51
|
Gabbasov ZA, Agapov AA, Saburova OS, Komlev AE, Soboleva EL, Akchurin RS, Smirnov VN. Circulating stromal osteonectin-positive progenitor cells and stenotic coronary atherosclerosis. Can J Physiol Pharmacol 2007; 85:295-300. [PMID: 17612637 DOI: 10.1139/y07-001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The level of circulating stromal progenitor cells carrying osteonectin (ON), a marker of osteogenic differentiation, was evaluated by flow cytometry in blood of patients with coronary artery disease (CAD). Ninety-nine patients with CAD were included into the study. Coronary angiography of all patients showed critical stenosis of at least 2 coronary arteries or their major branches. The control groups included 8 patients without CAD and 19 healthy volunteers. In control patients, no lesions of the coronary bed were found by angiography. The absence of CAD in the volunteers was confirmed by bicycle stress test. The content of ON-positive cells in blood was examined in various populations of lymphocyte-like cells. It was found that the number of ON+ lymphocyte-like cells with CD41 positivity in blood of patients without coronary stenosis (0.27%+/-0.11%, mean+/-SD) did not differ significantly from corresponding value in healthy volunteers (0.26%+/-0.07%, p=0.94). In CAD patients, the percent of these ON+ cells was 1.01%+/-0.49% and was significantly higher than in blood of healthy volunteers (p<0.0001) and patients without CAD (p<0.0001). High content of ON+ lymphocyte-like cells with CD41 positivity in blood may serve as noninvasive marker of arterial atherosclerosis.
Collapse
Affiliation(s)
- Zufar A Gabbasov
- Laboratory of Stem Cells, Institute of Experimental Cardiology, Cardiology Research Center, 3rd Cherepkovskaya st, 15A, 121 552 Moscow, Russia.
| | | | | | | | | | | | | |
Collapse
|
52
|
Tanaka K, Sata M, Natori T, Kim-Kaneyama JR, Nose K, Shibanuma M, Hirata Y, Nagai R. Circulating progenitor cells contribute to neointimal formation in nonirradiated chimeric mice. FASEB J 2007; 22:428-36. [PMID: 17848623 DOI: 10.1096/fj.06-6884com] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Recent evidence suggests that bone marrow-derived cells may contribute to repair and lesion formation following vascular injury. In most studies, bone marrow-derived cells were tracked by transplanting exogenous cells into bone marrow that had been compromised by irradiation. It remains to be determined whether endogenous circulating progenitors actually contribute to arterial remodeling under physiological conditions. Here, we established a parabiotic model in which two mice were conjoined subcutaneously without any vascular anastomosis. When wild-type mice were joined with transgenic mice that expressed green fluorescent protein (GFP) in all tissues, GFP-positive cells were detected not only in the peripheral blood but also in the bone marrow of the wild-type mice. The femoral arteries of the wild-type mice were mechanically injured by insertion of a large wire. At 4 wk, there was neointima hyperplasia that mainly consisted of alpha-smooth muscle actin-positive cells. GFP-positive cells were readily detected in the neointima (14.8+/-4.5%) and media (31.1+/-8.8%) of the injured artery. Some GFP-positive cells expressed alpha-smooth muscle actin or an endothelial cell marker. These results indicate that circulating progenitors contribute to re-endothelialization and neointimal formation after mechanical vascular injury even in nonirradiated mice.
Collapse
Affiliation(s)
- Kimie Tanaka
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Ball SG, Shuttleworth CA, Kielty CM. Mesenchymal stem cells and neovascularization: role of platelet-derived growth factor receptors. J Cell Mol Med 2007; 11:1012-30. [PMID: 17979880 PMCID: PMC4401270 DOI: 10.1111/j.1582-4934.2007.00120.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 08/27/2007] [Indexed: 12/19/2022] Open
Abstract
There is now accumulating evidence that bone marrow-derived mesenchymal stem cells (MSCs) make an important contribution to postnatal vasculogenesis, especially during tissue ischaemia and tumour vascularization. Identifying mechanisms which regulate the role of MSCs in vasculogenesis is a key therapeutic objective, since while increased neovascularization can be advantageous during tissue ischaemia, it is deleterious during tumourigenesis. The potent angiogenic stimulant vascular endothelial growth factor (VEGF) is known to regulate MSC mobilization and recruitment to sites of neovascularization, as well as directing the differentiation of MSCs to a vascular cell fate. Despite the fact that MSCs did not express VEGF receptors, we have recently identified that VEGF-A can stimulate platelet-derived growth factor (PDGF) receptors, which regulates MSC migration and proliferation. This review focuses on the role of PDGF receptors in regulating the vascular cell fate of MSCs, with emphasis on the function of the novel VEGF-A/PDGF receptor signalling mechanism.
Collapse
Affiliation(s)
- Stephen G Ball
- UK Centre for Tissue Engineering
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | - C Adrian Shuttleworth
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | - Cay M Kielty
- UK Centre for Tissue Engineering
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
54
|
Qian H, Yang Y, Li J, Huang J, Dou K, Yang G. The role of vascular stem cells in atherogenesis and post-angioplasty restenosis. Ageing Res Rev 2007; 6:109-127. [PMID: 17324640 DOI: 10.1016/j.arr.2007.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 09/16/2006] [Accepted: 01/19/2007] [Indexed: 12/20/2022]
Abstract
It is well known that atherosclerosis prevails in elderly populations as ageing acts as a recognized risk factor for this disease. Although the pathogenic factors leading to atherosclerosis are highly heterogeneous, traditionally speaking, the causative risk factors include hyperlipidemia, hypertension, diabetes mellitus and smoking, which can damage to endothelial function, and subsequently promote lipid penetration and inflammatory cell infiltration. Damaged endothelial cells (ECs) may be replaced by neighboring cell division, while damaged smooth muscle cells (SMCs) may be replaced by medial SMCs emigrating into the intima during atherogenesis. However, this standpoint is challenged by recent findings that vascular progenitor/stem cells (VPCs) may contribute to atherogenesis and post-angioplasty restenosis. VPCs are a group of primitive cells that have the potential to produce mature, functional cells in the vascular wall. VPCs residing in bone marrow, vascular wall or circulating in the peripheral blood may be stimulated by a variety of pathogenic factors. These stem cells then participate in regeneration, repair and remodeling of the injured arterial wall. This new concept may bring about a great breakthrough in understanding the pathogenesis of atherosclerosis and develop novel therapeutic strategies for coronary heart disease. This article will mainly review the role of VPCs in atherogenesis, thus providing a novel understanding about the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Haiyan Qian
- Department of Cardiology, Fuwai Hospital and Cardiovascular Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, 167 BeiLiShi Road, Beijing 100037, PR China
| | | | | | | | | | | |
Collapse
|
55
|
Kähler CM, Wechselberger J, Hilbe W, Gschwendtner A, Colleselli D, Niederegger H, Boneberg EM, Spizzo G, Wendel A, Gunsilius E, Patsch JR, Hamacher J. Peripheral infusion of rat bone marrow derived endothelial progenitor cells leads to homing in acute lung injury. Respir Res 2007; 8:50. [PMID: 17620112 PMCID: PMC2000890 DOI: 10.1186/1465-9921-8-50] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Accepted: 07/09/2007] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Bone marrow-derived progenitors for both epithelial and endothelial cells have been observed in the lung. Besides mature endothelial cells (EC) that compose the adult vasculature, endothelial progenitor cells (EPC) are supposed to be released from the bone marrow into the peripheral blood after stimulation by distinct inflammatory injuries. Homing of ex vivo generated bone marrow-derived EPC into the injured lung has not been investigated so far. We therefore tested the hypothesis whether homing of EPC in damaged lung tissue occurs after intravenous administration. METHODS Ex vivo generated, characterized and cultivated rat bone marrow-derived EPC were investigated for proliferation and vasculogenic properties in vitro. EPC were tested for their homing in a left-sided rat lung transplant model mimicking a severe acute lung injury. EPC were transplanted into the host animal by peripheral administration into the femoral vein (10(6) cells). Rats were sacrificed 1, 4 or 9 days after lung transplantation and homing of EPC was evaluated by fluorescence microscopy. EPC were tested further for their involvement in vasculogenesis processes occurring in subcutaneously applied Matrigel in transplanted animals. RESULTS We demonstrate the integration of intravenously injected EPC into the tissue of the transplanted left lung suffering from acute lung injury. EPC were localized in vessel walls as well as in destructed lung tissue. Virtually no cells were found in the right lung or in other organs. However, few EPC were found in subcutaneous Matrigel in transplanted rats. CONCLUSION Transplanted EPC may play an important role in reestablishing the endothelial integrity in vessels after severe injury or at inflammatory sites and might further contribute to vascular repair or wound healing processes in severely damaged tissue. Therapeutic applications of EPC transplantation may ensue.
Collapse
Affiliation(s)
- Christian M Kähler
- Department of Internal Medicine, Division of General Internal Medicine, Pneumology Centre, Innsbruck Medical University, Austria
| | - Jutta Wechselberger
- Department of Internal Medicine, Division of General Internal Medicine, Pneumology Centre, Innsbruck Medical University, Austria
| | - Wolfgang Hilbe
- Department of Internal Medicine, Division of General Internal Medicine, Oncology Service, Innsbruck Medical University, Austria
| | | | - Daniela Colleselli
- Department of Internal Medicine, Division of General Internal Medicine, Pneumology Centre, Innsbruck Medical University, Austria
| | - Harald Niederegger
- Department of Experimental Pathology, Innsbruck Medical University, Austria
| | - Eva-Maria Boneberg
- Biotechnology Institute Thurgau, University of Konstanz, Tägerwilen, Switzerland
| | - Gilbert Spizzo
- Department of Internal Medicine, Division of Haematology and Oncology, Innsbruck Medical University, Austria
| | - Albrecht Wendel
- Biochemical Pharmacology, Faculty of Biology, University of Konstanz, Germany
| | - Eberhard Gunsilius
- Department of Internal Medicine, Division of Haematology and Oncology, Innsbruck Medical University, Austria
| | - Josef R Patsch
- Department of Internal Medicine, Division of General Internal Medicine, Pneumology Centre, Innsbruck Medical University, Austria
- Department of Internal Medicine, Division of General Internal Medicine, Oncology Service, Innsbruck Medical University, Austria
| | - Jürg Hamacher
- Biochemical Pharmacology, Faculty of Biology, University of Konstanz, Germany
- Pulmonary Division, Department of Internal Medicine, University Hospital of Homburg, University of Saarland, D-66421 Homburg, Germany
| |
Collapse
|
56
|
Muto A, Fitzgerald TN, Pimiento JM, Maloney S, Teso D, Paszkowiak JJ, Westvik TS, Kudo FA, Nishibe T, Dardik A. Smooth muscle cell signal transduction: implications of vascular biology for vascular surgeons. J Vasc Surg 2007; 45 Suppl A:A15-24. [PMID: 17544020 PMCID: PMC1939976 DOI: 10.1016/j.jvs.2007.02.061] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 02/17/2007] [Indexed: 12/31/2022]
Abstract
Vascular smooth muscle cells exhibit varied responses after vessel injury and surgical interventions, including phenotypic switching, migration, proliferation, protein synthesis, and apoptosis. Although the source of the smooth muscle cells that accumulate in the vascular wall is controversial, possibly reflecting migration from the adventitia, from the circulating blood, or in situ differentiation, the intracellular signal transduction pathways that control these processes are being defined. Some of these pathways include the Ras-mitogen-activated protein kinase, phosphatidylinositol 3-kinase-Akt, Rho, death receptor-caspase, and nitric oxide pathways. Signal transduction pathways provide amplification, redundancy, and control points within the cell and culminate in biologic responses. We review some of the signaling pathways activated within smooth muscle cells that contribute to smooth muscle cell heterogeneity and development of pathology such as restenosis and neointimal hyperplasia.
Collapse
MESH Headings
- Animals
- Apoptosis
- Bone Marrow Cells/metabolism
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Constriction, Pathologic/metabolism
- Constriction, Pathologic/pathology
- Extracellular Matrix/metabolism
- Humans
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Muscle, Smooth, Vascular/surgery
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Protein Kinases/metabolism
- Signal Transduction
- Stem Cells/metabolism
- Vascular Surgical Procedures/adverse effects
Collapse
Affiliation(s)
- Akihito Muto
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Tamara N Fitzgerald
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Jose M Pimiento
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Stephen Maloney
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Desarom Teso
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Jacek J Paszkowiak
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Tormod S Westvik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Fabio A Kudo
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alan Dardik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
57
|
Fréguin-Bouilland C, Alkhatib B, David N, Lallemand F, Henry JP, Godin M, Thuillez C, Plissonnier D. Low Molecular Weight Fucoidan Prevents Neointimal Hyperplasia After Aortic Allografting. Transplantation 2007; 83:1234-41. [PMID: 17496541 DOI: 10.1097/01.tp.0000261109.97928.9c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Fucoidan, a new low molecular weight sulfated polysaccharide (LMWF), has previously been shown to mobilize bone marrow-derived progenitors cells via stimulation of stromal derived factor (SDF)-1 release. Mobilized progenitor cells have been suggested to repair intimal lesions after immune-mediated endothelial injury and thus prevent intimal proliferation. The aim of this study was to evaluate the effect of LMWF treatment in a rat aortic allograft model of transplant arteriosclerosis (TA). METHODS Aortic grafts were performed in Brown Norway (BN, donor) and Lewis (Lew, recipient) rats. The recipient rats were treated with LMWF (5 mg/kg/day) and sacrificed at 30 days. To determine the role of SDF-1 in mediating the effects of LMWF, a specific inhibitor of the SDF-1 receptor CXCR4, AMD 3100 (20 microg/kg/day), was used. The grafted segments were evaluated by morphometric (histochemical) analyses. RESULTS Untreated aortic allografts exhibited severe intimal proliferation, indicative of TA. In contrast, LMWF treatment significantly prevented allograft intimal proliferation as compared with controls (5.7+/-3 vs. 66.2+/-6 microm, P<0.01) and permitted a normalization of the intima/media ratio (0.1+/-0.1 vs. 1.7+/-0.3, P<0.01). Further, LMWF treatment stimulated allograft reendothelialization, as evidenced by strong intimal endothelial nitric oxide synthase antibody and CD31 signals. Unexpectedly, AMD treatment failed to prevent the protective effect of LMWF on intimal thickening and AMD treatment alone was found to reduced intimal proliferation in allografts. CONCLUSIONS We found that LMWF treatment reduced intimal thickness and induced the presence of an endothelial cell lining in the vascular graft at 30 days. Our findings may suggest a novel therapeutic strategy in the prevention of TA.
Collapse
|
58
|
Abstract
The identification of circulating endothelial progenitor cells (EPCs) has prompted an explosion of interest in postnatal vasculogenesis and the role of this mechanism in human health and disease. Previously considered restricted to the embryonic phase, the differentiation in situ of progenitor cells to vascular endothelium is now known to occur in the adult. A role for EPCs in the modulation of angiogenesis has also been recognized. These cells are enriched in the mononuclear cell fraction of peripheral blood but have also been isolated from bone marrow, the vessel wall, and a number of other organs and tissues. Accumulating data suggest an important vasculoprotective function for EPCs, although a maladaptive role underpinning a variety of angiogenesis-dependent diseases is also being investigated. Encouraging results observed with experimental and early human trials of EPC-based regenerative therapies have further underscored the significance of this recently discovered cell type. Notwithstanding the scope and pace of these developments, a number of challenges remain: the precise ontogeny and lineage of these cells is unknown, the true extent to which EPCs participate in neovascularization and vascular repair is still uncertain, and the efficacy of EPC-based regenerative therapies has yet to be proven in randomized controlled trials.
Collapse
Affiliation(s)
- Brendan Doyle
- Molecular Medicine Programm, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
59
|
Tanaka K, Sata M. Therapeutic Application of Bone Marrow-derived Progenitor Cells for Vascular Diseases: Magicbullets Having the Good Without the Bad? INT J GERONTOL 2007. [DOI: 10.1016/s1873-9598(08)70019-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
60
|
Yin X, Mayr M, Xiao Q, Wang W, Xu Q. Proteomic analysis reveals higher demand for antioxidant protection in embryonic stem cell-derived smooth muscle cells. Proteomics 2007; 6:6437-46. [PMID: 17163435 DOI: 10.1002/pmic.200600351] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Embryonic stem (ES) cells can differentiate into vascular smooth muscle cells (SMCs), but differences in protein composition, function and behaviour between stem cell-derived and mature SMCs remain to be characterized. Using differential in gel electrophoresis (DIGE) and MS, we identified 146 proteins that differed between ES cell-derived SMCs (esSMCs) and aortic SMCs, including proteins involved in DNA maintenance (higher in esSMCs), cytoskeletal proteins and calcium-binding proteins (higher in aortic SMCs). Notably, esSMCs showed decreased expression of mitochondrial, but a compensatory increase of cytosolic antioxidants. Subsequent experiments revealed that mitochondrial-derived reactive oxygen species (ROS) were markedly increased in esSMCs. Despite a three-fold rise in glutathione (GSH) reductase activity, esSMCs had lower levels of reduced GSH, and depletion of GSH by diethyl maleate or inhibition of GSH reductase by carmustine (BCNU) resulted in more pronounced cell death compared to aortic SMCs, while addition of antioxidants improved the viability of esSMCs. We present the first proteomic analysis of esSMCs demonstrating that stem cell-derived SMCs are more sensitive to oxidative stress due to increased generation of mitochondrial-derived ROS and require additional antioxidant protection for survival.
Collapse
Affiliation(s)
- Xiaoke Yin
- Cardiovascular Division, School of Medicine, King's College, University of London, UK
| | | | | | | | | |
Collapse
|
61
|
Wilkinson FL, Liu Y, Rucka AK, Jeziorska M, Hoyland JA, Heagerty AM, Canfield AE, Alexander MY. Contribution of VCAF-positive cells to neovascularization and calcification in atherosclerotic plaque development. J Pathol 2007; 211:362-9. [PMID: 17154367 PMCID: PMC1868967 DOI: 10.1002/path.2114] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Accepted: 10/30/2006] [Indexed: 12/21/2022]
Abstract
Calcification of the vessel wall is a regulated process with many similarities to osteogenesis. Progenitor cells may play a role in this process. Previously, we identified a novel gene, Vascular Calcification Associated Factor (VCAF), which was shown to be important in pericyte osteogenic differentiation. The aim of this study was to determine the localization and expression pattern of VCAF in human cells and tissues. Immunohistochemical analysis of seven atherosclerotic arteries confirmed VCAF protein expression within calcified lesions. In addition, individual VCAF-positive cells were detected within the intima and adventitia in areas where sporadic 3G5-positive pericytes were localized. Furthermore, VCAF-positive cells were identified in newly formed microvessels in association with CD34-positive/CD146-positive/c-kit-positive cells as well as in intact CD31-positive endothelium in internal mammary arteries. Western blot analysis confirmed the presence of VCAF (18 kD) in protein lysates extracted from human smooth muscle cells, endothelial cells, macrophages, and osteoblasts. In fracture callus samples from three patients, VCAF was detected in osteoblasts and microvessels. This study demonstrates the presence of VCAF in neovessels and raises the possibility that VCAF could be a new marker for vascular progenitor cells involved in a number of differentiation pathways. These data may have implications for the prevention or treatment of vascular disease.
Collapse
Affiliation(s)
- FL Wilkinson
- Cardiovascular Research Group, Division of Cardiovascular and Endocrine Sciences, University of Manchester, Manchester, UK
| | - Y Liu
- Cardiovascular Research Group, Division of Cardiovascular and Endocrine Sciences, University of Manchester, Manchester, UK
| | - AK Rucka
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - M Jeziorska
- Division of Laboratory and Regenerative Medicine, University of Manchester, Manchester, UK
| | - JA Hoyland
- Division of Laboratory and Regenerative Medicine, University of Manchester, Manchester, UK
| | - AM Heagerty
- Cardiovascular Research Group, Division of Cardiovascular and Endocrine Sciences, University of Manchester, Manchester, UK
| | - AE Canfield
- Cardiovascular Research Group, Division of Cardiovascular and Endocrine Sciences, University of Manchester, Manchester, UK
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - MY Alexander
- Cardiovascular Research Group, Division of Cardiovascular and Endocrine Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
62
|
Pelisek J, Fuchs AT, Kuehnl A, Tian W, Kuhlmann MT, Rolland PH, Mekkaoui C, Gaedtke L, Nikol S. C-type natriuretic peptide for reduction of restenosis: gene transfer is superior over single peptide administration. J Gene Med 2006; 8:835-44. [PMID: 16602133 DOI: 10.1002/jgm.905] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Restenosis is still a significant clinical problem limiting the long-term therapeutic success following balloon dilation or stent implantation. New approaches are necessary inhibiting neointima formation and simultaneously promoting re-endothelialization. Therefore, long-term therapeutic effects of adventitial liposome-mediated C-type natriuretic protein (CNP) gene and CNP peptide applications in a porcine model for restenosis post-angioplasty were investigated. METHODS For in vitro applications, primary cultures of porcine vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) were used. Gene transfer was performed with cationic lipid DOCSPER [1,3-dioleoyloxy-2-(N5-carbamoylspermine)propane]. In vivo treatment of pig femoral arteries was adventitial using a needle injection catheter following balloon angioplasty. Arteries were investigated by angiography, Evan's blue staining, histomorphometry, immunohistochemistry, PCR and RT-PCR. RESULTS Using CNP gene transfer in vitro, 29.4+/-7.2% reduction of cell proliferation in VSMCs was observed. In ECs, the CNP gene did not compromise cellular growth. For the CNP peptide the optimal concentration was 1 mM with 50.7+/-11.3% reduction of VSMC proliferation and 12.1+/-5.3% enhancement of growth of ECs. Three weeks following application in vivo complete re-endothelialization was observed in all treated groups. At 3 months significant reduction of neointima formation was observed using CNP gene vs. CNP peptide (85.9+/-7.8% vs. 63.3+/-27.6% reduction, P<0.05) compared to control treatment. CONCLUSION Periadventitial liposome-mediated CNP gene transfer in vivo resulted in a significant long-term reduction of neointima formation without compromising endothelial repair and was superior over single CNP peptide administration. Advantages of CNP are its physiological origin and simultaneous inhibition of VSMC proliferation and promotion of EC growth.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Animals
- Base Sequence
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Constriction, Pathologic
- DNA/genetics
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Femoral Artery/drug effects
- Femoral Artery/pathology
- Gene Expression
- Gene Transfer Techniques
- Liposomes
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Natriuretic Peptide, C-Type/administration & dosage
- Natriuretic Peptide, C-Type/genetics
- Sus scrofa
Collapse
Affiliation(s)
- Jaroslav Pelisek
- Department of Cardiology and Angiology, Westfaelische Wilhelm University, Muenster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Zeng L, Xiao Q, Margariti A, Zhang Z, Zampetaki A, Patel S, Capogrossi MC, Hu Y, Xu Q. HDAC3 is crucial in shear- and VEGF-induced stem cell differentiation toward endothelial cells. ACTA ACUST UNITED AC 2006; 174:1059-69. [PMID: 16982804 PMCID: PMC2064396 DOI: 10.1083/jcb.200605113] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Reendothelialization involves endothelial progenitor cell (EPC) homing, proliferation, and differentiation, which may be influenced by fluid shear stress and local flow pattern. This study aims to elucidate the role of laminar flow on embryonic stem (ES) cell differentiation and the underlying mechanism. We demonstrated that laminar flow enhanced ES cell–derived progenitor cell proliferation and differentiation into endothelial cells (ECs). Laminar flow stabilized and activated histone deacetylase 3 (HDAC3) through the Flk-1–PI3K–Akt pathway, which in turn deacetylated p53, leading to p21 activation. A similar signal pathway was detected in vascular endothelial growth factor–induced EC differentiation. HDAC3 and p21 were detected in blood vessels during embryogenesis. Local transfer of ES cell–derived EPC incorporated into injured femoral artery and reduced neointima formation in a mouse model. These data suggest that shear stress is a key regulator for stem cell differentiation into EC, especially in EPC differentiation, which can be used for vascular repair, and that the Flk-1–PI3K–Akt–HDAC3–p53–p21 pathway is crucial in such a process.
Collapse
Affiliation(s)
- Lingfang Zeng
- Department of Cardiac and Vascular Sciences, St. George's, University of London, London SW17 0RE, England, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Kusuyama T, Omura T, Nishiya D, Enomoto S, Matsumoto R, Murata T, Takeuchi K, Yoshikawa J, Yoshiyama M. The effects of HMG-CoA reductase inhibitor on vascular progenitor cells. J Pharmacol Sci 2006; 101:344-9. [PMID: 16891763 DOI: 10.1254/jphs.fp0060102] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Circulating bone marrow-derived vascular progenitor cells contribute to angiogenesis, atherosclerosis, and the response to vascular injury. These vascular progenitor cells consist of two cell groups, endothelial progenitor cells (EPCs) and smooth muscle progenitor cells (SMPCs). Although HMG-CoA reductase inhibitors (statins) have been reported to inhibit atherosclerosis partially by increased EPCs, the effects of statins on SMPCs are unclear. Therefore, we investigated the relationship between EPCs and SMPCs and whether pravastatin has atheroprotective effects on SMPCs. Peripheral mononuclear cells (MNCs) were isolated and cultured on fibronectin-coated dishes in SMPC medium. MNCs were stained with acetylated low density lipoprotein and lectin, or alpha-smooth muscle actin, and cell numbers were counted. mRNA expression and vascular endothelial growth factor (VEGF) protein synthesis of MNCs were evaluated. Pravastatin significantly increased the number of EPC and decreased the number of SMPC. mRNA expression of VEGF, endothelial nitric oxide synthase, VEGF receptor-2 (KDR), and Akt were up-regulated, and VEGF secretion was increased by pravastatin. The present study demonstrated that pravastatin has promotive effects on the differentiation from MNCs to EPC cells, while inhibitory effects to SMPC cells. Our findings suggest a previously unreported mechanism of the effect of statin therapy on vascular progenitor cells.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Cells, Cultured
- Culture Media, Conditioned/metabolism
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Immunohistochemistry
- Lectins/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Lipoproteins, LDL/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nitric Oxide Synthase Type III/genetics
- Platelet Endothelial Cell Adhesion Molecule-1/genetics
- Pravastatin/pharmacology
- Proto-Oncogene Proteins c-akt/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
- Time Factors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-2/genetics
Collapse
Affiliation(s)
- Takanori Kusuyama
- Department of Internal Medicine and Cardiology, Osaka City University Medical School, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
Strong evidence suggests that bone marrow-derived cells play a role in physiological and pathological blood vessel growth in the adult, both by augmenting angiogenesis through the secretion of angiogenic growth factors and by providing a rich source of progenitor cells that can differentiate into mature vascular endothelial cells. This is a true paradigm shift, since adult neovascularization processes were thought to be limited to angiogenesis. The cells that are critical to postnatal blood vessel growth – endothelial progenitor cells – may be analogous to the embryonic angioblast, in that they can circulate, proliferate and participate in the development of vascular networks by differentiating in situ, probably via the formation of cell clusters into mature endothelial cells. Therefore, initial reports have seen analogs to the process of vasculogenesis in the embryo, where the de novo synthesis of vessels occurs through the formation of blood island-like clusters, which subsequently connect and eventually form systemic vasculature. Recent work implicates precursors of endothelial cells in such processes as myocardial ischemia and infarction, limb ischemia, wound healing, atherosclerosis, endogenous endothelial repair and tumor vascularization. These new insights into the vascular biology of endothelial regeneration and repair led to the development of new cell therapeutic strategies to enhance adult neovascularization and re-endothelialization in ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Christoph Kalka
- Swiss Cardiovascular Center, Department of Vascular Medicine, Inselspital, Freiburgstrasse, 3010 Bern, Switzerland
| | - Stefano Di Santo
- Swiss Cardiovascular Center, Department of Vascular Medicine, Inselspital, University Hospital of Bern, Switzerland
| |
Collapse
|
66
|
Falk E. Pathogenesis of atherosclerosis. J Am Coll Cardiol 2006; 47:C7-12. [PMID: 16631513 DOI: 10.1016/j.jacc.2005.09.068] [Citation(s) in RCA: 1004] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Revised: 09/12/2005] [Accepted: 09/19/2005] [Indexed: 01/17/2023]
Abstract
Atherosclerosis is a multifocal, smoldering, immunoinflammatory disease of medium-sized and large arteries fuelled by lipids. Endothelial cells, leukocytes, and intimal smooth muscle cells are the major players in the development of this disease. The most devastating consequences of atherosclerosis, such as heart attack and stroke, are caused by superimposed thrombosis. Therefore, the vital question is not why atherosclerosis develops but rather why atherosclerosis, after years of indolent growth, suddenly becomes complicated with luminal thrombosis. If thrombosis-prone plaques could be detected and thrombosis averted, atherosclerosis would be a much more benign disease. Approximately 76% of all fatal coronary thrombi are precipitated by plaque rupture. Plaque rupture is a more frequent cause of coronary thrombosis in men (approximately 80%) than in women (approximately 60%). Ruptured plaques are characterized by a large lipid-rich core, a thin fibrous cap that contains few smooth muscle cells and many macrophages, angiogenesis, adventitial inflammation, and outward remodeling. Plaque rupture is the most common cause of coronary thrombosis. Ruptured plaques and, by inference, rupture-prone plaques have characteristic pathoanatomical features that might be useful for their detection in vivo by imaging. This article describes the pathogenesis of atherosclerosis, how it begets thrombosis, and the possibility to detect thrombosis-prone plaques and prevent heart attack.
Collapse
Affiliation(s)
- Erling Falk
- Department of Cardiology, Aarhus University Hospital (Skejby), Aarhus, Denmark.
| |
Collapse
|
67
|
Zhang LN, Wilson DW, da Cunha V, Sullivan ME, Vergona R, Rutledge JC, Wang YX. Endothelial NO Synthase Deficiency Promotes Smooth Muscle Progenitor Cells in Association With Upregulation of Stromal Cell-Derived Factor-1α in a Mouse Model of Carotid Artery Ligation. Arterioscler Thromb Vasc Biol 2006; 26:765-72. [PMID: 16456092 DOI: 10.1161/01.atv.0000207319.28254.8c] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Endothelial NO deficiency (endothelial NO synthase [eNOS]–knockout [KO]) enhanced smooth muscle cell (SMC)–rich neointimal lesion formation in a mouse model of carotid artery ligation (CAL). Recent evidence indicated that stromal cell-derived factor-1α (SDF-1α)–mediated recruitment of circulating SMC progenitor cells substantially contributed to the SMC-rich neointimal hyperplasia induced by vascular injury. The goal of this study was to investigate the effects of eNOS deficiency on the expression of SDF-1α and mobilization of circulating SMC progenitor cells in CAL model.
Methods and Results—
Two- to 3-month-old C57BL/6J wild-type (WT) and eNOS-KO mice were evaluated 1, 2, or 4 weeks after CAL. CAL-induced expression of SDF-1α, as detected by immunohistochemical staining and further quantified by ELISA in the ligated carotid arteries, was moderate and transient with a peak at 1 week in WT mice. SDF-1α expression was significantly higher at 1 week and persisted through 2 weeks in eNOS-KO mice. CAL was associated with increased circulating stem cell antigen-1+ (Sca-1
+
)/c-Kit
−
/Lin
−
cells (interpreted as SMC progenitor cells), which peaked at 1 week in WT mice. This effect was also significantly greater and longer-lasting in eNOS-KO than WT mice. The number of circulating Sca-1
+
/c-Kit
−
/Lin
−
cells was positively correlated with the expression of SDF-1α but not vascular endothelial growth factor in the ligated carotid arteries. Furthermore, immunostaining showed abundant Sca-1–positive cells in the adventitia of the 1-week ligated carotid arteries from eNOS-KO mice but not in WT mice. We also determined that eNOS deficiency enhanced CAL-induced intimal cell proliferation in the ligated arteries as detected by proliferating cell nuclear antigen staining but did not induce cell apoptosis as detected by staining for active caspase-3.
Conclusion—
Our results indicate that eNOS deficiency exacerbates CAL-induced expression of SDF-1α and its receptor CXCR4. This is correlated with an increase in Sca-1
+
cells in peripheral blood and adventitia, which may contribute to vascular remodeling and SMC-rich neointimal lesion formation. This suggests that constitutive eNOS inhibits SDF-1α expression and provides an important vasculoprotective mechanism for intact endothelium to limit SMC proliferation and recruitment in response to vascular injury.
Collapse
Affiliation(s)
- Le-Ning Zhang
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA 95616, USA.
| | | | | | | | | | | | | |
Collapse
|
68
|
Yin X, Mayr M, Xiao Q, Mayr U, Tarelli E, Wait R, Wang W, Xu Q. Proteomic dataset of Sca-1+ progenitor cells. Proteomics 2006; 5:4533-45. [PMID: 16240289 DOI: 10.1002/pmic.200402044] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Embryonic stem cells (ES cells) can differentiate into endothelial cells and smooth muscle cells (SMCs), which participate in vascular angiogenesis. In this study, we differentiated mouse ES cells into Sca-1(+) cells, which have the potential to serve as vascular progenitor cells, and mapped their proteome by 2-DE using a pH 3-10 non-linear gradient and 12% SDS-polyacrylamide gels. A subset of 300 protein spots was analysed and mapped, with 241 protein spots being identified by their PMF using MALDI-TOF MS or by partial amino acid sequencing using MS/MS. Our protein map is the first of Sca-1(+) progenitor cells and will facilitate the identification of proteins differentially expressed during stem cell differentiation. The proteome of adult arterial SMCs is described in an accompanying paper (in this issue, DOI 10.1002/pmic.200402045). All data are made accessible on our website http://www.vascular-proteomics.com.
Collapse
Affiliation(s)
- Xiaoke Yin
- Department of Cardiac and Vascular Sciences, St. George's, University of London, UK
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Berk BC. Vascular Smooth Muscle. Vasc Med 2006. [DOI: 10.1016/b978-0-7216-0284-4.50008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
70
|
Zhang L, Yang R, Han ZC. Transplantation of umbilical cord blood-derived endothelial progenitor cells: a promising method of therapeutic revascularisation. Eur J Haematol 2006; 76:1-8. [PMID: 16343265 DOI: 10.1111/j.1600-0609.2005.00579.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Therapeutic neovascularisation by endothelial progenitor cells (EPCs) mediated vascular regeneration is becoming a novel option for the treatment of ischaemic diseases. Recently, human umbilical cord blood (CB) has been found to contain a large number of EPCs and transplantation of CB EPCs led to a successful salvage of the ischaemic limbs through improvement in blood perfusion, indicating the feasibility of using CB cells for therapeutic revascularisation. This review will summarise recent studies in therapeutic revascularisation using CB cells and discuss the potential clinical utilisation of CB cells in ischaemic diseases.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Experimental Hematology, National Research Center of Stem Cell Engineering and Technology, Institute of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | | | | |
Collapse
|
71
|
Tan H, Jiang X, Yang F, Li Z, Liao D, Trial J, Magera MJ, Durante W, Yang X, Wang H. Hyperhomocysteinemia inhibits post-injury reendothelialization in mice. Cardiovasc Res 2006; 69:253-62. [PMID: 16226235 PMCID: PMC4400842 DOI: 10.1016/j.cardiores.2005.08.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 08/26/2005] [Accepted: 08/29/2005] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Hyperhomocysteinemia (HHcy) is a risk factor for cardiovascular disease and has been reported to inhibit endothelial cell (EC) growth. Notwithstanding, precisely how HHcy regulates EC growth in vivo remains unknown. In this study, we established a mouse model of endothelial injury and reendothelialization and examined the role and mechanism of HHcy in endothelial repair. METHODS AND RESULTS A mouse model of carotid artery air-dry endothelium denudation and reendothelialization was established and used to evaluate post-injury endothelial repair in mice with the gene deletion of cystathionine-beta-synthase (CBS). Moderate and severe HHcy were induced in CBS+/+ and CBS-/+ mice through a high-methionine diet. Post-injury reendothelialization, which correlated with increased post-injury neointima formation, was impaired in severe HHcy mice. To elucidate the underlying mechanism, we examined circulating endothelial progenitor cells (EPC) in HHcy mice and studied the effect of homocysteine (Hcy) on proliferation, migration, and adhesion of human umbilical vein endothelial cells (HUVEC). The peripheral EPC population was not significantly altered in HHcy mice. Hcy had a profound inhibitory effect on EC proliferation and migration at physiologically relevant concentrations and inhibited EC adhesion at concentrations of 200 microM and higher. CONCLUSION We have established a convenient and accurate mouse model of carotid injury in which the reendothelialization process can be precisely quantified. In addition, we have observed impaired reendothelialization and increased neointimal formation in severe HHcy mice. The capacity of Hcy to inhibit proliferation and migration of EC may be responsible for impaired reendothelialization and contribute to arteriosclerosis in HHcy.
Collapse
Affiliation(s)
- Hongmei Tan
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
- Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Xiaohua Jiang
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| | - Fan Yang
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| | - Zhaohui Li
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| | - Dan Liao
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| | - JoAnn Trial
- Department of Medicine, Baylor College of Medicine, United States
| | | | - William Durante
- Department of Medicine, Baylor College of Medicine, United States
- Department of Pharmacology, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| | - Xiaofeng Yang
- Department of Medicine, Baylor College of Medicine, United States
| | - Hong Wang
- Department of Medicine, Baylor College of Medicine, United States
- VA Medical Center, Houston, Texas 77030, United States
| |
Collapse
|
72
|
Joubert P, Lajoie-Kadoch S, Labonté I, Gounni AS, Maghni K, Wellemans V, Chakir J, Laviolette M, Hamid Q, Lamkhioued B. CCR3 expression and function in asthmatic airway smooth muscle cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:2702-8. [PMID: 16081847 DOI: 10.4049/jimmunol.175.4.2702] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Asthma is characterized by an increase in airway smooth muscle mass and a decreased distance between the smooth muscle layer and the epithelium. Furthermore, there is evidence to indicate that airway smooth muscle cells (ASMC) express a wide variety of receptors involved in the immune response. The aims of this study were to examine the expression of CCR3 on ASMC, to compare this expression between asthmatic and nonasthmatic subjects, and to determine the implications of CCR3 expression in the migration of ASMC. We first demonstrated that ASMC constitutively express CCR3 at both mRNA and protein levels. Interestingly, TNF-alpha increases ASMC surface expression of CCR3 from 33 to 74%. Furthermore, using FACS analysis, we found that ASMC CCR3 is expressed to a greater degree in asthmatic vs control subjects (95 vs 75%). Functionality of the receptor was demonstrated by calcium assay; the addition of CCR3 ligand eotaxin to ASMC resulted in an increase in intracellular calcium production. Interestingly, ASMC was seen to demonstrate a positive chemotactic response to eotaxin. Indeed, ASMC significantly migrated toward 100 ng/ml eotaxin (2.2-fold increase, compared with control). In conclusion, the expression of CCR3 by ASMC is increased in asthmatics, and our data show that a CCR3 ligand such as eotaxin induces migration of ASMC in vitro. These results may suggest that eotaxin could be involved in the increased smooth muscle mass observed in asthmatics through the activation of CCR3.
Collapse
MESH Headings
- Adult
- Asthma/immunology
- Asthma/metabolism
- Bronchi/cytology
- Bronchi/immunology
- Bronchi/metabolism
- Calcium/metabolism
- Cell Movement/immunology
- Cells, Cultured
- Chemokine CCL11
- Chemokines, CC/metabolism
- Chemokines, CC/pharmacology
- Humans
- Immunohistochemistry
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Ligands
- Monocyte Chemoattractant Proteins/metabolism
- Monocyte Chemoattractant Proteins/pharmacology
- Muscle, Smooth/cytology
- Muscle, Smooth/immunology
- Muscle, Smooth/metabolism
- RNA, Messenger/biosynthesis
- Receptors, CCR3
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/physiology
- Trachea/cytology
- Trachea/immunology
- Trachea/metabolism
- Tumor Necrosis Factor-alpha/physiology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Philippe Joubert
- Meakins-Christie Laboratories, McGill University, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Sata M, Fukuda D, Tanaka K, Kaneda Y, Yashiro H, Shirakawa I. The role of circulating precursors in vascular repair and lesion formation. J Cell Mol Med 2005; 9:557-68. [PMID: 16202205 PMCID: PMC6741295 DOI: 10.1111/j.1582-4934.2005.tb00488.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The accumulation of smooth muscle cells (SMCs) plays a principal role in atherogenesis, post-angioplasty restenosis and transplantation-associated vasculopathy. Therefore, much effort has been expended in targeting the migration and proliferation of medial smooth muscle cells to prevent occlusive vascular remodeling. Recent evidence suggests that bone marrow-derived circulating precursors can also give rise to endothelial cells and smooth muscle cells that contribute to vascular repair, remodeling, and lesion formation under physiological and pathological conditions. This article overviews recent findings on circulating vascular progenitor cells and describes potential therapeutic strategies that target these cells to treat occlusive vascular diseases.
Collapse
Affiliation(s)
- Masataka Sata
- Department of Cardiovascular Medicine, University of Tokyo, Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | | | | | | | | | | |
Collapse
|
74
|
Affiliation(s)
- Michael Simons
- Angiogenesis Research Center and Section of Cardiology, Department of Medicine, Dartmouth Medical School, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03756, USA.
| |
Collapse
|
75
|
Reyes M, Li S, Foraker J, Kimura E, Chamberlain JS. Donor origin of multipotent adult progenitor cells in radiation chimeras. Blood 2005; 106:3646-9. [PMID: 16099880 DOI: 10.1182/blood-2004-12-4603] [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: 12/17/2022] Open
Abstract
Multipotent adult progenitor cells (MAPCs) are bone marrow-derived stem cells that have extensive in vitro expansion capacity and can differentiate in vivo and in vitro into tissue cells of all 3 germinal layers: ectoderm, mesoderm, and endoderm. The origin of MAPCs within bone marrow is unknown. MAPCs are believed to be derived from the bone marrow stroma compartment as they are isolated within the adherent cell component. Numerous studies of bone marrow chimeras in the human and the mouse point to a host origin of bone marrow stromal cells. Mesenchymal stem cells (MSCs), which coexist with stromal cells, have also been proven to be of host origin after allogeneic bone marrow transplantation in numerous studies. We report here that following syngeneic bone marrow transplants into lethally irradiated C57BL6 mice, MAPCs are of donor origin.
Collapse
Affiliation(s)
- Morayma Reyes
- Department of Neurology, University of Washington School of Medicine, Seattle, 98195-7720, USA
| | | | | | | | | |
Collapse
|
76
|
Gulati R, Lerman A, Simari RD. Therapeutic uses of autologous endothelial cells for vascular disease. Clin Sci (Lond) 2005; 109:27-37. [PMID: 15966869 DOI: 10.1042/cs20050002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endothelial cells play important structural and functional roles in vascular homoeostasis. Perturbations in endothelial cell number and function are directly involved with the initiation and progression of multiple cardiovascular diseases, including atherosclerosis, hypertension and congestive heart failure. Attempts to modify these disorders have included pharmacological strategies to improve vascular and thus endothelial function. A goal of biological approaches to these disorders is the delivery of endothelial cells that might act to provide beneficial endothelial-derived factors. However, this approach has generally been limited by the lack of readily available autologous endothelial cells for delivery. The isolation of circulation-derived endothelial progenitor cells allows for direct access to autologous endothelial cells for preclinical and clinical studies. Preclinical studies using autologous endothelial cells have demonstrated beneficial effects when delivered in animal models of vascular injury and grafting. These effects are related to the endothelial nature of the cells and may be paracrine in nature. Ongoing studies are aimed at defining the nature of these effects and optimizing delivery strategies cognizant of these mechanisms.
Collapse
Affiliation(s)
- Rajiv Gulati
- Department of Cardiovascular Medicine, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
77
|
Hodges YK, Reese SM, Pahl PMB, Horwitz LD. Paradoxical Effects of Iron Chelation on Growth of Vascular Endothelial Cells. J Cardiovasc Pharmacol 2005; 45:539-44. [PMID: 15897780 DOI: 10.1097/01.fjc.0000159659.78675.4a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Endothelial cell (EC) and vascular smooth muscle cell (VSMC) interactions play critical roles in restenosis following vascular injury. We examined the effects of intracellular iron chelation on endothelial cell cycle progression and VSMC modulation of endothelial cell growth. A diffusible, lipid-soluble iron chelator that rapidly enters cells, desferri-exochelin 772SM (D-Exo), was studied in human endothelial cells and VSMCs. In both cell types D-Exo reversibly halted cell cycle progression from G0/G1 phase to S phase and from S phase to G2/M phase and increased expression of hypoxia-inducible factor 1alpha (HIF-1alpha). D-Exo increased secretion of vascular endothelial growth factor (VEGF), a downstream target of HIF-1alpha, in VSMCs, but there was no VEGF production in endothelial cells. D-Exo was 25-fold more potent than the lipid-insoluble iron chelator deferoxamine, which does not readily enter cells. Intracellular iron chelation with D-Exo directly inhibits endothelial cell growth but indirectly stimulates endothelial cell growth by increasing VEGF release by VSMCs.
Collapse
Affiliation(s)
- Yvonne K Hodges
- University of Colorado Health Sciences Center, Department of Medicine, Division of Cardiology, Denver, Colorado 80262, USA
| | | | | | | |
Collapse
|
78
|
Simosa HF, Wang G, Sui X, Peterson T, Narra V, Altieri DC, Conte MS. Survivin expression is up-regulated in vascular injury and identifies a distinct cellular phenotype. J Vasc Surg 2005; 41:682-90. [PMID: 15874934 DOI: 10.1016/j.jvs.2005.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVES The healing response to vascular injury is characterized by neointimal thickening. Proliferation and phenotypic transformation of vascular smooth muscle cells (SMCs) have been implicated in this process. We sought to investigate the role of survivin, a dual regulator of cell proliferation and apoptosis, in lesion formation after diverse forms of vascular injury. METHODS Rabbits underwent either carotid interposition vein grafting (n = 17) or bilateral femoral balloon injury (BI; n = 29); some in the BI group were placed on a high-cholesterol diet. A subset of BI arteries were treated with local adenoviral gene delivery of a survivin dominant negative-mutant (AdT34A) versus vector or saline controls. Survivin expression in vessels was analyzed by quantitative reverse transcriptase polymerase chain reaction (RT-PCR) and by immunohistochemistry (IHC), which also included markers of SMC differentiation. Specimens of human tissue including failed lower extremity bypass grafts and carotid plaque were also examined. RESULTS RT-PCR and IHC demonstrated increased survivin expression in all experimental models, colocalizing at early times with proliferating and alpha-actin-expressing cells but was largely absent in mature, contractile SMCs. Delivery of AdT34A after BI attenuated neointimal hyperplasia. CONCLUSION These studies provide strong evidence supporting a role for survivin in the cellular response to vascular injury. CLINICAL RELEVANCE The regulation of cell proliferation, death, and phenotype after vascular interventions remains incompletely understood. We investigated the role of the inhibitor of apoptosis protein survivin in diverse models of vascular injury. The results suggest that survivin is an important modulator of the generalized vascular injury response and may represent a relevant target for therapies targeting intimal hyperplasia.
Collapse
Affiliation(s)
- Hector F Simosa
- Division of Vascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
79
|
Shoji M, Sata M, Fukuda D, Tanaka K, Sato T, Iso Y, Shibata M, Suzuki H, Koba S, Geshi E, Katagiri T. Temporal and spatial characterization of cellular constituents during neointimal hyperplasia after vascular injury: Potential contribution of bone-marrow-derived progenitors to arterial remodeling. Cardiovasc Pathol 2005; 13:306-12. [PMID: 15556776 DOI: 10.1016/j.carpath.2004.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2004] [Revised: 07/26/2004] [Accepted: 08/10/2004] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Exuberant smooth muscle cells (SMCs) hyperplasia is the major cause of postangioplasty restenosis. We suggested that circulating smooth muscle progenitor cells might contribute to lesion formation after vascular injury. METHODS We extensively investigated the cellular constituents during neointimal formation after mechanical vascular injury. RESULTS A large wire was inserted into the mouse femoral artery, causing complete endothelial denudation and marked enlargement of the lumen with massive apoptosis of medial SMCs. At 2 h, the injured artery remained dilated with a thin media containing very few cells. A scanning electron microscopy showed fibrin and platelet deposition at the luminal side. One week after the injury, CD45-positive hematopoietic cells accumulated at the luminal side. Those CD45-positive cells gradually disappeared, whereas neointimal hyperplasia was formed with alpha-smooth muscle actin (SMA) positive cells. Bone marrow cells and peripheral mononuclear cells differentiated into alpha-SMA-positive cells in the presence of PDGF and basic FGF. Moreover, in bone marrow chimeric mice, bone-marrow-derived cells substantially contributed to neointimal hyperplasia after wire injury. CONCLUSION These results suggest that early accumulation of hematopoietic cells may play a role in the pathogenesis of SMC hyperplasia under certain circumstances.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Cells/metabolism
- Cell Differentiation
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Femoral Artery/injuries
- Femoral Artery/metabolism
- Femoral Artery/ultrastructure
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Hyperplasia
- Immunohistochemistry
- Leukocyte Common Antigens/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Male
- Mice
- Mice, Inbred C3H
- Microscopy, Electron, Scanning
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/ultrastructure
- RNA, Messenger/metabolism
- Tunica Intima/metabolism
- Tunica Intima/pathology
- Tunica Intima/physiopathology
Collapse
Affiliation(s)
- Makoto Shoji
- Third Department of Internal Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Miller AM, McPhaden AR, Preston A, Wadsworth RM, Wainwright CL. TNFα increases the inflammatory response to vascular balloon injury without accelerating neointimal formation. Atherosclerosis 2005; 179:51-9. [PMID: 15721009 DOI: 10.1016/j.atherosclerosis.2004.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 09/06/2004] [Accepted: 10/08/2004] [Indexed: 10/26/2022]
Abstract
There is now clear evidence for a contributory role of inflammatory processes to restenosis following vascular balloon injury and stent implantation. The aim of the present study was to study the effects of TNFalpha, administered locally in vivo immediately following balloon angioplasty, on the leukocyte adhesive response and extent of neointimal formation in a rabbit model of subclavian artery injury. Initial in vitro studies were performed with normal isolated artery rings to assess the vascular adhesive response to TNFalpha or IL-1beta. Pre-incubation with either cytokine prior to addition of (51)Cr-labelled leukocytes enhanced the adhesion of leukocytes to the artery in both a time- and concentration-dependent manner. Although both cytokines induced an increase in the expression of the adhesion molecules ICAM-1 and VCAM-1, only antibodies to ICAM-1 blocked the enhanced adhesion induced by the cytokines. In artery segments retrieved from rabbits that had previously undergone subclavian artery angioplasty either 24 h or 8 days previously, there was an injury-induced increase in adhesion of leukocytes assessed ex vivo. In segments obtained from rabbits that received a 15 min local infusion of TNFalpha (2 ng/min) to the injured artery immediately after the angioplasty procedure, leukocyte adhesion assessed ex vivo was further significantly enhanced. The pro-adhesive effect of TNFalpha was associated with an increased expression of both ICAM-1 and VCAM-1. However, TNFalpha administration did not alter the extent of neointimal formation observed 8 days after injury. These findings suggest that while TNFalpha may play a role following vascular injury, it does not act alone to induce neointimal formation. Thus anti-inflammatory strategies targeted at multiple cytokines may be more appropriate than targeting a single cytokine to reduce the response to vascular injury.
Collapse
Affiliation(s)
- Ashley M Miller
- Department of Physiology and Pharmacology, University of Strathclyde, Glasgow G4 0NR, UK
| | | | | | | | | |
Collapse
|
81
|
Araújo JDD, Araújo Filho JDD, Ciorlin E, Ruiz MA, Ruiz LP, Greco OT, Lago MR, Ardito RV. A terapia celular no tratamento da isquemia crítica dos membros inferiores. J Vasc Bras 2005. [DOI: 10.1590/s1677-54492005000400011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Os autores fazem um histórico sobre as pesquisas com células-tronco embrionárias e do cordão umbilical, suas respectivas vantagens e desvantagens. Seguem com as discussões sobre células-tronco adultas, sua definição, histórico, fontes e participação nos processos de regeneração tecidual, particularmente no endotélio. Ressaltam a importância de fatores que mobilizam as células-tronco adultas a partir da medula óssea: citocinas, angiopoietinas e outros fatores de crescimento. As células-tronco adultas mobilizam-se sob a forma de células endoteliais progenitoras, que têm origem comum com as células endoteliais a partir dos hemangioblastos. Os fatores de mobilização manifestam-se em condições de hipoxia e fazem com que as células endoteliais progenitoras se localizem nos locais de isquemia para produzir a neovasculogênese, que se faz por três possíveis mecanismos: a angiogênese (formação de novos capilares a partir de brotos de capilares já existentes), a arteriogênese (relacionada à circulação colateral) e a vasculogênese (vasos realmente novos). Fazem, a seguir, uma análise da literatura relativa à experimentação animal e aos estudos clínicos. Concluem ressaltando que as células-tronco adultas, embora tenham um grande potencial de uso, ainda demandam muito estudo e pesquisa para se firmar como método terapêutico.
Collapse
Affiliation(s)
| | - José Dalmo de Araújo Filho
- Instituto de Moléstias Cardiovasculares de São José do Rio Preto; Sociedade Brasileira de Angiologia e Cirurgia Vascular; Associação Médica Brasileira
| | - Emerson Ciorlin
- Instituto de Moléstias Cardiovasculares de São José do Rio Preto; Sociedade Brasileira de Angiologia e Cirurgia Vascular; Associação Médica Brasileira
| | - Milton Artur Ruiz
- Faculdade de Medicina de São José do Rio Preto; Hospital de Base de São José do Rio Preto; Instituto de Moléstias Cardiovasculares
| | - Lílian Piron Ruiz
- Hospital de Base de São José do Rio Preto; Instituto de Moléstias Cardiovasculares
| | | | | | | |
Collapse
|
82
|
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
|
83
|
Sata M. Molecular strategies to treat vascular diseases: circulating vascular progenitor cell as a potential target for prophylactic treatment of atherosclerosis. Circ J 2004; 67:983-91. [PMID: 14639011 DOI: 10.1253/circj.67.983] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerosis is responsible for more than half of all deaths in Western countries. Numerous studies have reported that accumulation of smooth muscle cells (SMCs) plays a principal role in atherogenesis, post-angioplasty restenosis and transplantation-associated vasculopathy. Although much effort has been devoted to targeting the migration and proliferation of medial SMCs, effective therapy to prevent occlusive vascular remodeling has not been established. Recently, it was suggested that bone marrow-derived precursors can give rise to vascular cells that contribute to the repair, remodeling, and lesion formation of the arterial wall under certain circumstances. This review highlights the recent findings on circulating vascular precursors and describes the potential therapeutic strategies for vascular diseases, targeting mobilization, homing, differentiation and proliferation of circulating progenitor cells.
Collapse
Affiliation(s)
- Masataka Sata
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Tokyo and PRESTO, Japan Science and Technology Agency, Kawaguchi.
| |
Collapse
|
84
|
Konishi A, Aizawa T, Mohan A, Korshunov VA, Berk BC. Hydrogen Peroxide Activates the Gas6-Axl Pathway in Vascular Smooth Muscle Cells. J Biol Chem 2004; 279:28766-70. [PMID: 15123721 DOI: 10.1074/jbc.m401977200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Axl, a receptor tyrosine kinase, is involved in cell survival, proliferation, and migration. We have shown that Axl expression increases in the neointima of balloon-injured rat carotids. Because oxidative stress is known to play a major role in remodeling of injured vessels, we hypothesized that H(2)O(2) might activate Axl by promoting autophosphorylation. H(2)O(2) rapidly stimulated Axl tyrosine phosphorylation in rat vascular smooth muscle cells within 1 min that was maximal at 5 min (6-fold). The response to H(2)O(2) was concentration-dependent with EC(50) of approximately 500 microm. Axl phosphorylation was partly dependent on production of its endogenous ligand, growth arrest gene 6 (Gas6), because Axl-Fc, a fragment of Axl extracellular domain that neutralizes Gas6, inhibited H(2)O(2)-induced Axl phosphorylation by 50%. Axl phosphorylation by H(2)O(2) was also attenuated by warfarin, which inhibits Gas6 activity by preventing post-translational modification. In intact vessels Axl was phosphorylated by H(2)O(2), and Axl phosphorylation was inhibited by warfarin treatment in balloon-injured carotids. Akt, a downstream target of Axl, was phosphorylated by H(2)O(2)in Axl(+/+) mouse aorta but significantly inhibited in Axl(-/-) aorta. Intimal proliferation was decreased significantly in a cuff injury model in Axl(-/-) mice compared with Axl(+/+) mice. In summary, Axl is an important signaling mediator for oxidative stress in cultured vascular smooth muscle cells and intact vessels and may represent an important therapeutic target for vascular remodeling and response to injury.
Collapse
Affiliation(s)
- Atsushi Konishi
- Center for Cardiovascular Research, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
85
|
|
86
|
Abstract
The elderly are particularly susceptible to disease, yet the biological mechanisms that bring about this susceptibility remain unclear. Using atherosclerosis as a model chronic illness, we review how recent studies of bone marrow-derived vascular repair systems in mice and humans provide new insights into the causes and potential cures for age-related illness. Organisms are born with a finite capacity for stem cell-mediated repair after chronic exposure to tissue injury. Once that capacity is exhausted, a cycle of pathological inflammation ensues and leads to overt disease manifestations. Augmentation of stem cell-mediated repair systems may provide a novel means of treating or preventing many age-related illnesses.
Collapse
|