1051
|
Abstract
Statins promote the proliferation, migration, and survival of endothelial cells and bone marrow-derived endothelial progenitor cells (angioblasts) by stimulating the serine/threonine protein kinase Akt (also known as protein kinase B) pathway. Like vascular endothelial growth factor (VEGF), the statins promote angiogenesis and vasculogenesis. Therefore, Akt activation may explain some of the beneficial effects of the statins, including postnatal neovascularization.
Collapse
Affiliation(s)
- Joan Llevadot
- Servicio de Cardiología, Centro Cardiovascular Sant Jordi, Barcelona, Spain.
| | | |
Collapse
|
1052
|
Li TS, Hamano K, Suzuki K, Ito H, Zempo N, Matsuzaki M. Improved angiogenic potency by implantation of ex vivo hypoxia prestimulated bone marrow cells in rats. Am J Physiol Heart Circ Physiol 2002; 283:H468-73. [PMID: 12124190 DOI: 10.1152/ajpheart.00261.2002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Therapeutic angiogenesis can be induced by local implantation of bone marrow cells. We tried to enhance the angiogenic potential of this treatment by ex vivo hypoxia stimulation of bone marrow cells before implantation. Bone marrow cells were collected and cultured at 33 degrees C under 2% O(2)-5% CO(2)-90% N(2) (hypoxia) or 95% air-5% CO(2) (normoxia). Cells were also injected into the ischemic hindlimb of rats after 24 h of culture. Hypoxia culture increased the mRNA expression of vascular endothelial growth factor (VEGF), vascular endothelial (VE)-cadherin, and fetal liver kinase-1 (Flk-1) from 2.5- to fivefold in bone marrow cells. The levels of VEGF protein in the ischemic hindlimb were significantly higher 1 and 3 days after implantation with hypoxia-cultured cells than with normoxia-cultured or noncultured cells. The microvessel density and blood flow rate in the ischemic hindlimbs were also significantly (P < 0.001) higher 2 wk after implantation with hypoxia-cultured cells (89.7 +/- 5.5%) than with normoxia-cultured cells (67.0 +/- 9.6%) or noncultured cells (70.4 +/- 7.7%). Ex vivo hypoxia stimulation increased the VEGF mRNA expression and endothelial differentiation of bone marrow cells, which together contributed to improved therapeutic angiogenesis in the ischemic hindlimb after implantation.
Collapse
Affiliation(s)
- Tao-Sheng Li
- Department of Bioregulation, Yamaguchi University School of Medicine, 1-1-1 Monami-Kogushi, Ube, Yamaguchi, Japan 755-8505
| | | | | | | | | | | |
Collapse
|
1053
|
Murayama T, Tepper OM, Silver M, Ma H, Losordo DW, Isner JM, Asahara T, Kalka C. Determination of bone marrow-derived endothelial progenitor cell significance in angiogenic growth factor-induced neovascularization in vivo. Exp Hematol 2002; 30:967-72. [PMID: 12160849 DOI: 10.1016/s0301-472x(02)00867-6] [Citation(s) in RCA: 200] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Our laboratory and others recently provided evidence indicating that endothelial progenitor cells (EPCs) participate in postnatal neovascularization. However, the extent to which EPCs contribute to adult neovascularization remains unclear. To address this issue, we investigated the quantitative contribution of EPCs to newly formed vascular structures in an in vivo Matrigel plug assay and corneal micropocket assay. MATERIALS AND METHODS Lethally irradiated FVB mice were transplanted with bone marrow (BM) mononuclear cells from transgenic mice constitutively expressing beta-galactosidase (beta-gal) encoded by the lacZ gene regulated by an endothelial-specific tie-2 promoter. Reconstitution of the transplanted BM leads to the expression of lacZ in mice, which is restricted to BM cells expressing tie-2. RESULTS Four weeks after BM transplantation (BMT), tie-2/lacZ/BMT mice were implanted with either Matrigel containing fibroblast growth factor-2 subcutaneously or with a vascular endothelial growth factor pellet into the cornea. After 7 days, the Matrigel plug or the cornea was removed and analyzed by X-gal staining or immunostaining for beta-gal. X-gal staining of the Matrigel plug identified 5.7% +/- 1.2% of endothelial cells (ECs) as cells originated from BM-derived EPCs, whereas the more sensitive technique of immunofluorescence identified 26.5% +/- 0.9% of ECs. Similarly, EPC-derived cells comprised 5.0% +/- 2.4% and 17.7% +/- 3.6% of the ECs in corneal neovascularization identified by X-gal staining and immunohistochemistry, respectively. Ki67 staining of the corneal tissue documented that the majority of EPC-derived cells were actively proliferating in situ. CONCLUSION These findings suggest that BM-derived EPCs make a significant contribution to angiogenic growth factor-induced neovascularization that may account for up to 26% of all ECs.
Collapse
Affiliation(s)
- Toshinori Murayama
- Division of Cardiovascular Research, Department of Medicine, St. Elizabeth's Medical Center of Boston, Tufts University School of Medicine, Brighton, Mass 02135-2997, USA
| | | | | | | | | | | | | | | |
Collapse
|
1054
|
Soeda S, Shimada T, Koyanagi S, Yokomatsu T, Murano T, Shibuya S, Shimeno H. An attempt to promote neo-vascularization by employing a newly synthesized inhibitor of protein tyrosine phosphatase. FEBS Lett 2002; 524:54-8. [PMID: 12135741 DOI: 10.1016/s0014-5793(02)03002-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Vascular endothelial growth factor (VEGF) and its receptors play a key role in angiogenesis. VEGF receptor-2 (VEGFR-2) has a tyrosine kinase domain, and, once activated, induces the phosphorylation of cytoplasmic signaling proteins. The phosphorylated VEGFR-2 may be a substrate for intracellular protein tyrosine phosphatases (PTPs) which prevent VEGF signaling. We synthesized a series of alpha,alpha-difluoro(phenyl)methylphosphonic acids (DFPMPAs) which inhibit the action of PTP. In this study, we test their effects on VEGF-induced angiogenesis. DFPMPA-3, the most effective inhibitor of human PTP-1B, promoted tube formation by human umbilical vein endothelial cells (HUVEC) on Matrigel more effectively than any other DFPMPAs. The inhibitor promoted the VEGF-induced proliferation and migration of HUVEC by inhibiting the dephosphorylation of VEGFR-2. Its effectiveness was proven through neo-vascularization in mice. The present findings suggest that targeting PTP to promote therapeutic neo-vascularization may be a potential strategy.
Collapse
Affiliation(s)
- Shinji Soeda
- Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
1055
|
Sakakibara Y, Nishimura K, Tambara K, Yamamoto M, Lu F, Tabata Y, Komeda M. Prevascularization with gelatin microspheres containing basic fibroblast growth factor enhances the benefits of cardiomyocyte transplantation. J Thorac Cardiovasc Surg 2002; 124:50-6. [PMID: 12091808 DOI: 10.1067/mtc.2002.121293] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The effects of cell transplantation on the ischemic failing heart have already been documented. However, the area in and around infarct regions is not a good environment for cells to survive in because they are exposed to poor conditions in which certain requirements cannot be adequately supplied. We therefore designed a study to investigate the efficacy of prevascularization in ischemic regions before cell transplantation. METHODS Rats with myocardial infarction were randomized into 4 groups: 11 rats received a culture medium injection to the left ventricular wall (control group), 11 received fetal cardiomyocyte transplantation (TX group), 11 received gelatin hydrogel microspheres incorporating basic fibroblast growth factor (FGF group), and 11 received basic fibroblast growth factor pretreatment sequentially, followed by cardiomyocyte transplantation (FGF-TX group). Four weeks later, left ventricular function was assessed by means of echocardiography and cardiac catheterization. RESULTS In the FGF and FGF-TX groups neovascularization was found in the scar tissue 1 week later. The TX, FGF, and FGF-TX groups showed better fractional shortening than the control group (TX, FGF, FGF-TX, and control: 28% +/- 4.4%, 24% +/- 8.6%, 27% +/- 7.3%, and 17% +/- 4.6%, respectively; P <.01). Left ventricular maximum time-varying elastance was higher in the FGF-TX group than in the TX and FGF groups (FGF-TX, TX, and FGF: 0.52 +/- 0.23, 0.30 +/- 0.08, and 0.27 +/- 0.20 mm Hg/microL, respectively; P <.01). Histologically, more transplanted cells survived in the FGF-TX group than in the TX group. CONCLUSIONS Prevascularization with basic fibroblast growth factor-incorporated microspheres enhances the benefits of cardiomyocyte transplantation. We expect that this system will contribute to regeneration medicine through its extensive application to other growth factors.
Collapse
Affiliation(s)
- Yutaka Sakakibara
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
1056
|
Abstract
Observations made in the last few years support the existence of pathways, in adult humans and rodents, that allow adult stem cells to be surprisingly flexible in their differentiation repertoires. Termed plasticity, this property allows adult stem cells, assumed, until now, to be committed to generating a fixed range of progeny, to switch, when they have been relocated, to make other specialized sets of cells appropriate to their new niche. Reprogramming of some adult stem cells can occur in vivo; the stem cells normally resident in bone marrow appear particularly flexible and are able to contribute usefully to multiple recipient organs. This process produces cells with specialized structural and metabolic adaptations commensurate with their new locations. In a few examples, the degree of support is sufficient to assist or even rescue recipient mice from genetic defects. Some studies provide evidence for the expansion of the reprogrammed cells locally, but in most it remains possible that cells arrive and redifferentiate, but are no longer stem cells. Nevertheless, the fact that appropriately differentiated cells are delivered deep within organs simply by injection of bone marrow cells should make us think differently about the way that organs regenerate and repair. Migratory pathways for stem cells in adult organisms may exist that could be exploited to effect repairs using an individual's own stem cells, perhaps after gene therapy. Logical extensions of this concept are that a transplanted organ would become affected by the genetic susceptibilities of the recipient, alleles that re-express themselves via marrow-derived stem cells, and that plasticity after bone marrow transplantation would also transfer different phenotypes, affecting important parameters such as susceptibility to long-term complications of diabetes, or the ability to metabolize drugs in the liver. This article reviews some of the evidence for stem cell plasticity in rodents and man.
Collapse
Affiliation(s)
- Richard Poulsom
- Histopathology Unit, Cancer Research UK, London Research Institute, London, UK.
| | | | | | | |
Collapse
|
1057
|
Scholz D, Ziegelhoeffer T, Helisch A, Wagner S, Friedrich C, Podzuweit T, Schaper W. Contribution of arteriogenesis and angiogenesis to postocclusive hindlimb perfusion in mice. J Mol Cell Cardiol 2002; 34:775-87. [PMID: 12099717 DOI: 10.1006/jmcc.2002.2013] [Citation(s) in RCA: 256] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
UNLABELLED The goal of this study was to examine the mechanisms of vascular growth that lead to the restoration of perfusion in a peripheral vascular disease model in mice. We monitored blood flow recovery and measured vascular growth in inbred strains of mice following femoral artery occlusion. Acute collateral blood flow to the hindlimb was lowest in Balb/C mice, causing intense ischemia, and showed a slower recovery (more than 21 days to 50% normal) than C57Bl/6 which had a 7-fold higher acute collateral flow and a fast recovery (3 days). Collateral vessels were enlarged by proliferation of ECs and SMCs. Capillary density increased in the lower limbs of Balb/Cs (1.7-fold) and of sv129s. Tissue oxygen saturation recovered faster than flow in all strains. Morphometry of mature collaterals showed a diameter increase of 2.1-2.4 fold. The increase in total vessel wall area exceeded that of the femoral artery by 1.4-fold and the common lumenal area by 1.6-fold. Infusion of the growth factor peptide FGF-2 by osmotic minipump accelerated arteriogenesis but inhibited the angiogenic response probably because it prevented ischemia. CONCLUSION the speed of arteriogenesis is inversely related to the intensity of ischemia, and arteriogenesis is by far the most efficient mechanism to increase blood flow after femoral artery occlusion. De novo arteriogenesis was not observed.
Collapse
Affiliation(s)
- Dimitri Scholz
- Department of Exp. Cardiology, Max-Planck-Institute, Bad Nauheim, Germany.
| | | | | | | | | | | | | |
Collapse
|
1058
|
Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama T, Nishimura H, Losordo DW, Asahara T, Isner JM. Statin therapy accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells. Circulation 2002; 105:3017-24. [PMID: 12081997 DOI: 10.1161/01.cir.0000018166.84319.55] [Citation(s) in RCA: 689] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Primary and secondary prevention trials suggest that statins possess favorable effects independent of cholesterol reduction. We investigated whether statin therapy may also accelerate reendothelialization after carotid balloon injury. METHODS AND RESULTS Simvastatin treatment in 34 male Sprague-Dawley rats accelerated reendothelialization of the balloon-injured arterial segments (reendothelialized area at 2 weeks, 12.3+/-1.8 versus 5.4+/-1.1 mm2, P< 0.01) and resulted in a dose-dependent (0.2 or 1 mg/kg IP) significant reduction in neointimal thickening at 2, 3, and 4 weeks compared with saline-injected controls (n=18). To elucidate the mechanism, we investigated the contribution of bone marrow-derived endothelial progenitor cells (EPCs) by bone marrow transplantation from Tie2/lacZ mice to background mice or nude rats. X-gal staining of mouse carotid artery specimens revealed a 2.9-fold increase in the number of beta-gal-positive cells per square millimeter appearing on the carotid artery luminal surface at 2 weeks, and double-fluorescence immunohistochemistry disclosed a significant 5-fold increase in the number of double-positive cells (beta-gal, isolectin B4) on the luminal surface in carotid arteries of statin-treated nude rats (20+/-3 versus 4+/-1 cells/mm surface length, P<0.005). Statins increased circulating rat EPCs (2.4-fold at 2 weeks and 2.5-fold at 4 weeks, P<0.001) and induced adhesiveness of cultured human EPCs by upregulation of the integrin subunits alpha5, beta1, alpha(v), and beta5 of human EPCs as shown by reverse transcription-polymerase chain reaction and fluorescence-activated cell sorting. CONCLUSIONS These findings establish additional mechanisms by which statins may specifically preempt disordered vascular wall pathology and constitute physiological evidence that EPC mobilization represents a functionally relevant consequence of statin therapy.
Collapse
Affiliation(s)
- Dirk H Walter
- Department of Medicine (Cardiovascular Research), St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Mass 02135, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1059
|
Edelberg JM, Tang L, Hattori K, Lyden D, Rafii S. Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function. Circ Res 2002; 90:E89-93. [PMID: 12039806 DOI: 10.1161/01.res.0000020861.20064.7e] [Citation(s) in RCA: 228] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Delivery of young bone marrow-derived stem cells offers a novel approach for restoring the impaired senescent cardiac angiogenic function that may underlie the increased morbidity and mortality associated with ischemic heart disease in older individuals. Recently, we reported that alterations in endothelial cells of the aging heart lead to a dysregulation in the cardiac myocyte platelet-derived growth factor (PDGF)-B-induced paracrine pathway, which contributes to impaired cardiac angiogenic function. Based on these results, we hypothesized that cellular restoration of the PDGF pathway by bone marrow-derived endothelial precursor cells (EPCs) could reverse the aging-associated decline in angiogenic activity. In vitro studies revealed that young murine (3-month-old) bone marrow-derived EPCs recapitulated the cardiac myocyte-induced expression of PDGF-B, whereas EPCs from the bone marrow of aging mice (18-month-old) did not express PDGF-B when cultured in the presence of cardiac myocytes. Transplantation of young, but not old, genetically marked syngeneic bone marrow cells into intact, unirradiated aging mice that populated the endogenous senescent murine bone marrow incorporated into the neovasculature of subsequently transplanted syngeneic neonatal myocardium. Moreover, the young bone marrow-derived EPCs restored the senescent host angiogenic PDGF-B induction pathway and cardiac angiogenesis, with graft survival and myocardial activity in the aging murine host (cardiac allograft viability: 3-month-old controls, 8/8; 18-month-old controls, 1/8; 18-month-old donors receiving bone marrow from 3-month-old mice, 15/16; or 18-month-old mice, 0/6; P<0.05). These results may offer a foundation for the development of novel therapies for the prevention and treatment of cardiovascular disease associated with aging.
Collapse
Affiliation(s)
- Jay M Edelberg
- Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA.
| | | | | | | | | |
Collapse
|
1060
|
Marchetti S, Gimond C, Iljin K, Bourcier C, Alitalo K, Pouysségur J, Pagès G. Endothelial cells genetically selected from differentiating mouse embryonic stem cells incorporate at sites of neovascularization in vivo. J Cell Sci 2002; 115:2075-85. [PMID: 11973349 DOI: 10.1242/jcs.115.10.2075] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Large scale purification of endothelial cells is of great interest as it could improve tissue transplantation, reperfusion of ischemic tissues and treatment of pathologies in which an endothelial cell dysfunction exists. In this study, we describe a novel genetic approach that selects for endothelial cells from differentiating embryonic stem (ES) cells. Our strategy is based on the establishment of ES-cell clones that carry an integrated puromycin resistance gene under the control of a vascular endothelium-specific promoter, tie-1. Using EGFP as a reporter gene, we first confirmed the endothelial specificity of the tie-1 promoter in the embryoid body model and in cells differentiated in 2D cultures. Subsequently, tie-1-EGFP ES cells were used as recipients for the tie-1-driven puror transgene. The resulting stable clones were expanded and differentiated for seven days in the presence of VEGF before puromycin selection. As expected, puromycin-resistant cells were positive for EGFP and also expressed several endothelial markers, including CD31, CD34,VEGFR-1, VEGFR-2, Tie-1, VE-cadherin and ICAM-2. Release from the puromycin selection resulted in the appearance of α-smooth muscle actin-positive cells. Such cells became more numerous when the population was cultured on laminin-1 or in the presence of TGF-β1, two known inducers of smooth muscle cell differentiation. The hypothesis that endothelial cells or their progenitors may differentiate towards a smooth muscle cell phenotype was further supported by the presence of cells expressing both CD31 andα-smooth muscle actin markers. Finally, we show that purified endothelial cells can incorporate into the neovasculature of transplanted tumors in nude mice. Taken together, these results suggest that application of endothelial lineage selection to differentiating ES cells may become a useful approach for future pro-angiogenic and endothelial cell replacement therapies.
Collapse
Affiliation(s)
- Sandrine Marchetti
- Institute of Signaling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre Antoine Lacassagne, 33 avenue de Valombrose, Nice, France
| | | | | | | | | | | | | |
Collapse
|
1061
|
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) exert potent vasculoprotective effects. However, the potential contribution to angiogenesis is controversial. In the present study, we demonstrate that atorvastatin dose-dependently affects endothelial cell migration and angiogenesis. In vivo relevant concentrations of 0.01 to 0.1 micromol/L atorvastatin or mevastatin promote the migration of mature endothelial cells and tube formation. Moreover, atorvastatin also increases migration and the potency to form vessel structures of circulating endothelial progenitor cells, which may contribute to vasculogenesis. In contrast, higher concentrations (>0.1 micromol/L atorvastatin) block angiogenesis and migration by inducing endothelial cell apoptosis. The dose-dependent promigratory and proangiogenic effects of atorvastatin on mature endothelial cells are correlated with the activation of the phosphatidylinositol 3-kinase-Akt pathway, as determined by the phosphorylation of Akt and endothelial NO synthase (eNOS) at Ser1177. In addition, the stimulation of migration and tube formation was blocked by phosphatidylinositol 3-kinase inhibitors. In contrast, the well-established stabilization of eNOS mRNA was achieved only at higher concentrations, suggesting that posttranscriptional activation rather than an increase in eNOS expression mediates the proangiogenic effect of atorvastatin. Taken together, these data suggest that statins exert a double-edged role in angiogenesis signaling by promoting the migration of mature endothelial cells and endothelial progenitor cells at low concentrations, whereas the antiangiogenic effects were achieved only at high concentrations.
Collapse
Affiliation(s)
- Carmen Urbich
- Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Frankfurt, Germany
| | | | | | | |
Collapse
|
1062
|
Iwaguro H, Yamaguchi JI, Kalka C, Murasawa S, Masuda H, Hayashi SI, Silver M, Li T, Isner JM, Asahara T. Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation 2002; 105:732-8. [PMID: 11839630 DOI: 10.1161/hc0602.103673] [Citation(s) in RCA: 379] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Previous studies have established that bone marrow-derived endothelial progenitor cells (EPCs) are present in the systemic circulation. In the current study, we investigated the hypothesis that gene transfer can be used to achieve phenotypic modulation of EPCs. METHODS AND RESULTS In vitro, ex vivo murine vascular endothelial growth factor (VEGF) 164 gene transfer augmented EPC proliferative activity and enhanced adhesion and incorporation of EPCs into quiescent as well as activated endothelial cell monolayers. To determine if such phenotypic modulation may facilitate therapeutic neovascularization, heterologous EPCs transduced with adenovirus encoding VEGF were administered to athymic nude mice with hindlimb ischemia; neovascularization and blood flow recovery were both improved, and limb necrosis/autoamputation were reduced by 63.7% in comparison with control animals. The dose of EPCs used for the in vivo experiments was 30 times less than that required in previous trials of EPC transplantation to improve ischemic limb salvage. Necropsy analysis of animals that received DiI-labeled VEGF-transduced EPCs confirmed that enhanced EPC incorporation demonstrated in vitro contributed to in vivo neovascularization as well. CONCLUSIONS In vitro, VEGF EPC gene transfer enhances EPC proliferation, adhesion, and incorporation into endothelial cell monolayers. In vivo, gene-modified EPCs facilitate the strategy of cell transplantation to augment naturally impaired neovascularization in an animal model of experimentally induced limb ischemia.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cell Count
- Cell Division/drug effects
- Cell Division/physiology
- Cells, Cultured
- Dendritic Cells
- Disease Models, Animal
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/metabolism
- Endothelial Growth Factors/pharmacology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Gene Transfer, Horizontal
- Genetic Therapy/methods
- Genetic Vectors/genetics
- Genetic Vectors/metabolism
- Genetic Vectors/pharmacology
- Humans
- Ischemia/drug therapy
- Ischemia/pathology
- Ischemia/physiopathology
- Lymphokines/genetics
- Lymphokines/metabolism
- Lymphokines/pharmacology
- Mice
- Mice, Nude
- Microcirculation/drug effects
- Microcirculation/metabolism
- Microcirculation/pathology
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Neovascularization, Physiologic/drug effects
- Regeneration/drug effects
- Stem Cell Transplantation
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
- Transgenes
- Treatment Outcome
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Hideki Iwaguro
- Division of Cardiovascular Research and Medicine, St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Mass 02135, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1063
|
Abstract
Somatic gene therapy of vascular diseases is a promising new field in modern medicine. Recent advancements in gene transfer technology have greatly evolved our understanding of the pathophysiologic role of candidate disease genes. With this knowledge, the expression of selective gene products provides the means to test the therapeutic use of gene therapy in a multitude of medical conditions. In addition, with the completion of genome sequencing programs, gene transfer can be used also to study the biologic function of novel genes in vivo. Novel genes are delivered to targeted tissue via several different vehicles. These vectors include adenoviruses, retroviruses, plasmids, plasmid/liposomes, and oligonucleotides. However, each one of these vectors has inherent limitations. Further investigations into developing delivery systems that not only allow for efficient, targeted gene transfer, but also are stable and nonimmunogenic, will optimize the clinical application of gene therapy in vascular diseases. This review further discusses the available mode of gene delivery and examines six major areas in vascular gene therapy, namely prevention of restenosis, thrombosis, hypertension, atherosclerosis, peripheral vascular disease in congestive heart failure, and ischemia. Although we highlight some of the recent advances in the use of gene therapy in treating vascular disease discovered primarily during the past two years, many excellent studies published during that period are not included in this review due to space limitations. The following is a selective review of practical uses of gene transfer therapy in vascular diseases. This review primarily covers work performed in the last 2 years. For earlier work, the reader may refer to several excellent review articles. For instance, Belalcazer et al. (6) reviewed general aspects of somatic gene therapy and the different vehicles used for the delivery of therapeutic genes. Gene therapy in restenosis and stimulation of angiogenesis in the cardiac muscle are discussed in reviews by several investigators (13,26,57,74,83). In another review, Meyerson et al. (43) discuss advances in gene therapy for vascular proliferative disorders and chronic peripheral and cardiac ischemia.
Collapse
Affiliation(s)
- M J McKay
- Department of Medicine, Sarver Heart Center, Cardiology Section 111C, University of Arizona, 3601 South 6th Avenue, Tucson, AZ 85723, USA
| | | |
Collapse
|
1064
|
Abstract
Gene therapy is proving likely to be a viable alternative to conventional therapies in coronary artery disease and heart failure. Phase 1 clinical trials indicate high levels of safety and clinical benefits with gene therapy using angiogenic growth factors in myocardial ischaemia. Although gene therapy for heart failure is still at the pre-clinical stage, experimental data indicate that therapeutic angiogenesis using short-term gene expression may elicit functional improvement in affected individuals.
Collapse
Affiliation(s)
- Jeffrey M Isner
- Department of Medicne, St. Elizabeth's Medical Center, Tufts School of Medicine, Boston, MA 02135, USA
| |
Collapse
|
1065
|
Harraz M, Jiao C, Hanlon HD, Hartley RS, Schatteman GC. CD34- blood-derived human endothelial cell progenitors. Stem Cells 2002; 19:304-12. [PMID: 11463950 DOI: 10.1634/stemcells.19-4-304] [Citation(s) in RCA: 215] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A subset of adult peripheral blood leukocytes functions as endothelial cell progenitors called angioblasts. They can incorporate into the vasculature in animal models of neovascularization and accelerate the restoration of blood flow to mouse ischemic limbs. Earlier reports suggested that CD34-expressing (CD34+) but not CD34+ cell-depleted (CD34-) leukocytes can differentiate into endothelial cells (EC) in vitro and in vivo. Recent findings suggest that CD14+ cells, which are typically CD34-, also have angioblast-like properties in vitro. To determine the identity of angioblasts, the potential of CD34+, CD34-, CD34- CD14+, and CD34- CD14- cells to produce EC was compared. We show that a subset of monocyte (CD34- CD14+)-enriched cells can take on an EC-like phenotype in culture, but that the EC-like cells also express dendritic cell antigens. These findings suggest that monocytes differentiate into macrophages, dendritic cells, or EC depending on environmental cues. The data also demonstrate that angioblasts are more abundant in the blood than previously thought. Finally, we demonstrate that CD34- and CD34- CD14+ cells incorporate into the endothelium of blood vessels in mouse ischemic limbs. However, incorporation of these cells requires co-injection with CD34+ cells, indicating that leukocyte-leukocyte interactions may play a critical role in governing angioblast behavior in vivo.
Collapse
Affiliation(s)
- M Harraz
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
1066
|
Reynolds LE, Wyder L, Lively JC, Taverna D, Robinson SD, Huang X, Sheppard D, Hynes RO, Hodivala-Dilke KM. Enhanced pathological angiogenesis in mice lacking beta3 integrin or beta3 and beta5 integrins. Nat Med 2002; 8:27-34. [PMID: 11786903 DOI: 10.1038/nm0102-27] [Citation(s) in RCA: 487] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibition of alphavbeta3 or alphavbeta5 integrin function has been reported to suppress neovascularization and tumor growth, suggesting that these integrins are critical modulators of angiogenesis. Here we report that mice lacking beta3 integrins or both beta3 and beta5 integrins not only support tumorigenesis, but have enhanced tumor growth as well. Moreover, the tumors in these integrin-deficient mice display enhanced angiogenesis, strongly suggesting that neither beta3 nor beta5 integrins are essential for neovascularization. We also observed that angiogenic responses to hypoxia and vascular endothelial growth factor (VEGF) are augmented significantly in the absence of beta3 integrins. We found no evidence that the expression or functions of other integrins were altered as a consequence of the beta3 deficiency, but we did observe elevated levels of VEGF receptor-2 (also called Flk-1) in beta3-null endothelial cells. These data indicate that alphavbeta3 and alphavbeta5 integrins are not essential for vascular development or pathological angiogenesis and highlight the need for further evaluation of the mechanisms of action of alphav-integrin antagonists in anti-angiogenic therapeutics.
Collapse
Affiliation(s)
- Louise E Reynolds
- Cell Adhesion and Disease Laboratory, Richard Dimbleby Department, Imperial Cancer Research Fund, St. Thomas' Hospital, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
1067
|
Francis SC, Raizada MK, Mangi AA, Melo LG, Dzau VJ, Vale PR, Isner JM, Losordo DW, Chao J, Katovich MJ, Berecek KH. Genetic targeting for cardiovascular therapeutics: are we near the summit or just beginning the climb? Physiol Genomics 2001; 7:79-94. [PMID: 11773594 DOI: 10.1152/physiolgenomics.00073.2001] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This article is based on an Experimental Biology symposium held in April 2001 and presents the current status of gene therapy for cardiovascular diseases in experimental studies and clinical trials. Evidence for the use of gene therapy to limit neointimal hyperplasia and confer myocardial protection was presented, and it was found that augmenting local nitric oxide (NO) production using gene transfer (GT) of NO synthase or interruption of cell cycle progression through a genetic transfer of cell cycle regulatory genes limited vascular smooth muscle hyperplasia in animal models and infra-inguinal bypass patients. The results of application of vascular endothelial growth factor (VEGF) GT strategies for therapeutic angiogenesis in critical limb and myocardial ischemia in pilot clinical trials was reviewed. In addition, experimental evidence was presented that genetic manipulation of peptide systems (i.e., the renin-angiotensin II system and the kallikrein-kinin system) was effective in the treatment of systemic cardiovascular diseases such as hypertension, heart failure, and renal failure. Although, as of yet, there are no well controlled human trials proving the clinical benefits of gene therapy for cardiovascular diseases, the data presented here in animal models and in human subjects show that genetic targeting is a promising and encouraging modality, not only for the treatment and long-term control of cardiovascular diseases, but for their prevention as well.
Collapse
Affiliation(s)
- S C Francis
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida Brain Institute, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1068
|
Feraud O, Cao Y, Vittet D. Embryonic stem cell-derived embryoid bodies development in collagen gels recapitulates sprouting angiogenesis. J Transl Med 2001; 81:1669-81. [PMID: 11742037 DOI: 10.1038/labinvest.3780380] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The formation of new blood vessels proceeds by both vasculogenesis and angiogenesis. The development of models, which fully recapitulate spatio-temporal events involved during these processes, are crucial to fully understand their mechanisms of regulation. In vitro differentiation of murine embryonic stem (ES) cells has been shown to be a useful tool to investigate factors and genes potentially involved in vasculogenesis (Hirashima et al, 1999; Risau et al, 1988; Vittet et al, 1996; Wang et al, 1992; Wartenberg et al, 1998). We asked here whether this model system can also recapitulate angiogenesis, which may offer new means to study mechanisms involved in this process. ES-derived embryoid bodies (EBs) obtained after 11 days of differentiation, in which a primitive vascular network had formed, were then subcultured into a type I collagen matrix. In the presence of angiogenic growth factors, EBs rapidly developed branching pseudopods. Whole mount immunostainings with a PECAM antibody revealed that more than 75% EBs displayed, within a few days, a large number of endothelial outgrowths that can give tube-like structures with concomitant differentiation of alpha-smooth muscle actin positive cells, thus evoking sprouting angiogenesis. High expression levels of flk1 (VEGFR2), flt1 (VEGFR1), tie-1, and tie-2 are also found, indicating that budding endothelial cells displayed an angiogenic phenotype. The endothelial sprouting response was specifically induced by angiogenic factors with a major contribution of vascular endothelial growth factor (VEGF). Known angiostatic agents, such as platelet factor 4 (PF4), angiostatin, and endostatin inhibited the formation of endothelial sprouts induced by angiogenic factors. Moreover, consistent with the in vivo phenotype, VE-cadherin deficient EBs failed to develop angiogenesis in this model. ES cell differentiation can then recapitulate, in addition to vasculogenesis, the early stages of sprouting angiogenesis. This model system, in which genetic modifications can be easily introduced, may be of particular interest to investigate unsolved questions and molecular mechanisms involved in blood vessel formation.
Collapse
Affiliation(s)
- O Feraud
- Laboratoire de Transgenèse et Différenciation Cellulaire, CEA Grenoble, DBMS, Grenoble, France
| | | | | |
Collapse
|
1069
|
Badylak SF, Park K, Peppas N, McCabe G, Yoder M. Marrow-derived cells populate scaffolds composed of xenogeneic extracellular matrix. Exp Hematol 2001; 29:1310-8. [PMID: 11698127 DOI: 10.1016/s0301-472x(01)00729-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The source of cells that participate in wound repair directly affects outcome. The extracellular matrix (ECM) and other acellular biomaterials have been used as therapeutic scaffolds for cell attachment and proliferation and as templates for tissue repair. The ECM consists of structural and functional proteins that influence cell attachment, gene expression patterns, and the differentiation of cells. OBJECTIVE The objective of this study was to determine if the composition of acellular matrix scaffolds affects the recruitment of bone marrow-derived cellular elements that populate the scaffolds in vivo. METHODS Scaffolds composed of porcine tissue ECM, purified Type I collagen, poly(L)lactic coglycolic acid (PLGA), or a mixture of porcine ECM and PLGA were implanted into subcutaneous pouches on the dorsum of mice. The origin of cells that populated the matrices was determined by first performing bone marrow transplantation to convert the marrow of glucose phosphate isomerase 1b (Gpi-1(b)) mice to cells expressing glucose phosphate isomerase 1a (Gpi-1(a)). RESULTS A significant increase in Gpi-1(a) expressing cells was present in sites implanted with the porcine ECM compared to sites implanted with either Type I collagen or PLGA. Use of recipient mice transplanted with marrow cells that expressed beta-galactosidase confirmed that the majority of cells that populated and remodeled the naturally occurring porcine ECM were marrow derived. Addition of porcine ECM to the PLGA scaffold caused a significant increase in the number of marrow-derived cells that became part of the remodeled implant site. CONCLUSION The composition of bioscaffolds affects the cellular recruitment pattern during tissue repair. ECM scaffolds facilitate the recruitment of marrow-derived cells into sites of remodeling.
Collapse
Affiliation(s)
- S F Badylak
- Department of Biomedical Engineering, Purdue University, 1296 Potter Building, West Lafayette, IN 47907-1296, USA.
| | | | | | | | | |
Collapse
|
1070
|
Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A, Heissig B, Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett NR, Crystal RG, Moore MA, Hajjar KA, Manova K, Benezra R, Rafii S. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 2001; 7:1194-201. [PMID: 11689883 DOI: 10.1038/nm1101-1194] [Citation(s) in RCA: 1372] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1+/-Id3-/- host, which were associated with VEGF-receptor-1-positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.
Collapse
Affiliation(s)
- D Lyden
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1071
|
Stevens T, Rosenberg R, Aird W, Quertermous T, Johnson FL, Garcia JG, Hebbel RP, Tuder RM, Garfinkel S. NHLBI workshop report: endothelial cell phenotypes in heart, lung, and blood diseases. Am J Physiol Cell Physiol 2001; 281:C1422-33. [PMID: 11600404 DOI: 10.1152/ajpcell.2001.281.5.c1422] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelium critically regulates systemic and pulmonary vascular function, playing a central role in hemostasis, inflammation, vasoregulation, angiogenesis, and vascular growth. Indeed, the endothelium integrates signals originating in the circulation with those in the vessel wall to coordinate vascular function. This highly metabolic role differs significantly from the historic view of endothelium, in which it was considered to be merely an inert barrier. New lines of evidence may further change our understanding of endothelium, in regard to both its origin and function. Embryological studies suggest that the endothelium arises from different sites, including angiogenesis of endothelium from macrovascular segments and vasculogenesis of endothelium from microcirculatory segments. These findings suggest an inherent phenotypic distinction between endothelial populations based on their developmental origin. Similarly, diverse environmental cues influence endothelial cell phenotype, critical to not only normal function but also the function of a diseased vessel. Consequently, an improved understanding of site-specific endothelial cell function is essential, particularly with consideration to environmental stimuli present both in the healthy vessel and in development of vasculopathic disease states. The need to examine endothelial cell phenotypes in the context of vascular function served as the basis for a recent workshop sponsored by the National Heart, Lung, and Blood Institute (NHLBI). This report is a synopsis of pertinent topics that were discussed, and future goals and research opportunities identified by the participants of the workshop are presented.
Collapse
Affiliation(s)
- T Stevens
- Department of Pharmacology, University of South Alabama College of Medicine, Mobile, Alabama 36688, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1072
|
Vale PR, Losordo DW, Symes JF, Isner JM. [Growth factors for therapeutic angiogenesis in cardiovascular diseases]. Rev Esp Cardiol 2001; 54:1210-24. [PMID: 11591302 DOI: 10.1016/s0300-8932(01)76480-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Therapeutic angiogenesis based on the administration of growth factors with angiogenic activity allows enhancement of collateral vessels able to palliate insufficient tissue perfusion secondary to obstruction of native arteries. At present, this type of therapy is addressed to patients that fail to respond to conventional treatment (surgical or percutaneous revascularization). The most extensively investigated angiogenic growth factors are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). These cytokines can be administered either as recombinant proteins or as the genes encoding for these proteins. Both approaches have pros and cons that are under investigation in animal models and in clinical studies. Although clinical trials consist so far of small, often non-randomized series, preliminary results are promising. For example, administration of VEGF or FGF has been associated to objective evidence of increased tissue perfusion in patients with myocardial ischemia, and to a significant improvement of pain and ischemia in patients with peripheral arterial disease. Contrarily to expected, these interventions have been associated to scant adverse side effects, although larger clinical trials will be necessary in order to prove the safety and effectiveness of these interventions. Nevertheless, it seems clear that it is feasible to induce effective therapeutic angiogenesis in selected patients without significant associated toxicity.
Collapse
Affiliation(s)
- P R Vale
- Departamento de Cardiología, St. Elizabeth's Medical Center, Tuft's University School of Medicine, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
1073
|
Young HE, Steele TA, Bray RA, Hudson J, Floyd JA, Hawkins K, Thomas K, Austin T, Edwards C, Cuzzourt J, Duenzl M, Lucas PA, Black AC. Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. THE ANATOMICAL RECORD 2001; 264:51-62. [PMID: 11505371 DOI: 10.1002/ar.1128] [Citation(s) in RCA: 337] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study details the profile of 13 cell surface cluster differentiation markers on human reserve stem cells derived from connective tissues. Stem cells were isolated from the connective tissues of dermis and skeletal muscle derived from fetal, mature, and geriatric humans. An insulin/dexamethasone phenotypic bioassay was used to determine the identity of the stem cells from each population. All populations contained lineage-committed myogenic, adipogenic, chondrogenic, and osteogenic progenitor stem cells as well as lineage-uncommitted pluripotent stem cells capable of forming muscle, adipocytes, cartilage, bone, fibroblasts, and endothelial cells. Flow cytometric analysis of adult stem cell populations revealed positive staining for CD34 and CD90 and negative staining for CD3, CD4, CD8, CD11c, CD33, CD36, CD38, CD45, CD117, Glycophorin-A, and HLA DR-II.
Collapse
Affiliation(s)
- H E Young
- Division of Basic Medical Science, Mercer University School of Medicine, Macon, Georgia 31207, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1074
|
Kaushal S, Amiel GE, Guleserian KJ, Shapira OM, Perry T, Sutherland FW, Rabkin E, Moran AM, Schoen FJ, Atala A, Soker S, Bischoff J, Mayer JE. Functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. Nat Med 2001; 7:1035-40. [PMID: 11533707 PMCID: PMC2818999 DOI: 10.1038/nm0901-1035] [Citation(s) in RCA: 587] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Arterial conduits are increasingly preferred for surgical bypass because of inherent functional properties conferred by arterial endothelial cells, especially nitric oxide production in response to physiologic stimuli. Here we tested whether endothelial progenitor cells (EPCs) can replace arterial endothelial cells and promote patency in tissue-engineered small-diameter blood vessels (4 mm). We isolated EPCs from peripheral blood of sheep, expanded them ex vivo and then seeded them on decellularized porcine iliac vessels. EPC-seeded grafts remained patent for 130 days as a carotid interposition graft in sheep, whereas non-seeded grafts occluded within 15 days. The EPC-explanted grafts exhibited contractile activity and nitric-oxide-mediated vascular relaxation that were similar to native carotid arteries. These results indicate that EPCs can function similarly to arterial endothelial cells and thereby confer longer vascular-graft survival. Due to their unique properties, EPCs might have other general applications for tissue-engineered structures and in treating vascular diseases.
Collapse
Affiliation(s)
- Sunjay Kaushal
- Department of Cardiac Surgery, Children’s Hospital, Boston, MA 02115
- Department of Surgery, Children’s Hospital, Boston, MA 02115
| | - Gilad E. Amiel
- Department of Urology, Children’s Hospital, Boston, MA 02115
| | | | - Oz M. Shapira
- Department of Cardiothoracic Surgery, Boston University School of Medicine, Boston, MA 02118
| | - Tjorvi Perry
- Department of Cardiac Surgery, Children’s Hospital, Boston, MA 02115
| | | | - Elena Rabkin
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
| | - Adrian M. Moran
- Department of Cardiology, Children’s Hospital, Boston, MA 02115
| | | | - Anthony Atala
- Department of Urology, Children’s Hospital, Boston, MA 02115
| | - Shay Soker
- Department of Urology, Children’s Hospital, Boston, MA 02115
| | - Joyce Bischoff
- Department of Surgery, Children’s Hospital, Boston, MA 02115
| | - John E. Mayer
- Department of Cardiac Surgery, Children’s Hospital, Boston, MA 02115
| |
Collapse
|
1075
|
Ojeifo JO, Lee HR, Rezza P, Su N, Zwiebel JA. Endothelial cell-based systemic gene therapy of metastatic melanoma. Cancer Gene Ther 2001; 8:636-48. [PMID: 11593332 DOI: 10.1038/sj.cgt.7700356] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2001] [Indexed: 11/08/2022]
Abstract
Cancer metastasis accounts for a significant proportion of morbidity and mortality in patients. Effective means of treating disseminated disease remains elusive. The purpose of this study was to determine whether genetically modified endothelial cells (GMEC) can selectively target and deliver recombinant therapeutic molecules to sites of tumor metastases. Following the establishment of lung metastases of 4T1 mammary tumor in mice, intravenously (i.v.) administered, lacZ transgene-expressing endothelial cells (lacZ-GMEC) accumulated at the tumor sites. An average of 32% and 90% of the pulmonary metastases were X-gal stained following one and three tail vein injections of 10(5) lacZ-GMEC, respectively. The linear pattern of X-gal staining seen within the tumor sites and the histological appearance of the tumor vasculature were consistent with the incorporation of lacZ-GMEC into blood vessels. In C57Bl/6 mice harboring lung metastases of melanoma, the administration of three sequential i.v. injections of 10(5) endothelial cells expressing a human interleukin 2 transgene abrogated the tumor metastases and prolonged survival of the animals. These results demonstrate that i.v.-administered GMEC can selectively accumulate, survive, and stably express exogenous genes at multiple tumor sites. These findings support a role for i.v.-administered GMEC as a potential therapeutic strategy for the systemic treatment of cancer metastases.
Collapse
Affiliation(s)
- J O Ojeifo
- Department of Oncology, Georgetown University Medical Center, N.W. Washington, DC 20007, USA
| | | | | | | | | |
Collapse
|
1076
|
|
1077
|
Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Tsutsumi Y, Ozono R, Masaki H, Mori Y, Iba O, Tateishi E, Kosaki A, Shintani S, Murohara T, Imaizumi T, Iwasaka T. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation 2001; 104:1046-52. [PMID: 11524400 DOI: 10.1161/hc3501.093817] [Citation(s) in RCA: 588] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Bone marrow implantation (BMI) was shown to enhance angiogenesis in a rat ischemic heart model. This preclinical study using a swine model was designed to test the safety and therapeutic effectiveness of BMI. METHODS AND RESULTS BM-derived mononuclear cells (BM-MNCs) were injected into a zone made ischemic by coronary artery ligation. Three weeks after BMI, regional blood flow and capillary densities were significantly higher (4.6- and 2.8-fold, respectively), and cardiac function was improved. Angiography revealed that there was a marked increase (5.7-fold) in number of visible collateral vessels. Implantation of porcine coronary microvascular endothelial cells (CMECs) did not cause any significant increase in capillary densities. Labeled BM-MNCs were incorporated into approximately 31% of neocapillaries and corresponded to approximately 8.7% of macrophages but did not actively survive as myoblasts or fibroblasts. There was no bone formation by osteoblasts or malignant ventricular arrhythmia. Time-dependent changes in plasma levels for cardiac enzymes (troponin I and creatine kinase-MB) did not differ between the BMI, CMEC, and medium-alone implantation groups. BM-MNCs contained 16% of endothelial-lineage cells and expressed basic fibroblast growth factor>>vascular endothelial growth factor>angiopoietin 1 mRNAs, and their cardiac levels were significantly upregulated by BMI. Cardiac interleukin-1beta and tumor necrosis factor-alpha mRNA expression were also induced by BMI but not by CMEC implantation. BM-MNCs were actively differentiated to endothelial cells in vitro and formed network structure with human umbilical vein endothelial cells. CONCLUSIONS BMI may constitute a novel safety strategy for achieving optimal therapeutic angiogenesis by the natural ability of the BM cells to secrete potent angiogenic ligands and cytokines as well as to be incorporated into foci of neovascularization.
Collapse
Affiliation(s)
- H Kamihata
- Department of Medicine II and Cardiovascular Center, Kansai Medical University, Moriguchi, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1078
|
Emanueli C, Madeddu P. Angiogenesis gene therapy to rescue ischaemic tissues: achievements and future directions. Br J Pharmacol 2001; 133:951-8. [PMID: 11487503 PMCID: PMC1572862 DOI: 10.1038/sj.bjp.0704155] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ischaemic diseases are characterized by an impaired supply of blood resulting from narrowed or blocked arteries that starve tissues of needed nutrients and oxygen. Coronary-atherosclerosis induced myocardial infarction is one of the leading causes of mortality in developed countries. Ischaemic disease also affects the lower extremities. Considerable advances in both surgical bypassing and percutaneous revascularization techniques have been reached. However, many patients cannot benefit from these therapies because of the extension of arterial occlusion and/or microcirculation impairment. Consequently, the need for alternative therapeutic strategies is compelling. An innovative approach consists of stimulating collateral vessel growth, a natural host defence response that intervenes upon occurrence of critical reduction in tissue perfusion (Isner & Asahara, 1999). This review will debate the relevance of therapeutic angiogenesis for promotion of tissue repair. The following issues will receive attention: (a) vascular growth patterns, (b) delivery systems for angiogenesis gene transfer, (c) achievements of therapeutic angiogenesis in myocardial and peripheral ischaemia, and (d) future directions to improve effectiveness and safety of vascular gene therapy.
Collapse
Affiliation(s)
- Costanza Emanueli
- Cardiovascular Medicine and Gene Therapy Section, National Laboratory of the National Institute of Biostructures and Biosystems, Osilo, Italy
| | - Paolo Madeddu
- Cardiovascular Medicine and Gene Therapy Section, National Laboratory of the National Institute of Biostructures and Biosystems, Osilo, Italy
- Department of Internal Medicine, Medical University of Sassari, Sassari, Italy
- Author for correspondence:
| |
Collapse
|
1079
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1080
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1081
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1082
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ Res 2001; 89:E1-7. [PMID: 11440984 DOI: 10.1161/hh1301.093953] [Citation(s) in RCA: 1599] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow-derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by approximately 40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (R=-0.394, P=0.002) and CD34-/KDR-positive cells (R=-0.537, P<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (P<0.001) and CD34-/KDR-positive cells (P=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (P=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (R=-0.484, P=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (P=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- M Vasa
- Division of Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1083
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1084
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1085
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1086
|
Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and Migratory Activity of Circulating Endothelial Progenitor Cells Inversely Correlate With Risk Factors for Coronary Artery Disease. Circ Res 2001. [DOI: 10.1161/hh1301.093953 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
—Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow–derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by ≈40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (
R
=−0.394,
P
=0.002) and CD34-/KDR-positive cells (
R
=−0.537,
P
<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (
P
<0.001) and CD34-/KDR-positive cells (
P
=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (
P
=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (
R
=−0.484,
P
=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (
P
=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- Mariuca Vasa
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stephan Fichtlscherer
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Alexandra Aicher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Klaudia Adler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Carmen Urbich
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Hans Martin
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Division of Molecular Cardiology, Department of Internal Medicine IV (M.V., S.F., A.A., K.A., C.U., A.M.Z., S.D.), and Department of Hematology, Internal Medicine III (H.M.), University of Frankfurt, Germany
| |
Collapse
|
1087
|
Vasa M, Fichtlscherer S, Adler K, Aicher A, Martin H, Zeiher AM, Dimmeler S. Increase in circulating endothelial progenitor cells by statin therapy in patients with stable coronary artery disease. Circulation 2001; 103:2885-90. [PMID: 11413075 DOI: 10.1161/hc2401.092816] [Citation(s) in RCA: 674] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Therapeutic neovascularization may constitute an important strategy to salvage tissue from critical ischemia. Circulating bone marrow-derived endothelial progenitor cells (EPCs) were shown to augment the neovascularization of ischemic tissue. In addition to lipid-lowering activity, hydroxymethyl glutaryl coenzyme A reductase inhibitors (statins) reportedly promote the neovascularization of ischemic tissue in normocholesterolemic animals. Methods and Results-Fifteen patients with angiographically documented stable coronary artery disease (CAD) were prospectively treated with 40 mg of atorvastatin per day for 4 weeks. Before and weekly after the initiation of statin therapy, EPCs were isolated from peripheral blood and counted. In addition, the number of hematopoietic precursor cells positive for CD34, CD133, and CD34/kinase insert domain receptor was analyzed. Statin treatment of patients with stable CAD was associated with an approximately 1.5-fold increase in the number of circulating EPCs by 1 week after initiation of treatment; this was followed by sustained increased levels to approximately 3-fold throughout the 4-week study period. Moreover, the number of CD34/kinase insert domain receptor-positive hematopoietic progenitor cells was significantly augmented after 4 weeks of therapy. Atorvastatin treatment increased the further functional activity of EPCs, as assessed by their migratory capacity. CONCLUSION The results of the present study define a novel mechanism of action of statin treatment in patients with stable CAD: the augmentation of circulating EPCs with enhanced functional activity. Given the well-established role of EPCs of participating in repair after ischemic injury, stimulation of EPCs by statins may contribute to the clinical benefit of statin therapy in patients with CAD.
Collapse
Affiliation(s)
- M Vasa
- Division of Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Theodor-Stern-Kai 7, Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
1088
|
Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A, Sasaki K, Shimada T, Oike Y, Imaizumi T. Mobilization of endothelial progenitor cells in patients with acute myocardial infarction. Circulation 2001; 103:2776-9. [PMID: 11401930 DOI: 10.1161/hc2301.092122] [Citation(s) in RCA: 763] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) circulate in adult peripheral blood (PB) and contribute to neovascularization. However, little is known regarding whether EPCs and their putative precursor, CD34-positive mononuclear cells (MNC(CD34+)), are mobilized into PB in acute ischemic events in humans. METHODS AND RESULTS Flow cytometry revealed that circulating MNC(CD34+) counts significantly increased in patients with acute myocardial infarction (n=16), peaking on day 7 after onset, whereas they were unchanged in control subjects (n=8) who had no evidence of cardiac ischemia. During culture, PB-MNCs formed multiple cell clusters, and EPC-like attaching cells with endothelial cell lineage markers (CD31, vascular endothelial cadherin, and kinase insert domain receptor) sprouted from clusters. In patients with acute myocardial infarction, more cell clusters and EPCs developed from cultured PB-MNCs obtained on day 7 than those on day 1. Plasma levels of vascular endothelial growth factor significantly increased, peaking on day 7, and they positively correlated with circulating MNC(CD34+) counts (r=0.35, P=0.01). CONCLUSIONS This is the first clinical demonstration showing that lineage-committed EPCs and MNC(CD34+), their putative precursors, are mobilized during an acute ischemic event in humans.
Collapse
Affiliation(s)
- S Shintani
- Cardiovascular Research Institute and Department of Internal Medicine III, Kurume University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1089
|
Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 2001; 7:430-6. [PMID: 11283669 DOI: 10.1038/86498] [Citation(s) in RCA: 1772] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Left ventricular remodeling is a major cause of progressive heart failure and death after myocardial infarction. Although neoangiogenesis within the infarcted tissue is an integral component of the remodeling process, the capillary network is unable to support the greater demands of the hypertrophied myocardium, resulting in progressive loss of viable tissue, infarct extension and fibrous replacement. Here we show that bone marrow from adult humans contains endothelial precursors with phenotypic and functional characteristics of embryonic hemangioblasts, and that these can be used to directly induce new blood vessel formation in the infarct-bed (vasculogenesis) and proliferation of preexisting vasculature (angiogenesis) after experimental myocardial infarction. The neoangiogenesis resulted in decreased apoptosis of hypertrophied myocytes in the peri-infarct region, long-term salvage and survival of viable myocardium, reduction in collagen deposition and sustained improvement in cardiac function. The use of cytokine-mobilized autologous human bone-marrow-derived angioblasts for revascularization of infarcted myocardium (alone or in conjunction with currently used therapies) has the potential to significantly reduce morbidity and mortality associated with left ventricular remodeling.
Collapse
Affiliation(s)
- A A Kocher
- Department of Surgery, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1090
|
Abstract
In animal models of ischemia, a large body of evidence indicates that administration of angiogenic growth factors, either as recombinant protein or by gene transfer, can augment nutrient perfusion through neovascularization. While many cytokines have angiogenic activity, the best studied both in animal models and clinical trials are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). Clinical trials of therapeutic angiogenesis in patients with end-stage coronary artery disease have shown large increases in exercise time and marked reductions in symptoms of angina, as well as objective evidence of improved perfusion and left ventricular function. Larger scale placebo-controlled trials have been limited to intracoronary and intravenous administration of recombinant protein, and have not yet shown significant improvement in either exercise time or angina when compared to placebo. Larger scale placebo-controlled studies of gene transfer are in progress. Future clinical studies will be required to determine the optimal dose, formulation, route of administration and combinations of growth factors, as well as the requirement for endothelial progenitor cell or stem cell supplementation, to provide effective and safe therapeutic myocardial angiogenesis.
Collapse
Affiliation(s)
- S B Freedman
- Divisions of Cardiology and Vascular Medicine, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
1091
|
Yang J, Nagavarapu U, Relloma K, Sjaastad MD, Moss WC, Passaniti A, Herron GS. Telomerized human microvasculature is functional in vivo. Nat Biotechnol 2001; 19:219-24. [PMID: 11231553 DOI: 10.1038/85655] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previously we showed the superior in vitro survival of human telomerase reverse transcriptase (hTERT)-transduced human endothelial cells (EC). Here we show that retroviral-mediated transduction of hTERT in human dermal microvascular EC (HDMEC) results in cell lines that form microvascular structures when subcutaneously implanted in severe combined immunodeficiency (SCID) mice. Anti-human type IV collagen basement membrane immunoreactivity and visualization of enhanced green fluorescent protein (eGFP)-labeled microvessels confirmed the human origin of these capillaries. No human vasculature was observed after implantation of HT1080 fibrosarcoma cells, 293 human embryonic kidney cells, or human skin fibroblasts. Intravascular red fluorescent microspheres injected into host circulation were found within green "telomerized" microvessels, indicating functional murine-human vessel anastamoses. Whereas primary HDMEC-derived vessel density decreased with time, telomerized HDMEC maintained durable vessels six weeks after xenografting. Modulation of implant vessel density by exposure to different angiogenic and angiostatic factors demonstrated the utility of this system for the study of human microvascular remodeling in vivo.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Basement Membrane/chemistry
- Capillaries/drug effects
- Capillaries/growth & development
- Cells, Cultured
- Chimera
- Collagen/analysis
- Collagen/pharmacology
- Collagen Type XVIII
- Dermis/blood supply
- Endostatins
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/transplantation
- Fibroblasts
- Humans
- Mice
- Mice, SCID
- Microcirculation/drug effects
- Microcirculation/growth & development
- Microspheres
- Models, Animal
- Neovascularization, Physiologic/drug effects
- Peptide Fragments/pharmacology
- Telomerase/genetics
- Telomerase/metabolism
- Telomere/genetics
- Transduction, Genetic
- Transplantation, Heterologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J Yang
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
1092
|
Shintani S, Murohara T, Ikeda H, Ueno T, Sasaki K, Duan J, Imaizumi T. Augmentation of postnatal neovascularization with autologous bone marrow transplantation. Circulation 2001; 103:897-903. [PMID: 11171801 DOI: 10.1161/01.cir.103.6.897] [Citation(s) in RCA: 286] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) have been identified in adult human peripheral blood. Because circulating EPCs should originate from bone marrow (BM), we examined whether BM mononuclear cells (BM-MNCs) can give rise to functional EPCs and whether transplantation of autologous BM-MNCs might augment angiogenesis and collateral vessel formation in a rabbit model of hindlimb ischemia. METHODS AND RESULTS Rabbit BM-MNCs were isolated by centrifugation through a Histopaque density gradient and cultured on fibronectin. EPCs developed from BM-MNCs in vitro, as assessed by acetylated LDL incorporation, nitric oxide (NO) release, and expression of von Willebrand factor and lectin binding. Unilateral hindlimb ischemia was surgically induced in rabbits (n=8), and fluorescence-labeled autologous BM-MNCs were transplanted into the ischemic tissues. Two weeks after transplantation, fluorescence microscopy revealed that transplanted cells were incorporated into the capillary network among preserved skeletal myocytes. In contrast, transplanted autologous BM-fibroblasts did not participate in EC capillary network formation (n=5). Then, in an additional 27 rabbits, saline (control; n=8), autologous BM-MNCs (n=13; 6.9+/-2.2x10(6) cells/animal), or BM-fibroblasts (n=6; 6.5+/-1.5x10(6) cells/animal) were injected into the ischemic tissues at postoperative day 7. Four weeks after transplantation, the BM-MNC-transplanted group had more angiographically detectable collateral vessels (angiographic score: 1.5+/-0.34 versus 0.94+/-0.26 and 1.1+/-0.14; P:<0.05), a higher capillary density (23+/-5.8 versus 10+/-1.9 and 11+/-0.8 per field; P:<0.001), and a greater laser Doppler blood perfusion index (505+/-155 versus 361+/-35 and 358+/-22 U; P:<0.05) than the control and BM-fibroblast-transplanted groups. CONCLUSIONS Direct local transplantation of autologous BM-MNCs seems to be a useful strategy for therapeutic neovascularization in ischemic tissues in adults, consistent with "therapeutic vasculogenesis."
Collapse
Affiliation(s)
- S Shintani
- Cardiovascular Research Institute and the Department of Internal Medicine III, Kurume University School of Medicine, Kurume, Japan
| | | | | | | | | | | | | |
Collapse
|
1093
|
Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI, Uchida S, Masuda H, Silver M, Ma H, Kearney M, Isner JM, Asahara T. Therapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemia. Circulation 2001; 103:634-7. [PMID: 11156872 DOI: 10.1161/01.cir.103.5.634] [Citation(s) in RCA: 750] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We investigated the therapeutic potential of ex vivo expanded endothelial progenitor cells (EPCs) for myocardial neovascularization. METHODS AND RESULTS Peripheral blood mononuclear cells obtained from healthy human adults were cultured in EPC medium and harvested 7 days later. Myocardial ischemia was induced by ligating the left anterior descending coronary artery in male Hsd:RH-rnu (athymic nude) rats. A total of 10(6) EPCs labeled with 1,1'-dioctadecyl-1 to 3,3,3',3'-tetramethylindocarbocyanine perchlorate were injected intravenously 3 hours after the induction of myocardial ischemia. Seven days later, fluorescence-conjugated Bandeiraea simplicifolia lectin I was administered intravenously, and the rats were immediately killed. Fluorescence microscopy revealed that transplanted EPCs accumulated in the ischemic area and incorporated into foci of myocardial neovascularization. To determine the impact on left ventricular function, 5 rats (EPC group) were injected intravenously with 10(6) EPCs 3 hours after ischemia; 5 other rats (control group) received culture media. Echocardiography, performed just before and 28 days after ischemia, disclosed ventricular dimensions that were significantly smaller and fractional shortening that was significantly greater in the EPC group than in the control group by day 28. Regional wall motion was better preserved in the EPC group. After euthanization on day 28, necropsy examination disclosed that capillary density was significantly greater in the EPC group than in the control group. Moreover, the extent of left ventricular scarring was significantly less in rats receiving EPCs than in controls. Immunohistochemistry revealed capillaries that were positive for human-specific endothelial cells. CONCLUSIONS Ex vivo expanded EPCs incorporate into foci of myocardial neovascularization and have a favorable impact on the preservation of left ventricular function.
Collapse
Affiliation(s)
- A Kawamoto
- Division of Cardiovascular Research, St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1094
|
Abstract
It is generally accepted that vasculogenesis is limited to early embryogenesis and is believed not to occur in adult, whereas angiogenesis occurs in both the developing embryo and postnatal life. However, the distinction between them is not absolute, because both require endothelial cell proliferation and migration and three-dimensional reorganization of newly formed blood vessels, nor are they mutually exclusive, inasmuch as angioblasts can be incorporated into expanding pre-existing blood vessels. Recent observations indicate that vasculogenesis may not be restricted to early embryogenesis, but may also have a physiological role or contribute to the pathology of vascular diseases in adults. The major evidence in favor of this new view comes from: (i) demonstration of the presence of circulating endothelial cells and endothelial precursor cells; (ii) newly described mechanisms of blood vessel formation in tumor growth. The potential biomedical applications of endothelial precursor cells and the new opportunities for the development of new forms of tumor-targeted treatments are discussed.
Collapse
Affiliation(s)
- D Ribatti
- Department of Human Anatomy and Histology, University of Bari Medical School, Piazza G. Cesare, 11, Policlinico, I-70124, Bari, Italy.
| | | | | | | | | |
Collapse
|
1095
|
Young HE, Steele TA, Bray RA, Hudson J, Floyd JA, Hawkins K, Thomas K, Austin T, Edwards C, Cuzzourt J, Duenzl M, Lucas PA, Black AC. Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. ACTA ACUST UNITED AC 2001. [DOI: 10.1002/ar.1128 [pii]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1096
|
Embryonic central nervous system angiogenesis does not involve blood-borne endothelial progenitors. J Comp Neurol 2001. [DOI: 10.1002/cne.1066] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
1097
|
Isner JM. Tissue responses to ischemia: local and remote responses for preserving perfusion of ischemic muscle. J Clin Invest 2000; 106:615-9. [PMID: 10974011 PMCID: PMC381295 DOI: 10.1172/jci10961] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- J M Isner
- Tufts University School of Medicine and the Department of Medicine (Vascular Medicine) and Biomedical Research, St. Elizabeth's Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
1098
|
Bertolini F, Mancuso P, Gobbi A, Pruneri G. The thin red line: angiogenesis in normal and malignant hematopoiesis. Exp Hematol 2000; 28:993-1000. [PMID: 11008011 DOI: 10.1016/s0301-472x(00)00508-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This review describes the current knowledge about cell subsets involved in vasculogenesis (i.e., differentiation of endothelial cells from mesodermal precursors) and angiogenesis (i.e., blood vessel generation from pre-existing vessels), together with recent findings about angiogenesis and antiangiogenic therapies in hematopoietic malignancies such as leukemia, lymphoma, myeloma, and myelodysplastic syndromes.
Collapse
Affiliation(s)
- F Bertolini
- Division of Hematology-Oncology, IRCCS European Institute of Oncology, Milan, Italy.
| | | | | | | |
Collapse
|
1099
|
Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. Blood-derived angioblasts accelerate blood-flow restoration in diabetic mice. J Clin Invest 2000; 106:571-8. [PMID: 10953032 PMCID: PMC380249 DOI: 10.1172/jci9087] [Citation(s) in RCA: 359] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/1999] [Accepted: 07/05/2000] [Indexed: 11/17/2022] Open
Abstract
Endothelial cell progenitors, angioblasts, have been detected in the peripheral blood of adult humans, mice, and rabbits. These cells have been shown to incorporate into the endothelium of newly forming blood vessels in pathological and nonpathological conditions. Here we investigated the possibility that the CD34-expressing leukocytes (CD34(+) cells) that appear to be enriched for angioblasts could be used to accelerate the rate of blood-flow restoration in nondiabetic and diabetic mice undergoing neovascularization due to hindlimb ischemia. CD34(+) cells did not accelerate the restoration of flow in nondiabetic mice, but dramatically increased it in diabetic mice. Furthermore, CD34(+) cells derived from type 1 diabetics produced fewer differentiated endothelial cells in culture than did their type 2 diabetic- or nondiabetic-derived counterparts. In vitro experiments suggest that hyperglycemia per se does not alter the ability of angioblasts to differentiate or of angioblast-derived endothelial cells to proliferate. In contrast, hyperinsulinemia may enhance angioblast differentiation but impair angioblast-derived endothelial cell survival or proliferation. Our findings suggest that CD34(+) cells may be a useful tool for therapeutic angiogenesis in diabetics.
Collapse
Affiliation(s)
- G C Schatteman
- Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52245, USA.
| | | | | | | | | |
Collapse
|
1100
|
Murohara T, Ikeda H, Duan J, Shintani S, Sasaki KI, Eguchi H, Onitsuka I, Matsui K, Imaizumi T. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J Clin Invest 2000; 105:1527-36. [PMID: 10841511 PMCID: PMC300847 DOI: 10.1172/jci8296] [Citation(s) in RCA: 639] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/1999] [Accepted: 04/10/2000] [Indexed: 12/15/2022] Open
Abstract
Endothelial precursor cells (EPCs) have been identified in adult peripheral blood. We examined whether EPCs could be isolated from umbilical cord blood, a rich source for hematopoietic progenitors, and whether in vivo transplantation of EPCs could modulate postnatal neovascularization. Numerous cell clusters, spindle-shaped and attaching (AT) cells, and cord-like structures developed from culture of cord blood mononuclear cells (MNCs). Fluorescence-trace experiments revealed that cell clusters, AT cells, and cord-like structures predominantly were derived from CD34-positive MNCs (MNC(CD34+)). AT cells and cell clusters could be generated more efficiently from cord blood MNCs than from adult peripheral blood MNCs. AT cells incorporated acetylated-LDL, released nitric oxide, and expressed KDR, VE-cadherin, CD31, and von Willebrand factor but not CD45. Locally transplanted AT cells survived and participated in capillary networks in the ischemic tissues of immunodeficient nude rats in vivo. AT cells thus had multiple endothelial phenotypes and were defined as a major population of EPCs. Furthermore, laser Doppler and immunohistochemical analyses revealed that EPC transplantation quantitatively augmented neovascularization and blood flow in the ischemic hindlimb. In conclusion, umbilical cord blood is a valuable source of EPCs, and transplantation of cord blood-derived EPCs represents a promising strategy for modulating postnatal neovascularization.
Collapse
Affiliation(s)
- T Murohara
- The Cardiovascular Research Institute, Department of Internal Medicine III, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|