101
|
Lee HJ, Choi CW, Kim EK, Kim HS, Kim BI, Choi JH. Granulocyte colony-stimulating factor reduces hyperoxia-induced alveolarization inhibition by increasing angiogenic factors. Neonatology 2012; 101:278-84. [PMID: 22286224 DOI: 10.1159/000335285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/22/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND Granulocyte colony-stimulating factor (G-CSF) is known to mobilize endothelial progenitor cells (EPCs) from bone marrow. EPCs reportedly promote neovascularization and participate in the repair of lung structure in adult animals. OBJECTIVE We tested the hypothesis that G-CSF contributes to alveolar growth by increasing the production of angiogenic growth factor in the lungs of hyperoxia-exposed neonatal mice. METHODS Neonatal mice were exposed to hyperoxia (80%) or room air (RA) for 7 days and treated with G-CSF (50 μg/kg/day) or vehicle for 5 days. Blood was subjected to flow cytometry to gate for CD45(dim/-)/Sca-1(+)/CD133(+)/vascular endothelial growth factor (VEGF) receptor-2 (VEGFR2) to define the EPC population at day 7. RESULTS The percentage of EPCs in the peripheral blood and VEGF and VEGFR2 levels in the lungs of neonatal mice exposed to hyperoxia were significantly reduced compared to those of mice kept in RA. G-CSF significantly increased EPCs in the peripheral blood, and VEGF and VEGFR2 levels in the lungs of both mice exposed to hyperoxia and mice kept in RA. G-CSF restored alveolarization inhibited by hyperoxia without altering normal alveolarization under RA. CONCLUSION G-CSF restored alveolarization inhibited by hyperoxia in the developing lungs and this alveolarization-enhancing effect of G-CSF is associated with mobilization of EPCs and upregulation of VEGF signaling.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
102
|
Agudelo CA, Tachibana Y, Hurtado AF, Ose T, Iida H, Yamaoka T. The use of magnetic resonance cell tracking to monitor endothelial progenitor cells in a rat hindlimb ischemic model. Biomaterials 2011; 33:2439-48. [PMID: 22206594 DOI: 10.1016/j.biomaterials.2011.11.075] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 11/25/2011] [Indexed: 10/14/2022]
Abstract
A water-soluble magnetic resonance imaging (MRI) contrast agent, Dextran mono-N-succinimidyl 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate-gadolinium(3+) (Dex-DOTA-Gd(3+)), was shown to enable monitoring of the anatomical migration and the survival period of transplanted stem cells for up to 1 month. Gadolinium molecules in the cells were rapidly eliminated from the site and excreted upon cell death. Endothelial progenitor cells (EPCs) transplanted into the inguinal femoral muscle of rats migrated distally through the knee in rats after hindlimb ischemia but did not migrate in non-ischemic rats. Interestingly, the survival period of transplanted EPCs was notably prolonged in the ischemic limb, indicating that EPCs are required by the ischemic tissues and that the fate of transplanted EPCs was affected by the disease. Compared to the commonly used particle type of MRI contrast agents, the system described in this study is expected to be invaluable to help clarifying the process of stem cell transplantation therapy.
Collapse
Affiliation(s)
- Carlos A Agudelo
- Department of Biomedical Engineering, National Cardiovascular Center Research Institute, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | |
Collapse
|
103
|
Abstract
Tumor necrosis factor-alpha (TNF-α) binds to two distinct receptors, TNFR1/p55 and TNFR2/p75. TNF-α is implicated in the processes of tumor growth, survival, differentiation, invasion, metastases, secretion of cytokines and pro-angiogenic factors. We have shown that TNFR2/p75 signaling promotes ischemia-induced angiogenesis via modulation of several angiogenic growth factors. We hypothesized that TNFR2/p75 may promote tumor growth and angiogenesis. Growth of mouse Lewis lung carcinoma (LLC1) and/or mouse melanoma B16 cell was evaluated in wild type (WT), p75 knockout (KO) and double p55KO/p75KO mouse tumor xenograft models. Compared to WT and p55KO/p75KO mice, growth of tumors in p75KO mice was significantly decreased (two-fold) in both LLC and B16 tumors. Tumor growth inhibition was correlated with decreases in VEGF expression and capillary density, as well as bone marrow (BM)-derived endothelial progenitor cells (EPCs) incorporation into the functional capillary network, and an increase in apoptotic cells in LLC xenografts. Gene array analysis of tumor tissues showed a decrease in gene expression in pathways that promote tumor angiogenesis and cell survival. Blocking p75 by shRNA in cultured LLCs led to increases in TNF-mediated apoptosis, as well as decreases in the constitutive and TNF-mediated expression of angiogenic growth factors (VEGF, HGF, PLGF), and SDF-1α receptor CXCR4. In summary, p75 is essential for tumor angiogenesis and survival in highly vascularized murine lung tumor xenografts. Blocking p75 expression may lead to tumor regression. This may represent new and effective therapy against lung neoplasms and potentially tumors of other origin.
Collapse
|
104
|
Mitsos S, Katsanos K, Koletsis E, Kagadis GC, Anastasiou N, Diamantopoulos A, Karnabatidis D, Dougenis D. Therapeutic angiogenesis for myocardial ischemia revisited: basic biological concepts and focus on latest clinical trials. Angiogenesis 2011; 15:1-22. [PMID: 22120824 DOI: 10.1007/s10456-011-9240-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/04/2011] [Indexed: 12/24/2022]
Abstract
Therapeutic angiogenesis is based on the premise that the development of new blood vessels can be augmented by exogenous administration of the appropriate growth factors. Over the last years, successful preclinical studies and promising results of early clinical trials have created great excitement about the potential of therapeutic angiogenesis for patients with advanced ischemic heart disease. The authors provide an overview of the biology of angiogenesis, the basic characteristics of angiogenic factors, and the different routes of their delivery. They discuss experimental studies in animal models of myocardial ischemia and outline available clinical studies on therapeutic angiogenesis for myocardial ischemia. Related safety issues are also addressed followed by a critical perspective about the future of proangiogenic therapies for ischemic cardiovascular disorders. Despite the established proof of concept and reasonable safety, however, results of the latest trials on therapeutic angiogenesis for myocardial ischemia have provided inconsistent results and the definite means of inducing clinically useful therapeutic angiogenesis remain elusive. More studies are required to gain further insights into the biology of angiogenesis and address pharmacological limitations of current approaches of angiogenic therapy. The authors hope and envisage that in the not-too-distant future, these investigative efforts will lead to important new strategies for treatment of myocardial ischemic syndromes. Means of non-invasive individualized pharmacological therapeutic neovascularization may be the next major advance in the treatment of ischaemic heart disease.
Collapse
Affiliation(s)
- Sofoclis Mitsos
- Department of Cardiothoracic Surgery, Onassion Cardiac Surgery Center, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
105
|
Schroeter MR, Stein S, Heida NM, Leifheit-Nestler M, Cheng IF, Gogiraju R, Christiansen H, Maier LS, Shah AM, Hasenfuss G, Konstantinides S, Schäfer K. Leptin promotes the mobilization of vascular progenitor cells and neovascularization by NOX2-mediated activation of MMP9. Cardiovasc Res 2011; 93:170-80. [PMID: 22065732 DOI: 10.1093/cvr/cvr275] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Bone marrow (BM) progenitors participate in new vessel formation and endothelial repair. The leptin receptor (ObR) is expressed on hematopoietic cells; however, the effects of leptin on BM progenitor cells and their angiogenic potential are unknown. METHODS AND RESULTS In the present study, we show that the short-term administration of leptin (over five consecutive days) into wild-type mice increased the number of circulating, BM-derived sca-1(+), flk-1(+) vascular progenitors, 95 ± 1.7% of which also expressed ObR. Ex vivo stimulation of BM cells with leptin enhanced the expression of NADPH oxidase isoform 2 (NOX2), and the leptin-induced increase in reactive oxygen species production, matrix metalloproteinase-9 (MMP9) expression and circulating soluble KitL levels was absent in mice lacking NOX2. Furthermore, intraperitoneal injections of leptin improved perfusion and increased the number of BM-derived, CD31-positive endothelial cells in ischaemic hindlimbs after femoral artery ligation. The effects of leptin on the mobilization of sca-1(+), flk-1(+) cells and neovascularization were abolished in mice transplanted with BM from ObR-deficient and in NOX2(-/-) mice. CONCLUSION Our findings suggest that the angiogenic effects of leptin involve sca-1(+), flk-1(+) vascular progenitor cells mobilized from the BM in response to ObR-mediated activation of NOX2, increased MMP9 expression, and sKitL release.
Collapse
Affiliation(s)
- Marco R Schroeter
- Department of Cardiology and Pulmonary Medicine, Georg August University of Goettingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Huang M, Nguyen P, Jia F, Hu S, Gong Y, de Almeida PE, Wang L, Nag D, Kay MA, Giaccia AJ, Robbins RC, Wu JC. Double knockdown of prolyl hydroxylase and factor-inhibiting hypoxia-inducible factor with nonviral minicircle gene therapy enhances stem cell mobilization and angiogenesis after myocardial infarction. Circulation 2011; 124:S46-54. [PMID: 21911818 DOI: 10.1161/circulationaha.110.014019] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Under normoxic conditions, hypoxia-inducible factor (HIF)-1α is rapidly degraded by 2 hydroxylases: prolyl hydroxylase (PHD) and factor-inhibiting HIF-1 (FIH). Because HIF-1α mediates the cardioprotective response to ischemic injury, its upregulation may be an effective therapeutic option for ischemic heart failure. METHODS AND RESULTS PHD and FIH were cloned from mouse embryonic stem cells. The best candidate short hairpin (sh) sequences for inhibiting PHD isoenzyme 2 and FIH were inserted into novel, nonviral, minicircle vectors. In vitro studies after cell transfection of mouse C2C12 myoblasts, HL-1 atrial myocytes, and c-kit(+) cardiac progenitor cells demonstrated higher expression of angiogenesis factors in the double-knockdown group compared with the single-knockdown and short hairpin scramble control groups. To confirm in vitro data, shRNA minicircle vectors were injected intramyocardially after left anterior descending coronary artery ligation in adult FVB mice (n=60). Functional studies using MRI, echocardiography, and pressure-volume loops showed greater improvement in cardiac function in the double-knockdown group. To assess mechanisms of this functional recovery, we performed a cell trafficking experiment, which demonstrated significantly greater recruitment of bone marrow cells to the ischemic myocardium in the double-knockdown group. Fluorescence-activated cell sorting showed significantly higher activation of endogenous c-kit(+) cardiac progenitor cells. Immunostaining showed increased neovascularization and decreased apoptosis in areas of injured myocardium. Finally, western blots and laser-capture microdissection analysis confirmed upregulation of HIF-1α protein and angiogenesis genes, respectively. CONCLUSIONS We demonstrated that HIF-1α upregulation by double knockdown of PHD and FIH synergistically increases stem cell mobilization and myocardial angiogenesis, leading to improved cardiac function.
Collapse
Affiliation(s)
- Mei Huang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Phase 1-2 study of docetaxel plus aflibercept in patients with recurrent ovarian, primary peritoneal, or fallopian tube cancer. Lancet Oncol 2011; 12:1109-17. [PMID: 21992853 DOI: 10.1016/s1470-2045(11)70244-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Biologically targeted therapies have been postulated as a viable strategy to improve outcomes for women with ovarian cancer. We assessed the safety, tolerance, pharmacokinetics, relevant circulating and image-derived biomarkers, and clinical activity of combination aflibercept and docetaxel in this population. METHODS For the phase 1 (pharmacokinetic) study, eligible patients had measurable, recurrent or persistent epithelial ovarian, primary peritoneal, or fallopian tube carcinoma with a maximum of two prior chemotherapy regimens. Aflibercept was administered intravenously over three dose levels (2, 4, or 6 mg/kg; one dose every 21 days) to identify the maximum tolerated dose for the phase 2 study. Pharmacokinetics were assessed and dynamic imaging was done during a lead-in phase with single-agent aflibercept (cycle 0) and during combination therapy with intravenous docetaxel (75 mg/m(2)). Eligibility for the phase 2 study was the same as for phase 1. Patients were enrolled in a two-stage design and given aflibercept 6 mg/kg intravenously and docetaxel 75 mg/m(2) intravenously, every 3 weeks. The primary endpoint was objective response rate (ORR) as assessed by Response Evaluation Criteria in Solid Tumors version 1.0. The trial has completed enrolment and all patients are now off study. The trial is registered at ClinicalTrials.gov, number NCT00436501. FINDINGS From the phase 1 study, the recommended phase 2 doses of aflibercept and docetaxel were found to be 6 mg/kg and 75 mg/m(2), respectively. Log-linear pharmacokinetics (for unbound aflibercept) were observed for the three dose levels. No dose-limiting toxicities were noted. 46 evaluable patients were enrolled in the phase 2 trial; 33 were platinum resistant (15 refractory) and 13 were platinum sensitive. The confirmed ORR was 54% (25 of 46; 11 patients had a complete response and 14 had a partial response). Grade 3-4 toxicities observed in more than two patients (5%) were: neutropenia in 37 patients (80%); leucopenia in 25 patients (54%); fatigue in 23 patients (50%); dyspnoea in ten patients (22%); and stomatitis in three patients (7%). Adverse events specifically associated with aflibercept were grade 1-2 hypertension in five patients (11%), and grade 2 proteinuria in one patient (2%). INTERPRETATION Combination aflibercept plus docetaxel can be safely administered at the dose and schedule reported here, and is associated with substantial antitumour activity. These findings suggest that further clinical development of this combination in ovarian cancer is warranted. FUNDING US National Cancer Institute, US Department of Defense, Sanofi-Aventis, Gynecologic Cancer Foundation, Marcus Foundation, and the Commonwealth Foundation.
Collapse
|
108
|
Laschke M, Giebels C, Menger M. Vasculogenesis: a new piece of the endometriosis puzzle. Hum Reprod Update 2011; 17:628-636. [DOI: 10.1093/humupd/dmr023] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
109
|
Affiliation(s)
| | - Jeffrey J.D. Henry
- Department of Bioengineering, University of California, Berkeley, California 94720;
| |
Collapse
|
110
|
Abstract
Accumulating evidence indicates that the mobilization and recruitment of circulating or tissue-resident progenitor cells that give rise to endothelial cells (ECs) and smooth muscle cells (SMCs) can participate in atherosclerosis, neointima hyperplasia after arterial injury, and transplant arteriosclerosis. It is believed that endothelial progenitor cells do exist and can repair and rejuvenate the arteries under physiologic conditions; however, they may also contribute to lesion formation by influencing plaque stability in advanced atherosclerotic plaque under specific pathologic conditions. At the same time, smooth muscle progenitors, despite their capacity to expedite lesion formation during restenosis, may serve to promote atherosclerotic plaque stabilization by producing extracellular matrix proteins. This profound evidence provides support to the hypothesis that both endothelial and smooth muscle progenitors may act as a double-edged sword in the pathogenesis of arteriosclerosis. Therefore, the understanding of the regulatory networks that control endothelial and smooth muscle progenitor differentiation is undoubtedly fundamental both for basic research and for improving current therapeutic avenues for atherosclerosis. We update the progress in progenitor cell study related to the development of arteriosclerosis, focusing specifically on the role of progenitor cells in lesion formation and discuss the controversial issues that regard the origins, frequency, and impact of the progenitors in the disease.
Collapse
Affiliation(s)
- Paola Campagnolo
- Cardiovascular Division, King's College London BHF Centre, London, England
| | | | | |
Collapse
|
111
|
Abstract
OBJECTIVES The concept of neovascularization in response to tissue ischemia was recently extended by the finding of postnatal vasculogenesis through circulating endothelial progenitor cells (EPCs). The aim of this study was to assess the role of acute ischemia for EPC mobilization in patients with peripheral arterial occlusive disease (PAOD) and in healthy volunteers. METHODS The number of circulating EPCs was analyzed by flow cytometry in PAOD patients (n = 23) with exercise-induced limb ischemia for up to 72 h after a maximal treadmill test and in healthy volunteers (n = 17) who underwent a 15-min suprasystolic occlusion of one lower extremity to induce limb ischemia. Plasma concentrations of vascular endothelial growth factor, basic fibroblast growth factor, tumor necrosis factor-α, and granulocyte macrophage-colony stimulating factor were determined by ELISA. RESULTS EPCs (CD 34 pos/KDRpos) increased significantly in both PAOD patients from 82 ± 20 to 256 ± 52 (P < 0.05) and healthy volunteers from 144 ± 39 to 590 ± 61 cells per 1 million events (P < 0.05) in response to induced ischemia, with a maximum after 24 h and returned to baseline within 72 h. The relative increase in EPC numbers was significantly lower in patients with PAOD as compared with healthy volunteers (P < 0.05). Plasma levels of vascular endothelial growth factor increased from 27.4 ± 3.1 to 126.4 ± 12 pg/ml in patients with PAOD (P < 0.05) and from 30.7 ± 6.1 to 134.1 ± 12.4 pg/ml in healthy volunteers (P < 0.05). CONCLUSION Both patients with symptomatic PAOD and healthy volunteers respond to a single episode of limb ischemia with a time-dependent increase in circulating EPCs. The increase of EPC numbers in response to ischemia is reduced when vascular disease is present, underlining the reduced vasculogenic potential of patients with PAOD.
Collapse
|
112
|
bFGF and PDGF-BB have a synergistic effect on the proliferation, migration and VEGF release of endothelial progenitor cells. Cell Biol Int 2011; 35:545-51. [PMID: 20961291 DOI: 10.1042/cbi20100401] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We have investigated the synergistic effects of bFGF (basic fibroblast growth factor) and PDGF-BB (platelet-derived growth factor-BB) on the proliferation, migration and VEGF (vascular endothelial growth factor) release of EPCs (endothelial progenitor cells). The proliferation of EPCs was assayed by MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium]. EPCs migration was detected using the Transwell system. Real-time PCR was used to assess the transcription of PDGFRβ mRNA. PLC-γ (phospholipase C gamma) expression and VEGF release were analysed by Western blot and ELISA. bFGF and PDGF-BB could, respectively, or synergistically, promote the proliferation and migration of EPCs, and these effects of bFGF and PDGF-BB were implemented by enhancing PDGFRβ mRNA, PLC-γ and VEGF expression, while inhibitor of PDGF receptor kinase (AG1296) and the selective PLC inhibitor (U73122) could block these effects of bFGF and PDGF-BB. In the meantime, we proved that the amplification by bFGF and PDGF-BB-stimulated PDGFRβ mRNA, PLC-γ and VEGF expression was abrogated by anti-bFGF antibody, AG1296 and U73122. These results strongly suggest that the proliferation and migration of EPCs may depend on bFGF and/or PDGF-BB by PDGFRβ/PLC-γ signalling pathway, and bFGF and/or PDGF-BB stimulate VEGF release at a point downstream from PDGFRβ/PLC-γ in EPCs.
Collapse
|
113
|
Wei HJ, Wang D, Chen JL, Xu Y, Lei P, Jiang RC, Liu L, Dong JF, Zhang JN. Mobilization of circulating endothelial progenitor cells after endovascular therapy for ruptured cerebral aneurysms. Neurosci Lett 2011; 498:114-8. [DOI: 10.1016/j.neulet.2011.04.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 04/19/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
|
114
|
Santini MP, Lexow J, Borsellino G, Slonimski E, Zarrinpashneh E, Poggioli T, Rosenthal N. IGF-1Ea induces vessel formation after injury and mediates bone marrow and heart cross-talk through the expression of specific cytokines. Biochem Biophys Res Commun 2011; 410:201-7. [PMID: 21621517 DOI: 10.1016/j.bbrc.2011.05.081] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 05/16/2011] [Indexed: 11/20/2022]
Abstract
The aim of this study was to investigate whether supplemental IGF-1Ea transgene expression induces activation of local cardiac and bone marrow stem cell population to mediate mammalian heart repair. In physiologic conditions, cardiac overexpression of the IGF-1Ea propeptide is associated with an enrichment of c-Kit/Sca-1 positive side population cells in the bone marrow and the occurrence of an endothelial-primed CD34 positive side population in the heart. This cellular profile is shown here to correlate with the expression of cytokines involved in stem cell mobilization and vessel formation. This molecular and cellular interplay favored IGF-1Ea-mediated vessel formation in injured hearts. The physiologic and pathologic connection between cytokines and stem cells in response to IGF-1Ea may represent an important model to understand how to elicit endogenous reparative signaling.
Collapse
Affiliation(s)
- Maria Paola Santini
- Heart Science Centre, National Heart and Lung Institute, Imperial College London, UK.
| | | | | | | | | | | | | |
Collapse
|
115
|
Bhatt RS, Zurita AJ, O'Neill A, Norden-Zfoni A, Zhang L, Wu HK, Wen PY, George D, Sukhatme VP, Atkins MB, Heymach JV. Increased mobilisation of circulating endothelial progenitors in von Hippel-Lindau disease and renal cell carcinoma. Br J Cancer 2011; 105:112-7. [PMID: 21673679 PMCID: PMC3137404 DOI: 10.1038/bjc.2011.186] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Circulating endothelial cells (CECs) are a candidate biomarker for monitoring angiogenesis in cancer. Circulating endothelial cell subsets are mobilised by angiogenic mediators. Because of the highly angiogenic phenotype of renal cell carcinoma (RCC), we sought to assess the potential of CECs as a marker of RCC in patients with von Hippel-Lindau (VHL) disease and those with sporadic RCC. Methods: We performed multicolour flow cytometry to enumerate CECs in patients with RCC, patients with VHL disease with and without RCC, and normal subjects. Two subsets of CECs were evaluated: mature CECs (mCECs) and circulating endothelial progenitors (CEPs). Results: In patients with VHL disease and RCC and those with sporadic RCC (N=10), CEPs and the CEP:mCEC ratio were higher than in normal subjects (N=17) (median CEPs: 0.97 vs 0.19 cells μl−1, respectively, P<0.01; median CEP:mCEC: 0.92 vs 0.58, respectively, P=0.04). However, in patients with VHL without RCC, CECs were not increased. In paired pre- and post-nephrectomy RCC patient samples (N=20), CEPs decreased after surgery (median difference 0.02 cells μl−1, −0.06 to 1.2; P=0.05). Conclusion: Circulating endothelial progenitors were elevated in RCC, but not in patients with VHL without RCC. Circulating endothelial progenitor enumeration merits further investigation as a monitoring strategy for patients with VHL.
Collapse
Affiliation(s)
- R S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, 375 Longwood Avenue, MASCO 426, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Xiao-Yun X, Zhao-Hui M, Ke C, Hong-Hui H, Yan-Hong X. Glucagon-like peptide-1 improves proliferation and differentiation of endothelial progenitor cells via upregulating VEGF generation. Med Sci Monit 2011; 17:BR35-41. [PMID: 21278683 PMCID: PMC3524715 DOI: 10.12659/msm.881383] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Glucagon-like peptide-1(GLP-1), released from enteroendocrine cells of the intestine, exerted cardiovascular protective effect. Circulating endothelial progenitor cells (EPCs) play an important role in maintaining endothelial integrity regulating neovascularization and reendothelialization after endothelial injury. Vascular endothelial growth factor (VEGF) is an important cytokine in the process of EPCs vascular differentiation and proliferation. Material/Methods This study was designed to investigate the association between VEGF changes and the proliferation/differentiation function of EPCs in the presence of GLP-1. Results We demonstrated that GLP-1 markedly enhanced the EPCs proliferation and expression of EC-specific markers, and simultaneously upregulated VEGF secretion in EPCs. Exogenous VEGF augmented EPCs proliferation/differentiation abilities in a dose-dependent manner. However, all of the beneficial effects of GLP-1 were suppressed by anti-VEGFmAb or the KDR-specific tyrosine kinase inhibitor SU1498. Conclusions These findings suggest that GLP-1 improves VEGF generation, which contributed to improvement of EPCs biological function, partly by tyrosine kinase KDR. VEGF is a necessary intermediate, mediating the effects of GLP-1 on EPCs. These changes offer a novel explanation that upregulation EPCs bioactivities may be one of the mechanisms of GLP-1 cardiovascular protective effect.
Collapse
Affiliation(s)
- Xie Xiao-Yun
- Department of Endocrinology, 3rd Xiangya Hospital, Central South University, Hunan Province, Changsha, China
| | | | | | | | | |
Collapse
|
117
|
Agudelo CA, Tachibana Y, Noboru T, Iida H, Yamaoka T. Long-term in vivo magnetic resonance imaging tracking of endothelial progenitor cells transplanted in rat ischemic limbs and their angiogenic potential. Tissue Eng Part A 2011; 17:2079-89. [PMID: 21466415 DOI: 10.1089/ten.tea.2010.0482] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Stem cell therapy has been used to repair ischemic tissues in the limbs, in myocardial infarctions, and in the brain. To understand the mechanisms of healing, a contrast agent capable of inducing sufficient magnetic resonance (MR) contrast would be useful in providing fundamental information about the cell migration and incorporation into the ischemic tissue. A magnetic resonance imaging contrast agent composed of dextran and gadolinium chelate was synthesized. Hydroxyl groups of dextran were activated with 1,1'-carbonylbis-1H-imidazole and reacted with propanediamine to obtain aminated dextran. This modified polymer was then reacted with mono-N-succinimidyl 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate, then with fluorescein isothiocyanate, and finally reacted with gadolinium chloride solution (Dex-DOTA-Gd3(+)). Endothelial progenitor cells (EPCs) were selected as a stem cell model for magnetic resonance imaging tracking. Cells were isolated from the bone marrow harvested from the femurs and tibias of rats. Dex-DOTA-Gd3(+) was then introduced into the EPCs by electroporation. The intracellular stability and cytotoxicity of Dex-DOTA-Gd3(+) were evaluated in vitro. Dex-DOTA-Gd3(+)-labeled EPCs were transplanted into a rat model of ischemic limb, and MR images were acquired. Dex-DOTA-Gd3(+) was found to efficiently label EPCs over a long duration without significant cytotoxicity. This provides an MR signal sufficient for tracking the EPCs intramuscularly injected into the limb.
Collapse
Affiliation(s)
- Carlos A Agudelo
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
118
|
Shah VK, Shalia KK. Stem Cell Therapy in Acute Myocardial Infarction: A Pot of Gold or Pandora's Box. Stem Cells Int 2011; 2011:536758. [PMID: 21804827 PMCID: PMC3142872 DOI: 10.4061/2011/536758] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/18/2010] [Accepted: 12/29/2010] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy for conditions characterized by myocyte loss in myocardial infarction and heart failure is intuitively appealing. Stem cells from various sources, including heart itself in preclinical and animal studies, have shown the potential to improve the function of ventricular muscle after ischaemic injury. The clinical experience from worldwide studies have indicated the safety profile but with modest benefits. The predominant mechanisms of transplanted cells for improving cardiac function have pointed towards paracrine effects rather than transdifferentiation into cardiomyocytes. Thus, further investigations should be encouraged towards bench side and bedside to resolve various issues for ensuring the correct type and dosing of cells, time, and method of delivery and identify correct mechanism of functional improvement. An interdisciplinary effort at the scientific, clinical, and the government front will bring successful realization of this therapy for healing the heart and may convert what seems now a Pandora's Box into a Pot of Gold.
Collapse
Affiliation(s)
- V K Shah
- Interventional Cardiologist, Sir H.N. Hospital and Research Centre, Raja Rammohan Roy Road, Mumbai 400 004, India
| | | |
Collapse
|
119
|
|
120
|
Thang OHD, Serné EH, Grooteman MPC, Smulders YM, ter Wee PM, Tangelder GJ, Nubé MJ. Capillary rarefaction in advanced chronic kidney disease is associated with high phosphorus and bicarbonate levels. Nephrol Dial Transplant 2011; 26:3529-36. [PMID: 21414968 DOI: 10.1093/ndt/gfr089] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In patients with chronic kidney disease (CKD), disorders of mineral metabolism are associated with vascular calcifications and mortality. Microvascular dysfunction, by affecting flow resistance and tissue perfusion, may explain the cardiovascular sequelae of CKD-associated disorders of mineral metabolism. We investigated whether advanced CKD is associated with a decrease in the functional and structural number of capillaries in skin and subsequently whether capillary rarefaction is related to mineral metabolism. METHODS Capillary density was measured by nailfold microscopy in 19 predialysis and 35 CKD Stage 5 (CKD5) patients and 19 controls. In CKD patients, calcium, phosphorus, parathyroid hormone, 25-hydroxyvitaminD3 (25vitD3) and 1,25-dihydroxyvitaminD3 (1,25vitD3) were analysed as well. RESULTS Capillary density at baseline was 42 ± 15/mm(2) in predialysis patients, 45 ± 17/mm(2) in CKD5 patients and 56 ± 20/mm(2) in controls (patients versus controls, respectively, P < 0.05 and P = 0.05). Absolute capillary recruitment during post-occlusive reactive hyperaemia was 17 ± 7/mm(2), 14 ± 6/mm(2) and 23 ± 8/mm(2), respectively (P < 0.05 for both patients and controls). Capillary density during venous occlusion was 59 ± 20/mm(2), 59 ± 21/mm(2) and 77 ± 21/mm(2), respectively (P < 0.05 for both patients and controls). In multiple regression analysis, both serum phosphorus and bicarbonate values were independently and inversely associated with capillary density at baseline (r(2) of model = 19%) as well as during venous occlusion (r(2) of model = 28%). Furthermore, both serum phosphorus and bicarbonate were inversely and female gender positively correlated with capillary density during recruitment (r(2) of model = 37%). CONCLUSION Advanced CKD is characterized by an impaired functional and structural capillary density in skin, which is related to both high phosphorus and bicarbonate values.
Collapse
Affiliation(s)
- Oanh H D Thang
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
121
|
Endothelial progenitor cells: novel biomarker and promising cell therapy for cardiovascular disease. Clin Sci (Lond) 2011; 120:263-83. [PMID: 21143202 DOI: 10.1042/cs20100429] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone-marrow-derived EPCs (endothelial progenitor cells) play an integral role in the regulation and protection of the endothelium, as well as new vessel formation. Peripheral circulating EPC number and function are robust biomarkers of vascular risk for a multitude of diseases, particularly CVD (cardiovascular disease). Importantly, using EPCs as a biomarker is independent of both traditional and non-traditional risk factors (e.g. hypertension, hypercholesterolaemia and C-reactive protein), with infused ex vivo-expanded EPCs showing potential for improved endothelial function and either reducing the risk of events or enhancing recovery from ischaemia. However, as the number of existing cardiovascular risk factors is variable between patients, simple EPC counts do not adequately describe vascular disease risk in all clinical conditions and, as such, the risk of CVD remains. It is likely that this limitation is attributable to variation in the definition of EPCs, as well as a difference in the interaction between EPCs and other cells involved in vascular control such as pericytes, smooth muscle cells and macrophages. For EPCs to be used regularly in clinical practice, agreement on definitions of EPC subtypes is needed, and recognition that function of EPCs (rather than number) may be a better marker of vascular risk in certain CVD risk states. The present review focuses on the identification of measures to improve individual risk stratification and, further, to potentially individualize patient care to address specific EPC functional abnormalities. Herein, we describe that future therapeutic use of EPCs will probably rely on a combination of strategies, including optimization of the function of adjunct cell types to prime tissues for the effect of EPCs.
Collapse
|
122
|
Lenk K, Uhlemann M, Schuler G, Adams V. Role of endothelial progenitor cells in the beneficial effects of physical exercise on atherosclerosis and coronary artery disease. J Appl Physiol (1985) 2011; 111:321-8. [PMID: 21350026 DOI: 10.1152/japplphysiol.01464.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In clinical trials as well as in several animal experiments it is evident that physical exercise is a powerful tool to positively influence the development and/or progression of atherosclerosis and coronary artery disease (CAD). The main target of physical exercise seems to be the maintenance of an intact endothelial cell layer. Since the discovery that endothelial progenitor cells (EPCs) are present in the circulation and the knowledge that exercise, either as a single exercise bout or an exercise training program, have the potency to mobilize EPCs from the bone marrow, the contribution of the EPCs for the preservation or repair of the endothelial cell layer is still under debate. Either the EPCs differentiate into mature endothelial cells, or they stimulate via a paracrine mechanism mature endothelial cells to proliferate. It is still unclear, if the exercise-induced mobilization of EPCs is casually related to the improvement of endothelial function. This review will discuss the role of endothelial progenitor cells in the beneficial effects of physical exercise on atherosclerosis and coronary artery disease.
Collapse
Affiliation(s)
- Karsten Lenk
- University Leipzig, Heart Center, Department of Internal Medicine/Cardiology, Leipzig, Germany
| | | | | | | |
Collapse
|
123
|
Li M, Ho JCY, Lai KWH, Au KKW, Xu A, Cheung BMY, Lam KSL, Tse HF. The decrement in circulating endothelial progenitor cells (EPCs) in type 2 diabetes is independent of the severity of the hypoadiponectemia. Diabetes Metab Res Rev 2011; 27:185-94. [PMID: 21294240 DOI: 10.1002/dmrr.1159] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (DM) is associated with a decreased level of circulating endothelial progenitor cells (EPCs) and adiponectin. Experimental studies suggest a potential link between hypoadiponectinaemia and the depletion of the EPC level. This study investigated the relationships between adiponectin level and EPC in patients with type 2 DM. METHODS A total of 95 type 2 DM patients (58.5 ± 8.8 years, 42 men) and 95 age- and sex-matched healthy controls were recruited. Circulating EPC levels were determined by flow cytometry using CD133(+), CD34(+), CD133(+) /KDR(+) and CD34(+) /KDR(+) as surface markers. Plasma adiponectin levels were measured by enzyme-linked immunosorbent assay. EPC function was studied by in vitro tube formation and migration assay. RESULTS The levels of CD133(+) (p < 0.001) and CD133(+) /KDR(+) (p < 0.001) EPCs were independently associated with the presence of type 2 DM. The levels of CD34(+) (p = 0.004) and CD34(+) /KDR(+) (p = 0.013) EPCs were independently associated with haemoglobin A(1c). Nevertheless, there was no relationship between the number of EPCs and adiponectin level. Tube formation assay showed impaired pro-angiogenic function of EPC in DM patients compared with controls (p = 0.007). Interestingly, adiponectin supplementation (5 µg/mL) increased tube formation by 17.6% in EPCs from DM patients (p = 0.002). It also significantly enhanced cell migration by 35.9% in EPCs from DM patients (p = 0.01). CONCLUSIONS We detected no relationship between the reduction in the level of EPC and in the level of total adiponectin in blood from patients with type 2 diabetes. EPC from patients with diabetes were stimulated when exposed to adiponectin in the test tube, findings that warrant further study.
Collapse
Affiliation(s)
- Mingfang Li
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Kränkel N, Spinetti G, Amadesi S, Madeddu P. Targeting stem cell niches and trafficking for cardiovascular therapy. Pharmacol Ther 2011; 129:62-81. [PMID: 20965213 PMCID: PMC3017934 DOI: 10.1016/j.pharmthera.2010.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/06/2010] [Indexed: 12/12/2022]
Abstract
Regenerative cardiovascular medicine is the frontline of 21st-century health care. Cell therapy trials using bone marrow progenitor cells documented that the approach is feasible, safe and potentially beneficial in patients with ischemic disease. However, cardiovascular prevention and rehabilitation strategies should aim to conserve the pristine healing capacity of a healthy organism as well as reactivate it under disease conditions. This requires an increased understanding of stem cell microenvironment and trafficking mechanisms. Engagement and disengagement of stem cells of the osteoblastic niche is a dynamic process, finely tuned to allow low amounts of cells move out of the bone marrow and into the circulation on a regular basis. The balance is altered under stress situations, like tissue injury or ischemia, leading to remarkably increased cell egression. Individual populations of circulating progenitor cells could give rise to mature tissue cells (e.g. endothelial cells or cardiomyocytes), while the majority may differentiate to leukocytes, affecting the environment of homing sites in a paracrine way, e.g. promoting endothelial survival, proliferation and function, as well as attenuating or enhancing inflammation. This review focuses on the dynamics of the stem cell niche in healthy and disease conditions and on therapeutic means to direct stem cell/progenitor cell mobilization and recruitment into improved tissue repair.
Collapse
Affiliation(s)
- Nicolle Kränkel
- Institute of Physiology/Cardiovascular Research, University of Zürich, and Cardiovascular Center, Cardiology, University Hospital Zurich, Zürich, Switzerland
| | | | - Silvia Amadesi
- Experimental Cardiovascular Medicine, University of Bristol, Bristol, UK
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
125
|
Jinturkar KA, Rathi MN, Misra A. Gene Delivery Using Physical Methods. CHALLENGES IN DELIVERY OF THERAPEUTIC GENOMICS AND PROTEOMICS 2011:83-126. [DOI: 10.1016/b978-0-12-384964-9.00003-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
126
|
Kishore R, Tkebuchava T, Sasi SP, Silver M, Gilbert HY, Yoon YS, Park HY, Thorne T, Losordo DW, Goukassian DA. Tumor necrosis factor-α signaling via TNFR1/p55 is deleterious whereas TNFR2/p75 signaling is protective in adult infarct myocardium. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:433-48. [PMID: 21153348 PMCID: PMC3076133 DOI: 10.1007/978-1-4419-6612-4_45] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Raj Kishore
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Tengiz Tkebuchava
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Sharath P. Sasi
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Marcy Silver
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Hu-Ya Gilbert
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Young-Sup Yoon
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Hee-Young Park
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Tina Thorne
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Douglas W. Losordo
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - David A. Goukassian
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
127
|
Capobianco S, Chennamaneni V, Mittal M, Zhang N, Zhang C. Endothelial progenitor cells as factors in neovascularization and endothelial repair. World J Cardiol 2010; 2:411-20. [PMID: 21191542 PMCID: PMC3011136 DOI: 10.4330/wjc.v2.i12.411] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/20/2010] [Accepted: 09/27/2010] [Indexed: 02/06/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous population of cells that are provided by the bone marrow and other adult tissue in both animals and humans. They express both hematopoietic and endothelial surface markers, which challenge the classic dogma that the presumed differentiation of cells into angioblasts and subsequent endothelial and vascular differentiation occurred exclusively in embryonic development. This breakthrough stimulated research to understand the mechanism(s) underlying their physiologic function to allow development of new therapeutic options. One focus has been on their ability to form new vessels in injured tissues, and another has been on their ability to repair endothelial damage and restore both monolayer integrity and endothelial function in denuded vessels. Moreover, measures of their density have been shown to be a better predictor of cardiovascular events, both in healthy and coronary artery disease populations than the classical tools used in the clinic to evaluate the risk stratification. In the present paper we review the effects of EPCs on revascularization and endothelial repair in animal models and human studies, in an attempt to better understand their function, which may lead to potential advancement in clinical management.
Collapse
Affiliation(s)
- Stefano Capobianco
- Stefano Capobianco, Department of Cardiology, Gaetano Rummo Hospital, Via Dell'Angelo 1, 82100 Benevento, Italy
| | | | | | | | | |
Collapse
|
128
|
Abstract
OBJECTIVE Sepsis and acute lung injury continue to be major causes of morbidity and mortality worldwide despite advances in our understanding of pathophysiology and the discovery of new management strategies. Recent investigations show that stem cells may be beneficial as prognostic biomarkers and novel therapeutic strategies in these syndromes. This article reviews the potential use of endogenous adult tissue-derived stem cells in sepsis and acute lung injury as prognostic markers and also as exogenous cell-based therapy. DATA SOURCES A directed systematic search of the medical literature using PubMed and OVID, with particular emphasis on the time period after 2002, was done to evaluate topics related to 1) the epidemiology and pathophysiology of sepsis and acute lung injury; and 2) the definition, characterization, and potential use of stem cells in these diseases. DATA SYNTHESIS AND FINDINGS: When available, preferential consideration was given to prospective nonrandomized clinical and preclinical studies. CONCLUSIONS Stem cells have shown significant promise in the field of critical care both for 1) prognostic value and 2) treatment strategies. Although several recent studies have identified the potential benefit of stem cells in sepsis and acute lung injury, further investigations are needed to more completely understand stem cells and their potential prognostic and therapeutic value.
Collapse
|
129
|
Wang DH, Li FR, Zhang Y, Wang YQ, Yuan FH. Conditioned medium from renal tubular epithelial cells stimulated by hypoxia influences rat bone marrow-derived endothelial progenitor cells. Ren Fail 2010; 32:863-70. [PMID: 20662701 DOI: 10.3109/0886022x.2010.494806] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND AIMS It has been well documented that endothelial progenitor cells (EPCs) participate in neovascularization in adults and that rarefaction of peritubular capillaries (PTCs) is closely associated with progressive kidney disease. Therefore, we investigated whether EPCs were influenced by conditioned medium (CM) of renal tubular epithelial cells (RTECs) stimulated by hypoxia, to provide evidence for EPCs transplantation in vivo in the future. METHODS Mononuclear cells of rat bone marrow were isolated by density gradient centrifugation and were cultured according to previously described techniques. RTECs were cultured primarily with routine tissue block adhering wall method. In addition, CM was harvested from RTECs cultivated for 48 h in 5% O(2). EPCs proliferation and migration were evaluated by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and transwell. The protein and mRNA expression of stromal cell-derived factor (SDF-1), vascular endothelial growth factor (VEGF), angiogenin 1 (Ang-1), and C-X-C chemokine receptor 4 (CXCR4) was separately assessed by Western blot, enzyme-linked immunosorbent assay (ELISA), and reverse transcriptase-polymerase chain reaction (RT-PCR) methods. RESULTS We showed that hypoxia increased the expression of SDF-1 and VEGF in RTEC. In addition, hypoxic CM improved proliferation, migration, and expression of VEGF, Ang-1, and CXCR4 of EPCs. CONCLUSIONS These results suggest that hypoxic CM improves neovascularization of EPCs and may also be considered as therapeutic agents to supply the potent origin of reconstituion of PTCs of progressive kidney disease.
Collapse
Affiliation(s)
- Dai-hong Wang
- Department of Nephrology, Xinqiao Hospital, Chongqing, PR China
| | | | | | | | | |
Collapse
|
130
|
Tokunaga M, Liu ML, Nagai T, Iwanaga K, Matsuura K, Takahashi T, Kanda M, Kondo N, Wang P, Naito AT, Komuro I. Implantation of cardiac progenitor cells using self-assembling peptide improves cardiac function after myocardial infarction. J Mol Cell Cardiol 2010; 49:972-83. [DOI: 10.1016/j.yjmcc.2010.09.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 08/20/2010] [Accepted: 09/15/2010] [Indexed: 12/15/2022]
|
131
|
Napoli C, Hayashi T, Cacciatore F, Casamassimi A, Casini C, Al-Omran M, Ignarro LJ. Endothelial progenitor cells as therapeutic agents in the microcirculation: an update. Atherosclerosis 2010; 215:9-22. [PMID: 21126740 DOI: 10.1016/j.atherosclerosis.2010.10.039] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 09/28/2010] [Accepted: 10/25/2010] [Indexed: 12/15/2022]
Abstract
This review evaluates novel beneficial effects of circulating endothelial progenitor cells (EPCs) as shown by several preclinical studies and clinical trials carried out to test the safety and feasibility of using EPCs. There are 31 registered clinical trials (and many others still ongoing) and 19 published studies. EPCs originate in the bone marrow and migrate into the bloodstream where they undergo a differentiation program leading to major changes in their antigenic characteristics. EPCs lose typical progenitor markers and acquire endothelial markers, and two important receptors, (VEGFR and CXCR-4), which recruit circulating EPCs to damaged or ischemic microcirculatory (homing to damaged tissues) beds. Overall, therapeutic angiogenesis will likely change the face of regenerative medicine in the next decade with many patients worldwide predicted to benefit from these treatments.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of General Pathology, Division of Clinical Pathology and Excellence Research Center on Cardiovascular Diseases, 1st School of Medicine, II University of Naples, 80138 Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
132
|
Pesce M, Burba I, Gambini E, Prandi F, Pompilio G, Capogrossi MC. Endothelial and cardiac progenitors: boosting, conditioning and (re)programming for cardiovascular repair. Pharmacol Ther 2010; 129:50-61. [PMID: 21035506 DOI: 10.1016/j.pharmthera.2010.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/06/2010] [Indexed: 12/26/2022]
Abstract
Preclinical studies performed in cell culture and animal systems have shown the outstanding ability of stem cells to repair ischemic heart and lower limbs by promoting the formation of new blood vessels and new myocytes. In contrast, clinical studies of stem cell administration in patients with myocardial ischemia have revealed only modest, although promising, results. Basic investigations have shown the feasibility of adult cells reprogramming into pluripotent cells by defined factors, thus opening the way to the devise of protocols to ex vivo derive virtually unexhausted cellular pools. In contrast, cellular and molecular studies have indicated that risk factors limit adult-derived stem cell survival, proliferation and engraftment in ischemic tissues. The use of fully reprogrammed cells raises safety concerns; therefore, adult cells remain a primary option for clinicians interested in therapeutic cardiovascular repair. Pharmacologic approaches have been devised to restore the cardiovascular repair ability of failing progenitors from patients at risk. In the present contribution, the most advanced pharmacologic approaches to (re)program, boost, and condition endothelial and cardiac progenitor cells to enhance cardiovascular regeneration are discussed.
Collapse
Affiliation(s)
- Maurizio Pesce
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
133
|
Beohar N, Rapp J, Pandya S, Losordo DW. Rebuilding the damaged heart: the potential of cytokines and growth factors in the treatment of ischemic heart disease. J Am Coll Cardiol 2010; 56:1287-97. [PMID: 20888519 PMCID: PMC3123891 DOI: 10.1016/j.jacc.2010.05.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 04/21/2010] [Accepted: 05/10/2010] [Indexed: 12/15/2022]
Abstract
Cytokine therapy promises to provide a noninvasive treatment option for ischemic heart disease. Cytokines are thought to influence angiogenesis directly via effects on endothelial cells or indirectly through progenitor cell-based mechanisms or by activating the expression of other angiogenic agents. Several cytokines mobilize progenitor cells from the bone marrow or are involved in the homing of mobilized cells to ischemic tissue. The recruited cells contribute to myocardial regeneration both as a structural component of the regenerating tissue and by secreting angiogenic or antiapoptotic factors, including cytokines. To date, randomized, controlled clinical trials have not reproduced the efficacy observed in pre-clinical and small-scale clinical investigations. Nevertheless, the list of promising cytokines continues to grow, and combinations of cytokines, with or without concurrent progenitor cell therapy, warrant further investigation.
Collapse
Affiliation(s)
- Nirat Beohar
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Cardiology, Northwestern University and Northwestern Memorial Hospital, Chicago, Illinois
| | - Jonathan Rapp
- Division of Cardiology, Northwestern University and Northwestern Memorial Hospital, Chicago, Illinois
- Department of Cardiology, Ochsner Clinic Foundation, New Orleans, Louisiana
| | - Sanjay Pandya
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Douglas W. Losordo
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Cardiology, Northwestern University and Northwestern Memorial Hospital, Chicago, Illinois
- Program in Cardiovascular Regenerative Medicine, Bluhm Cardiovascular Institute, Northwestern Memorial Hospital, Chicago, Illinois
| |
Collapse
|
134
|
Tilling L, Chowienczyk P, Clapp B. Progenitors in motion: mechanisms of mobilization of endothelial progenitor cells. Br J Clin Pharmacol 2010; 68:484-92. [PMID: 19843051 DOI: 10.1111/j.1365-2125.2009.03486.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Endothelial progenitor cells are a population of bone marrow-derived mononuclear cells thought to engage in endothelial repair and hence are considered potential therapeutic agents in many pathological conditions. The mechanism of their exit from the bone marrow to the circulation and damaged tissues, termed mobilization, has not been fully elucidated. Despite this, several pharmacological interventions have been shown to influence mobilization of these specialized cells. Here we review the current understanding of their mobilization.
Collapse
|
135
|
Padfield GJ, Tura O, Haeck MLA, Short A, Freyer E, Barclay GR, Newby DE, Mills NL. Circulating endothelial progenitor cells are not affected by acute systemic inflammation. Am J Physiol Heart Circ Physiol 2010; 298:H2054-61. [PMID: 20382859 PMCID: PMC2886634 DOI: 10.1152/ajpheart.00921.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 04/01/2010] [Indexed: 01/27/2023]
Abstract
Vascular injury causes acute systemic inflammation and mobilizes endothelial progenitor cells (EPCs) and endothelial cell (EC) colony-forming units (EC-CFUs). Whether such mobilization occurs as part of a nonspecific acute phase response or is a phenomenon specific to vascular injury remains unclear. We aimed to determine the effect of acute systemic inflammation on EPCs and EC-CFU mobilization in the absence of vascular injury. Salmonella typhus vaccination was used as a model of acute systemic inflammation. In a double-blind randomized crossover study, 12 healthy volunteers received S. typhus vaccination or placebo. Phenotypic EPC populations enumerated by flow cytometry [CD34(+)VEGF receptor (VEGF)R-2(+)CD133(+), CD14(+)VEGFR-2(+)Tie2(+), CD45(-)CD34(+), as a surrogate for late outgrowth EPCs, and CD34(+)CXCR-4(+)], EC-CFUs, and serum cytokine concentrations (high sensitivity C-reactive protein, IL-6, and stromal-derived factor-1) were quantified during the first 7 days. Vaccination increased circulating leukocyte (9.8 + or - 0.6 vs. 5.1 + or - 0.2 x 10(9) cells/l, P < 0.0001), serum IL-6 [0.95 (0-1.7) vs. 0 (0-0) ng/l, P = 0.016], and VEGF-A [60 (45-94) vs. 43 (21-64) pg/l, P = 0.006] concentrations at 6 h and serum high sensitivity C-reactive protein at 24 h [2.7 (1.4-3.6) vs. 0.4 (0.2-0.8) mg/l, P = 0.037]. Vaccination caused a 56.7 + or - 7.6% increase in CD14(+) cells at 6 h (P < 0.001) and a 22.4 + or - 6.9% increase in CD34(+) cells at 7 days (P = 0.04). EC-CFUs, putative vascular progenitors, and the serum stromal-derived factor-1 concentration were unaffected throughout the study period (P > 0.05 for all). In conclusion, acute systemic inflammation causes nonspecific mobilization of hematopoietic progenitor cells, although it does not selectively mobilize putative vascular progenitors. We suggest that systemic inflammation is not the primary stimulus for EPC mobilization after acute vascular injury.
Collapse
Affiliation(s)
- Gareth J Padfield
- Centre for Cardiovascular Science, Univ. of Edinburgh, Chancellor's Bldg., Edinburgh EH16 4SU, UK.
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Zubair AC, Malik S, Paulsen A, Ishikawa M, McCoy C, Adams PX, Amrani D, Costa M. Evaluation of mobilized peripheral blood CD34(+) cells from patients with severe coronary artery disease as a source of endothelial progenitor cells. Cytotherapy 2010; 12:178-89. [PMID: 20078384 DOI: 10.3109/14653240903493409] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS The distinction between hematopoietic stem cells (HSC) and endothelial progenitor cells (EPC) is poorly defined. Co-expression of CD34 antigen with vascular endothelial growth factor (VEGF) receptor (VEGFR2) is currently used to define EPC ( 1 ). METHODS We evaluated the phenotypic and genomic characteristics of peripheral blood-derived CD34(+) cells in 22 granulocyte-colony-stimulating factor (G-CSF)-mobilized patients with severe coronary artery disease and assessed the influence of cell selection and storage on CD34(+) cell characteristics. RESULTS The median CD34(+) cell contents in the products before and after enrichment with the Isolex 300i Magnetic Cell Selection System were 0.2% and 82.5%, respectively. Cell-cycle analysis showed that 80% of CD34(+) cells were in G0 stage; 70% of the isolated CD34(+) cells co-expressed CD133, a marker for more immature progenitors. However, less than 5% of the isolated CD34(+) cells co-expressed the notch receptor Jagged-1 (CD339) and only 2% of the isolated CD34(+) population were positive for VEGFR2 (CD309). Molecular assessment of the isolated CD34(+) cells demonstrated extremely low expression of VEGFR2 and endothelial nitric oxide synthase (eNOS) and high expression of VEGF-A. Overnight storage at 4 degrees C did not significantly affect CD34(+) cell counts and viability. Storage in liquid nitrogen for 7 weeks did not affect the percentage of CD34(+) cells but was associated with a 26% drop in cell viability. CONCLUSIONS We have demonstrated that the majority of isolated CD34(+) cells consist of immature and quiescent cells that lack prototypic markers of EPC. High VEGF-A gene expression might be one of the mechanisms for CD34(+) cell-induced angiogenesis.
Collapse
Affiliation(s)
- Abba C Zubair
- Transfusion Medicine, Department of Pathology, Mayo Clinic, Jacksonville, Florida 32224, USA.
| | | | | | | | | | | | | | | |
Collapse
|
137
|
Everaert BR, Van Craenenbroeck EM, Hoymans VY, Haine SE, Van Nassauw L, Conraads VM, Timmermans JP, Vrints CJ. Current perspective of pathophysiological and interventional effects on endothelial progenitor cell biology: focus on PI3K/AKT/eNOS pathway. Int J Cardiol 2010; 144:350-66. [PMID: 20444511 DOI: 10.1016/j.ijcard.2010.04.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/04/2010] [Indexed: 12/24/2022]
Abstract
For more than a decade, endothelial progenitor cells (EPCs) have been implicated in cardiovascular homeostasis. EPCs are believed to reside within the bone marrow in close contact with surrounding stromal cells, and, under stimulation of pro-inflammatory cytokines, EPCs are mobilized out of the bone marrow. Hereafter circulating EPCs home to peripheral tissues, undergoing further proliferation and differentiation. Under certain pathophysiologic conditions this process seems to be blunted, resulting in a reduced capacity of EPCs to engage in vasculogenesis at sites of endothelial injury or tissue ischemia. In this review, we focus on the effects of traditional cardiovascular risk factors on EPC biology and we explore whether pharmacological, dietary and lifestyle interventions can favorably restore EPC mobilization, differentiation, homing and angiogenic properties. Because the PI3K/Akt/eNOS pathway plays a pivotal role in the process of EPC mobilization, migration and homing, we specifically emphasize the involvement of PI3K, Akt and eNOS in EPC biology under these different (patho)physiologic conditions. (Pre)clinically used drugs or lifestyle interventions that have been shown to ameliorate EPC biology are reviewed. These treatment strategies remain attractive targets to restore the regenerative capacity of EPCs in cardiovascular diseases.
Collapse
Affiliation(s)
- Bert R Everaert
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Young PP, Ardestani S, Li B. Myeloid cells in cancer progression: unique subtypes and their roles in tumor growth, vascularity, and host immune suppression. CANCER MICROENVIRONMENT 2010; 4:1-11. [PMID: 21505557 PMCID: PMC3047625 DOI: 10.1007/s12307-010-0045-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 03/23/2010] [Indexed: 12/22/2022]
Abstract
Leukocytic infiltrates, particularly myeloid cells, can stimulate an anti-tumor immune response, but more often they stimulate tumor development, including promoting invasion, tumor growth, angiogenesis, and metastasis. Distinct myeloid phenotypes are being characterized that have been shown to promote tumor growth, angiogenesis, and metastasis. This review provides an overview of myeloid differentiation and spotlights specific pro-tumorogenic myeloid populations and their role in cancer progression. Efforts to characterize these pro-tumorogenic myeloid cell immunophenotypes may lead to novel targets for cancer therapy.
Collapse
|
139
|
Choi YH, Neef K, Reher M, Liakopoulos OJ, Zeriouh M, Wittwer T, Stamm C, Madershahian N, Teschendorf P, Wahlers T. The influence of pre-operative risk on the number of circulating endothelial progenitor cells during cardiopulmonary bypass. Cytotherapy 2010; 12:79-87. [PMID: 19929452 DOI: 10.3109/14653240903377029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS The number of circulating endothelial progenitor cells (EPC) depends on cytokine release and is also associated with cardiovascular risk factors. During cardiopulmonary bypass (CPB) the endothelium is the first organ to be affected by mechanical and immunologic stimuli. We hypothesized that the magnitude of EPC mobilization by CPB correlates with the pre-operative cardiovascular morbidity profile. METHODS EPC were quantified in blood samples from 30 patients who underwent cardiac surgery by magnetic bead isolation and fluorescence-activated cell sorting (FACS) analysis, based on concomitant expression of CD34, CD133 and CD309. Patients were divided into two groups based on the European System for Cardiac Operative Risk Evaluation (EuroSCORE): low risk (LR) and high risk (HR). Ten healthy volunteers served as controls. Samples were obtained before the start of CPB and at 1 and 24 h post-operatively. Plasma samples were collected for determination of release levels of cytokines and growth factors. RESULTS All CPB patients showed a significantly reduced basal number of EPC compared with healthy individuals (LR 5.60 +/- 0.39/mL, HR 3.89 +/- 0.34/ mL, versus control 0.807 +/- 0.82/mL, P = 0.012 versus LR, P< 0.001 versus HR). CPB induced EPC release that peaked 1 h after surgery (pre-operative 4.79 +/- 0.32/mL, 1 h 57.49 +/- 5.31/mL, 24 h 6.67 +/- 1.05/mL, P< 0.001 pre-operative versus 1 h, P< 0.001 pre-operative versus 24 h) and was associated with the duration of CPB. However, EPC release was significantly attenuated in HR patients (33.09 +/- 3.58/mL versus 81.89 +/- 4.36/mL at 1 h after CPB, P < 0.0001) and inversely correlated with the pre-operative EuroSCORE. Serum granulocyte-colony-stimulating factor (G-CSF), stem cell factor (SCF) and vascular endothelial growth factor (VEGF) levels increased throughout the observation period and were also correlated with the EPC count. CONCLUSIONS Cardiovascular risk factors influence the mobilization of EPC from the bone marrow after stimulation by CPB. This could be secondary to impaired mobilization or the result of increased EPC turnover, and may have implications for future cell therapy strategies in cardiac surgical patients.
Collapse
Affiliation(s)
- Yeong-Hoon Choi
- Heart Center of the University of Cologne, Department of Cardiothoracic Surgery, Cologne, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Asosingh K, Hanson JD, Cheng G, Aronica MA, Erzurum SC. Allergen-induced, eotaxin-rich, proangiogenic bone marrow progenitors: a blood-borne cellular envoy for lung eosinophilia. J Allergy Clin Immunol 2010; 125:918-25. [PMID: 20227754 PMCID: PMC2850950 DOI: 10.1016/j.jaci.2010.01.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 12/15/2009] [Accepted: 01/07/2010] [Indexed: 01/21/2023]
Abstract
BACKGROUND Eosinophilic inflammation is closely related to angiogenesis in asthmatic airway remodeling. In ovalbumin (OVA)-sensitized mice bone marrow-derived, proangiogenic endothelial progenitor cells (EPCs) are rapidly recruited into the lungs after OVA aerosol challenge and promptly followed by mobilization and recruitment of eosinophils. OBJECTIVE We hypothesized that bone marrow-derived EPCs initiate the recruitment of eosinophils through expression of the eosinophil chemoattractant eotaxin-1. METHODS EPCs were isolated from an OVA murine model of allergic airway inflammation and from asthmatic patients. Endothelial and smooth muscle cells were isolated from mice. Eotaxin-1 expression was analyzed by means of immunofluorescence, real-time PCR, or ELISA. In vivo recruitment of eosinophils by EPCs was analyzed in mice. RESULTS Circulating EPCs of asthmatic patients had higher levels of eotaxin-1 compared with those seen in control subjects. In the murine model OVA allergen exposure augmented eotaxin-1 mRNA and protein levels in EPCs. The EPCs from OVA-sensitized and OVA-challenged mice released high levels of eotaxin-1 on contact with lung endothelial cells from sensitized and challenged mice but not from control animals and not on contact with cardiac or hepatic endothelial cells from sensitized and challenged mice. Intranasal administration of the eotaxin-rich media overlying cultures of EPCs caused recruitment into the lungs, confirming functional chemoattractant activity. CONCLUSIONS Bone marrow-derived EPCs are early responders to environmental allergen exposures and initiate a parallel switch to a proangiogenic and proeosinophilic environment in the lungs of asthmatic patients.
Collapse
Affiliation(s)
- Kewal Asosingh
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA. and
| | | | | | | | | |
Collapse
|
141
|
Abebe W, Mozaffari M. Endothelial dysfunction in diabetes: potential application of circulating markers as advanced diagnostic and prognostic tools. EPMA J 2010. [PMID: 23199039 PMCID: PMC3405304 DOI: 10.1007/s13167-010-0012-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Endothelial dysfunction is a predisposing factor for vascular disease in diabetes, which contributes significantly to the mortality of diabetic patients. The currently utilized assessment methods of endothelial function/dysfunction in humans are associated with various limitations. Circulating endothelial-derived/associated markers have been proposed as potential alternatives for evaluation of the endothelium in condition of vascular disorders. These indicators include von Willebrand factor, soluble thrombomodulin, soluble E-selectin, asymmetric dimethylarginine, tissue plasminogen activator, endothelial microparticles, circulating endothelial cells and circulating endothelial progenitor cells. While tentative evidence is available for most of these biomarkers to serve as reliable sources of information, their usefulness for routine clinical applications has not yet been established. Thus, circulating endothelial markers are currently the subject of intense research interest and it is anticipated that as more information becomes available their improved quantification will provide a suitable diagnostic and prognostic tool for vascular events in diabetes and related diseases.
Collapse
Affiliation(s)
- Worku Abebe
- Department of Oral Biology, CL 2140, School of Dentistry, Medical College of Georgia, Augusta, GA 30912-1128 USA
| | | |
Collapse
|
142
|
Li Calzi S, Neu MB, Shaw LC, Grant MB. Endothelial progenitor dysfunction in the pathogenesis of diabetic retinopathy: treatment concept to correct diabetes-associated deficits. EPMA J 2010; 1:88-100. [PMID: 21494317 PMCID: PMC3008583 DOI: 10.1007/s13167-010-0011-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 02/10/2010] [Indexed: 11/29/2022]
Abstract
Progressive obliteration of the retinal microvessels is a characteristic of diabetic retinopathy and the resultant retinal ischemia can lead to sight-threatening macular edema, macular ischemia and ultimately preretinal neovascularization. Bone marrow derived endothelial progenitor cells (EPCs) play a critical role in vascular maintenance and repair. There is still great debate about the most appropriate markers that define an EPC. EPCs can be isolated using cell sorting by surface phenotype selection or in vitro cell culture. For freshly isolated cells, EPC cell sorting is heavily dependent on the surface markers used; EPCs can also be isolated by in vitro propagation of heterogeneous mixtures of cells in culture using adhesion to specific substrates and cell growth characteristics. in vitro isolation enables consistent reproducibility and using this approach at least two distinct types of EPCs with different angiogenic properties have been identified from adult peripheral and umbilical cord blood; early EPCs (eEPCs) and late outgrowth endothelial progenitor cells (OECs). Emerging studies demonstrate the potential of these cells in revascularization of ischemic/injured retina in animal models of retinal disease. Since ischemic retinopathies are leading causes of blindness, they are a potential disease target for EPC-based therapy. In this chapter, we summarize the current knowledge about EPCs and discuss the possibility of cellular therapy for treatment of diabetic macular ischemia and the vasodegenerative phase of diabetic retinopathy. We also report current pharmacological options that can be utilized to correct diabetes associated defects in EPCs so as to enhance the therapeutic utility of these cells.
Collapse
Affiliation(s)
- Sergio Li Calzi
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Matthew B. Neu
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Lynn C. Shaw
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Maria B. Grant
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Box 100267, ARB, 1600 SW Archer Road, Gainesville, FL 32610 USA
| |
Collapse
|
143
|
Probst-Cousin S, Neundörfer B, Heuss D. Microvasculopathic neuromuscular diseases: Lessons from hypoxia-inducible factors. Neuromuscul Disord 2010; 20:192-7. [DOI: 10.1016/j.nmd.2010.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 12/21/2009] [Accepted: 01/07/2010] [Indexed: 10/19/2022]
|
144
|
Distler JHW, Beyer C, Schett G, Lüscher TF, Gay S, Distler O. Endothelial progenitor cells: novel players in the pathogenesis of rheumatic diseases. ACTA ACUST UNITED AC 2010; 60:3168-79. [PMID: 19877034 DOI: 10.1002/art.24921] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jörg H W Distler
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | |
Collapse
|
145
|
Takayama M, Kuramoto Y, Okuyama R, Yamasaki K, Aiba S. The Exudate of Pressure Ulcers Contains a Substantial Amount of Vascular Endothelial Growth Factor. TOHOKU J EXP MED 2010; 221:315-9. [DOI: 10.1620/tjem.221.315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Mariko Takayama
- Department of Dermatology, Tohoku University Graduate School of Medicine
| | | | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine
| | - Kenshi Yamasaki
- Department of Dermatology, Tohoku University Graduate School of Medicine
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine
| |
Collapse
|
146
|
Bhatwadekar AD, Shaw LC, Grant MB. Promise of endothelial progenitor cell for treatment of diabetic retinopathy. Expert Rev Endocrinol Metab 2010; 5:29-37. [PMID: 23678364 PMCID: PMC3652409 DOI: 10.1586/eem.09.75] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progressive obliteration of the retinal microvessels is a characteristic of diabetic retinopathy. The resultant retinal ischemia leads to sight-threatening neovascularization and macular edema. Bone marrow-derived endothelial progenitor cells play a critical role in vascular maintenance and repair and forms the basis of cellular therapy for revascularization of ischemic myocardium and ischemic limbs. Emerging studies show potential of these cells in revascularization of ischemic retina and this review summarizes this possibility. We also report current pharmacological options to correct diabetes-associated defects in endothelial progenitor cells for their therapeutic transfer.
Collapse
Affiliation(s)
- Ashay D Bhatwadekar
- Dept of Pharmacology and Therapeutics, University of Florida, 1600 SW Archer Road Gainesville, FL 32610-0267, USA Tel.: + 1 352 392 9006 Fax: + 1 352 392 9696
| | - Lynn C Shaw
- Dept of Pharmacology and Therapeutics, University of Florida, 1600 SW Archer Road Gainesville, FL 32610-0267, USA Tel.: + 1 352 392 8020 Fax: + 1 352 392 9696
| | - Maria B Grant
- Author for correspondence Department of Pharmacology and Therapeutics, University of Florida, 1600 SW Archer Road Gainesville, FL 32610-0267, USA Tel: + 1 352 846 0978 Fax: + 1 352 392 9696
| |
Collapse
|
147
|
Nesselmann C, Li W, Ma N, Steinhoff G. Stem cell-mediated neovascularization in heart repair. Ther Adv Cardiovasc Dis 2009; 4:27-42. [PMID: 20042449 DOI: 10.1177/1753944709353338] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Accumulating clinical and experimental evidence indicates that stem cells from various sources are promising in the treatment of cardiac dysfunction. They may be incorporated into neovascular foci and thus contribute to postnatal physiological and pathological vasculogenesis and/or produce a variety of growth factors for angiogenesis and cytokines that home other stem cells from other organs for cardiac regeneration. This review focuses on the neovascularization of stem cells from different sources in cardiac repair, with emphasis on adult stem cells.
Collapse
|
148
|
High serum levels of growth factors are associated with good outcome in intracerebral hemorrhage. J Cereb Blood Flow Metab 2009; 29:1968-74. [PMID: 19756022 DOI: 10.1038/jcbfm.2009.182] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In experimental models, growth factors (GFs) such as vascular endothelial growth factor (VEGF), Angiopoietin 1 (Ang-1), or granulocyte-colony stimulating factor (G-CSF) mediate brain recovery after intracerebral hemorrhage (ICH). Our aim was to study the association between serum levels of GF and clinical outcome in patients with ICH. A total of 95 patients with primary ICH (male, 66.3%; mean age, 67.8+/-9.8 years) were prospectively included in the study within 12 h from symptoms onset. The main outcome variable was good functional outcome at 3 months (modified Rankin scale < or =2). Median serum levels of GF at 72 h from stroke onset were significantly higher in patients with good outcome (n=39) compared with those with poor outcome (all P<0.0001). Serum levels of VEGF > or =330 pg/mL, G-CSF > or =413 pg/mL, and Ang-1 > or =35 ng/mL at 72 h were independently associated with good functional outcome (odds ratio (OR), 11.2; 95% confidence interval (CI): 2.9 to 43.0; OR, 19.6; 95% CI: 3.9 to 97.9; and OR, 14.7; 95% CI: 3.6 to 60.0, respectively), neurologic improvement (all P<0.0001) and reduced residual cavity at 3 months (all P<0.01). These results illustrate that high serum levels of GF are associated with good functional outcome and reduced lesion volume in ICH.
Collapse
|
149
|
Abstract
Acute ischemic injury and chronic cardiomyopathies can cause irreversible loss of cardiac tissue leading to heart failure. Cellular therapy offers a new paradigm for treatment of heart disease. Stem cell therapies in animal models show that transplantation of various cell preparations improves ventricular function after injury. The first clinical trials in patients produced some encouraging results, despite limited evidence for the long-term survival of transplanted cells. Ongoing research at the bench and the bedside aims to compare sources of donor cells, test methods of cell delivery, improve myocardial homing, bolster cell survival, and promote cardiomyocyte differentiation. This article reviews progress toward these goals.
Collapse
Affiliation(s)
- John A. Schoenhard
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University, MRB IV P425C, 2213 Garland Avenue, Nashville, TN 37232 USA
- Department of Cell and Developmental Biology, Vanderbilt University, MRB IV P425C, 2213 Garland Avenue, Nashville, TN 37232 USA
| | - Antonis K. Hatzopoulos
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University, MRB IV P425C, 2213 Garland Avenue, Nashville, TN 37232 USA
- Department of Cell and Developmental Biology, Vanderbilt University, MRB IV P425C, 2213 Garland Avenue, Nashville, TN 37232 USA
| |
Collapse
|
150
|
Grimaldi A, Banfi S, Gerosa L, Tettamanti G, Noonan DM, Valvassori R, de Eguileor M. Identification, isolation and expansion of myoendothelial cells involved in leech muscle regeneration. PLoS One 2009; 4:e7652. [PMID: 19876402 PMCID: PMC2765724 DOI: 10.1371/journal.pone.0007652] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 10/07/2009] [Indexed: 01/23/2023] Open
Abstract
Adult skeletal muscle in vertebrates contains myoendothelial cells that express both myogenic and endothelial markers, and which are able to differentiate into myogenic cells to contribute to muscle regeneration. In spite of intensive research efforts, numerous questions remain regarding the role of cytokine signalling on myoendothelial cell differentiation and muscle regeneration. Here we used Hirudo medicinalis (Annelid, leech) as an emerging new model to study myoendothelial cells and muscle regeneration. Although the leech has relative anatomical simplicity, it shows a striking similarity with vertebrate responses and is a reliable model for studying a variety of basic events, such as tissue repair. Double immunohistochemical analysis were used to characterize myoendothelial cells in leeches and, by injecting in vivo the matrigel biopolymer supplemented with the cytokine Vascular Endothelial Growth Factor (VEGF), we were able to isolate this specific cell population expressing myogenic and endothelial markers. We then evaluated the effect of VEGF on these cells in vitro. Our data indicate that, similar to that proposed for vertebrates, myoendothelial cells of the leech directly participate in myogenesis both in vivo and in vitro, and that VEGF secretion is involved in the recruitment and expansion of these muscle progenitor cells.
Collapse
Affiliation(s)
- Annalisa Grimaldi
- Department of Biotechnology and Molecular Sciences, University of Insubria, Varese, Italy.
| | | | | | | | | | | | | |
Collapse
|