201
|
Park KW, Hwang KK, Cho HJ, Hur J, Yang HM, Yoon CH, Kang HJ, Oh BH, Park YB, Kim HS. Simvastatin enhances endothelial differentiation of peripheral blood mononuclear cells in hypercholesterolemic patients and induces pro-angiogenic cytokine IL-8 secretion from monocytes. Clin Chim Acta 2007; 388:156-66. [PMID: 18037374 DOI: 10.1016/j.cca.2007.10.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2007] [Revised: 10/19/2007] [Accepted: 10/23/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Statins are known to have pleiotropic effects. We examined the effect and mechanism of simvastatin therapy on EPC differentiation and pro-angiogenic cytokines in patients with hypercholesterolemia. METHODS Twenty-two hypercholesterolemia patients without any other modifiable cardiovascular risk factors or history of previous lipid-lowering therapy were given simvastatin 20 mg/day for 4 weeks. Blood were drawn pre- and post-therapy. The in vitro effects of simvastatin were studied in a separate set of experiments. RESULTS Simvastatin treatment significantly increased the number of DiI-acLDL, UEA-1 lectin double-positive EPCs and facilitated its appearance. By FACS analysis of freshly isolated PBMNCs, KDR (+) cells increased after simvastatin treatment while there were no differences in CD34, AC133, and VE-cadherin. Also, serum concentration of IL-8 was markedly increased, while VEGF was only slightly increased. In vitro, PBMNCs co-cultured with simvastatin showed increased cluster formation at day 7, and simvastatin facilitated the appearance and networking of EPCs compared with vehicle. Simvastatin-co-cultured PBMNCs showed significantly increased KDR (+) cells, in contrast to CD34, CD31, and VE-Cadherin (+) cells. In response to simvastatin, IL-8 was mainly increased in monocyte culture supernatants while VEGF increased in smooth muscle cell culture supernatants. These cytokines were associated with increased EPC migratory function. The increase in IL-8 secretion from monocytes by statin treatment was associated with phosphorylation and inactivation of GSK3beta, which was reversed by constitutive activation of GSK-3beta. CONCLUSION Simvastatin enhances endothelial differentiation of peripheral blood mononuclear cells in patients with hypercholesterolemia and increases pro-angiogenic cytokine IL-8 secretion from monocytes. Our results may explain the pro-angiogenic effects associated with statin therapy and offer further evidence of statin pleiotropism.
Collapse
Affiliation(s)
- Kyung Woo Park
- National Research Laboratory for Cardiovascular Stem Cell, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Ballard VLT, Edelberg JM. Targets for regulating angiogenesis in the ageing endothelium. Expert Opin Ther Targets 2007; 11:1385-99. [DOI: 10.1517/14728222.11.11.1385] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
203
|
Tongers J, Losordo DW. Frontiers in Nephrology: The Evolving Therapeutic Applications of Endothelial Progenitor Cells. J Am Soc Nephrol 2007; 18:2843-52. [DOI: 10.1681/asn.2007050597] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
204
|
de la Peña M, Barceló A, Barbe F, Piérola J, Pons J, Rimbau E, Ayllón O, Agustí AGN. Endothelial function and circulating endothelial progenitor cells in patients with sleep apnea syndrome. ACTA ACUST UNITED AC 2007; 76:28-32. [PMID: 17921670 DOI: 10.1159/000109643] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 06/16/2007] [Indexed: 12/22/2022]
Abstract
BACKGROUND Endothelial dysfunction and cardiovascular diseases are frequent in patients with obstructive sleep apnea (OSA). Circulating endothelial progenitor cells (EPCs) contribute to repair dysfunctional endothelium and have been related to increased cardiovascular risk. OBJECTIVES We tested the hypothesis that the number of circulating EPCs may be altered in OSA patients. METHODS EPCs (CD34+ VEGF-R2+) were isolated and quantified from peripheral blood samples of OSA patients (n = 13) and healthy controls (n = 13) matched for age and sex. All subjects were free of any other known cardiovascular risk factors. The plasma levels of vascular endothelial growth factor (VEGF) were also determined, and the endothelium-dependent and endothelium-independent vascular function was assessed in all subjects. RESULTS Patients with OSA had lower levels of EPCs (p < 0.05) and higher plasma levels of VEGF (p < 0.05) than controls. Endothelial function was not different between OSA and controls. CONCLUSIONS Patients with OSA free of any other known cardiovascular risk factor show a reduced number of circulating EPCs and an increase in plasma VEGF levels. These alterations may contribute to future endothelial dysfunction in these patients.
Collapse
Affiliation(s)
- Mónica de la Peña
- Servei de Pneumología, Hospital Universitari Son Dureta, Palma de Mallorca, España.
| | | | | | | | | | | | | | | |
Collapse
|
205
|
Rossi A, Maione P, Ferrara C, Del Gaizo F, Guerriero C, Nicolella D, Palazzolo G, Falanga M, Colantuoni G, Gridelli C. New angiogenic agents and non-small cell lung cancer: current results and future development. Target Oncol 2007. [DOI: 10.1007/s11523-007-0060-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
206
|
Renault MA, Losordo DW. Therapeutic myocardial angiogenesis. Microvasc Res 2007; 74:159-71. [PMID: 17950369 PMCID: PMC2172411 DOI: 10.1016/j.mvr.2007.08.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2007] [Revised: 08/15/2007] [Accepted: 08/17/2007] [Indexed: 01/23/2023]
Abstract
Armed with an improved understanding of the mediators of angiogenesis, physicians and scientists have made significant efforts at harnessing this naturally occurring process in order to treat patients with a variety of peripheral vascular and coronary ischemic syndromes. There is a growing population of patients with end-stage coronary artery disease (CAD) who are no longer candidates for mechanical revascularization, yet suffer from chronic myocardial ischemia who may benefit from regeneration of the depleted microvasculature.
Collapse
Affiliation(s)
- Marie-Ange Renault
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 12-703, Chicago, IL 60611, USA
| | | |
Collapse
|
207
|
Abstract
PURPOSE OF REVIEW Peripheral arterial disease is a common disease that has few treatment options. Angiogenesis is defined as the growth of new blood vessels from preexisting vasculature. Therapeutic angiogenesis is an investigational method that uses vascular growth to alleviate disorders of tissue ischemia, such as coronary artery disease and peripheral arterial disease. There have been tremendous changes in the field of therapeutic angiogenesis over the past decade, and there is much promise for the future. RECENT FINDINGS Initial preclinical work with cytokine growth factor delivery resulted in a great deal of enthusiasm, but larger clinical studies have failed to achieve similar success. With an increased understanding of the complex mechanisms involved in angiogenesis, gene therapy and cell therapy have moved to the forefront of therapeutic angiogenesis. Novel therapies which target multiple different angiogenic pathways are also being developed and tested. SUMMARY Therapeutic angiogenesis is an exciting field that continues to evolve. This review will focus on the different growth factors being used, their routes of delivery, the results of clinical trials, and some of the novel therapies being developed.
Collapse
Affiliation(s)
- William S Jones
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
208
|
Okazaki T, Ebihara S, Asada M, Yamanda S, Saijo Y, Shiraishi Y, Ebihara T, Niu K, Mei H, Arai H, Yambe T. Macrophage colony-stimulating factor improves cardiac function after ischemic injury by inducing vascular endothelial growth factor production and survival of cardiomyocytes. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1093-103. [PMID: 17717142 PMCID: PMC1988861 DOI: 10.2353/ajpath.2007.061191] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Macrophage colony-stimulating factor (M-CSF), known as a hematopoietic growth factor, induces vascular endothelial growth factor (VEGF) production from skeletal muscles. However, the effects of M-CSF on cardiomyocytes have not been reported. Here, we show M-CSF increases VEGF production from cardiomyocytes, protects cardiomyocytes and myotubes from cell death, and improves cardiac function after ischemic injury. In mice, M-CSF increased VEGF production in hearts and in freshly isolated cardiomyocytes, which showed M-CSF receptor expression. In rat cell line H9c2 cardiomyocytes and myotubes, M-CSF induced VEGF production via the Akt signaling pathway, and M-CSF pretreatment protected these cells from H(2)O(2)-induced cell death. M-CSF activated Akt and extracellular signal-regulated kinase signaling pathways and up-regulated downstream anti-apoptotic Bcl-xL expression in these cells. Using goats as a large animal model of myocardial infarction, we found that M-CSF treatment after the onset of myocardial infarction by permanent coronary artery ligation promoted angiogenesis in ischemic hearts but did not reduce the infarct area. M-CSF pretreatment of the goat myocardial infarction model by coronary artery occlusion-reperfusion improved cardiac function, as assessed by hemodynamic parameters and echocardiography. These results suggest M-CSF might be a novel therapeutic agent for ischemic heart disease.
Collapse
Affiliation(s)
- Tatsuma Okazaki
- Department of Geriatrics and Gerontology, Tohoku University School of Medicine, Seiryo-machi 1-1, Aoba-ku, Sendai 980-8574, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Bartsch T, Brehm M, Zeus T, Kögler G, Wernet P, Strauer BE. Transplantation of autologous mononuclear bone marrow stem cells in patients with peripheral arterial disease (the TAM-PAD study). Clin Res Cardiol 2007; 96:891-9. [PMID: 17694378 DOI: 10.1007/s00392-007-0569-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Accepted: 07/03/2007] [Indexed: 01/14/2023]
Abstract
OBJECTIVES For patients with severe, chronic limb ischemia in many cases interventional or surgical treatment is not possible anymore. In the past, both intramuscular and intraarterial transplantation of autologous BMCs had been proved therapeutically beneficial. The TAM-PAD study is the first one to analyze combined intraarterial and intramuscular BMC transplantation in its acute and long-term effects. METHODS 13 patients with chronically ischemic limbs due to peripheral arterial disease (Fontaine stage IIb) were recruited and underwent follow-up examinations after 2 and 13 months. Mononuclear cells from bone marrow were injected intramuscular and intraarterial into the ischemic limb. RESULTS In contrast to the control group, after 2 months the pain-free walking distance of the transplanted patients significantly increased (from 147 +/- 90 to 500 +/- 614 m, p = 0.001). Furthermore the ankle-brachial index was significantly improved (at rest from 0.66+/-0.18 to 0.80+/-0.15, p = 0.003, after stress from 0.64 +/- 0.19 to 0.76 +/- 0.16, p = 0.006). Similar improvement was documented in capillary-venous oxygen-saturation (thigh from 59 +/- 9 to 66 +/- 5, p = 0.005, lower leg from 56 +/- 14 to 63 +/- 5, p = 0.021) and venous occlusion plethysmography (rest from 2.1 +/- 0.7 to 2.5 +/- 0.7, p = 0.009, mean reactive hyperemia from 5.3 +/- 1.8 to 7.2 +/- 1.8, p = 0.003, and peak flow from 7.2 +/- 3.2 to 10.8 +/- 2.8, p = 0.002). After 13 months these positive effects persisted at their improved level. No side effects or complications were monitored. CONCLUSIONS Combined intraarterial and intramuscular transplantation of autologous mononuclear bone marrow stem cells is a clinically feasible and minimally invasive therapeutic option for patients with severe chronic peripheral occlusive arterial disease.
Collapse
Affiliation(s)
- T Bartsch
- Department of Medicine, Division of Cardiology Pneumology and Angiology, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
210
|
Zhao Q, Liu Z, Wang Z, Yang C, Liu J, Lu J. Effect of prepro-calcitonin gene-related peptide-expressing endothelial progenitor cells on pulmonary hypertension. Ann Thorac Surg 2007; 84:544-52. [PMID: 17643632 DOI: 10.1016/j.athoracsur.2007.03.067] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 03/18/2007] [Accepted: 03/20/2007] [Indexed: 11/26/2022]
Abstract
BACKGROUND Calcitonin gene-related peptide (CGRP) is a potent smooth muscle cell proliferation inhibitor and vasodilator. It is now believed that CGRP plays an important role in maintaining a low pulmonary vascular resistance. We evaluated the therapeutic effect of intravenously administered CGRP-expressing endothelial progenitor cells (EPCs) on left-to-right shunt-induced pulmonary hypertension in rats. METHODS Endothelial progenitor cells were obtained from cultured human peripheral blood mononuclear cells. The genetic sequence for CGRP was subcloned into cultured EPCs by human expression plasmid. Pulmonary hypertension was established in immunodeficient rats with an abdominal aorta to inferior vena cava shunt operation. The transfected EPCs were injected through the left jugular vein at 10 weeks after the shunt operation. Mean pulmonary artery pressure and total pulmonary vascular resistance were detected with right cardiac catheterization at 4 weeks. The distribution of EPCs in the lung tissue was examined with immunofluorescence technique. Histopathologic changes in the structure of the pulmonary arteries was observed with electron microscopy and subjected to computerized image analysis. RESULTS The lungs of rats transplanted with CGRP-expressing EPCs demonstrated a decrease in both mean pulmonary artery pressure (17.64 +/- 0.79 versus 22.08 +/- 0.95 mm Hg; p = 0.018) and total pulmonary vascular resistance (1.26 +/- 0.07 versus 2.45 +/- 0.18 mm Hg x min/mL; p = 0.037) at 4 weeks. Immunofluorescence revealed that intravenously administered cells were incorporated into the pulmonary vasculature. Pulmonary vascular remodeling was remarkably attenuated with the administration of CGRP-expressing EPCs. CONCLUSIONS The transplantation of CGRP-expressing EPCs may effectively attenuate established pulmonary hypertension and exert reversal effects on pulmonary vascular remodeling. Our findings suggest that the therapy based on the combination of both CGRP transfection and EPCs may be a potentially useful strategy for the treatment of pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Qiang Zhao
- Division of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
211
|
Abstract
The discovery of endothelial progenitor cells has generated considerable interest in the field of vascular biology. These cells arise from a population of circulating mononuclear cells and have the capacity to form new blood vessels and contribute to vascular repair. Circulating endothelial progenitor cell numbers are reduced in patients with cardiovascular risk factors and in the presence of endothelial dysfunction, but are increased in response to ischaemia, oestrogens and drug therapy. They have been studied in pathologies from cardiovascular and renal disease to rheumatoid arthritis and pre-eclampsia. Pregnancy is a challenge to the maternal vascular system, requiring systemic adaptation and pronounced local changes in the uterus. Diseases of pregnancy such as pre-eclampsia and gestational diabetes increase the risk of pregnancy complications and are associated with endothelial dysfunction. We propose that endothelial progenitor cells have an important role in the regulation and maintenance of the vasculature during pregnancy. This review summarises our current understanding of endothelial progenitor cells, with specific reference to their role in angiogenesis and human pregnancy.
Collapse
Affiliation(s)
- Amy O Robb
- Centre for Reproductive Biology, Reproductive and Developmental Sciences, The University of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SU United Kingdom
| | | | | | | |
Collapse
|
212
|
Lapergue B, Mohammad A, Shuaib A. Endothelial progenitor cells and cerebrovascular diseases. Prog Neurobiol 2007; 83:349-62. [PMID: 17884277 DOI: 10.1016/j.pneurobio.2007.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Revised: 06/26/2007] [Accepted: 08/03/2007] [Indexed: 01/09/2023]
Abstract
Identifying factors that may increase the risk of stroke and assessing if treatment of such conditions may lower that risk are important in the management of cerebrovascular disease. Tobacco smoking, poor diet, hypertension and hyperlipidemia remain the major risk factors, and treatment of these conditions has been shown to significantly reduce stroke. In recent years, research has shown that stem cells from a variety of sources can be used as a tool to study and prevent the events that lead to stroke. In this regard, a population of adult stem cells, called endothelial progenitor cells (EPCs), have been identified in peripheral blood and may play an important role in tissue vascularization and endothelium homeostasis in the adult. Most of the studies on EPCs have been carried out on patients with cardiovascular diseases; however, there is emerging evidence which suggests that the introduction or mobilization of EPCs can restore tissue vascularization even after cerebrovascular diseases (CVD), such as ischemic stroke or intracerebral haemorrhage. In this review, we discuss the present level of knowledge about the characteristics of EPCs, their possible therapeutic role in CVD and how they could alter clinical practice in the future.
Collapse
Affiliation(s)
- Bertrand Lapergue
- Stroke Research Unit, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Alberta, Canada
| | | | | |
Collapse
|
213
|
Vertesaljai M, Piroth Z, Fontos G, Andreka G, Font G, Szantho G, Lueff S, Reti M, Masszi T, Ablonczy L, Juhasz ED, Simor T, Turner MS, Andreka P. Drugs, gene transfer, signaling factors: a bench to bedside approach to myocardial stem cell therapy. Heart Fail Rev 2007; 13:227-44. [PMID: 17668319 DOI: 10.1007/s10741-007-9047-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 07/10/2007] [Indexed: 12/13/2022]
Abstract
In the past few years, the dogma that the heart is a terminally differentiated organ has been challenged. Evidence from preclinical investigations emerged that there are cells, even in the heart itself, that may be able to restore impaired cardiac function after myocardial infarction. Although the exact mechanisms by which the infarcted heart can be repaired by stem cells are not yet fully defined, there is a new optimism among cardiologists that this treatment will prove successful in addressing the cause of heart failure after myocardial infarction-myocyte loss. Despite the promising preliminary data of human myocardial stem cell trials, scientists have also focused on the possibility of enhancing the underlying mechanisms of stem cell repair to gain healthier myocardial tissue. Attempts to induce neo-angiogenesis by transfecting stem cells with signaling factors (such as VEGF), to raise the number of endothelial progenitor cells with medical treatments (such as statins), to transfect stem cells with heat shock protein 70 (as a cardioprotective agent against ischemia) and to enhance the healing process after myocardial infarction with the use of various forms of stimulating factors (G-CSF, SCF, GM-CSF) have been made with notable results. In this article, we summarize the evidence from preclinical and clinical myocardial stem cell studies that have addressed the possibility of enhancing the regenerative capacity of cells used after myocardial infarction.
Collapse
Affiliation(s)
- Marton Vertesaljai
- Department of Adult Cardiology, Gottsegen Hungarian Institute of Cardiology, Haller u. 29, Budapest 1096, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Benndorf RA, Gehling UM, Appel D, Maas R, Schwedhelm E, Schlagner K, Silberhorn E, Hossfeld DK, Rogiers X, Böger R. Mobilization of putative high-proliferative-potential endothelial colony-forming cells during antihypertensive treatment in patients with essential hypertension. Stem Cells Dev 2007; 16:329-38. [PMID: 17521243 DOI: 10.1089/scd.2006.0074] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Recent studies have shown that in response to vascular damage or ischemia, bone marrow-derived endothelial progenitor cells (EPCs) are recruited into the circulation. To investigate whether antihypertensive treatment has an influence on the number of circulating EPCs, patients with essential hypertension were treated either with the angiotensin receptor antagonist telmisartan, the calcium channel blocker nisoldipine, or their combination for 6 weeks. At baseline and after 3 and 6 weeks of treatment, EPCs were identified and quantified by fluorescence-activated cell sorting (FACS) analysis and by their capacity to generate colony-forming units of the endothelial lineage (CFU-EC) in a methylcellulose-based assay. During treatment, patients in the nisoldipine groups, but not in the telmisartan group, showed a significant mobilization of EPCs, which in part had the capacity to generate large-sized colonies comprising more than 1,000 cells. Moreover, a remarkable correlation between the number of CFU-EC and the number of circulating CD133(+)/CD34(+)/CD146(+) cells was observed, thereby providing strong evidence that cells with this phenotype represent functional EPCs. No correlation was found between the numbers of CFU-EC and the blood pressure levels at any time point during the treatment. Hence, nisoldipine-induced mobilization of EPCs might represent a novel mechanism by which this antihypertensive compound independently of its blood pressure-lowering effect contributes to vasoprotection in patients with essential hypertension.
Collapse
Affiliation(s)
- Ralf A Benndorf
- Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Clinical Pharmacology, University Hospital Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Sugihara S, Yamamoto Y, Matsuura T, Narazaki G, Yamasaki A, Igawa G, Matsubara K, Miake J, Igawa O, Shigemasa C, Hisatome I. Age-related BM-MNC dysfunction hampers neovascularization. Mech Ageing Dev 2007; 128:511-6. [PMID: 17688912 DOI: 10.1016/j.mad.2007.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 06/22/2007] [Accepted: 06/23/2007] [Indexed: 01/11/2023]
Abstract
Although ischemia-induced neovascularization is reportedly impaired with aging, the effect of aged-bone marrow mononuclear cells (BM-MNCs) on neovascularization has not been investigated. The neovascularization capacity of BM-MNCs obtained from 8-week-old mice (young) was compared to those obtained from 18-month-old mice (old), both in vivo and in vitro. Neovascularization in ischemic limbs was significantly impaired in old mice. Whereas transplantation of young BM-MNCs significantly improved blood perfusion, tissue capillary density, and vascular endothelial growth factor (VEGF) production in transplanted ischemic limbs, no such effects were observed with old BM-MNCs. Old BM-MNCs also showed a significant impairment of in vitro VEGF production and migratory capacity in response to VEGF. The number of Dil/lectin-positive cells was significantly lower in old mice, but there was no difference in the number of AC133(+)/CD34(+) and CD34(+)/VEGF-R2(+) positive cells between young and old BM-MNCs. Transplantation of young BM-MNCs improved neovascularization and VEGF production in the ischemic limbs of old recipients, with results that were similar to those obtained in young recipients. These results indicate that the neovascularization capacity of transplanted BM-MNCs is impaired with aging. However, aging does not hamper the revitalization of neovascularization in the murine host in response to transplantation of young BM-MNCs.
Collapse
Affiliation(s)
- Shinobu Sugihara
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Tottori University Graduate School of Medical Science, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Abstract
Congestive heart failure remains the leading cause of morbidity and mortality in the developed world. Current therapies do not address the underlying pathophysiology of this disease, namely, the progressive loss of functional cardiomyocytes. The notion of repairing or regenerating lost myocardium via cell-based therapies remains highly appealing. The recent identification of adult stem cells, including both cardiac stem/progenitor cells and bone marrow stem cells, has triggered an explosive interest in using these cells for physiologically relevant cardiomyogenesis. Enthusiasm for cardiac regeneration via cell therapy has further been fueled by the many encouraging reports in both animals and human studies. Further intensive research in basic science and clinical arenas are needed to make this next great frontier in cardiovascular regenerative medicine a reality. In this review, we focus on the role of bone marrow-derived stem cells and cardiac stem/progenitor cells in cardiomyocyte homeostasis and myocardial repair and regeneration, as well as provide a brief overview of current clinical trials using cell-based therapeutic approaches in patients with heart disease.
Collapse
Affiliation(s)
- Ronglih Liao
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
217
|
Asosingh K, Swaidani S, Aronica M, Erzurum SC. Th1- and Th2-dependent endothelial progenitor cell recruitment and angiogenic switch in asthma. THE JOURNAL OF IMMUNOLOGY 2007; 178:6482-94. [PMID: 17475878 DOI: 10.4049/jimmunol.178.10.6482] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Increased numbers of submucosal vessels are a consistent pathologic component of asthmatic airway remodeling. However, the relationship between new vessel formation and asthmatic inflammatory response is unknown. We hypothesized that angiogenesis is a primary event during the initiation of airway inflammation and is linked to the recruitment of bone marrow-derived endothelial progenitor cells (EPC). To test this hypothesis, circulating EPC and EPC-derived endothelial cell colony formation of individuals with asthma or allergic rhinitis and health controls was evaluated. Circulating EPC were increased in asthma, highly proliferative, and exhibited enhanced incorporation into endothelial cell tubes as compared with controls. In an acute allergen challenge murine asthma model, EPC mobilization occurred within hours of challenge and mobilized EPC were selectively recruited into the challenged lungs of sensitized animals, but not into other organs. EPC recruitment was Th1 and Th2 dependent and was temporally associated with an increased microvessel density that was noted within 48 h of allergen challenge, indicating an early switch to an angiogenic lung environment. A chronic allergen challenge model provided evidence that EPC recruitment to the lung persisted and was associated with increasing microvessel density over time. Thus, a Th1- and Th2-dependent angiogenic switch with EPC mobilization, recruitment, and increased lung vessel formation occurs early but becomes a sustained and cumulative component of the allergen-induced asthmatic response.
Collapse
Affiliation(s)
- Kewal Asosingh
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
218
|
Rullman E, Rundqvist H, Wågsäter D, Fischer H, Eriksson P, Sundberg CJ, Jansson E, Gustafsson T. A single bout of exercise activates matrix metalloproteinase in human skeletal muscle. J Appl Physiol (1985) 2007; 102:2346-51. [PMID: 17255365 DOI: 10.1152/japplphysiol.00822.2006] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The aims of this study were 1) to characterize changes in matrix metalloproteinase (MMP), endostatin, and vascular endothelial growth factor (VEGF)-A expression in skeletal muscle in response to a single bout of exercise in humans; and 2) to determine if any exchange of endostatin and VEGF-A between circulation and the exercising leg is associated with a change in the tissue expression or plasma concentration of these factors. Ten healthy males performed 65 min of cycle exercise, and muscle biopsies were obtained from the vastus lateralis muscle at rest and immediately and 120 min after exercise. In the muscle biopsies, measurements of mRNA expression levels of MMP-2, MMP-9, MMP-14, and tissue inhibitor of metalloproteinase; VEGF and endostatin protein levels; and MMP activities were performed. Femoral arterial and venous concentrations of VEGF-A and endostatin were determined before, during, and 120 min after exercise. A single bout of exercise increased MMP-9 mRNA and activated MMP-9 protein in skeletal muscle. No measurable increase of endostatin was observed in the skeletal muscle or in plasma following exercise. A concurrent increase in skeletal muscle VEGF-A mRNA and protein levels was induced by exercise, with no signs of peripheral uptake from the circulation. However, a decrease in plasma VEGF-A concentration occurred following exercise. Thus 1) a single bout of exercise activated the MMP system without any resulting change in tissue endostatin protein levels, and 2) the increased VEGF-A protein levels are due to changes in the skeletal muscle tissue itself. Other mechanisms are responsible for the observed exercise-induced decrease in VEGF-A in plasma.
Collapse
Affiliation(s)
- E Rullman
- Department of Laboratory Medicine, Division of Clinical Physiology, C1-88, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
219
|
Bougioukas I, Didilis V, Ypsilantis P, Giatromanolaki A, Sivridis E, Lialiaris T, Mikroulis D, Simopoulos C, Bougioukas G. Intramyocardial injection of low-dose basic fibroblast growth factor or vascular endothelial growth factor induces angiogenesis in the infarcted rabbit myocardium. Cardiovasc Pathol 2007; 16:63-8. [PMID: 17317537 DOI: 10.1016/j.carpath.2006.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2005] [Revised: 07/24/2006] [Accepted: 08/30/2006] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Myocardial angiogenesis after the systemic administration of basic fibroblast growth factor or vascular endothelial growth factor at high therapeutic doses has been implicated in the occurrence of side effects that may undermine their safety. The aim of this study was to investigate the angiogenic effects of the intramyocardial administration of recombinant human basic fibroblast growth factor or vascular endothelial growth factor protein, at low doses, in the infarcted rabbit myocardium. METHODS AND RESULTS Twenty-five New Zealand White rabbits were divided into five groups (n=5) and subjected to coronary artery ligation after lateral thoracotomy, inducing acute myocardial infarction. Five minutes later, the following substances were injected intramyocardially into the infarcted area: (a) normal saline (controls); (b) 6.25 or 12.5 mug of recombinant human basic fibroblast growth factor protein (basic fibroblast growth factor-1 group or basic fibroblast growth factor-2 group); or (c) 5 or 10 microg of recombinant human vascular endothelial growth factor 165 protein (vascular endothelial growth factor-1 group or vascular endothelial growth factor-2 group). On the 21st postoperative day, the animals were euthanized, and their hearts were subjected to histopathological examination and immunohistochemical assessment of vascular density in the infarcted area. The alkaline phosphatase anti-alkaline phosphatase procedure and the primary monoclonal antibody JC70 were used. Histopathological examination confirmed the induction of myocardial infarction. Vascular density was significantly increased (P<.004) in all treatment groups (in mean+/-S.E. vessels/x 200 optical field: basic fibroblast growth factor-1: 85.8+/-10.9; basic fibroblast growth factor-2: 76.6+/-3.7; vascular endothelial growth factor-1: 73.4+/-3.2; vascular endothelial growth factor-2: 89.5+/-5.2) compared to that in controls (58.9+/-4.9 vessels/x 200 optical field). Vascular density in the vascular endothelial growth factor-2 group was significantly higher than that in the vascular endothelial growth factor-1 group (P<.001). CONCLUSIONS Low doses of recombinant human basic fibroblast growth factor or vascular endothelial growth factor protein, when administered intramyocardially, stimulate angiogenesis in the infarcted myocardium.
Collapse
Affiliation(s)
- Ioannis Bougioukas
- Department of Cardiothoracic Surgery, School of Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Dragana, 68 100 Alexandroupolis, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Abstract
Recent studies indicate that portions of ischemic and tumor neovasculature are derived by neovasculogenesis, whereby bone marrow (BM)-derived circulating endothelial progenitor cells (EPCs) home to sites of regenerative or malignant growth and contribute to blood vessel formation. Recent data from animal models suggest that a variety of cell types, including unfractionated BM mononuclear cells and those obtained by ex vivo expansion of human peripheral blood or enriched progenitors, can function as EPCs to promote tissue vasculogenesis, regeneration, and repair when introduced in vivo. The promising preclinical results have led to several human clinical trials using BM as a potential source of EPCs in cardiac repair as well as ongoing basic research on using EPCs in tissue engineering or as cell therapy to target tumor growth.
Collapse
Affiliation(s)
- Pampee P Young
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | |
Collapse
|
221
|
Affiliation(s)
- Otmar Pfister
- Whitaker Cardiovascular Institute, Boston, MA 02118, USA
| | | | | |
Collapse
|
222
|
Bartsch T, Brehm M, Zeus T, Strauer BE. Autologous mononuclear stem cell transplantation in patients with peripheral occlusive arterial disease. J Cardiovasc Nurs 2007; 21:430-2. [PMID: 17293730 DOI: 10.1097/00005082-200611000-00003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
UNLABELLED Patients with chronic peripheral occlusive arterial disease often are not candidates for conventional revascularization procedures. Preclinical trials have shown that the transplantation of autologous bone marrow cells induces and increases the collateral vessel formation. We analyzed the clinical benefit of combined intraarterial and intramuscular transplantation of adult autologous mononuclear bone marrow stem cells in patients with lower-limb peripheral occlusive arterial disease. METHODS Patients with severe peripheral occlusive arterial disease and a reduced walking distance (Fontaine stage II or III) were included. Bone marrow was harvested from the hip under local anesthesia and mononuclear cells were transplanted intramuscularly and intraarterially into the ischemic limb after isolation under good manufacturing practice conditions. RESULTS After 2 months, pain-free walking distance increased 3.7-fold. Furthermore, the ankle-brachial index was significantly improved at rest and after exercise. Similar improvements could be documented by capillary-venous oxygen saturation and venous occlusion plethysmography. No side effects or complications were detected during transplantation and during time of follow-up. CONCLUSIONS Combined intraarterial and intramuscular transplantation of autologous mononuclear bone marrow stem cells is a clinically feasible and minimally invasive therapeutic option for patients with severe, chronic peripheral occlusive arterial disease.
Collapse
Affiliation(s)
- Thomas Bartsch
- Division of Cardiology, Pneumology, and Angiology, Department of Internal Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | | | | | | |
Collapse
|
223
|
Eguchi M, Masuda H, Asahara T. Endothelial progenitor cells for postnatal vasculogenesis. Clin Exp Nephrol 2007; 11:18-25. [PMID: 17384994 DOI: 10.1007/s10157-006-0448-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Bone marrow-derived endothelial progenitor cells (EPCs) are present in the systemic circulation, are augmented in response to certain cytokines and/or tissue ischemia, and are home to--as well as incorporate into--sites of neovascularization. On the basis of these aspects, EPCs have attractive potential therapeutic applications for cardiovascular ischemic diseases as a novel cell-based strategy, mainly via a vasculogenesis mechanism. This review provides an update of the biology of EPCs, as well as highlighting the potential use of these cells for therapeutic regeneration.
Collapse
Affiliation(s)
- Masamichi Eguchi
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Bohseidai, Isehara, Kanagawa, 259-1193, Japan
| | | | | |
Collapse
|
224
|
Wang XX, Zhang FR, Shang YP, Zhu JH, Xie XD, Tao QM, Zhu JH, Chen JZ. Transplantation of autologous endothelial progenitor cells may be beneficial in patients with idiopathic pulmonary arterial hypertension: a pilot randomized controlled trial. J Am Coll Cardiol 2007; 49:1566-71. [PMID: 17418297 DOI: 10.1016/j.jacc.2006.12.037] [Citation(s) in RCA: 222] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 12/06/2006] [Accepted: 12/08/2006] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The goal of this study was to investigate the feasibility, safety, and initial clinical outcome of intravenous infusion of autologous endothelial progenitor cells (EPCs) in patients with idiopathic pulmonary arterial hypertension (IPAH). BACKGROUND Experimental data suggest that transplantation of EPCs attenuates monocrotaline-induced pulmonary hypertension in rats and dogs. In addition, clinical studies suggest that autologous progenitor cell transplantation is feasible and safe in patients with ischemic diseases. METHODS We conducted a prospective, randomized trial comparing the effects of EPC transplantation plus conventional therapy with those of conventional therapy alone in patients with IPAH. The primary end point was change in the 6-min walk distance using a standardized protocol. The secondary end points were changes in hemodynamic variables as assessed by right heart catheterization. RESULTS After 12 weeks of follow-up, the mean distance walked in 6 min increased by 48.2 m in the cell infusion group (from 263 +/- 42 m to 312 +/- 34 m), and an increase of 5.7 m occurred in the conventional therapy group (from 264 +/- 42 m to 270 +/- 44 m). The mean difference between the 2 groups was 42.5 m (95% confidence interval 28.7 to 56.3 m, p < 0.001). The patients in the cell infusion group also had significant improvement in mean pulmonary artery pressure, pulmonary vascular resistance, and cardiac output. There were no severe adverse events with cell infusion. CONCLUSIONS This preliminary study showed that intravenous infusion of autologous EPCs seemed to be feasible and safe, and might have beneficial effects on exercise capacity and pulmonary hemodynamics in patients with IPAH. (Safety and Efficacy Study of Transplantation of EPCs to Treat Idiopathic Pulmonary Arterial Hypertension; http://www.clinicaltrials.gov/ct/show/NCT00257413?order=1; NCT00257413).
Collapse
Affiliation(s)
- Xing-Xiang Wang
- Department of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Smadja DM, Cornet A, Emmerich J, Aiach M, Gaussem P. Endothelial progenitor cells: characterization, in vitro expansion, and prospects for autologous cell therapy. Cell Biol Toxicol 2007; 23:223-39. [PMID: 17370127 DOI: 10.1007/s10565-007-0177-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 12/15/2006] [Indexed: 01/14/2023]
Abstract
Injection of hematopoietic stem cells or endothelial progenitor cells (EPCs) expanded ex vivo has been shown to augment neovascularization in adult patients, but the precise origin and identity of the cell population responsible for these clinical benefits are controversial. The limited quantity of EPCs in the circulation has been the main obstacle to clinical trials. Several authors have therefore attempted to expand these cells ex vivo in order to obtain a homogeneous cell therapy product. One possible means of expanding EPCs ex vivo is to activate the thrombin receptor PAR-1 with the specific peptide SFLLRN. Indeed, PAR-1 activation promotes cell proliferation and C-X-C chemokine receptor type 4 (CXCR4) dependent migration and differentiation, with an overall angiogenic effect. This review summarizes the results and rationale of clinical trials of angiogenic therapy, the nature of EPCs, the different methods of ex vivo expansion, and current methods of quantification.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/pharmacology
- Animal Testing Alternatives
- Animals
- Bone Marrow Transplantation/methods
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Clinical Trials as Topic
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/transplantation
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/physiology
- Humans
- Ischemia/therapy
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Peptide Fragments/pharmacology
- Receptor, PAR-1/drug effects
- Receptor, PAR-1/metabolism
- Receptors, CXCR4/drug effects
- Receptors, CXCR4/metabolism
- Transplantation, Autologous
Collapse
|
226
|
Shantsila E, Watson T, Lip GYH. Endothelial progenitor cells in cardiovascular disorders. J Am Coll Cardiol 2007; 49:741-52. [PMID: 17306702 DOI: 10.1016/j.jacc.2006.09.050] [Citation(s) in RCA: 317] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 09/27/2006] [Accepted: 11/28/2006] [Indexed: 12/22/2022]
Abstract
The important role of the vascular endothelium in cardiovascular health is increasingly recognized. However, mature endothelial cells possess limited regenerative capacity. There is therefore much interest in circulating endothelial progenitor cells (EPCs) among the scientific community, especially into their purported role in maintenance of endothelial integrity and function, as well as postnatal neovascularization. It has been suggested that these cells might not only be responsible for the continuous recovery of the endothelium after injury/damage, but also might take part in angiogenesis, giving the hope of new treatment opportunities. Indeed, there is accumulating evidence showing reduced availability and impaired EPC function in the presence of both cardiovascular disease and associated comorbid risk factors. Thus, many studies into the potential for use of EPCs in the clinical setting are being undertaken. The goal of this review article is to provide an overview of data relevant to the clinical role of EPCs and perspectives for treatment of cardiovascular disorders.
Collapse
Affiliation(s)
- Eduard Shantsila
- Haemostasis, Thrombosis, and Vascular Biology Unit, University Department of Medicine, City Hospital, Birmingham, England
| | | | | |
Collapse
|
227
|
Cardier JE, Rivas B, Romano E, Rothman AL, Perez-Perez C, Ochoa M, Caceres AM, Cardier M, Guevara N, Giovannetti R. Evidence of vascular damage in dengue disease: demonstration of high levels of soluble cell adhesion molecules and circulating endothelial cells. ACTA ACUST UNITED AC 2007; 13:335-40. [PMID: 17090406 DOI: 10.1080/10623320600972135] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Clinical evidence suggests that vascular damage plays a key role in the pathophysiology of dengue hemorrhagic fever (DHF). In this study, the authors tested this hypothesis by examining the levels of soluble intercellular adhesion molecule and vascular cell adhesion molecule (sICAM-1 and sVCAM-1), and the presence of circulating endothelial cells (CECs), as evidence of vascular damage, in peripheral blood from DHF patients (n=13). A significant increase in plasma levels of sICAM-1 (n=12) and sVCAM-1 (n=13) was detected by enzyme-linked immunosorbent assay (ELISA) in DHF patients, compared with healthy individuals. Increased numbers of CECs, as detected by the expression of endothelial cell markers (ICAM-1, platelet cell adhesion molecule [PCAM]-1, and CD36) with flow cytometry, were observed in DHF patients (n=4), compared to healthy subjects. The high levels of sICAM-1 and sVCAM-1, together with the presence of CECs in DHF patients, provide further evidence of endothelium damage and activation in DHF patients.
Collapse
Affiliation(s)
- José E Cardier
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Venezuela.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Tara S, Miyamoto M, Asoh S, Ishii N, Yasutake M, Takagi G, Takano T, Ohta S. Transduction of the anti-apoptotic PTD-FNK protein improves the efficiency of transplantation of bone marrow mononuclear cells. J Mol Cell Cardiol 2007; 42:489-97. [PMID: 17229437 DOI: 10.1016/j.yjmcc.2006.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 10/13/2006] [Accepted: 11/17/2006] [Indexed: 11/26/2022]
Abstract
Since most transplanted cells rapidly die in an ischemic environment by hypoxia and hyponutrition, it is crucial to know how to protect transplanted cells for improving transplantation efficiency. We examined whether the transduction of an artificial anti-cell death protein (PTD-FNK) into bone marrow mononuclear cells (BM-MNCs) prevents cell death and improves the transplantation efficiency of BM-MNCs in ischemic regions. Rat bone marrow cells were prepared from the femur and tibia and cultured on dishes precoated with human fibronectin in the absence of serum. BM-MNCs transduced with PTD-FNK survived better than those without the protein (P<0.008) and retained the potential to differentiate into endothelial progenitor cells (EPCs), as judged by the uptake of an acetylated low-density lipoprotein and the ability to bind lectin. Next, we used a co-culture system comprising human umbilical vein endothelial cells (HUVECs) and fibroblasts to examine angiogenic potential. HUVECs pretreated with PTD-FNK survived and formed a blood-vessel-like structure better than untreated cells (P<0.001). When BM-MNCs expressing EGFP were transplanted into ischemic areas of a male rat ischemic hindlimb model, the cells pretreated with PTD-FNK were incorporated into blood vessel with a higher efficiency than the untreated BM-MNCs (P=0.03). BM-MNCs protected through transduction of PTD-FNK maintained their angiogenic potential. Thus, PTD-FNK improves the transplantation efficiency of BM-MNCs into ischemic regions.
Collapse
Affiliation(s)
- Shuhei Tara
- Department of Biochemistry and Cell Biology, Institute of Development and Aging Sciences, Graduate School of Medicine, Nippon Medical School, Kawasaki, Kanagawa 211-8533, Japan
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Capla JM, Grogan RH, Callaghan MJ, Galiano RD, Tepper OM, Ceradini DJ, Gurtner GC. Diabetes impairs endothelial progenitor cell-mediated blood vessel formation in response to hypoxia. Plast Reconstr Surg 2007; 119:59-70. [PMID: 17255657 DOI: 10.1097/01.prs.0000244830.16906.3f] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Diabetics suffer from vascular dysfunction with increased risks of coronary artery disease and peripheral vascular disease secondary to an impaired ability to respond to tissue ischemia. Because endothelial progenitor cells are known to home to sites of ischemia and participate in new blood vessel growth, the authors examined the effects of diabetes on human endothelial progenitor cell function and peripheral tissue signaling in hypoxia, and determined whether these cells might be a useful cell-based therapy for diabetic vascular complications. METHODS Circulating human endothelial progenitor cells from type 2 diabetic patients and controls were isolated and subjected to in vitro adhesion, migration, and proliferation assays (n = 5). Cell mobilization and recruitment were studied in vivo in diabetic and nondiabetic environments (n = 6). Exogenous human diabetic and normal cells were analyzed for therapeutic efficacy in a murine ischemia model (n = 6). RESULTS Adhesion, migration, and proliferation of human diabetic endothelial progenitor cells in response to hypoxia was significantly reduced compared with controls. In diabetic mice, cell mobilization from the bone marrow and recruitment into ischemic tissue was significantly reduced compared with controls. Normal cells injected systemically as replacement therapy in a diabetic mouse increased but did not normalize ischemic tissue survival. CONCLUSIONS These findings suggest that diabetes causes defects in both the endothelial progenitor cell and peripheral tissue responses to hypoxia. These changes in endothelial progenitor cell function and signaling offer a novel explanation for the poor clinical outcome of type 2 diabetics following ischemic events. Based on these findings, it is unlikely that endothelial progenitor cell-based cellular therapies will be able to prevent diabetic complications.
Collapse
Affiliation(s)
- Jennifer M Capla
- New York, N.Y.; and Stanford, Calif. From the Laboratory of Microvascular Research and Vascular Tissue Engineering, Institute of Reconstructive Plastic Surgery, New York University Medical Center; and Department of Surgery, Stanford University
| | | | | | | | | | | | | |
Collapse
|
230
|
Bluff JE, O'Ceallaigh S, O'Kane S, Ferguson MWJ, Ireland G. The microcirculation in acute murine cutaneous incisional wounds shows a spatial and temporal variation in the functionality of vessels. Wound Repair Regen 2007; 14:434-42. [PMID: 16939571 DOI: 10.1111/j.1743-6109.2006.00142.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A mouse perfusion model using fluorescently labeled dextran has been developed to investigate the functionality of blood vessels during cutaneous wound healing. By immunostaining cryostat sections of perfused wounds with antibodies that identify vessels, we were able to assess their functionality. There was an increase in the proportion of CD31(+)-perfused vessels in all wound regions with time, although the vessels of the wound margins and superficial granulation tissue (GT) took the longest to become perfused. More than 50% of the latter vessels were not perfused at 10 days postwounding. This is consistent with the growth of functional vessels from the wound base proceeding to the more superficial GT. The CD34 marker was expressed by a subpopulation of CD31(+) vessels. However, in contrast to CD31(+) vessels, the functionality of CD34(+) vessels did not change significantly with time and 50-75% of CD34(+) vessels in the GT and wound margins were nonfunctional. This might be explained either by apoptosis of the CD34(+) vessels or the loss of the marker with time. This study has important implications for assays of wound-healing angiogenesis based on histology and immunohistochemical markers for vessels, because vessel functionality differs both spatially and temporally during wound healing.
Collapse
Affiliation(s)
- Joanne E Bluff
- UK Centre for Tissue Engineering, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
231
|
Shah PB, Losordo DW. Angiogenesis. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
232
|
|
233
|
Mehra N, Penning M, Maas J, Beerepoot LV, van Daal N, van Gils CH, Giles RH, Voest EE. Progenitor marker CD133 mRNA is elevated in peripheral blood of cancer patients with bone metastases. Clin Cancer Res 2006; 12:4859-66. [PMID: 16914572 DOI: 10.1158/1078-0432.ccr-06-0422] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE We examined whether RNA expression of CD133, a surface molecule expressed on progenitors from hematopoietic and endothelial lineages, and CD146, a pan-endothelial marker, are increased in the blood of cancer patients and whether these factors correlate with patient characteristics and are predictive factors of survival. EXPERIMENTAL DESIGN We developed a real-time quantification method (nuclear acid sequence-based amplification) to determine expression of CD146 and CD133 mRNA in the peripheral blood mononuclear cells of 131 progressive cancer patients, 37 healthy volunteers, and 5 patients who received granulocyte colony-stimulating factor. Overall survival and other clinicopathologic variables were obtained. Cox proportional hazards studies were done. RESULTS We show that patients with metastatic disease have a significant increase in CD133 mRNA (P = 0.03), specifically patients with bone metastasis (P < 0.001). Cancer patients with high CD133 mRNA expression, using a defined cutoff value, show a decreased survival compared with patients with low or undetectable CD133 expression (21% versus 45% cumulative survival, respectively, after 20 months; P = 0.01). Among patients with metastasis to the bone, cumulative survival was 22%, compared with 61% for patients with high or low CD133 levels (P = 0.004). Multivariate analysis showed that CD133 expression is an independent predictor for overall survival in patients with bone metastases. CD146 mRNA was not increased in patients with cancer, nor did it correlate with clinical variables or survival. CONCLUSION CD133, but not CD146, mRNA expression is increased in cancer patients with metastatic disease, specifically with bone metastasis. In addition, CD133 mRNA expression seems to be an independent prognostic factor for overall survival.
Collapse
MESH Headings
- AC133 Antigen
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Antigens, CD/genetics
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Bone Neoplasms/blood
- Bone Neoplasms/genetics
- Bone Neoplasms/secondary
- CD146 Antigen/biosynthesis
- CD146 Antigen/blood
- CD146 Antigen/genetics
- Female
- Glycoproteins/biosynthesis
- Glycoproteins/blood
- Glycoproteins/genetics
- Humans
- Leukocytes, Mononuclear/metabolism
- Male
- Middle Aged
- Neoplasm Metastasis
- Neoplasms/blood
- Neoplasms/genetics
- Neoplasms/pathology
- Peptides/blood
- Peptides/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/blood
- RNA, Messenger/genetics
- Reproducibility of Results
Collapse
Affiliation(s)
- Niven Mehra
- Department of Medical Oncology and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Heeschen C, Chang E, Aicher A, Cooke JP. Endothelial progenitor cells participate in nicotine-mediated angiogenesis. J Am Coll Cardiol 2006; 48:2553-60. [PMID: 17174197 DOI: 10.1016/j.jacc.2006.07.066] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 07/10/2006] [Accepted: 07/24/2006] [Indexed: 11/18/2022]
Abstract
OBJECTIVES We aimed to determine the role of endothelial progenitor cells (EPCs) in cholinergic angiogenesis. BACKGROUND Recently, we provided evidence for a new angiogenic pathway mediated by endothelial nicotinic acetylcholine receptors (nAChR). Increasing evidence suggests that circulating EPCs also contribute to postnatal neovascularization by homing to sites of neovascularization, a process termed postnatal vasculogenesis. Therefore, we investigated whether nAChR activation increases mobilization and/or recruitment of EPCs to a site of angiogenesis. METHODS To identify EPCs from reservoirs both inside and outside of the bone marrow and to avoid the adverse effects of total body irradiation, we employed a murine parabiosis model with tie-2-LacZ FvB/N mice connected to wild-type FvB/N mice and induced unilateral hind limb ischemia in the wild-type animal. RESULTS Administration of nicotine increased capillary density in the ischemic hind limb, and increased soluble Kit ligand plasma levels. The effect of systemic administration was greater than that of local delivery of nicotine (45% vs. 76% increase in capillary density by comparison to vehicle control, intramuscular vs. oral administration of nicotine; p < 0.05). Ischemia-induced incorporation of EPC in the control group was rare, but was increased 5-fold by systemic administration of nicotine. Exposure to nicotine in vitro increased EPC count and EPC transmigration. Finally, systemic administration of nicotine increased EPC number in the bone marrow and spleen during hind limb ischemia. CONCLUSIONS Nicotine treatment increased the number of EPCs in the bone marrow and spleen, and increased their incorporation into the vasculature of ischemic tissue. Administration of nicotine increased markers of EPC mobilization. This study indicates that the known angiogenic effect of nicotine may be mediated in part by mobilization of precursor cells.
Collapse
Affiliation(s)
- Christopher Heeschen
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
235
|
Shintani S, Kusano K, Ii M, Iwakura A, Heyd L, Curry C, Wecker A, Gavin M, Ma H, Kearney M, Silver M, Thorne T, Murohara T, Losordo DW. Synergistic effect of combined intramyocardial CD34+ cells and VEGF2 gene therapy after MI. ACTA ACUST UNITED AC 2006; 3 Suppl 1:S123-8. [PMID: 16501618 DOI: 10.1038/ncpcardio0430] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2005] [Accepted: 10/27/2005] [Indexed: 12/14/2022]
Abstract
Previous studies have shown that local angiogenic gene therapy acts, in part, by recruiting endothelial progenitor cells (EPCs) to ischemic tissue. Recent data indicate that patients with the most severe vascular disease may have insufficient or deficient EPCs and the poorest response to angiogenic therapy. Accordingly, we hypothesized that combining human CD34(+) cell implantation with local vascular endothelial growth factor 2 (phVEGF2) gene therapy might overcome these deficiencies. The addition of VEGF2 to EPC cultures resulted in significant and dose-dependent decreases in EPC apoptosis. Phosphorylated Akt (p-Akt) was increased in VEGF2-treated EPCs. In vivo, myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in 34 immunodeficient rats. The animals were then randomized to one of four treatment groups: cell therapy alone with human CD34(+) cells; VEGF2 gene therapy alone; combination therapy with CD34(+) cells plus phVEGF2; or CD34(-) cells and 50 microg empty plasmid. Four weeks after MI, animals treated with combination therapy showed improved fractional shortening, increased capillary density, and reduced infarct size compared with the other three groups. Combination therapy was also associated with an increased number of circulating EPCs 1 week after MI. Combined subtherapeutic doses of cell and gene therapy result in a significant therapeutic effect compared to monotherapy. This approach may overcome therapeutic failures (e.g. inability of certain patients to mobilize sufficient EPCs) and may also offer safety advantages by allowing lower dosing strategies.
Collapse
Affiliation(s)
- Satoshi Shintani
- Division of Cardiovascular Research, Caritas St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Alsberg E, von Recum HA, Mahoney MJ. Environmental cues to guide stem cell fate decision for tissue engineering applications. Expert Opin Biol Ther 2006; 6:847-66. [PMID: 16918253 DOI: 10.1517/14712598.6.9.847] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The human body contains a variety of stem cells capable of both repeated self-renewal and production of specialised, differentiated progeny. Critical to the implementation of these cells in tissue engineering strategies is a thorough understanding of which external signals in the stem cell microenvironment provide cues to control their fate decision in terms of proliferation or differentiation into a desired, specific phenotype. These signals must then be incorporated into tissue regeneration approaches for regulated exposure to stem cells. The precise spatial and temporal presentation of factors directing stem cell behaviour is extremely important during embryogenesis, development and natural healing events, and it is possible that this level of control will be vital to the success of many regenerative therapies. This review covers existing tissue engineering approaches to guide the differentiation of three disparate stem cell populations: mesenchymal, neural and endothelial. These progenitor cells will be of central importance in many future connective, neural and vascular tissue regeneration technologies.
Collapse
Affiliation(s)
- Eben Alsberg
- Case Western Reserve University, Department of Biomedical Engineering, 10900 Euclid Avenue, Wickenden Building, Room 204, Cleveland, OH 44106-7207, USA.
| | | | | |
Collapse
|
237
|
Abstract
Several landmark clinical trials suggest that 3-hydroxyl-3-methylglutaryl coenzyme A reductase inhibitors (statins) have additional cardiovascular protective activity that may function independently of their ability to lower serum cholesterol. The cardiovascular protective effects of statins are partly caused by the activation of postnatal neovascularization. At therapeutic doses, statins promote proliferation, migration and survival of endothelial cells, induce mobilization and differentiation of bone marrow-derived endothelial progenitor cells by stimulating the serine/threonine protein kinase Akt (also known as protein kinase B) and nitric oxide (NO) signal pathway. However, at excessive doses, statins may decrease protein isoprenylation as well as inhibit endothelial cell growth and migration. NO is an important signaling molecule that regulates a wide range of physiological and pathological processes in different tissues. There is substantial evidence that effective neovascularization requires endothelium-derived NO. Statins have pleiotropic effects on the expression and activity of endothelial nitric oxide synthase (eNOS) and lead to improved NO bioavailability. NO plays an important role in the effects of statins on neovascularization. In this review, we focus on the effects of statins on neovascularization and highlight specific novel targets, such as endothelial progenitor cells and NO.
Collapse
Affiliation(s)
- Feng Xia Ma
- State Key Laboratory of Experimental Hematology and National Research Center for Stem Cell Engineering and Technology, Institute of Hematology, Chinese Academy of Medical Sciences & Peking Union of Medical College, Tianjin, China
| | | |
Collapse
|
238
|
Abstract
Over the past decade, the old idea that the bone marrow contains endothelial cell precursors has become an area of renewed interest. While some still believe that there are no endothelial precursors in the blood, even among those who do, there is no consensus as to what they are or what they do. In this review, we describe the problems in identifying endothelial cells and conclude that expression of endothelial nitric oxide synthase may be the most reliable antigenic indicator of the phenotype. The evidence for two different classes of endothelial precursors is also presented. We suggest that, though there is no single endothelial cell precursor, we may be able to use these phenotypic variations to our advantage in better understanding their biology. We also discuss how a variety of genetic, epigenetic, and methodological differences can account for the seemingly contradictory findings on the physiological relevance of bone marrow-derived precursors in normal vascular maintenance and in response to injury. Data on the impact of tumor type and location on the contribution of bone marrow-derived cells to the tumor vasculature are also presented. These data provide hope that we may ultimately be able to predict those tumors in which bone marrow-derived cells will have a significant contribution and design therapies accordingly. Finally, factors that regulate bone marrow cell recruitment to and function in the endothelium are beginning to be identified, and several of these, including stromal derived factor 1, monocyte chemoattractant factor-1, and vascular endothelial growth factor are discussed.
Collapse
Affiliation(s)
- Gina C Schatteman
- Integrative Physiology FH412, Univ. of Iowa, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
239
|
Miriuka SG, Rao V, Peterson M, Tumiati L, Delgado DH, Mohan R, Ramzy D, Stewart D, Ross HJ, Waddell TK. mTOR inhibition induces endothelial progenitor cell death. Am J Transplant 2006; 6:2069-79. [PMID: 16796720 DOI: 10.1111/j.1600-6143.2006.01433.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Immunosuppressants are necessary to prevent graft rejection after solid organ transplantation. However, they are also known to have significant side effects, including endothelial toxicity. Endothelial progenitor cells originate in the bone marrow and are recognized by their angiogenic and endothelial reparative properties. The effects of the immunosuppressants cyclosporine A (CyA), tacrolimus and rapamycin were analyzed on endothelial progenitor-like cells. Rapamycin induced rapid cell death, even at concentrations much lower than those used clinically, in peripheral blood mononuclear cells (PBMC) cultured to favor outgrowth of endothelial progenitors. Cyclosporine A and tacrolimus had no significant effects at clinical concentrations. The effect of rapamycin was specific to endothelial progenitor cells, in particular to the early stages of differentiation, as a lesser effect was observed in late outgrowth endothelial progenitors, mature aortic endothelial cells, and macrophages derived from the same PBMCs. The mechanism of cell death appeared to be apoptosis; however, its induction was probably multifactorial and did not depend on caspase or cathepsin activation. In conclusion, rapamycin induces endothelial progenitor cell death, possibly because it blocks survival signals given by growth factors critically required by these cells.
Collapse
Affiliation(s)
- S G Miriuka
- Department of Medicine, Toronto General Hospital, Faculty of Medicine, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Asahara T. Stem cell biology for vascular regeneration. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2006:111-29. [PMID: 16080290 DOI: 10.1007/3-540-37644-5_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The isolation of endothelial progenitor cells (EPCs) derived from bone marrow (BM) was one epoch-making event for the recognition of neovessel formation in adults occurring as physiological and pathological responses. The finding that EPCs home to sites of neovascularization and differentiate into endothelial cells (ECs) in situ is consistent with vasculogenesis, a critical paradigm that has been well described for embryonic neovascularization, but proposed recently in adults in which a reservoir of stem or progenitor cells contribute to vascular organogenesis. EPCs have also been considered as therapeutic agents to supply the potent origin of neovascularization under pathological conditions. This chapter highlights an update of EPC biology as well as its potential use for therapeutic regeneration.
Collapse
Affiliation(s)
- T Asahara
- Riken Center for Developmental Biology, Kobe, Japan.
| |
Collapse
|
241
|
Laufs U, Urhausen A, Werner N, Scharhag J, Heitz A, Kissner G, Böhm M, Kindermann W, Nickenig G. Running exercise of different duration and intensity: effect on endothelial progenitor cells in healthy subjects. ACTA ACUST UNITED AC 2006; 12:407-14. [PMID: 16079651 DOI: 10.1097/01.hjr.0000174823.87269.2e] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Increased numbers of circulating endothelial progenitor cells (EPC) are associated with improved vascular function. Exercise is a central component of the primary prevention of vascular diseases. The effect of physical activity on circulating EPC in healthy individuals is not known. DESIGN A prospective crossover study. METHODS AND RESULTS In order to study a potential link between the extent of physical exercise and progenitor cells in humans, EPC were quantified by flow cytometry and cell culture in 25 healthy volunteers undergoing three protocols of running exercise. Intensive running, defined as 30 min at 100% of the velocity of the individual anaerobic threshold (IAT; approximately 82% maximal oxygen consumption; VO2max), as well as moderate running with 30 min at 80% of the velocity of the IAT ( approximately 68% VO2max), increased circulating EPC numbers to 235+/-93% and 263+/-106% of control levels, respectively. However, moderate short-term running for 10 min did not upregulate EPC counts. The maximum increase in circulating EPC numbers was observed 10-30 min after intensive running. Exercise increased EPC migratory and colony-forming capacity. CONCLUSIONS Intensive and moderate exercising for 30 min, but not for 10 min, increased circulating levels of EPC, which may represent an important beneficial outcome of physical exercise. The data support the notion that increased numbers of EPC correlate with cardiovascular health and suggest EPC quantification as a novel surrogate parameter of the vascular effects of exercising.
Collapse
Affiliation(s)
- Ulrich Laufs
- Medizinische Klinik und Poliklinik der Universität des Saarlandes, Innere Medizin III, 66421 Homburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Gridelli C, Rossi A, Maione P. New antiangiogenetic agents and non-small cell lung cancer. Crit Rev Oncol Hematol 2006; 60:76-86. [PMID: 16843002 DOI: 10.1016/j.critrevonc.2006.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Revised: 01/27/2006] [Accepted: 01/27/2006] [Indexed: 01/02/2023] Open
Abstract
New blood vessel formation, known as angiogenesis is a fundamental event in the process of tumor growth and metastatic dissemination. Due to its central role in tumor angiogenesis, the vascular endothelial growth factor (VEGF) and its receptor have been a major focus of basic research and drug development in the field of oncology, including the treatment of non-small cell lung cancer (NSCLC). Approaches targeting VEGF include monoclonal antibodies and vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs). Bevacizumab (Avastin) is an anti-VEGF recombinant humanized monoclonal antibody. A very recent randomized phase III trial demonstrated a statistically significant advantage in median survival favouring the combination of bevacizumab plus chemotherapy versus chemotherapy alone in the treatment of advanced non-squamous NSCLC. This study represents the first evidence of superior efficacy of targeted therapy combined with chemotherapy over chemotherapy alone in the treatment of NSCLC. ZD6474 is an orally bioavailable inhibitor of VEGFR-2 tyrosine kinase. First evidences of antitumor activity and its excellent toxicity profile make it a promising targeted agent for the treatment of NSCLC. A recent phase I/II study examined the combination of Epidermal Growth Factor Receptor (EGFR)-TKI erlotinib and bevacizumab in patients with non-squamous stage IIIB/IV NSCLC. Data on antitumor activity of this combination have to be considered very promising. Clinical trials of multiple targeted therapy may represent the second generation studies in the treatment of NSCLC.
Collapse
Affiliation(s)
- C Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Contrada Amoretta, Avellino, Italy.
| | | | | |
Collapse
|
243
|
Gaballa MA, Sunkomat JNE, Thai H, Morkin E, Ewy G, Goldman S. Grafting an acellular 3-dimensional collagen scaffold onto a non-transmural infarcted myocardium induces neo-angiogenesis and reduces cardiac remodeling. J Heart Lung Transplant 2006; 25:946-54. [PMID: 16890116 DOI: 10.1016/j.healun.2006.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 04/11/2006] [Accepted: 04/17/2006] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND This study was designed to determine whether tissue engineering could be used to reduce ventricular remodeling in a rat model of non-transmural, non-ST-elevation myocardial infarction. METHODS We grafted an acellular 3-dimensional (3D) collagen type 1 scaffold (solid porous foam) onto infarcted myocardium in rats. Three weeks after grafting, the scaffold was integrated into the myocardium and retarded cardiac remodeling by reducing left ventricular (LV) dilation. The LV inner and outer diameters, measured at the equator at zero LV pressure, decreased (p < 0.05) from 11,040 +/- 212 to 9,144 +/- 135 microm, and 13,469 +/- 187 to 11,673 +/- 104 microm (N = 12), after scaffold transplantation onto infarcted myocardium. The scaffold also shifted the LV pressure-volume curve to the left toward control and induced neo-angiogenesis (700 +/- 25 vs 75 +/- 11 neo-vessels/cm2, N = 5, p < 0.05). These vessels (75 +/- 11%) ranged in diameter from 25 to 100 microm and connected to the native coronary vasculature. Systemic treatment with granulocyte-colony stimulating factor (G-CSF), 50 microg/kg/day for 5 days immediately after myocardial injury, increased (p < 0.05) neo-vascular density from 700 +/- 25 to 978 +/- 57 neo-vessels/cm2. CONCLUSIONS A 3D collagen type 1 scaffold grafted onto an injured myocardium induced neo-vessel formation and reduced LV remodeling. Treatment with G-CSF further increased the number of vessels in the myocardium, possibly due to mobilization of bone marrow cells.
Collapse
Affiliation(s)
- Mohamed A Gaballa
- Section of Cardiology, University of Arizona Sarver Heart Center, Tucson, Arizona, USA
| | | | | | | | | | | |
Collapse
|
244
|
Abstract
Strong evidence suggests that bone marrow-derived cells play a role in physiological and pathological blood vessel growth in the adult, both by augmenting angiogenesis through the secretion of angiogenic growth factors and by providing a rich source of progenitor cells that can differentiate into mature vascular endothelial cells. This is a true paradigm shift, since adult neovascularization processes were thought to be limited to angiogenesis. The cells that are critical to postnatal blood vessel growth – endothelial progenitor cells – may be analogous to the embryonic angioblast, in that they can circulate, proliferate and participate in the development of vascular networks by differentiating in situ, probably via the formation of cell clusters into mature endothelial cells. Therefore, initial reports have seen analogs to the process of vasculogenesis in the embryo, where the de novo synthesis of vessels occurs through the formation of blood island-like clusters, which subsequently connect and eventually form systemic vasculature. Recent work implicates precursors of endothelial cells in such processes as myocardial ischemia and infarction, limb ischemia, wound healing, atherosclerosis, endogenous endothelial repair and tumor vascularization. These new insights into the vascular biology of endothelial regeneration and repair led to the development of new cell therapeutic strategies to enhance adult neovascularization and re-endothelialization in ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Christoph Kalka
- Swiss Cardiovascular Center, Department of Vascular Medicine, Inselspital, Freiburgstrasse, 3010 Bern, Switzerland
| | - Stefano Di Santo
- Swiss Cardiovascular Center, Department of Vascular Medicine, Inselspital, University Hospital of Bern, Switzerland
| |
Collapse
|
245
|
Romagnani P, Lasagni L, Romagnani S. Peripheral blood as a source of stem cells for regenerative medicine. Expert Opin Biol Ther 2006; 6:193-202. [PMID: 16503730 DOI: 10.1517/14712598.6.3.193] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Converging evidence indicates that peripheral blood (PB) contains stem cells (SCs) with multidifferentiation potential, thus representing a potential source for regenerative medicine in several human disorders, as has also been confirmed by promising results obtained in several preliminary clinical trials. In addition to the classic haematopoietic SCs, PB also harbours endothelial progenitor cells, mesenchymal SCs, tissue-committed SCs and monocyte-like SCs. In spite of a series of different names and/or definitions, a large overlap seems to exist among surface markers, functions and origin of these different SC types. This review analyses the different subsets of SCs described in PB, the different hypotheses suggested to explain their origin, and the possible mechanisms that provide the basis for their biological potential.
Collapse
Affiliation(s)
- Paola Romagnani
- University of Florence, Center for Research, Transfer and High Education DENOthe, Viale Pieraccini 6, Florence 50139, Italy.
| | | | | |
Collapse
|
246
|
Li B, Sharpe EE, Maupin AB, Teleron AA, Pyle AL, Carmeliet P, Young PP. VEGF and PlGF promote adult vasculogenesis by enhancing EPC recruitment and vessel formation at the site of tumor neovascularization. FASEB J 2006; 20:1495-7. [PMID: 16754748 DOI: 10.1096/fj.05-5137fje] [Citation(s) in RCA: 271] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There are growing data to suggest that tissue hypoxia represents a critical force that drives adult vasculogenesis. Vascular endothelial growth factor (VEGF) expression is dramatically up-regulated by hypoxia and results in enhanced neovascularization. Although the role of VEGF in angiogenesis has been well characterized, its role in adult vasculogenesis remains poorly understood. We used two distinct murine bone marrow transplantation (BMT) models to demonstrate that increased VEGF levels at the site of tumor growth promoted vasculogenesis in vivo. This effect of VEGF was downstream of its effect to enhance either mobilization or survival of circulating endothelial progenitor cells (EPCs). Both VEGFR1 (flt1) and VEGFR2 (flk1) are expressed on culture expanded human EPCs. Previous studies suggest that the effect of VEGF on endothelial cell migration is primarily mediated via VEGFR2; however, VEGF-induced EPC migration in vitro was mediated by both receptors, suggesting that VEGF-VEGFR1 interactions in EPCs are distinct from differentiated endothelial cells. We used specific blocking antibodies to these receptors to demonstrate that VEGFR1 plays an important role in human EPC recruitment to tumors. These findings were further supported by our finding that tumor-associated placental growth factor (PlGF), a VEGFR1-specific agonist, increased tumor vasculogenesis in a murine BMT model. We further showed that both VEGF receptors were necessary for the formation of functional vessels derived from exogenously administered human ex vivo expanded EPCs. Our data suggest local VEGF and/or PlGF expression promote vasculogenesis; VEGF plays a role in EPC recruitment and subsequent formation of functional vessels.
Collapse
Affiliation(s)
- Bin Li
- Department of Pathology, Vanderbilt University School of Medicine, 1161 21st Ave. South, C2217 MCN, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | |
Collapse
|
247
|
Liew A, Barry F, O'Brien T. Endothelial progenitor cells: diagnostic and therapeutic considerations. Bioessays 2006; 28:261-70. [PMID: 16479582 DOI: 10.1002/bies.20372] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Endothelial progenitor cells (EPCs) may be defined as adherent cells derived from peripheral blood- or bone marrow-derived mononuclear cells demonstrating acLDL uptake and isolectin-binding capacity. The number of circulating EPCs inversely correlates with the number of cardiovascular risk factors and is reduced in cardiovascular disease. This measurement may therefore serves as a surrogate marker for cardiovascular disease risk. EPC numbers can be modified by various means. However, the effectiveness of risk-factor modification on EPC number and function is currently unknown. Furthermore, EPCs may be used as a potential therapy for a variety of vascular disease states including ischaemia, restenosis and pulmonary hypertension. This review provides an update on multiple factors that affect EPC number as well as highlighting the potential use of EPCs as a novel marker of vascular dysfunction. Furthermore, potential gene- and/or EPC-based approaches to a number of vascular disease states are explored.
Collapse
Affiliation(s)
- Aaron Liew
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway
| | | | | |
Collapse
|
248
|
Allende A, Madigan MC, Provis JM. Endothelial cell proliferation in the choriocapillaris during human retinal differentiation. Br J Ophthalmol 2006; 90:1046-51. [PMID: 16613918 PMCID: PMC1857208 DOI: 10.1136/bjo.2006.092080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Differentiation patterns of the neural retina and its retinal vasculature are not well matched. The foveal region differentiates first, however the central retina is not vascularised until late in gestation. The authors explored the hypothesis that higher rates of endothelial cell proliferation in the choriocapillaris of the central retina might compensate for the slow growth of central retinal vessels, providing supplementary nutrients to the region during the early stages of neuronal maturation. METHODS Frozen sections of five human fetal eyes (14-18.5 weeks' gestation), were examined for Ki-67 and CD34 immunoreactivity using confocal microscopy. Measurements of choriocapillaris area and the number of proliferating choroidal endothelial cells were used to calculate the rate of choroidal endothelial proliferation at five different chorioretinal locations. RESULTS The choriocapillaris area is consistently greater in the foveal region than at other locations and increases progressively with age. A higher rate of endothelial cell proliferation was found in parts of the choriocapillaris associated with the undifferentiated (proliferating) neural retina, compared with the differentiated, central region. CONCLUSION The findings suggest that mechanisms regulating proliferation and growth of the choroidal vasculature are independent of differentiation in the neural retina, and are thus profoundly different from mechanisms that regulate formation of the retinal vasculature.
Collapse
Affiliation(s)
- A Allende
- Discipline of Clinical Ophthalmology and Save Sight Institute, University of Sydney, GPO 4337, Sydney, NSW 2001, Australia.
| | | | | |
Collapse
|
249
|
Abstract
The concept of an Endothelial Progenitor Cell (EPC) that participates in adult angiogenesis is less than a decade old, yet it has received a great deal of attention due to its potential for cell-based clinical therapies in many pathologies. However, controversy remains as to the identity of this bone marrow-derived cell type and its ability to give rise to new endothelium in the adult. Reports on the contribution of EPCs to new vessels in ischemic tissue or tumors vary widely, ranging from 80-90% to negligible. As researchers hone their ability to identify, isolate, and expand these cells by their markers and functionality, mounting evidence suggests that they might constitute multiple, but related cell types. At least two general phenotypes have emerged from studies of bone marrow-derived cells contributing to angiogenesis: one that incorporates into the endothelial wall directly contributing to vascular expansion and another that is able to home to neovessels, but it locates behind the endothelial wall. Nonetheless, experimental evidence indicates that this second cell type supports the viability of newly formed vessels and thus it is equally relevant to neovascular growth. As our understanding of neovascularization in pathologic states expands, a more clear definition of the multiple cellular components required for the process will shed light into new models of therapeutic intervention. The identification of a cell type that could be isolated, expanded and infused into a patient would be very useful for promoting angiogenesis in ischemia, myocardial infarct and other pathologies.
Collapse
Affiliation(s)
- Chad L Barber
- Molecular Biology Institute, University of California, Los Angeles, 90095, USA
| | | |
Collapse
|
250
|
Jin YL, Enzan H, Kuroda N, Hayashi Y, Toi M, Miyazaki E, Hamauzu T, Hiroi M, Guo LM, Shen ZS, Saibara T. Vascularization in tissue remodeling after rat hepatic necrosis induced by dimethylnitrosamine. Med Mol Morphol 2006; 39:33-43. [PMID: 16575513 DOI: 10.1007/s00795-005-0306-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 11/14/2005] [Indexed: 01/31/2023]
Abstract
We observed postnecrotic tissue remodeling to examine vascularization in adult rat livers. Livers, bone marrow, and peripheral blood from rats at 24 h to 14 days after an injection of dimethylnitrosamine (DMN) were examined by light microscopic, immunohistochemical, and ultrastructural methods. Numerous ED-1 (a marker for rat monocytes/macrophages)-positive round mononuclear cells infiltrated in the necrotic areas at 36 h after DMN treatment. On day 5, when necrotic tissues were removed, some of the cells were transformed from round to spindle in shape. On day 7, these cells were contacted with residual reticulin fibers and became positive for SE-1, a marker of hepatic sinusoidal endothelial cells and Tie-1, an endothelial cell-specific surface receptor, associated with frequent occurrence of ED-1/SE-1 and ED-1/Tie-1 double-positive spindle cells. Ultrastructurally, the spindle cells simultaneously showed phagocytosis and endothelial cell-like morphology. With time necrotic areas diminished, and on day 14, the necrotic tissues were almost replaced by regenerated liver tissues and thin bundles of central-to-central bridging fibrosis. Bone marrow from 12 h to day 2 showed an increase of BrdU-positive mononuclear cells. Some of them were positive for ED-1. The BrdU-labeled and ED-1-positive cells appeared as early as 12 h after DMN injection and reached a peak in number at 36 h. They were similar in structure to ED-1-positive cells in necrotic liver tissues. These findings suggest that round mononuclear ED-1-positive cells proliferate first in bone marrow after DMN treatment, reach necrotic areas of the liver through the circulation, and differentiate to sinusoidal endothelial cells. Namely, hepatic sinusoids in DMN-induced necrotic areas may partly be reorganized possibly by vasculogenesis.
Collapse
Affiliation(s)
- Yu-Lan Jin
- Department of Pathology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|