activated smooth muscle cells. These cells, which proliferate in response to bFGF, can thus be killed by a conjugate of bFGF and the ribosome-inactivating enzyme, saporin (which, by itself, does not enter the cells). Quiescent smooth muscle cells and other cells that have few FGF receptors are not killed. In vivo, bFGF-saporin transiently inhibits smooth muscle cell proliferation and neointimal accumulation after balloon injury to the rat carotid artery. Delivery of saporin, diagnostic imaging agents, or antisense oligodeoxynucleotides might be made even more selective by linking these substances to antibodies against the extracellular domains of the putative FGF receptor isoform specific for activated smooth muscle cells.
Collapse
Affiliation(s)
- W Casscells
- Texas Heart Institute and University of Texas Medical School, Houston, TX 77225-0345 USA
| | | | | |
Collapse
|
57
|
Koskinas KC, Chatzizisis YS, Antoniadis AP, Giannoglou GD. Role of endothelial shear stress in stent restenosis and thrombosis: pathophysiologic mechanisms and implications for clinical translation. J Am Coll Cardiol 2012; 59:1337-49. [PMID: 22480478 DOI: 10.1016/j.jacc.2011.10.903] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 09/13/2011] [Accepted: 10/27/2011] [Indexed: 10/28/2022]
Abstract
Restenosis and thrombosis are potentially fatal complications of coronary stenting with a recognized multifactorial etiology. The effect of documented risk factors, however, cannot explain the preponderance of certain lesion types, stent designs, and implantation configurations for the development of these complications. Local hemodynamic factors, low endothelial shear stress (ESS) in particular, are long known to critically affect the natural history of atherosclerosis. Increasing evidence now suggests that ESS may also contribute to the development of restenosis and thrombosis upon stenting of atherosclerotic plaques, in conjunction with well-appreciated risk factors. In this review, we present in vivo and mechanistic evidence associating ESS with the localization and progression of neointimal hyperplasia and in-stent clotting. Clinical studies have associated stent design features with the risk of restenosis. Importantly, computational simulations extend these observations by directly linking specific stent geometry and positioning characteristics with the post-stenting hemodynamic milieu and with the stent's thrombogenicity and pro-restenotic potential, thereby indicating ways to clinical translation. An enhanced understanding of the pathophysiologic role of ESS in restenosis and thrombosis might dictate hemodynamically favorable stent designs and deployment configurations to reduce the potential for late lumen loss and thrombotic obstruction. Recent methodologies for in vivo ESS profiling at a clinical level might allow for early identification of patients at high risk for the development of restenosis or thrombosis and might thereby guide individualized, risk-tailored treatment strategies to prevent devastating complications of endovascular interventions.
Collapse
Affiliation(s)
- Konstantinos C Koskinas
- 1st Cardiology Department, AHEPA University Hospital, Aristole University Medical School, Thessaloniki, Greece
| | | | | | | |
Collapse
|
58
|
Antirestenotic mechanisms of everolimus on human coronary artery smooth muscle cells: inhibition of human coronary artery smooth muscle cell proliferation, but not migration. J Cardiovasc Pharmacol 2012; 59:165-74. [PMID: 21983747 DOI: 10.1097/fjc.0b013e31823a39c7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Everolimus, a pharmaceutical component of drug-eluting stents, inhibits coronary vessel restenosis, but the antirestenotic mechanisms of action remain unclear. Here, we describe the effects of everolimus on key contributors to vessel restenosis, smooth muscle cell proliferation, and migration. In a dose-dependent fashion, everolimus reduced human coronary artery smooth muscle cell (HCASMC) proliferation without toxicity in a bimodal fashion, with accentuated potency occurring at 10 μM. Everolimus arrested the majority of HCASMCs in G1-phase, whereas it reduced the fraction of cells in S-phase at doses that inhibited DNA synthesis (bromodeoxyuridine incorporation). Consistent with this, Western blotting demonstrated that everolimus reduced activation and expression of G1-phase cell cycle progression factors, including p70S6K and cyclin D, respectively, decreased levels of proliferating cell nuclear antigen, and attenuated growth factor/serum-induced phosphorylation of the cell cycle phase transition intermediate, retinoblastoma protein. Everolimus did not, however, affect HCASMC migration. These observations suggest that everolimus acts as an antiproliferative, but not antimigratory, compound to account for at least some of the clinical efficacy exhibited by this drug as an antirestenotic agent. Moreover, everolimus-induced inhibition of the mammalian target of rapamycin complex 1 and regulation of cyclin-mediated cell cycle progression actions likely account for the antiproliferative effects of this compound on HCASMCs.
Collapse
|
59
|
Clever YP, Cremers B, Krauss B, Böhm M, Speck U, Laufs U, Scheller B. Paclitaxel and sirolimus differentially affect growth and motility of endothelial progenitor cells and coronary artery smooth muscle cells. EUROINTERVENTION 2012; 7 Suppl K:K32-42. [PMID: 22027725 DOI: 10.4244/eijv7ska6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIMS EPC and hCASMC play an important role in the pathogenesis of restenosis and stent thrombosis. Drug-coated balloon catheters exert a local, short-term application of antiproliferative agents. This study investigates the time-dependent influence on growth and motility of paclitaxel and sirolimus alone and combined with the coating additive iopromide on EPC and hCASMC. METHODS AND RESULTS Treatment of cultured human EPC and hCASMC with paclitaxel and sirolimus 1.5 and 15 µM for three seconds, three minutes and 24 hours, alone or combined with iopromide 0.197 M, resulted in a concentration- and time- dependent inhibition of proliferation and of migration. Paclitaxel and sirolimus increase apoptosis in either cell type. However, the effects of paclitaxel and sirolimus differed between the cell types: short-term exposure with paclitaxel leads to stronger inhibition of cell-density and apoptosis of hCASMC compared to EPC. In comparison to paclitaxel, short-term incubation with sirolimus showed a more effective inhibition of cell-density and migration as well as increased apoptosis in EPC in contrast to hCASMC. The effects of paclitaxel and sirolimus were increased in combination with iopromide. Interestingly, the antiproliferative effect of the paclitaxel-iopromide formulation on hCASMC was more potent compared to its effect on EPC. Endothelialisation in a porcine coronary stent model was similar with drug-coated balloons and uncoated controls, whereas it was delayed with drug-eluting stents. CONCLUSION After short-term application, paclitaxel and sirolimus show differential, cell-specific effects on EPC and hCASMC. Iopromide used as a coating agent intensifies these effects.
Collapse
Affiliation(s)
- Yvonne P Clever
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | | | | | | | | | | | | |
Collapse
|
60
|
Rocchiccioli S, Ucciferri N, Comelli L, Trivella MG, Citti L, Cecchettini A. Proteomics changes in adhesion molecules: a driving force for vascular smooth muscle cell phenotypic switch. MOLECULAR BIOSYSTEMS 2012; 8:1052-9. [DOI: 10.1039/c2mb05470a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
61
|
Wiedon A, Tölle M, Bastine J, Schuchardt M, Huang T, Jankowski V, Jankowski J, Zidek W, van der Giet M. Uridine adenosine tetraphosphate (Up4A) is a strong inductor of smooth muscle cell migration via activation of the P2Y2 receptor and cross-communication to the PDGF receptor. Biochem Biophys Res Commun 2011; 417:1035-40. [PMID: 22214933 DOI: 10.1016/j.bbrc.2011.12.088] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 12/19/2011] [Indexed: 11/28/2022]
Abstract
The recently discovered dinucleotide uridine adenosine tetraphosphate (Up(4)A) was found in human plasma and characterized as endothelium-derived vasoconstrictive factor (EDCF). A further study revealed a positive correlation between Up(4)A and vascular smooth muscle cell (VSMC) proliferation. Due to the dominant role of migration in the formation of atherosclerotic lesions our aim was to investigate the migration stimulating potential of Up(4)A. Indeed, we found a strong chemoattractant effect of Up(4)A on VSMC by using a modified Boyden chamber. This migration dramatically depends on osteopontin secretion (OPN) revealed by the reduction of the migration signal down to 23% during simultaneous incubation with an OPN-blocking antibody. Due to inhibitory patterns using specific and unspecific purinoreceptor inhibitors, Up(4)A mediates it's migratory signal mainly via the P2Y(2). The signaling behind the receptor was investigated with luminex technique and revealed an activation of the extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathway. By use of the specific PDGF receptor (PDGFR) inhibitor AG1296 and siRNA technique against PDGFR-β we found a strongly reduced migration signal after Up(4)A stimulation in the PDGFR-β knockdown cells compared to control cells. In this study, we present substantiate data that Up(4)A exhibits migration stimulating potential probably involving the signaling cascade of MEK1 and ERK1/2 as well as the matrix protein OPN. We further suggest that the initiation of the migration process occurs predominant through direct activation of the P2Y(2) by Up(4)A and via transactivation of the PDGFR.
Collapse
Affiliation(s)
- Annette Wiedon
- Charité - Universitätsmedizin Berlin, CharitéCentrum, Department of Nephrology, Campus Benjamin Franklin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Esteban V, Méndez-Barbero N, Jiménez-Borreguero LJ, Roqué M, Novensá L, García-Redondo AB, Salaices M, Vila L, Arbonés ML, Campanero MR, Redondo JM. Regulator of calcineurin 1 mediates pathological vascular wall remodeling. ACTA ACUST UNITED AC 2011; 208:2125-39. [PMID: 21930771 PMCID: PMC3182048 DOI: 10.1084/jem.20110503] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Angiotensin-II–driven calcineurin activation and regulator of calcineurin-1 (Rcan-1) expression is required for pathological vascular remodeling in mice. Artery wall remodeling, a major feature of diseases such as hypertension, restenosis, atherosclerosis, and aneurysm, involves changes in the tunica media mass that reduce or increase the vessel lumen. The identification of molecules involved in vessel remodeling could aid the development of improved treatments for these pathologies. Angiotensin II (AngII) is a key effector of aortic wall remodeling that contributes to aneurysm formation and restenosis through incompletely defined signaling pathways. We show that AngII induces vascular smooth muscle cell (VSMC) migration and vessel remodeling in mouse models of restenosis and aneurysm. These effects were prevented by pharmacological inhibition of calcineurin (CN) or lentiviral delivery of CN-inhibitory peptides. Whole-genome analysis revealed >1,500 AngII-regulated genes in VSMCs, with just 11 of them requiring CN activation. Of these, the most sensitive to CN activation was regulator of CN 1 (Rcan1). Rcan1 was strongly activated by AngII in vitro and in vivo and was required for AngII-induced VSMC migration. Remarkably, Rcan1−/− mice were resistant to AngII-induced aneurysm and restenosis. Our results indicate that aneurysm formation and restenosis share mechanistic elements and identify Rcan1 as a potential therapeutic target for prevention of aneurysm and restenosis progression.
Collapse
Affiliation(s)
- Vanesa Esteban
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Walker T, Müller I, Raabe C, Nohe B, Zanke C, Ziemer G, Wendel HP. Effective silencing of adhesion molecules on venous endothelial cells for protection of venous bypass grafts. Eur J Cardiothorac Surg 2011; 40:1241-7. [PMID: 21429759 DOI: 10.1016/j.ejcts.2011.01.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The patency of venous conduits after aortocoronary bypass grafting is still not satisfactory and needs to be improved. Atherosclerotic alterations mediated by adhesion molecules triggering the transmigration of leukocytes are regarded as one of the major causes for venous graft failure. This study deals with short interfering RNA (siRNA)-mediated silencing of adhesion molecule expression on venous endothelial cells, which could lead to a new therapeutic strategy, resulting in improved patency rates by inhibiting early graft alterations. METHODS Primary human venous endothelial cells (HVECs) were cultured in a newly developed perfusion model and subsequently transfected with specific siRNAs targeting three different adhesion molecules (the E-selectin (ESELE), the intercellular adhesion molecule 1 (ICAM-1), and the vascular adhesion molecule (VCAM-1)), followed by stimulation with tumor necrosis factor-alpha (TNF-α). Isolated leukocytes were perfused under physiological shear stress conditions, and their attachment to HVEC after single and triple transfection was quantified. RESULTS siRNA transfection effectively knocks down adhesion molecule expression on venous endothelial cells, which subsequently reduces leukocyte attachment. Leukocyte adhesion to activated HVEC was significantly reduced after transfection by specific siRNAs in each case compared to the controls (p<0.05). Transfection with a mixture of all three siRNA sequences improved this effect even more (p<0.05). CONCLUSION For the first time, a functional protection of HUEC in a model simulating physiologic vascular conditions by using nonviral transfection of the cells in a setup with high relevance for clinical applicability was demonstrated. Therefore, siRNA transfection of bypass material may develop into a new therapeutic option to improve the quality of venous graft material in the future.
Collapse
Affiliation(s)
- Tobias Walker
- Department of Thoracic, Cardiac, and Vascular Surgery, Tuebingen University Hospital, Hoppe Seyler Strasse 3, 72076 Tuebingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
64
|
|
65
|
|
66
|
Sarkar J, Gou D, Turaka P, Viktorova E, Ramchandran R, Raj JU. MicroRNA-21 plays a role in hypoxia-mediated pulmonary artery smooth muscle cell proliferation and migration. Am J Physiol Lung Cell Mol Physiol 2010; 299:L861-71. [PMID: 20693317 PMCID: PMC3006273 DOI: 10.1152/ajplung.00201.2010] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 08/03/2010] [Indexed: 12/18/2022] Open
Abstract
Hypoxia stimulates pulmonary artery smooth muscle cell (PASMC) proliferation. Recent studies have implicated an important role for microRNAs (miRNAs) in hypoxia-mediated responses in various cellular processes, including cell proliferation. In this study, we investigated the role of microRNA-21 (miR-21) in hypoxia-induced PASMC proliferation and migration. We first demonstrated that miR-21 expression increased by ∼3-fold in human PASMC after 6 h of hypoxia (3% O₂) and remained high (∼2-fold) after 24 h of hypoxia. Knockdown of miR-21 with anti-miR-21 inhibitors significantly reduced hypoxia-induced cell proliferation, whereas miR-21 overexpression in normoxia enhanced cell proliferation. We also found that miR-21 is essential for hypoxia-induced cell migration. Protein expression of miR-21 target genes, specifically programmed cell death protein 4 (PDCD4), Sprouty 2 (SPRY2), and peroxisome proliferator-activated receptor-α (PPARα), was decreased in hypoxia and in PASMC overexpressing miR-21 in normoxia and increased in hypoxic cells in which miR-21 was knocked down. In addition, PPARα 3'-untranslated region (UTR) luciferase-based reporter gene assays demonstrated that PPARα is a direct target of miR-21. Taken together, our findings indicate that miR-21 plays a significant role in hypoxia-induced pulmonary vascular smooth muscle cell proliferation and migration by regulating multiple gene targets.
Collapse
Affiliation(s)
- Joy Sarkar
- Dept. of Pediatrics, University of Illinois College of Medicine at Chicago, 60612, USA.
| | | | | | | | | | | |
Collapse
|
67
|
Kwak KW, Cho KS, Hahn OJ, Lee KH, Lee BY, Ko JJ, Chung KH. Biological effects of fucoidan isolated from Fucus vesiculosus on thrombosis and vascular cells. THE KOREAN JOURNAL OF HEMATOLOGY 2010; 45:51-7. [PMID: 21120163 PMCID: PMC2983006 DOI: 10.5045/kjh.2010.45.1.51] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 11/18/2009] [Accepted: 11/28/2009] [Indexed: 12/02/2022]
Abstract
Background Fucoidan is a highly sulfated glycosaminoglycan, which has a molecular structure similar to that of heparin. The antithrombotic effects of fucoidan in vitro have been widely reported, but its antithrombotic effects in vivo as well as its other biological properties in vitro have not been well investigated. Methods This study investigated the effects and mechanism of fucoidan from Fucus vesiculosus on thrombosis both in vitro and in vivo. A ferric chloride-induced mouse carotid artery thrombosis model was used to determine the antithrombotic effects of fucoidan in vivo. Additionally, changes in the levels of proinflammatory cytokines and chemokines were examined in vascular cells treated with fucoidan. Results In vivo studies employing a ferric chloride-induced mouse carotid artery thrombosis model indicated that fucoidan had a stronger antithrombotic activity than heparin. Further, vascular cells treated with fucoidan demonstrated a decrease in proinflammatory cytokine and chemokine production as well as inhibition of proliferation. Conclusion The major findings of this study showed that fucoidan has a stronger antithrombotic effect than heparin in vivo and that fucoidan has an inhibitory effect on proinflammatory cytokine production and proliferation of vascular cells.
Collapse
Affiliation(s)
- Kyu-Won Kwak
- Thrombosis and Vascular Biochemistry Lab., Department of Biochemistry, College of Medicine, CHA University, Seongnam, Korea
| | | | | | | | | | | | | |
Collapse
|
68
|
Karpurapu M, Wang D, Van Quyen D, Kim TK, Kundumani-Sridharan V, Pulusani S, Rao GN. Cyclin D1 is a bona fide target gene of NFATc1 and is sufficient in the mediation of injury-induced vascular wall remodeling. J Biol Chem 2009; 285:3510-23. [PMID: 19933579 DOI: 10.1074/jbc.m109.063727] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Platelet-derived growth factor BB induced cyclin D1 expression in a time- and nuclear factor of activated T cells (NFAT)-dependent manner in human aortic smooth muscle cells (HASMCs), and blockade of NFATs prevented HASMC DNA synthesis and their cell cycle progression from G(1) to S phase. Selective inhibition of NFATc1 by its small interfering RNA also blocked HASMC proliferation and migration. Characterization of the cyclin D1 promoter revealed the presence of several NFAT binding sites, and the site at nucleotide -1333 was found to be sufficient in mediating platelet-derived growth factor BB-induced cyclin D1 promoter-luciferase reporter gene activity. In addition to its role in cell cycle progression, cyclin D1 mediated HASMC migration in an NFATc1-dependent manner. Balloon injury-induced cyclin D1-CDK4 activity requires NFAT activation, and adenovirus-mediated transduction of cyclin D1 was found to be sufficient to overcome the blockade effect of NFATs by VIVIT on balloon injury-induced vascular wall remodeling events, including smooth muscle cell migration from the medial to luminal region, their proliferation in the intimal region, and neointima formation. Together, these results provide more mechanistic evidence for the role of NFATs, particularly NFATc1, in the regulation of HASMC proliferation and migration as well as vascular wall remodeling. NFATc1 could be a potential therapeutic target against the renarrowing of artery after angioplasty.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Shirotani M, Yui Y, Kawai C. Restenosis after Coronary Angioplasty: Pathogenesis of Neointimal Thickening Initiated by Endothelial Loss. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329309100951] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
70
|
Zhang X, Wang X, Keshav V, Wang X, Johanas J, Leisk G, Kaplan DL. Dynamic culture conditions to generate silk-based tissue-engineered vascular grafts. Biomaterials 2009; 30:3213-23. [PMID: 19232717 PMCID: PMC2732107 DOI: 10.1016/j.biomaterials.2009.02.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 02/02/2009] [Indexed: 11/26/2022]
Abstract
Tissue engineering is an alternative approach for the preparation of small-diameter (<6mm) vascular grafts due to the potential to control thrombosis, anastomotic cellular hyperplasia and matrix production. This control also requires the maintenance of graft patency in vivo, appropriate mechanical properties and the formation of a functional endothelium. As a first step in generating such tissue-engineered vascular grafts (TEVGs), our objective was to develop a tissue-engineered construct that mimicked the structure of blood vessels using tubular electrospun silk fibroin scaffolds (ESFSs) with suitable mechanical properties. Human coronary artery smooth muscle cells (HCASMCs) and human aortic endothelial cells (HAECs) were sequentially seeded onto the luminal surface of the tubular scaffolds and cultivated under physiological pulsatile flow. The results demonstrated that TEVGs under dynamic flow conditions had better outcome than static culture controls in terms of cell proliferation and alignment, ECM production and cell phenotype based on transcript and protein level assessments. The metabolic activity of HCASMCs present in the TEGs indicated the advantage of dynamic flow over static culture in effective nutrient and oxygen distribution to the cells. A matrigel coating as a basement membrane mimic for ECM significantly improved endothelium coverage and retention under physiological shear forces. The results demonstrate the successful integration of vascular cells into silk electrospun tubular scaffolds as a step toward the development of a TEVG similar to native vessels in terms of vascular cell outcomes and mechanical properties.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Xiuli Wang
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Vinny Keshav
- Department of Biology, Tufts University, 163 Packard Avenue, Medford, MA 02155, USA
| | - Xiaoqin Wang
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Jacqueline Johanas
- Department of Mechanical Engineering, Tufts University, 200 College Avenue, Medford, MA 02155
| | - Gary Leisk
- Department of Mechanical Engineering, Tufts University, 200 College Avenue, Medford, MA 02155
| | - David L Kaplan
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
71
|
Lord MS, Yu W, Cheng B, Simmons A, Poole-Warren L, Whitelock JM. The modulation of platelet and endothelial cell adhesion to vascular graft materials by perlecan. Biomaterials 2009; 30:4898-906. [PMID: 19540587 DOI: 10.1016/j.biomaterials.2009.05.063] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/25/2009] [Indexed: 10/20/2022]
Abstract
Controlled neo-endothelialisation is critical to the patency of small diameter vascular grafts. Endothelialisation and platelet adhesion to purified endothelial cell-derived perlecan, the major heparan sulfate (HS) proteoglycan in basement membranes, were investigated using in vivo and in vitro assays. Expanded polytetrafluoroethylene (ePTFE) vascular grafts were coated with perlecan and tested in an ovine carotid interposition model for a period of 6 weeks and assessed using light and scanning microscopy. Enhanced endothelial cell growth and reduced platelet adhesion were observed on the perlecan coated grafts when compared to uncoated controls implanted in the same sheep (n=5). Perlecan was also found to stimulate endothelial cell proliferation in vitro over a period of 6 days in the presence of plasma proteins and fibroblastic growth factor 2 (FGF-2), however in the absence of FGF-2 endothelial cell growth could not be maintained during this period. Perlecan was found to be anti-adhesive for platelets, however after removal of the HS chains attached to perlecan, platelet adhesion and aggregation were supported. These results suggest a role for HS chains of perlecan in improving graft patency by selectively promoting endothelial cell proliferation while modulating platelet adhesion.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, The University of New South Wales, Gate 11, Botany St, Randwick, Sydney, NSW 2052, Australia.
| | | | | | | | | | | |
Collapse
|
72
|
Zou Y, Qi Y, Roztocil E, Davies MG. Patterns of gelatinase activation induced by injury in the murine femoral artery. J Surg Res 2009; 154:135-42. [PMID: 19101695 PMCID: PMC2698815 DOI: 10.1016/j.jss.2008.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 04/30/2008] [Accepted: 05/21/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND Intimal hyperplasia remains the principal lesion in the development of restenosis after vessel wall injury. Modulation of the extracellular matrix by proteases is a pivotal component of the response to injury. The aim of this study is to characterize the changes in gelatinase (MMP-2/TIMP-2 and MMP-9/TIMP-1) systems in a murine model. METHODS The murine femoral wire injury model was used in which a microwire is passed through a branch of the femoral and used to denude the common femoral artery. Pluronic gel was used to apply a proteass inhibitor (GM6001) to the exterior of the vessels. Specimens were perfusion-fixed and sections were stained with hematoxylin and eosin and Movat's stain such that morphometry could be performed by using an ImagePro system. Additional specimens of femoral artery were also harvested and snap frozen for Western blotting and zymography to allow for the study of gelatinase expression and activation. Contralateral vessels were used as controls. RESULTS MMP-2 activity increased significantly at day 1, peaked again at day 7, and then showed a continual decline in activity to day 56. MMP-9 activity peaked early at day 3 and declined thereafter. Protein expression for both MMP-2 and MMP-9 increased significantly after injury and both were maximal at day 14. There was an initial decrease in TIMP-1 and TIMP-2 expression and activity after injury until day 5. Both expression and activation gradually increased thereafter to level out by day 21 and correlated well with the early increases in MMP-2 and MMP-9 activity and their subsequent decline. Local application of protease inhibitor (GM6001) within a pluronic gel decreased cell proliferation, and at 14 d there was a decrease in intimal hyperplasia. CONCLUSIONS These data demonstrate that femoral wire injury in the mouse is associated with a time-dependent increase in gelatinase activity. Cell proliferation is associated with increased MMP-2/MMP-9 activity and decreased TIMP-2/TIMP-1 activity, whereas migration is associated with increased in MMP-2 activity. Modulation of proteases and their inhibitors control the vessels' response to injury.
Collapse
Affiliation(s)
- Yiping Zou
- Vascular Biology and Therapeutics Program, Methodist DeBakey Heart and Vascular Center, Department of Cardiovascular Surgery, The Methodist Hospital, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
73
|
A nanoscale drug-entrapment strategy for hydrogel-based systems for the delivery of poorly soluble drugs. Biomaterials 2009; 30:2102-11. [DOI: 10.1016/j.biomaterials.2008.12.047] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2008] [Accepted: 12/19/2008] [Indexed: 11/20/2022]
|
74
|
Rochefort GY, Mondon C, Machet MC, Halimi JM, Eder V, Antier D. Intravascular ultrasound allows a hyperplasia diagnosis in vivo in rat as accurate as histomorphometry. ULTRASONICS 2009; 49:145-148. [PMID: 18947847 DOI: 10.1016/j.ultras.2008.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 08/21/2008] [Accepted: 08/26/2008] [Indexed: 05/27/2023]
Abstract
OBJECTIVE AND MOTIVATION Intravascular ultrasound (IVUS) allows in vivo invasive intra-luminal real-time examination of the arterial wall structure. In this study, we aimed to validate for the first time the in vivo IVUS performing as a diagnostic tool by comparison to the well-established histomorphometry approach, in the largely used rat model of carotid angioplasty model that mimics the angioplasty procedure in humans. METHODS Atherosclerotic lesions were allowed to develop during four weeks after balloon catheter inflation of the left carotid artery, whereas the intact right carotid artery was used as control. Four weeks after injury, a Boston Scientific 40MHz device to perform IVUS exams in vivo on both carotid arteries. Then, both carotid arteries were examined in vitro by histomorphometry and correlation between IVUS and histomorphometric parameters (plaque plus media cross-sectional areas [CSA] and eccentricity index) were researched. RESULTS After ANOVA analysis, comparative statistical analysis showed significant correlations between IVUS and histomorphometry when examining the intact right carotid artery (r=0.662 with p<0.003 for plaque plus media CSA; r=0.774 with p<0.002 for eccentricity index), but also when exploring the injured left carotid artery (r=0.805 with p<0.0001 for plaque plus media CSA; r=0.775 with p<0.002 for eccentricity index). CONCLUSIONS AND OUTCOME We report here for the first time the ability of IVUS to study therapeutic vascular effects in vivo in alive rats. This result is of major importance since it will allow this device to be used for restenosis drug testing in rat model of carotid angioplasty.
Collapse
Affiliation(s)
- Gaël Y Rochefort
- Laboratoire de Physiologie de la Paroi Artérielle (LABPART)-EA 3852-IFR135, Université François Rabelais, Tours Cedex 1, France.
| | | | | | | | | | | |
Collapse
|
75
|
Sphingosine-1-phosphate promotes lymphangiogenesis by stimulating S1P1/Gi/PLC/Ca2+ signaling pathways. Blood 2008; 112:1129-38. [PMID: 18541717 DOI: 10.1182/blood-2007-11-125203] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The lymphatic system plays pivotal roles in mediating tissue fluid homeostasis and immunity, and excessive lymphatic vessel formation is implicated in many pathological conditions, which include inflammation and tumor metastasis. However, the molecular mechanisms that regulate lymphatic vessel formation remain poorly characterized. Sphingosine-1-phosphate (S1P) is a potent bioactive lipid that is implicated in a variety of biologic processes such as inflammatory responses and angiogenesis. Here, we first report that S1P acts as a lymphangiogenic mediator. S1P induced migration, capillary-like tube formation, and intracellular Ca(2+) mobilization, but not proliferation, in human lymphatic endothelial cells (HLECs) in vitro. Moreover, a Matrigel plug assay demonstrated that S1P promoted the outgrowth of new lymphatic vessels in vivo. HLECs expressed S1P1 and S1P3, and both RNA interference-mediated down-regulation of S1P1 and an S1P1 antagonist significantly blocked S1P-mediated lymphangiogenesis. Furthermore, pertussis toxin, U73122, and BAPTA-AM efficiently blocked S1P-induced in vitro lymphangiogenesis and intracellular Ca(2+) mobilization of HLECs, indicating that S1P promotes lymphangiogenesis by stimulating S1P1/G(i)/phospholipase C/Ca(2+) signaling pathways. Our results suggest that S1P is the first lymphangiogenic bioactive lipid to be identified, and that S1P and its receptors might serve as new therapeutic targets against inflammatory diseases and lymphatic metastasis in tumors.
Collapse
|
76
|
Miyake T, Aoki M, Morishita R. Inhibition of anastomotic intimal hyperplasia using a chimeric decoy strategy against NFkappaB and E2F in a rabbit model. Cardiovasc Res 2008; 79:706-14. [PMID: 18515844 DOI: 10.1093/cvr/cvn139] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Neointimal formation remains a major limitation after arterial reconstruction. To overcome this problem, we focused on two important transcription factors, nuclear factor-kappaB (NFkappaB) and E2F. The purpose of this study was to determine the effects of simultaneous inhibition of these transcription factors on the formation of neointimal hyperplasia. METHODS AND RESULTS We employed chimeric decoy oligodeoxynucleotides (ODN) to inhibit both NFkappaB and E2F simultaneously, and examined the effects of chimeric decoy ODN on the proliferation and migration of cultured vascular cells and on the formation of neointimal hyperplasia using prosthetic graft placement in a rabbit hypercholesterolemia model. Our in vitro study demonstrated that transfection of chimeric decoy ODN inhibited platelet-derived growth factor (PDGF)-induced proliferation and migration of vascular smooth muscle cells, whereas endothelial cell proliferation was not inhibited. In an in vivo study, treatment with chimeric decoy ODN significantly inhibited proximal and distal anastomotic intimal hyperplasia, and accelerated re-endothelialization. alpha-Smooth muscle actin (alpha-SMA)-positive cell proliferation was inhibited at the anastomotic sites. Expression of PDGF-BB and PDGF receptor-beta was also suppressed by chimeric decoy ODN, resulting in a reduction of alpha-SMA-positive cell accumulation. In addition, chimeric decoy ODN treatment inhibited macrophage accumulation, which was accompanied by a reduction of vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1 gene expression. CONCLUSION The present study demonstrates the feasibility of chimeric decoy ODN treatment for preventing neointimal formation. This strategy might be useful to improve the clinical outcome after arterial reconstruction.
Collapse
Affiliation(s)
- Takashi Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
77
|
IWAI K, TAKAHASHI T, NAKAHASHI T, NOMURA K, ATSUMI M, ZENG L, ISHIGAMI K, KANDA T, YAMAGUCHI N, MORIMOTO S. Immobilization Stress Inhibits Intimal Fibromuscular Proliferation in the Process of Arterial Remodeling in Rats. Hypertens Res 2008; 31:977-86. [DOI: 10.1291/hypres.31.977] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
78
|
Human vascular smooth muscle cells and endothelial cells cocultured on polyglycolic acid (70/30) scaffold in tissue engineered vascular graft. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200708010-00007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
79
|
Yang M, Kahn AM. Insulin-inhibited and stimulated cultured vascular smooth muscle cell migration are related to divergent effects on protein phosphatase-2A and autonomous calcium/calmodulin-dependent protein kinase II. Atherosclerosis 2007; 196:227-233. [PMID: 17553505 DOI: 10.1016/j.atherosclerosis.2007.04.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 03/30/2007] [Accepted: 04/30/2007] [Indexed: 11/26/2022]
Abstract
Insulin, in the permissive presence of nitric oxide (NO), stimulates cGMP production which inhibits autonomous calcium/calmodulin-dependent protein kinase II (CaM kinase II) thereby inhibiting cultured vascular smooth muscle cell (VSMC) migration. In the presence of angiotensin II (Ang II), insulin stimulates NAD(P)H oxidase activity leading to increased VSMC migration. We wished to see whether insulin-stimulated cGMP stimulates protein phosphatase-2A (PP-2A) thereby inhibiting autonomous CaM kinase II and migration, and whether insulin, in the presence of Ang II, inhibits PP-2A and stimulates autonomous CaM kinase II in a NAD(P)H oxidase-dependent manner. One nanomole per litre of insulin in the presence of NO, or 50 micromol/L 8-Br-cGMP stimulated PP-2A activity by 46+/-6 and 247+/-23%, respectively (both P<0.05), and 8-Br-cGMP inhibited autonomous CaM kinase II activity by 67+/-9% (P<0.05) by a 10 nmol/L okadaic acid-sensitive pathway. Insulin plus Ang II inhibited PP-2A activity by 57+/-7% (P<0.05) and stimulated autonomous CaM kinase II activity by 120+/-14% (P<0.05), both by an apocynin-sensitive pathway. 8-Br-cGMP-inhibited VSMC migration was blocked by okadaic acid. It is concluded that insulin in the presence of NO stimulates cGMP which stimulates PP-2A activity causing inhibition of autonomous CaM kinase II activity and thus VSMC migration, and that insulin in the presence of Ang II inhibits PP-2A and stimulates autonomous CaM kinase II activities by a NAD(P)H oxidase-dependent mechanism which are associated with insulin-stimulated NAD(P)H oxidase-dependent migration.
Collapse
Affiliation(s)
- Ming Yang
- Division of Nephrology and Hypertension, Department of Medicine, Medical School, University of Texas Health Science Center, Houston, United States
| | - Andrew M Kahn
- Division of Nephrology and Hypertension, Department of Medicine, Medical School, University of Texas Health Science Center, Houston, United States.
| |
Collapse
|
80
|
Seye CI, Kong Q, Yu N, Gonzalez FA, Erb L, Weisman GA. P2 receptors in atherosclerosis and postangioplasty restenosis. Purinergic Signal 2007; 3:153-62. [PMID: 18404429 PMCID: PMC2096770 DOI: 10.1007/s11302-006-9047-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Accepted: 04/20/2006] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is an immunoinflammatory process that involves complex interactions between the vessel wall and blood components and is thought to be initiated by endothelial dysfunction [Ross (Nature 362:801–09, 1993); Fuster et al. (N Engl J Med 326:242–50, 1992); Davies and Woolf (Br Heart J 69:S3–S11, 1993)]. Extracellular nucleotides that are released from a variety of arterial and blood cells [Di Virgilio and Solini (Br J Pharmacol 135:831–42, 2002)] can bind to P2 receptors and modulate proliferation and migration of smooth muscle cells (SMC), which are known to be involved in intimal hyperplasia that accompanies atherosclerosis and postangioplasty restenosis [Lafont et al. (Circ Res 76:996–002, 1995)]. In addition, P2 receptors mediate many other functions including platelet aggregation, leukocyte adherence, and arterial vasomotricity. A direct pathological role of P2 receptors is reinforced by recent evidence showing that upregulation and activation of P2Y2 receptors in rabbit arteries mediates intimal hyperplasia [Seye et al. (Circulation 106:2720–726, 2002)]. In addition, upregulation of functional P2Y receptors also has been demonstrated in the basilar artery of the rat double-hemorrhage model [Carpenter et al. (Stroke 32:516–22, 2001)] and in coronary artery of diabetic dyslipidemic pigs [Hill et al. (J Vasc Res 38:432–43, 2001)]. It has been proposed that upregulation of P2Y receptors may be a potential diagnostic indicator for the early stages of atherosclerosis [Elmaleh et al. (Proc Natl Acad Sci U S A 95:691–95, 1998)]. Therefore, particular effort must be made to understand the consequences of nucleotide release from cells in the cardiovascular system and the subsequent effects of P2 nucleotide receptor activation in blood vessels, which may reveal novel therapeutic strategies for atherosclerosis and restenosis after angioplasty.
Collapse
Affiliation(s)
- Cheikh I Seye
- Department of Biochemistry, University of Missouri-Columbia, 540C Life Sciences Center, 1201 Rollins Road, Columbia, MO, 65211-7310, USA,
| | | | | | | | | | | |
Collapse
|
81
|
Nakagawa M, Ohno T, Maruyama R, Okubo M, Nagatsu A, Inoue M, Tanabe H, Takemura G, Minatoguchi S, Fujiwara H. Sesquiterpene Lactone Suppresses Vascular Smooth Muscle Cell Proliferation and Migration via Inhibition of Cell Cycle Progression. Biol Pharm Bull 2007; 30:1754-7. [PMID: 17827734 DOI: 10.1248/bpb.30.1754] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation and migration are involved in restenosis following percutaneous transluminal angioplasty (PTCA) as well as in the development and progression of atherosclerosis. We investigated the mechanisms underlying the inhibitory effect of the sesquiterpene 3-oxo-5alphaH,8betaH-eudesma-1,4(15),7(11)-trien-8,12-olide (1) on rat VSMC proliferation and migration. VSMCs were isolated from rat aorta, and then the effect of 1 on cell proliferation and migration was examined using methylthiazolyldiphenyl-tetrazolium bromide (MTT) and chemotaxis assays, respectively. Compound 1 had a potent inhibitory effect on fetal calf serum-induced VSMC proliferation. This effect correlated with reduced expression of cyclin D(1). In addition, 1 also inhibited platelet derived growth factor (PDGF)-induced migration of VSMCs. These results indicate that 1 is a promising candidate for additional biological evaluation to further define its potential as an inhibitory modulator of VSMC responses that contribute to restenosis following PTCA and to the development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Munehiro Nakagawa
- Second Department of Internal Medicine, Gifu University School of Medicine, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Miller LW. Heart Transplantation: Indications, Outcome, and Long-Term Complications. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_67] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
83
|
Xu H, Shi D, Chen K. Inhibition of vascular remodelling in a porcine coronary injury model by herbal extract XS0601. Chin Med 2006; 1:2. [PMID: 17302965 PMCID: PMC1761146 DOI: 10.1186/1749-8546-1-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Accepted: 11/23/2006] [Indexed: 11/17/2022] Open
Abstract
Background Arterial remodelling is a major pathologic change of restenosis after percutaneous coronary intervention (PCI). Our previous studies showed that XS0601 (consisting of Chuangxingol and paeoniflorin) had some effects on the prevention of restenosis after PCI. Therefore, the purpose of this study was to examine whether and how its mechanism was related to the regulation of the arterial remodelling after endothelial injury by balloon dilation. Methods Twenty Chinese mini-pigs were randomized into four groups: control, probucol, low-dose XS0601 and high-dose XS0601 group before oversized balloon injury of the left anterior descending coronary arteries. Starting from two days before balloon injury, the mini-pigs in the treated group were administered with probucol (2 g/day) and XS0601 (0.02 g/kg/day for low dose; 0.04 g/kg/day for high dose) for four weeks after balloon injury. The animals receiving balloon injury alone were used as control. Morphometric and angiographic analysis of the injured arteries were performed. Results The contribution of intimal hyperplasia and arterial remodelling to angiographic late lumen loss was 41% and 59% respectively. XS0601 markedly inhibited proliferation of smooth muscle cells (SMCs) and transformation of SMCs from contractile to synthetic phenotype in neointima, inhibited hyperplasia-related indices of morphometric analysis and reduce late angiographic lumen loss. The reduction of the late angiographic lumen loss resulting from vascular remodelling was greater after XS0601 treatment. Conclusion Both intimal hyperplasia and vascular remodelling are attributed to late lumen loss in this porcine coronary injury model. XS0601 markedly reduced angiographic late lumen loss resulting from intimal hyperplasia, vascular remodelling and XS0601 may be a potential agent to prevent restenosis after PCI.
Collapse
Affiliation(s)
- Hao Xu
- National Integrative Medicine Centre for Cardiovascular Diseases, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
| | - Dazhuo Shi
- Division of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medicial Sciences, 1 Xiyuan Caochang, Haidian District, Beijing 100091, China
| | - Keji Chen
- National Integrative Medicine Centre for Cardiovascular Diseases, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
- Division of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medicial Sciences, 1 Xiyuan Caochang, Haidian District, Beijing 100091, China
| |
Collapse
|
84
|
Schrepfer S, Deuse T, Sultan KR, Haddad M, Böger R, Münzel T, Schäfer H, Pelletier MP, Robbins RC, Reichenspurner H. Inhibition of Restenosis Development after Mechanical Injury: A New Field of Application for Malononitrilamides? Cardiology 2006; 108:128-37. [PMID: 17028423 DOI: 10.1159/000096037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Accepted: 07/06/2006] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate the efficacy of the malononitrilamide FK778 to prevent vascular smooth muscle cell (SMC) migration/proliferation, and vascular fibrosis, the key events in restenosis development using in vivo and in vitro studies. BACKGROUND Since the high rate of restenosis after percutaneous transluminal coronary angioplasty limited its long-term success, the implementation of locally delivered antiproliferative/immunosuppressive agents became advantageous. METHODS Rats underwent balloon denudation of the abdominal aorta and received sirolimus, tacrolimus, or FK778 for 28 days in varying doses. Aortas were harvested for histologic evaluation, profibrotic gene expression, and organ chamber studies. Antifibrotic, antiproliferative and antimigratory effects of the immunosuppressants were further evaluated in vitro. RESULTS Histology of untreated animals revealed marked intimal hyperplasia with moderate luminal obliteration. Neointima formation was dose-dependently attenuated by all three agents with FK778 and sirolimus being most efficacious. Organ chamber relaxation studies showed a leftward shift of the nitroglycerin and the acetylcholine dose-responses in all treatment groups, indicating diminished endothelial dysfunction. In vivo, only FK778 treatment revealed a significant downregulation of the TGF-beta/vasorin system which could be explained by upregulation of the TGF-beta-inhibitory mediator SMAD7. In vitro, FK778 showed most potent antiproliferative and antimigratory effects on SMC compared with sirolimus and tacrolimus. Only the antiproliferative effect of FK778 was due to pyrimidine synthesis blockade and could be reversed by uridine supplementation. CONCLUSIONS The malononitrilamide FK778 proved highly efficacious against restenosis development by targeting two major components of intimal hyperplasia: SMC proliferation/migration and vascular fibrosis. Thus, the introduction of malononitrilamide-loaded stents may be a promising effort for future strategies.
Collapse
MESH Headings
- Alkynes/pharmacokinetics
- Alkynes/pharmacology
- Alkynes/therapeutic use
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Carrier Proteins/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Endothelium, Vascular/drug effects
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/prevention & control
- Hyperplasia/pathology
- Hyperplasia/prevention & control
- Immunosuppressive Agents/pharmacokinetics
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Isoxazoles/pharmacokinetics
- Isoxazoles/pharmacology
- Isoxazoles/therapeutic use
- Male
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Nitriles/pharmacokinetics
- Nitriles/pharmacology
- Nitriles/therapeutic use
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred Lew
- Sirolimus/therapeutic use
- Smad7 Protein/metabolism
- Tacrolimus/therapeutic use
- Transforming Growth Factor beta1/metabolism
- Tunica Intima/drug effects
- Tunica Intima/pathology
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Sonja Schrepfer
- Department of Cardiovascular Surgery, University Heart Center Hamburg, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Steele C, Fioretto ET, Sasahara THC, Guidi WL, de Lima AR, Ribeiro AACM, Loesch A. On the atrophy of the internal carotid artery in capybara. Cell Tissue Res 2006; 326:737-48. [PMID: 16826374 DOI: 10.1007/s00441-006-0218-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Accepted: 04/11/2006] [Indexed: 01/07/2023]
Abstract
Capybara might be a useful model for studying changes in cerebral circulation as the natural atrophy of the internal carotid artery (ICA) occurs in this animal at maturation. In this study, confocal and electron microscopy combined with immunohistochemical techniques were applied in order to reveal the changes in morphology and innervation to the proximal part of ICA in young (6-month-old) and mature (12-month-old) capybaras. Some features of the basilar artery (BA) were also revealed. The ICA of young animals degenerated to a ligamentous cord in mature animals. Immunolabelling positive for pan-neuronal marker protein gene product 9.5 but negative for tyrosine hydroxylase was observed in the proximal part of ICA at both ages examined. Axon varicosities positive for synaptophysin were present in the adventitia of ICA of young animals but were absent in the ligamentous cord of mature animals. In the ICA of young animals, adventitial connective tissue invaded the media suggesting that the process of regression of this artery began within the first 6 months of life. An increase in size of the BA was found in mature animals indicating increased blood flow in the vertebro-basilar system, possibly making capybara susceptible to cerebrovascular pathology (e.g. stroke). Capybara may therefore provide a natural model for studying adaptive responses to ICA regression/occlusion.
Collapse
Affiliation(s)
- Caroline Steele
- Department of Anatomy and Developmental Biology, Hampstead Campus, Royal Free and University College Medical School, University College London, London NW3 2PF, UK
| | | | | | | | | | | | | |
Collapse
|
86
|
Seye CI, Kong Q, Yu N, Gonzalez FA, Erb L, Weisman GA. P2 receptors in atherosclerosis and postangioplasty restenosis. Purinergic Signal 2006; 2:471-80. [PMID: 18404484 PMCID: PMC2096650 DOI: 10.1007/s11302-006-9015-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Accepted: 05/08/2006] [Indexed: 02/14/2023] Open
Abstract
Atherosclerosis is an immunoinflammatory process that involves complex interactions between the vessel wall and blood components and is thought to be initiated by endothelial dysfunction [1-3]. Extracellular nucleotides that are released from a variety of arterial and blood cells [4] can bind to P2 receptors and modulate proliferation and migration of smooth muscle cells (SMC), which is known to be involved in intimal hyperplasia that accompanies atherosclerosis and postangioplasty restenosis [5]. In addition, P2 receptors mediate many other functions, including platelet aggregation, leukocyte adherence, and arterial vasomotoricity. A direct pathological role of P2 receptors is reinforced by recent evidence showing that up-regulation and activation of P2Y(2) receptors in rabbit arteries mediates intimal hyperplasia [6]. In addition, up-regulation of functional P2Y receptors also has been demonstrated in the basilar artery of the rat double-hemorrhage model [7] and in coronary arteries of diabetic dyslipidemic pigs [8]. It has been proposed that up-regulation of P2Y receptors may be a potential diagnostic indicator for the early stages of atherosclerosis [9]. Therefore, particular effort must be made to understand the consequences of nucleotide release from cells in the cardiovascular system and the subsequent effects of P2 nucleotide receptor activation in blood vessels, which may reveal novel therapeutic strategies for atherosclerosis and restenosis after angioplasty.
Collapse
Affiliation(s)
- Cheikh I Seye
- Department of Biochemistry, 540C Life Sciences Center, University of Missouri-Columbia, 1201 Rollins Road, Columbia, MO, 65211-7310, USA,
| | | | | | | | | | | |
Collapse
|
87
|
Srivastava S, Ramana KV, Tammali R, Srivastava SK, Bhatnagar A. Contribution of aldose reductase to diabetic hyperproliferation of vascular smooth muscle cells. Diabetes 2006; 55:901-10. [PMID: 16567509 PMCID: PMC3463958 DOI: 10.2337/diabetes.55.04.06.db05-0932] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The objective of this study was to determine whether the polyol pathway enzyme aldose reductase mediates diabetes abnormalities in vascular smooth muscle cell (SMC) growth. Aldose reductase inhibitors (tolrestat or sorbinil) or antisense aldose reductase mRNA prevented hyperproliferation of cultured rat aortic SMCs induced by high glucose. Cell cycle progression in the presence of high glucose was blocked by tolrestat, which induced a G0-G1 phase growth arrest. In situ, diabetes increased SMC growth and intimal hyperplasia in balloon-injured carotid arteries of streptozotocin-treated rats, when examined 7 or 14 days after injury. Treatment with tolrestat (15 mg x kg(-1) x day(-1)) diminished intimal hyperplasia and decreased SMC content of the lesion by 25%. Although tolrestat treatment increased immunoreactivity of the lesion with antibodies raised against protein adducts of the lipid peroxidation product 4-hydroxy trans-2-nonenal, no compensatory increase in lesion fibrosis was observed. Collectively, these results suggest that inhibition of aldose reductase prevents glucose-induced stimulation of SMC growth in culture and in situ. Even though inhibition of aldose reductase increases vascular oxidative stress, this approach may be useful in preventing abnormal SMC growth in vessels of diabetic patients.
Collapse
Affiliation(s)
- Sanjay Srivastava
- Division of Cardiology, Department of Medicine, Delia Baxter Building, 580 S. Preston St., Room 421B, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|
88
|
Perivenous application of fibrin glue prevents the early injury of jugular vein graft to arterial circulation in rabbits. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200602020-00006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
89
|
Lukasik SM, Cierpicki T, Borloz M, Grembecka J, Everett A, Bushweller JH. High resolution structure of the HDGF PWWP domain: a potential DNA binding domain. Protein Sci 2005; 15:314-23. [PMID: 16384999 PMCID: PMC2242466 DOI: 10.1110/ps.051751706] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Hepatoma Derived Growth Factor (HDGF) is an endogenous nuclear-targeted mitogen that is linked with human disease. HDGF is a member of the weakly conserved PWWP domain family. This 70-amino acid motif, originally identified from the WHSC1 gene, has been found in more than 60 eukaryotic proteins. In addition to the PWWP domain, many proteins in this class contain known chromatin remodeling domains, suggesting a role for HDGF in chromatin remodeling. We have determined the NMR structure of the HDGF PWWP domain to high resolution using a combination of NOEs, J-couplings, and dipolar couplings. Comparison of this structure to a previously determined structure of the HDGF PWWP domain shows a significant difference in the C-terminal region. Comparison to structures of other PWWP domains shows a high degree of similarity to the PWWP domain structures from Dnmt3b and mHRP. The results of selected and amplified binding assay and NMR titrations with DNA suggest that the HDGF PWWP domain may function as a nonspecific DNA-binding domain. Based on the NMR titrations, we propose a model of the interaction of the PWWP domain with DNA.
Collapse
Affiliation(s)
- Stephen M Lukasik
- Department of Chemistry, University of Virginia, Charlottesville, VA 22906, USA
| | | | | | | | | | | |
Collapse
|
90
|
Vara DS, Salacinski HJ, Kannan RY, Bordenave L, Hamilton G, Seifalian AM. Cardiovascular tissue engineering: state of the art. ACTA ACUST UNITED AC 2005; 53:599-612. [PMID: 16364812 DOI: 10.1016/j.patbio.2004.12.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Accepted: 12/03/2004] [Indexed: 11/18/2022]
Abstract
In patients requiring coronary or peripheral vascular bypass procedures, autogenous arterial or vein grafts remain as the conduit of choice even in the case of redo patients. It is in this class of redo patients that often natural tissue of suitable quality becomes unavailable; so that prosthetic material is then used. Prosthetic grafts are liable to fail due to graft occlusion caused by surface thrombogenicity and lack of elasticity. To prevent this, seeding of the graft lumen with endothelial cells has been undertaken and recent clinical studies have evidenced patency rates approaching reasonable vein grafts. Recent advances have also looked at developing a completely artificial biological graft engineered from the patient's cells with surface and viscoelastic properties similar to autogenous vessels. This review encompasses both endothelialisation of grafts and the construction of biological cardiovascular conduits.
Collapse
Affiliation(s)
- Dina S Vara
- Biomaterial and Tissue Engineering Centre (BTEC), University Department of Surgery, Royal Free and University College Medical School, University College London, Rowland Hill Street, London NW3 2PF, UK
| | | | | | | | | | | |
Collapse
|
91
|
Amabile PG, Wang DS, Kao EY, Lee J, Elkins CJ, Yuksel E, Hilfiker PR, Waugh JM, Dake MD. Directed Migration of Smooth Muscle Cells to Engineer Plaque-Resistant Vein Grafts. J Endovasc Ther 2005; 12:667-75. [PMID: 16363896 DOI: 10.1583/04-1268ra.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE To test the hypothesis that controlled perivascular release of tissue plasminogen activator (tPA) can generate cleaved extracellular matrix (ECM) chemotactic gradients to guide the migration of vascular smooth muscle cells (SMCs) away from the lumen, thereby limiting neointima formation. METHODS This hypothesis was tested in rabbit models in which the perivascular surface of vein bypass grafts was treated with microspheres releasing tPA (MS-tPA), microspheres containing no drug (MS-blank), or phosphate buffered saline (PBS). Vein graft segments harvested after 7 days were then evaluated for elastin content, proliferating SMCs, intima-to-media (I/M) ratio, and inflammation; late impact on neointima formation was also examined. RESULTS The 7-day results demonstrated cleaved elastin gradients and proliferating SMCs that assumed a more peripheral distribution in the MS-tPA group than MS-blank and PBS controls (p<0.05). At 28 days, vein grafts treated with MS-tPA showed a mean I/M ratio (0.35+/-0.04) that was 63.5% lower than PBS controls (0.96+/-0.07, p<0.005) and 43.5% lower than MS-blank specimens (0.62+/-0.08, p<0.05). CONCLUSIONS Perivascular release of tPA modifies ECM gradients, directionally guides SMC migration away from the lumen, and limits neointima formation.
Collapse
Affiliation(s)
- Philippe G Amabile
- Department of Cardiovascular and Interventional Radiology, Stanford University School of Medicine, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Parry TJ, Brosius R, Thyagarajan R, Carter D, Argentieri D, Falotico R, Siekierka J. Drug-eluting stents: Sirolimus and paclitaxel differentially affect cultured cells and injured arteries. Eur J Pharmacol 2005; 524:19-29. [PMID: 16271360 DOI: 10.1016/j.ejphar.2005.09.042] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 09/01/2005] [Indexed: 12/13/2022]
Abstract
Sirolimus and paclitaxel eluted from stents inhibit cell proliferation and other cellular processes by dramatically different mechanisms. In this study, the effects of sirolimus and paclitaxel on cultured human coronary artery smooth muscle and endothelial cell function or cell cycle changes in balloon-injured arteries were directly compared. Both sirolimus and paclitaxel inhibited smooth muscle and endothelial cell proliferation. However, only paclitaxel inhibited smooth muscle and endothelial cell migration at low (nM) concentrations. Sirolimus arrested smooth muscle and endothelial cells in the G0/G1 phase of the cell cycle without inducing apoptosis while paclitaxel produced apoptosis in both cell types at low nanomolar concentrations. Although both agents blocked neointimal formation, sirolimus applied locally to injured rat carotid arteries increased the percentage of cycling vascular cells in G0/G1 without inducing apoptosis while paclitaxel increased the percentage of cycling cells in S and G2/M phases while inducing apoptosis. These results suggest that sirolimus reduces neointimal hyperplasia through a cytostatic mechanism while paclitaxel produces apoptotic cell death.
Collapse
Affiliation(s)
- Tom J Parry
- Cordis Corporation, a Johnson and Johnson Company, Warren, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Williams C, Wick TM. Endothelial cell-smooth muscle cell co-culture in a perfusion bioreactor system. Ann Biomed Eng 2005; 33:920-8. [PMID: 16060532 DOI: 10.1007/s10439-005-3238-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Vascular endothelial cells (EC) are exposed to a complex biomechanical environment in vivo and are responsible for relaying important messages to the underlying tissue. EC and smooth muscle cells (SMC) communicate to regulate vascular development and function. In this work, a vascular perfusion bioreactor is used to grow tubular constructs seeded with EC and SMC under pulsatile shear stress in long-term co-culture to study the effects of EC on SMC function. SMC seeded into porous poly(glycolic acid) tubular scaffolds are cultured in the bioreactor for 25 days. Constructs are seeded with EC on day 10 or day 23 creating 2-day (short-term) or 15-day (long-term) EC and SMC co-cultures. Long-term EC-SMC co-culture significantly increases cell proliferation and downregulates collagen and proteoglycan deposition compared to short-term co-culture. After 25 days of culture, 15-day co-culture constructs have a more uniform cell distribution across the construct thickness and SMC express a more contractile phenotype compared to 2-day co-culture constructs. These data demonstrate strong interactions between SMC and EC in the bioreactor under physiologically relevant conditions. Thus, the vascular construct perfusion bioreactor is an important tool to investigate cell-cell and cell-extracellular matrix interactions in vascular cell biology and tissue engineering.
Collapse
Affiliation(s)
- Chrysanthi Williams
- School of Chemical & Biomolecular Engineering, Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | | |
Collapse
|
94
|
Lin J, Friesen MT, Bocangel P, Cheung D, Rawszer K, Wigle JT. Characterization of Mesenchyme Homeobox 2 (MEOX2) transcription factor binding to RING finger protein 10. Mol Cell Biochem 2005; 275:75-84. [PMID: 16335786 DOI: 10.1007/s11010-005-0823-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The molecular mechanisms by which Mesenchyme Homeobox 2 (Meox2) regulates the proliferation, differentiation and migration of vascular smooth muscle cells and cardiomyocytes are not known. The discovery of MEOX2 binding proteins will aid in understanding how MEOX2 functions as a regulator of these key cellular processes. To identify MEOX2 binding proteins, a yeast two-hybrid screen of a human heart cDNA library was performed using a deleted MEOX2 bait protein that does not contain the histidine/glutamine rich region (MEOX2deltaHQ). Eleven putative interacting proteins were identified including RING finger protein 10 (RNF10). In vitro pull-down assays and co-immunoprecipitation studies in mammalian cells further supported the yeast data demonstrating RNF10 bound to MEOX2. The minimal RNF10 binding region of MEOX2 was determined to be a central region between the histidine/glutamine rich domain and the homeodomain (amino acids 101-185). The amino terminal region of RNF10, containing the RING finger domain, was not essential for the binding to MEOX2. Our results also demonstrated that MEOX2 activation of the p21WAF1 promoter was enhanced by RNF10 co-expression.
Collapse
Affiliation(s)
- Jijin Lin
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba
| | | | | | | | | | | |
Collapse
|
95
|
Wilson WRW, Schwalbe EC, Jones JL, Bell PRF, Thompson MM. Matrix metalloproteinase 8 (neutrophil collagenase) in the pathogenesis of abdominal aortic aneurysm. Br J Surg 2005; 92:828-33. [PMID: 15918165 DOI: 10.1002/bjs.4993] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Loss of elastin is the initiating event in abdominal aortic aneurysm (AAA) formation, whereas loss of collagen is required for continued expansion. The elastolytic matrix metalloproteinases (MMPs) 2 and 9 are well described, but the source of excessive collagenolysis remains undefined. The aim of this study was to determine the expression of MMP-8, a potent type I collagenase, in normal aorta and AAA.
Methods
Infrarenal aortic biopsies were taken from 40 AAA and ten age-matched normal aortas. The concentrations of MMP-8 protein and its inhibitors, tissue inhibitor of metalloproteinase (TIMP) 1 and TIMP-2, were quantified by enzyme-linked immunosorbent assay. Immunohistochemistry was used to localize MMP-8 expression.
Results
MMP-8 concentrations were significantly raised in AAA compared with normal aorta (active MMP-8: 4·5 versus 0·5 ng per mg protein, P < 0·001; total MMP-8: 16·6 versus 2·8 ng per mg protein, P < 0·001). Levels of TIMP-1 and TIMP-2 were significantly lower in AAA than in normal aortic samples (TIMP-1: 142·2 versus 302·8 ng per mg protein; P = 0·010; TIMP-2: 9·2 versus 33·1 ng per mg protein, P < 0·001). Immunohistochemistry localized MMP-8 to mesenchymal cells within the adventitia of the aortic wall.
Conclusion
The high concentration of MMP-8 in aortic aneurysms represents a potent pathway for collagen degradation, and hence aneurysm formation and expansion.
Collapse
Affiliation(s)
- W R W Wilson
- Department of Vascular Surgery, St George's Hospital Medical School, London, UK
| | | | | | | | | |
Collapse
|
96
|
Yang S, Huang XY. Ca2+ influx through L-type Ca2+ channels controls the trailing tail contraction in growth factor-induced fibroblast cell migration. J Biol Chem 2005; 280:27130-7. [PMID: 15911622 DOI: 10.1074/jbc.m501625200] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth factor-induced cell migration underlies various physiological and pathological processes. The mechanisms by which growth factors regulate cell migration are not completely understood. Although intracellular elevation of Ca2+ is known to be critical in cell migration, the source of this Ca2+ elevation and the mechanism by which Ca2+ modulates this process in fibroblast cells are not well defined. Here we show that increase of cellular Ca2+ through Ca2+ influx, rather than Ca2+ release from intracellular stores, is essential for growth factor-induced fibroblast cell migration. Voltage-gated L-type Ca2+ channels, previously known to exist in excitable cells such as neurons and muscle cells, are shown here to be present in fibroblasts as well. Furthermore, these channels are responsible for the Ca2+ influx. L-type Ca2+ channel inhibitors block growth factor-induced Ca2+ influx and fibroblast cell migration. One mechanism by which Ca2+ signals control cell migration is to regulate the contraction of the trailing edge of migrating fibroblasts; this process is controlled by the small GTPase Rho in fast migrating cells such as leukocytes. Downstream of Ca2+, both calmodulin and myosin light chain kinase, but not calcineurin, are involved leading to phosphorylation of the myosin light chain at the trailing end. Thus, trailing edge contraction is critically regulated by Ca2+ influx through L-type Ca2+ channels in growth factor-induced fibroblast cell migration.
Collapse
Affiliation(s)
- Shengyu Yang
- Department of Physiology, Cornell University Weill Medical College, New York, New York 10021, USA
| | | |
Collapse
|
97
|
Jang WG, Kim HS, Park KG, Park YB, Yoon KH, Han SW, Hur SH, Park KS, Lee IK. Analysis of proteome and transcriptome of tumor necrosis factor ? stimulated vascular smooth muscle cells with or without alpha lipoic acid. Proteomics 2004; 4:3383-93. [PMID: 15378733 DOI: 10.1002/pmic.200400972] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play an important role in the development and progression of atherosclerosis. Tumor necrosis factor alpha (TNFalpha), a cytokine secreted by VSMCs and macrophages in atherosclerotic lesions, regulates a variety of cellular functions of inflammatory cells and VSMCs by promoting cell growth and motility, which are critical for the initiation and progression of vascularlesions. Alpha lipoic acid (ALA), a well known antioxidant, acts as a pyruvate dehydrogenase cofactor in mitochondrial metabolism. Recently, we reported that ALA has many beneficial effects on vascular cells in atherosclerosis. The aim of the current study was to examine VSMCs, treated for 24 hours with TNFalpha (10 ng/mL) in the presence or absence of ALA (2 mM), for differential protein and genes expression using two-dimensional gel electrophoresis (2-DE) and DNA microarray analysis, respectively. Using 2-DE, we identified proteins whose expression changed by at least 2.5-fold after TNFalpha stimulation. Proteins up-regulated by TNFalpha that were subsequently down-regulated in the presence of ALA were identified by matrix-assisted laser desorption/ionization-time of flight mass spectrometry as plasminogen activator inhibitor-2, fetal liver LKB-interacting protein, osteoblast-specific factor 2, glucosidase II, cyclin-dependent kinase 3, endoplasmin precursor and glutathione synthetase. TNFalpha down-regulated proteins that were up-regulated in the presence of ALA were keratin 19, eukaryotic translation elongation factor and Rho GDP dissociation inhibitor alpha. Gene expression analysis using DNA microarray tools confirmed the up-regulation or down-regulation of some, but not all, of the proteins observed in ALA challenged, TNFalpha-treated cells. This data should provide valuable information about the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Won Gu Jang
- Department of Genetic Engineering, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Strategies that enhance the number of endothelial cells (ECs) in the vessel wall following injury may limit complications such as thrombosis, vasospasm, and neointimal formation through reconstitution of a luminal barrier and cellular secretion of paracrine factors. Proof of principle has been demonstrated by studies in which mature ECs, culture expanded from harvested vascular tissue, were seeded in the arterial wall following balloon injury. The recent identification of circulating cells capable of developing an endothelial phenotype, including progenitor cells, has raised the possibility of using blood-derived cells as therapeutic agents. This article reviews data suggesting that such cells confer vascular protective effects after injury, raising the potential for novel, autologous approaches to the treatment of vascular disease.
Collapse
Affiliation(s)
- Rajiv Gulati
- Department of Cardiovascular Medicine, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
99
|
Veinot JP, Prichett-Pejic W, Picard P, Parks W, Schwartz R, Seidah NG, Chretien M. Implications of proprotein Convertase 5 (PC5) in the arterial restenotic process in a porcine model. Cardiovasc Pathol 2004; 13:241-50. [PMID: 15358338 DOI: 10.1016/j.carpath.2004.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Revised: 04/16/2004] [Accepted: 05/10/2004] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Convertases (PCs), especially PC5, have been detected in various layers of atherosclerotic and injured arteries. We postulate that PCs could be important enzymes in vascular disease thus studied PC5 expression in a porcine balloon and stent coronary arterial vascular injury model. METHODS Immunohistochemistry and in situ hybridization of slides of porcine arteries from paraffin blocks were studied 1, 7, 14 and 28 days post injury. RESULTS Immunohistochemistry studies show expression of PC5 in control artery endothelial cells, weak medial smooth muscle cell (SMC) staining and strong staining in the small nerves of the adventitia. At 7, 14 and 28 days postinjury, there is strong positive PC5 staining of the neointimal cells and the adventitial vasa vasora and myofibroblasts. Colocalization immunohistochemistry confirms the smooth muscle staining properties of the myofibroblast-like cells in both these locations. Single-label immunohistochemistry studies show the same cells to stain strongly positive with TGF-B, PDGF, matrix metalloproteinase-2 (MMP-2) and MMP-9. CONCLUSION PC5 may be involved in the process of arterial injury via its effect on growth factors (GFs) and mediators. These preliminary observations suggest that the convertases, especially PC5, represent a target for future study in the process of arterial injury.
Collapse
Affiliation(s)
- John P Veinot
- Department of Laboratory Medicine, Division of Anatomical Pathology, Ottawa Hospital, 1053 Carling Avenue, Ottawa, Ontario, Canada K1Y 4E9.
| | | | | | | | | | | | | |
Collapse
|
100
|
Shimada K, Miyauchi K, Mokuno H, Watanabe Y, Iwama Y, Shigekiyo M, Matsumoto M, Okazaki S, Tanimoto K, Kurata T, Sato H, Daida H. Promoter polymorphism in the CD14 gene and concentration of soluble CD14 in patients with in-stent restenosis after elective coronary stenting. Int J Cardiol 2004; 94:87-92. [PMID: 14996480 DOI: 10.1016/j.ijcard.2003.05.007] [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] [Received: 05/31/2002] [Revised: 04/01/2003] [Accepted: 05/12/2003] [Indexed: 12/13/2022]
Abstract
BACKGROUND Activated monocytes/macrophages, neutrophils, endothelial cells and smooth muscle cells participate in the restenosis processes. Monocytes/macrophages and neutrophils are activated by lipopolysaccharide (LPS) via CD14. Endothelial cells and smooth muscle cells are also stimulated by soluble CD14 (sCD14)-LPS complexes. METHODS We tested the hypothesis that C(-260)-->T polymorphism of the CD14 gene and sCD14 might be predictors for in-stent restenosis. We analyzed 129 consecutive patients who underwent elective coronary stenting. The restenosis was defined as > or =50% diameter stenosis at follow-up angiography. RESULTS The prevalence of the T/T genotype and the concentration of sCD14 were significantly higher in the restenosis group than in the no-restenosis group. This CD14 polymorphism also affected the levels of sCD14, therefore, we divided the patients into four groups. The loss index was 24.8% in C/C or C/T and < or =50th percentile of sCD14, 35.9% in T/T and < or =50th percentile of sCD14, 44.2% in C/C or C/T and >50th percentile of sCD14, and 49.1% in T/T and >50th percentile of sCD14 (P=0.02). The restenosis rate was 10.0%, 26.7%, 26.2% and 50.0% in each group, respectively (P=0.003). In the multivariate analysis, T/T and >50th percentile of sCD14 was the independent predictor for in-stent restenosis. CONCLUSIONS This study showed that the T/T genotype with a high level of sCD14 is an independent predictor of in-stent restenosis. The activation of monocytes/macrophages, endothelial cells and smooth muscle cells mediated by CD14 and/or sCD14 may play an important role in the restenosis processes.
Collapse
Affiliation(s)
- Kazunori Shimada
- Department of Cardiology, Juntendo University School of Medicine, 2-1-1 Hongo Bunkyo, Tokyo 113-8421, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|