151
|
Kawamoto A, Ii M, Asahara T. Vascular Regeneration: Endothelial Progenitor Cell Therapy for Ischemic Diseases. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
152
|
Marrotte EJ, Chen DD, Hakim JS, Chen AF. Manganese superoxide dismutase expression in endothelial progenitor cells accelerates wound healing in diabetic mice. J Clin Invest 2010; 120:4207-19. [PMID: 21060152 DOI: 10.1172/jci36858] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 09/08/2010] [Indexed: 12/15/2022] Open
Abstract
Amputation as a result of impaired wound healing is a serious complication of diabetes. Inadequate angiogenesis contributes to poor wound healing in diabetic patients. Endothelial progenitor cells (EPCs) normally augment angiogenesis and wound repair but are functionally impaired in diabetics. Here we report that decreased expression of manganese superoxide dismutase (MnSOD) in EPCs contributes to impaired would healing in a mouse model of type 2 diabetes. A decreased frequency of circulating EPCs was detected in type 2 diabetic (db/db) mice, and when isolated, these cells exhibited decreased expression and activity of MnSOD. Wound healing and angiogenesis were markedly delayed in diabetic mice compared with normal controls. For cell therapy, topical transplantation of EPCs onto excisional wounds in diabetic mice demonstrated that diabetic EPCs were less effective than normal EPCs at accelerating wound closure. Transplantation of diabetic EPCs after MnSOD gene therapy restored their ability to mediate angiogenesis and wound repair. Conversely, siRNA-mediated knockdown of MnSOD in normal EPCs reduced their activity in diabetic wound healing assays. Increasing the number of transplanted diabetic EPCs also improved the rate of wound closure. Our findings demonstrate that cell therapy using diabetic EPCs after ex vivo MnSOD gene transfer accelerates their ability to heal wounds in a mouse model of type 2 diabetes.
Collapse
Affiliation(s)
- Eric J Marrotte
- 1Department of Surgery, Vascular Medicine Institute, McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
153
|
Napoli C, Hayashi T, Cacciatore F, Casamassimi A, Casini C, Al-Omran M, Ignarro LJ. Endothelial progenitor cells as therapeutic agents in the microcirculation: an update. Atherosclerosis 2010; 215:9-22. [PMID: 21126740 DOI: 10.1016/j.atherosclerosis.2010.10.039] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 09/28/2010] [Accepted: 10/25/2010] [Indexed: 12/15/2022]
Abstract
This review evaluates novel beneficial effects of circulating endothelial progenitor cells (EPCs) as shown by several preclinical studies and clinical trials carried out to test the safety and feasibility of using EPCs. There are 31 registered clinical trials (and many others still ongoing) and 19 published studies. EPCs originate in the bone marrow and migrate into the bloodstream where they undergo a differentiation program leading to major changes in their antigenic characteristics. EPCs lose typical progenitor markers and acquire endothelial markers, and two important receptors, (VEGFR and CXCR-4), which recruit circulating EPCs to damaged or ischemic microcirculatory (homing to damaged tissues) beds. Overall, therapeutic angiogenesis will likely change the face of regenerative medicine in the next decade with many patients worldwide predicted to benefit from these treatments.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of General Pathology, Division of Clinical Pathology and Excellence Research Center on Cardiovascular Diseases, 1st School of Medicine, II University of Naples, 80138 Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
154
|
Pesce M, Burba I, Gambini E, Prandi F, Pompilio G, Capogrossi MC. Endothelial and cardiac progenitors: boosting, conditioning and (re)programming for cardiovascular repair. Pharmacol Ther 2010; 129:50-61. [PMID: 21035506 DOI: 10.1016/j.pharmthera.2010.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/06/2010] [Indexed: 12/26/2022]
Abstract
Preclinical studies performed in cell culture and animal systems have shown the outstanding ability of stem cells to repair ischemic heart and lower limbs by promoting the formation of new blood vessels and new myocytes. In contrast, clinical studies of stem cell administration in patients with myocardial ischemia have revealed only modest, although promising, results. Basic investigations have shown the feasibility of adult cells reprogramming into pluripotent cells by defined factors, thus opening the way to the devise of protocols to ex vivo derive virtually unexhausted cellular pools. In contrast, cellular and molecular studies have indicated that risk factors limit adult-derived stem cell survival, proliferation and engraftment in ischemic tissues. The use of fully reprogrammed cells raises safety concerns; therefore, adult cells remain a primary option for clinicians interested in therapeutic cardiovascular repair. Pharmacologic approaches have been devised to restore the cardiovascular repair ability of failing progenitors from patients at risk. In the present contribution, the most advanced pharmacologic approaches to (re)program, boost, and condition endothelial and cardiac progenitor cells to enhance cardiovascular regeneration are discussed.
Collapse
Affiliation(s)
- Maurizio Pesce
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
155
|
Yuan L, Wu MJ, Sun HY, Xiong J, Zhang Y, Liu CY, Fu LL, Liu DM, Liu HQ, Mei CL. VEGF-modified human embryonic mesenchymal stem cell implantation enhances protection against cisplatin-induced acute kidney injury. Am J Physiol Renal Physiol 2010; 300:F207-18. [PMID: 20943766 DOI: 10.1152/ajprenal.00073.2010] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The implantation of mesenchymal stem cells (MSC) has been reported as a new technique to restore renal tubular structure and improve renal function in acute kidney injury (AKI). Vascular endothelial growth factor (VEGF) plays an important role in the renoprotective function of MSC. Whether upregulation of VEGF by a combination of MSC and VEGF gene transfer could enhance the protective effect of MSC in AKI is not clear. We investigated the effects of VEGF-modified human embryonic MSC (VEGF-hMSC) in healing cisplatin-injured renal tubular epithelial cells (TCMK-1) with a coculture system. We found that TCMK-1 viability declined 3 days after cisplatin pretreatment and that coculture with VEGF-hMSC enhanced cell protection via mitogenic and antiapoptotic actions. In addition, administration of VEGF-hMSC in a nude mouse model of cisplatin-induced kidney injury offered better protective effects on renal function, tubular structure, and survival as represented by increased cell proliferation, decreased cellular apoptosis, and improved peritubular capillary density. These data suggest that VEGF-modified hMSC implantation could provide advanced benefits in the protection against AKI by increasing antiapoptosis effects and improving microcirculation and cell proliferation.
Collapse
Affiliation(s)
- Li Yuan
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Saltzman AJ, Choi SW, Dabreo A, Baur WE, Weiss E, Nguyen K, Ishibashi F, Celestin FF, Karia DH, Pandian NG, Karas RH, Waxman S. Endothelial progenitor cells delivered into the pericardial space incorporate into areas of ischemic myocardium. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2010; 11:241-8. [DOI: 10.1016/j.carrev.2009.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 05/28/2009] [Accepted: 06/04/2009] [Indexed: 11/29/2022]
|
157
|
Johnson CA, Sarwate S, Miller RJ, O'Brien WD. A temporal study of ultrasound contrast agent-induced changes in capillary density. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2010; 29:1267-75. [PMID: 20733181 PMCID: PMC3069919 DOI: 10.7863/jum.2010.29.9.1267] [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] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
OBJECTIVE The ability of ultrasound (US) and ultrasound contrast agents (UCAs) to induce angiogenesis has been explored as a means of restoring blood flow to ischemic muscle. Because UCAs demonstrate an increasing percentage of collapse cavitation with increasing US pressure (Pr), this study sought to explore the effects of a US Pr that produces 100% collapse cavitation, determine the capillary density changes, and determine the time point of angiogenic rebound in a normal animal model. METHODS Using a 1-MHz focused transducer and a peak rarefactional US Pr of 3.8 MPa, rat gracilis muscles were exposed to US, and bioeffects were assessed. Capillary density, as a measure of angiogenesis, was examined. As an additional measure, inflammatory cells were quantified via a color threshold analysis to detect the presence of CD31 and CD34 as a percentage of the total section on stained slides. Six groups (0, 3, 6, 13, 20, and 27 days postexposure [DPE]; n = 3 each) and 5 cage controls were used to characterize the angiogenic response. RESULTS Ultrasound-UCA treatment caused the capillary density to decrease acutely (0 DPE) by 70% and inflammatory cells to increase by up to 250%. The angiogenic rebound was observed at 3 DPE but did not return to control levels by 27 DPE, suggesting an incomplete healing response. CONCLUSIONS Capillary destruction and inflammation played an important role in the angiogenic response induced by US-UCA. Exposure that causes 100% collapse cavitation causes capillary destruction from which normal rats are unable to recover and suggests a nontherapeutic effect.
Collapse
Affiliation(s)
- Chenara A Johnson
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
158
|
Cao L, Li L, Yang H, Yin H. Overexpression of P-selectin glycoprotein ligand-1 enhances adhesive properties of endothelial progenitor cells through Syk activation. Acta Biochim Biophys Sin (Shanghai) 2010; 42:507-14. [PMID: 20705591 DOI: 10.1093/abbs/gmq057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1) not only functions as an anchor molecule to capture monocytes and other leukocytes to endothelial cells in ischemic tissue by its interaction with P-selectin, but also transduces signals to initiate firm adhesion. Endothelial progenitor cells are derived from monocytes and play a very important role in neovascularization. Transplantation of endothelial progenitor cells is a promising therapeutic strategy to improve treatment of ischemic disease such as myocardial and cerebral infarction; however, its efficacy is now limited by the fact that few of the transplanted cells adhere to and accumulate in the ischemic tissue. In this study we aimed to investigate whether the overexpression of PSGL-1 gene promotes endothelial progenitor cells adhesion activity and explore the underlying mechanisms. We found that after transfection with human PSGL-1 gene, endothelial progenitor cells exhibited higher affinity to activated human umbilical vein endothelial cells or recombined P-selectin/ICAM-1 monolayer. The overexpression of PSGL-1-enhanced beta2-integrin expression on endothelial progenitor cells surface, and this effect was Syk dependent. The specific Syk inhibitor abolished the elevating effect of overexpression of PSGL-1 on surface beta2-integrin expression and the adhesive affinity of endothelial progenitor cells. These results suggested that Syk plays a key role in signal transduction downstream of PSGL-1 in endothelial progenitor cells, and the overexpression of PSGL-1 improves endothelial progenitor cells adhesive properties through enhanced activation of Syk and following integrin activation.
Collapse
Affiliation(s)
- Li Cao
- Department of Neurology, Beijing Anzhen Hospital of Capital Medical University, China
| | | | | | | |
Collapse
|
159
|
Wang W, Li W, Ou L, Flick E, Mark P, Nesselmann C, Lux CA, Gatzen HH, Kaminski A, Liebold A, Lützow K, Lendlein A, Li RK, Steinhoff G, Ma N. Polyethylenimine-mediated gene delivery into human bone marrow mesenchymal stem cells from patients. J Cell Mol Med 2010; 15:1989-98. [PMID: 20629995 PMCID: PMC3918054 DOI: 10.1111/j.1582-4934.2010.01130.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) derived from adult bone marrow has been proposed as a potential therapeutic approach for post-infarction left ventricular (LV) dysfunction. However, age-related functional decline of stem cells has restricted their clinical benefits after transplantation into the infarcted myocardium. The limitations imposed on patient cells could be addressed by genetic modification of stem cells. This study was designed to improve our understanding of genetic modification of human bone marrow derived mesenchymal stem cells (hMSCs) by polyethylenimine (PEI, branched with Mw 25 kD), one of non-viral vectors that show promise in stem cell genetic modification, in the context of cardiac regeneration for patients. We optimized the PEI-mediated reporter gene transfection into hMSCs, evaluated whether transfection efficiency is associated with gender or age of the cell donors, analysed the influence of cell cycle on transfection and investigated the transfer of therapeutic vascular endothelial growth factor gene (VEGF). hMSCs were isolated from patients with cardiovascular disease aged from 41 to 85 years. Optimization of gene delivery to hMSCs was carried out based on the particle size of the PEI/DNA complexes, N/P ratio of complexes, DNA dosage and cell viability. The highest efficiency with the cell viability near 60% was achieved at N/P ratio 2 and 6.0 μg DNA/cm2. The average transfection efficiency for all tested samples, middle-age group (<65 years), old-age group (>65 years), female group and male group was 4.32%, 3.85%, 4.52%, 4.14% and 4.38%, respectively. The transfection efficiency did not show any correlation either with the age or the gender of the donors. Statistically, there were two subpopulations in the donors; and transfection efficiency in each subpopulation was linearly related to the cell percentage in S phase. No significant phenotypic differences were observed between these two subpopulations. Furthermore, PEI-mediated therapeutic gene VEGF transfer could significantly enhance the expression level.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Hagikura K, Fukuda N, Yokoyama SI, Yuxin L, Kusumi Y, Matsumoto T, Ikeda Y, Kunimoto S, Takayama T, Jumabay M, Mitsumata M, Saito S, Hirayama A, Mugishima H. Low invasive angiogenic therapy for myocardial infarction by retrograde transplantation of mononuclear cells expressing the VEGF gene. Int J Cardiol 2010; 142:56-64. [PMID: 19167769 DOI: 10.1016/j.ijcard.2008.12.108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 12/03/2008] [Accepted: 12/12/2008] [Indexed: 01/10/2023]
Abstract
BACKGROUND Although transplantation of mononuclear cells (MNCs) induces angiogenesis in myocardial infarction, transplantation requires a large amount of bone marrow or peripheral blood cells. We examined the effects of transplantation of peripheral MNCs expressing an exogenous vascular endothelial growth factor (VEGF) gene in a pig model of acute myocardial infarction (AMI). METHODS MNCs were isolated from 20 ml peripheral blood from pigs and transfected with 10 microg of human VEGF165 plasmid (phVEGF). Myocardial infarction was induced by occlusion of the mid portion of the left anterior descending coronary artery (LAD) in anesthetized pigs. At 4 h after total occlusion, 5 x 10(6) VEGF-transfected MNCs were retrogradely transplanted into the pig via the coronary vein. Cardiac function, neovascularization and histology of the ischemic tissue were evaluated 4 weeks after transplantation. RESULTS MNCs expressing hVEGF and infused via the coronary vein were efficiently delivered the heart in pigs with myocardial infarction. Transplantation of MNCs expressing hVEGF significantly increased left ventricular (LV) function, collateral vessels, and capillary density in heart from AMI model pigs. Transplantation of MNCs expressing hVEGF increased the wall thickness of the scar in the heart after AMI. CONCLUSIONS Retrograde transplantation of peripheral blood MNCs expressing hVEGF efficiently induced angiogenesis and improved the impaired LV function in hearts of pigs with AMI. These findings indicate that angiogenic cells and gene therapy may be useful to treat ischemic heart disease.
Collapse
Affiliation(s)
- Kazuhiro Hagikura
- Department of Advanced Medicine, Nihon University School of Medicine, Division of Cell Regeneration and Transplantation, 30-1, Oyaguchi, Kami-machi, Itabashi-ku, 173-8610, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Hu JJ, Sun C, Lan L, Chen YW, Li DG. Therapeutic effect of transplanting beta(2)m(-)/Thy1(+) bone marrow-derived hepatocyte stem cells transduced with lentiviral-mediated HGF gene into CCl(4)-injured rats. J Gene Med 2010; 12:244-54. [PMID: 20143305 DOI: 10.1002/jgm.1439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND beta(2)m(-)/Thy1(+) bone marrow-derived hepatocyte stem cells (BDHSCs) isolated from the bone marrow of cholestatic rats by magnetic bead cell sorting consistently express characteristics of both stem and liver cells. These stem cells may be good vehicles for gene transfer. Administration of exogenous hepatocyte growth factor (HGF) may be potentially useful for the treatment of liver fibrosis. Because lentiviral vectors integrate stably into the host-cell genome of nondividing and dividing cells, it may efficiently transfect beta(2)m(-)/Thy1(+) BDHSCs in vitro and secrete high-level HGF consistently. Transplantation of beta(2)m(-)/Thy1(+) BDHSCs transduced with lentiviral vectors containing the HGF gene may reduce liver fibrosis in rats. METHODS Lentiviral vectors expressing HGF were constructed and used to transduce beta(2)m(-)/Thy1(+) BDHSCs sorted from cholestatic rats in vitro. Transduction efficiency was evaluated and then these cells were transplanted into rats through the portal vein. Liver function as well as histological and immunohistochemical examinations were carried out to assess the therapeutic efficacy on liver fibrosis. RESULTS We demonstrated that high-level exogenous HGF was detected in supernatants after beta(2)m(-)/Thy1(+) BDHSCs were transfected with lentiviral vectors expressing HGF. Transplantation of transduced beta(2)m(-)/Thy1(+) BDHSCs significantly enhanced liver function and attenuated liver fibrosis in vivo. CONCLUSIONS The present study indicates that transplantation of beta(2)m(-)/Thy1(+) BDHSCs overexpressing the HGF gene may offer a novel approach for promoting liver function and reverse liver fibrosis.
Collapse
Affiliation(s)
- Jun-Jie Hu
- Department of Gastroenterology, Xinhua Hospital, College of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
162
|
Yang JX, Tang WL, Wang XX. Superparamagnetic iron oxide nanoparticles may affect endothelial progenitor cell migration ability and adhesion capacity. Cytotherapy 2010; 12:251-9. [PMID: 20196696 DOI: 10.3109/14653240903446910] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AIMS Cell labeling with superparamagnetic iron oxide (SPIO) nanoparticles enables non-invasive tracking of transplanted cells. The aim of this study was to investigate whether SPIO nanoparticles have an effect on endothelial progenitor cell (EPC) functional activity and the feasibility of a protocol for labeling swine- and rat-origin EPC using SPIO nanoparticles at an optimized low dosage. METHODS EPC were isolated from the peripheral blood of swine and bone marrow of rat and characterized. After ex vivo cultivation, EPC were labeled with SPIO nanoparticles (to make a series of final concentrations, 50, 100, 200 and 400 microg/mL) or vehicle control. We also investigated the long-term effects of 200 microg/mL SPIO nanoparticles on EPC (4, 8, 12 and 16 days after labeling). The labeling efficiency was tested through Prussian blue (PB) staining and the intracellular iron uptake was also measured quantitatively and confirmed. EPC proliferation and migration were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and transwell chamber assay, respectively. An EPC adhesion assay was performed by replating the cells on fibronectin-coated dishes and then counting the adherent cells. EPC apoptosis was evaluated using an Annexin V-FITC apoptosis kit. RESULTS SPIO nanoparticles impaired EPC migration and promoted EPC adhesion. EPC proliferation and apoptosis were not affected. SPIO nanoparticles could label EPC efficiently at 200 microg/mL overnight without significantly affecting EPC functional activity. CONCLUSIONS SPIO nanoparticles impaired the EPC migration ability and promoted the EPC adhesion capacity. EPC could be labeled efficiently at an appropriate concentration (200 microg/mL) without significantly affecting their functional activity.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- Department of Cardiology, The First Affiliated Hospital, Medical School of Zhejiang University, Hangzhou, China
| | | | | |
Collapse
|
163
|
Schuhmann NK, Pozzoli O, Sallach J, Huber A, Avitabile D, Perabo L, Rappl G, Capogrossi MC, Hallek M, Pesce M, Büning H. Gene transfer into human cord blood-derived CD34(+) cells by adeno-associated viral vectors. Exp Hematol 2010; 38:707-17. [PMID: 20447441 DOI: 10.1016/j.exphem.2010.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 04/08/2010] [Accepted: 04/27/2010] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Bone marrow-derived CD34(+) cells are currently used in clinical trials in patients with ischemic heart disease. An option to enhance activity of injected progenitors may be offered by genetic engineering of progenitor cells with angiogenic growth factors. Recombinant adeno-associated viral vectors (rAAV) have emerged as a leading gene transfer systems. In contrast to other vector systems in use for genetic engineering of CD34(+) cells, rAAV-mediated gene expression does not depend on vector integration. This is relevant for application in regenerative medicine of ischemic tissues, where transient transgene expression is likely sufficient to achieve therapeutic benefits. MATERIALS AND METHODS We compared three different human AAV serotypes, packaged as pseudotypes by a helper virus-free production method, for their transduction efficiency in human cord blood-derived CD34(+) cells. We further assessed the impact of vector genome conformation, of alpha(v)beta(5) and alpha(5)beta(1) integrin availability and of the transcription-modulating drugs retinoic acid and Trichostatin A on rAAV-mediated human CD34(+) cell transduction. RESULTS We provide, for the first time, evidence that hCD34(+) cells can be reproducibly transduced with high efficiency by self-complementary rAAV2 without inducing cytotoxicity or interfering with their differentiation potential. We further show the involvement of alpha(5)beta(1) integrin as a crucial AAV2 internalization receptor and a function for transcription-modulating drugs in enhancing rAAV-mediated transgene expression. CONCLUSION This study represents a first step toward translation of a combined cellular/rAAV-based therapy of ischemic disease.
Collapse
|
164
|
Jarajapu YPR, Grant MB. The promise of cell-based therapies for diabetic complications: challenges and solutions. Circ Res 2010; 106:854-69. [PMID: 20299675 DOI: 10.1161/circresaha.109.213140] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The discovery of endothelial progenitor cells (EPCs) in human peripheral blood advanced the field of cell-based therapeutics for many pathological conditions. Despite the lack of agreement about the existence and characteristics of EPCs, autologous EPC populations represent a novel treatment option for complications requiring therapeutic revascularization and vascular repair. Patients with diabetic complications represent a population of patients that may benefit from cellular therapy yet their broadly dysfunctional cells may limit the feasibility of this approach. Diabetic EPCs have decreased migratory prowess and reduced proliferative capacity and an altered cytokine/growth factor secretory profile that can accelerate deleterious repair mechanisms rather than support proper vascular repair. Furthermore, the diabetic environment poses additional challenges for the autologous transplantation of cells. The present review is focused on correcting diabetic EPC dysfunction and the challenges involved in the application of cell-based therapies for treatment of diabetic vascular complications. In addition, ex vivo and in vivo functional manipulation(s) of EPCs to overcome these hurdles are discussed.
Collapse
Affiliation(s)
- Yagna P R Jarajapu
- Pharmacology and Therapeutics, College of Medicine, University of Florida, PO Box 100267, Gainesville, FL 32610-0267, USA
| | | |
Collapse
|
165
|
Zhang P, Baxter J, Vinod K, Tulenko TN, Di Muzio PJ. Endothelial differentiation of amniotic fluid-derived stem cells: synergism of biochemical and shear force stimuli. Stem Cells Dev 2010; 18:1299-308. [PMID: 19508152 DOI: 10.1089/scd.2008.0331] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human amniotic fluid-derived stem (AFS) cells possess several advantages over embryonic and adult stem cells, as evidenced by expression of both types of stem cell markers and ability to differentiate into cells of all three germ layers. Herein, we examine endothelial differentiation of AFS cells in response to growth factors, shear force, and hypoxia. We isolated human AFS cells from amniotic fluid samples (1-4 cc/specimen) obtained from patients undergoing amniocentesis at 15-18 weeks of gestation (n = 10). Isolates maintained in nondifferentiating medium expressed the stem cell markers CD13, CD29, CD44, CD90, CD105, OCT-4, and SSEA-4 through passage 8. After 3 weeks of culture in endothelial growth media-2 (EGM-2), the stem cells exhibited an endothelial-like morphology, formed cord-like structures when plated on Matrigel, and uptook acetylated LDL/lectin. Additionally, mRNA and protein levels of CD31 and von Willebrand factor (vWF) significantly increased in response to culture in EGM-2, with further up-regulation when stimulated by physiological levels (12 dyne/cm(2)) of shear force. Culture in hypoxic conditions (5% O(2)) resulted in significant expression of vascular endothelial growth factor (VEGF) and placental growth factor (PGF) mRNA. This study suggests that AFS cells, isolated from minute amounts of amniotic fluid, acquire endothelial cell characteristics when stimulated by growth factors and shear force, and produce angiogenic factors (VEGF, PGF, and hepatocyte growth factor [HGF]) in response to hypoxia. Thus, amniotic fluid represents a rich source of mesenchymal stem cells potentially suitable for use in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|
166
|
Zhang Y, Wong S, Laflèche J, Crowe S, Mesana TG, Suuronen EJ, Ruel M. In vitro functional comparison of therapeutically relevant human vasculogenic progenitor cells used for cardiac cell therapy. J Thorac Cardiovasc Surg 2010; 140:216-24, 224.e1-4. [PMID: 20167338 DOI: 10.1016/j.jtcvs.2009.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 10/15/2009] [Accepted: 11/03/2009] [Indexed: 01/11/2023]
Abstract
OBJECTIVE In cardiac cell therapy almost every cell type tested experimentally has yielded some benefit. However, there is a lack of studies directly comparing the function of various stem/progenitor cell populations. This study describes the expansion of peripheral blood CD133(+) cells and compares their functional properties with those of other commonly used human progenitor cell populations. METHODS CD133(+) cells were generated from the CD133(-) fraction of peripheral blood, either serially (pooled-derived) or after 14 days of culture (derived). Their phenotypic, migratory, and vasculogenic properties were compared with those of 4 commonly used progenitor cell populations in vitro. RESULTS Serial expansion resulted in an 11-fold increase in the number of CD133(+) cells. The proportion of derived CD133(+) cells collected between 0 and 8 days also expressing CD34 and vascular endothelial growth factor receptor 2 was similar (approximately 60%, P = .41). Adherent, 4-day cultured endothelial progenitor cells demonstrated enhanced migration compared with each of the other 5 cell populations (all P < or = .002). The migration of derived CD133(+) progenitors was enhanced by coculture with CD133(-) cells or their supernatant (P < .05). In vitro vasculogenesis assays revealed that derived and pooled-derived CD133(+) cells had superior vasculogenic potential compared with other progenitor populations (P < or = .03). CONCLUSIONS A novel source of expandable CD133(+) cells can be generated from the CD133(-) fraction of peripheral blood. The CD133 phenotypic marker translates into the cell being vasculogenically more potent in vitro, which could be beneficial to inducing vasculogenesis in the ischemic heart. Furthermore, intercellular interactions appear important for improving the therapeutic efficacy of cell transplantation.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
167
|
Zakharova L, Mastroeni D, Mutlu N, Molina M, Goldman S, Diethrich E, Gaballa MA. Transplantation of cardiac progenitor cell sheet onto infarcted heart promotes cardiogenesis and improves function. Cardiovasc Res 2010; 87:40-9. [PMID: 20118202 DOI: 10.1093/cvr/cvq027] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIMS Cell-based therapy for myocardial infarction (MI) holds great promise; however, the ideal cell type and delivery system have not been established. Obstacles in the field are the massive cell death after direct injection and the small percentage of surviving cells differentiating into cardiomyocytes. To overcome these challenges we designed a novel study to deliver cardiac progenitor cells as a cell sheet. METHODS AND RESULTS Cell sheets composed of rat or human cardiac progenitor cells (cardiospheres), and cardiac stromal cells were transplanted onto the infarcted myocardium after coronary artery ligation in rats. Three weeks later, transplanted cells survived, proliferated, and differentiated into cardiomyocytes (14.6 +/- 4.7%). Cell sheet transplantation suppressed cardiac wall thinning and increased capillary density (194 +/- 20 vs. 97 +/- 24 per mm(2), P < 0.05) compared with the untreated MI. Cell migration from the sheet was observed along the necrotic trails within the infarcted area. The migrated cells were located in the vicinity of stromal-derived factor (SDF-1) released from the injured myocardium, and about 20% of these cells expressed CXCR4, suggesting that the SDF-1/CXCR4 axis plays, at least, a role in cell migration. Transplantation of cell sheets resulted in a preservation of cardiac contractile function after MI, as was shown by a greater ejection fraction and lower left ventricular end diastolic pressure compared with untreated MI. CONCLUSION The scaffold-free cardiosphere-derived cell sheet approach seeks to efficiently deliver cells and increase cell survival. These transplanted cells effectively rescue myocardium function after infarction by promoting not only neovascularization but also inducing a significant level of cardiomyogenesis.
Collapse
Affiliation(s)
- Liudmila Zakharova
- Cardiovascular Research Laboratory, Center for Cardiovascular Research, Banner Sun Health Research Institute, 10515 W. Santa Fe Drive, Sun City, AZ 85351, USA
| | | | | | | | | | | | | |
Collapse
|
168
|
Germani A, Di Campli C, Pompilio G, Biglioli P, Capogrossi MC. Regenerative therapy in peripheral artery disease. Cardiovasc Ther 2010; 27:289-304. [PMID: 19903190 DOI: 10.1111/j.1755-5922.2009.00105.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Patients with peripheral artery disease (PAD) and critical limb ischemia are the main candidates for limb amputations and have a poor life expectancy. Frequently, these patients are not eligible for either surgical or percutaneous interventions aimed at mechanical revascularization. Therefore, new strategies need to be identified to offer these patients a viable therapeutic option. Gene and cell therapy hold great promise for the treatment of peripheral vascular diseases because, in animal models, local delivery of growth factors and endothelial progenitor cells result in new blood vessel formation and regeneration of ischemic tissues. In this article, are reviewed phase I and phase II gene, and cell therapy clinical trials in patients with PAD.
Collapse
|
169
|
The promotion of endothelial progenitor cells recruitment by nerve growth factors in tissue-engineered blood vessels. Biomaterials 2009; 31:1636-45. [PMID: 20006381 DOI: 10.1016/j.biomaterials.2009.11.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Accepted: 11/16/2009] [Indexed: 11/21/2022]
Abstract
Endothelial progenitor cells (EPCs) mobilization and homing are critical to the development of an anti-thrombosis and anti-stenosis tissue-engineered blood vessel. The growth and activation of blood vessels are supported by nerves. We investigated whether nerve growth factors (NGF) can promote EPCs mobilization and endothelialization of tissue-engineered blood vessels. In vitro, NGF promoted EPCs to form more colonies, stimulated human EPCs to differentiate into endothelial cells, and significantly enhanced EPCs migration. Flow cytometric analysis revealed that NGF treatment increased the number of EPCs in the peripheral circulation of C57BL/6 mice. Furthermore, the treatment of human EPCs with NGF facilitated their homing into wire-injured carotid arteries after injection into mice. Decellularized rat blood vessel matrix was incubated with EDC cross-linked collagen and bound to NGF protein using the bifunctional coupling agent N-succinmidyl3-(2-pyridyldit-hio) propionate (SPDP). The NGF-bound tissue-engineered blood vessel was implanted into rat carotid artery for 1 week and 1 month. NGF-bound blood vessels possessed significantly higher levels of endothelialization and patency than controls did. These results demonstrated that NGF can markedly increase EPCs mobilization and homing to vascular grafts. Neurotrophic factors such as NGF have a therapeutic potential for the construction of tissue-engineered blood vessels in vivo.
Collapse
|
170
|
Silvestre JS. [Proangiogenic cell-based therapy for treatment of ischemic diseases]. Med Sci (Paris) 2009; 25:931-8. [PMID: 19951667 DOI: 10.1051/medsci/20092511931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The application of endothelial progenitor cells (EPC) cell-based therapy for regenerative medicine constitutes a promising therapeutic avenue for the treatment of cardiovascular diseases. Based on experimental studies demonstrating that bone marrow-, blood- or tissue-derived stem/progenitor cells improve the functional recovery after ischemia, clinical trials were initiated to address this new therapeutic concept. Although autolougous cell therapy was shown to improve perfusion and function of ischemic tissues, a number of issues remain to be adressed. The nature of the mobilizing, migratory and homing signals, and the mechanisms of action need to be identified and further defined. In addition, strategies to enhance homing, survival and therapeutic potential of EPC need to be developped to improve therapeutic effect and counteract EPC dysfunction in aged patients with cardiovascular risk factors. The present review article will discuss the mechanisms of action of different types of adult stem cells and several approaches to improve their therapeutic efficiency.
Collapse
Affiliation(s)
- Jean-Sébastien Silvestre
- Paris-Cardiovascular Research Center-Inserm U970, Hôpital européen Georges Pompidou, 56, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
171
|
Shmilovich H, Ben-Shoshan J, Tal R, Afek A, Barshack I, Maysel-Auslander S, Harats D, Keren G, George J. B-type natriuretic peptide enhances vasculogenesis by promoting number and functional properties of early endothelial progenitor cells. Tissue Eng Part A 2009; 15:2741-9. [PMID: 19275472 DOI: 10.1089/ten.tea.2008.0414] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To test the hypothesis that B-type natriuretic peptide (BNP) acts as a potent vasculogenic agent by enhancing the number, proliferation, adhesion, and migration of endothelial progenitor cells (EPCs). BACKGROUND BNP is a neurohormonal peptide that predicts outcome and used for treatment in chronic heart failure patients. It has been shown to promote angiogenesis in experimental animals. EPCs have been demonstrated to contribute to postnatal angiogenesis and vasculogenesis. METHODS The number of EPC colony forming units (CFU) and levels of N-terminal ProBNP were assayed in patients with severe, yet controlled, New York Heart Association (NYHA) II-IV heart failure. The in vitro effects of BNP on early EPC-CFU numbers, proliferation, migration, adhesive, and vascular tube formation capacities were studied using human and murine systems. The effects of in vivo BNP administration on Sca-1/Flk-1 progenitors and on vasculogenesis in the hindlimb ischemia model were then assayed in wild-type mice. RESULTS A significant correlation was found between circulating N-terminal ProBNP levels and EPC-CFU numbers. We observed a dose-dependent effect of BNP on the numbers of CFU and proliferation capacity of human EPCs as well as on their adhesive, migratory, and tube formation properties, in vitro. Systemic BNP administration to mice led to a significant increase in bone marrow Sca-1/Flk-1 EPCs and improvement in blood flow and capillary density in the ischemic limbs of mice. CONCLUSIONS BNP promotes vessel growth by increasing the number of endothelial progenitors and enhancing their functional properties. These provasculogenic properties of BNP could account for some of its beneficial effects in chronic heart failure patients and may be harnessed for the purpose of improving collateral formation in ischemic subjects.
Collapse
Affiliation(s)
- Haim Shmilovich
- Department of Cardiology, Tel Aviv Sourasky Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Higuchi T, Anton M, Saraste A, Dumler K, Pelisek J, Nekolla SG, Bengel FM, Schwaiger M. Reporter gene PET for monitoring survival of transplanted endothelial progenitor cells in the rat heart after pretreatment with VEGF and atorvastatin. J Nucl Med 2009; 50:1881-6. [PMID: 19837770 DOI: 10.2967/jnumed.109.067801] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED It has been suggested that vascular endothelial growth factor (VEGF) and statins enhance the survival, proliferation, and function of endothelial progenitor cells (EPCs). We investigated whether reporter gene PET can be used to detect the effects of atorvastatin and VEGF on survival of EPCs after transplantation in the rat heart. METHODS Healthy nude rats received an intramyocardial injection of 4 million human EPCs retrovirally transduced with the sodium/iodide symporter gene for reporter gene imaging. Reporter gene expression was imaged at days 1 and 3 after injection on a small-animal PET scanner with (124)I, and the presence of EPCs was confirmed by immunohistochemistry with human CD31 antibodies. The control group received EPCs transduced only with the reporter gene, whereas treatment groups received oral atorvastatin (10 mg/kg/d) and EPCs cotransduced with adenoviral vectors encoding VEGF in addition to sodium/iodide symporter. RESULTS Immunohistochemistry showed more EPCs at the site of injection after atorvastatin treatment and in the presence of VEGF expression in EPCs than in controls. PET successfully visualized EPCs as focal (124)I accumulation at the site of injection. The quantitative amount of (124)I accumulation assessed by PET was significantly higher in the pretreatment than control group. Autoradiography confirmed (124)I accumulation in the myocardium that correlated with the number of EPCs. CONCLUSION Early survival of transplanted EPCs in the rat myocardium is prolonged by pretreatment with a combination of atorvastatin and VEGF. Reporter gene PET, by successfully quantifying the effect, is an attractive tool for monitoring stem cell survival in vivo.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
173
|
Zisa D, Shabbir A, Suzuki G, Lee T. Vascular endothelial growth factor (VEGF) as a key therapeutic trophic factor in bone marrow mesenchymal stem cell-mediated cardiac repair. Biochem Biophys Res Commun 2009; 390:834-8. [PMID: 19836359 DOI: 10.1016/j.bbrc.2009.10.058] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 10/13/2009] [Indexed: 12/16/2022]
Abstract
We recently demonstrated a novel effective therapeutic regimen for treating hamster heart failure based on injection of bone marrow mesenchymal stem cells (MSCs) or MSC-conditioned medium into the skeletal muscle. The work highlights an important cardiac repair mechanism mediated by the myriad of trophic factors derived from the injected MSCs and local musculature that can be explored for non-invasive stem cell therapy. While this therapeutic regimen provides the ultimate proof that MSC-based cardiac repair is mediated by the trophic actions independent of MSC differentiation or stemness, the trophic factors responsible for cardiac regeneration after MSC therapy remain largely undefined. Toward this aim, we took advantage of the finding that human and porcine MSCs exhibit species-related differences in expression of trophic factors. We demonstrate that human MSCs when compared to porcine MSCs express and secrete 5-fold less vascular endothelial growth factor (VEGF) in conditioned medium (40+/-5 and 225+/-17 pg/ml VEGF, respectively). This deficit in VEGF output was associated with compromised cardiac therapeutic efficacy of human MSC-conditioned medium. Over-expression of VEGF in human MSCs however completely restored the therapeutic potency of the conditioned medium. This finding indicates VEGF as a key therapeutic trophic factor in MSC-mediated myocardial regeneration, and demonstrates the feasibility of human MSC therapy using trophic factor-based cell-free strategies, which can eliminate the concern of potential stem cell transformation.
Collapse
Affiliation(s)
- David Zisa
- Department of Biochemistry and Center for Research in Cardiovascular Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
174
|
Genetic engineering of human stem cells for enhanced angiogenesis using biodegradable polymeric nanoparticles. Proc Natl Acad Sci U S A 2009; 107:3317-22. [PMID: 19805054 DOI: 10.1073/pnas.0905432106] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cells hold great potential as cell-based therapies to promote vascularization and tissue regeneration. However, the use of stem cells alone to promote angiogenesis remains limited because of insufficient expression of angiogenic factors and low cell viability after transplantation. Here, we have developed vascular endothelial growth factor (VEGF) high-expressing, transiently modified stem cells for the purposes of promoting angiogenesis. Nonviral, biodegradable polymeric nanoparticles were developed to deliver hVEGF gene to human mesenchymal stem cells (hMSCs) and human embryonic stem cell-derived cells (hESdCs). Treated stem cells demonstrated markedly enhanced hVEGF production, cell viability, and engraftment into target tissues. S.c. implantation of scaffolds seeded with VEGF-expressing stem cells (hMSCs and hESdCs) led to 2- to 4-fold-higher vessel densities 2 weeks after implantation, compared with control cells or cells transfected with VEGF by using Lipofectamine 2000, a leading commercial reagent. Four weeks after intramuscular injection into mouse ischemic hindlimbs, genetically modified hMSCs substantially enhanced angiogenesis and limb salvage while reducing muscle degeneration and tissue fibrosis. These results indicate that stem cells engineered with biodegradable polymer nanoparticles may be therapeutic tools for vascularizing tissue constructs and treating ischemic disease.
Collapse
|
175
|
Garolla A, D'Incà R, Checchin D, Biagioli A, De Toni L, Nicoletti V, Scarpa M, Bolzonello E, Sturniolo GC, Foresta C. Reduced endothelial progenitor cell number and function in inflammatory bowel disease: a possible link to the pathogenesis. Am J Gastroenterol 2009; 104:2500-7. [PMID: 19568231 DOI: 10.1038/ajg.2009.332] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Circulating endothelial progenitor cells (EPCs) are essential for endothelial repair and vascular healing. Patients with inflammatory bowel disease (IBD) may suffer from endothelial dysfunction. Reduced EPC number, impaired mobilization, or increased EPC apoptosis may be crucial in this phenomenon. The aim of our study was to investigate the number and function of EPCs in patients with IBD and to assess their endothelial function. METHODS In 100 IBD patients (47 ulcerative colitis (UC) and 53 Crohn's disease (CD)) and 50 healthy controls, EPC number, CXC motif receptor 4 (CXCR4) expression, the percentage of apoptotic circulating EPCs, and the number of colony-forming units were evaluated. Endothelial dysfunction was assessed by luteinizing hormone (LH), follicle stimulating hormone (FSH), and testosterone levels, and in a subgroup of patients, brachial artery flow-mediated dilation (FMD) was measured. Kruskal-Wallis ANOVA (analysis of variance), Mann-Whitney U two-tailed, and Spearman's rank correlation tests were used to assess differences. RESULTS EPC number was significantly lower in UC patients (39.6 (95% confidence interval (95% CI): 30.7-48.6)) and in CD patients (43.1 (95% CI: 35.9-50.4)) than in healthy controls (97.1 (95% CI: 88.3-105.9)), (P<0.001). LH and FSH levels and CXCR4 expression on EPCs did not significantly differ from controls. Testosterone concentrations and FMD were lower in UC patients. Number of apoptotic EPCs was higher in both UC and CD patients with an impaired ability to generate colony in vitro. CONCLUSIONS We hypothesize that in IBD patients, apoptosis contributes to the reduction of circulating EPC number and to their ability to proliferate in vitro. As this condition represents a risk factor for cardiovascular disease, endothelial function should be evaluated in these patients.
Collapse
Affiliation(s)
- Andrea Garolla
- Department of Histology, Microbiology, and Medical Biotechnologies, Center for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Yamamoto Y, Matsuura T, Narazaki G, Sugitani M, Tanaka K, Maeda A, Shiota G, Sato K, Yoshida A, Hisatome I. Synergistic effects of autologous cell and hepatocyte growth factor gene therapy for neovascularization in a murine model of hindlimb ischemia. Am J Physiol Heart Circ Physiol 2009; 297:H1329-36. [DOI: 10.1152/ajpheart.00321.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Autologous cell implantation and angiogenic gene therapy have been evaluated in critical limb ischemic patients. Here, we compared the features of these strategies individually and in combination. C57BL/6J mice with ischemic hindlimbs were injected with adherent mononuclear cells (aMNCs) from bone marrow or adenovirus encoding the hepatocyte growth factor (HGF) gene (Ad-HGF). Under comparable angiogenic conditions, 10 × 105 aMNCs produced significantly higher amounts of VEGF and FGF-2 and stimulated the number of arterioles in ischemic muscle compared with 1 × 108 plaque-forming units (pfu) of Ad-HGF. Ad-HGF produced 10 times more HGF in ischemic muscle compared with aMNCs. Injection of 0.3 × 105 aMNCs previously transfected with Ad-HGF (aMNC/Ad-HGF) increased blood flow and elevated the numbers of capillaries and arterioles to levels comparable with that seen with 10 × 105 aMNCs or 1 × 108 pfu of Ad-HGF. Hypoxic conditions induced the apoptotic death of aMNCs. However, coincubation with HGF or aMNC/Ad-HGF protected cells against apoptosis. HGF stimulated the migration of aMNCs, and the migration capacity of the aMNC/Ad-HGF group was significantly higher than that in the aMNC/Ad-LacZ group. In conclusion, cell-based HGF gene therapy decreased the number of cells required for neovascularization. This strategy can be an effective angiogenic therapy.
Collapse
Affiliation(s)
- Yasutaka Yamamoto
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Takashi Matsuura
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Genta Narazaki
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Miyoko Sugitani
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Kohei Tanaka
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Akihiro Maeda
- Division of Molecular Biology, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, and
| | - Goshi Shiota
- Division of Molecular and Genetic Medicine, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Kenzo Sato
- Division of Molecular Biology, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, and
| | - Akio Yoshida
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| |
Collapse
|
177
|
Zisa D, Shabbir A, Mastri M, Suzuki G, Lee T. Intramuscular VEGF repairs the failing heart: role of host-derived growth factors and mobilization of progenitor cells. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1503-15. [PMID: 19759338 DOI: 10.1152/ajpregu.00227.2009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Skeletal muscle produces a myriad of mitogenic factors possessing cardiovascular regulatory effects that can be explored for cardiac repair. Given the reported findings that VEGF may modulate muscle regeneration, we investigated the therapeutic effects of chronic injections of low doses of human recombinant VEGF-A(165) (0.1-1 microg/kg) into the dystrophic hamstring muscle in a hereditary hamster model of heart failure and muscular dystrophy. In vitro, VEGF stimulated proliferation, migration, and growth factor production of cultured C2C12 skeletal myocytes. VEGF also induced production of HGF, IGF2, and VEGF by skeletal muscle. Analysis of skeletal muscle revealed an increase in myocyte nuclear [531 +/- 12 VEGF 1 microg/kg vs. 364 +/- 19 for saline (number/mm(2)) saline] and capillary [591 +/- 80 VEGF 1 microg/kg vs. 342 +/- 21 for saline (number/mm(2))] densities. Skeletal muscle analysis revealed an increase in Ki67(+) nuclei in the VEGF 1 microg/kg group compared with saline. In addition, VEGF mobilized c-kit(+), CD31(+), and CXCR4(+) progenitor cells. Mobilization of progenitor cells was consistent with higher SDF-1 concentrations found in hamstring, plasma, and heart in the VEGF group. Echocardiogram analysis demonstrated improvement in left ventricular ejection fraction (0.60 +/- 0.02 VEGF 1 microg/kg vs. 0.45 +/- 0.01 mm for saline) and an attenuation in ventricular dilation [5.59 +/- 0.12 VEGF 1 microg/kg vs. 6.03 +/- 0.09 for saline (mm)] 5 wk after initiating therapy. Hearts exhibited higher cardiomyocyte nuclear [845 +/- 22 VEGF 1 microg/kg vs. 519 +/- 40 for saline (number/mm(2))] and capillary [2,159 +/- 119 VEGF 1 microg/kg vs. 1,590 +/- 66 for saline (number/mm(2))] densities. Myocardial analysis revealed approximately 2.5 fold increase in Ki67+ cells and approximately 2.8-fold increase in c-kit(+) cells in the VEGF group, which provides evidence for cardiomyocyte regeneration and progenitor cell expansion. This study provides novel evidence of a salutary effect of VEGF in the cardiomyopathic hamster via induction of myogenic growth factor production by skeletal muscle and mobilization of progenitor cells, which resulted in attenuation of cardiomyopathy and repair of the heart.
Collapse
Affiliation(s)
- David Zisa
- Department of Biochemistry and Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York 14214, USA
| | | | | | | | | |
Collapse
|
178
|
Chen J, Song M, Yu S, Gao P, Yu Y, Wang H, Huang L. Advanced glycation endproducts alter functions and promote apoptosis in endothelial progenitor cells through receptor for advanced glycation endproducts mediate overpression of cell oxidant stress. Mol Cell Biochem 2009; 335:137-46. [PMID: 19756958 DOI: 10.1007/s11010-009-0250-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 09/02/2009] [Indexed: 01/09/2023]
Abstract
Endothelial progenitor cells (EPCs) play an important role in preventing atherosclerosis. The factors that regulate the function of EPCs are not completely clear. Increased formation of advanced glycation endproducts (AGEs) is generally regarded as one of the main mechanisms responsible for vascular damage in patients with diabetes and atherosclerosis. AGEs lead to the generation of reactive oxygen species (ROS) and part of the regenerative capacity of EPCs seems to be due to their low baseline ROS levels and reduced sensitivity to ROS-induced cell apoptosis. Therefore, we tested the hypothesis that AGEs can alter functions and promote apoptosis in EPCs through overpress cell oxidant stress. EPCs, isolated from bone marrow, were cultured in the absence or presence of AGEs (50, 100, and 200 microg/ml). A modified Boyden's chamber was used to assess the migration of EPCs and the number of recultured EPCs was counted to measure the adhesiveness function. MTT assay was used to determine the proliferation function. ROS were analyzed using the ROS assay kit. A spectrophotometer was used to assess superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) activity, and PCR was used to test mRNA expression of SOD and GSH-PX. SiRNA was used to block receptor for advanced glycation endproducts (RAGEs) expression. Apoptosis was evaluated by Annexin V immunostaining and TUNEL staining. Co-culturing with AGEs increases ROS production, decreases anti-oxidant defenses, overpresses oxidant stress, inhibits the proliferation, migration, and adhesion of EPCs, and induces EPCs apoptosis. In addition, these effects were attenuated during block RAGE protein expression by siRNA. AGEs may serve to impair EPCs functions through RAGE-mediate oxidant stress, and promote EPCs sensitivity toward oxidative-stress-mediated apoptosis, which indicates a new pathophysiological mechanism of disturbed vascular adaptation in atherosclerosis and suggests that lower levels of AGEs might improve the success of progenitor cell therapy.
Collapse
Affiliation(s)
- Jianfei Chen
- Department of Cardiology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
179
|
Park SJ, Baek SH, Oh MK, Choi SH, Park EH, Kim NH, Shin JC, Kim IS. Enhancement of angiogenic and vasculogenic potential of endothelial progenitor cells by haptoglobin. FEBS Lett 2009; 583:3235-40. [PMID: 19751729 DOI: 10.1016/j.febslet.2009.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 10/20/2022]
Abstract
Endothelial progenitor cells (EPCs) were transfected with the haptoglobin (Hp) gene to investigate the effect of Hp on cell function. Hp potentiated the gene expression of various pro-angiogenic factors in the EPCs. The Hp-modified EPCs also increased in vitro tube formation on Matrigel compared with control cells. In hindlimb ischaemia models, Hp-EPCs showed a greater ability for improving blood perfusion and recovery from ischaemic injury. These results indicate that Hp improves EPC function in neovasculogenesis, which suggests that ex vivo modification of EPCs with the Hp gene can be applied to the treatment of vascular damage.
Collapse
Affiliation(s)
- Seon-Joo Park
- Department of Natural Sciences, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
180
|
Fujinaga H, Baker CD, Ryan SL, Markham NE, Seedorf GJ, Balasubramaniam V, Abman SH. Hyperoxia disrupts vascular endothelial growth factor-nitric oxide signaling and decreases growth of endothelial colony-forming cells from preterm infants. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1160-9. [PMID: 19734318 DOI: 10.1152/ajplung.00234.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Exposure of preterm infants to hyperoxia impairs vascular growth, contributing to the development of bronchopulmonary dysplasia and retinopathy of prematurity. Disruption of vascular endothelial growth factor (VEGF)-nitric oxide (NO) signaling impairs vascular growth. Endothelial progenitor cells (EPCs) may play an important role in vascular growth. Endothelial colony-forming cells (ECFCs), a type of EPC, from human preterm cord blood are more susceptible to hyperoxia-induced growth impairment than term ECFCs. Therefore, we hypothesized that hyperoxia disrupts VEGF-NO signaling and impairs growth in preterm ECFCs and that exogenous VEGF or NO preserves growth in hyperoxia. Growth kinetics of preterm cord blood-derived ECFCs (gestational ages, 27-34 wk) were assessed in room air (RA) and hyperoxia (40-50% oxygen) with or without VEGF, NO, or N(omega)-nitro-l-arginine. VEGF, VEGF receptor-2 (VEGFR-2), and endothelial NO synthase (eNOS) protein expression and NO production were compared. Compared with RA controls, hyperoxia significantly decreased growth, VEGFR-2 and eNOS expression, and NO production. VEGF treatment restored growth in hyperoxia to values measured in RA controls and significantly increased eNOS expression in hyperoxia. NO treatment also increased growth in hyperoxia. N(omega)-nitro-l-arginine treatment inhibited VEGF-augmented growth in RA and hyperoxia. We conclude that hyperoxia decreases growth and disrupts VEGF-NO signaling in human preterm ECFCs. VEGF treatment restores growth in hyperoxia by increasing NO production. NO treatment also increases growth during hyperoxia. Exogenous VEGF or NO may protect preterm ECFCs from the adverse effects of hyperoxia and preservation of ECFC function may improve outcomes of preterm infants.
Collapse
Affiliation(s)
- Hideshi Fujinaga
- Pediatric Heart Lung Center. Univ. of Colorado Denver School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
181
|
Mechanism of Sustained Release of Vascular Endothelial Growth Factor in Accelerating Experimental Diabetic Healing. J Invest Dermatol 2009; 129:2275-87. [DOI: 10.1038/jid.2009.26] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
182
|
Koga M, Sudo R, Abe Y, Yamamoto K, Ando J, Ikeda M, Tanishita K. Contribution of Rat Endothelial Progenitor Cells on Three-Dimensional Network FormationIn Vitro. Tissue Eng Part A 2009; 15:2727-39. [DOI: 10.1089/ten.tea.2008.0417] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Masaki Koga
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Yoshinori Abe
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Kimiko Yamamoto
- Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Joji Ando
- Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mariko Ikeda
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Kazuo Tanishita
- Department of System Design Engineering, Keio University, Yokohama, Japan
| |
Collapse
|
183
|
Sander AL, Jakob H, Henrich D, Powerski M, Witt H, Dimmeler S, Barker J, Marzi I, Frank J. Systemic transplantation of progenitor cells accelerates wound epithelialization and neovascularization in the hairless mouse ear wound model. J Surg Res 2009; 165:165-70. [PMID: 20036386 DOI: 10.1016/j.jss.2009.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 06/11/2009] [Accepted: 07/02/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND Impaired wound healing due to local injury, infection, or systemic diseases, such as diabetes, is a major clinical problem. Recent studies have shown that endothelial progenitor cells (EPC) isolated from peripheral blood, bone marrow, as well as the spleen accumulate in granulation tissue at the site of neovascularization, causing secretion of growth factors and cytokines and thus accelerating wound healing. MATERIALS AND METHODS In the present study, we transplanted systemic EPC and then measured epithelialization and neovascularization in the hairless mouse ear wound model. RESULTS Systemic EPC transplantation significantly accelerated epithelialization and neovascularization compared with control wounds receiving phosphate-buffered saline without calcium and magnesium (PBS). The EPC group had significantly higher vascular density than did the PBS-treated group as determined by immunohistochemistry for CD31 and CD90. Fluorescence microscopy revealed accumulation "homing" of the transplanted EPC at the sites of neovascularization in the granulation tissue throughout healing. Furthermore, transplantation of EPC also increased the expression of the angiogenic cytokine stromal cell-derived factor 1α (SDF1α). CONCLUSIONS This appears to be the first demonstration of EPC recruitment to the site of wound neovascularization throughout the healing process. These findings demonstrate that transplanting systemic EPC into "normal" healing wounds promotes epithelialization and neovascularization and thus could be an useful method for accelerating wound healing.
Collapse
Affiliation(s)
- Anna L Sander
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University, Frankfurt am Main, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Effects of insulin resistance on endothelial progenitor cells and vascular repair. Clin Sci (Lond) 2009; 117:173-90. [PMID: 19630751 DOI: 10.1042/cs20080263] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Insulin resistance, a key feature of obesity, the metabolic syndrome and Type 2 diabetes mellitus, results in an array of metabolic and vascular phenomena which ultimately promote the development of atherosclerosis. Endothelial dysfunction is intricately related to insulin resistance through the parallel stimulatory effects of insulin on glucose disposal in metabolic tissues and NO production in the endothelium. Perturbations characteristic of insulin resistance, including dyslipidaemia, inflammation and oxidative stress, may jeopardize the structural or functional integrity of the endothelium. Recent evidence suggests that endothelial damage is mitigated by endogenous reparative processes which mediate endothelial regeneration. EPCs (endothelial progenitor cells) are circulating cells which have been identified as mediators of endothelial repair. Several of the abnormalities associated with insulin resistance, including reduced NO bioavailability, increased production of ROS (reactive oxygen species) and down-regulation of intracellular signalling pathways, have the potential to disrupt EPC function. Improvement in the number and function of EPCs may contribute to the protective actions of evidence-based therapies to reduce cardiometabolic risk. In the present article, we review the putative effects of insulin resistance on EPCs, discuss the underlying mechanisms and highlight potential therapeutic manoeuvres which could improve vascular repair in individuals with insulin resistance.
Collapse
|
185
|
|
186
|
Yu JX, Huang XF, Lv WM, Ye CS, Peng XZ, Zhang H, Xiao LB, Wang SM. Combination of stromal-derived factor-1alpha and vascular endothelial growth factor gene-modified endothelial progenitor cells is more effective for ischemic neovascularization. J Vasc Surg 2009; 50:608-16. [PMID: 19595531 DOI: 10.1016/j.jvs.2009.05.049] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 05/13/2009] [Accepted: 05/13/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Recruitment and entrapment of bone marrow-derived endothelial progenitor cells (EPCs) is important in vascular endothelial growth factor (VEGF)-induced angiogenesis. EPC mobilization and differentiation are modulated by stromal-derived factor-1alpha (SDF-1alpha/CXCL12), another important chemokine. In this study, we investigated the hypothesis that SDF-1alpha and VEGF might act synergistically on EPC-mediated vasculogenesis. METHODS EPCs were isolated and cultured from human peripheral blood, then transduced with retroviral vectors pBabe containing human VEGF(165) complimentary DNA (Td/V-EPCs) and pBabe wild-type (Td/p-EPCs). EPC migration activity was investigated with a modified Boyden chamber assay. EPC apoptosis induced by serum starvation was studied by annexin V assays. The combined effect of local administration of SDF-1alpha and Td/V-EPC transplantation on neovascularization was investigated in a murine model of hind limb ischemia. RESULTS Over-expression of hVEGF(165) increased SDF-1alpha-mediated EPC migration. SDF-1alpha-mediated migration was significantly increased when EPCs were modified with VEGF (Td/V-EPCs) vs when VEGF was not present (Td/p-EPCs) or when VEGF alone was present (Td/V-EPCs; 196.8 +/- 15.2, 81.2 +/- 9.8, and 67.4 +/- 7.4/mm(2), respectively P < .001). SDF-1alpha combined with VEGF reduced serum starvation-induced apoptosis of EPCs more than SDF-1alpha or VEGF alone (P < .001). To determine the effect of this combination in vivo, SDF-1alpha was locally injected alone into the ischemic hind limb muscle of nude mice or combined with systemically injected Td/V-EPCs. The SDF-1alpha plus VEGF group showed significantly increased local accumulation of EPCs, blood-flow recovery, and capillary density compared with the other groups. The ratio of ischemic/normal blood flow in Td/V-EPCs plus SDF-1alpha group was significantly higher (P < .01), as was capillary density (capillaries/mm(2)), an index of neovascularization (Td/V-EPCs plus SDF-1alpha group, 863 +/- 31; no treatment, 395 +/-13; SDF-1alpha, 520 +/- 29; Td/p-EPCs, 448 +/- 28; Td/p-EPCs plus SDF-1alpha, 620 +/- 29; Td/V-EPCs, 570 +/- 30; P < .01). To investigate a possible mechanistic basis, we showed that VEGF up-regulated the receptor for SDF-1alpha, CXCR4, on EPCs in vitro. CONCLUSION The combination of SDF-1alpha and VEGF greatly increases EPC-mediated angiogenesis. The use VEGF and SDF-1alpha together, rather than alone, will be a novel and efficient angiogenesis strategy to provide therapeutic neovascularization.
Collapse
Affiliation(s)
- Jian-Xing Yu
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Wahl P, Brixius K, Bloch W. Exercise‐induced stem cell activation and its implication for cardiovascular and skeletal muscle regeneration. MINIM INVASIV THER 2009; 17:91-9. [DOI: 10.1080/13645700801969816] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
188
|
|
189
|
Kealy B, Liew A, McMahon JM, Ritter T, O'Doherty A, Hoare M, Greiser U, Vaughan EE, Maenz M, O'Shea C, Barry F, O'Brien T. Comparison of Viral and Nonviral Vectors for Gene Transfer to Human Endothelial Progenitor Cells. Tissue Eng Part C Methods 2009; 15:223-31. [DOI: 10.1089/ten.tec.2008.0323] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Brian Kealy
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Aaron Liew
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Jill M. McMahon
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Aideen O'Doherty
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Melissa Hoare
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Udo Greiser
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Erin E. Vaughan
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Martin Maenz
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Ciara O'Shea
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Frank Barry
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science & Department of Medicine, National University of Ireland, Galway, Republic of Ireland
| |
Collapse
|
190
|
Chen SY, Wang F, Yan XY, Zhou Q, Ling Q, Ling JX, Rong YZ, Li YG. Autologous transplantation of EPCs encoding FGF1 gene promotes neovascularization in a porcine model of chronic myocardial ischemia. Int J Cardiol 2009; 135:223-32. [DOI: 10.1016/j.ijcard.2008.12.193] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Revised: 11/19/2008] [Accepted: 12/13/2008] [Indexed: 01/08/2023]
|
191
|
Genetic engineering with endothelial nitric oxide synthase improves functional properties of endothelial progenitor cells from patients with coronary artery disease: an in vitro study. Basic Res Cardiol 2009; 104:739-49. [DOI: 10.1007/s00395-009-0039-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/07/2009] [Accepted: 05/13/2009] [Indexed: 12/23/2022]
|
192
|
Bhatwadekar AD, Glenn JV, Curtis TM, Grant MB, Stitt AW, Gardiner TA. Retinal endothelial cell apoptosis stimulates recruitment of endothelial progenitor cells. Invest Ophthalmol Vis Sci 2009; 50:4967-73. [PMID: 19474402 DOI: 10.1167/iovs.09-3616] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Bone marrow-derived endothelial progenitor cells (EPCs) contribute to vascular repair although it is uncertain how local endothelial cell apoptosis influences their reparative function. This study was conducted to determine how the presence of apoptotic bodies at sites of endothelial damage may influence participation of EPCs in retinal microvascular repair. METHODS Microlesions of apoptotic cell death were created in monolayers of retinal microvascular endothelial cells (RMECs) by using the photodynamic drug verteporfin. The adhesion of early-EPCs to these lesions was studied before detachment of the apoptotic cells or after their removal from the wound site. Apoptotic bodies were fed to normal RMECs and mRNA levels for adhesion molecules were analyzed. RESULTS Endothelial lesions where apoptotic bodies were left attached at the wound site showed a fivefold enhancement in EPC recruitment (P < 0.05) compared with lesions where the apoptotic cells had been removed. In intact RMEC monolayers exposed to apoptotic bodies, expression of ICAM, VCAM, and E-selectin was upregulated by 5- to 15-fold (P < 0.05-0.001). EPCs showed a characteristic chemotactic response (P < 0.05) to conditioned medium obtained from apoptotic bodies, whereas analysis of the medium showed significantly increased levels of VEGF, IL-8, IL-6, and TNF-alpha when compared to control medium; SDF-1 remained unchanged. CONCLUSIONS The data indicate that apoptotic bodies derived from retinal capillary endothelium mediate release of proangiogenic cytokines and chemokines and induce adhesion molecule expression in a manner that facilitates EPC recruitment.
Collapse
Affiliation(s)
- Ashay D Bhatwadekar
- Centre for Vision Science, Queen's University Belfast, Royal Victoria Hospital, Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | |
Collapse
|
193
|
Ruifrok WPT, de Boer RA, Iwakura A, Silver M, Kusano K, Tio RA, Losordo DW. Estradiol-induced, endothelial progenitor cell-mediated neovascularization in male mice with hind-limb ischemia. Vasc Med 2009; 14:29-36. [PMID: 19144777 DOI: 10.1177/1358863x08096666] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated whether administration of estradiol to male mice augments mobilization of bone marrow-derived endothelial progenitor cells (EPC) and incorporation into foci of neovascularization after hind-limb ischemia, thereby contributing to blood flow restoration. Mice were randomized and implanted with placebo pellets or pellets containing low-dose estradiol (0.39 mg) or high-dose estradiol (1.7 mg). Hind-limb ischemia was induced by unilateral resection of the left femoral artery 1 week after pellet implantation, then EPC mobilization and functional recovery was evaluated. EPC recruitment was assessed in mice transplanted with bone marrow from transgenic donors expressing beta-galactosidase driven by the Tie-2 promoter. EPC culture assay performed 2 weeks after pellet implantation revealed a significantly greater (p<0.05) number of circulating EPCs in the high-dose estradiol group than in the low-dose estradiol and placebo groups. At 3 and 4 weeks after induction of hind-limb ischemia, perfusion was significantly greater (p<0.05) in high-dose estradiol mice than in mice implanted with the low-dose estradiol or placebo pellets. At 1 and 4 weeks after hind-limb ischemia surgery, more bone marrow-derived EPCs, identified as beta-galactosidase-positive cells, were observed in ischemic regions from high-dose estradiol animals than in low-dose (p<0.05) or placebo groups (p<0.05). These results indicate that estradiol dose-dependently increases the levels of EPCs in peripheral blood in male animals, improves the recovery of blood flow, and decreases limb necrosis after hind-limb ischemia, and that this enhancement occurs, in part, through augmentation of EPC mobilization and greater incorporation of bone marrow-derived EPCs into foci of neovascularization.
Collapse
Affiliation(s)
- Willem-Peter T Ruifrok
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
194
|
Zeoli A, Dentelli P, Brizzi MF. Endothelial progenitor cells and their potential clinical implication in cardiovascular disorders. J Endocrinol Invest 2009; 32:370-82. [PMID: 19636208 DOI: 10.1007/bf03345729] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Risk factors associated with cardiovascular diseases reduce the availability of endothelial progenitor cells (EPC) by affecting their mobilization and integration into injured vascular sites. The existence of a bone marrow reservoir of EPC has attracted interest, especially as target for therapeutic intervention in different pathological settings. Among the cardiovascular risk factors, hypertension has been shown to be a strongest predictor of EPC migratory impairment. However, at present, data concerning EPC biology are still limited. In this article we provide an overview of data relevant to their potential clinical implications in cardiovascular disorders. In addition, the recent advances in understanding the role of EPC in the pathophysiology of hypertension are discussed.
Collapse
Affiliation(s)
- A Zeoli
- Department of Internal Medicine, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | | | | |
Collapse
|
195
|
Protective roles of HDL, apoA-I and mimetic peptide on endothelial function: Through endothelial cells and endothelial progenitor cells. Int J Cardiol 2009; 133:286-92. [DOI: 10.1016/j.ijcard.2008.11.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 11/08/2008] [Indexed: 12/21/2022]
|
196
|
Sekiguchi H, Ii M, Losordo DW. The relative potency and safety of endothelial progenitor cells and unselected mononuclear cells for recovery from myocardial infarction and ischemia. J Cell Physiol 2009; 219:235-42. [PMID: 19115244 DOI: 10.1002/jcp.21672] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endothelial progenitor cells (EPCs) are a subset of the total mononuclear cell population (tMNCs) that possess an enhanced potential for differentiation within the endothelial-cell lineage. Typically, EPCs are selected from tMNCs via the expression of both hematopoietic stem-cell markers and endothelial-cell markers, such as CD34, or by culturing tMNCs in media selective for endothelial cells. Both EPCs and tMNCs participate in vascular growth and regeneration, and their potential use for treatment of myocardial injury or disease has been evaluated in early-phase clinical studies. Direct comparisons between EPCs and tMNCs are rare, but the available evidence appears to favor EPCs, particularly CD34+ cells, and the potency of EPCs may be increased as much as 30-fold through genetic modification. However, these observations must be interpreted with caution because clinical investigations of EPC therapy are ongoing. We anticipate that with continued development, EPC therapy will become a safe and effective treatment option for patients with acute myocardial infarction or chronic ischemic disease.
Collapse
Affiliation(s)
- Haruki Sekiguchi
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
197
|
Gulati R, Simari RD. Defining the potential for cell therapy for vascular disease using animal models. Dis Model Mech 2009; 2:130-7. [PMID: 19259386 PMCID: PMC2650189 DOI: 10.1242/dmm.000562] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell-based therapeutics are currently being developed for a wide array of unmet medical needs. As obstructive vascular disease is the major cause of mortality in the world, cell-based strategies aimed at developing novel therapies or improving current therapies are currently under study. These studies are based on the evolving understanding of the biology of vascular progenitor cells, which has in turn led to the availability of well-defined sources of vascular cells for delivery. Crucial to the development of these approaches is the preclinical testing of cell delivery in animal models. This review highlights the crucial steps involved in the selection of cell sources and generation, delivery approaches, animal models to be used, and endpoints to be studied, in the context of cell delivery for vascular disease. Furthermore, the development of cell delivery to induce angiogenesis in ischemic limbs and to improve the response to large vessel injury will be discussed.
Collapse
Affiliation(s)
- Rajiv Gulati
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert D. Simari
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
198
|
Murasawa S, Asahara T. Cardiogenic potential of endothelial progenitor cells. Ther Adv Cardiovasc Dis 2009; 2:341-8. [PMID: 19124432 DOI: 10.1177/1753944708096283] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Transplantation of endothelial progenitor cells (EPCs) are one of the promising strategies to recover the capillary flow in ischaemic diseases such as ischaemic heart disease and peripheral artery disease (PAD) in the leg. However, our previous and another group's works suggested the scarcity of the number of EPCs in peripheral blood might cause insufficient effect for the ischaemic diseases. There are several strategies to overcome this issue, such as (1) in vivo EPC expansion; (2) ex vivo EPC expansion; (3) local (not systemic) EPC injection; and (4) modification of EPC by gene transfer. Recent publications from our own and other groups have reported the possibility of cardiogenic potential of EPCs. We would like to focus on the strategies of EPC transplantation and cardiomyogenesis of EPCs in this review.
Collapse
Affiliation(s)
- Satoshi Murasawa
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Japan.
| | | |
Collapse
|
199
|
Möbius-Winkler S, Höllriegel R, Schuler G, Adams V. Endothelial progenitor cells: implications for cardiovascular disease. Cytometry A 2009; 75:25-37. [PMID: 19009636 DOI: 10.1002/cyto.a.20669] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial progenitor cells (EPCs) reside in the bone marrow and are mobilized into the circulation by specific stimuli such as certain drugs, ischemia, and exercise training. Once in the circulation EPCs are thought to participate in the maintenance of the endothelial cell layer. Recently it was clearly demonstrated that the amount and function of EPCs is significantly impaired in different cardiovascular diseases. Furthermore, the level of circulating EPCs predicts the occurrence of cardiovascular events and death from cardiovascular causes and may help to identify patients at increased cardiovascular risk. After demonstrating the beneficial effect of applied EPCs in several animal experiments, these cells were also used to treat humans with different cardiovascular diseases. This review will focus on the characterization and liberation of EPCs from the bone marrow, as well as on the most important clinical cardiovascular diseases for which EPCs were used therapeutically.
Collapse
Affiliation(s)
- Sven Möbius-Winkler
- Department of Cardiology and Internal Medicine, University Leipzig - Heart Center Leipzig, Leipzig, Germany
| | | | | | | |
Collapse
|
200
|
Khoo CP, Pozzilli P, Alison MR. Endothelial progenitor cells and their potential therapeutic applications. Regen Med 2009; 3:863-76. [PMID: 18947309 DOI: 10.2217/17460751.3.6.863] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are derived from the bone marrow (BM) and peripheral blood (PB), contributing to tissue repair in various pathological conditions via the formation of new blood vessels, that is, neovascularization. EPCs can be mobilized into the circulation in response to growth factors and cytokines released following stimuli such as vascular trauma, wounding and cancer. EPCs are involved in vasculogenesis during embryogenesis, but are now recognized to have a significant bearing upon disease outcome through their contribution to neovascularization in a variety of pathological states in adulthood. EPCs exist in very small numbers, especially in circulating blood in adults where they only account for 0.01% of all cells. We discuss the contribution and potential therapeutic applications of EPCs in disease, also noting the prognostic value of PB EPC numbers, especially in heart disease and cancer.
Collapse
Affiliation(s)
- Cheen P Khoo
- ICMS, Centre for Diabetes & Metabolic Medicine (DMM), Barts & The London School of Medicine & Dentistry, Queen Mary University of London, 4 Newark Street, London E12AT, UK.
| | | | | |
Collapse
|