201
|
Abstract
Improvements in the care of patients with ischemic cardiovascular disease have led to improved survival but also a burgeoning population of patients with advanced ischemic heart disease. Cell therapies offer a novel approach toward cardiac "rejuvenation" via stimulation of new blood vessel growth, enhancing tissue perfusion, and via preservation or even regeneration of myocardial tissue, leading to improvements in cardiac performance after myocardial infarction and in patients with advanced heart failure. Here, we summarize and offer some thoughts on the state of the field of cell therapy for ischemic heart disease, targeting three separate conditions that have been the subject of significant clinical research: enhancing left ventricular recovery after MI, improving outcomes and symptoms in patients with congestive heart failure (CHF), and treatment of patients with refractory angina, despite maximal medical therapy.
Collapse
Affiliation(s)
- Thomas J Povsic
- Duke Clinical Research Institute and Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
202
|
Jagged-1 Signaling in the Bone Marrow Microenvironment Promotes Endothelial Progenitor Cell Expansion and Commitment of CD133+ Human Cord Blood Cells for Postnatal Vasculogenesis. PLoS One 2016; 11:e0166660. [PMID: 27846321 PMCID: PMC5112804 DOI: 10.1371/journal.pone.0166660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/01/2016] [Indexed: 01/23/2023] Open
Abstract
Notch signaling is involved in cell fate decisions during murine vascular development and hematopoiesis in the microenvironment of bone marrow. To investigate the close relationship between hematopoietic stem cells and human endothelial progenitor cells (EPCs) in the bone marrow niche, we examined the effects of Notch signals [Jagged-1 and Delta-like ligand (Dll)-1] on the proliferation and differentiation of human CD133+ cell-derived EPCs. We established stromal systems using HESS-5 murine bone marrow cells transfected with human Jagged-1 (hJagged-1) or human Dll-1 (hDll-1). CD133+ cord blood cells were co-cultured with the stromal cells for 7 days, and then their proliferation, differentiation, and EPC colony formation was evaluated. We found that hJagged-1 induced the proliferation and differentiation of CD133+ cord blood EPCs. In contrast, hDll-1 had little effect. CD133+ cells stimulated by hJagged-1 differentiated into CD31+/KDR+ cells, expressed vascular endothelial growth factor-A, and showed enhanced EPC colony formation compared with CD133+ cells stimulated by hDll-1. To evaluate the angiogenic properties of hJagged-1- and hDll-1-stimulated EPCs in vivo, we transplanted these cells into the ischemic hindlimbs of nude mice. Transplantation of EPCs stimulated by hJagged-1, but not hDll-1, increased regional blood flow and capillary density in ischemic hindlimb muscles. This is the first study to show that human Notch signaling influences EPC proliferation and differentiation in the bone marrow microenvironment. Human Jagged-1 induced the proliferation and differentiation of CD133+ cord blood progenitors compared with hDll-1. Thus, hJagged-1 signaling in the bone marrow niche may be used to expand EPCs for therapeutic angiogenesis.
Collapse
|
203
|
Abplanalp WT, Conklin DJ, Cantor JM, Ginsberg MH, Wysoczynski M, Bhatnagar A, O'Toole TE. Enhanced Integrin α4β1-Mediated Adhesion Contributes to a Mobilization Defect of Endothelial Progenitor Cells in Diabetes. Diabetes 2016; 65:3505-3515. [PMID: 27495221 PMCID: PMC5079633 DOI: 10.2337/db16-0634] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
Diabetes is associated with a deficit of circulating endothelial progenitor cells (EPCs), which has been attributed to their defective mobilization from the bone marrow. The basis for this mobilization defect is not completely understood, and we sought to determine if hyperglycemic conditions enhanced EPC adhesion. We found that culturing EPCs in high glucose media increased adhesion to bone marrow stromal cells. This enhanced adhesion was associated with decreased expression of protein kinase A regulatory subunit 1β (PRKAR1β), activation of protein kinase A (PKA), and phosphorylation of α4-integrin on serine 988. This potentiated adhesion was reversed by treatment with a PKA inhibitor, overexpression of PRKAR1β, or expression of a phosphorylation-defective α4-integrin variant (α4[S988A]). Using a model of type 1 diabetes, we showed that α4(S988A)-expressing mice have more circulating EPCs than their wild-type counterparts. Moreover, diabetic α4(S988A) mice demonstrate enhanced revascularization after hind limb ischemia. Thus, we have identified a novel signaling mechanism activating PKA in diabetes (downregulation of an inhibitory regulatory subunit) that leads to deficits of circulating EPCs and impaired vascular repair, which could be reversed by α4-integrin mutation.
Collapse
Affiliation(s)
- Wesley T Abplanalp
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
| | - Joseph M Cantor
- Department of Medicine, University of California, San Diego, San Diego, CA
| | - Mark H Ginsberg
- Department of Medicine, University of California, San Diego, San Diego, CA
| | | | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Timothy E O'Toole
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
| |
Collapse
|
204
|
Xiang W, Hu ZL, He XJ, Dang XQ. Intravenous transfusion of endothelial progenitor cells that overexpress vitamin D receptor inhibits atherosclerosis in apoE-deficient mice. Biomed Pharmacother 2016; 84:1233-1242. [PMID: 27810779 DOI: 10.1016/j.biopha.2016.10.042] [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: 08/08/2016] [Revised: 10/07/2016] [Accepted: 10/17/2016] [Indexed: 10/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are widely used for angiogenic therapies, as well as predictive biomarkers to assess cardiovascular disease risk. However, it is unknown that whether overexpressed vitamin D receptor (VDR) in EPCs could help EPCs counteracting atherosclerotic risks. Here, we study intravenous transplantation of genetically modified EPCs over-expressing VDR in regulating endothelial dysfunction and spontaneously arising atherosclerotic plaques of ApoE-deficient mice. Firstly, we found that over-expression of VDR in EPCs could reduce atherosclerotic plaque formation in transplanted ApoE-/- mice. In addition, the concentration of serum HDL-C in ovVDR-EPCs group was much higher than that in control groups (ApoE-/- mice without injection or injected with fresh medium or adenovirus vector). While concentrations of serum total cholesterol, LDL-C, apoB and Lp (a) were negatively correlated with the expression level of VDR. What's more, improved serum concentration of NO and elevated serum and vessel wall expression of eNOS were observed in ovVDR-EPCs group. Furthermore, reduced expression and activity of MMP2, and elevated expression and activity of TIMP2 were detected in ovVDR-EPCs group. Taken together, intravenous transfusion of EPCs that overexpress VDR significantly inhibited atherosclerosis in ApoE-deficient mice and could be used as a potential method for angiogenic therapy.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Pediatrics, Hainan Provincial Maternal Hospital, Hainan province, 570006, China
| | - Zhi-Lan Hu
- Department of Nephropathy, Children's Medical Center, The Second Xiangya Hospital, Central South University, Hunan province, 410000, China
| | - Xiao-Jie He
- Department of Nephropathy, Children's Medical Center, The Second Xiangya Hospital, Central South University, Hunan province, 410000, China.
| | - Xi-Qiang Dang
- Department of Nephropathy, Children's Medical Center, The Second Xiangya Hospital, Central South University, Hunan province, 410000, China
| |
Collapse
|
205
|
Paek SJ, Park WJ, Shin HS, Choi MG, Kwon KH, Choi EJ. Diseases having an influence on inhibition of angiogenesis as risk factors of osteonecrosis of the jaw. J Korean Assoc Oral Maxillofac Surg 2016; 42:271-277. [PMID: 27847735 PMCID: PMC5104869 DOI: 10.5125/jkaoms.2016.42.5.271] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/08/2016] [Accepted: 09/16/2016] [Indexed: 12/13/2022] Open
Abstract
Objectives The objective of this study was to retrospectively investigate the association of diseases having an influence on inhibition of angiogenesis such as hypertension, diabetes mellitus type II, hypercholesterolemia, and rheumatoid arthritis (RA) with the development of osteonecrosis of the jaws. Materials and Methods The 135 patients were allocated into 4 groups of bisphosphonate-related osteonecrosis of the jaw (BRONJ) group (1A); non-BRONJ group (1B); osteonecrosis of the jaw (ONJ) group (2A); and control group (2B), according to histologic results and use of bisphosphonate. This retrospective study was conducted with patients who were treated in one institute from 2012 to 2013. Fisher's exact test and logistic regression analysis were used to analyze the odds ratios of diseases having an influence on inhibition of angiogenesis for development of ONJ. Results The effects of diabetes and hypertension were not statistically significant on development of ONJ. When not considering bisphosphonate use, RA exhibited a high odds ratio of 3.23 (P=0.094), while hyperlipidemia showed an odds ratio of 2.10 (P=0.144) for development of ONJ. More than one disease that had an influence on inhibition of angiogenesis showed a statistically significant odds ratio of 2.54 (P=0.012) for development of ONJ. Conclusion Patients without diseases having an influence on inhibition of angiogenesis were at less risk for developing ONJ.
Collapse
Affiliation(s)
- Seung Jae Paek
- Department of Oral and Maxillofacial Surgery, Wonkwang University Dental Hospital, Wonkwang University, Iksan, Korea
| | - Won-Jong Park
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Ho-Sung Shin
- Department of Society Dentistry, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Moon-Gi Choi
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Kyung-Hwan Kwon
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Eun Joo Choi
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University, Iksan, Korea
| |
Collapse
|
206
|
Wils J, Favre J, Bellien J. Modulating putative endothelial progenitor cells for the treatment of endothelial dysfunction and cardiovascular complications in diabetes. Pharmacol Ther 2016; 170:98-115. [PMID: 27773788 DOI: 10.1016/j.pharmthera.2016.10.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetes induces a decrease in the number and function of different pro-angiogenic cell types generically designated as putative endothelial progenitor cells (EPC), which encompasses cells from myeloid origin that act in a paracrine fashion to promote angiogenesis and putative "true" EPC that contribute to endothelial replacement. This not only compromises neovasculogenesis in ischemic tissues but also impairs, at an early stage, the reendotheliziation process at sites of injury, contributing to the development of endothelial dysfunction and cardiovascular complications. Hyperglycemia, insulin resistance and dyslipidemia promote putative EPC dysregulation by affecting the SDF-1/CXCR-4 and NO pathways and the p53/SIRT1/p66Shc axis that contribute to their mobilization, migration, homing and vasculogenic properties. To optimize the clinical management of patients with hypoglycemic agents, statins and renin-angiotensin system inhibitors, which display pleiotropic effects on putative EPC, is a first step to improve their number and angiogenic potential but specific strategies are needed. Among them, mobilizing therapies based on G-CSF, erythropoietin or CXCR-4 antagonism have been developed to increase putative EPC number to treat ischemic diseases with or without prior cell isolation and transplantation. Growth factors, genetic and pharmacological strategies are also evaluated to improve ex vivo cultured EPC function before transplantation. Moreover, pharmacological agents increasing in vivo the bioavailability of NO and other endothelial factors demonstrated beneficial effects on neovascularization in diabetic ischemic models but their effects on endothelial dysfunction remain poorly evaluated. More experiments are warranted to develop orally available drugs and specific agents targeting p66Shc to reverse putative EPC dysfunction in the expected goal of preventing endothelial dysfunction and diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Julien Wils
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Julie Favre
- MITOVASC Institute, Angers, France; Centre National de la Recherche Scientifique (CNRS) UMR 6214, Angers, France; INSERM U1083, Angers, France; University of Angers, Angers, France
| | - Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
207
|
Nielsen N, Laustsen C, Bertelsen LB. 13C dynamic nuclear polarization for measuring metabolic flux in endothelial progenitor cells. Exp Cell Res 2016; 349:95-100. [PMID: 27720669 DOI: 10.1016/j.yexcr.2016.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022]
Abstract
Endothelial progenitor cells (EPCs) represent a heterogeneous cell population that is believed to be involved in vasculogenesis. With the purpose of enhancing endothelial repair, EPCs could have a potential for future cell therapies. Due to the low amount of EPCs in the peripheral circulating blood, in vitro expansion is needed before administration to recipients and the effects of in vitro culturing is still an under-evaluated field with little knowledge of how the cells change over time in culture. The aim of this study was to use hyperpolarised carbon-13 magnetic resonance spectroscopy to profile important metabolic pathways in a population of progenitor cells and to show that cell culturing in 3D scaffolds seem to block the metabolic processes that leads to cell senescence. The metabolic breakdown of hyperpolarized [1-13C]pyruvate was followed after injection of the substrate to a bioreactor system with EPCs either adhered to 3D printed scaffolds or kept in cell suspension. The pyruvate-to-lactate conversion was elevated in suspension of EPCs compared to the EPCs adhered to scaffolds. Furthermore in the setup with EPCs in suspension, an increase in lactate production was seen over time indicating that the older the cultures of EPCs was before using the cells for cell suspension experiments, the more lactate they produce, compared to a constant lactate level in the cells adhered to scaffolds. It could therefore be stated that cells grown first in 2D culture and subsequent prepared for cell suspension show a metabolism with higher lactate production consistent with cells senescence processes compared to cells grown first at 2D culture and subsequent in the 3D printed scaffolds, where metabolism shows no sign of metabolic shifting during the monitored period.
Collapse
Affiliation(s)
- Nathalie Nielsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Lotte Bonde Bertelsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| |
Collapse
|
208
|
Van Pham P, Vu NB, Truong MTH, Huynh OT, Nguyen HT, Pham HL, Phan NK. Hepatocyte growth factor improves direct reprogramming of fibroblasts towards endothelial progenitor cells via ETV2 transduction. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0045-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
209
|
Tsukada S, Masuda H, Jung SY, Yun J, Kang S, Kim DY, Park JH, Ji ST, Kwon SM, Asahara T. Impaired development and dysfunction of endothelial progenitor cells in type 2 diabetic mice. DIABETES & METABOLISM 2016; 43:154-162. [PMID: 27638126 DOI: 10.1016/j.diabet.2016.07.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/02/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
AIM Dysfunction of circulating endothelial progenitor cells (EPCs) has been shown to affect the development of microvascular diseases in diabetes patients. The aim of this study was to elucidate the development and mechanical dysfunction of EPCs in type 2 diabetes (T2D). METHODS The colony-forming capacity of EPCs and differentiation potential of bone marrow (BM) c-Kit(+)/Sca-I(+) lineage-negative mononuclear cells (KSL) were examined in T2D mice, db/db mice and KKAy mice, using EPC colony-forming assay (EPC-CFA). RESULTS T2D mice had fewer BM stem/progenitor cells, and proliferation of KSL was lowest in the BM of db/db mice. In T2D mice, the frequency of large colony-forming units (CFUs) derived from BM-KSL was highly reduced, indicating dysfunction of differentiation into mature EPCs. Only a small number of BM-derived progenitors [CD34(+) KSL cells], which contribute to the supply of EPCs for postnatal neovascularization, was also found. Furthermore, in terms of their plasticity to transdifferentiate into various cell types, BM-KSL exhibited a greater potential to differentiate into granulocyte macrophages (GMs) than into other cell types. CONCLUSION T2D affected EPC colony formation and differentiation of stem cells to mature EPCs or haematopoietic cells. These data suggest opposing regulatory mechanisms for differentiation into mature EPCs and GMs in T2D mice.
Collapse
Affiliation(s)
- S Tsukada
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan
| | - H Masuda
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan
| | - S Y Jung
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - J Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S Kang
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - D Y Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - J H Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S T Ji
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S-M Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea; Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, Republic of Korea.
| | - T Asahara
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan; Stem Cell Translational Research Laboratory, Center For Developmental Biology, RIKEN, Kobe, Japan; Kobe Institute of Biomedical Research and Innovation, Kobe, Japan.
| |
Collapse
|
210
|
NAMPT regulates senescence, proliferation, and migration of endothelial progenitor cells through the SIRT1 AS lncRNA/miR-22/SIRT1 pathway. Biochem Biophys Res Commun 2016; 478:1382-8. [DOI: 10.1016/j.bbrc.2016.08.133] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 08/24/2016] [Indexed: 11/24/2022]
|
211
|
Yang J, Zhang X, Zhao Z, Li X, Wang X, Chen M, Song B, Ii M, Shen Z. Regulatory roles of interferon-inducible protein 204 on differentiation and vasculogenic activity of endothelial progenitor cells. Stem Cell Res Ther 2016; 7:111. [PMID: 27514835 PMCID: PMC4981987 DOI: 10.1186/s13287-016-0365-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/09/2016] [Accepted: 07/14/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) have shown great potential in angiogenesis either by their differentiation into endothelial cells or by secretion of angiogenic factors. Interferon-inducible protein 204 (Ifi204) has been reported to participate in the regulation of cell growth and differentiation. However, its role in differentiation of EPCs remains unknown. We proposed that Ifi204 could modulate the differentiation and regenerative abilities of EPCs. METHODS Ifi204-expressing lentivirus and Ifi204 siRNA were introduced into EPCs to overexpress and suppress the expression of Ifi204. Using fluorescence-activated cell sorting, immunocytochemistry, and quantitative PCR, endothelial markers including CD31, VE-cadherin, and vWF were detected in the modified EPCs. An in-vitro incorporation assay and a colony-forming assay were also performed. RESULTS Evidence showed that Ifi204 inhibition decreased the endothelial differentiation and vasculogenic activities of EPCs in vitro. In mice with hindlimb ischemia, downregulation of Ifi204 in EPCs, which was tracked by our newly synthesized nanofluorogen, impaired neovascularization, with a corresponding reduction in hindlimb blood reperfusion by postoperative day 14. CONCLUSIONS Ifi204 is required for EPC differentiation and neovascularization in vitro and in vivo. The regulatory roles of Ifi204 in EPC differentiation may benefit the clinical therapy of ischemic vascular diseases.
Collapse
Affiliation(s)
- Junjie Yang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
- Institute for Cardiovascular Science, Soochow University, 708 Renmin Road, Suzhou, 215006 China
| | - Xiaofei Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
| | - Zhenao Zhao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
| | - Xizhe Li
- Department of Cardiovascular Surgery, Affiliated Shanghai 1st People’s Hospital, Shanghai Jiaotong University, Shanghai, 200080 China
| | - Xu Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
| | - Ming Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
| | - Bo Song
- Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215006 China
| | - Masaaki Ii
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686 Japan
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, 188 Shizi Street, Suzhou, 215006 China
| |
Collapse
|
212
|
Significant improvement of direct reprogramming efficacy of fibroblasts into progenitor endothelial cells by ETV2 and hypoxia. Stem Cell Res Ther 2016; 7:104. [PMID: 27488544 PMCID: PMC4973107 DOI: 10.1186/s13287-016-0368-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/18/2016] [Indexed: 12/22/2022] Open
Abstract
Background Endothelial progenitor cell (EPC) transplantation is a promising therapy for ischemic diseases such as ischemic myocardial infarction and hindlimb ischemia. However, limitation of EPC sources remains a major obstacle. Direct reprogramming has become a powerful tool to produce EPCs from fibroblasts. Some recent efforts successfully directly reprogrammed human fibroblasts into functional EPCs; however, the procedure efficacy was low. This study therefore aimed to improve the efficacy of direct reprogramming of human fibroblasts to functional EPCs. Methods Human fibroblasts isolated from foreskin were directly reprogrammed into EPCs by viral ETV2 transduction. Reprogramming efficacy was improved by culturing transduced fibroblasts in hypoxia conditions (5 % oxygen). Phenotype analyses confirmed that single-factor ETV2 transduction successfully reprogrammed dermal fibroblasts into functional EPCs. Results Hypoxia treatment during the reprogramming procedure increased the efficacy of reprogramming from 1.21 ± 0.61 % in normoxia conditions to 7.52 ± 2.31 % in hypoxia conditions. Induced EPCs in hypoxia conditions exhibited functional EPC phenotypes similar to those in normoxia conditions, such as expression of CD31 and VEGFR2, and expressed endothelial gene profiles similar to human umbilical vascular endothelial cells. These cells also formed capillary-like networks in vitro. Conclusion Our study demonstrates a new simple method to increase the reprogramming efficacy of human fibroblasts to EPCs using ETV2 and hypoxia.
Collapse
|
213
|
Endothelial progenitor cells accelerate the resolution of deep vein thrombosis. Vascul Pharmacol 2016; 83:10-6. [DOI: 10.1016/j.vph.2015.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/27/2015] [Accepted: 07/11/2015] [Indexed: 11/23/2022]
|
214
|
Cho H, Balaji S, Hone NL, Moles CM, Sheikh AQ, Crombleholme TM, Keswani SG, Narmoneva DA. Diabetic wound healing in a MMP9-/- mouse model. Wound Repair Regen 2016; 24:829-840. [PMID: 27292154 DOI: 10.1111/wrr.12453] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/12/2016] [Indexed: 12/12/2022]
Abstract
Reduced mobilization of endothelial progenitor cells (EPCs) from the bone marrow (BM) and impaired EPC recruitment into the wound represent a fundamental deficiency in the chronic ulcers. However, mechanistic understanding of the role of BM-derived EPCs in cutaneous wound neovascularization and healing remains incomplete, which impedes development of EPC-based wound healing therapies. The objective of this study was to determine the role of EPCs in wound neovascularization and healing both under normal conditions and using single deficiency (EPC) or double-deficiency (EPC + diabetes) models of wound healing. MMP9 knockout (MMP9 KO) mouse model was utilized, where impaired EPC mobilization can be rescued by stem cell factor (SCF). The hypotheses were: (1) MMP9 KO mice exhibit impaired wound neovascularization and healing, which are further exacerbated with diabetes; (2) these impairments can be rescued by SCF administration. Full-thickness excisional wounds with silicone splints to minimize contraction were created on MMP9 KO mice with/without streptozotocin-induced diabetes in the presence or absence of tail-vein injected SCF. Wound morphology, vascularization, inflammation, and EPC mobilization and recruitment were quantified at day 7 postwounding. Results demonstrate no difference in wound closure and granulation tissue area between any groups. MMP9 deficiency significantly impairs wound neovascularization, increases inflammation, decreases collagen deposition, and decreases peripheral blood EPC (pb-EPC) counts when compared with wild-type (WT). Diabetes further increases inflammation, but does not cause further impairment in vascularization, as compared with MMP9 KO group. SCF improves neovascularization and increases EPCs to WT levels (both nondiabetic and diabetic MMP9 KO groups), while exacerbating inflammation in all groups. SCF rescues EPC-deficiency and impaired wound neovascularization in both diabetic and nondiabetic MMP9 KO mice. Overall, the results demonstrate that BM-derived EPCs play a significant role during wound neovascularization and that the SCF-based therapy with controlled inflammation could be a viable approach to enhance healing in chronic diabetic wounds.
Collapse
Affiliation(s)
- Hongkwan Cho
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Swathi Balaji
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio.,Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Natalie L Hone
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Chad M Moles
- Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Abdul Q Sheikh
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Timothy M Crombleholme
- Children's Hospital Colorado and the University of Colorado School of Medicine, Aurora, Colorado
| | - Sundeep G Keswani
- Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Daria A Narmoneva
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
215
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
216
|
Xia X, Yu Y, Zhang L, Ma Y, Wang H. Inhibitor of DNA binding 1 regulates cell cycle progression of endothelial progenitor cells through induction of Wnt2 expression. Mol Med Rep 2016; 14:2016-24. [PMID: 27432753 PMCID: PMC4991734 DOI: 10.3892/mmr.2016.5491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 06/15/2016] [Indexed: 11/28/2022] Open
Abstract
Endothelial injury is a risk factor for atherosclerosis. Endothelial progenitor cell (EPC) proliferation contributes to vascular injury repair. Overexpression of inhibitor of DNA binding 1 (Id1) significantly promotes EPC proliferation; however, the underlying molecular mechanism remains to be fully elucidated. The present study investigated the role of Id1 in cell cycle regulation of EPCs, which is closely associated with proliferation. Overexpression of Id1 increased the proportion of EPCs in the S/G2M phase and significantly increased cyclin D1 expression levels, while knockdown of Id1 arrested the cell cycle progression of EPCs in the G1 phase and inhibited cyclin D1 expression levels. In addition, it was demonstrated that Id1 upregulated wingless-type mouse mammary tumor virus integration site family member 2 (Wnt2) expression levels and promoted β-catenin accumulation and nuclear translocation. Furthermore, Wnt2 knockdown counteracted the effects of Id1 on cell cycle progression of EPCs. In conclusion, the results of the present study indicate that Id1 promoted Wnt2 expression, which accelerated cell cycle progression from G1 to S phase. This suggests that Id1 may promote cell cycle progression of EPCs, and that Wnt2 may be important in Id1 regulation of the cell cycle of EPCs.
Collapse
Affiliation(s)
- Xi Xia
- Postgraduate Department, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yang Yu
- Department of Cardiology, Institute of Cardiovascular Science of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Li Zhang
- Department of Geriatrics, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnan 650032, P.R. China
| | - Yang Ma
- Department of Geriatrics, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnan 650032, P.R. China
| | - Hong Wang
- Department of Geriatrics, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
217
|
Kidani Y, Miki Y, Nomimura N, Minakawa S, Tanaka N, Miyoshi H, Wakabayashi K, Kudo Y. The therapeutic effect of CD133+ cells derived from human umbilical cord blood on neonatal mouse hypoxic-ischemic encephalopathy model. Life Sci 2016; 157:108-115. [DOI: 10.1016/j.lfs.2016.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/26/2016] [Accepted: 06/04/2016] [Indexed: 12/22/2022]
|
218
|
Gallagher KA, Goldstein LJ, Thom SR, Velazquez OC. Hyperbaric Oxygen and Bone Marrow–Derived Endothelial Progenitor Cells in Diabetic Wound Healing. Vascular 2016; 14:328-37. [PMID: 17150153 DOI: 10.2310/6670.2006.00057] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Endothelial progenitor cells (EPCs) are the key cellular effectors of postnatal vasculogenesis and play a central role in wound healing. In diabetes, there is a significant impairment in the number and function of circulating and wound-tissue EPC. Recent evidence indicates, that tissue-level hyperoxia achieved by therapeutic hyperbaric oxygen protocols (HBO2) can increase the mobilization of EPC from the bone marrow into peripheral blood. In this paper we review the recent reports on hyperoxia-mediated mobilization of bone marrow-derived EPC and postulate avenues of future research in this area as it applies to improving healing in chronic wounds affected by diabetes and peripheral arterial disease (PAD).
Collapse
Affiliation(s)
- Katherine A Gallagher
- Department of Surgery, Institute for Environmental Medicine, University of Pennsylvania Medical Center, Philadelphia 19124, USA
| | | | | | | |
Collapse
|
219
|
Chao TH, Chen IC, Li YH, Lee PT, Tseng SY. Plasma Levels of Proprotein Convertase Subtilisin/Kexin Type 9 Are Elevated in Patients With Peripheral Artery Disease and Associated With Metabolic Disorders and Dysfunction in Circulating Progenitor Cells. J Am Heart Assoc 2016; 5:JAHA.116.003497. [PMID: 27207972 PMCID: PMC4889209 DOI: 10.1161/jaha.116.003497] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is involved in cholesterol homeostasis, inflammation, and oxidative stress. This study investigated the association of plasma PCSK9 levels with the presence and severity of peripheral artery disease (PAD) and with parameters of endothelial homeostasis. METHODS AND RESULTS A post hoc analysis of 2 randomized trials (115 patients, 44 with PAD and 71 without atherosclerotic disease) was conducted. Patients with PAD had significantly higher plasma PCSK9 levels than those without (471.6±29.6 versus 302.4±16.1 ng/mL, P<0.001). Parameters for glucose homeostasis, endothelial progenitor cell functions, apoptotic circulating endothelial cell counts, and plasma levels of vascular endothelial growth factor-A165 and oxidized low-density lipoprotein were correlated with PCSK9 concentration. By multivariable linear regression analysis, presence of PAD, plasma glucose or hemoglobin A1c levels, apoptotic circulating endothelial cell counts, and vascular endothelial growth factor-A165 concentration were found to be associated with PCSK9 levels after multivariable adjustment. Patients with extensive involvement of PAD or with severe PAD had significantly higher PCSK9 levels than those without PAD. Computed tomographic angiography showed that the numbers of chronic total occlusion sites and vessels involved were positively associated with PCSK9 levels in patients with PAD (r=0.40, P=0.01, and r=0.36, P=0.02, respectively). CONCLUSION PCSK9 levels were significantly higher in patients with PAD, especially those with advanced PAD. Further large-scale studies examining the effect of PCSK9-targeting therapies or the modification of PCSK9 levels on cardiovascular outcomes in this clinical setting are warranted. CLINICAL TRIAL REGISTRATION Cohort 1: URL: ClinicalTrials.gov. Unique identifier: NCT01952756; cohort 2: URL: ClinicalTrials.gov. Unique identifier: NCT02194686.
Collapse
Affiliation(s)
- Ting-Hsing Chao
- Department of Internal Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - I-Chih Chen
- Department of Internal Medicine, Tainan Municipal Hospital, Tainan, Taiwan
| | - Yi-Heng Li
- Department of Internal Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Po-Tseng Lee
- Department of Internal Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Shih-Ya Tseng
- Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
220
|
Simsekyilmaz S, Liehn EA, Weinandy S, Schreiber F, Megens RTA, Theelen W, Smeets R, Jockenhövel S, Gries T, Möller M, Klee D, Weber C, Zernecke A. Targeting In-Stent-Stenosis with RGD- and CXCL1-Coated Mini-Stents in Mice. PLoS One 2016; 11:e0155829. [PMID: 27192172 PMCID: PMC4871500 DOI: 10.1371/journal.pone.0155829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/04/2016] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic lesions that critically narrow the artery can necessitate an angioplasty and stent implantation. Long-term therapeutic effects, however, are limited by excessive arterial remodeling. We here employed a miniaturized nitinol-stent coated with star-shaped polyethylenglycole (star-PEG), and evaluated its bio-functionalization with RGD and CXCL1 for improving in-stent stenosis after implantation into carotid arteries of mice. Nitinol foils or stents (bare metal) were coated with star-PEG, and bio-functionalized with RGD, or RGD/CXCL1. Cell adhesion to star-PEG-coated nitinol foils was unaltered or reduced, whereas bio-functionalization with RGD but foremost RGD/CXCL1 increased adhesion of early angiogenic outgrowth cells (EOCs) and endothelial cells but not smooth muscle cells when compared with bare metal foils. Stimulation of cells with RGD/CXCL1 furthermore increased the proliferation of EOCs. In vivo, bio-functionalization with RGD/CXCL1 significantly reduced neointima formation and thrombus formation, and increased re-endothelialization in apoE-/- carotid arteries compared with bare-metal nitinol stents, star-PEG-coated stents, and stents bio-functionalized with RGD only. Bio-functionalization of star-PEG-coated nitinol-stents with RGD/CXCL1 reduced in-stent neointima formation. By supporting the adhesion and proliferation of endothelial progenitor cells, RGD/CXCL1 coating of stents may help to accelerate endothelial repair after stent implantation, and thus may harbor the potential to limit the complication of in-stent restenosis in clinical approaches.
Collapse
Affiliation(s)
- Sakine Simsekyilmaz
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
- Biochemistry Institute, Justus-Liebig-University, Giessen, Germany
| | - Elisa A. Liehn
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
- Human Genetic Laboratory, University of Medicine and Pharmacy, Craiova, Romania
- IZKF Aachen, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Weinandy
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
- Department of Applied Medical Engineering, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Fabian Schreiber
- Institute for Textile Technology, RWTH Aachen University, Aachen, Germany
| | - Remco T. A. Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Munich, Germany
| | - Wendy Theelen
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, Center of Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Jockenhövel
- Department of Applied Medical Engineering, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Thomas Gries
- Institute for Textile Technology, RWTH Aachen University, Aachen, Germany
| | - Martin Möller
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
| | - Doris Klee
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alma Zernecke
- Institute for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
221
|
Aschbacher K, Derakhshandeh R, Flores AJ, Narayan S, Mendes WB, Springer ML. Circulating angiogenic cell function is inhibited by cortisol in vitro and associated with psychological stress and cortisol in vivo. Psychoneuroendocrinology 2016; 67:216-23. [PMID: 26925833 PMCID: PMC4808379 DOI: 10.1016/j.psyneuen.2016.02.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 02/07/2023]
Abstract
Psychological stress and glucocorticoids are associated with heightened cardiovascular disease risk. We investigated whether stress or cortisol would be associated with reduced circulating angiogenic cell (CAC) function, an index of impaired vascular repair. We hypothesized that minority-race individuals who experience threat in interracial interactions would exhibit reduced CAC function, and that this link might be explained by cortisol. To test this experimentally, we recruited 106 African American participants for a laboratory interracial interaction task, in which they received socially evaluative feedback from Caucasian confederates. On a separate day, a subset of 32 participants (mean age=26years, 47% female) enrolled in a separate biological substudy and provided blood samples for CAC isolation and salivary samples to quantify the morning peak in cortisol (the cortisol awakening response, CAR). CAC function was quantified using cell culture assays of migration to vascular endothelial growth factor (VEGF) and secretion of VEGF into the culture medium. Heightened threat in response to an interracial interaction and trait anxiety in vivo were both associated with poorer CAC migratory function in vitro. Further, threat and poorer sustained attention during the interracial interaction were associated with a higher CAR, which in turn, was related to lower CAC sensitivity to glucocorticoids. In vitro, higher doses of cortisol impaired CAC migratory function and VEGF protein secretion. The glucocorticoid receptor antagonist RU486 reversed this functional impairment. These data identify a novel, neuroendocrine pathway by which psychological stress may reduce CAC function, with potential implications for cardiovascular health.
Collapse
Affiliation(s)
- Kirstin Aschbacher
- Department of Psychiatry, University of California, 3333 California Street, San Francisco, CA 94143, United States; The Institute for Integrative Health, 1407 Fleet Street, Baltimore, MD 21231, United States.
| | | | | | | | | | | |
Collapse
|
222
|
Nakamura T, Koga H, Iwamoto H, Tsutsumi V, Imamura Y, Naitou M, Masuda A, Ikezono Y, Abe M, Wada F, Sakaue T, Ueno T, Ii M, Alev C, Kawamoto A, Asahara T, Torimura T. Ex vivo expansion of circulating CD34(+) cells enhances the regenerative effect on rat liver cirrhosis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16025. [PMID: 27162932 PMCID: PMC4847556 DOI: 10.1038/mtm.2016.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/11/2016] [Accepted: 02/16/2016] [Indexed: 02/07/2023]
Abstract
Ex vivo expansion of autologous cells is indispensable for cell transplantation therapy of patients with liver cirrhosis. The aim of this study was to investigate the efficacy of human ex vivo-expanded CD34+ cells for treatment of cirrhotic rat liver. Recipient rats were intraperitoneally injected with CCl4 twice weekly for 3 weeks before administration of CD34+ cells. CCl4 was then re-administered twice weekly for 3 more weeks, and the rats were sacrificed. Saline, nonexpanded or expanded CD34+ cells were injected via the spleen. After 7 days, CD34+ cells were effectively expanded in a serum-free culture medium. Expanded CD34+ cells were also increasingly positive for cell surface markers of VE-cadherin, VEGF receptor-2, and Tie-2. The expression of proangiogenic growth factors and adhesion molecules in expanded CD34+ cells increased compared with nonexpanded CD34+ cells. Expanded CD34+ cell transplantation reduced liver fibrosis, with a decrease of αSMA+ cells. Assessments of hepatocyte and sinusoidal endothelial cell proliferative activity indicated the superior potency of expanded CD34+ cells over non-expanded CD34+ cells. The inhibition of integrin αvβ3 and αvβ5 disturbed the engraftment of transplanted CD34+ cells and aggravated liver fibrosis. These findings suggest that expanded CD34+ cells enhanced the preventive efficacy of cell transplantation in a cirrhotic model.
Collapse
Affiliation(s)
- Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Victor Tsutsumi
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies , Mexico City, Mexico
| | - Yasuko Imamura
- Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University , Kurume, Japan
| | - Masako Naitou
- Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University , Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Yu Ikezono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Mitsuhiko Abe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Fumitaka Wada
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takato Ueno
- Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University , Kurume, Japan
| | - Masaaki Ii
- Group of Translational Stem Cell Research, Department of Pharmacology, Osaka Medical College , Takatsuki, Japan
| | - Cantas Alev
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University , Kyoto, Japan
| | - Atsuhiko Kawamoto
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation , Kyoto, Japan
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine , Isehara, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine , Kurume, Japan
| |
Collapse
|
223
|
The development and characterization of SDF1α-elastin-like-peptide nanoparticles for wound healing. J Control Release 2016; 232:238-47. [PMID: 27094603 DOI: 10.1016/j.jconrel.2016.04.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/18/2022]
Abstract
Chronic skin wounds are characterized by poor re-epithelialization, angiogenesis and granulation. Previous work has demonstrated that topical stromal cell-derived growth factor-1 (SDF1) promotes neovascularization, resulting in faster re-epithelialization of skin wounds in diabetic mice. However, the clinical usefulness of such bioactive peptides is limited because they are rapidly degraded in the wound environment due to high levels of proteases. Here, we describe the development of a recombinant fusion protein comprised of SDF1 and an elastin-like peptide that confers the ability to self-assemble into nanoparticles. The fusion protein and recombinant human SDF1 showed similar binding characteristics, as indicated by the measured equilibrium dissociation constant (Kd) for the binding of free SDF1 or the fusion protein to the CXCR4 receptor. The biological activity of SDF1-ELP, as measured by intracellular calcium release in HL60 cells was dose dependent, and also very similar to that of free SDF1. In contrast, the biological activity of SDF1-ELP in vivo was significantly superior to that of free SDF1. When applied to full thickness skin wounds in diabetic mice, wounds treated with SDF1-ELP nanoparticles were 95% closed by day 21, and fully closed by day 28, while wounds treated with free SDF1, ELP alone, or vehicle were only 80% closed by day 21, and took 42days to fully close. In addition, the SDF1-ELP nanoparticles significantly increased the epidermal and dermal layer of the healed wound, as compared to the other groups. These results indicate that SDF1-ELP fusion protein nanoparticles are promising agents for the treatment of chronic skin wounds.
Collapse
|
224
|
|
225
|
Tseng SY, Chao TH, Li YH, Liu PY, Lee CH, Cho CL, Wu HL, Chen JH. Cilostazol improves high glucose-induced impaired angiogenesis in human endothelial progenitor cells and vascular endothelial cells as well as enhances vasculoangiogenesis in hyperglycemic mice mediated by the adenosine monophosphate-activated protein kinase pathway. J Vasc Surg 2016; 63:1051-62.e3. [DOI: 10.1016/j.jvs.2014.10.103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/28/2014] [Indexed: 02/06/2023]
|
226
|
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
|
227
|
Flex A, Biscetti F, Iachininoto MG, Nuzzolo ER, Orlando N, Capodimonti S, Angelini F, Valentini CG, Bianchi M, Larocca LM, Martini M, Teofili L. Human cord blood endothelial progenitors promote post-ischemic angiogenesis in immunocompetent mouse model. Thromb Res 2016; 141:106-11. [PMID: 26994683 DOI: 10.1016/j.thromres.2016.03.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Human cord blood (CB) endothelial colony forming cells (ECFCs) are endowed with high vascular regenerative ability in immunodeficient mice, but their immunogenicity and susceptibility to rejection in immunocompetent models has yet to be explored. METHODS We injected CB ECFCs in non-immuno-suppressed C57BL/6J mice after having induced the hindlimb ischemia and we investigated their contribution to the recovery from the ischemic injury. Human ECFCs (hECFCs) were administered by intramuscular injection and hindlimb blood perfusion was measured by laser Doppler analysis at 7-day intervals for 28days after treatment. Mice were sacrificed after 7 and 28days and immunohistochemistry for specific human (CD31) and mouse (von Willebrand factor) endothelial antigens was carried out. Before euthanasia, blood samples to assess cytokines and angiogenic growth factor levels were collected. RESULTS Mice injected with hECFCs showed a prompter and greater recovery of blood flow than controls. Several endothelial cells of human origin were detected at day7 after injection and their number declined progressively. Likewise, a progressive increase of mouse-derived vascular structures were observed, paralleled by the amplified endogenous production of various soluble mediators of angiogenesis, including Vascular Endothelial Growth Factor and Fibroblast Growth Factor. CONCLUSIONS Overall, our findings are consistent with the hypothesis that human ECFCs might expand the endogenous vascular repair potential of recipients and support their possible HLA-independent unconventional use.
Collapse
Affiliation(s)
- Andrea Flex
- Department of Internal Medicine, Catholic University, Rome, Italy
| | | | | | | | | | | | - Flavia Angelini
- Department of Internal Medicine, Catholic University, Rome, Italy
| | | | - Maria Bianchi
- Institute of Hematology, Catholic University, Rome, Italy
| | | | | | | |
Collapse
|
228
|
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: 147] [Impact Index Per Article: 16.3] [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
|
229
|
Cha MJ, Choi E, Lee S, Song BW, Yoon C, Hwang KC. The microRNA-dependent cell fate of multipotent stromal cells differentiating to endothelial cells. Exp Cell Res 2016; 341:139-146. [PMID: 26854694 DOI: 10.1016/j.yexcr.2016.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/15/2023]
Abstract
In the endothelial recovery process, bone marrow-derived MSCs are a potential source of cells for both research and therapy, and their capacities to self-renew and to differentiate into all the cell types in the human body make them a promising therapeutic agent for remodeling cellular differentiation and a valuable resource for the treatment of many diseases. Based on the results provided in a miRNA database, we selected miRNAs with unique targets in cell fate-related signaling pathways. The tested miRNAs targeting GSK-3β (miR-26a), platelet-derived growth factor receptor, and CD133 (miR-26a and miR-29b) induced MSC differentiation into functional ECs, whereas miRNAs targeting VEGF receptor (miR-15, miR-144, miR-145, and miR-329) inhibited MSC differentiation into ECs through VEGF stimulation. In addition, the expression levels of these miRNAs were correlated with in vivo physiological endothelial recovery processes. These findings indicate that the miRNA expression profile is distinct for cells in different stages of differentiation from MSCs to ECs and that specific miRNAs can function as regulators of endothelialization.
Collapse
Affiliation(s)
- Min-Ji Cha
- Institute for Integrative Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea; Comprehensive Care Hospital for Cancer Patients, Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea; Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea
| | - Eunhyun Choi
- Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea; Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea
| | - Seahyoung Lee
- Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea; Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea
| | - Byeong-Wook Song
- Institute for Integrative Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea; Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea
| | - Cheesoon Yoon
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea; Department of Cardiovascular & Thoracic Surgery, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea
| | - Ki-Chul Hwang
- Catholic Kwandong University International St. Mary's Hospital, Incheon 22711, Republic of Korea; Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do 25601, Republic of Korea.
| |
Collapse
|
230
|
Pedralli ML, Waclawovsky G, Camacho A, Markoski MM, Castro I, Lehnen AM. Study of endothelial function response to exercise training in hypertensive individuals (SEFRET): study protocol for a randomized controlled trial. Trials 2016; 17:84. [PMID: 26873336 PMCID: PMC4752791 DOI: 10.1186/s13063-016-1210-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/03/2016] [Indexed: 02/06/2023] Open
Abstract
Background Endothelial dysfunction is a hallmark of diabetes mellitus and systemic arterial hypertension (SAH) and an early maker for atherosclerosis. Aerobic exercise training is known to enhance endothelial function, but little is understood about the effects of resistance or combined exercise training on endothelial function. The aim of this study is to investigate the effect of a 12-week aerobic (AT), resistance (RT), or combined (aerobic and resistance, CT) training program on endothelial function and assess associated effects on blood pressure in individuals with SAH. Methods/design Eighty-one subjects with SAH aged 18 to 70 years will be selected and randomly assigned to three types of exercise training: AT, RT or CT. The study will involve the following procedures and tests: anamnesis, anthropometric assessment, echocardiography, blood pressure measurements through ambulatory blood pressure monitoring, flow-mediated dilation, ergospirometry, one repetition maximum test (1-RM), and blood collection (number of circulating endothelial progenitor cells, number of circulating endothelial microparticles, lipid profile, glucose, glycated hemoglobin, and creatinine). The AT intervention will consist of a 40-min exercise session with progressive intensities ranging from 50 to 75 % of heart rate reserve. The RT intervention will consist of a 40-minute session with four sets of six to 12 repetitions with a rest period of 60 to 90 seconds between each set and each type of exercise. Weight loads will be adjusted to 60 to 80 % of 1-RM for six types of exercise. The CT intervention will consist of a 20-min aerobic exercise session, followed by an additional 20-min resistance exercise session; each resistance exercise will have two sets less to match the total training volume. Discussion The study results are expected evidence of cardiovascular protective effects of different types of exercise training through the modulation of endothelial function in hypertensive individuals. Knowing the magnitude of improvement of endothelium-dependent vasodilation for the different types of exercise training can provide scientific evidence for the prescription of exercise programs for vascular protection targeting hypertensive individuals. Trial registration The Brazilian Clinical Trials Registry (http://www.ensaiosclinicos.gov.br/) under RBR-9ygmdn and dated 1 March 2015.
Collapse
Affiliation(s)
- Marinei Lopes Pedralli
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil. .,Universidade Luterana do Brasil - ULBRA, Torres, Rio Grande do Sul, Brazil.
| | - Gustavo Waclawovsky
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil.
| | - Augusto Camacho
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil.
| | - Melissa Medeiros Markoski
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil.
| | - Iran Castro
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil.
| | - Alexandre Machado Lehnen
- Laboratório de Investigação Clínica, Instituto de Cardiologia / Fundação Universitária de Cardiologia - ICFUC, Av. Princesa Isabel, 395. Bairro Santana, 90620 001, Porto Alegre, RS, Brazil. .,Faculdade Sogipa de Educação Física, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
231
|
Cheng HS, Fish JE. Neovascularization Driven by MicroRNA Delivery to the Endothelium. Arterioscler Thromb Vasc Biol 2016; 35:2263-5. [PMID: 26490274 DOI: 10.1161/atvbaha.115.306558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Henry S Cheng
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| | - Jason E Fish
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada.
| |
Collapse
|
232
|
Kong L, Du X, Hu N, Li W, Wang W, Wei S, Zhuang H, Li X, Li C. Downregulation of let-7e-5p contributes to endothelial progenitor cell dysfunction in deep vein thrombosis via targeting FASLG. Thromb Res 2016; 138:30-36. [DOI: 10.1016/j.thromres.2015.12.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/01/2015] [Accepted: 12/23/2015] [Indexed: 01/07/2023]
|
233
|
Jang HK, Kim BS, Han J, Yoon JK, Lee JR, Jeong GJ, Shin JY. Therapeutic angiogenesis using tumor cell-conditioned medium. Biotechnol Prog 2016; 32:456-64. [DOI: 10.1002/btpr.2226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/31/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Hyeon-Ki Jang
- Interdisciplinary Program for Bioengineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Byung-Soo Kim
- Interdisciplinary Program for Bioengineering; Seoul National University; Seoul 151-744 Republic of Korea
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
- Bio-MAX Inst.; Inst. for Chemical Processes, Seoul National University; Seoul 151-744 Republic of Korea
| | - Jin Han
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Ju-Ro Lee
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| | - Jung-Youn Shin
- School of Chemical and Biological Engineering; Seoul National University; Seoul 151-744 Republic of Korea
| |
Collapse
|
234
|
Kong L, Hu N, Du X, Wang W, Chen H, Li W, Wei S, Zhuang H, Li X, Li C. Upregulation of miR-483-3p contributes to endothelial progenitor cells dysfunction in deep vein thrombosis patients via SRF. J Transl Med 2016; 14:23. [PMID: 26801758 PMCID: PMC4724160 DOI: 10.1186/s12967-016-0775-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/06/2016] [Indexed: 12/20/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) contribute to recanalization of deep vein thrombosis (DVT). This study aimed to detect miRNA expression profiles in EPCs from patients with DVT and characterize the role of miRNA in EPCs dysfunction. Methods EPCs was isolated from DVT patients and control subjects, and miRNA expression profiles were compared to screen differential miRNAs. The candidate miRNAs were confirmed by RT-PCR analysis. The targets of miRNA were identified by bioinformatics analyses, luciferase reporter assay and gene expression analyses. The apoptosis, migration and tube formation of EPCs were examined by flow cytometry, transwell assay and matrigel tube formation assay. A rat model of venous thrombosis was established as in vivo model. Results We identified miR-483-3p as a candidate miRNA upregulated in EPCs from DVT patients. By using miR-483-3p agomir and antagomir, we demonstrated that miR-483-3p decreased the migration and tube formation while increased the apoptosis of EPCs. Moreover, we identified serum response factor (SRF) as the target of miR-483-3p, and showed that SRF knockdown decreased the migration and tube formation while increased the apoptosis of EPCs. In addition, miR-483-3p inhibition led to enhanced ability of homing and thrombus resolution of EPCs in rat model of venous thrombosis. Conclusions miR-483-3p is upregulated in EPCs from DVT patients, and it targets SRF to decrease EPCs migration and tube formation and increase apoptosis in vitro, while decrease EPCs homing and thrombus resolution in vivo. MiR-483-3p is a potential therapeutic target in DVT treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0775-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lingshang Kong
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Nan Hu
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Xiaolong Du
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Hong Chen
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Wendong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Sen Wei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Hao Zhuang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| | - Chenglong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Rd, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
235
|
Di Santo S, Fuchs AL, Periasamy R, Seiler S, Widmer HR. The Cytoprotective Effects of Human Endothelial Progenitor Cell-Conditioned Medium Against an Ischemic Insult Are Not Dependent on VEGF and IL-8. Cell Transplant 2016; 25:735-47. [PMID: 26776768 DOI: 10.3727/096368916x690458] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Endothelial progenitor cells (EPCs) promote revascularization and tissue repair mainly by paracrine actions. In the present study, we investigated whether EPC-secreted factors in the form of conditioned medium (EPC-CM) can protect cultured brain microvascular endothelial cells against an ischemic insult. Furthermore, we addressed the type of factors that are involved in the EPC-CM-mediated functions. For that purpose, rat brain-derived endothelial cells (rBCEC4 cell line) were exposed to EPC-CM pretreated with proteolytic digestion, heat inactivation, and lipid extraction. Moreover, the involvement of VEGF and IL-8, as canonical angiogenic factors, was investigated by means of neutralizing antibodies. We demonstrated that EPC-CM significantly protected the rBCEC4 cells against an ischemic insult mimicked by induced oxygen-glucose deprivation followed by reoxygenation. The cytoprotective effect was displayed by higher viable cell numbers and reduced caspase 3/7 activity. Heat inactivation, proteolytic digestion, and lipid extraction resulted in a significantly reduced EPC-CM-dependent increase in rBCEC4 viability, tube formation, and survival following the ischemic challenge. Notably, VEGF and IL-8 neutralization did not affect the actions of EPC-CM on rBCEC4 under both standard and ischemic conditions. In summary, our findings show that paracrine factors released by EPCs activate an angiogenic and cytoprotective response on brain microvascular cells and that the activity of EPC-CM relies on the concerted action of nonproteinaceous and proteinaceous factors but do not directly involve VEGF and IL-8.
Collapse
Affiliation(s)
- Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
| | | | | | | | | |
Collapse
|
236
|
Samura M, Morikage N, Suehiro K, Tanaka Y, Nakamura T, Nishimoto A, Ueno K, Hosoyama T, Hamano K. Combinatorial Treatment with Apelin-13 Enhances the Therapeutic Efficacy of a Preconditioned Cell-Based Therapy for Peripheral Ischemia. Sci Rep 2016; 6:19379. [PMID: 26763337 PMCID: PMC4725909 DOI: 10.1038/srep19379] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/08/2015] [Indexed: 11/23/2022] Open
Abstract
Hypoxic pretreatment of peripheral blood mononuclear cells (PBMNCs) enhances therapeutic angiogenesis in ischemic tissues after cell transplantation. However, newly formed vessels generated using this approach are immature and insufficient for promoting functional recovery from severe ischemia. In this study, we examined whether apelin-13, a regulator of vessel maturation, could be an effective promoter of therapeutic angiogenesis, following severe limb ischemia. Combinatorial treatment of hypoxic preconditioned PBMNCs with apelin-13 resulted in increased blood perfusion and vascular reactivity in ischemic mouse hindlimbs compared with a monotherapy comprising each factor. Apelin-13 upregulated expression of PDGF-BB and TGF-β1 in hypoxic PBMNCs, as well as that of PDGFR-β in vascular smooth muscle cells (VSMCs). Proliferation and migration of VSMCs treated with apelin-13 was accelerated in the presence of PDGF-BB. Interestingly, expression of an apelin receptor, APJ, in PBMNC was increased under hypoxia but not under normoxia. In addition, an in vitro angiogenesis assay using a co-culture model comprising mouse thoracic aorta, hypoxic PBMNCs, and apelin-13 demonstrated that combinatorial treatment recruited mural cells to sprouted vessel outgrowths from the aortic ring, thereby promoting neovessel maturation. Thus, combinatorial injection of hypoxic PBMNCs and apelin-13 could be an effective therapeutic strategy for patients with severe ischemic diseases.
Collapse
Affiliation(s)
- Makoto Samura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Kotaro Suehiro
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Yuya Tanaka
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Tamami Nakamura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Arata Nishimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Tohru Hosoyama
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0836, Japan
| |
Collapse
|
237
|
Chen Q, Varga M, Wang X, Haddad DJ, An S, Medzikovic L, Derakhshandeh R, Kostyushev DS, Zhang Y, Clifford BT, Luu E, Danforth OM, Rafikov R, Gong W, Black SM, Suchkov SV, Fineman JR, Heiss C, Aschbacher K, Yeghiazarians Y, Springer ML. Overexpression of Nitric Oxide Synthase Restores Circulating Angiogenic Cell Function in Patients With Coronary Artery Disease: Implications for Autologous Cell Therapy for Myocardial Infarction. J Am Heart Assoc 2016; 5:e002257. [PMID: 26738788 PMCID: PMC4859354 DOI: 10.1161/jaha.115.002257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/25/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Circulating angiogenic cells (CACs) are peripheral blood cells whose functional capacity inversely correlates with cardiovascular risk and that have therapeutic benefits in animal models of cardiovascular disease. However, donor age and disease state influence the efficacy of autologous cell therapy. We sought to determine whether age or coronary artery disease (CAD) impairs the therapeutic potential of CACs for myocardial infarction (MI) and whether the use of ex vivo gene therapy to overexpress endothelial nitric oxide (NO) synthase (eNOS) overcomes these defects. METHODS AND RESULTS We recruited 40 volunteers varying by sex, age (< or ≥45 years), and CAD and subjected their CACs to well-established functional tests. Age and CAD were associated with reduced CAC intrinsic migration (but not specific response to vascular endothelial growth factor, adherence of CACs to endothelial tubes, eNOS mRNA and protein levels, and NO production. To determine how CAC function influences therapeutic potential, we injected the 2 most functional and the 2 least functional CAC isolates into mouse hearts post MI. The high-function isolates substantially improved cardiac function, whereas the low-function isolates led to cardiac function only slightly better than vehicle control. Transduction of the worst isolate with eNOS cDNA adenovirus increased NO production, migration, and cardiac function of post-MI mice implanted with the CACs. Transduction of the best isolate with eNOS small interfering RNA adenovirus reduced all of these capabilities. CONCLUSIONS Age and CAD impair multiple functions of CACs and limit therapeutic potential for the treatment of MI. eNOS gene therapy in CACs from older donors or those with CAD has the potential to improve autologous cell therapy outcomes.
Collapse
Affiliation(s)
- Qiumei Chen
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Monika Varga
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Xiaoyin Wang
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Daniel J. Haddad
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Songtao An
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Lejla Medzikovic
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | - Ronak Derakhshandeh
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | | | - Yan Zhang
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | - Brian T. Clifford
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | - Emmy Luu
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | - Olivia M. Danforth
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
| | | | - Wenhui Gong
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCA
| | | | | | - Jeffrey R. Fineman
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCA
| | - Christian Heiss
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
| | - Kirstin Aschbacher
- Department of PsychiatryUniversity of California, San FranciscoSan FranciscoCA
| | - Yerem Yeghiazarians
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
- Eli & Edythe Broad Institute of Regeneration Medicine and Stem Cell ResearchUniversity of California, San FranciscoSan FranciscoCA
| | - Matthew L. Springer
- Cardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCA
- Division of CardiologyUniversity of California, San FranciscoSan FranciscoCA
- Eli & Edythe Broad Institute of Regeneration Medicine and Stem Cell ResearchUniversity of California, San FranciscoSan FranciscoCA
| |
Collapse
|
238
|
Feito MJ, Serrano MC, Oñaderra M, Matesanz MC, Sánchez-Salcedo S, Arcos D, Vallet-Regí M, Portolés MT. Effects of immobilized VEGF on endothelial progenitor cells cultured on silicon substituted and nanocrystalline hydroxyapatites. RSC Adv 2016. [DOI: 10.1039/c6ra19154a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Immobilized VEGF effects on angiogenic cells cultured on silicon substituted and nanocrystalline hydroxyapatites.
Collapse
Affiliation(s)
- M. J. Feito
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry
- Universidad Complutense de Madrid
- Spain
- Instituto de Investigación Sanitaria San Carlos IdISSC
- Spain
| | - M. C. Serrano
- Hospital Nacional de Parapléjicos Servicio de Salud de Castilla-La Mancha
- Toledo
- Spain
| | - M. Oñaderra
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry
- Universidad Complutense de Madrid
- Spain
| | - M. C. Matesanz
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry
- Universidad Complutense de Madrid
- Spain
| | - S. Sánchez-Salcedo
- Department of Inorganic and Bioinorganic Chemistry/Faculty of Pharmacy
- Universidad Complutense de Madrid
- Instituto de Investigación Hospital 12 de Octubre i+12
- Spain
- Networking Research Center on Bioengineering/Biomaterials and Nanomedicine
| | - D. Arcos
- Department of Inorganic and Bioinorganic Chemistry/Faculty of Pharmacy
- Universidad Complutense de Madrid
- Instituto de Investigación Hospital 12 de Octubre i+12
- Spain
- Networking Research Center on Bioengineering/Biomaterials and Nanomedicine
| | - M. Vallet-Regí
- Department of Inorganic and Bioinorganic Chemistry/Faculty of Pharmacy
- Universidad Complutense de Madrid
- Instituto de Investigación Hospital 12 de Octubre i+12
- Spain
- Networking Research Center on Bioengineering/Biomaterials and Nanomedicine
| | - M. T. Portolés
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry
- Universidad Complutense de Madrid
- Spain
- Instituto de Investigación Sanitaria San Carlos IdISSC
- Spain
| |
Collapse
|
239
|
Rioja AY, Tiruvannamalai Annamalai R, Paris S, Putnam AJ, Stegemann JP. Endothelial sprouting and network formation in collagen- and fibrin-based modular microbeads. Acta Biomater 2016; 29:33-41. [PMID: 26481042 PMCID: PMC4681647 DOI: 10.1016/j.actbio.2015.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Abstract
A modular tissue engineering approach may have advantages over current therapies in providing rapid and sustained revascularization of ischemic tissue. In this study, modular protein microbeads were prepared from pure fibrin (FIB) and collagen-fibrin composites (COL-FIB) using a simple water-in-oil emulsification technique. Human endothelial cells and fibroblasts were embedded directly in the microbead matrix. The resulting microbeads were generally spheroidal with a diameter of 100-200μm. Cell viability was high (75-80% viable) in microbeads, but was marginally lower than in bulk hydrogels of corresponding composition (85-90% viable). Cell proliferation was significantly greater in COL-FIB microbeads after two weeks in culture, compared to pure FIB microbeads. Upon embedding of microbeads in a surrounding fibrin hydrogel, endothelial cell networks formed inside the microbead matrix and extended into the surrounding matrix. The number of vessel segments, average segment length, and number of branch points was higher in FIB samples, compared to COL-FIB samples, resulting in significantly longer total vessel networks. Anastomosis of vessel networks from adjacent microbeads was also observed. These studies demonstrate that primitive vessel networks can be formed by modular protein microbeads containing embedded endothelial cells and fibroblasts. Such microbeads may find utility as prevascularized tissue modules that can be delivered minimally invasively as a therapy to restore blood flow to ischemic tissues. STATEMENT OF SIGNIFICANCE Vascularization is critically important for tissue engineering and regenerative medicine, and materials that support and/or promote neovascularization are of value both for translational applications and for mechanistic studies and discovery-based research. Therefore, we fabricated small modular microbeads formulated from pure fibrin (FIB) and collagen-fibrin (COL-FIB) containing endothelial cells and supportive fibroblasts. We explored how cells encapsulated within these materials form microvessel-like networks both within and outside of the microbeads when embedded in larger 3D matrices. FIB microbeads were found to initiate more extensive sprouting into the surrounding ECM in vitro. These results represent an important step towards our goal of developing injectable biomaterial modules containing preformed vascular units that can rapidly restore vascularization to an ischemic tissue in vivo.
Collapse
Affiliation(s)
- Ana Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | | | - Spencer Paris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| |
Collapse
|
240
|
Sukmawati D, Tanaka R, Ito-Hirano R, Fujimura S, Hayashi A, Itoh S, Mizuno H, Daida H. The role of Notch signaling in diabetic endothelial progenitor cells dysfunction. J Diabetes Complications 2016; 30:12-20. [PMID: 26598222 DOI: 10.1016/j.jdiacomp.2015.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/06/2015] [Accepted: 09/24/2015] [Indexed: 12/24/2022]
Abstract
AIMS To investigate the role of Notch signaling pathway in vasculogenic dysfunction of diabetic EPCs (DM-EPCs). METHODS The study was performed in mice and diabetes was induced with Streptozotocin. The functional consequences of Notch pathway modulation were studied by assessment of colony forming capacity (EPC colony forming assay), EPC differentiation capacity (% of definitive EPC-CFU (dEPC-CFU)), circulating EPCs (EPC culture assay) and migrated cells (migration assay); in the presence of Notch inhibitor (γ-secretase inhibitors (GSI)) compared to control. Notch pathway and VEGF involvement in DM- EPCs were assessed by gene expression (RT-qPCR). RESULTS DM demonstrated to increase Notch pathway expression in bone marrow (BM) EPCs followed by lower EPC-CFU number, EPCs differentiation capacity, number of circulating EPCs, migrated cells and VEGF expression compared to control (p<0.05). Inhibition of Notch pathway by GSI rescued vasculogenic dysfunction in DM-EPCs as represented by increase in EPC-CFU number, differentiation capacity and number of circulating EPCs (p<0.05). CONCLUSION Our findings indicate the involvement of Notch pathway in mediating DM-EPCs dysfunction including less number of EPC-CFU, circulating EPCs and migrated cell number compared to control. Further in vitro inhibition of Notch pathway by GSI rescued DM-EPC dysfunction. Therefore targeting Notch pathway in DM may provide a target to restore DM-EPC dysfunction.
Collapse
Affiliation(s)
- Dewi Sukmawati
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan; Department of Cardiovascular Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan; Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Jalan Salemba Raya No. 6 Jakarta Pusat, 10430, Indonesia.
| | - Rica Tanaka
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Rie Ito-Hirano
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Satoshi Fujimura
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Ayato Hayashi
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Seigo Itoh
- Department of Cardiovascular Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Hiroshi Mizuno
- Department of Plastic Reconstructive Surgery, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
241
|
Kalyva A, Marketou ME, Parthenakis FI, Pontikoglou C, Kontaraki JE, Maragkoudakis S, Petousis S, Chlouverakis G, Papadaki HA, Vardas PE. Endothelial progenitor cells as markers of severity in hypertrophic cardiomyopathy. Eur J Heart Fail 2015; 18:179-84. [PMID: 26696595 DOI: 10.1002/ejhf.436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 01/05/2023] Open
Abstract
AIMS Endothelial progenitor cells (EPCs) are bone marrow-derived cells that are mobilized into the circulation to migrate and differentiate into mature endothelial cells contributing to post-natal physiological and pathological neovascularization. In this study, we evaluated circulating EPCs in patients with hypertrophic cardiomyopathy (HCM) and examined a potential association with clinical parameters of the disease. METHODS AND RESULTS We included 40 HCM patients and 23 healthy individuals. Using flow cytometry we measured EPCs in peripheral blood as two subpopulations of CD45-/CD34+/VEGFR2+ and CD45-/CD34+/CD133+ cells. Circulating CD45-/CD34+/VEGFR2+ cells were significantly increased in HCM patients in comparison with the controls (0.000238 ± 0.0003136 vs. 0.000057 ± 0.0001316, respectively, P = 0.002). However, there was no significant difference in the number of circulating CD45-/CD34+/CD133+ cells (0.003079 ± 0.0033288 vs. 0.002065 ± 0.0022173, respectively, P = 0.153). The CD45-/CD34+/VEGFR2+ subpopulation revealed a moderate correlation with LV mass index (r = 0.35, P = 0.026), while both EPC subpopulation levels showed strong positive correlations with th E/e' ratio (r = 0.423, P = 0.007 for CD45-/CD34+/VEGFR2+ and r = 0.572, P < 0.001 for CD45-/CD34+/CD133+). CONCLUSION HCM patients showed an increased mobilization of EPCs compared with healthy individuals that correlated with diastolic dysfunction. Our findings may open up new dimensions in the pathophysiology, prognostication, and treatment of HCM.
Collapse
Affiliation(s)
- Athanasia Kalyva
- Molecular Cardiology Laboratory, School of Medicine, University of Crete, Greece
| | - Maria E Marketou
- Department of Cardiology, Heraklion University Hospital, Crete, Greece
| | | | | | - Joanna E Kontaraki
- Molecular Cardiology Laboratory, School of Medicine, University of Crete, Greece
| | | | | | | | - Helen A Papadaki
- Department of Haematology, Heraklion University Hospital, Crete, Greece
| | - Panos E Vardas
- Department of Cardiology, Heraklion University Hospital, Crete, Greece
| |
Collapse
|
242
|
Field S, Uyttenhove C, Stroobant V, Cheou P, Donckers D, Coutelier JP, Simpson PT, Cummings MC, Saunus JM, Reid LE, Kutasovic JR, McNicol AM, Kim BR, Kim JH, Lakhani SR, Neville AM, Van Snick J, Jat PS. Novel highly specific anti-periostin antibodies uncover the functional importance of the fascilin 1-1 domain and highlight preferential expression of periostin in aggressive breast cancer. Int J Cancer 2015; 138:1959-70. [PMID: 26619948 DOI: 10.1002/ijc.29946] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 11/04/2015] [Indexed: 01/07/2023]
Abstract
Periostin (POSTN), a secreted homodimeric protein that binds integrins αvβ3, αvβ5, and α6β4, was originally found to be expressed in fetal tissues and in the adult upon injury particularly bone fractures due to its role in remodelling and repair. Recently it was found to be over-expressed in human breast cancer and a variety of other tumour types including head and neck squamous cell carcinoma, where its overexpression correlates with increased tumour invasion. Progress in studying its functional role in tumour pathogenesis has been hampered by the paucity of antibodies for its specific and sensitive detection. It has proven very difficult to obtain monoclonal antibodies (mAbs) against this highly conserved protein but we report here that combining infection of mice with lactate dehydrogenase elevating virus (LDV), a B cell activating arterivirus, with conjugation of human POSTN to ovalbumin as an immunogenic carrier, enabled us to develop six mAbs recognizing both human and mouse POSTN and inhibiting its binding to αvβ3 integrin. Two of the mAbs, MPB4B1 and MPC5B4, were tested and found to inhibit POSTN-induced migration of human endothelial colony forming cells. All six mAbs recognized amino acids 136-51 (APSNEAWDNLDSDIRR) within the POSTN fascilin (FAS) 1-1 domain revealing the functional importance of this motif; this was further highlighted by the ability of aa 136-151 peptide to inhibit integrin-mediated cell migration. Immunohistochemistry using MPC5B4, indicated that breast tumour cell POSTN expression was a strong prognostic indicator, along with tumour size, lymph node, and human epidermal growth factor receptor 2 (HER2) status.
Collapse
Affiliation(s)
- Sarah Field
- University of Oxford Branch, Ludwig Cancer Research, Oxford, United Kingdom.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Catherine Uyttenhove
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique De Louvain, Brussels, Belgium
| | | | - Paméla Cheou
- de Duve Institute, Université Catholique De Louvain, Brussels, Belgium
| | | | | | - Peter T Simpson
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,Cancer Genetics Laboratory, QIMR Berghofer Medical Research Institute, Queensland, Herston, Australia.,The University of Queensland, School of Medicine, Discipline of Molecular & Cellular Pathology, Herston, Brisbane, Australia
| | - Margaret C Cummings
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,The University of Queensland, School of Medicine, Discipline of Molecular & Cellular Pathology, Herston, Brisbane, Australia.,Pathology Queensland, The Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - Jodi M Saunus
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,Cancer Genetics Laboratory, QIMR Berghofer Medical Research Institute, Queensland, Herston, Australia
| | - Lynne E Reid
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,Cancer Genetics Laboratory, QIMR Berghofer Medical Research Institute, Queensland, Herston, Australia
| | - Jamie R Kutasovic
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,Cancer Genetics Laboratory, QIMR Berghofer Medical Research Institute, Queensland, Herston, Australia
| | - Anne Marie McNicol
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,The University of Queensland, School of Medicine, Discipline of Molecular & Cellular Pathology, Herston, Brisbane, Australia
| | - Ba Reun Kim
- Medical Research Centre for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea.,Department of Physiology, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Jae Ho Kim
- Medical Research Centre for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea.,Department of Physiology, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Sunil R Lakhani
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, Australia.,The University of Queensland, School of Medicine, Discipline of Molecular & Cellular Pathology, Herston, Brisbane, Australia.,Pathology Queensland, The Royal Brisbane & Women's Hospital, Brisbane, Australia
| | | | - Jacques Van Snick
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique De Louvain, Brussels, Belgium
| | - Parmjit S Jat
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom.,MRC Prion Unit, UCL Institute of Neurology, Queen Square, London, United Kingdom
| |
Collapse
|
243
|
Evaluation of the clinical relevance and limitations of current pre-clinical models of peripheral artery disease. Clin Sci (Lond) 2015; 130:127-50. [DOI: 10.1042/cs20150435] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peripheral artery disease (PAD) has recognized treatment deficiencies requiring the discovery of novel interventions. This article describes current animal models of PAD and discusses their advantages and disadvantages. There is a need for models which more directly simulate the characteristics of human PAD, such as acute-on-chronic presentation, presence of established risk factors and impairment of physical activity.
Collapse
|
244
|
Mata MF, Lopes JP, Ishikawa M, Alaiti MA, Cabral JM, da Silva CL, Costa MA. Scaling up the ex vivo expansion of human circulating CD34+progenitor cells with upregulation of angiogenic and anti-inflammatory potential. Cytotherapy 2015; 17:1777-84. [DOI: 10.1016/j.jcyt.2015.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/16/2015] [Accepted: 09/11/2015] [Indexed: 01/27/2023]
|
245
|
Yin J, Huang F, Yi Y, Yin L, Peng D. EGCG attenuates atherosclerosis through the Jagged-1/Notch pathway. Int J Mol Med 2015; 37:398-406. [PMID: 26648562 DOI: 10.3892/ijmm.2015.2422] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 11/19/2015] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis is the most common cause of cardiovascular diseases worldwide. Oxidized low-density lipoprotein (ox-LDL) is a particularly important risk factor in the pathogenesis of atherosclerosis. Accumulating evidence has indicated that epigallocatechin-3-gallate (EGCG; a catechin found in the popular beverage, greent tea) protects against ox-LDL-induced atherosclerosis. However, the underlying mechanisms remain unclear. In the present study, ox-LDL (100 mg/l) induced damage to, and the apoptosis of human umbilical vein endothelial cells (HUVECs) by reducing endothelial nitric oxide synthase (eNOS) expression and promoting inducible nitric oxide synthase (iNOS) expression; these effects were abrogated by the addition of 50 µM EGCG. Furthermore, ox-LDL rapidly activated the membrane translocation of p22phox, and altered the protein expression of Jagged-1 and Notch pathway-related proteins [Math1, hairy and enhancer of split (HES)1 and HES5]; these effects were also prevented by pre-treatment with 50 µM EGCG. In addition, Jagged-1 played a significant role in the EGCG-mediated protection against ox-LDL-induced apoptosis and ox-LDL‑diminished cell adhesion in the HUVECs. Finally, EGCG inhibited high-fat diet (HFD)-induced atherosclerosis in apolipoprotein E (ApoE) knockout (ApoE-KO) mice through the Jagged-1/Notch pathway. Taken together, these findings demonstrate that 50 µM EGCG protects against ox-LDL-induced endothelial dysfunction through the Jagged-1/Notch signaling pathway. Moreover, our data provide insight into the possible molecular mechanisms through which EGCG attenuates ox-LDL‑induced vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Jianguo Yin
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Fang Huang
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410011, P.R. China
| | - Yuhong Yi
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Liang Yin
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Daoquan Peng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
246
|
Xu SM, Liang T. Clinical observation of the application of autologous peripheral blood stem cell transplantation for the treatment of diabetic foot gangrene. Exp Ther Med 2015; 11:283-288. [PMID: 26889255 DOI: 10.3892/etm.2015.2888] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 08/20/2015] [Indexed: 01/27/2023] Open
Abstract
The aim of the present study was to investigate the optimal mobilization plan in autologous peripheral blood stem cell transplantation for the treatment of diabetic foot and to observe its clinical curative effect. A total of 127 patients with diabetic foot were treated with different doses of granulocyte colony stimulating factor (G-CSF) to mobilize their hematopoietic stem cells. Subsequently, the extracted stem cell suspension was injected into the ischemic lower extremities along the blood vessels in the areas presenting with pathological changes. Following the treatment, the intermittent claudication distance, skin temperature, ankle brachial index and pain scores of the patients were evaluated. In addition, the associations among the mobilization time, doses and peripheral blood CD34+ level were analyzed. The collection efficiency of the stem cells was associated with the dose of G-CSF and the mobilization time. Following the injection of the autologous peripheral blood stem cell suspension, the ischemic area of the patients was improved significantly. In conclusion, autologous peripheral blood stem cell transplantation can promote the establishment of collateral circulation in patients with diabetic foot, and the optimal time for gathering stem cells is closely correlated with the peripheral blood CD34+ level.
Collapse
Affiliation(s)
- Shi-Min Xu
- Department of Orthopedics, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Ting Liang
- Department of Administration, People's Hospital of Langfang, Langfang, Hebei 065000, P.R. China
| |
Collapse
|
247
|
Chang MY, Huang TT, Chen CH, Cheng B, Hwang SM, Hsieh PCH. Injection of Human Cord Blood Cells With Hyaluronan Improves Postinfarction Cardiac Repair in Pigs. Stem Cells Transl Med 2015; 5:56-66. [PMID: 26574556 DOI: 10.5966/sctm.2015-0092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 09/16/2015] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Recent clinical trials using autologous bone marrow or peripheral blood cells to treat myocardial infarction (MI) show controversial results, although the treatment has a good safety profile. These discrepancies are likely caused by factors such as aging, systemic inflammation, and cell processing procedures, all of which might impair the regenerative capability of the cells used. Here, we tested whether injection of human cord blood mononuclear cells (CB-MNCs) combined with hyaluronan (HA) hydrogel improves cell therapy efficacy in a pig MI model. A total of 34 minipigs were divided into 5 groups: sham operation (Sham), surgically induced-MI plus injection with normal saline (MI+NS), HA only (MI+HA), CB-MNC only (MI+CB-MNC), or CB-MNC combined with HA (MI+CB-MNC/HA). Two months after the surgery, injection of MI+CB-MNC/HA showed the highest left ventricle ejection fraction (51.32%±0.81%) compared with MI+NS (42.87%±0.97%, p<.001), MI+HA (44.2%±0.63%, p<.001), and MI+CB-MNC (46.17%±0.39%, p<.001) groups. The hemodynamics data showed that MI+CB-MNC/HA improved the systolic function (+dp/dt) and diastolic function (-dp/dt) as opposed to the other experimental groups, of which the CB-MNC alone group only modestly improved the systolic function (+dp/dt). In addition, CB-MNC alone or combined with HA injection significantly decreased the scar area and promoted angiogenesis in the infarcted region. Together, these results indicate that combined CB-MNC and HA treatment improves heart performance and may be a promising treatment for ischemic heart diseases. SIGNIFICANCE This study using healthy human cord blood mononuclear cells (CB-MNCs) to treat myocardial infarction provides preclinical evidence that combined injection of hyaluronan and human CB-MNCs after myocardial infarction significantly increases cell retention in the peri-infarct area, improves cardiac performance, and prevents cardiac remodeling. Moreover, using healthy cells to replace dysfunctional autologous cells may constitute a better strategy to achieve heart repair and regeneration.
Collapse
Affiliation(s)
- Ming-Yao Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan, Republic of China Institute of Clinical Medicine, and National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Tzu-Ting Huang
- Institute of Clinical Medicine, and National Cheng Kung University, Tainan, Taiwan, Republic of China Department of Life Science, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Chien-Hsi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Bill Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China
| | - Patrick C H Hsieh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan, Republic of China Institute of Clinical Medicine, and National Cheng Kung University, Tainan, Taiwan, Republic of China Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| |
Collapse
|
248
|
Yıldırım C, Favre J, Weijers EM, Fontijn RD, van Wijhe MH, van Vliet SJ, Boon RA, Koolwijk P, van der Pouw Kraan TCTM, Horrevoets AJG. IFN-β affects the angiogenic potential of circulating angiogenic cells by activating calpain 1. Am J Physiol Heart Circ Physiol 2015; 309:H1667-78. [DOI: 10.1152/ajpheart.00810.2014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 09/01/2015] [Indexed: 01/15/2023]
Abstract
Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-β (IFN-β) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-β might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-β significantly reduced the number of CACs by 18–64%. Apoptosis was not induced by IFN-β, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-β impaired adhesion to, and spreading on, fibronectin, which was dependent on α5β1 (VLA-5)-integrin. IFN-β affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-β. We show that the IFN-β-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.
Collapse
Affiliation(s)
- Cansu Yıldırım
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Julie Favre
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Ruud D. Fontijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michiel H. van Wijhe
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Reinier A. Boon
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | | | - Anton J. G. Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
249
|
Repeated treatment with bone marrow cell secretory products maintains long-term renoprotection in experimental chronic kidney disease: a placebo-controlled trial. Can J Kidney Health Dis 2015; 2:44. [PMID: 26568839 PMCID: PMC4644292 DOI: 10.1186/s40697-015-0082-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/30/2015] [Indexed: 12/04/2022] Open
Abstract
Background Bone marrow-derived early outgrowth cells (EOCs) secrete soluble factors that exert potent renoprotective effects, such that infusion of their conditioned medium recapitulates the affects of the cells themselves. Objectives The objective of this study is to test whether the protective effect of conditioned medium infusion wanes with time and whether tachyphylaxis occurs with repeated administration. Design This is a placebo-controlled animal study. Setting The study was conducted at St. Michael’s Hospital, Toronto, Ontario, Canada. Subjects Fischer 344 (F344) rats were used in this study. Measurements The following were measured: (1) urinary protein:creatinine ratio, (2) glomerular filtration rate, (3) systolic blood pressure, (4) body weight, (5) glomerular endothelial cell density, and (6) glomerular and tubulointerstitial type IV collagen deposition. Methods Subtotally nephrectomized F344 rats, a model of progressive chronic kidney disease, were randomized 4 weeks post-surgery to receive thrice-weekly intravenous injections of concentrated EOC-conditioned medium (EOC CM) or unconditioned medium (UCM) over 2 weeks. Three animal groups were studied, according to whether they were administered conditioned medium: once (Initial Therapy Only group), twice (Repeat Therapy group), or not at all (No Therapy group). Results Following initial therapy, EOC CM-treated animals excreted less urinary protein, a marker of renal injury, than their UCM-treated counterparts. At 10 weeks post-subtotal nephrectomy, however, mean urinary protein excretion in conditioned medium-treated animals was fourfold greater than at the completion of the initial treatment course. At this time point, conditioned medium-treated animals were randomized to receive a second course of either conditioned medium (Repeat Therapy group) or unconditioned medium (Initial Therapy Only group). At study end (14 weeks post-subtotal nephrectomy), Repeat Therapy animals demonstrated higher glomerular filtration rate, less proteinuria, preserved renal microvasculature, and diminished fibrosis when compared with the No Therapy group. Initial Therapy Only animals exhibited an intermediate effect. Limitations Testing the effect of EOC-conditioned medium in a single model of chronic kidney disease (CKD) has limitations. Conclusions These findings suggest that early outgrowth cell-derived factors, while renoprotective, have a limited duration of action. Repeated administration of these factors, however, is able to extend the duration of efficacy and attenuate the progression of experimental chronic kidney disease.
Collapse
|
250
|
Song W, Kaufman DS, Shen W. Efficient generation of endothelial cells from human pluripotent stem cells and characterization of their functional properties. J Biomed Mater Res A 2015; 104:678-687. [PMID: 26519950 DOI: 10.1002/jbm.a.35607] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/24/2015] [Accepted: 10/29/2015] [Indexed: 01/15/2023]
Abstract
Although endothelial cells (ECs) have been derived from human pluripotent stem cells (hPSCs), large-scale generation of hPSC-ECs remains challenging and their functions are not well characterized. Here we report a simple and efficient three-stage method that allows generation of approximately 98 and 9500 ECs on day 16 and day 34, respectively, from each human embryonic stem cell (hESC) input. The functional properties of hESC-ECs derived in the presence and absence of a TGFβ-inhibitory molecule SB431542 were characterized and compared with those of human umbilical vein endothelial cells (HUVECs). Confluent monolayers formed by SB431542 + hESC-ECs, SB431542- hESC-ECs, and HUVECs showed similar permeability to 10,000 Da dextran, but these cells exhibited striking differences in forming tube-like structures in 3D fibrin gels. The SB431542 + hESC-ECs were most potent in forming tube-like structures regardless of whether VEGF and bFGF were present in the medium; less potent SB431542- hESC-ECs and HUVECs responded differently to VEGF and bFGF, which significantly enhanced the ability of HUVECs to form tube-like structures but had little impact on SB431542- hESC-ECs. This study offers an efficient approach to large-scale hPSC-EC production and suggests that the phenotypes and functions of hPSC-ECs derived under different conditions need to be thoroughly examined before their use in technology development. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 678-687, 2016.
Collapse
Affiliation(s)
- Wei Song
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, 55455
| | - Dan S Kaufman
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, 55455
| | - Wei Shen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, 55455
| |
Collapse
|