301
|
Lutz AH, Blumenthal JB, Landers-Ramos RQ, Prior SJ. Exercise-induced endothelial progenitor cell mobilization is attenuated in impaired glucose tolerance and type 2 diabetes. J Appl Physiol (1985) 2016; 121:36-41. [PMID: 27197857 DOI: 10.1152/japplphysiol.00349.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022] Open
Abstract
Circulating endothelial progenitor cells (EPCs) contribute to vascular homeostasis and are fewer in those with type 2 diabetes mellitus (T2DM) compared with normal glucose tolerance (NGT), suggesting a link between EPCs and T2DM-associated vasculopathies. The purpose of this study was to assess EPC number and mobilization by acute submaximal exercise in older adults with NGT, impaired glucose tolerance (IGT) or T2DM. We tested the hypothesis that EPC mobilization is lower in IGT compared with NGT and further reduced in older adults with T2DM. Forty-five older (50-75 yr of age) men and women with NGT (n = 18), IGT (n = 10), or T2DM (n = 17) were characterized and underwent submaximal aerobic exercise tests with blood sampling for enumeration of vascular endothelial growth factor receptor 2+ (VEGFR2+) cells, CD34+ hematopoetic progenitor cells, and CD34+/VEGFR2+ EPCs by flow cytometry before and after exercise. Basal EPC number was 65 and 61% lower in the IGT and T2DM groups, respectively, compared with the NGT group (P < 0.05). EPC number increased 23% after acute exercise in the NGT group (P < 0.01), but did not change in the IGT or T2DM groups. Before and after exercise, VEGFR2+ cell number was lower in a stepwise manner across the NGT, IGT, and T2DM groups (P < 0.05). Basal CD34+ cell number was lower in the IGT group compared with NGT (P < 0.05), but did not change after exercise in any group. These findings suggest a CD34+/VEGFR2+ EPC mobilization defect in IGT and T2DM that could play a role in the cardiovascular diseases and capillary rarefaction associated with insulin resistance.
Collapse
Affiliation(s)
- Andrew H Lutz
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - Jacob B Blumenthal
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - Rian Q Landers-Ramos
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - Steven J Prior
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| |
Collapse
|
302
|
Patschan D, Kribben A, Müller GA. Postischemic microvasculopathy and endothelial progenitor cell-based therapy in ischemic AKI: update and perspectives. Am J Physiol Renal Physiol 2016; 311:F382-94. [PMID: 27194716 DOI: 10.1152/ajprenal.00232.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/15/2016] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) dramatically increases mortality of hospitalized patients. Incidences have been increased in recent years. The most frequent cause is transient renal hypoperfusion or ischemia which induces significant tubular cell dysfunction/damage. In addition, two further events take place: interstitial inflammation and microvasculopathy (MV). The latter evolves within minutes to hours postischemia and may result in permanent deterioration of the peritubular capillary network, ultimately increasing the risk for chronic kidney disease (CKD) in the long term. In recent years, our understanding of the molecular/cellular processes responsible for acute and sustained microvasculopathy has increasingly been expanded. The methodical approaches for visualizing impaired peritubular blood flow and increased vascular permeability have been optimized, even allowing the depiction of tissue abnormalities in a three-dimensional manner. In addition, endothelial dysfunction, a hallmark of MV, has increasingly been recognized as an inductor of both vascular malfunction and interstitial inflammation. In this regard, so-called regulated necrosis of the endothelium could potentially play a role in postischemic inflammation. Endothelial progenitor cells (EPCs), represented by at least two major subpopulations, have been shown to promote vascular repair in experimental AKI, not only in the short but also in the long term. The discussion about the true biology of the cells continues. It has been proposed that early EPCs are most likely myelomonocytic in nature, and thus they may simply be termed proangiogenic cells (PACs). Nevertheless, they reliably protect certain types of tissues/organs from ischemia-induced damage, mostly by modulating the perivascular microenvironment in an indirect manner. The aim of the present review is to summarize the current knowledge on postischemic MV and EPC-mediated renal repair.
Collapse
Affiliation(s)
- D Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Georg-August-University, Göttingen, Germany; and
| | - A Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - G A Müller
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Georg-August-University, Göttingen, Germany; and
| |
Collapse
|
303
|
Deguelin inhibits vasculogenic function of endothelial progenitor cells in tumor progression and metastasis via suppression of focal adhesion. Oncotarget 2016; 6:16588-600. [PMID: 26078334 PMCID: PMC4599291 DOI: 10.18632/oncotarget.3752] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022] Open
Abstract
Deguelin is a nature-derived chemopreventive drug. Endothelial progenitor cells (EPCs) are bone-marrow (BM)-derived key components to induce new blood vessels in early tumorigenesis and metastasis. Here we determined whether deguelin inhibits EPC function in vitro and in vivo at doses not affecting cancer cell apoptosis. Deguelin significantly reduced the number of EPC colony forming units of BM-derived c-kit+/sca-1+ mononuclear cells (MNCs), proliferation, migration, and adhesion to endothelial cell monolayers, and suppressed incorporation of EPC into tube-like vessel networks when co-cultured with endothelial cells. Deguelin caused cell cycle arrest at G1 without induction of apoptosis in EPC. In a mouse tumor xenograft model, tumor growth, lung metastasis and tumor-induced circulating EPCs were supressed by deguelin treatment (2 mg/kg). In mice tranplanted with GFP-expressing BM-MNCs, deguelin reduced the co-localization of CD31 and GFP, suggesting suppression of BM-derived EPC incoporation into tumor vessels. Interestingly, focal adhesion kinase (FAK)-integrin-linked kinase (ILK) activation and actin polymerization were repressed by deguelin. Decreased number of focal adhesions and a depolarized morphology was found in deguelin-treated EPCs. Taken together, our results suggest that the deguelin inhibits tumorigenesis and metastasis via EPC suppression and that suppression of focal adhesion by FAK-integrin-ILK-dependent actin remodeling is a key underlying molecular mechanism.
Collapse
|
304
|
Roan JN, Luo CY, Tsai MD, Wu IS, Chang SW, Huang CC, Tsai YS, Lam CF. Mobilization of Endothelial Progenitor Cells Following Creation of Arteriovenous Access in Patients with End-Stage Renal Disease. ACTA CARDIOLOGICA SINICA 2016; 31:24-32. [PMID: 27122843 DOI: 10.6515/acs20140310e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND A patent arteriovenous (AV) fistula induces activation of regional vascular endothelium and vascular shear force. Shear stress is an important physiological force in mobilizing endothelial progenitor cells (EPCs). This study aimed to explore the perioperative changes of circulating EPC levels for patients who require hemodialysis and underwent radiocephalic fistula operation. METHODS This prospective cohort study included patients who received a radiocephalic fistula surgery when they were between 25 and 65 years of age. The subjects were followed for 90 days postoperatively for any stenotic events or immaturity of the fistula. Blood samples were obtained on the day before surgery and at postoperation day (POD) 3 and 30. CD133+/KDR+ cells, defined as EPCs, were analyzed using flow cytometry. Blood flow of the fistula was followed on POD 3 and 30. RESULTS A total of 30 patients were enrolled in the study from July 2009 to December 2011. One patient dropped out of the study and seven patients developed a stenotic (or immature) AV fistula (7/29, 24.1%). There were positive linear relationships between EPC numbers and shear rate postoperatively, which were more significant on POD 30. In addition, postoperative mobilization of EPCs was significantly higher in patients who developed a stenotic fistula than those without. CONCLUSIONS The mobilization of circulating EPCs correlated with a compromised arteriovenous fistula. The biological significance of increased EPC numbers need to be determined in future studies. KEY WORDS Arteriovenous fistula; Endothelial progenitor cells.
Collapse
Affiliation(s)
- Jun-Neng Roan
- Institute of Clinical Medicine; ; Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital; ; Department of Surgery, Tainan Municipal Hospital
| | - Chwan-Yau Luo
- Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital
| | - Mang-Da Tsai
- Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital
| | - I-Shuan Wu
- Department of Surgery, Tainan Municipal Hospital
| | - Shih-Wei Chang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan
| | - Chien-Chi Huang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan
| | | | - Chen-Fuh Lam
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan; ; Department of Anesthesiology, Buddhist Tzu-Chi General Hospital and Tzu-Chi University School of Medicine, Hualien, Taiwan
| |
Collapse
|
305
|
Bogoslovsky T, Maric D, Gong Y, Qu B, Yang K, Spatz M, Hallenbeck J, Diaz-Arrastia R. Preservation and enumeration of endothelial progenitor and endothelial cells from peripheral blood for clinical trials. Biomark Med 2016; 9:625-37. [PMID: 26174838 DOI: 10.2217/bmm.15.34] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIMS Endothelial progenitor cells (EPCs) are markers of vascular repair. Increased numbers of circulating endothelial cells (ECs) are associated with endothelial damage. MATERIALS & METHODS We enumerated EPC-EC by using Enrichment kit with addition of anti-human CD146-PE/Cy7 from peripheral blood mononuclear cell (PBMC) isolated either by red blood cell (RBC) lysing solution or by Ficoll centrifugation, and from fresh and preserved samples. PBMCs were quantified by flow cytometry. RESULTS RBC lysis yielded higher percentage of PBMC (p = 0.0242) and higher numbers of PBMC/ml (p = 0.0039) than Ficoll. Absolute numbers of CD34(+)CD133(+)VEGFR2(+) and CD146(+)CD34(+)VEGFR2(+) were higher (p = 0.0117 for both), when isolated by RBC lysis than by Ficoll, when no difference in other subsets was found. Cryopreservation at -160°C and -80°C and short-term preservation at room temperature decreased EPC-EC. CONCLUSIONS Our data support use of fresh samples and isolation of PBMC from human blood by RBC lysis for enumeration of EPC and EC.
Collapse
Affiliation(s)
- Tanya Bogoslovsky
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of Health Sciences, 12725 Twinbrook Pkwy, Rockville, MD 20852, USA.,National Institute of Neurological Disorders and Stroke, Stroke Branch, 10 Center Drive, Bethesda, MD 20814, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow Cytometry Core Facility, 49 Convent Drive, Bethesda, MD 20814, USA
| | - Yunhua Gong
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of Health Sciences, 12725 Twinbrook Pkwy, Rockville, MD 20852, USA
| | - Baoxi Qu
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of Health Sciences, 12725 Twinbrook Pkwy, Rockville, MD 20852, USA
| | - Kelly Yang
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, MD 20814, USA
| | - Maria Spatz
- National Institute of Neurological Disorders and Stroke, Stroke Branch, 10 Center Drive, Bethesda, MD 20814, USA
| | - John Hallenbeck
- National Institute of Neurological Disorders and Stroke, Stroke Branch, 10 Center Drive, Bethesda, MD 20814, USA
| | - Ramon Diaz-Arrastia
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of Health Sciences, 12725 Twinbrook Pkwy, Rockville, MD 20852, USA
| |
Collapse
|
306
|
Takizawa S, Nagata E, Nakayama T, Masuda H, Asahara T. Recent Progress in Endothelial Progenitor Cell Culture Systems: Potential for Stroke Therapy. Neurol Med Chir (Tokyo) 2016; 56:302-9. [PMID: 27041632 PMCID: PMC4908073 DOI: 10.2176/nmc.ra.2016-0027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Endothelial progenitor cells (EPCs) participate in endothelial repair and angiogenesis due to their abilities to differentiate into endothelial cells and to secrete protective cytokines and growth factors. Consequently, there is considerable interest in cell therapy with EPCs isolated from peripheral blood to treat various ischemic injuries. Quality and quantity-controlled culture systems to obtain mononuclear cells enriched in EPCs with well-defined angiogenic and anti-inflammatory phenotypes have recently been developed, and increasing evidence from animal models and clinical trials supports the idea that transplantation of EPCs contributes to the regenerative process in ischemic organs and is effective for the therapy of ischemic cerebral injury. Here, we briefly describe the general characteristics of EPCs, and we review recent developments in culture systems and applications of EPCs and EPC-enriched cell populations to treat ischemic stroke.
Collapse
Affiliation(s)
- Shunya Takizawa
- Department of Neurology, Tokai University School of Medicine
| | | | | | | | | |
Collapse
|
307
|
Patschan D, Schwarze K, Henze E, Patschan S, Müller GA. The endothelial-to-mesenchymal transition and endothelial cilia in EPC-mediated postischemic kidney protection. Am J Physiol Renal Physiol 2016; 310:F679-F687. [DOI: 10.1152/ajprenal.00306.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/19/2016] [Indexed: 12/12/2022] Open
Abstract
Renal ischemia induces peritubular capillary rarefication and fibrosis, with the latter partly resulting from the endothelial-to-mesenchymal transition (EndoMT). Endothelial cilia transmit blood flow-associated forces into the cell. Early endothelial progenitor cells (eEPCs) have been shown to protect mice from acute kidney injury in the short term. The aim of the present study was to analyze midterm consequences of eEPC treatment in the context of endothelial cilia and the EndoMT. Male C57/Bl6N mice were subjected to unilateral renal ischemia postuninephrectomy. Syngeneic murine eEPCs were systemically injected at the time of reperfusion. Animals were investigated 1, 4, and 6 wk later. Cultured mature endothelial cells were exposed to a variable flow with versus without eEPC supernatant incubation. Systemically injected eEPCs reduced serum creatinine levels at week 1 (35 and 45 min) and week 4 (45 min). Interstitial fibrosis was significantly diminished by cell treatment at all time points as well. The EndoMT was less pronounced at week 4 (35 min) and week 6 (45 min). eEPC supernatant reduced α-smooth muscle actin expression and α-tubulin abundance in flow-treated cultured mature endothelial cells, and percentages of cilium-positive cells increased. The loss of peritubular capillaries was prevented by eEPCs. Intrarenal endothelial α-tubulin decreased postischemia and was further reduced by eEPC administration. We conclude that eEPCs are capable of reorganizing the endothelial cytoskeleton in an indirect manner, ultimately resulting in stabilization of the endothelial ciliome. The investigation indicates an antimesenchymal role of endothelial cilia in the process of postischemic tissue fibrosis/EndoMT.
Collapse
Affiliation(s)
- D. Patschan
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - K. Schwarze
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - E. Henze
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - S. Patschan
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - G. A. Müller
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
308
|
Zhao H, Yun W, Zhang Q, Cai X, Li X, Hui G, Zhou X, Ni J. Mobilization of Circulating Endothelial Progenitor Cells by dl-3-n-Butylphthalide in Acute Ischemic Stroke Patients. J Stroke Cerebrovasc Dis 2016; 25:752-60. [DOI: 10.1016/j.jstrokecerebrovasdis.2015.11.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/01/2015] [Accepted: 11/14/2015] [Indexed: 01/01/2023] Open
|
309
|
Rurali E, Bassetti B, Perrucci GL, Zanobini M, Malafronte C, Achilli F, Gambini E. BM ageing: Implication for cell therapy with EPCs. Mech Ageing Dev 2016; 159:4-13. [PMID: 27045606 DOI: 10.1016/j.mad.2016.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/22/2016] [Accepted: 04/01/2016] [Indexed: 12/17/2022]
Abstract
The bone marrow (BM) is a well-recognized source of stem/progenitor cells for cell therapy in cardiovascular diseases (CVDs). Preclinical and clinical studies suggest that endothelial progenitor cells (EPCs) contribute to reparative process of vascular endothelium and participate in angiogenesis. As for all organs and cells across the lifespan, BM and EPCs are negatively impacted by ageing due to microenvironment modifications and EPC progressive dysfunctions. The encouraging results in terms of neovascularization observed in young animals after EPC administration were mitigated in aged patients treated for ischemic CVDs. The limited efficacy of EPC-based therapy in clinical setting might be ascribed at least partly to ageing. In this review, we comprehensively discussed the age-related changes of BM and EPCs and their implication for cardiovascular cell-therapies. Finally, we examined alternative approaches under investigation to enhance EPC potency.
Collapse
Affiliation(s)
- Erica Rurali
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Marco Zanobini
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Felice Achilli
- Cardiology Department, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
310
|
Haberzettl P, McCracken JP, Bhatnagar A, Conklin DJ. Insulin sensitizers prevent fine particulate matter-induced vascular insulin resistance and changes in endothelial progenitor cell homeostasis. Am J Physiol Heart Circ Physiol 2016; 310:H1423-38. [PMID: 27016579 DOI: 10.1152/ajpheart.00369.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 03/23/2016] [Indexed: 12/15/2022]
Abstract
Exposure to fine particular matter (PM2.5) increases the risk of developing cardiovascular disease and Type 2 diabetes. Because blood vessels are sensitive targets of air pollutant exposure, we examined the effects of concentrated ambient PM2.5 (CAP) on vascular insulin sensitivity and circulating levels of endothelial progenitor cells (EPCs), which reflect cardiovascular health. We found that CAP exposure for 9 days decreased insulin-stimulated Akt phosphorylation in the aorta of mice maintained on control diet. This change was accompanied by the induction of IL-1β and increases in the abundance of cleaved IL-18 and p10 subunit of Casp-1, consistent with the activation of the inflammasome pathway. CAP exposure also suppressed circulating levels of EPCs (Flk-1(+)/Sca-1(+) cells), while enhancing the bone marrow abundance of these cells. Although similar changes in vascular insulin signaling and EPC levels were observed in mice fed high-fat diet, CAP exposure did not exacerbate diet-induced changes in vascular insulin resistance or EPC homeostasis. Treatment with an insulin sensitizer, metformin or rosiglitazone, prevented CAP-induced vascular insulin resistance and NF-κB and inflammasome activation and restored peripheral blood and bone marrow EPC levels. These findings suggest that PM2.5 exposure induces diet-independent vascular insulin resistance and inflammation and prevents EPC mobilization, and that this EPC mobilization defect could be mediated by vascular insulin resistance. Impaired vascular insulin sensitivity may be an important mechanism underlying PM2.5-induced vascular injury, and pharmacological sensitization to insulin action could potentially prevent deficits in vascular repair and mitigate vascular inflammation due to exposure to elevated levels of ambient air pollution.
Collapse
Affiliation(s)
- Petra Haberzettl
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - James P McCracken
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel J Conklin
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
311
|
Significance of endothelial progenitor cells (EPC) for tumorigenesis of head and neck squamous cell carcinoma (HNSCC): possible marker of tumor progression and neovascularization? Clin Oral Investig 2016; 20:2293-2300. [PMID: 26993659 DOI: 10.1007/s00784-016-1785-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/04/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Angiogenesis and neovascularisation plays a crucial role for tumorigenesis and tumor progression in head and neck squamous cell carcinoma (HNSCC). The aim of our study was to investigate the neovascularization capacity by endothelial progenitor cells (EPC) in tumor patient as a possible predictor for tumor progression and tumor stage. MATERIALS AND METHODS Therefore, we investigated the cell number and biologic activity by cell migration and colony-forming ability of EPC. Cells were isolated from the peripheral venous blood of 79 patients who suffer HNSCC in different stages of disease. Thirty-three healthy individuals served as the control group. RESULTS Significantly increased biological activities were reflected by expression of the migration rate (1027 ± 1510) in comparison to the control group (632 ± 269) and the clonal potency measured by colony-forming unit (CFU) (tumor patients (19.7 ± 12.3) vs. control group (10.84 ± 4.8)). To determine whether or not EPC number can be used as a valid prognostic marker for clinical outcome of tumor patients, we furthermore compared a "high EPC-number-subgroup" (HI) with a "low EPC-number-subgroup" (LO) in a Kaplan-Meier survival curve. The HI-subgroup shows herein clearly a worse outcome. CONCLUSIONS Our findings indicate a possible pathway for EPC to play a critical role in the vasculogenesis and consequently in the progression of HNSCC. CLINICAL RELEVANCE Our findings could serve as possible predictors for the neovascularisation potential in HNSCC tumor patients.
Collapse
|
312
|
Danova M, Comolli G, Manzoni M, Torchio M, Mazzini G. Flow cytometric analysis of circulating endothelial cells and endothelial progenitors for clinical purposes in oncology: A critical evaluation. Mol Clin Oncol 2016; 4:909-917. [PMID: 27284422 DOI: 10.3892/mco.2016.823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/11/2015] [Indexed: 12/19/2022] Open
Abstract
Malignant tumors are characterized by uncontrolled cell growth and metastatic spread, with a pivotal importance of the phenomenon of angiogenesis. For this reason, research has focused on the development of agents targeting the vascular component of the tumor microenvironment and regulating the angiogenic switch. As a result, the therapeutic inhibition of angiogenesis has become an important component of anticancer treatment, however, its utility is partly limited by the lack of an established methodology to assess its efficacy in vivo. Circulating endothelial cells (CECs), which are rare in healthy subjects and significantly increased in different tumor types, represent a promising tool for monitoring the tumor clinical outcome and the treatment response. A cell population circulating into the blood also able to form endothelial colonies in vitro and to promote vasculogenesis is represented by endothelial progenitor cells (EPCs). The number of both of these cell types is extremely low and they cannot be identified using a single marker, therefore, in absence of a definite consensus on their phenotype, require discrimination using combinations of antigens. Multiparameter flow cytometry (FCM) is ideal for rapid processing of high numbers of cells per second and is commonly utilized to quantify CECs and EPCs, however, remains technically challenging since there is as yet no standardized protocol for the identification and enumeration of these rare events. Methodology in studies on CECs and/or EPCs as clinical biomarkers in oncology is heterogeneous and data have been obtained from different studies leading to conflicting conclusions. The present review presented a critical review of the issues that limit the comparability of results of the most significant studies employing FCM for CEC and/or EPC detection in patients with cancer.
Collapse
Affiliation(s)
- Marco Danova
- Internal Medicine and Medical Oncology, Vigevano Hospital, ASST Pavia, I-27029 Vigevano, Italy
| | - Giuditta Comolli
- Microbiology and Virology, Biotechnology Laboratories, IRCCS San Matteo Foundation, I-27100 Pavia, Italy
| | | | - Martina Torchio
- Internal Medicine and Medical Oncology, Vigevano Hospital, ASST Pavia, I-27029 Vigevano, Italy
| | - Giuliano Mazzini
- Molecular Genetics Institute, National Research Council and Biology and Biotechnology Department 'L. Spallanzani', University of Pavia, I-27100 Pavia, Italy
| |
Collapse
|
313
|
Lee JH, Lee SH, Choi SH, Asahara T, Kwon SM. The sulfated polysaccharide fucoidan rescues senescence of endothelial colony-forming cells for ischemic repair. Stem Cells 2016; 33:1939-51. [PMID: 25693733 DOI: 10.1002/stem.1973] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/15/2015] [Indexed: 01/08/2023]
Abstract
The efficacy of cell therapy using endothelial colony-forming cells (ECFCs) in the treatment of ischemia is limited by the replicative senescence of isolated ECFCs in vitro. Such senescence must therefore be overcome in order for such cell therapies to be clinically applicable. This study aimed to investigate the potential of sulfated polysaccharide fucoidan to rescue ECFCs from cellular senescence and to improve in vivo vascular repair by ECFCs. Fucoidan-preconditioning of senescent ECFCs was shown by flow cytometry to restore the expression of functional ECFC surface markers (CD34, c-Kit, VEGFR2, and CXCR4) and stimulate the in vitro tube formation capacity of ECFCs. Fucoidan also promoted the expression of cell cycle-associated proteins (cyclin E, Cdk2, cyclin D1, and Cdk4) in senescent ECFCs, significantly reversed cellular senescence, and increased the proliferation of ECFCs via the FAK, Akt, and ERK signaling pathways. Fucoidan was found to enhance the survival, proliferation, incorporation, and endothelial differentiation of senescent ECFCs transplanted in ischemic tissues in a murine hind limb ischemia model. Moreover, ECFC-induced functional recovery and limb salvage were markedly improved by fucoidan pretreatment of ECFCs. To our knowledge, the findings of our study are the first to demonstrate that fucoidan enhances the neovasculogenic potential of ECFCs by rescuing them from replicative cellular senescence. Pretreatment of ECFCs with fucoidan may thus provide a novel strategy for the application of senescent stem cells to therapeutic neovascularization.
Collapse
Affiliation(s)
- Jun Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sang Hun Lee
- Soonchunhyang Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Yongsan-gu, Seoul, Korea.,Department of Biochemistry, School of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Sung Hyun Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, Korea
| | - Takayuki Asahara
- Department Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
314
|
Cui Y, Sun Q, Liu Z. Ambient particulate matter exposure and cardiovascular diseases: a focus on progenitor and stem cells. J Cell Mol Med 2016; 20:782-93. [PMID: 26988063 PMCID: PMC4831366 DOI: 10.1111/jcmm.12822] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/29/2016] [Indexed: 12/13/2022] Open
Abstract
Air pollution is a major challenge to public health. Ambient fine particulate matter (PM) is the key component for air pollution, and associated with significant mortality. The majority of the mortality following PM exposure is related to cardiovascular diseases. However, the mechanisms for the adverse effects of PM exposure on cardiovascular system remain largely unknown and under active investigation. Endothelial dysfunction or injury is considered one of the major factors that contribute to the development of cardiovascular diseases such as atherosclerosis and coronary heart disease. Endothelial progenitor cells (EPCs) play a critical role in maintaining the structural and functional integrity of vasculature. Particulate matter exposure significantly suppressed the number and function of EPCs in animals and humans. However, the mechanisms for the detrimental effects of PM on EPCs remain to be fully defined. One of the important mechanisms might be related to increased level of reactive oxygen species (ROS) and inflammation. Bone marrow (BM) is a major source of EPCs. Thus, the number and function of EPCs could be intimately associated with the population and functional status of stem cells (SCs) in the BM. Bone marrow stem cells and other SCs have the potential for cardiovascular regeneration and repair. The present review is focused on summarizing the detrimental effects of PM exposure on EPCs and SCs, and potential mechanisms including ROS formation as well as clinical implications.
Collapse
Affiliation(s)
- Yuqi Cui
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Qinghua Sun
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zhenguo Liu
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
315
|
Chong MSK, Ng WK, Chan JKY. Concise Review: Endothelial Progenitor Cells in Regenerative Medicine: Applications and Challenges. Stem Cells Transl Med 2016; 5:530-8. [PMID: 26956207 DOI: 10.5966/sctm.2015-0227] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Endothelial progenitor cells (EPCs) are currently being studied as candidate cell sources for revascularization strategies. Significant advances have been made in understanding the biology of EPCs, and preclinical studies have demonstrated the vasculogenic, angiogenic, and beneficial paracrine effects of transplanted EPCs in the treatment of ischemic diseases. Despite these promising results, widespread clinical acceptance of EPCs for clinical therapies remains hampered by several challenges. The present study provides a concise summary of the different EPC populations being studied for ischemic therapies and their known roles in the healing of ischemic tissues. The challenges and issues surrounding the use of EPCs and the current strategies being developed to improve the harvest efficiency and functionality of EPCs for application in regenerative medicine are discussed. SIGNIFICANCE Endothelial progenitor cells (EPCs) have immense clinical value for cardiovascular therapies. The present study provides a concise description of the EPC subpopulations being evaluated for clinical applications. The current major lines of investigation involving preclinical and clinical evaluations of EPCs are discussed, and significant gaps limiting the translation of EPCs are highlighted. The present report could be useful for clinicians and clinical researchers with interests in ischemic therapy and for basic scientists working in the related fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mark Seow Khoon Chong
- School of Chemical and Biochemical Engineering, Nanyang Technological University, Singapore
| | - Wei Kai Ng
- School of Chemical and Biochemical Engineering, Nanyang Technological University, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore Department of Obstetrics and Gynaecology, National University of Singapore, Singapore
| |
Collapse
|
316
|
Paneni F, Costantino S, Kränkel N, Cosentino F, Lüscher TF. Reprogramming ageing and longevity genes restores paracrine angiogenic properties of early outgrowth cells. Eur Heart J 2016; 37:1733-7. [DOI: 10.1093/eurheartj/ehw073] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/01/2016] [Indexed: 01/20/2023] Open
|
317
|
Zeng H, Jiang Y, Tang H, Ren Z, Zeng G, Yang Z. Abnormal phosphorylation of Tie2/Akt/eNOS signaling pathway and decreased number or function of circulating endothelial progenitor cells in prehypertensive premenopausal women with diabetes mellitus. BMC Endocr Disord 2016; 16:13. [PMID: 26936372 PMCID: PMC4776390 DOI: 10.1186/s12902-016-0093-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUNDS The number and activity of circulating endothelial progenitor cells (EPCs) in prehypertension is preserved in premenopausal women. However, whether this favorable effect still exists in prehypertensive premenopausal women with diabetes is not clear. METHODS This study compared the number and functional activity of circulating EPCs in normotensive or prehypertensive premenopausal women without diabetes mellitus and normotensive or prehypertensive premenopausal women with diabetes mellitus, evaluated the vascular endothelial function in each groups, and investigated the possible underlying mechanism. RESULTS We found that compared with normotensive premenopausal women, the number and function of circulating EPCs, as well as endothelial function evaluated by flow-mediated dilatation (FMD) in prehypertensive premenopausal women were preserved. In parallel, the Tie2/Akt/eNOS signaling pathway and the plasma NO level or NO secretion of circulating EPCs in prehypertensive premenopausal women was also retained. However, in presence of normotension or prehypertension with diabetes mellitus, the number or function of circulating EPCs and FMD in premenopausal women decreased. Similarly, the phosphorylation of Tie2/Akt/eNOS signaling pathway and the plasma NO level or NO secretion of circulating EPCs was reduced in prehypertension premenopausal with diabetes mellitus. CONCLUSION The present findings firstly demonstrate that the unfavorable effects of diabetes mellitus on number and activity of circulating EPCs in prehypertension premenopausal women, which is at least partially related to the abnormal phosphorylation of Tie2/Akt/eNOS signaling pathway and subsequently reduced nitric oxide bioavailability. The Tie2/Akt/eNOS signaling pathway may be a potential target of vascular protection in prehypertensive premenopausal women with diabetes mellitus.
Collapse
Affiliation(s)
- Haitao Zeng
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Yanping Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Hailin Tang
- Cancer Center, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Zi Ren
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Gaofeng Zeng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Yang
- Department of Hypertension & Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
318
|
Hong X, Le Bras A, Margariti A, Xu Q. Reprogramming towards endothelial cells for vascular regeneration. Genes Dis 2016; 3:186-197. [PMID: 30258888 PMCID: PMC6147164 DOI: 10.1016/j.gendis.2016.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/11/2016] [Indexed: 01/08/2023] Open
Abstract
Endothelial damage and dysfunction are implicated in cardiovascular pathological changes and the development of vascular diseases. In view of the fact that the spontaneous endothelial cell (EC) regeneration is a slow and insufficient process, it is of great significance to explore alternative cell sources capable of generating functional ECs to repair damaged endothelium. Indeed, recent achievements of cell reprogramming to convert somatic cells to other cell types provide new powerful approaches to study endothelial regeneration. Based on progress in the research field, the present review aims to summarize the strategies and mechanisms of generating endothelial cells through reprogramming from somatic cells, and to examine what this means for the potential application of cell therapy in the clinic.
Collapse
Affiliation(s)
- Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Alexandra Le Bras
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Andriana Margariti
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
319
|
Banyard DA, Adnani BO, Melkumyan S, Araniego CA, Widgerow AD. Endothelial progenitor cells and burn injury - exploring the relationship. BURNS & TRAUMA 2016; 4:4. [PMID: 27574674 PMCID: PMC4964096 DOI: 10.1186/s41038-016-0028-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/13/2016] [Indexed: 12/25/2022]
Abstract
Burn wounds result in varying degrees of soft tissue damage that are typically graded clinically. Recently a key participant in neovascularization, the endothelial progenitor cell, has been the subject of intense cardiovascular research to explore whether it can serve as a biomarker for vascular injury. In this review, we examine the identity of the endothelial progenitor cell as well as the evidence that support its role as a key responder after burn insult. While there is conflicting evidence with regards to the delta of endothelial progenitor cell mobilization and burn severity, it is clear that they play an important role in wound healing. Systematic and controlled studies are needed to clarify this relationship, and whether this population can serve as a biomarker for burn severity.
Collapse
Affiliation(s)
- Derek A Banyard
- Department of Plastic Surgery, Center for Tissue Engineering, University of California, Irvine, 200S Manchester Ave, Ste 650, Orange, CA 92868 USA
| | - Blake O Adnani
- Department of Plastic Surgery, Center for Tissue Engineering, University of California, Irvine, 200S Manchester Ave, Ste 650, Orange, CA 92868 USA
| | - Satenik Melkumyan
- Department of Plastic Surgery, Center for Tissue Engineering, University of California, Irvine, 200S Manchester Ave, Ste 650, Orange, CA 92868 USA
| | - Cheryl Ann Araniego
- Department of Plastic Surgery, Center for Tissue Engineering, University of California, Irvine, 200S Manchester Ave, Ste 650, Orange, CA 92868 USA
| | - Alan D Widgerow
- Department of Plastic Surgery, Center for Tissue Engineering, University of California, Irvine, 200S Manchester Ave, Ste 650, Orange, CA 92868 USA
| |
Collapse
|
320
|
Ma Q, Xia X, Tao Q, Lu K, Shen J, Xu Q, Hu X, Tang Y, Block NL, Webster KA, Schally AV, Wang J, Yu H. Profound Actions of an Agonist of Growth Hormone-Releasing Hormone on Angiogenic Therapy by Mesenchymal Stem Cells. Arterioscler Thromb Vasc Biol 2016; 36:663-672. [PMID: 26868211 DOI: 10.1161/atvbaha.116.307126] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 01/21/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The efficiency of cell therapy is limited by poor cell survival and engraftment. Here, we studied the effect of the growth hormone-releasing hormone agonist, JI-34, on mesenchymal stem cell (MSC) survival and angiogenic therapy in a mouse model of critical limb ischemia. APPROACH AND RESULTS Mouse bone marrow-derived MSCs were incubated with or without 10(-8) mol/L JI-34 for 24 hours. MSCs were then exposed to hypoxia and serum deprivation to detect the effect of preconditioning on cell apoptosis, migration, and tube formation. For in vivo tests, critical limb ischemia was induced by femoral artery ligation. After surgery, mice received 50 μL phosphate-buffered saline or with 1×10(6) MSCs or with 1×10(6) JI-34-reconditioned MSCs. Treatment of MSCs with JI-34 improved MSC viability and mobility and markedly enhanced their capability to promote endothelial tube formation in vitro. These effects were paralleled by an increased phosphorylation and nuclear translocation of signal transducer and activator of transcription 3. In vivo, JI-34 pretreatment enhanced the engraftment of MSCs into ischemic hindlimb muscles and augmented reperfusion and limb salvage compared with untreated MSCs. Significantly more vasculature and proliferating CD31(+) and CD34(+) cells were detected in ischemic muscles that received MSCs treated with JI-34. CONCLUSIONS Our studies demonstrate a novel role for JI-34 to markedly improve therapeutic angiogenesis in hindlimb ischemia by increasing the viability and mobility of MSCs. These findings support additional studies to explore the full potential of growth hormone-releasing hormone agonists to augment cell therapy in the management of ischemia.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Antigens, CD34/metabolism
- Apoptosis/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Growth Hormone-Releasing Hormone/agonists
- Growth Hormone-Releasing Hormone/analogs & derivatives
- Growth Hormone-Releasing Hormone/metabolism
- Growth Hormone-Releasing Hormone/pharmacology
- Hindlimb
- Ischemia/metabolism
- Ischemia/physiopathology
- Ischemia/therapy
- Male
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Muscle, Skeletal/blood supply
- Neovascularization, Physiologic
- Peptide Fragments/pharmacology
- Phosphorylation
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Receptors, Neuropeptide/agonists
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/agonists
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- STAT3 Transcription Factor/metabolism
- Time Factors
Collapse
|
321
|
Siavashi V, Nassiri SM, Rahbarghazi R, Vafaei R, Sariri R. ECM-Dependence of Endothelial Progenitor Cell Features. J Cell Biochem 2016; 117:1934-46. [DOI: 10.1002/jcb.25492] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/08/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Vahid Siavashi
- Department of Biology; Faculty of Sciences; University of Guilan; Rasht Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Rana Vafaei
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Reyhaneh Sariri
- Department of Biology; Faculty of Sciences; University of Guilan; Rasht Iran
| |
Collapse
|
322
|
Almubarak S, Nethercott H, Freeberg M, Beaudon C, Jha A, Jackson W, Marcucio R, Miclau T, Healy K, Bahney C. Tissue engineering strategies for promoting vascularized bone regeneration. Bone 2016; 83:197-209. [PMID: 26608518 PMCID: PMC4911893 DOI: 10.1016/j.bone.2015.11.011] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/06/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
This review focuses on current tissue engineering strategies for promoting vascularized bone regeneration. We review the role of angiogenic growth factors in promoting vascularized bone regeneration and discuss the different therapeutic strategies for controlled/sustained growth factor delivery. Next, we address the therapeutic uses of stem cells in vascularized bone regeneration. Specifically, this review addresses the concept of co-culture using osteogenic and vasculogenic stem cells, and how adipose derived stem cells compare to bone marrow derived mesenchymal stem cells in the promotion of angiogenesis. We conclude this review with a discussion of a novel approach to bone regeneration through a cartilage intermediate, and discuss why it has the potential to be more effective than traditional bone grafting methods.
Collapse
Affiliation(s)
- Sarah Almubarak
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Hubert Nethercott
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Marie Freeberg
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Caroline Beaudon
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Amit Jha
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Wesley Jackson
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin Healy
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States.
| |
Collapse
|
323
|
Wong WT, Cooke JP. Therapeutic transdifferentiation of human fibroblasts into endothelial cells using forced expression of lineage-specific transcription factors. J Tissue Eng 2016; 7:2041731416628329. [PMID: 27081470 PMCID: PMC4820020 DOI: 10.1177/2041731416628329] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/31/2015] [Indexed: 12/14/2022] Open
Abstract
Transdifferentiation is the direct conversion from one somatic cell type into another desired somatic cell type. This reprogramming method offers an attractive approach for regenerative medicine. Here, we demonstrate that neonatal fibroblasts can be transdifferentiated into endothelial cells using only four endothelial transcription factors, namely, ETV2, FLI1, GATA2, and KLF4. We observed a significant up-regulation of endothelial genes including KDR, CD31, CD144, and vWF in human neonatal foreskin (BJ) fibroblasts infected with the lentiviral construct encoding the open reading frame of the four transcription factors. We observed morphological changes in BJ fibroblasts from the fibroblastic spindle shape into a more endothelial-like cobblestone structures. Fluorescence-activated cell sorting analysis revealed that ~16% of the infected cells with the lentiviral constructs encoding 4F expressed CD31. The sorted cells were allowed to expand for 2 weeks and these cells were immunostained and found to express endothelial markers CD31. The induced endothelial cells also incorporated fluorescence-labeled acetylated low-density lipoprotein and efficiently formed capillary-like networks when seeded on Matrigel. These results suggested that the induced endothelial cells were functional in vitro. Taken together, we successfully demonstrated the direct conversion of human neonatal fibroblasts into endothelial cells by transduction of lentiviral constructs encoding endothelial lineage-specific transcription factors ETV2, FLI1, GATA2, and KLF4. The directed differentiation of fibroblasts into endothelial cells may have significant utility in diseases characterized by fibrosis and loss of microvasculature.
Collapse
Affiliation(s)
- Wing Tak Wong
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
324
|
Watt J, Kennedy S, Ahmed N, Hayhurst J, McClure JD, Berry C, Wadsworth RM, Oldroyd KG. The relationship between oxidised LDL, endothelial progenitor cells and coronary endothelial function in patients with CHD. Open Heart 2016; 3:e000342. [PMID: 26848395 PMCID: PMC4731840 DOI: 10.1136/openhrt-2015-000342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/18/2015] [Accepted: 12/20/2015] [Indexed: 12/28/2022] Open
Abstract
Objective The balance between coronary endothelial dysfunction and repair is influenced by many protective and deleterious factors circulating in the blood. We studied the relationship between oxidised low-density lipoprotein (oxLDL), circulating endothelial progenitor cells (EPCs) and coronary endothelial function in patients with stable coronary heart disease (CHD). Methods 33 patients with stable CHD were studied. Plasma oxLDL was measured using ELISA, coronary endothelial function was assessed using intracoronary acetylcholine infusion and EPCs were quantified using flow cytometry for CD34+/KDR+ cells. Results Plasma oxLDL correlated positively with the number of EPCs in the blood (r=0.46, p=0.02). There was a positive correlation between the number of circulating EPCs and coronary endothelial function (r=0.42, p=0.04). There was no significant correlation between oxLDL and coronary endothelial function. Conclusions Plasma levels of oxLDL are associated with increased circulating EPCs in the blood of patients with CHD, which may reflect a host-repair response to endothelial injury. Patients with stable CHD had a high prevalence of coronary endothelial dysfunction, which was associated with lower numbers of circulating EPCs, suggesting a mechanistic link between endothelial dysfunction and the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Jonathan Watt
- West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital , Glasgow , UK
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow , UK
| | - Nadeem Ahmed
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow , UK
| | - James Hayhurst
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow , UK
| | - John D McClure
- West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital , Glasgow , UK
| | - Colin Berry
- West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital , Glasgow , UK
| | - Roger M Wadsworth
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow , UK
| | - Keith G Oldroyd
- West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital , Glasgow , UK
| |
Collapse
|
325
|
Cianciolo G, Capelli I, Cappuccilli M, Schillaci R, Cozzolino M, La Manna G. Calcifying circulating cells: an uncharted area in the setting of vascular calcification in CKD patients. Clin Kidney J 2016; 9:280-6. [PMID: 26985381 PMCID: PMC4792620 DOI: 10.1093/ckj/sfv145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/27/2015] [Indexed: 12/16/2022] Open
Abstract
Vascular calcification, occurring during late-stage vascular and valvular disease, is highly associated with chronic kidney disease-mineral and bone disorders (CKD-MBD), representing a major risk factor for cardiovascular morbidity and mortality. The hallmark of vascular calcification, which involves both media and intima, is represented by the activation of cells committed to an osteogenic programme. Several studies have analysed the role of circulating calcifying cells (CCCs) in vascular calcification. CCCs are bone marrow (BM)-derived cells with an osteogenic phenotype, participating in intima calcification processes and defined by osteocalcin and bone alkaline phosphatase expression. The identification of CCCs in diabetes and atherosclerosis is the most recent, intriguing and yet uncharted chapter in the scenario of the bone-vascular axis. Whether osteogenic shift occurs in the BM, the bloodstream or both, is not known, and also the factors promoting CCC formation have not been identified. However, it is possible to recognize a common pathogenic commitment of inflammation in atherosclerosis and diabetes, in which metabolic control may also have a role. Currently available studies in patients without CKD did not find an association of CCCs with markers of bone metabolism. Preliminary data on CKD patients indicate an implication of mineral bone disease in vascular calcification, as a consequence of functional and anatomic integrity interruption of BM niches. Given the pivotal role that parathyroid hormone and osteoblasts play in regulating expansion, mobilization and homing of haematopoietic stem/progenitors cells, CKD-MBD could promote CCC formation.
Collapse
Affiliation(s)
- Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Irene Capelli
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Maria Cappuccilli
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Roberto Schillaci
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Mario Cozzolino
- Nephrology and Dialysis, S. Paolo Hospital, Department of Health Sciences (DISS) , University of Milan , Milan , Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| |
Collapse
|
326
|
Kotlinowski J, Jozkowicz A. PPAR Gamma and Angiogenesis: Endothelial Cells Perspective. J Diabetes Res 2016; 2016:8492353. [PMID: 28053991 PMCID: PMC5174176 DOI: 10.1155/2016/8492353] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/22/2016] [Accepted: 11/01/2016] [Indexed: 12/24/2022] Open
Abstract
We summarize the current knowledge concerning PPARγ function in angiogenesis. We discuss the mechanisms of action for PPARγ and its role in vasculature development and homeostasis, focusing on endothelial cells, endothelial progenitor cells, and bone marrow-derived proangiogenic cells.
Collapse
Affiliation(s)
- Jerzy Kotlinowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- *Alicja Jozkowicz:
| |
Collapse
|
327
|
Endothelial Progenitor Cells for Diagnosis and Prognosis in Cardiovascular Disease. Stem Cells Int 2015; 2016:8043792. [PMID: 26839569 PMCID: PMC4709789 DOI: 10.1155/2016/8043792] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/16/2015] [Accepted: 09/20/2015] [Indexed: 12/12/2022] Open
Abstract
Objective. To identify, evaluate, and synthesize evidence on the predictive power of circulating endothelial progenitor cells (EPCs) in cardiovascular disease, through a systematic review of quantitative studies. Data Sources. MEDLINE was searched using keywords related to "endothelial progenitor cells" and "endothelium" and, for the different categories, respectively, "smoking"; "blood pressure"; "diabetes mellitus" or "insulin resistance"; "dyslipidemia"; "aging" or "elderly"; "angina pectoris" or "myocardial infarction"; "stroke" or "cerebrovascular disease"; "homocysteine"; "C-reactive protein"; "vitamin D". Study Selection. Database hits were evaluated against explicit inclusion criteria. From 927 database hits, 43 quantitative studies were included. Data Syntheses. EPC count has been suggested for cardiovascular risk estimation in the clinical practice, since it is currently accepted that EPCs can work as proangiogenic support cells, maintaining their importance as regenerative/reparative potential, and also as prognostic markers. Conclusions. EPCs showed an important role in identifying cardiovascular risk conditions, and to suggest their evaluation as predictor of outcomes appears to be reasonable in different defined clinical settings. Due to their capability of proliferation, circulation, and the development of functional progeny, great interest has been directed to therapeutic use of progenitor cells in atherosclerotic diseases. This trial is registered with registration number: Prospero CRD42015023717.
Collapse
|
328
|
Diabetes-Induced Oxidative Stress in Endothelial Progenitor Cells May Be Sustained by a Positive Feedback Loop Involving High Mobility Group Box-1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:1943918. [PMID: 26798412 PMCID: PMC4698939 DOI: 10.1155/2016/1943918] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/21/2022]
Abstract
Oxidative stress is considered to be a critical factor in diabetes-induced endothelial progenitor cell (EPC) dysfunction, although the underlying mechanisms are not fully understood. In this study, we investigated the role of high mobility group box-1 (HMGB-1) in diabetes-induced oxidative stress. HMGB-1 was upregulated in both serum and bone marrow-derived monocytes from diabetic mice compared with control mice. In vitro, advanced glycation end productions (AGEs) induced, expression of HMGB-1 in EPCs and in cell culture supernatants in a dose-dependent manner. However, inhibition of oxidative stress with N-acetylcysteine (NAC) partially inhibited the induction of HMGB-1 induced by AGEs. Furthermore, p66shc expression in EPCs induced by AGEs was abrogated by incubation with glycyrrhizin (Gly), while increased superoxide dismutase (SOD) activity in cell culture supernatants was observed in the Gly treated group. Thus, HMGB-1 may play an important role in diabetes-induced oxidative stress in EPCs via a positive feedback loop involving the AGE/reactive oxygen species/HMGB-1 pathway.
Collapse
|
329
|
Functional and Biological Role of Endothelial Precursor Cells in Tumour Progression: A New Potential Therapeutic Target in Haematological Malignancies. Stem Cells Int 2015; 2016:7954580. [PMID: 26788072 PMCID: PMC4691637 DOI: 10.1155/2016/7954580] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/19/2015] [Accepted: 08/10/2015] [Indexed: 12/11/2022] Open
Abstract
It was believed that vasculogenesis occurred only during embryo life and that postnatal formation of vessels arose from angiogenesis. Recent findings demonstrate the existence of Endothelial Precursor Cells (EPCs), which take partin postnatal vasculogenesis. EPCs are recruited from the bone marrow under the stimulation of growth factors and cytokines and reach the sites of neovascularization in both physiological and pathological conditions such as malignancies where they contribute to the “angiogenic switch” and tumor progression. An implementation of circulating EPCs in the bloodstream of patients with haematological malignancies has been demonstrated. This increase is strictly related to the bone marrow microvessel density and correlated with a poor prognosis. The EPCs characterization is a very complex process and still under investigation. This literature review aims to provide an overview of the functional and biological role of EPCs in haematological malignancies and to investigate their potential as a new cancer therapeutic target.
Collapse
|
330
|
Aso Y, Jojima T, Iijima T, Suzuki K, Terasawa T, Fukushima M, Momobayashi A, Hara K, Takebayashi K, Kasai K, Inukai T. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, increases the number of circulating CD34⁺CXCR4⁺ cells in patients with type 2 diabetes. Endocrine 2015. [PMID: 26209038 DOI: 10.1007/s12020-015-0688-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated the effects of sitagliptin, a dipeptidyl peptidase (DPP)-4 inhibitor, on the number of circulating CD34(+)CXCR4(+)cells, a candidate for endothelial progenitor cells (EPCs), plasma levels of stromal cell-derived factor (SDF)-1α, a ligand for CXCR4 receptor and a substrate for DPP-4, and plasma levels of interferon-inducible protein (IP)-10, for a substrate for DPP-4, in patients with type 2 diabetes. We studied 30 consecutive patients with type 2 diabetes who had poor glycemic control despite treatment with metformin and/or sulfonylurea. Thirty diabetic patients were randomized in a 2:1 ratio into a sitagliptin (50 mg/day) treatment group or an active placebo group (glimepiride 1 mg/day) for 12 weeks. Both groups showed similar improvements in glycemic control. The number of circulating CD34(+)CXCR4(+) cells was increased from 30.5 (20.0, 47.0)/10(6) cells at baseline to 55.5 (31.5, 80.5)/10(6) cells at 12 weeks of treatment with 50 mg/day sitagliptin (P = 0.0014), while showing no significant changes in patients treated with glimepiride. Plasma levels of SDF-1α and IP-10, both physiological substrates of endogenous DPP-4 and chemokines, were significantly decreased at 12 weeks of sitagliptin treatment. In conclusion, treatment with sitagliptin increased the number of circulating CD34(+)CXCR4(+) cells by approximately 2-fold in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yoshimasa Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan.
| | - T Jojima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - T Iijima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - K Suzuki
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kita-Kobayashi, Shimotsuga, Mibu, Tochigi, 321-0293, Japan
| | - T Terasawa
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - M Fukushima
- LSI Medience Corp., Itabashi-Ku, Tokyo, Japan
| | | | - K Hara
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - K Takebayashi
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| | - K Kasai
- Department of Medicine, Ishibashi General Hospital, Shimotsuke, Tochigi, Japan
| | - T Inukai
- Department of Internal Medicine, Koshigaya Hospital, Dokkyo Medical University, Koshigaya, Saitama, Japan
| |
Collapse
|
331
|
Fadini GP, Bonora BM, Marcuzzo G, Marescotti MC, Cappellari R, Pantano G, Sanzari MC, Duran X, Vendrell J, Plebani M, Avogaro A. Circulating Stem Cells Associate With Adiposity and Future Metabolic Deterioration in Healthy Subjects. J Clin Endocrinol Metab 2015; 100:4570-8. [PMID: 26469382 DOI: 10.1210/jc.2015-2867] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Obesity and metabolic syndrome are associated with mild leukocytosis, but whether hematopoietic stem/progenitor cells (HSPCs) play a role in metabolic deterioration is unknown. OBJECTIVE Our objective was to analyze the cross-sectional and longitudinal associations between CD34(+) HSPCs, adiposity, and metabolic syndrome features. DESIGN This is a cross-sectional study on 242 participants, 155 of whom were followed and included in a longitudinal assessment. SETTING This study took place in a tertiary referral center for metabolic diseases. PARTICIPANTS Healthy working individuals attending a cardiovascular screening program (total n = 3158) and having a baseline measure of circulating CD34(+) cells participated. MAIN OUTCOME MEASURES We collected demographic and anthropometric data, cardiovascular risk factors, and metabolic syndrome parameters. RESULTS Participants (34.7% males, mean age 45.9 ± 0.5 years) were free from diabetes and cardiovascular disease. Cross-sectionally, absolute CD34(+) cell counts were directly correlated with body mass index and waist circumference, inversely correlated with high-density lipoprotein cholesterol and the quantitative insulin sensitivity check index, and were higher in individuals with the metabolic syndrome. The hematopoietic component contributed most to the association of CD34(+) cells with adiposity. During a 6.3-year follow-up, high absolute levels of CD34(+) cells were associated with increasing waist circumference, declining quantitative insulin sensitivity check index and high-density lipoprotein cholesterol, and with incidence of metabolic syndrome. Relative CD34(+) cell counts showed weaker associations with metabolic parameters than absolute levels, but were longitudinally associated with increasing waist circumference and metabolic syndrome development. CONCLUSIONS A mild elevation of circulating CD34(+) progenitor cells, reflecting expansion of HSPCs, is associated with adiposity and future metabolic deterioration in healthy individuals.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Benedetta Maria Bonora
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Giorgio Marcuzzo
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Maria Cristina Marescotti
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Roberta Cappellari
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Giorgia Pantano
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Maria Colomba Sanzari
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Xavier Duran
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Joan Vendrell
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Mario Plebani
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Angelo Avogaro
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| |
Collapse
|
332
|
Huang Z, Zhang X, Zhang H, Ye R, Xiong Y, Sun W, Li Y, Liu X. Reduced endothelial progenitor cells in extracranial arterial stenosis but not intracranial arterial stenosis. J Vasc Surg 2015; 62:1539-45. [DOI: 10.1016/j.jvs.2015.07.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/23/2015] [Indexed: 11/27/2022]
|
333
|
Hirase T. Capturing VE-Cadherin-Positive Endothelial Progenitor Cells for in-stent Vascular Repair. J Atheroscler Thromb 2015; 23:46-7. [PMID: 26549735 DOI: 10.5551/jat.ed027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Tetsuaki Hirase
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute
| |
Collapse
|
334
|
Bou Khzam L, Bouchereau O, Boulahya R, Hachem A, Zaid Y, Abou-Saleh H, Merhi Y. Early outgrowth cells versus endothelial colony forming cells functions in platelet aggregation. J Transl Med 2015; 13:353. [PMID: 26552480 PMCID: PMC4640203 DOI: 10.1186/s12967-015-0723-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/02/2015] [Indexed: 12/26/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) have been implicated in neoangiogenesis, endothelial repair and cell-based therapies for cardiovascular diseases. We have previously shown that the recruitment of EPCs to sites of vascular lesions is facilitated by platelets where EPCs, in turn, modulate platelet function and thrombosis. However, EPCs encompass a heterogeneous population of progenitor cells that may exert different effects on platelet function. Recent evidence suggests the existence of two EPC subtypes: early outgrowth cells (EOCs) and endothelial colony-forming cells (ECFCs). We aimed at characterizing these two EPC subtypes and at identifying their role in platelet aggregation. Methods EOCs and ECFCs were generated from human peripheral blood mononuclear cells (PBMCs) seeded in conditioned media on fibronectin and collagen, respectively. The morphological, phenotypical and functional characteristics of EOCs and ECFCs were assessed by optical and confocal laser scanning microscopes, cell surface markers expression, and Matrigel tube formation. The impact of EOCs and ECFCs on platelet aggregation was monitored in collagen-induced optical aggregometry and compared with PBMCs and human umbilical vein endothelial cells (HUVECs). The levels of the anti-platelet agents’ nitric oxide (NO) and prostacyclin (PGI2) released from cultured cells as well as the expression of their respective producing enzymes NO synthases (NOS) and cyclooxygenases (COX) were also assessed. Results We showed that EOCs display a monocytic-like phenotype whereas ECFCs have an endothelial-like phenotype. We demonstrated that both EOCs and ECFCs and their supernatants inhibited platelet aggregation; however ECFCs were more efficient than EOCs. This could be related to the release of significantly higher amounts of NO and PGI2 from ECFCs, in comparison to EOCs. Indeed, ECFCs, like HUVECs, constitutively express the endothelial (eNOS)—and inducible (iNOS)—NOS isoforms, and COX-1 and weakly express COX-2, whereas EOCs do not constitutively express these NO and PGI2 producing enzymes. Conclusion The different morphological, phenotypic and more importantly the release of the anti-aggregating agents PGI2 and NO in each EPC subtype are implicated in their respective roles in platelet function and thus, may be linked to the increased efficiency of ECFCs in inhibiting platelet aggregation as compared to EOCs.
Collapse
Affiliation(s)
- Lara Bou Khzam
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada. .,Department of Biochemistry, Weill Cornell Medical College in Qatar, Doha, Qatar.
| | - Olivier Bouchereau
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada.
| | - Rahma Boulahya
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada.
| | - Ahmed Hachem
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada.
| | - Younes Zaid
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada.
| | | | - Yahye Merhi
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, QC, H1T 1C8, Canada. .,Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
335
|
Buchmann N, Norman K, Steinhagen-Thiessen E, Demuth I, Eckardt R. Lung function in elderly subjects with metabolic syndrome and type II diabetes : Data from the Berlin Aging Study II. Z Gerontol Geriatr 2015:10.1007/s00391-015-0958-0. [PMID: 26508108 DOI: 10.1007/s00391-015-0958-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 07/15/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Previous studies have indicated a relationship between type II diabetes (T2D), metabolic syndrome (MetS) and pulmonary function but the pathological mechanism responsible remains unclear. The aim of the current analysis within the Berlin Aging Study II (BASE-II) was to investigate the influence of abdominal obesity and muscle mass on pulmonary function in subjects with T2D and MetS. MATERIAL AND METHODS A prebronchodilator pulmonary function test was carried out in 1369 subjects from the BASE-II (mean age 69 ± 4 years, 51.6 % women) where T2D was defined according to the German Diabetes Association (DDG) criteria, MetS according to the criteria of the International Diabetes Foundation (IDF), American Heart Association (AHA) and National Heart, Lung and Blood Institute (NHLBI) criteria from 2009 and pulmonary obstruction (obstructive lung disease, OLD) by the Global Initiative for Chronic Obstructive Lung Disease (GOLD) criteria of a forced expiratory volume in 1 s (FEV1 and forced vital capacity (FVC) ratio < 70 %. RESULTS Of the subjects 50.9 % achieved a sufficient level of quality according to the GOLD guidelines and were analyzed with respect to the research question. The FEV1 and FVC were decreased in study participants with T2D and MetS and the lung volume decreased with an increasing number of MetS criteria. Parameters of body composition, such as waist circumference and muscle mass had a significant influence on lung volumes, independent of MetS or T2D. DISCUSSION In this study MetS and T2D were associated with decreased lung volumes; however, muscle mass and abdominal obesity proved to be the most important factors influencing pulmonary function and could thus form the link between pulmonary function and MetS or T2D. Measurement of grip strength for the determination of muscle mass and waist circumference for determining abdominal obesity could contribute to the interpretation of the results of pulmonary function tests.
Collapse
Affiliation(s)
- Nikolaus Buchmann
- Research Group on Geriatrics, Charité - Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13347, Berlin, Germany.
| | - Kristina Norman
- Research Group on Geriatrics, Charité - Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13347, Berlin, Germany
| | | | - Ilja Demuth
- Research Group on Geriatrics, Charité - Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13347, Berlin, Germany
- Institute of Medical and Human Genetics, Charité - Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13347, Berlin, Germany
| | - Rahel Eckardt
- Research Group on Geriatrics, Charité - Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13347, Berlin, Germany
| |
Collapse
|
336
|
Buchmann N, Norman K, Steinhagen-Thiessen E, Demuth I, Eckardt R. Lungenfunktion bei älteren Probanden mit metabolischem Syndrom und Typ-2-Diabetes. Z Gerontol Geriatr 2015; 49:405-15. [DOI: 10.1007/s00391-015-0959-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 07/15/2015] [Indexed: 01/01/2023]
|
337
|
Obeid J, Nguyen T, Cellucci T, Larché MJ, Timmons BW. Effects of acute exercise on circulating endothelial and progenitor cells in children and adolescents with juvenile idiopathic arthritis and healthy controls: a pilot study. Pediatr Rheumatol Online J 2015; 13:41. [PMID: 26458943 PMCID: PMC4604015 DOI: 10.1186/s12969-015-0038-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/30/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Youth with juvenile idiopathic arthritis (JIA) may be at risk of poor cardiovascular health. Circulating endothelial progenitor cells (EPCs) and circulating endothelial cells (CECs) are markers of cardiovascular repair and damage, respectively, and respond to exercise. The objectives of this study were to compare resting levels of EPCs and CECs in JIA and controls, and to assess the effects of distinct types of exercise on EPCs and CECs in JIA and controls. METHODS Seven youth with JIA and six controls completed 3 visits. First, aerobic fitness was assessed. Participants then performed either moderate intensity, continuous exercise (MICE) or high intensity, intermittent exercise (HIIE) on separate days. Blood samples were collected at the beginning (REST), mid-point (MID) and end of exercise (POST) for determination of EPCs (CD31(+)CD34(bright)CD45(dim)CD133(+)) and CECs (CD31(bright)CD34(+)CD45(-)CD133(-)) by flow cytometry. Between group differences in EPCs and CECs were examined using two-way ANOVA, followed by Tukey's HSD post hoc, where appropriate. Statistical significance set at p ≤ 0.05. RESULTS Both EPCs and CECs were similar between groups at REST (p = 0.18-0.94). During MICE, EPCs remained unchanged in JIA (p = 0.95) but increased significantly at POST in controls (REST: 0.91 ± 0.55 × 10(6) cells/L vs. POST: 1.53 ± 0.36 × 10(6) cells/L, p = 0.04). Compared with controls, lower levels of EPCs were observed in JIA at MID (0.48 ± 0.50 × 10(6) cells/L vs. 1.10 ± 0.39 × 10(6) cells/L, p = 0.01) and POST (0.38 ± 0.34 × 10(6) cells/L vs. 1.53 ± 0.36 × 10(6) cells/L, p < 0.001) during MICE. No changes were detected in CECs with MICE in JIA and controls (p = 0.69). Neither EPCs nor CECs were modified with HIIE (p = 0.28-0.69). CONCLUSION Youth with JIA demonstrated a blunted EPC response to MICE when compared with controls. Future work should examine factors that may increase or normalize EPC mobilization in JIA.
Collapse
Affiliation(s)
- Joyce Obeid
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Thanh Nguyen
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Tania Cellucci
- Department of Pediatrics, Division of Rheumatology, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Maggie J. Larché
- Departments of Medicine and Pediatrics, Division of Rheumatology, McMaster University, Hamilton, ON Canada ,Department of Medicine, Charlton Medical Centre, 25 Charlton Ave E, Suite 702, Hamilton, L8N1Y2 ON Canada
| | - Brian W. Timmons
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1 ON Canada ,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1 ON Canada
| |
Collapse
|
338
|
Sradnick J, Rong S, Luedemann A, Parmentier SP, Bartaun C, Todorov VT, Gueler F, Hugo CP, Hohenstein B. Extrarenal Progenitor Cells Do Not Contribute to Renal Endothelial Repair. J Am Soc Nephrol 2015; 27:1714-26. [PMID: 26453608 DOI: 10.1681/asn.2015030321] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/18/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) may be relevant contributors to endothelial cell (EC) repair in various organ systems. In this study, we investigated the potential role of EPCs in renal EC repair. We analyzed the major EPC subtypes in murine kidneys, blood, and spleens after induction of selective EC injury using the concanavalin A/anti-concanavalin A model and after ischemia/reperfusion (I/R) injury as well as the potential of extrarenal cells to substitute for injured local EC. Bone marrow transplantation (BMTx), kidney transplantation, or a combination of both were performed before EC injury to allow distinction of extrarenal or BM-derived cells from intrinsic renal cells. During endothelial regeneration, cells expressing markers of endothelial colony-forming cells (ECFCs) were the most abundant EPC subtype in kidneys, but were not detected in blood or spleen. Few cells expressing markers of EC colony-forming units (EC-CFUs) were detected. In BM chimeric mice (C57BL/6 with tandem dimer Tomato-positive [tdT+] BM cells), circulating and splenic EC-CFUs were BM-derived (tdT+), whereas cells positive for ECFC markers in kidneys were not. Indeed, most BM-derived tdT+ cells in injured kidneys were inflammatory cells. Kidneys from C57BL/6 donors transplanted into tdT+ recipients with or without prior BMTx from C57BL/6 mice were negative for BM-derived or extrarenal ECFCs. Overall, extrarenal cells did not substitute for any intrinsic ECs. These results demonstrate that endothelial repair in mouse kidneys with acute endothelial lesions depends exclusively on local mechanisms.
Collapse
Affiliation(s)
- Jan Sradnick
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Song Rong
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Anika Luedemann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Simon P Parmentier
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Christoph Bartaun
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Vladimir T Todorov
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Faikah Gueler
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Christian P Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Bernd Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| |
Collapse
|
339
|
Protective effect of Astragalus polysaccharide on endothelial progenitor cells injured by thrombin. Int J Biol Macromol 2015; 82:711-8. [PMID: 26434517 DOI: 10.1016/j.ijbiomac.2015.09.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/27/2015] [Accepted: 09/28/2015] [Indexed: 01/22/2023]
Abstract
Several studies have demonstrated that Astragalus polysaccharide (APS) has a protective effect on endothelial cells damaged by various factors. To examine the role of APS in the endothelial inflammatory response, rat bone marrow endothelial progenitor cells (EPCs) were isolated by density gradient centrifugation and identified by immunohistochemistry, then we established a model of inflammatory injury induced by thrombin and measured the effects of APS on EPC viability and proliferation by MTT assays. We also assayed the effect APS had on the inflammatory response, by examining the nuclear factor kappa B (NF-κB) signaling pathway, as well as the expression of intercellular adhesion molecule-1 (ICAM-1), vascular endothelial growth factor (VEGF) and its receptors Flt-1 and KDR. Results demonstrated that EPCs were damaged by thrombin, and APS appeared to inhibit this damage. APS suppressed thrombin-induced ICAM-1 expression by blocking NF-κB signaling in rat bone marrow EPCs, and up-regulating expression of VEGF and its receptors. We believed that APS may be used to treat and prevent EPC injury-related diseases.
Collapse
|
340
|
Full GMP-compliant validation of bone marrow-derived human CD133(+) cells as advanced therapy medicinal product for refractory ischemic cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:473159. [PMID: 26495296 PMCID: PMC4606188 DOI: 10.1155/2015/473159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/11/2015] [Indexed: 11/24/2022]
Abstract
According to the European Medicine Agency (EMA) regulatory frameworks, Advanced Therapy Medicinal Products (ATMP) represent a new category of drugs in which the active ingredient consists of cells, genes, or tissues. ATMP-CD133 has been widely investigated in controlled clinical trials for cardiovascular diseases, making CD133+ cells one of the most well characterized cell-derived drugs in this field. To ensure high quality and safety standards for clinical use, the manufacturing process must be accomplished in certified facilities following standard operative procedures (SOPs). In the present work, we report the fully compliant GMP-grade production of ATMP-CD133 which aims to address the treatment of chronic refractory ischemic heart failure. Starting from bone marrow (BM), ATMP-CD133 manufacturing output yielded a median of 6.66 × 106 of CD133+ cells (range 2.85 × 106–30.84 × 106), with a viability ranged between 96,03% and 99,97% (median 99,87%) and a median purity of CD133+ cells of 90,60% (range 81,40%–96,20%). Based on these results we defined our final release criteria for ATMP-CD133: purity ≥ 70%, viability ≥ 80%, cellularity between 1 and 12 × 106 cells, sterile, and endotoxin-free. The abovementioned criteria are currently applied in our Phase I clinical trial (RECARDIO Trial).
Collapse
|
341
|
Joo HJ, Song S, Seo HR, Shin JH, Choi SC, Park JH, Yu CW, Hong SJ, Lim DS. Human endothelial colony forming cells from adult peripheral blood have enhanced sprouting angiogenic potential through up-regulating VEGFR2 signaling. Int J Cardiol 2015; 197:33-43. [DOI: 10.1016/j.ijcard.2015.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 05/03/2015] [Accepted: 06/12/2015] [Indexed: 12/27/2022]
|
342
|
Ma F, Morancho A, Montaner J, Rosell A. Endothelial progenitor cells and revascularization following stroke. Brain Res 2015; 1623:150-9. [DOI: 10.1016/j.brainres.2015.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 01/02/2023]
|
343
|
DeJarnett N, Yeager R, Conklin DJ, Lee J, O'Toole TE, McCracken J, Abplanalp W, Srivastava S, Riggs DW, Hamzeh I, Wagner S, Chugh A, DeFilippis A, Ciszewski T, Wyatt B, Becher C, Higdon D, Ramos KS, Tollerud DJ, Myers JA, Rai SN, Shah J, Zafar N, Krishnasamy SS, Prabhu SD, Bhatnagar A. Residential Proximity to Major Roadways Is Associated With Increased Levels of AC133+ Circulating Angiogenic Cells. Arterioscler Thromb Vasc Biol 2015; 35:2468-77. [PMID: 26293462 DOI: 10.1161/atvbaha.115.305724] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 08/04/2015] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Previous studies have shown that residential proximity to a roadway is associated with increased cardiovascular disease risk. Yet, the nature of this association remains unclear, and its effect on individual cardiovascular disease risk factors has not been assessed. The objective of this study was to determine whether residential proximity to roadways influences systemic inflammation and the levels of circulating angiogenic cells. APPROACH AND RESULTS In a cross-sectional study, cardiovascular disease risk factors, blood levels of C-reactive protein, and 15 antigenically defined circulating angiogenic cell populations were measured in participants (n=316) with moderate-to-high cardiovascular disease risk. Attributes of roadways surrounding residential locations were assessed using geographic information systems. Associations between road proximity and cardiovascular indices were analyzed using generalized linear models. Close proximity (<50 m) to a major roadway was associated with lower income and higher rates of smoking but not C-reactive protein levels. After adjustment for potential confounders, the levels of circulating angiogenic cells in peripheral blood were significantly elevated in people living in close proximity to a major roadway (CD31(+)/AC133(+), AC133(+), CD34(+)/AC133(+), and CD34(+)/45(dim)/AC133(+) cells) and positively associated with road segment distance (CD31(+)/AC133(+), AC133(+), and CD34(+)/AC133(+) cells), traffic intensity (CD31(+)/AC133(+) and AC133(+) cells), and distance-weighted traffic intensity (CD31(+)/34(+)/45(+)/AC133(+) cells). CONCLUSIONS Living close to a major roadway is associated with elevated levels of circulating cells positive for the early stem marker AC133(+). This may reflect an increased need for vascular repair. Levels of these cells in peripheral blood may be a sensitive index of cardiovascular injury because of residential proximity to roadways.
Collapse
Affiliation(s)
- Natasha DeJarnett
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Ray Yeager
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Daniel J Conklin
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Jongmin Lee
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Timothy E O'Toole
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - James McCracken
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Wes Abplanalp
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Sanjay Srivastava
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Daniel W Riggs
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Ihab Hamzeh
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Stephen Wagner
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Atul Chugh
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Andrew DeFilippis
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Tiffany Ciszewski
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Brad Wyatt
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Carrie Becher
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Deirdre Higdon
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Kenneth S Ramos
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - David J Tollerud
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - John A Myers
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Shesh N Rai
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Jasmit Shah
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Nagma Zafar
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Sathya S Krishnasamy
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Sumanth D Prabhu
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| | - Aruni Bhatnagar
- From the Diabetes and Obesity Center (N.D., R.Y., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., A.C., A.D., T.C., B.W., S.N.R., J.S., N.Z., S.S.K., A.B.), Department of Environmental and Occupational Health Sciences (N.D., R.Y., D.J.T.), Institute of Molecular Cardiology (N.D., D.J.C., J.L., T.E.O., J.M., W.A., S.S., D.W.R., S.W., A.C., A.D., T.C., B.W., C.B., D.H., J.S., S.D.P., A.B.), Department of Pediatrics (J.A.M.), Department of Bioinformatics and Biostatics (S.N.R.), Biostatistics Shared Facility, JG Brown Cancer Center (S.N.R.), Division of Endocrinology, Metabolism and Diabetes, Department of Medicine (S.S.K.), and Department of Biochemistry and Molecular Biology (K.S.R., A.B.), University of Louisville, KY; Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (I.H.); Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.); and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (S.D.P.)
| |
Collapse
|
344
|
Maroun-Eid C, Ortega-Hernández A, Abad M, García-Donaire JA, Barbero A, Reinares L, Martell-Claros N, Gómez-Garre D. [Circulating endothelial progenitor cell levels in treated hypertensive patients]. HIPERTENSION Y RIESGO VASCULAR 2015; 32:142-50. [PMID: 26486462 DOI: 10.1016/j.hipert.2015.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/02/2015] [Accepted: 07/13/2015] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Most optimally treated hypertensive patients still have an around 50% increased risk of any cardiovascular event, suggesting the possible existence of unidentified risk factors. In the last years there has been evidence of the essential role of circulating endothelial progenitor cells (EPCs) in the maintenance of endothelial integrity and function, increasing the interest in their involvement in cardiovascular disease. In this study, the circulating levels of EPCs and vascular endothelial growth factor (VEGF) are investigated in treated hypertensive patients with adequate control of blood pressure (BP). MATERIAL AND METHODS Blood samples were collected from treated hypertensive patients with controlled BP. Plasma levels of EPCs CD34+/KDR+ and CD34+/VE-cadherin+ were quantified by flow cytometry. Plasma concentration of VEGF was determined by ELISA. A group of healthy subjects without cardiovascular risk factors was included as controls. RESULTS A total of 108 hypertensive patients were included (61±12 years, 47.2% men) of which 82.4% showed BP<140/90 mmHg, 91.7% and 81.5% controlled diabetes (HbA1c <7%) and cLDL (<130 or 100 mg/dL), respectively, and 85.2% were non-smokers. Around 45% of them were obese. Although patients had cardiovascular parameters within normal ranges, they showed significantly lower levels of CD34+/KDR+ and CD34+/VE-cadherin+ compared with healthy control group, although plasma VEGF concentration was higher in patients than in controls. CONCLUSIONS Despite an optimal treatment, hypertensive patients show a decreased number of circulating EPCs that could be, at least in part, responsible for their residual cardiovascular risk, suggesting that these cells could be a therapeutic target.
Collapse
Affiliation(s)
- C Maroun-Eid
- Unidad de Hipertensión, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, España
| | - A Ortega-Hernández
- Laboratorio de Biología Vascular, Hospital Clínico San Carlos-IdISSC, Madrid, España
| | - M Abad
- Unidad de Hipertensión, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, España
| | - J A García-Donaire
- Unidad de Hipertensión, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, España
| | - A Barbero
- Unidad de Hipertensión, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, España
| | - L Reinares
- Unidad de Lípidos, Hospital Clínico San Carlos-IdISSC, Madrid, España
| | - N Martell-Claros
- Unidad de Hipertensión, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, España
| | - D Gómez-Garre
- Laboratorio de Biología Vascular, Hospital Clínico San Carlos-IdISSC, Madrid, España.
| |
Collapse
|
345
|
Lo Gullo A, Mandraffino G, Bagnato G, Aragona CO, Imbalzano E, D’Ascola A, Rotondo F, Cinquegrani A, Mormina E, Saitta C, Versace AG, Sardo MA, Lo Gullo R, Loddo S, Saitta A. Vitamin D Status in Rheumatoid Arthritis: Inflammation, Arterial Stiffness and Circulating Progenitor Cell Number. PLoS One 2015; 10:e0134602. [PMID: 26241902 PMCID: PMC4524708 DOI: 10.1371/journal.pone.0134602] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/12/2015] [Indexed: 12/11/2022] Open
Abstract
Background and Aims Suboptimal vitamin D status was recently acknowledged as an independent predictor of cardiovascular diseases and all-cause mortality in several clinical settings, and its serum levels are commonly reduced in Rheumatoid Arthritis (RA). Patients affected by RA present accelerated atherosclerosis and increased cardiovascular morbidity and mortality with respect to the general population. In RA, it has been reported an impairment of the number and the activity of circulating proangiogenic haematopoietic cells (PHCs), including CD34+, that may play a role in endothelial homeostasis. The purpose of the study is to investigate the association between vitamin D levels and PHCs, inflammatory markers, and arterial stiffening in patients with RA. Methods and Results CD34+ cells were isolated from 27 RA patients and 41 controls. Vitamin D levels, C-reactive protein (CRP), fibrinogen, pulse wave velocity (PWV), and carotid intima-media thickness (cIMT) were also evaluated. CD34+ count and vitamin D levels were lower in RA patients as compared to controls, while fibrinogen, CRP, PWV and cIMT were higher in RA patients. CD34+ cell number appeared to be associated with vitamin D levels, and negatively correlated to fibrinogen and early atherosclerosis markers (PWV and cIMT); vitamin D levels appear also to be inversely associated to fibrinogen. Conclusions RA patients with moderate disease activity presented with low vitamin D levels, low CD34+ cell count, increased PWV and cIMT; we found that vitamin D deficiency is associated to CD34+ cell reduction in peripheral blood, and with fibrinogen levels. This suggests that vitamin D might contribute to endothelial homeostasis in patients with RA.
Collapse
Affiliation(s)
- Alberto Lo Gullo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- * E-mail:
| | - Gianluca Bagnato
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Angela D’Ascola
- Department of Biochemical, Physiological and Nutritional Sciences, University of Messina, Messina, Italy
| | - Francesco Rotondo
- Department of General Surgery and Oncology, University of Messina, Messina, Italy
| | - Antonella Cinquegrani
- Department of Biomedical Sciences and of Morphologic and Functional Images, University of Messina, Messina, Italy
| | - Enricomaria Mormina
- Department of Biomedical Sciences and of Morphologic and Functional Images, University of Messina, Messina, Italy
| | - Carlo Saitta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Maria Adriana Sardo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Renato Lo Gullo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Saverio Loddo
- Department of Biochemical, Physiological and Nutritional Sciences, University of Messina, Messina, Italy
| | - Antonino Saitta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
346
|
Fadini GP, Fiala M, Cappellari R, Danna M, Park S, Poncina N, Menegazzo L, Albiero M, DiPersio J, Stockerl-Goldstein K, Avogaro A. Diabetes Limits Stem Cell Mobilization Following G-CSF but Not Plerixafor. Diabetes 2015; 64:2969-77. [PMID: 25804941 PMCID: PMC4512229 DOI: 10.2337/db15-0077] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/18/2015] [Indexed: 12/19/2022]
Abstract
Previous studies suggest that diabetes impairs hematopoietic stem cell (HSC) mobilization in response to granulocyte colony-stimulating factor (G-CSF). In this study, we tested whether the CXCR4 antagonist plerixafor, differently from G-CSF, is effective in mobilizing HSCs in patients with diabetes. In a prospective study, individuals with and without diabetes (n = 10/group) were administered plerixafor to compare CD34(+) HSC mobilization; plerixafor was equally able to mobilize CD34(+) HSCs in the two groups, whereas in historical data, G-CSF was less effective in patients with diabetes. In a retrospective autologous transplantation study conducted on 706 patients, diabetes was associated with poorer mobilization in patients who received G-CSF with/without chemotherapy, whereas it was not in patients who received G-CSF plus plerixafor. Similarly in an allogeneic transplantation study (n = 335), diabetes was associated with poorer mobilization in patients who received G-CSF. Patients with diabetes who received G-CSF without plerixafor had a lower probability of reaching >50/μL CD34(+) HSCs, independent from confounding variables. In conclusion, diabetes negatively impacted HSC mobilization after G-CSF with or without chemotherapy but had no effect on mobilization induced by G-CSF with plerixafor. This finding has major implications for the care of patients with diabetes undergoing stem cell mobilization and transplantation and for the vascular regenerative potential of bone marrow stem cells.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Mark Fiala
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Roberta Cappellari
- Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marianna Danna
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy
| | - Soo Park
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Nicol Poncina
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Lisa Menegazzo
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Mattia Albiero
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - John DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Keith Stockerl-Goldstein
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Angelo Avogaro
- Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
347
|
Kim J, Kim M, Jeong Y, Lee WB, Park H, Kwon JY, Kim YM, Hwang D, Kwon YG. BMP9 Induces Cord Blood-Derived Endothelial Progenitor Cell Differentiation and Ischemic Neovascularization via ALK1. Arterioscler Thromb Vasc Biol 2015; 35:2020-31. [PMID: 26229139 DOI: 10.1161/atvbaha.115.306142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/20/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Modulating endothelial progenitor cells (EPCs) is essential for therapeutic angiogenesis, and thus various clinical trials involving EPCs are ongoing. However, the identification of environmental conditions and development of optimal methods are required to accelerate EPC-driven vasculogenesis. APPROACH AND RESULTS We evaluated gene expression profiles of cord blood-derived EPCs and endothelial cells to identify the key factors in EPC→endothelial cell differentiation and to show that transforming growth factor-β family members contribute to EPC differentiation. The expression levels of activin receptor-like kinase 1 (ALK1) and its high-affinity ligand, bone morphogenetic protein 9 (BMP9) were markedly changed in EPC→endothelial cell differentiation. Interestingly, BMP9 induced EPC→endothelial cell differentiation and EPC incorporation into vessel-like structures by acting on ALK1 expressed on EPCs in vitro. BMP9 also induced neovascularization in mice with hindlimb ischemia by increasing vessel formation and the incorporation of EPCs into vessels. Conversely, neovascularization was impaired when ALK1 signaling was blocked. Furthermore, EPCs exposed to either short- or long-term BMP9 stimulation demonstrated these functions in EPC-mediated neovascularization. CONCLUSIONS Collectively, our results indicated that BMP9/ALK1 augmented vasculogenesis and angiogenesis, and thereby enhanced neovascularization. Thus, we suggest that BMP9/ALK1 may improve the efficacy of EPC-based therapies for treating ischemic diseases.
Collapse
Affiliation(s)
- Jihye Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Minhyung Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Yoonjeong Jeong
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Wook-Bin Lee
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Hyojin Park
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Ja-Young Kwon
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Young-Myeong Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Daehee Hwang
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Young-Guen Kwon
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.).
| |
Collapse
|
348
|
Kanayasu-Toyoda T, Tanaka T, Ishii-Watabe A, Kitagawa H, Matsuyama A, Uchida E, Yamaguchi T. Angiogenic Role of MMP-2/9 Expressed on the Cell Surface of Early Endothelial Progenitor Cells/Myeloid Angiogenic Cells. J Cell Physiol 2015; 230:2763-75. [DOI: 10.1002/jcp.25002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/24/2015] [Indexed: 02/01/2023]
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Division of Biological Chemistry and Biologicals; National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku; Tokyo Japan
| | - Takeshi Tanaka
- Nihon Pharmaceutical University; Komuro 10281, Inamachi, Kitaadachigun; Saitama Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals; National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku; Tokyo Japan
| | - Hiroko Kitagawa
- Division of Biological Chemistry and Biologicals; National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku; Tokyo Japan
| | - Akifumi Matsuyama
- National Institute of Biomedical Innovation; 7-6-8 Asagi Saito Ibaraki-City; Osaka Japan
| | - Eriko Uchida
- Division of Cellular Gene Therapy Products; National Institute of Health Sciences; Tokyo Japan
| | - Teruhide Yamaguchi
- Division of Biological Chemistry and Biologicals; National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku; Tokyo Japan
| |
Collapse
|
349
|
Gu W, Sun W, Guo C, Yan Y, Liu M, Yao X, Yang B, Zheng J. Culture and Characterization of Circulating Endothelial Progenitor Cells in Patients with Renal Cell Carcinoma. J Urol 2015; 194:214-22. [PMID: 25659661 DOI: 10.1016/j.juro.2015.01.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Wenyu Gu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wei Sun
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yang Yan
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Min Liu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Junhua Zheng
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
350
|
Shih YT, Wang MC, Zhou J, Peng HH, Lee DY, Chiu JJ. Endothelial progenitors promote hepatocarcinoma intrahepatic metastasis through monocyte chemotactic protein-1 induction of microRNA-21. Gut 2015; 64:1132-47. [PMID: 24939570 DOI: 10.1136/gutjnl-2013-306302] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 05/27/2014] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Endothelial progenitor cells (EPCs) circulate with increased numbers in the peripheral blood of patients with highly-vascularised hepatocellular carcinoma (HCC) and contribute to angiogenesis and neovascularisation. We hypothesised that angiogenic EPCs, that is, colony forming unit-endothelial cells (CFU-ECs), and outgrowth EPCs, that is, endothelial colony-forming cells, may exert paracrine effects on the behaviours and metastatic capacities of human hepatoma cells. DESIGN Various molecular and functional approaches ranging from in vitro cell culture studies on molecular signalling to in vivo investigations on cell invasion and orthotropic transplantation models in mice and clinical specimens from patients with HCC were used. RESULTS Monocyte chemotactic protein-1 (MCP-1) was identified as a critical mediator released from CFU-ECs to contribute to the chemotaxis of Huh7 and Hep3B cells by inducing their microRNA-21 (miR-21) biogenesis through the C-C chemokine receptor-2/c-Jun N-terminal kinase/activator protein-1 signalling cascade. CFU-EC-induction of miR-21 in these cells activated their Rac1 and matrix metallopeptidase-9 by silencing Rho GTPase-activating protein-24 and tissue inhibitor of metalloproteinase-3, respectively, leading to increased cell mobility. MCP-1-induction of miR-21 induced epithelial-mesenchymal transformation of Huh7 cells in vitro and their intrahepatic metastatic capability in vivo. Moreover, increased numbers of MCP-1(+) EPCs and their positive correlations with miR-21 induction and metastatic stages in human HCC were found. CONCLUSIONS Our results provide new insights into the complexity of EPC-HCC interactions and indicate that anticancer therapies targeting either the MCP-1 released from angiogenic EPCs or the miR-21 biogenesis in HCC cells may prevent the malignant progression of primary tumours.
Collapse
Affiliation(s)
- Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Cun Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jing Zhou
- Department of Physiology and Pathophysiology, Basic Medical College, Peking University, Beijing, China Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA
| | - Hsin-Hsin Peng
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Ding-Yu Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|