251
|
Taniguchi E, Kin M, Torimura T, Nakamura T, Kumemura H, Hanada S, Hisamoto T, Yoshida T, Kawaguchi T, Baba S, Maeyama M, Koga H, Harada M, Kumashiro R, Ueno T, Mizuno S, Ikeda H, Imaizumi T, Murohara T, Sata M. Endothelial progenitor cell transplantation improves the survival following liver injury in mice. Gastroenterology 2006; 130:521-31. [PMID: 16472604 DOI: 10.1053/j.gastro.2005.10.050] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Accepted: 10/19/2005] [Indexed: 12/28/2022]
Abstract
BACKGROUND & AIMS Neovascularization, which is vital to the healing of injured tissues, recently has been found to include both angiogenesis, which involves in mature endothelial cells, and vasculogenesis, involving endothelial progenitor cells. The aim of this study was to clarify the possible roles of endothelial progenitor cells during postnatal liver regeneration. METHODS To determine how endothelial progenitor cells participate in liver regeneration, human or mouse endothelial progenitor cells were transplanted into the mice with carbon tetrachloride-induced acute liver injury. Survival rate of the mice in endothelial progenitor cell-transplanted and control groups was calculated. Separately, livers removed temporally from both groups were examined. RESULTS At an early stage, transplanted human endothelial progenitor cells were seen mainly surrounding hepatic central veins where hepatocytes showed extensive necrosis; later, the transplanted cells formed tubular structures. More of these cells were observed along hepatic sinusoids. Transplantation of human or mouse endothelial progenitor cells improved survival of the mice following liver injury (from 28.6% to 85.7%, P < .0005 and from 33.3% to 80.0%, P < .001, respectively), accompanied by greater proliferation of hepatocytes. Human endothelial progenitor cells produced several growth factors, such as hepatocyte growth factor, transforming growth factor-alpha, heparin-binding epidermal growth factor-like growth factor, and vascular endothelial growth factor, and also elicited endogenous growth factors. CONCLUSIONS Endogenous and exogenous growth factors and direct neovascularization after endothelial progenitor cell transplantation promoted liver regeneration, thus improving survival after liver injury. Transplantation of endothelial progenitor cells could represent a new therapeutic strategy for promoting liver regeneration.
Collapse
Affiliation(s)
- Eitaro Taniguchi
- Second Department of Medicine, Kurume University School of Medicine, Liver Cancer Division, Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Chen MC, Yip HK, Chen CJ, Yang CH, Wu CJ, Cheng CI, Chen YH, Chai HT, Lee CP, Chang HW. No Age-Related Change in Circulating Endothelial Progenitor Cells in Healthy Subjects. Int Heart J 2006; 47:95-105. [PMID: 16479045 DOI: 10.1536/ihj.47.95] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Endothelial progenitor cells were isolated from peripheral blood obtained from 32 healthy volunteers without cardiovascular risk factors who ranged in age from 20 to 61 years (mean [+/- SD] age, 34.1 +/- 9.6 years). The fractions of CD34(+) endothelial progenitor cells expressing kinase insert domain receptor-1, CD62E, or CD31 were analyzed with flow cytometry. Correlation analysis demonstrated that there was no significant correlation between subject age and the fraction of circulating CD34(+) mononuclear cells expressing kinase insert domain receptor-1 (P = 0.324; r = -0.180). Similarly, there was no significant correlation between subject age and the fraction of circulating CD34(+) mononuclear cells expressing CD62E (P = 0.496; r = -0.125) or the fraction of circulating CD34(+) mononuclear cells expressing CD31 (P = 0.245; r = -0.212). In conclusion, the experimental results showed that there was no age-related change in the basal level of circulating endothelial progenitor cells in healthy subjects without cardiovascular risk factors.
Collapse
Affiliation(s)
- Mien-Cheng Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital Kaohsiung, Kaohsiung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
253
|
Clancy RM. Preventing and repairing vascular damage in scleroderma: Should we focus beyond vasodilatation to recruitment of endothelial precursor cells? ACTA ACUST UNITED AC 2006; 54:1730-2. [PMID: 16729284 DOI: 10.1002/art.21900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
254
|
Mohri T, Fujio Y, Maeda M, Ito T, Iwakura T, Oshima Y, Uozumi Y, Segawa M, Yamamoto H, Kishimoto T, Azuma J. Leukemia inhibitory factor induces endothelial differentiation in cardiac stem cells. J Biol Chem 2005; 281:6442-7. [PMID: 16407199 DOI: 10.1074/jbc.m508969200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The importance of interleukin 6 (IL-6)-related cytokines in cardiac homeostasis has been studied extensively; however, little is known about their biological significance in cardiac stem cells. Here we describe that leukemia inhibitory factor (LIF), a member of IL-6-related cytokines, activated STAT3 and ERK1/2 in cardiac Sca-1+ stem cells. LIF stimulation resulted in the induction of endothelial cell-specific genes, including VE-cadherin, Flk-1, and CD31, whereas neither smooth muscle nor cardiac muscle marker genes such as GATA4, GATA6, Nkx-2.5, and calponin were up-regulated. Immunocytochemical examination showed that about 25% of total cells were positively stained with anti-CD31 antibody 14 days after LIF stimulation. Immunofluorescent microscopic analyses identified the Sca-1+ cells that were also positively stained with anti-von Willebrand factor antibody, indicating the differentiating process of Sca-1+ cells into the endothelial cells. IL-6, which did not activate STAT3 and ERK1/2, failed to induce the differentiation of cardiac stem cells into the endothelial cells. In cardiac stem cells, the transduction with dominant negative STAT3 abrogated the LIF-induced endothelial differentiation. And the inhibition of ERK1/2 with the MEK1/2 inhibitor U0126 also prevented the differentiation of Sca-1+ cells into endothelial cells. Thus, both STAT3 and ERK1/2 are required for LIF-mediated endothelial differentiation in cardiac stem cells. Collectively, it is proposed that LIF regulates the commitment of cardiac stem cells into the endothelial cell lineage, contributing to neovascularization in the process of tissue remodeling and/or regeneration.
Collapse
Affiliation(s)
- Tomomi Mohri
- Department of Clinical Evaluation of Medicines and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
255
|
Riha GM, Lin PH, Lumsden AB, Yao Q, Chen C. Review: application of stem cells for vascular tissue engineering. ACTA ACUST UNITED AC 2005; 11:1535-52. [PMID: 16259608 DOI: 10.1089/ten.2005.11.1535] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As the prevalence of vascular disease has continued to expand, the need for a suitable arterial replacement has prompted researchers to look beyond synthetic and autologous grafts toward the field of tissue engineering. Advances in vascular tissue engineering have utilized both mesenchymal and hematopoietic stem cells as a cell source in an attempt to create a fully engineered small-diameter graft. Stem cells offer enormous potential as a cell source because of their proliferative and growth potential, and the application of stem cell technology has far-reaching implications for future applications. The innovative use of stem cells for vascular tissue engineering has opened new possibilities for a fully engineered blood vessel. The purpose of this review is to summarize the current perspective on the use of stem cells for vascular tissue engineering. It focuses principally on the classes of stem cells used, techniques for differentiation scaffolding technology, and the successes and failures of models.
Collapse
Affiliation(s)
- Gordon M Riha
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
256
|
Okazaki T, Ebihara S, Asada M, Kanda A, Sasaki H, Yamaya M. Granulocyte colony-stimulating factor promotes tumor angiogenesis via increasing circulating endothelial progenitor cells and Gr1+CD11b+ cells in cancer animal models. Int Immunol 2005; 18:1-9. [PMID: 16352631 DOI: 10.1093/intimm/dxh334] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recombinant granulocyte colony-stimulating factor (G-CSF) is used for cancer patients with myelosuppression induced by chemotherapy. G-CSF has been reported to progress tumor growth and angiogenesis, but the precise mechanism of tumor angiogenesis activated by G-CSF has not been fully clarified. N-terminal-mutated recombinant human G-CSF administration increased WBCs and neutrophils in peripheral blood and reduced bone marrow stromal cell-derived factor-1 in mice, indicating its biological relevance. Mice were inoculated with Lewis lung carcinoma cells (LLCs) or KLN205 cells and treated with G-CSF. G-CSF accelerated tumor growth and intratumoral vessel density, while it did not accelerate proliferation of LLCs, KLN205 cells or human umbilical vein endothelial cells in vitro. In the absence of tumors, G-CSF did not increase circulating cells that displayed phenotypic characteristics of endothelial progenitor cells (EPCs). In the presence of tumors, G-CSF increased circulating EPCs. In addition, G-CSF treatment increased immune suppressor and endothelial cell-differentiating Gr1+CD11b+ cells in tumor-bearing mice. We conclude that G-CSF promotes tumor growth by activating tumor angiogenesis via increasing circulating EPCs and Gr1+CD11b+ cells in cancer animal models.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- CD11b Antigen/metabolism
- Carcinoma, Lewis Lung/blood
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Granulocyte Colony-Stimulating Factor/administration & dosage
- Granulocyte Colony-Stimulating Factor/adverse effects
- Humans
- Leukocyte Count
- Male
- Mice
- Myelopoiesis/drug effects
- Neovascularization, Pathologic/blood
- Neovascularization, Pathologic/chemically induced
- Neovascularization, Pathologic/pathology
- Receptors, Chemokine/metabolism
- Recombinant Proteins
- Stem Cells/metabolism
- Stem Cells/pathology
- Umbilical Veins/cytology
- Umbilical Veins/metabolism
Collapse
Affiliation(s)
- Tatsuma Okazaki
- Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Seiryo-machi 1-1, Aoba-ku, Sendai 980-8574, Japan.
| | | | | | | | | | | |
Collapse
|
257
|
Waltham M, Burnand K, Fenske C, Modarai B, Humphries J, Smith A. Vascular endothelial growth factor naked DNA gene transfer enhances thrombus recanalization and resolution. J Vasc Surg 2005; 42:1183-9. [PMID: 16376212 DOI: 10.1016/j.jvs.2005.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Accepted: 07/02/2005] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Enhancing thrombus resolution may reduce the long-term complications of venous thrombosis. The aim of this study was to examine whether a sustained release of vascular endothelial growth factor (VEGF) would further improve thrombus recanalization. METHODS Inferior caval vein thrombosis was induced in a cohort of 21 male Wistar rats. A plasmid encoding the human VEGF gene (phVEGF) was injected directly into thrombus (30 to 50 microg) and the muscle adjacent to the inferior vena cava (300 to 400 microg). A plasmid containing the gene encoding beta-galactosidase (pCMVbeta) was injected into the same sites of a separate cohort of rats to act as a control. Tissues were harvested after 1 and 2 weeks, and beta-galactosidase activity was measured to estimate transfection efficiency. Muscle and serum VEGF were measured by enzyme-inked immunosorbent assay. Thrombus size, recanalization, and organization were determined by computer-assisted image analysis. RESULTS The efficiency of control plasmid transfection into muscle was about 1%. No serum hVEGF was detected in phVEGF- or pCMVbeta-treated animals. Significantly raised levels of hVEGF (P < .01) were detected in the muscle injected with phVEGF after 2 weeks compared with control muscle. There was a significant reduction in thrombus size of 23% (P < .05) and 48% (P < .001) in phVEGF-treated animals compared with pCMVbeta-treated controls after 1 and 2 weeks, respectively. Thrombus recanalization was a significantly greater in the phVEGF-treated group after 2 weeks (mean 19% +/- 2% [SEM]) compared with controls (mean 13% +/- 2%, P < .01). There were no differences in the thrombus organization score. CONCLUSION VEGF gene therapy of venous thrombus resulted in smaller thrombi with greater recanalization. Angiogenic gene therapy may form the basis of a novel treatment that may improve the resolution of venous thrombi. CLINICAL RELEVANCE Deep vein thrombosis may lead to residual venous obstruction or reflux and result in post-thrombotic complications that are debilitating and have a substantial socioeconomic impact. Enhancing the resolution of venous thrombi may reduce post thrombotic complications.
Collapse
Affiliation(s)
- Matthew Waltham
- Cardiovascular Division, Academic Department of Surgery, GKT, King's College, St Thomas' Campus, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
258
|
Abstract
The prognosis for the majority of patients with lung cancer remains poor, and treatment strategies including newer generation chemotherapeutics have not improved survival. New approaches are required to further improve patient outcome and survival. Recently, key molecules involved in signal transduction pathways that contribute to tumor growth have been identified as therapeutic targets, particularly molecules involved in cellular proliferation and angiogenesis. Novel therapeutics that specifically target angiogenesis have shown promise as single agents and in combination with standard chemotherapy. The results of recent studies validate the use of this class of targeted therapeutics as an important new treatment modality in cancer therapy. This review will focus on a discussion of antiangiogenic therapeutic monoclonal antibodies in development for the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Alan B Sandler
- Department of Medical Oncology, Vanderbilt-Ingram Cancer Center, Nashville, TN 37232-6307, USA.
| |
Collapse
|
259
|
Abstract
Endothelial progenitor cells (EPCs) have been isolated from peripheral blood CD34, VEGFR-2, or AC 133 (CD133) antigen-positive cells, which may home to site of neovascularization and differentiate into endothelial cells in situ. Endothelial cells contribute to tumor angiogenesis, and can originate from sprouting or co-option of neighbouring pre-existing vessels. Emerging evidence indicate that bone marrow-derived circulating EPCs can contribute to tumor angiogenesis and growth of certain tumors. This review article will summarize the literature data concerning this new role played by EPCs in tumor angiogenesis.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari I-70124, Italy.
| |
Collapse
|
260
|
Sata M, Fukuda D, Tanaka K, Kaneda Y, Yashiro H, Shirakawa I. The role of circulating precursors in vascular repair and lesion formation. J Cell Mol Med 2005; 9:557-68. [PMID: 16202205 PMCID: PMC6741295 DOI: 10.1111/j.1582-4934.2005.tb00488.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The accumulation of smooth muscle cells (SMCs) plays a principal role in atherogenesis, post-angioplasty restenosis and transplantation-associated vasculopathy. Therefore, much effort has been expended in targeting the migration and proliferation of medial smooth muscle cells to prevent occlusive vascular remodeling. Recent evidence suggests that bone marrow-derived circulating precursors can also give rise to endothelial cells and smooth muscle cells that contribute to vascular repair, remodeling, and lesion formation under physiological and pathological conditions. This article overviews recent findings on circulating vascular progenitor cells and describes potential therapeutic strategies that target these cells to treat occlusive vascular diseases.
Collapse
Affiliation(s)
- Masataka Sata
- Department of Cardiovascular Medicine, University of Tokyo, Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | | | | | | | | | | |
Collapse
|
261
|
Okazaki T, Ebihara S, Takahashi H, Asada M, Kanda A, Sasaki H. Macrophage colony-stimulating factor induces vascular endothelial growth factor production in skeletal muscle and promotes tumor angiogenesis. THE JOURNAL OF IMMUNOLOGY 2005; 174:7531-8. [PMID: 15944252 DOI: 10.4049/jimmunol.174.12.7531] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although M-CSF has been used for myelosuppression due to chemotherapy in patients with solid tumors, the effect of exogenous M-CSF on tumor angiogenesis has not been studied. In this study we showed that M-CSF has the ability to accelerate solid tumor growth by enhancing angiogenesis with a novel mechanism. M-CSF accelerated intratumoral vessel density in tumors inoculated into mice, although it did not accelerate the proliferation of malignant cells and cultured endothelial cells in vitro. In both the absence and the presence of tumors, M-CSF significantly increased the circulating cells that displayed phenotypic characteristics of endothelial progenitor cells in mice. Moreover, M-CSF treatment induced the systemic elevation of vascular endothelial growth factor (VEGF). VEGFR-2 kinase inhibitor significantly impaired the effect of M-CSF on tumor growth. In vivo, M-CSF increased VEGF mRNA expression in skeletal muscles. Even after treatment with carageenan and anti-CD11b mAb in mice, M-CSF increased VEGF production in skeletal muscles, suggesting that systemic VEGF elevation was attributed to skeletal muscle VEGF production. In vitro, M-CSF increased VEGF production and activated the Akt signaling pathway in C2C12 myotubes. These results suggest that M-CSF promotes tumor growth by increasing endothelial progenitor cells and activating angiogenesis, and the effects of M-CSF are largely based on the induction of systemic VEGF from skeletal muscles.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/physiopathology
- Cell Line
- Cell Proliferation
- Endothelium, Vascular/immunology
- Endothelium, Vascular/pathology
- Humans
- Macrophage Colony-Stimulating Factor/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Microcirculation/immunology
- Microcirculation/pathology
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- NIH 3T3 Cells
- Neoplastic Cells, Circulating/immunology
- Neoplastic Cells, Circulating/pathology
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/physiopathology
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Signal Transduction/immunology
- Stem Cells/immunology
- Stem Cells/pathology
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/blood
Collapse
Affiliation(s)
- Tatsuma Okazaki
- Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | |
Collapse
|
262
|
Beaudry P, Force J, Naumov GN, Wang A, Baker CH, Ryan A, Soker S, Johnson BE, Folkman J, Heymach JV. Differential effects of vascular endothelial growth factor receptor-2 inhibitor ZD6474 on circulating endothelial progenitors and mature circulating endothelial cells: implications for use as a surrogate marker of antiangiogenic activity. Clin Cancer Res 2005; 11:3514-22. [PMID: 15867254 DOI: 10.1158/1078-0432.ccr-04-2271] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Circulating endothelial cells (CEC) comprise at least two distinct populations: bone marrow-derived circulating endothelial progenitors (CEP) and mature CECs derived from existing vasculature. We hypothesized that antiangiogenic agents may have differential effects on CEPs and mature CECs and that these changes may serve as a marker of biological activity. EXPERIMENTAL DESIGN The effect of angiogenesis inhibitors on CECs was evaluated by flow cytometry after vascular endothelial growth factor (VEGF)-induced mobilization and in mice bearing Lewis lung carcinoma (LLC). Tumor angiogenesis was evaluated in parallel by immunohistochemistry. RESULTS In nontumor-bearing mice, VEGF administration increased both mature CECs and CEPs. This increase was inhibited by the VEGF receptor 2 inhibitor ZD6474 as well as the VEGF inhibitor-soluble Flt-1. ZD6474 had no significant effect on CECs in the absence of exogenous VEGF stimulation. In contrast, LLC-bearing mice had an increase in mature CECs but not CEPs after 3 days of treatment with ZD6474. The increase in mature CECs was dose-dependent, accompanied by a decrease in tumor microvessel density, and preceded reduction in tumor volume. Treatment of LLC-bearing mice with the vascular targeting agent ZD6126 also increased mature CECs. CONCLUSIONS VEGF inhibitors can have differential effects on mature CECs and CEPs, and agents inhibiting tumor angiogenesis may cause a concomitant increase in mature CECs. This increase occurs in tumor-bearing but not in nontumor-bearing mice, suggesting that tumor endothelium is a potential source of mature CECs. Therefore, assessing both mature CECs and CEPs may provide insights into the mechanism of antiangiogenic agents and serve as an early surrogate marker of biological activity.
Collapse
Affiliation(s)
- Paul Beaudry
- Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Wang Y, Tägil K, Ripa RS, Nilsson JC, Carstensen S, Jørgensen E, Søndergaard L, Hesse B, Johnsen HE, Kastrup J. Effect of mobilization of bone marrow stem cells by granulocyte colony stimulating factor on clinical symptoms, left ventricular perfusion and function in patients with severe chronic ischemic heart disease. Int J Cardiol 2005; 100:477-83. [PMID: 15837093 DOI: 10.1016/j.ijcard.2004.12.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Accepted: 12/31/2004] [Indexed: 12/23/2022]
Abstract
OBJECTIVES A phase I safety and efficacy study with granulocyte colony stimulating factor (G-CSF) mobilization of bone marrow stem cells to induce vasculogenesis in patients with severe ischemic heart disease (IHD) was conducted. DESIGN, PATIENTS AND RESULTS 29 patients with IHD participated in the study. Thirteen patients were treated with G-CSF for 6 days and 16 patients served as controls. G-CSF treatment was without any serious adverse events. Four patients were "poor mobilizers" with a maximal increase in CD34+ cells to 5,000+/-700/mL blood (mean+/-S.D.) compared to 28,900+/-5,100/mL blood in "mobilizers". At the follow-up, G-CSF treated had improved in CCS classification, NTG consumption and angina attacks, but the controls only in CCS classification. No difference was seen between the two groups. The decline in NTG consumption tended to be significant in "mobilizers" compared to controls. Myocardial perfusion was unchanged at adenosine stress single photon emission computerized tomography (SPECT) or magnetic resonance images (MRI). Left ventricular ejection fraction decreased from 57% to 52% (p<0.01, MRI) and from 48% to 44% (p=0.07, SPECT) in G-CSF treated, but was unchanged measured with echocardiography. CONCLUSIONS Treatment by G-CSF improved symptoms but not signs of myocardial ischemia in patients with severe IHD. The effects seemed related to mobilization of stem cells. An adverse effect on ejection fraction could not be excluded.
Collapse
Affiliation(s)
- Yongzhong Wang
- Cardiac Catheterization Laboratory, The Heart Centre, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Burnham EL, Taylor WR, Quyyumi AA, Rojas M, Brigham KL, Moss M. Increased circulating endothelial progenitor cells are associated with survival in acute lung injury. Am J Respir Crit Care Med 2005; 172:854-60. [PMID: 15976374 DOI: 10.1164/rccm.200410-1325oc] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Repair of damaged endothelium is important in recovery from acute lung injury. In animal models, bone marrow-derived endothelial progenitor cells differentiate into mature endothelium and assist in repairing damaged vasculature. OBJECTIVES The quantity of endothelial progenitor cells in patients with acute lung injury is unknown. We hypothesize that increased numbers of circulating endothelial progenitor cells will be associated with an improved outcome in acute lung injury and the acute respiratory distress syndrome. METHODS Peripheral blood mononuclear cells from the buffy coat of patients with early acute lung injury (n=45), intubated control subjects (n=10), and healthy volunteers (n=7) were isolated using Ficoll density gradient centrifugation, and plated on fibronectin-coated cellware. After 24 hours, nonadherent cells were removed and replated on fibronectin-coated cellware at a concentration of 1x10(6) cells/well. Colony-forming units were counted after 7 days' incubation. MEASUREMENTS/MAIN RESULTS Endothelial progenitor cell colony numbers were significantly higher in patients with acute lung injury compared with healthy control subjects (p<0.05), but did not differ between patients with acute lung injury and intubated control subjects. However, in the 45 patients with acute lung injury, improved survival correlated with a higher colony count (p<0.04). Patients with acute lung injury with a colony count of >or= 35 had a mortality of 30%, compared with 61% in those with colony counts <35 (p<0.03), results that persisted in a multivariable analysis correcting for age, sex, and severity of illness. CONCLUSIONS An increased number of circulating endothelial progenitor cells in acute lung injury is associated with improved survival.
Collapse
Affiliation(s)
- Ellen L Burnham
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Emory University School of Medicine, and the Atlanta Venterans' Affairs Medical Center, Atlanta, GA 30335, USA.
| | | | | | | | | | | |
Collapse
|
265
|
Herbst RS, Onn A, Sandler A. Angiogenesis and lung cancer: prognostic and therapeutic implications. J Clin Oncol 2005; 23:3243-56. [PMID: 15886312 DOI: 10.1200/jco.2005.18.853] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lung cancer is the most common cause of cancer death worldwide, with most patients dying with metastatic disease. The prognosis for the majority of patients remains poor. It is evident that advances in the treatment of this and other tumor types will require new approaches, and recent research has focused on molecular-targeted therapies. A key therapeutic strategy is inhibition of specific processes essential for tumor vascular development (a concept known to be beneficial in colorectal cancer) and a range of such antiangiogenic agents are currently in development. The most promising of these target the proangiogenic vascular endothelial growth factor (VEGF), either by preventing VEGF-receptor binding or inhibiting downstream receptor signaling. However, other more direct approaches against tumor vasculature are also in development. Since antiangiogenic agents often exert an indirect, cytostatic effect, many are being evaluated in combination with conventional chemotherapies in order to optimize the anticancer effects of both strategies. Additionally, the combination of several antiangiogenic agents is also being explored. This has become possible given the large number of agents currently available. As part of this evaluation process, the assessment of surrogate markers of target inhibition and treatment effect is ongoing in the hope of identifying reliable surrogate markers to aid the development of this new generation of anticancer agents.
Collapse
Affiliation(s)
- Roy S Herbst
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 432, Houston, TX 77030-4009, USA.
| | | | | |
Collapse
|
266
|
Theopold C, Yao F, Eriksson E. Gene therapy in the treatment of lower extremity wounds. INT J LOW EXTR WOUND 2005; 3:69-79. [PMID: 15866792 DOI: 10.1177/1534734604265431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This article presents a brief overview of the etiology of chronic wounds of the lower extremities and their current medical and surgical treatment. Gene therapy as a potential tool for treating therapeutically challenging wounds is described in terms of the vectors employed in gene transfer, as well as the strategies used to promote wound healing. Results from animal model studies, as well as clinical trials, are presented.
Collapse
Affiliation(s)
- Christoph Theopold
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
267
|
Simman R, Craft C, McKinney B. Improved Survival of Ischemic Random Skin Flaps Through the Use of Bone Marrow Nonhematopoietic Stem Cells and Angiogenic Growth Factors. Ann Plast Surg 2005; 54:546-52. [PMID: 15838218 DOI: 10.1097/01.sap.0000158068.86576.73] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Surgical skin flaps are frequently used in plastic and reconstructive surgery to repair acquired or congenital defects. Necrosis is a common complication associated with these flaps postoperatively as a result of inadequate blood supply. Stem cells are precursor cells with the potential to differentiate into more specialized cells. Angiogenic factors act to direct cellular differentiation and organization to form new vascular elements. Our theory was that the combination of angiogenic growth factors with stem cells derived from the subject preoperatively would augment neovascularization, thereby increasing blood supply to the flap, which may ultimately improve flap survival. In phase I, 40 Lewis rats were randomized into 4 groups of 10. Random dorsal skin flaps were elevated and treated at the same time. The first group was injected with only medium, the second with stem cells, the third with stem cells and angiogenic factors, and the fourth with angiogenic growth factors. Millimetric measurements of flap viability at 7 and 14 days did not show any statistically significant differences between the studied groups. In phase II, 24 rats were also randomized into 4 groups of 6, but this time were treated 1 week before flap elevation. The viability measurements showed an increased rate of viability in the group in which stem cells and the angiogenic factors were administered simultaneously (84.5% +/- 3.2%) as compared with the unmodified control group (62.6% +/- 7.3%) or to the groups in which only precursor cells (60.4% +/- 7.9%) or angiogenic factors (62.3%+/- 10.1%). Increased blood supply brought by these manipulations is believed translated to increased tissue flap survival. Punch biopsies showed that "green fluorescent protein"-labeled precursor cells was noted to form luminal structures in the treated flaps. The vascular cast of all flaps was filled with Mercox plastic resin. After euthanasia, the soft tissues of the harvested flaps were dissolved and the remaining vascular cast was weighted. The weight-based ratio of the vascular composition was determined. The flaps injected with both stem cells and angiogenic factors showed higher values. We conclude that the administration of bone marrow stem cells with angiogenic factors 1 week before flap creation improves the survival of ischemic random skin flaps.
Collapse
Affiliation(s)
- Richard Simman
- East Tennessee State University, James H. Quillen College of Medicine, Department of Surgery, Division of Plastic Surgery, Johnson City, TN, USA.
| | | | | |
Collapse
|
268
|
Bellik L, Ledda F, Parenti A. Morphological and phenotypical characterization of human endothelial progenitor cells in an early stage of differentiation. FEBS Lett 2005; 579:2731-6. [PMID: 15862317 DOI: 10.1016/j.febslet.2005.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Revised: 03/29/2005] [Accepted: 04/06/2005] [Indexed: 10/25/2022]
Abstract
The exact phenotype and lineage of endothelial progenitor cells (EPCs) are still a matter of debate and different expansion protocols are used to obtain them. In this study, EPC expansion from peripheral blood mononuclear cells was analyzed within the first week of culture. Both the adherent and suspended cells, of which the latter usually discarded, were considered. We provide, for the first time, a systematic study of EPC phenotype and functional features within the first 3 days of culture. Moreover, within the 2nd day, both cellular fractions displayed a significant increase in endothelial marker expression which correlated with EPC properties.
Collapse
Affiliation(s)
- Lydia Bellik
- Department of Preclinical and Clinical Pharmacology, University of Florence, Italy
| | | | | |
Collapse
|
269
|
Affiliation(s)
- J M Hill
- The Laboratory of Molecular Biology; Cardiology Branch of the National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
270
|
Abstract
Postnatal vasculogenesis is considered to be involved in neovascularization of adult tissues, because bone marrow-derived endothelial progenitor cells (EPCs) were isolated from circulating mononuclear cells in peripheral blood and were shown to incorporate into sites of physiological and pathological neovascularization and to differentiate into mature endothelial cells. EPCs might have an attractive potential therapeutic application for cardiovascular ischemic diseases as a novel cell-based strategy mainly via a vasculogenesis mechanism.
Collapse
Affiliation(s)
- Satoshi Murasawa
- Department of Regenerative Medicine and Research, Kobe Institute of Biomedical Research and Innovation/RIKEN, Center for Developmental Biology, Kobe Japan.
| | | |
Collapse
|
271
|
Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med 2005; 8:498-508. [PMID: 15601578 PMCID: PMC6740289 DOI: 10.1111/j.1582-4934.2004.tb00474.x] [Citation(s) in RCA: 337] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bone marrow and peripheral blood of adults contain a special sub-type of progenitor cells which are able to differentiate into mature endothelial cells, thus contributing to re-endothelialization and neo-vascularization. These angiogenic cells have properties of embryonal angioblasts and were termed endothelial progenitor cells (EPCs). In general, three surface markers (CD133, CD34 and the vascular endothelial growth factor receptor-2) characterize the early functional angioblast, located predominantly in the bone marrow. Later, when migrating to the systemic circulation EPCs gradually lose their progenitor properties and start to express endothelial marker like VE-cadherin, endothelial nitric oxide synthase and von Willebrand factor. The number of circulating EPCs in healthy subjects is rather low and a variety of conditions or factors may further influence this number. In the context of possible therapeutic application of EPCs recent clinical studies employing these cells for neo-vascularization of ischemic organs have just been published. However, the specificity of the observed positive clinical effects, the mechanisms regulating the differentiation of EPCs and their homing to sites of injured tissue remain partially unknown at present.
Collapse
Affiliation(s)
- Mihail Hristov
- Department for Molecular Cardiovascular Research, University Hospital Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | | |
Collapse
|
272
|
Abstract
Recent evidences suggest that endothelial progenitor cells (EPCs) derived from bone marrow (BM) contribute to de novo vessel formation in adults occurring as physiological and pathological responses. Emerging preclinical trials have shown that EPCs home to sites of neovascularization after ischemic events in limb and myocardium. On the basis of these aspects, EPCs are expected to develop as a key strategy of therapeutic applications for the ischemic organs. Such clinical requirements of EPCs will tentatively accelerate the translational research aiming at the devices to acquire the optimized quality and quantity of EPCs. In this review, we attempt to discuss about biological features of EPCs and speculate on the clinical potential of EPCs for therapeutic neovascularization.
Collapse
Affiliation(s)
- Yo Iwami
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Bohseidai, Isehara, Kanagawa 259-1193, Japan
| | | | | |
Collapse
|
273
|
Kuroda S, Kondo H, Ohya K, Kasugai S. A New Technique With Calcium Phosphate Precipitate Enhances Efficiency of In Vivo Plasmid DNA Gene Transfer. J Pharmacol Sci 2005; 97:227-33. [PMID: 15699579 DOI: 10.1254/jphs.fp0040504] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
In vivo gene transfer with plasmid vector has been applied experimentally and clinically; however, the low level of gene transfer efficiency with plasmid vector is a problem. We speculated that the combination of calcium phosphate precipitate (CaP) and plasmid vector could solve this problem because CaP stabilizes plasmid DNA. In the present study, we used a plasmid exression vector encoding enhanced green fluorescent protein and combined the vector with CaP. Then, this combination was mixed with bovine type I atelocollagen. After incubating this mixture in phosphate-buffered saline, the amount of the plasmid DNA in the supernatant was low when the plasmid DNA was combined with CaP. Furthermore, the plasmid DNA, which was combined with CaP, was stable in DNase digestion in vitro. The plasmid vector with or without CaP, together with the atelocollagen, was transplanted subcutaneously or injected in the bone marrow of the femurs of rats. Then, the fluorescence was observed under a confocal laser scanning microscope and the fluorescence intensity in the tissue homogenates was measured. In these animal experiments, the fluorescence was extensive when the plasmid DNA was combined with CaP. These results indicate that our formula, collagen/CaP/DNA, appeared efficient for in vivo gene transfer.
Collapse
Affiliation(s)
- Shinji Kuroda
- Oral Implantology and Regenerative Dental Medicine, Department of Masticatory Function Rehabilitation, Tokyo Medical and Dental University, Japan.
| | | | | | | |
Collapse
|
274
|
Non-classical mechanisms of heart repair. Mol Cell Biochem 2005. [DOI: 10.1007/s11010-005-5117-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
275
|
Povsic TJ, Peterson ED. Progenitor cell therapy of ischemic heart disease: the new frontier. Future Cardiol 2005; 1:87-98. [DOI: 10.1517/14796678.1.1.87] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The discovery of circulating cells capable of differentiating into vascular structures (endothelial progenitor cells) has both altered our understanding of the pathophysiology of atherosclerosis and offered the possibility of using nature's reparative mechanisms to accelerate vascular restoration and regeneration. Epidemiologic studies indicate a correlation between cardiac risk factors, the presence of coronary disease and the numbers and function of endothelial progenitor cells. Preclinical animal models have demonstrated the therapeutic potential of cellular therapies for repair of ischemic myocardial damage. Several human trials investigating either the use of progenitor cells derived from bone marrow or peripheral blood, or pharmacologic therapies aimed at increasing mobilization of such cells for the treatment of ischemic heart disease, are currently underway. Some basic tenets of progenitor cell biology, key preclinical results suggesting the utility of this therapy, and the strengths and limitations of current human trials are briefly summarized, and the role of cellular therapies, today and in the future, are explored.
Collapse
Affiliation(s)
- Thomas J Povsic
- Duke University Medical Center, Division of Cardiology, Department of Medicine, Box 3126, Durham, NC 27710,USATel.: Fax:
| | - Eric D Peterson
- Duke University Medical Center, Division of Cardiology, Department of Medicine, Box 3126, Durham, NC 27710,USATel.: Fax:
| |
Collapse
|
276
|
Imanishi T, Hano T, Nishio I. Estrogen Reduces Angiotensin II-Induced Acceleration of Senescence in Endothelial Progenitor Cells. Hypertens Res 2005; 28:263-71. [PMID: 16097371 DOI: 10.1291/hypres.28.263] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The interaction among estrogen, angiotensin II (Ang II), and oxidative stress in endothelial progenitor cells (EPCs) remains unknown. We therefore investigated the potential effect of estrogen on Ang II-induced EPC oxidative stress and senescence in EPCs. EPCs were isolated from peripheral blood and characterized. Both reverse transcription (RT)-polymerase chain reaction (PCR) and Western blotting were used to assess gp91phox and angiotensin type 1 receptor (AT1R) expression. Immunofluorescence of nitrotyrosine provided evidence of peroxynitrite formation. Our data indicate that Ang II increased the expression of gp91phox mRNA and protein, and these effects were attenuated by 17beta-estradiol (E2). The exposure of cultured EPCs to Ang II (100 nmol/l) significantly accelerated the rate of senescence compared to that in control cells during 14 days in culture as determined by acidic beta-galactosidase staining, and this effect was significantly inhibited by E2 (p < 0.01). Because cellular senescence is critically influenced by telomerase, which elongates telomeres, we measured telomerase activity by using a PCR-ELISA-based assay. Ang II significantly diminished telomerase activity, although the effect was significantly reduced by pre-treatment with E2 (p < 0.01). Because we previously demonstrated that both the up-regulation of gp91phox and the acceleration of cellular senescence in Ang II-stimulated EPCs could be abolished by pre-treatment with the AT1R- specific antagonist, valsartan, we also explored the effect of estrogen on AT1R expression. Ang II increased AT1R mRNA and protein expression, and these increases were prevented by E2, suggesting that AT1R may at least partially mediate the inhibitory effect of E2 on Ang II-induced acceleration of senescence in EPCs. In conclusion, estrogen reduces Ang II-induced acceleration of senescence in EPCs partially through down-regulation of AT1R expression.
Collapse
Affiliation(s)
- Toshio Imanishi
- Department of Cardiovascular Medicine, Wakayama Medical University, 811-1, Kimiidera, Wakayama 641-8510, Japan.
| | | | | |
Collapse
|
277
|
Shah PB, Losordo DW. Non‐Viral Vectors for Gene Therapy: Clinical Trials in Cardiovascular Disease. NON-VIRAL VECTORS FOR GENE THERAPY, SECOND EDITION: PART 2 2005; 54:339-61. [PMID: 16096018 DOI: 10.1016/s0065-2660(05)54014-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The population of patients with end-stage symptomatic coronary and peripheral vascular disease is ever-expanding. Many of these patients no longer have options for mechanical revascularization, and despite maximal medical therapy, they remain physically limited due to angina or critical limb ischemia. The fundamental problem in these patients is insufficient blood supply to muscle due to severely diseased conduit vessels to the target tissue. Therefore, it seems logical that increasing the blood supply to ischemic tissue will relieve symptoms. One potential means to achieving this goal is via therapeutic angiogenesis. The molecular mechanisms behind vascular development are being elucidated, and animal models have shown that mediators of vascular development can be harnessed to produce new capillaries in ischemic tissue. These mediators include cytokines such as vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). Angiogenic cytokines can be delivered in several forms including recombinant protein or via gene delivery as a naked plasmid or via viral vector. This chapter will describe the clinical trial experience to date with delivery of non-viral gene therapy for therapeutic angiogenesis in humans with disabling myocardial ischemia and peripheral vascular disease.
Collapse
Affiliation(s)
- Pinak B Shah
- Division of Cardiology, Caritas St. Elizabeth's Medical Center Boston, Massachusetts 02135, USA
| | | |
Collapse
|
278
|
Takagi Y, Omura T, Yoshiyama M, Matsumoto R, Enomoto S, Kusuyama T, Nishiya D, Akioka K, Iwao H, Takeuchi K, Yoshikawa J. Granulocyte-Colony Stimulating Factor Augments Neovascularization Induced by Bone Marrow Transplantation in Rat Hindlimb Ischemia. J Pharmacol Sci 2005; 99:45-51. [PMID: 16127245 DOI: 10.1254/jphs.fp0040966] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Because granulocyte-colony stimulating factor (G-CSF) mobilizes bone marrow cells including endothelial progenitor cells, we examined whether G-CSF augments angiogenesis and collateral vessel formation induced by bone marrow-mononuclear cells transplantation (BMT). Unilateral hindlimb ischemia was surgically induced in Lewis rats. One week after surgery, administration of 100 mg/kg per day G-CSF significantly increased the laser Doppler blood perfusion index (LDBPI), number of angiographically detectable collateral vessels (angiographic score), and capillary density determined by alkaline phosphatase staining. In the BMT group (1 x 10(7) cells/rat) and the group with combined G-CSF treatment and BMT, LDBPI was significantly increased compared with that in the vehicle-treated group. In the BMT group, neovascularization was significantly increased as evidenced by the angiographic score and capillary density compared with the vehicle-treated group. Furthermore, the combination of G-CSF treatment and BMT augmented neovascularization compared with BMT alone, as evidenced by the angiographic score and capillary density. Moreover, G-CSF significantly increased vascular endothelial growth factor mRNA and fibroblast growth factor-2 mRNA in hindlimb muscle. In conclusion, G-CSF was found to augment neovascularization in rat hindlimb ischemia. Combined use of G-CSF treatment and BMT may be a useful strategy for therapeutic neovascularization in ischemic tissues.
Collapse
Affiliation(s)
- Yasuhiro Takagi
- Department of Internal Medicine and Cardiology, Osaka City University Graduate School of Medical Sciences, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Silvestre JS, Lévy BI. Hormones and the neovascularization process: role of angiotensin II. EXS 2004:77-93. [PMID: 15617472 DOI: 10.1007/3-7643-7311-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
|
280
|
Otani A, Dorrell MI, Kinder K, Moreno SK, Nusinowitz S, Banin E, Heckenlively J, Friedlander M. Rescue of retinal degeneration by intravitreally injected adult bone marrow-derived lineage-negative hematopoietic stem cells. J Clin Invest 2004; 114:765-74. [PMID: 15372100 PMCID: PMC516263 DOI: 10.1172/jci21686] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2004] [Accepted: 07/13/2004] [Indexed: 12/26/2022] Open
Abstract
Inherited retinal degenerations afflict 1 in 3,500 individuals and are a heterogeneous group of diseases that result in profound vision loss, usually the result of retinal neuronal apoptosis. Atrophic changes in the retinal vasculature are also observed in many of these degenerations. While it is thought that this atrophy is secondary to diminished metabolic demand in the face of retinal degeneration, the precise relationship between the retinal neuronal and vascular degeneration is not clear. In this study we demonstrate that whenever a fraction of mouse or human adult bone marrow-derived stem cells (lineage-negative hematopoietic stem cells [Lin- HSCs]) containing endothelial precursors stabilizes and rescues retinal blood vessels that would ordinarily completely degenerate, a dramatic neurotrophic rescue effect is also observed. Retinal nuclear layers are preserved in 2 mouse models of retinal degeneration, rd1 and rd10, and detectable, albeit severely abnormal, electroretinogram recordings are observed in rescued mice at times when they are never observed in control-treated or untreated eyes. The normal mouse retina consists predominantly of rods, but the rescued cells after treatment with Lin- HSCs are nearly all cones. Microarray analysis of rescued retinas demonstrates significant upregulation of many antiapoptotic genes, including small heat shock proteins and transcription factors. These results suggest a new paradigm for thinking about the relationship between vasculature and associated retinal neuronal tissue as well as a potential treatment for delaying the progression of vision loss associated with retinal degeneration regardless of the underlying genetic defect.
Collapse
Affiliation(s)
- Atsushi Otani
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
281
|
Ishizawa K, Kubo H, Yamada M, Kobayashi S, Suzuki T, Mizuno S, Nakamura T, Sasaki H. Hepatocyte growth factor induces angiogenesis in injured lungs through mobilizing endothelial progenitor cells. Biochem Biophys Res Commun 2004; 324:276-80. [PMID: 15465014 DOI: 10.1016/j.bbrc.2004.09.049] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Indexed: 11/22/2022]
Abstract
Circulating endothelial progenitor cells (EPCs) play a pivotal role in angiogenesis. Hepatocyte growth factor (HGF) is known to induce proliferation and motility in endothelial cells, and to play a role in mitogenic and morphogenic actions. However, the role of HGF in EPC mobilization has not been clearly described yet. We investigated the effect of HGF on mobilizing EPCs and on angiogenesis in elastase-induced lung injury. HGF significantly increased the triple-positive (Sca-1(+), Flk-1(+), and c-kit(+)) fraction in peripheral mononuclear cells in mice. The bone marrow-derived cells were recruited into the injured lungs, where they differentiated to capillary endothelial cells. HGF induced proliferation of both bone marrow-derived and resident endothelial cells in the alveolar wall. In conclusion, the present study suggests that HGF induces EPC mobilization from the bone marrow and enhances the proliferation of endothelial cells in vivo. These complex effects induced by HGF orchestrate pulmonary regeneration in emphysematous lung parenchyma.
Collapse
Affiliation(s)
- Kota Ishizawa
- Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Miyagi 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
282
|
Beerepoot LV, Mehra N, Linschoten F, Jorna AS, Lisman T, Verheul HMW, Voest EE. Circulating endothelial cells in cancer patients do not express tissue factor. Cancer Lett 2004; 213:241-8. [PMID: 15327840 DOI: 10.1016/j.canlet.2004.04.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Revised: 04/16/2004] [Accepted: 04/16/2004] [Indexed: 11/28/2022]
Abstract
Numbers of circulating endothelial cells (CECs) are increased in cancer patients with progressive disease. Also, cancer patients have an increased risk for thrombotic events, being negatively associated with prognosis. Tissue factor (TF), the physiological initiator of coagulation, is present on the surface of many extravascular cells. In 34 samples from cancer patients and in seven from volunteers, CECs were quantified (with endothelium-specific anti-CD146 beads), and TF-activity assessed with a chromogenic assay. All samples displayed very limited TF-activity (patients: 1.6+/-3.1 microU; volunteers: 0.94+/-1.7 microU FXa/100 CECs, P = 0.30 by Mann-Whitney test). After in vitro TNFalpha-stimulation, CECs from both cancer patients and volunteers showed substantially increased TF-activity (endogenous activity: 17.3+/-6.4 microU; after TNFalpha-stimulation: 73.8+/-34.3 microU FXa; P = 0.028, Wilcoxon signed ranks test), reflecting the potential of CECs to generate biologically active TF. As the chromogenic assay determines a mean cellular TF-activity, we also analyzed immunohistochemical TF-antigen expression on CEC subsets. TF-antigen expression was undetectable. CECs isolated from cancer patients do not express TF. CECs can generate functional TF after stimulation and may therefore play a role in (intratumoral) coagulation induction and tumor angiogenesis.
Collapse
Affiliation(s)
- Laurens V Beerepoot
- Laboratory of Experimental Oncology, Department of Medical Oncology, HP F02.126, University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, 3508 GA, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
283
|
Chalajour F, Treede H, Ebrahimnejad A, Lauke H, Reichenspurner H, Ergun S. Angiogenic activation of valvular endothelial cells in aortic valve stenosis. Exp Cell Res 2004; 298:455-64. [PMID: 15265693 DOI: 10.1016/j.yexcr.2004.04.034] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2004] [Revised: 04/16/2004] [Indexed: 11/30/2022]
Abstract
Here, we demonstrate the angiogenic response of valvular endothelial cells to aortic valve (AV) stenosis using a new ex vivo model of aortic leaflets. Histological analysis revealed neovascularization within the cusps of stenotic but not of non-stenotic aortic valves. Correspondingly, the number of capillary-like outgrowth in 3D collagen gel was significantly higher in stenotic than in non-stenotic valves. Capillary-like sprouting was developed significantly faster in stenotic than in non-stenotic valves. New capillary sprouts from stenotic aortic valves exhibited the endothelial cell markers CD31, CD34 and von-Willebrand factor (vWF) as well as carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1), Tie-2 and angiogenesis inhibitor endostatin. Western blot analyses revealed a significant increase of CEACAM1 and endostatin in stenotic aortic valve tissue. Electron microscopic examinations demonstrate that these capillary-like tubes are formed by endothelial cells containing Weibel-Palade bodies. Remarkably, inter-endothelial junctions are established and basement membrane material is partially deposited on the basal side of the endothelial tubes. Our data demonstrate the capillary-like sprout formation from aortic valves and suggest a role of angiogenesis in the pathogenesis of aortic valve stenosis. These data provide new insights into the mechanisms of valvular disorders and open new perspectives for prevention and early treatment of calcified aortic stenosis.
Collapse
MESH Headings
- Aged
- Antigens, CD/metabolism
- Antigens, CD34/metabolism
- Antigens, Differentiation/metabolism
- Aortic Valve/growth & development
- Aortic Valve/pathology
- Aortic Valve/physiopathology
- Aortic Valve Stenosis/metabolism
- Aortic Valve Stenosis/pathology
- Aortic Valve Stenosis/physiopathology
- Basement Membrane/metabolism
- Basement Membrane/ultrastructure
- Capillaries/metabolism
- Capillaries/pathology
- Capillaries/physiopathology
- Cell Adhesion Molecules
- Endostatins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/ultrastructure
- Female
- Humans
- Intercellular Junctions/metabolism
- Intercellular Junctions/ultrastructure
- Male
- Microscopy, Electron
- Models, Biological
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/physiopathology
- Organ Culture Techniques
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Receptor, TIE-2/metabolism
- Weibel-Palade Bodies/metabolism
- Weibel-Palade Bodies/ultrastructure
- von Willebrand Factor/metabolism
Collapse
Affiliation(s)
- Fariba Chalajour
- Department of Cardiovascular Surgery, University Hospital Eppendorf, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
284
|
Keswani SG, Katz AB, Lim FY, Zoltick P, Radu A, Alaee D, Herlyn M, Crombleholme TM. Adenoviral mediated gene transfer of PDGF-B enhances wound healing in type I and type II diabetic wounds. Wound Repair Regen 2004; 12:497-504. [PMID: 15453831 DOI: 10.1111/j.1067-1927.2004.12501.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have shown that the genetically diabetic mouse (C57BLKS/J-m+/+Lepr(db)) has a wound healing and neovascularization deficit associated with an inability to recruit endothelial precursor cells (EPCs) to the wound. This may account for a fundamental mechanism in impaired diabetic wound healing. We hypothesized that the adenoviral mediated overexpression of platelet-derived growth factor-B (PDGF-B) would enhance wound healing, improve neovascularization, and recruit EPCs to the epithelial wound in three diabetic mouse models. Eight-mm full-thickness flank wounds were made in db/db, nonobese NOD/Ltj, streptozotocin, and C57BLKS/J mice. Wounds were treated with either 1 x 10(8) PFU Ad-PDGF-B or Ad LacZ or phosphate buffered saline solution. Wounds harvested at seven days were analyzed for epithelial gap, blood vessel density, granulation tissue area, and EPCs per high powered field. All three diabetic models have a significant wound healing and neovascularization defect compared to C57BLKS/J controls. Adenoviral-PDGF-B treatment significantly enhanced epithelial gap closure in db/db, streptozotocin, and nonobese NOD/Ltj mice as compared to diabetic phosphate buffered saline solution or Ad LacZ controls. A similar increase in the formation of granulation tissue and vessel density was also observed. All three models had reduced levels of GATA-2 positive EPCs in the wound bed that was corrected by the adenoviral mediated gene transfer of PDGF. EPC recruitment was positively correlated with neovascularization and wound healing. Three different diabetic models have a wound healing impairment and a decreased ability to recruit EPCs. The vulnerary effect of adenoviral mediated gene therapy with PDGF-B significantly enhanced wound healing and neovascularization in diabetic wounds. The PDGF-B mediated augmentation of EPC recruitment to the wound bed may be a fundamental mechanism of these results.
Collapse
Affiliation(s)
- Sundeep G Keswani
- Division of General, Thoracic, and Fetal Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
285
|
Tepper OM, Capla JM, Galiano RD, Ceradini DJ, Callaghan MJ, Kleinman ME, Gurtner GC. Adult vasculogenesis occurs through in situ recruitment, proliferation, and tubulization of circulating bone marrow-derived cells. Blood 2004; 105:1068-77. [PMID: 15388583 DOI: 10.1182/blood-2004-03-1051] [Citation(s) in RCA: 320] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Ischemia is a known stimulus for vascular growth. Bone marrow (BM)-derived endothelial progenitor cells (EPCs) are believed to contribute to new blood vessel growth, but the mechanism for this contribution is unknown. To elucidate how BM cells are able to form new blood vessels, a novel murine model of soft tissue ischemia was developed in lethally irradiated mice with BM reconstituted from either tie2/lacZ or ROSA/green fluorescent protein (GFP) mice (n = 24). BM-derived EPCs were recruited to ischemic tissue within 72 hours, and the extent of recruitment was directly proportional to the degree of tissue ischemia. At 7 days, there were persistently elevated levels of vascular endothelial growth factor (VEGF) (2.5-fold) and circulating VEGF receptor-2/CD11(-) (flk-1(+)/CD11(-)) cells (18-fold) which correlated with increased numbers of BM-derived EPCs within ischemic tissue. The cells were initially located extravascularly as proliferative clusters. By day 14, these clusters coalesced into vascular cords, which became functional vessels by day 21. In vitro examination of human EPCs from healthy volunteers (n = 10) confirmed that EPC proliferation, adhesion, and chemotaxis were all significantly stimulated in hypoxic conditions. We conclude that BM-derived cells produce new blood vessels via localized recruitment, proliferation, and differentiation of circulating cells in a sequence of events markedly different from existing paradigms of angiogenesis.
Collapse
Affiliation(s)
- Oren M Tepper
- Laboratory of Microvascular Research and Vascular Tissue Engineering, New York University Medical Center, NY, USA
| | | | | | | | | | | | | |
Collapse
|
286
|
Otani A, Dorrell MI, Kinder K, Moreno SK, Nusinowitz S, Banin E, Heckenlively J, Friedlander M. Rescue of retinal degeneration by intravitreally injected adult bone marrow–derived lineage-negative hematopoietic stem cells. J Clin Invest 2004. [DOI: 10.1172/jci200421686] [Citation(s) in RCA: 211] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
287
|
Dimmeler S, Zeiher AM. Vascular repair by circulating endothelial progenitor cells: the missing link in atherosclerosis? J Mol Med (Berl) 2004; 82:671-7. [PMID: 15322703 DOI: 10.1007/s00109-004-0580-x] [Citation(s) in RCA: 232] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The integrity and functional activity of the endothelial monolayer play a crucial role in the prevention of atherosclerosis. Increasing evidence suggests that risk factors for coronary artery disease increase endothelial cell apoptosis and lead to a disturbance in the endothelial monolayer. Recent insights suggest that the injured endothelial monolayer is regenerated by circulating bone marrow derived endothelial progenitor cells, which accelerates reendothelialization and limits atherosclerotic lesion formation. However, risk factors for coronary artery disease such as age and diabetes reduce the number and functional activity of these circulating endothelial progenitor cells, thus limiting the regenerative capacity. The impairment of stem/progenitor cells by risk factors may contribute to atherogenesis and atherosclerotic disease progression. We discuss this novel concept of endothelial regeneration and highlight possible novel strategies to interfere with the balance of injury and repair mechanisms.
Collapse
Affiliation(s)
- Stefanie Dimmeler
- Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | | |
Collapse
|
288
|
Walter DH, Zeiher AM, Dimmeler S. Effects of statins on endothelium and their contribution to neovascularization by mobilization of endothelial progenitor cells. Coron Artery Dis 2004; 15:235-42. [PMID: 15238818 DOI: 10.1097/01.mca.0000131572.14521.8a] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Statins are potent drugs with a variety of cardiovascular protective effects which appear to occur independent of cholesterol reduction. The vasculoprotective effects of statins might be due to their direct effect on endothelial cells leading to improved nitric oxide (NO) bioavailability. Mechanistically, statins induce endothelial nitric oxide synthesis (eNOS) mRNA stability in endothelial cells and promote eNOS activity through a PI3K/Akt dependent pathway. Novel targets of statins are pro-angiogenic actions including the mobilization and differentiation of bone marrow derived endothelial progenitor cells, which accelerate angiogenesis or re-endothelialization. The functional improvement and increased homing capacity of endothelial progenitor cells induced by statin treatment might reverse impaired functional regeneration capacities seen in patients with risk factors for coronary artery disease or documented active coronary artery disease.
Collapse
Affiliation(s)
- Dirk H Walter
- Division of Cardiology and Molecular Cardiology, University of Frankfurt, Frankfurt, Germany.
| | | | | |
Collapse
|
289
|
Melo LG, Pachori AS, Kong D, Gnecchi M, Wang K, Pratt RE, Dzau VJ. Gene and cell-based therapies for heart disease. FASEB J 2004; 18:648-63. [PMID: 15054087 DOI: 10.1096/fj.03-1171rev] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heart disease remains the prevalent cause of premature death and accounts for a significant proportion of all hospital admissions. Recent developments in understanding the molecular mechanisms of myocardial disease have led to the identification of new therapeutic targets, and the availability of vectors with enhanced myocardial tropism offers the opportunity for the design of gene therapies for both protection and rescue of the myocardium. Genetic therapies have been devised to treat complex diseases such as myocardial ischemia, heart failure, and inherited myopathies in various animal models. Some of these experimental therapies have made a successful transition to clinical trial and are being considered for use in human patients. The recent isolation of endothelial and cardiomyocyte precursor cells from adult bone marrow may permit the design of strategies for repair of the damaged heart. Cell-based therapies may have potential application in neovascularization and regeneration of ischemic and infarcted myocardium, in blood vessel reconstruction, and in bioengineering of artificial organs and prostheses. We expect that advances in the field will lead to the development of safer and more efficient vectors. The advent of genomic screening technology should allow the identification of novel therapeutic targets and facilitate the detection of disease-causing polymorphisms that may lead to the design of individualized gene and cell-based therapies.
Collapse
Affiliation(s)
- Luis G Melo
- Department of Physiology, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| | | | | | | | | | | | | |
Collapse
|
290
|
Csaky KG, Baffi JZ, Byrnes GA, Wolfe JD, Hilmer SC, Flippin J, Cousins SW. Recruitment of marrow-derived endothelial cells to experimental choroidal neovascularization by local expression of vascular endothelial growth factor. Exp Eye Res 2004; 78:1107-16. [PMID: 15109917 DOI: 10.1016/j.exer.2004.01.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2003] [Accepted: 01/28/2004] [Indexed: 12/01/2022]
Abstract
PURPOSE The question of whether adult animals maintain a reservoir of endothelial progenitor cells (EPCs) in the bone marrow that is involved in neovascularization is under investigation. The following study was undertaken to examine the potential contribution of EPCs to the development of choroidal neovascularization (CNV) in adult mice and to examine the role of local expression of vascular endothelial growth factor (VEGF) in this process. METHODS Lethally irradiated, adult female nude mice were engrafted with whole bone marrow isolated from male transgenic mice expressing LacZ driven by the endothelial specific Tie-2 promoter. Two months, following bone marrow reconstitution, confirmed by quantitative Taqman PCR, an E1-deleted adenoviral vector expressing vascular endothelial growth factor (165) (Ad.VEGF(165)) was injected subretinally to induce CNV, confirmed by collagen IV immunohistochemistry. Bone marrow-derived endothelial cells were detected using either X-gal staining or Y chromosome in situ hybridization. Y chromosome positive cells within the CNV were confirmed to be endothelial cells by lectin staining. RESULTS Subretinal Ad.VEGF(165) was capable of inducing CNV. Four-week old lesions were found to contain LacZ expressing cells within the CNV in bone marrow transplanted animals but not in negative control animals. Eighteen percent of all Y chromosome positive cells within the CNV were found to be lectin positive while 27% of all endothelial cells within the CNV were Y chromosome positive. CONCLUSION Engrafted bone marrow-derived EPCs were shown to differentiate into endothelial cells at the site of subretinal VEGF-induced CNV in mice. These results suggest that EPCs contribute to the formation of neovascularization and that subretinal expression of VEGF might play an important role in recruitment of these cells to the site of CNV.
Collapse
Affiliation(s)
- Karl G Csaky
- National Institutes of Health, National Eye Institute, Building 10-10B11, 9000 Rockville Pike, Bethesda, MD 20892-1857, USA.
| | | | | | | | | | | | | |
Collapse
|
291
|
Abstract
Platelet microparticles (PMP) are endogenous substances generated during the coagulation process in a hypercoagulable state. This study demonstrated that PMP promote the proliferation and survival, migration, and tube formation in human umbilical vein endothelial cells (HUVEC). Heat-treated PMP did not significantly decrease the angiogenic activity in HUVEC compared with that of the untreated PMP. Meanwhile when PMP were treated with activated charcoal, a procedure known to remove the lipid growth factors, the angiogenic activity was significantly reduced. These results suggest that the lipid component(s) of the PMP may be major active factor(s) and that protein component(s) may be minor contributor(s). PMP were also shown to augment endothelial progenitor cell differentiation in peripheral blood mononuclear cells. In addition, PMP-stimulated proliferation, chemotaxis and tube formation of the HUVEC was mediated via the Pertussis toxin-sensitive G protein, extracellular signal-regulated kinase and the phosphoinositide 3-kinase pathway. Herein, a new action of PMP was demonstrated to be a potent angiogenic stimulator. It is expected that in pathological states such as a growing tumour, PMP shed from the circulating platelets may reach adequate concentrations and that the elevated levels of PMP could contribute to florid formation of new blood vessels.
Collapse
Affiliation(s)
- Hyun Kyung Kim
- Hematologic Malignancies Branch, Division of Special Cancers, Research Institute & Hospital, National Cancer Centre, 809 Madu, Ilsan, Goyang, Gyeonggi 411-769, Republic of Korea.
| | | | | | | | | |
Collapse
|
292
|
Tamarat R, Silvestre JS, Le Ricousse-Roussanne S, Barateau V, Lecomte-Raclet L, Clergue M, Duriez M, Tobelem G, Lévy BI. Impairment in ischemia-induced neovascularization in diabetes: bone marrow mononuclear cell dysfunction and therapeutic potential of placenta growth factor treatment. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:457-66. [PMID: 14742252 PMCID: PMC1602274 DOI: 10.1016/s0002-9440(10)63136-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mechanisms that hinder ischemia-induced neovascularization in diabetes remain poorly understood. We hypothesized that endogenous bone marrow mononuclear cell (BM-MNC) dysfunction may contribute to the abrogated postischemic revascularization reaction associated with diabetes. We first analyzed the effect of diabetes (streptozotocin, 40 mg/kg) on BM-MNC pro-angiogenic potential in a model of surgically induced hindlimb ischemia. In nondiabetic animals, transplantation of BM-MNCs isolated from nondiabetic animals raised the ischemic/nonischemic angiographic score, capillary number, and blood flow recovery by 1.8-, 2.7-, and 2.2-fold, respectively, over that of PBS-injected nondiabetic animals (P < 0.05). Administration of diabetic BM-MNCs also improved the neovascularization reaction in ischemic hindlimbs of nondiabetic mice but to a lesser extent from that observed with nondiabetic BM-MNC transplantation. In diabetic mice, injection of nondiabetic BM-MNCs was still more efficient than that of diabetic BM-MNCs. Such BM-MNC dysfunction was associated with the impairment of diabetic BM-MNC capacity to differentiate into endothelial progenitor cells (EPCs) in vitro and to participate in vascular-like structure formation in a subcutaneous Matrigel plug. Placenta growth factor (PlGF) administration improved by sixfold the number of EPCs differentiated from diabetic BM-MNCs in vitro and enhanced ischemic/nonischemic angiographic score, capillary number and blood flow recovery by 1.9-, 1.5- and 1.6-fold, respectively, over that of untreated diabetic animals (P < 0.01). Endogenous BM-MNC pro-angiogenic potential was affected in diabetes. Therapeutic strategy based on PlGF administration restored such defects and improved postischemic neovascularization in diabetic mice.
Collapse
Affiliation(s)
- Radia Tamarat
- INSERM U541, Hôpital Lariboisière, IFR Circulation-Paris 7, Université Paris 7, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Smith EJ, Rothman MT. Antiproliferative coatings for the treatment of coronary heart disease:. what are the targets and which are the tools? J Interv Cardiol 2004; 16:475-83. [PMID: 14632944 DOI: 10.1046/j.1540-8183.2003.01058.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Since the advent of percutaneous coronary intervention (PCI) for stenosing coronary disease, restenosis has remained a clinical problem. Despite the emergence and evolution of coronary stents, the rate of restenosis following PCI is still 10-20%, and above 50% in high risk subgroups. With increased understanding of the pathophysiology of this process, a number of potential therapeutic targets have been identified, allowing the development of novel therapies against restenosis, which can now be delivered locally using stent platforms. Some of the reported clinical trial data utilizing drug-eluting stents (DES) have produced such profound reductions in clinical and angiographic restenosis that we have been tempted to believe we are on the brink of eradicating this process completely. As the initial excitement subsides, however, there is a need to decide whether these tools will remain effective in real-world interventional practice. In this article we review the pathophysiology of the restenotic process, and the biological targets of the DES therapies currently available in clinical practice. We attempt to define clinical target populations for DES therapy, and assess the impact on outcomes thus far. We consider the advantages that newly emergent stent coatings might offer, and whether targeting specific patient subgroups with unique antiproliferative agents may provide the best chance of limiting restenosis in high risk subgroups. Finally, we consider future strategies to prevent restenosis, with a movement away from the antiproliferative approach, and toward accelerating endothelialization.
Collapse
Affiliation(s)
- Elliot J Smith
- London Chest Hospital, Barts and the London NHS Trust, London, UK
| | | |
Collapse
|
294
|
Abstract
PURPOSE OF REVIEW Gene therapy utilizes viral and non-viral vectors to transfer genetic material into a host in the hope of treating disease. This article will review the potential applications of gene therapy in the treatment of cardiac and pulmonary diseases. RECENT FINDINGS The results from several phase I and II clinical trials have recently been published. In patients with ischemic heart disease, evidence of coronary revascularization has been observed after the delivery of angiogenic factors. Several trials have demonstrated a reduction in anginal symptoms, increases in exercise tolerance, and objective improvements in myocardial perfusion. Evidence of the transfer of therapeutic genes has been observed in human trials of inherited pulmonary diseases. Unfortunately, there has been little evidence of clinical efficacy in these studies. A variety of gene therapy strategies are being explored in the treatment of thoracic malignancies. Partial antitumor responses have occurred in some of the subjects enrolled in these studies. SUMMARY Significant progress has been made in the field of gene therapy in the past decade. Data from these early animal and human clinical trials will provide important information to guide future studies.
Collapse
Affiliation(s)
- Joseph W Szokol
- Department of Anesthesiology, Evanston Northwestern Healthcare, Evanston and Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60201, USA.
| | | | | | | |
Collapse
|
295
|
Park S, Tepper OM, Galiano RD, Capla JM, Baharestani S, Kleinman ME, Pelo CR, Levine JP, Gurtner GC. Selective recruitment of endothelial progenitor cells to ischemic tissues with increased neovascularization. Plast Reconstr Surg 2004; 113:284-93. [PMID: 14707648 DOI: 10.1097/01.prs.0000091169.51035.a5] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tissue ischemia remains a common problem in plastic surgery and one for which proangiogenic approaches have been investigated. Given the recent discovery of circulating endothelial stem or progenitor cells that are able to form new blood vessels, the authors sought to determine whether these cells might selectively traffic to regions of tissue ischemia and induce neovascularization. Endothelial progenitor cells were isolated from the peripheral blood of healthy human volunteers and expanded ex vivo for 7 days. Elevation of a cranially based random-pattern skin flap was performed in nude mice, after which they were injected with fluorescent-labeled endothelial progenitor cells (5 x 10(5); n = 15), fluorescent-labeled human microvascular endothelial cells (5 x 10(5); n = 15), or media alone (n = 15). Histologic examination demonstrated that endothelial progenitor cells were recruited to ischemic tissue and first appeared by postoperative day 3. Subsequently, endothelial progenitor cell numbers increased exponentially over time for the remainder of the study [0 cells/mm2 at day 0 (n = 3), 9.6 +/- 0.9 cells/mm2 at day 3 (n = 3), 24.6 +/- 1.5 cells/mm2 at day 7 (n = 3), and 196.3 +/- 9.6 cells/mm2 at day 14 (n = 9)]. At all time points, endothelial progenitor cells localized preferentially to ischemic tissue and healing wound edges, and were not observed in normal, uninjured tissues. Endothelial progenitor cell transplantation led to a statistically significant increase in vascular density in ischemic tissues by postoperative day 14 [28.7 +/- 1.2 in the endothelial progenitor cell group (n = 9) versus 18 +/- 1.1 in the control media group (n = 9) and 17.7 +/- 1.0 in the human microvascular endothelial cell group (n = 9; p < 0.01)]. Endothelial progenitor cell transplantation also showed trends toward increased flap survival [171.2 +/- 18 mm2 in the endothelial progenitor cell group (n = 12) versus 134.2 +/- 10 mm2 in the media group (n = 12) and 145.0 +/- 13 mm2 in the human microvascular endothelial cell group (n = 12)], but this did not reach statistical significance. These findings indicate that local tissue ischemia is a potent stimulus for the recruitment of circulating endothelial progenitor cells. Systemic delivery of endothelial progenitor cells increased neovascularization and suggests that autologous endothelial progenitor cell transplantation may have a role in the salvage of ischemic tissue.
Collapse
Affiliation(s)
- Sanghoon Park
- Laboratory of Microvascular Research and Vascular Tissue Engineering, Institute of Reconstructive Plastic Surgery, New York University, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
296
|
|
297
|
Tomita M, Yamada H, Adachi Y, Cui Y, Yamada E, Higuchi A, Minamino K, Suzuki Y, Matsumura M, Ikehara S. Choroidal neovascularization is provided by bone marrow cells. Stem Cells 2004; 22:21-26. [PMID: 14688388 DOI: 10.1634/stemcells.22-1-21] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Choroidal neovascularization (CNV) is a known cause of age-related macular degeneration (ARMD). Moreover, the most common cause of blindness in the elderly in advanced countries is ARMD with CNV. It has recently been shown that bone marrow cells (BMCs) can differentiate into various cell lineages in vitro and in vivo. Adults maintain a reservoir of hematopoietic stem cells included in BMCs that can enter the circulation to reach various organs in need of regeneration. It has recently been reported that endothelial progenitor cells (EPCs) included in BMCs are associated with neovascularization. We examine the role of BMCs in CNV using a model of CNV in adult mice. Using methods consisting of fractionated irradiation (6.0 Gy x 2) followed by bone marrow transplantation (BMT), adult mice were engrafted with whole BMCs isolated from transgenic mice expressing enhanced green fluorescent protein (EGFP). Three months after BMT, we confirmed that the hematopoietic cells in the recipients had been completely replaced with donor cells. We then carried out laser photocoagulation to induce CNV in chimeric mice (donor cells >95%). Two weeks after the laser photocoagulation, by which time CNV had occurred, immunohistochemical examination was carried out. The vascular wall cells of the CNV expressed both EGFP and CD31. These findings indicate that newly developed blood vessels in the CNV are derived from the BMCs and suggest that the inhibition of EPC mobilization from the bone marrow to the eyes could be a new approach to the fundamental treatment of CNV in ARMD.
Collapse
Affiliation(s)
- Minoru Tomita
- First Department of Pathology, Kansai Medical University, Moriguchi City, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
298
|
Henrich D, Hahn P, Wahl M, Wilhelm K, Dernbach E, Dimmeler S, Marzi I. Serum Derived from Multiple Trauma Patients Promotes the Differentiation of Endothelial Progenitor Cells In Vitro: Possible Role of Transforming Growth Factor-??1 and Vascular Endothelial Growth Factor165. Shock 2004; 21:13-6. [PMID: 14676678 DOI: 10.1097/01.shk.0000101669.49265.50] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ischemia in various organs and tissues takes place during and as a direct result of multiple trauma (MT). Bone marrow-derived endothelial progenitor cells (EPCs) are involved in neovascularization after ischemic incidences. Here, we report that serum derived from patients with MT stimulates differentiation of EPCs in vitro from peripheral blood mononuclear cells (PBMCs). EPCs were identified by DiL-Acetyl-LDL-uptake with concomitant UEA-I-lectin binding. A significant increase in EPC numbers was noted when PBMCs were cultivated for 72 h with the serum of MT patients (n = 25) obtained at 5 days. Furthermore, serum from MT patients enhanced the functional acting of EPCs to form prevascular structures in matrigel. Reverse transcription polymerase chain reaction analysis revealed gene expression of transforming growth factor (TGF)-beta1- and vascular endothelial growth factor (VEGF) receptors 1 and 2. Reverse transcription polymerase chain reaction analysis was based on further cultivated cell preparations, which contained at least 80% EPCs. Moreover, the addition of recombinant VEGF or low concentrations of TGF-beta increased EPC differentiation. In addition, neutralization of TGF-beta1 and of VEGF165 in MT serum using specific antibodies resulted in a significant decrease in EPC differentiation. Our data indicate that TGF-beta1 and VEGF165 play a pivotal role for EPC differentiation induced by serum of polytrauma patients.
Collapse
Affiliation(s)
- Dirk Henrich
- Department of Trauma Surgery, Johann-Wolfgang-Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
299
|
Abstract
Long before their existence was proven, work with blood islands pointed to the existence of hemangioblasts in the embryo, and it was widely accepted that such cells existed. In contrast, though evidence for adult hemangioblasts appeared at least as early as 1932, until quite recently, it was commonly assumed that there were no adult hemangioblasts. Over the past decade, these views have changed, and it is now generally accepted that a subset of bone marrow cells or their progeny can and do function as adult hemangioblasts. This chapter will examine the basic biology of bone marrow-derived hemangioblasts and endothelial cell progenitors (angioblasts) and the relationship of these adult cells to their embryonic counterparts. Efforts to define the endothelial cell progenitor phenotype will also be discussed, though to date, there is no consensus on the definitive adult phenotype, probably because there are multiple phenotypes and because the cells are plastic. Also examined are the putative roles of bone marrow-derived cells in vascular homeostasis and repair, including both their ability to differentiate and contribute directly to vascular repair, as well as to promote vascular growth by secreting pro-angiogenic factors. Finally, the use of bone marrow cells as therapeutic tools will be addressed.
Collapse
Affiliation(s)
- Gina C Schatteman
- Department of Exercise Science, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
300
|
Padilla L, Krötzsch E, Schalch P, Figueroa S, Miranda A, Rojas E, Esperante S, Villegas F, de la Garza AS, Di Silvio M. Administration of bone marrow cells into surgically induced fibrocollagenous tunnels induces angiogenesis in ischemic rat hindlimb model. Microsurgery 2003; 23:568-74. [PMID: 14705073 DOI: 10.1002/micr.10208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We established a comparative model of angiogenic induction in previously formed fibrocollagenous tunnels in rat inner thigh muscles. A unilateral hindlimb chronic ischemia model was performed in male Sprague-Dawley rats. A device was then inserted in the central portion of the inner thigh muscles. Vascularity in the ischemic limb was determined by means of an angiographic score, capillary/fiber ratio, and endothelial proliferation by histochemistry and immunohistochemistry. Autologous transplant of bone marrow, vascular endothelial growth factor (VEGF), or collagen-polyvinylpyrrolidone plus heparin induced significant vascularization of the ischemic hindlimb when compared to saline solution. However, the bone marrow group presented a higher angiographic score than the other two. No differences among groups were observed in capillary/fiber ratio or proliferation, except for the VEGF group, where capillary proliferating cells were significantly higher than in controls. Based on these results, bone marrow-derived progenitor cells may constitute a safe and viable alternative for the induction of therapeutic angiogenesis.
Collapse
MESH Headings
- Angiography
- Animals
- Biopsy, Needle
- Bone Marrow Cells
- Bone Marrow Transplantation/methods
- Disease Models, Animal
- Graft Survival
- Hindlimb/blood supply
- Immunohistochemistry
- Ischemia/diagnostic imaging
- Ischemia/pathology
- Ischemia/therapy
- Male
- Microscopy, Electron, Scanning
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/diagnostic imaging
- Muscle, Skeletal/ultrastructure
- Neovascularization, Physiologic/physiology
- Probability
- Rats
- Rats, Sprague-Dawley
- Sensitivity and Specificity
- Transplantation, Autologous
Collapse
Affiliation(s)
- Luis Padilla
- Department of Microsurgery and Experimental Surgery, Centro Médico Nacional 20 de Noviembre, I.S.S.SF.T.E., Mexico City, Mexico.
| | | | | | | | | | | | | | | | | | | |
Collapse
|