1
|
Liu Y, Niu Z, Wang X, Xiu C, Hu Y, Wang J, Lei Y, Yang J. Yiqihuoxue decoction (GSC) inhibits mitochondrial fission through the AMPK pathway to ameliorate EPCs senescence and optimize vascular aging transplantation regimens. Chin Med 2024; 19:143. [PMID: 39402613 PMCID: PMC11479513 DOI: 10.1186/s13020-024-01008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND During the aging process, the number and functional activity of endothelial progenitor cells (EPCs) are impaired, leading to the unsatisfactory efficacy of transplantation. Previous studies demonstrated that Yiqihuoxue decoction (Ginseng-Sanqi-Chuanxiong, GSC) exerts anti-vascular aging effects. The purpose of this study is to evaluated the effects of GSC on D-galactose (D-gal)induced senescence and the underlying mechanisms. METHODS The levels of cellular senescence-related markers P16, P21, P53, AMPK and p-AMPK were detected by Western blot analysis (WB). SA-β-gal staining was used to evaluate cell senescence. EPCs function was measured by CCK-8, Transwell cell migration and cell adhesion assay. The morphological changes of mitochondria were detected by confocal microscopy. The protein and mRNA expression of mitochondrial fusion fission Drp1, Mff, Fis1, Mfn1, Mfn2 and Opa1 in mitochondria were detect using WB and RT-qPCR. Mitochondrial membrane potential, mtROS and ATP of EPCs were measured using IF. H&E staining was used to observe the pathological changes and IMT of the aorta. The expressions of AGEs, MMP-2 and VEGF in aorta were measured using Immunohistochemical (IHC). The levels of SOD, MDA, NO and ET-1 in serum were detected by SOD, MDA and NO kits. RESULTS In vitro, GSC ameliorated the senescence of EPCs induced by D-gal and reduced the expression of P16, P21 and P53. The mitochondrial morphology of EPCs was restored, the expression of mitochondrial Drp1, Mff and Fis1 protein was decreased, the levels of mtROS and ATP were decreased, and mitochondrial function was improved. Meanwhile, the expression of AMPK and p-AMPK increased. The improvement effects of GSC on aging and mitochondrial morphology and function were were hindered after adding AMPK inhibitor. In vivo, GSC improved EPCs efficiency, ameliorated aortic structural disorder and decreased IMT in aging mice. The serum SOD level increased and MDA level decreased, indicating the improvement of antioxidant capacity. Increased NO content and ET-1 content suggested improvement of vascular endothelial function. The changes observed in SOD and MMP-2 suggested a reduction in vascular stiffness and the degree of vascular damage. The decreased expression of P21 and P53 indicates the delay of vascular senescence.
Collapse
Affiliation(s)
- Yinan Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zenghui Niu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xue Wang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chengkui Xiu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanhong Hu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiali Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Yan Lei
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jing Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Liu RS, Li B, Li WD, Du XL, Li XQ. miRNA-130 Promotes Migration and Angiogenesis of Endothelial Progenitor Cells Through PI3K/AKT/mTOR Pathways. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
<sec> <title>Aim:</title> In this study, we aimed to investigate the effects and mechanisms of miRNA-130a in human endothelial progenitor cells (EPCs) involved in Deep vein thrombosis (DVT). </sec> <sec> <title>Methods:</title>
EPCs were isolated and identified by cell morphology and surface marker detection. The effect of miR-130a on the migration, invasion and angiogenesis of EPCs in vitro were also detected. In addition, whether miR-130a is involved in the MMP-1 expression and Akt/PI3K/mTOR signaling
pathway was also demonstrated. </sec> <sec> <title>Results:</title> Results suggested that miRNA-130a promotes migration, invasion, and tube formation of EPCs by positively regulating the expression of MMP-1 through Akt/PI3K/mTOR signaling pathway.
</sec> <sec> <title>Conclusion:</title> Thus, as a potential therapeutic target, miRNA-130a may play an important role in the treatment of DVT. </sec>
Collapse
Affiliation(s)
- Ru-Sheng Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Suzhou, JS 512, China
| | - Bin Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Suzhou, JS 512, China
| | - Wen-Dong Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu Province, Nanjing, JS 25, China
| | - Xiao-Long Du
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu Province, Nanjing, JS 25, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Suzhou, JS 512, China
| |
Collapse
|
3
|
Jiang RC, Zheng XY, Yang SL, Shi HJ, Xi JH, Zou YJ, Dou HQ, Wang YJ, Qin Y, Zhang XL, Xiao Q. CD146 mediates the anti-apoptotic role of Netrin-1 in endothelial progenitor cells under hypoxic conditions. Mol Med Rep 2021; 25:5. [PMID: 34738629 PMCID: PMC8600420 DOI: 10.3892/mmr.2021.12521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/21/2021] [Indexed: 01/18/2023] Open
Abstract
Modulating the biological status of endothelial progenitor cells (EPCs), such as function and survival, is essential for therapeutic angiogenesis in ischemic vascular disease environments. This study aimed to explore the role and molecular mechanisms underlying Netrin-1 in the viability and angiogenic function of EPCs. EPCs were isolated from the bone barrow of adult C57/BL6 mice. The apoptosis and various functions of EPCs were analyzed in vitro by manipulating the expression of Netrin-1. The TUNEL assay was performed to detect apoptotic EPCs. Cell migration and tube formation assays were performed to detect EPC function. Trypan blue staining was performed to detect cell viability. Western blot analysis was performed to detect the protein expression levels of Netrin-1, CD146 and apoptotic factors. Quantitative PCR analysis was performed to detect the expression levels of Netrin-1 receptors. The results demonstrated that treatment with exogenous Netrin-1 promoted EPC migration and tube formation, whereas transfection with small interfering (si)RNA targeting Netrin-1 exhibited the opposite effects. Exogenous Netrin-1 protected EPCs from hypoxia-induced apoptosis, whereas the interruption of endogenous Netrin-1 enhancement under hypoxia by Netrin-1-siRNA exacerbated the apoptosis of EPCs. Furthermore, CD146, one of the immunoglobulin receptors activated by Netrin-1, was screened for in the present study. Results demonstrated that CD146 did not participate in Netrin-1-promoted EPC function, but mediated the anti-apoptotic effects of Netrin-1 in EPCs. In conclusion, Netrin-1 enhanced the angiogenic function of EPCs and alleviated hypoxia-induced apoptosis, which was mediated by CD146. This biological function of Netrin-1 may provide a potential therapeutic option to promote EPCs for the treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Ru-Chao Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Xue-Ying Zheng
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Sheng-Lan Yang
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Hai-Jie Shi
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Jia-Hui Xi
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Yong-Jian Zou
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Hua-Qian Dou
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Yun-Jing Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| | - Xiao-Ling Zhang
- Department of Neonatology, Maternal and Children Hospital of Guangdong Province, Guangzhou, Guangdong 510260, P.R. China
| | - Qing Xiao
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
4
|
Sun Y, Lu Y, Yin L, Liu Z. The Roles of Nanoparticles in Stem Cell-Based Therapy for Cardiovascular Disease. Front Bioeng Biotechnol 2020; 8:947. [PMID: 32923434 PMCID: PMC7457042 DOI: 10.3389/fbioe.2020.00947] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease (CVD) is currently one of the primary causes of mortality and morbidity worldwide. Nanoparticles (NPs) are playing increasingly important roles in regulating stem cell behavior because of their special features, including shape, size, aspect ratio, surface charge, and surface area. In terms of cardiac disease, NPs can facilitate gene delivery in stem cells, track the stem cells in vivo for long-term monitoring, and enhance retention after their transplantation. The advantages of applying NPs in peripheral vascular disease treatments include facilitating stem cell therapy, mimicking the extracellular matrix environment, and utilizing a safe non-viral gene delivery tool. However, the main limitation of NPs is toxicity, which is related to their size, shape, aspect ratio, and surface charge. Currently, there have been many animal models proving NPs’ potential in treating CVD, but no extensive applications of stem-cell therapy using NPs are available in clinical practice. In conclusion, NPs might have significant potential uses in clinical trials of CVD in the future, thereby meeting the changing needs of individual patients worldwide.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuexin Lu
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Yin
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Liu
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Tsai WC, Chiang WH, Wu CH, Li YC, Campbell M, Huang PH, Lin MW, Lin CH, Cheng SM, Chang PC, Cheng CC. miR-548aq-3p is a novel target of Far infrared radiation which predicts coronary artery disease endothelial colony forming cell responsiveness. Sci Rep 2020; 10:6805. [PMID: 32322002 PMCID: PMC7176637 DOI: 10.1038/s41598-020-63311-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/21/2020] [Indexed: 12/24/2022] Open
Abstract
Non-invasive far infrared radiation (FIR) has been observed to improve the health of patients with coronary artery disease (CAD). Endothelial colony forming cells (ECFCs) contribute to vascular repair and CAD. The goal of this study was to uncover the role of FIR in ECFCs function and to reveal potential biomarkers for indication of FIR therapy in CAD patients. FIR significantly enhanced in vitro migration (transwell assay) and tube formation (tube length) capacities in a subpopulation of CAD ECFCs. Clinical parameters associated with the responsiveness of ECFCs to FIR include smoking and gender. ECFCs from CAD patients that smoke did not respond to FIR in most cases. In contrast, ECFCs from females showed a higher responsiveness to FIR than ECFCs from males. To decipher the molecular mechanisms by which FIR modulates ECFCs functions, regardless of sex, RNA sequencing analysis was performed in both genders of FIR-responsive and FIR-non/unresponsive ECFCs. Gene Ontology (GO) analysis of FIR up-regulated genes indicated that the pathways enriched in FIR-responsive ECFCs were involved in cell viability, angiogenesis and transcription. Small RNA sequencing illustrated 18 and 14 miRNAs that are up- and down-regulated, respectively, in FIR-responsive CAD ECFCs in both genders. Among the top 5 up- and down-regulated miRNAs, down-regulation of miR-548aq-3p in CAD ECFCs after FIR treatment was observed in FIR-responsive CAD ECFCs by RT-qPCR. Down-regulation of miR-548aq-3p was correlated with the tube formation activity of CAD ECFCs enhanced by FIR. After establishment of the down-regulation of miR-548aq-3p by FIR in CAD ECFCs, we demonstrated through overexpression and knockdown experiments that miR-548aq-3p contributes to the inhibition of the tube formation of ECFCs. This study suggests the down-regulation of miR-548aq-3p by FIR may contribute to the improvement of ECFCs function, and represents a novel biomarker for therapeutic usage of FIR in CAD patients.
Collapse
Affiliation(s)
- Wei-Che Tsai
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hui Chiang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Hsien Wu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Yue-Cheng Li
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Mel Campbell
- UC Davis Cancer Center, University of California, Davis, California, USA
| | - Po-Hsun Huang
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital and Institute of Clinical Medicine, Taipei, Taiwan
| | - Ming-Wei Lin
- Institute of Public Health, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chi-Hung Lin
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan
| | - Shu-Meng Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Ching Chang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan. .,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.
| | - Cheng-Chung Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
6
|
Liu L, Cui Y, Li X, Que X, Xiao Y, Yang C, Zhang J, Xie X, Cowan PJ, Tian J, Hao H, Liu Z. Concomitant overexpression of triple antioxidant enzymes selectively increases circulating endothelial progenitor cells in mice with limb ischaemia. J Cell Mol Med 2019; 23:4019-4029. [PMID: 30973215 PMCID: PMC6533526 DOI: 10.1111/jcmm.14287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/01/2019] [Accepted: 03/02/2019] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a group of heterogeneous cells in bone marrow (BM) and blood. Ischaemia increases reactive oxygen species (ROS) production that regulates EPC number and function. The present study was conducted to determine if ischaemia‐induced ROS differentially regulated individual EPC subpopulations using a mouse model concomitantly overexpressing superoxide dismutase (SOD)1, SOD3 and glutathione peroxidase. Limb ischaemia was induced by femoral artery ligation in male transgenic mice with their wild‐type littermate as control. BM and blood cells were collected for EPCs analysis and mononuclear cell intracellular ROS production, apoptosis and proliferation at baseline, day 3 and day 21 after ischaemia. Cells positive for c‐Kit+/CD31+ or Sca‐1+/Flk‐1+ or CD34+/CD133+ or CD34+/Flk‐1+ were identified as EPCs. ischaemia significantly increased ROS production and cell apoptosis and decreased proliferation of circulating and BM mononuclear cells and increased BM and circulating EPCs levels. Overexpression of triple antioxidant enzymes effectively prevented ischaemia‐induced ROS production with significantly decreased cell apoptosis and preserved proliferation and significantly increased circulating EPCs level without significant changes in BM EPC populations, associated with enhanced recovery of blood flow and function of the ischemic limb. These data suggested that ischaemia‐induced ROS was differentially involved in the regulation of circulating EPC population.
Collapse
Affiliation(s)
- Lingjuan Liu
- Department of Cardiology, Children's hospital of Chongqing Medical University, Chongqing, China.,Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Yuqi Cui
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Xin Li
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Xingyi Que
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri.,Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Yuan Xiao
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Chunlin Yang
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Jia Zhang
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Xiaoyun Xie
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Peter J Cowan
- Department of Medicine, University of Melbourne, Melbourne, Australia.,Immunology Research Centre, St. Vincent's Hospital, Melbourne, Australia
| | - Jie Tian
- Department of Cardiology, Children's hospital of Chongqing Medical University, Chongqing, China
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
7
|
Osipova O, Saaya S, Karpenko A, Zakian S, Aboian E. Cell therapy of critical limb ischemia-problems and prospects. VASA 2019; 48:461-471. [PMID: 30969159 DOI: 10.1024/0301-1526/a000787] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell therapy is proposed for indirect revascularization for the patient's incurable by endovascular or surgical revascularization. The therapy with stem cells (SCs) or progenitor cells is assumed to be more efficient as compared with protein or gene therapy not only because of their direct vasculogenic properties, but also thanks to their paracrine effect via secretion of manifold biologically active substances. This review gives an overview of the potential of SC-based therapy for critical limb ischemia (CLI), putative mechanism underlying cell therapy, and comparison of cell therapy to angiogenesis gene therapy in CLI treatment. Human trial data and meta-analysis, as well as some problems of clinical trials and considerations for future SC-based therapy in CLI are also discussed.
Collapse
Affiliation(s)
- Olesia Osipova
- Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Shoraan Saaya
- Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Andrei Karpenko
- Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Suren Zakian
- Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Edouard Aboian
- Division of Vascular Surgery, Palo Alto Medical Foundation, Burlingame, USA
| |
Collapse
|
8
|
Continuous Passive Motion Promotes and Maintains Chondrogenesis in Autologous Endothelial Progenitor Cell-Loaded Porous PLGA Scaffolds during Osteochondral Defect Repair in a Rabbit Model. Int J Mol Sci 2019; 20:ijms20020259. [PMID: 30634691 PMCID: PMC6358980 DOI: 10.3390/ijms20020259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/27/2018] [Accepted: 01/02/2019] [Indexed: 11/19/2022] Open
Abstract
Continuous passive motion (CPM) is widely used after total knee replacement. In this study, we investigated the effect of CPM combined with cell-based construct-transplantation in osteochondral tissue engineering. We created osteochondral defects (3 mm in diameter and 3 mm in depth) in the medial femoral condyle of 36 knees and randomized them into three groups: ED (empty defect), EPC/PLGA (endothelial progenitor cells (EPCs) seeded in the poly lactic-co-glycolic acid (PLGA) scaffold), or EPC/PLGA/CPM (EPC/PLGA scaffold complemented with CPM starting one day after transplantation). We investigated the effects of CPM and the EPC/PLGA constructs on tissue restoration in weight-bearing sites by histological observation and micro-computed tomography (micro-CT) evaluation 4 and 12 weeks after implantation. After CPM, the EPC/PLGA construct exhibited early osteochondral regeneration and prevention of subchondral bone overgrowth and cartilage degeneration. CPM did not alter the microenvironment created by the construct; it up-regulated the expression of the extracellular matrix components (glycosaminoglycan and collagen), down-regulated bone formation, and induced the biosynthesis of lubricin, which appeared in the EPC/PLGA/CPM group after 12 weeks. CPM can provide promoting signals during osteochondral tissue engineering and achieve a synergistic effect when combined with EPC/PLGA transplantation, so it should be considered a non-invasive treatment to be adopted in clinical practices.
Collapse
|
9
|
Royer C, Bégin AAG, Plawinski L, Lévesque L, Durrieu MC, Laroche G. Validation of reference genes for real-time PCR of cord blood mononuclear cells, differentiating endothelial progenitor cells, and mature endothelial cells. Exp Cell Res 2018; 370:389-398. [PMID: 30146063 DOI: 10.1016/j.yexcr.2018.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/06/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
Abstract
In the last ten years, endothelial progenitor cells (EPCs) have gained interest as an attractive cell population in regenerative medicine for vascular applications. This population is defined as the precursor of endothelial mature cells (ECs) through a process of differentiation. To our knowledge, no single marker can be used to discriminate them from mature ECs. To effectively study their differentiation kinetics, gene expression must be assessed. Quantitative real-time PCR (RT-qPCR) is widely used to analyze gene expression. To minimize the impact of variances from RT-qPCR, a rigorous selection of reference genes must be performed prior to any experiments due to variations in experimental conditions. In this study, CD34+ mononuclear cells were extracted from human cord blood and differentiated into EPCs after seeding for a maximum period of 21 days. To choose the best combinations of reference genes, we compared the results of EPCs, CD34+ mononuclear cells, and mature endothelial cells to ensure that the differentiation kinetics did not affect the expression of our selected reference genes. The expression levels of seven genes, namely, YWHAZ, GAPDH, HPRT1, RPLP0, UBC, B2M, and TBP were thus compared. The algorithms geNorm, NormFinder, BestKeeper, and the Comparative ΔCt method were employed to assess the expression of each candidate gene. Overall results reveal that the expression stability of reference genes may differ depending on the statistical program used. YWHAZ, GAPDH, and UBC composed the optimal set of reference genes for the gene expression studies performed by RT-qPCR in our experimental conditions. This work can thus serve as a starting point for the selection of candidate reference genes to normalize the levels of gene expression in endothelial progenitor cell populations.
Collapse
Affiliation(s)
- Caroline Royer
- Univ. Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Pessac, France; CNRS, CBMN UMR5248, Pessac, France; Bordeaux INP, CBMN UMR5248, Pessac, France; Laboratoire d'Ingénierie de Surface, Centre de recherche du CHU de Québec - Université Laval, Hôpital Saint-François d'Assise, Québec, Canada; Département de Génie des Mines, de la Métallurgie et des Matériaux, Centre de Recherche sur les Matériaux Avancés, Université Laval, Québec, Canada
| | - Andrée-Anne Guay Bégin
- Laboratoire d'Ingénierie de Surface, Centre de recherche du CHU de Québec - Université Laval, Hôpital Saint-François d'Assise, Québec, Canada
| | - Laurent Plawinski
- Univ. Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Pessac, France; CNRS, CBMN UMR5248, Pessac, France; Bordeaux INP, CBMN UMR5248, Pessac, France
| | - Lucie Lévesque
- Laboratoire d'Ingénierie de Surface, Centre de recherche du CHU de Québec - Université Laval, Hôpital Saint-François d'Assise, Québec, Canada
| | - Marie-Christine Durrieu
- Univ. Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Pessac, France; CNRS, CBMN UMR5248, Pessac, France; Bordeaux INP, CBMN UMR5248, Pessac, France
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface, Centre de recherche du CHU de Québec - Université Laval, Hôpital Saint-François d'Assise, Québec, Canada; Département de Génie des Mines, de la Métallurgie et des Matériaux, Centre de Recherche sur les Matériaux Avancés, Université Laval, Québec, Canada.
| |
Collapse
|
10
|
Wang RY, Liu LH, Liu H, Wu KF, An J, Wang Q, Liu Y, Bai LJ, Qi BM, Qi BL, Zhang L. Nrf2 protects against diabetic dysfunction of endothelial progenitor cells via regulating cell senescence. Int J Mol Med 2018; 42:1327-1340. [PMID: 29901179 PMCID: PMC6089760 DOI: 10.3892/ijmm.2018.3727] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes is associated with an increased risk of cardio-vascular disease. A decrease in the number and functionality of endothelial progenitor cells (EPCs) leads to reduced endothelial repair and the development of cardiovascular disease. The aim of the present study was to explore the effect and underlying mechanisms of nuclear factor erythroid 2-related factor 2 (Nrf2) on EPC dysfunction caused by diabetic mellitus. The biological functions of EPCs in streptozotocin-induced diabetic mice were evaluated, including migration, proliferation, angiogenesis and the secretion of vascular endothelial growth factor (VEGF), stromal-derived growth factor (SDF) and nitric oxide (NO). Oxidative stress levels in diabetic EPCs were also assessed by detecting intracellular reactive oxygen species (ROS), superoxide dismutase (SOD) and malondialdehyde (MDA). EPC senescence was evaluated by measuring p16 and b-gal expression and observing the senescence-associated secretory phenotype. In addition, the function of EPCs and level of oxidative stress were assessed following Nrf2 silencing or activation. Nrf2 silencing resulted in a decrease of EPC biological functions, accelerated cell senescence and increased oxidative stress, as indicated by ROS and MDA upregulation accompanied with decreased SOD activity. Furthermore, Nrf2 silencing inhibited migration, proliferation and secretion in EPCs, while it increased oxidative stress and cell senescence. Nrf2 activation protected diabetic EPCs against the effects of oxidative stress and cell senescence, ameliorating the biological dysfunction of EPCs derived from mice with diabetes. In conclusion, Nrf2 overexpression protected against oxidative stress-induced functional damage in EPCs derived from diabetic mice by regulating cell senescence.
Collapse
Affiliation(s)
- Rui-Yun Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Li-Hua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongxia Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ke-Fei Wu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing An
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qian Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Li-Juan Bai
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ben-Ming Qi
- Department of Otorhinolaryngology, First People's Hospital of Yunnan Province, Kunming, Yunnan 650000, P.R. China
| | - Ben-Ling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lei Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
11
|
Tournois C, Pignon B, Sevestre MA, Al-Rifai R, Creuza V, Poitevin G, François C, Nguyen P. Cell therapy in critical limb ischemia: A comprehensive analysis of two cell therapy products. Cytotherapy 2017; 19:299-310. [DOI: 10.1016/j.jcyt.2016.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/13/2022]
|
12
|
Safety and Effectiveness of Bone Marrow Cell Concentrate in the Treatment of Chronic Critical Limb Ischemia Utilizing a Rapid Point-of-Care System. Stem Cells Int 2017; 2017:4137626. [PMID: 28194186 PMCID: PMC5282442 DOI: 10.1155/2017/4137626] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/14/2016] [Indexed: 01/09/2023] Open
Abstract
Critical limb ischemia (CLI) is the end stage of lower extremity peripheral vascular disease (PVD) in which severe obstruction of blood flow results in ischemic rest pain, ulcers and/or gangrene, and a significant risk of limb loss. This open-label, single-arm feasibility study evaluated the safety and therapeutic effectiveness of autologous bone marrow cell (aBMC) concentrate in revascularization of CLI patients utilizing a rapid point-of-care device. Seventeen (17) no-option CLI patients with ischemic rest pain were enrolled in the study. Single dose of aBMC, prepared utilizing an intraoperative point-of-care device, the Res-Q™ 60 BMC system, was injected intramuscularly into the afflicted limb and patients were followed up at regular intervals for 12 months. A statistically significant improvement in Ankle Brachial Index (ABI), Transcutaneous Oxygen Pressure (TcPO2), mean rest pain and intermittent claudication pain scores, wound/ ulcer healing, and 6-minute walking distance was observed following aBMC treatment. Major amputation-free survival (mAFS) rate and amputation-free rates (AFR) at 12 months were 70.6% and 82.3%, respectively. In conclusion, aBMC injections were well tolerated with improved tissue perfusion, confirming the safety, feasibility, and preliminary effectiveness of aBMC treatment in CLI patients.
Collapse
|
13
|
Brenner C, Kränkel N, Kühlenthal S, Israel L, Remm F, Fischer C, Herbach N, Speer T, Grabmaier U, Laskowski A, Gross L, Theiss H, Wanke R, Landmesser U, Franz WM. Short-term inhibition of DPP-4 enhances endothelial regeneration after acute arterial injury via enhanced recruitment of circulating progenitor cells. Int J Cardiol 2014; 177:266-75. [PMID: 25499391 DOI: 10.1016/j.ijcard.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/06/2014] [Accepted: 09/15/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Endothelial injuries regularly occur in atherosclerosis and during interventional therapies of the arterial occlusive disease. Disturbances in the endothelial integrity can lead to insufficient blood supply and bear the risk of thrombus formation and acute vascular occlusion. At present, effective therapeutics to restore endothelial integrity are barely available. We analyzed the effect of pharmacological DPP-4-inhibition by Sitagliptin on endogenous progenitor cell-based endothelial regeneration via the SDF-1α/CXCR4-axis after acute endothelial damage in a mouse model of carotid injury. METHODS AND RESULTS Induction of a defined endothelial injury was performed in the carotid artery of C57Bl/6 mice which led to a local upregulation of SDF-1α expression. Animals were treated with placebo, Sitagliptin or Sitagliptin+AMD3100. Using mass spectrometry we could prove that Sitagliptin prevented cleavage of the chemokine SDF-1α. Accordingly, increased SDF-1α concentrations enhanced recruitment of systemically applied and endogenous circulating CXCR4+ progenitor cells to the site of vascular injury followed by a significantly accelerated reendothelialization as compared to placebo-treated animals. Improved endothelial recovery, as well as recruitment of circulating CXCR4+ progenitor cells (CD133+, Flk1+), was reversed by CXCR4-antagonization through AMD3100. In addition, short-term Sitagliptin treatment did not significantly promote neointimal or medial hyperplasia. CONCLUSION Sitagliptin can accelerate endothelial regeneration after acute endothelial injury. DPP-4 inhibitors prevent degradation of the chemokine SDF-1α and thus improve the recruitment of regenerative circulating CXCR4+ progenitor cells which mediate local endothelial cell proliferation without adversely affecting vessel wall architecture.
Collapse
Affiliation(s)
- Christoph Brenner
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| | - Nicolle Kränkel
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Sarah Kühlenthal
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lars Israel
- Institute of Molecular Biology, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Friederike Remm
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Cornelia Fischer
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Timo Speer
- Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Ulrich Grabmaier
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Alexandra Laskowski
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lisa Gross
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Hans Theiss
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Ulf Landmesser
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Wolfgang-Michael Franz
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Hung HS, Yang YC, Lin YC, Lin SZ, Kao WC, Hsieh HH, Chu MY, Fu RH, Hsu SH. Regulation of human endothelial progenitor cell maturation by polyurethane nanocomposites. Biomaterials 2014; 35:6810-21. [DOI: 10.1016/j.biomaterials.2014.04.076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/22/2014] [Indexed: 12/24/2022]
|
15
|
Kang BJ, Lee SH, Kweon OK, Cho JY. Differentiation of canine adipose tissue–derived mesenchymal stem cells towards endothelial progenitor cells. Am J Vet Res 2014; 75:685-91. [DOI: 10.2460/ajvr.75.7.685] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Kim BR, Jang IH, Shin SH, Kwon YW, Heo SC, Choi EJ, Lee JS, Kim JH. Therapeutic angiogenesis in a murine model of limb ischemia by recombinant periostin and its fasciclin I domain. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1324-32. [PMID: 24834847 DOI: 10.1016/j.bbadis.2014.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 04/21/2014] [Accepted: 05/05/2014] [Indexed: 01/03/2023]
Abstract
Periostin, an extracellular matrix protein, is expressed in injured tissues, such as the heart with myocardial infarction, and promotes angiogenesis and tissue repair. However, the molecular mechanism associated with periostin-stimulated angiogenesis and tissue repair is still unclear. In order to clarify the role of periostin in neovascularization, we examined the effect of periostin in angiogenic potentials of human endothelial colony forming cells (ECFCs) in vitro and in an ischemic limb animal model. Recombinant periostin protein stimulated the migration and tube formation of ECFCs. To identify the functional domains of periostin implicated in angiogenesis, five fragments of periostin, including four repeating FAS-1 domains and a carboxyl terminal domain, were expressed in Escherichia coli and purified to homogeneity. Of the five different domains, the first FAS-1 domain stimulated the migration and tube formation of human ECFCs as potent as the whole periostin. Chemotactic migration of ECFCs induced by the full length and the first FAS-1 domain of periostin was abrogated by blocking antibodies against β3 and β5 integrins. Intramuscular injection of the full length and the first FAS-1 domain of periostin into the ischemic hindlimb of mice attenuated severe limb loss and promoted blood perfusion and homing of intravenously administered ECFCs to the ischemic limb. These results suggest that the first FAS-1 domain is responsible for periostin-induced migration and angiogenesis and it can be used as a therapeutic tool for treatment of peripheral artery occlusive disease by stimulating homing of ECFCs.
Collapse
Affiliation(s)
- Ba Reun Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Sang Hun Shin
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Yang Woo Kwon
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Soon Chul Heo
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Eun-Jung Choi
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Jung Sub Lee
- Department of Orthopaedic Surgery, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Jae Ho Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea; Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 626-770, Gyeongsangnam-do, Republic of Korea.
| |
Collapse
|
17
|
Quaranta P, Antonini S, Spiga S, Mazzanti B, Curcio M, Mulas G, Diana M, Marzola P, Mosca F, Longoni B. Co-transplantation of endothelial progenitor cells and pancreatic islets to induce long-lasting normoglycemia in streptozotocin-treated diabetic rats. PLoS One 2014; 9:e94783. [PMID: 24733186 PMCID: PMC3986409 DOI: 10.1371/journal.pone.0094783] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/20/2014] [Indexed: 01/09/2023] Open
Abstract
Graft vascularization is a crucial step to obtain stable normoglycemia in pancreatic islet transplantation. Endothelial progenitor cells (EPCs) contribute to neoangiogenesis and to the revascularization process during ischaemic events and play a key role in the response to pancreatic islet injury. In this work we co-transplanted EPCs and islets in the portal vein of chemically-induced diabetic rats to restore islet vascularization and to improve graft survival. Syngenic islets were transplanted, either alone or with EPCs derived from green fluorescent protein (GFP) transgenic rats, into the portal vein of streptozotocin-induced diabetic rats. Blood glucose levels were monitored and intraperitoneal glucose tolerance tests were performed. Real time-PCR was carried out to evaluate the gene expression of angiogenic factors. Diabetic-induced rats showed long-lasting (6 months) normoglycemia upon co-transplantation of syngenic islets and EPCs. After 3–5 days from transplantation, hyperglycaemic levels dropped to normal values and lasted unmodified as long as they were checked. Further, glucose tolerance tests revealed the animals' ability to produce insulin on-demand as indexed by a prompt response in blood glucose clearance. Graft neovascularization was evaluated by immunohistochemistry: for the first time the measure of endothelial thickness revealed a donor-EPC-related neovascularization supporting viable islets up to six months after transplant. Our results highlight the importance of a newly formed viable vascular network together with pancreatic islets to provide de novo adequate supply in order to obtain enduring normoglycemia and prevent diabetes-related long-term health hazards.
Collapse
Affiliation(s)
- Paola Quaranta
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Sara Antonini
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Benedetta Mazzanti
- Department of Clinical and Experimental Medicine, Haematology Section, University of Florence, Florence, Italy
| | - Michele Curcio
- U.O. Immunohaematology, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Marco Diana
- Department of Chemistry and Pharmacy, “G. Minardi” Laboratory of Cognitive Neuroscience, University of Sassari, Sassari, Italy
| | - Pasquina Marzola
- Department of Computer Science, University of Verona, Verona, Italy
| | - Franco Mosca
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Biancamaria Longoni
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
- * E-mail:
| |
Collapse
|
18
|
Bradykinin preconditioning improves therapeutic potential of human endothelial progenitor cells in infarcted myocardium. PLoS One 2013; 8:e81505. [PMID: 24312554 PMCID: PMC3846887 DOI: 10.1371/journal.pone.0081505] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/14/2013] [Indexed: 12/29/2022] Open
Abstract
Objectives Stem cell preconditioning (PC) is a powerful approach in reducing cell death after transplantation. We hypothesized that PC human endothelial progenitor cells (hEPCs) with bradykinin (BK) enhance cell survival, inhibit apoptosis and repair the infarcted myocardium. Methods The hEPCs were preconditioned with or without BK. The hEPCs apoptosis induced by hypoxia along with serum deprivation was determined by annexin V-fluorescein isothiocyanate/ propidium iodide staining. Cleaved caspase-3, Akt and eNOS expressions were determined by Western blots. Caspase-3 activity and vascular endothelial growth factor (VEGF) levels were assessed in hEPCs. For invivo studies, the survival and cardiomyocytes apoptosis of transplanted hEPCs were assessed using 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindodi- carbocyanine,4-chlorobenzenesul-fonate salt labeled hEPCs and TUNEL staining. Infarct size and cardiac function were measured at 10 days after transplantation, and the survival of transplanted hEPCs were visualized using near-infrared optical imaging. Results Invitro data showed a marked suppression in cell apoptosis following BK PC. The PC reduced caspase-3 activation, increased the Akt, eNOS phosphorylation and VEGF levels. Invivo data in preconditioned group showed a robust cell anti-apoptosis, reduction in infarct size, and significant improvement in cardiac function. The effects of BK PC were abrogated by the B2 receptor antagonist HOE140, the Akt and eNOS antagonists LY294002 and L-NAME, respectively. Conclusions The activation of B2 receptor-dependent PI3K/Akt/eNOS pathway by BK PC promotes VEGF secretion, hEPC survival and inhibits apoptosis, thereby improving cardiac function invivo. The BK PC hEPC transplantation for stem cell-based therapies is a novel approach that has potential for clinical used.
Collapse
|
19
|
Abstract
BACKGROUND Obstructive sleep apnea (OSA) occurs in 4% of middle-aged men and 2% of middle-aged women in the general population, and the prevalence is even higher in specific patient groups. OSA is an independent risk factor for a variety of cardiovascular diseases. Endothelial injury could be the pivotal determinant in the development of cardiovascular pathology in OSA. Endothelial damage ultimately represents a dynamic balance between the magnitude of injury and the capacity for repair. Bone marrow-derived endothelial progenitor cells (EPCs) within adult peripheral blood present a possible means of vascular maintenance that could home to sites of injury and restore endothelial integrity and normal function. METHODS We summarized pathogenetic mechanisms of OSA and searched for available studies on numbers and functions of EPCs in patients with OSA to explore the potential links between the numbers and functions of EPCs and OSA. In particular, we tried to elucidate the molecular mechanisms of the effects of OSA on EPCs. CONCLUSION Intermittent hypoxia cycles and sleep fragmentation are major pathophysiologic characters of OSA. Intermittent hypoxia acts as a trigger of oxidative stress, systemic inflammation, and sympathetic activation. Sleep fragmentation is associated with a burst of sympathetic activation and systemic inflammation. In most studies, a reduction in circulating EPCs has emerged. The possible mechanisms underlying the decrease in the number or function of EPCs include prolonged inflammation response, oxidative stress, increased sympathetic activation, physiological adaptive responses of tissue to hypoxia, reduced EPC mobilization, EPC apoptosis, and functional impairment in untreated OSA. Continuous positive airway pressure (CPAP) therapy for OSA affects the mobilization, apoptosis, and function of EPCs through preventing intermittent hypoxia episodes, improving sleep quality, and reducing systemic inflammation, oxidative stress levels, and sympathetic overactivation. To improve CPAP adherence, the medical staff should pay attention to making the titration trial a comfortable first CPAP experience for the patients; for example, using the most appropriate ventilators or proper humidification. It is also important to give the patients education and support about CPAP use in the follow-up, especially in the early stage of the treatment.
Collapse
Affiliation(s)
- Qing Wang
- The Second Respiratory Department of the First People’s Hospital of Kunming, Yunnan, People’s Republic of China
| | - Qi Wu
- Tianjin Haihe Hospital, Tianjin, People’s Republic of China
| | - Jing Feng
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, NC, USA
- Correspondence: Jing Feng, Respiratory Department of Tianjin Medical University General Hospital, Tianjin 300052, People’s Republic of China, Email
| | - Xin Sun
- Respiratory Department of Tianjin Haihe Hospital, Tianjin, People’s Republic of China
- Xin Sun, Respiratory Department of Tianjin Haihe Hospital, Tianjin 300350, People’s Republic of China, Email
| |
Collapse
|
20
|
Mechanisms underlying protective effects of trimetazidine on endothelial progenitor cells biological functions against H2O2-induced injury: Involvement of antioxidation and Akt/eNOS signaling pathways. Eur J Pharmacol 2013; 707:87-94. [DOI: 10.1016/j.ejphar.2013.03.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 11/22/2022]
|
21
|
Teraa M, Sprengers RW, Westerweel PE, Gremmels H, Goumans MJTH, Teerlink T, Moll FL, Verhaar MC. Bone marrow alterations and lower endothelial progenitor cell numbers in critical limb ischemia patients. PLoS One 2013; 8:e55592. [PMID: 23383236 PMCID: PMC3561321 DOI: 10.1371/journal.pone.0055592] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is characterized by lower extremity artery obstruction and a largely unexplained impaired ischemic neovascularization response. Bone marrow (BM) derived endothelial progenitor cells (EPC) contribute to neovascularization. We hypothesize that reduced levels and function of circulating progenitor cells and alterations in the BM contribute to impaired neovascularization in CLI. METHODS Levels of primitive (CD34(+) and CD133(+)) progenitors and CD34(+)KDR(+) EPC were analyzed using flow cytometry in blood and BM from 101 CLI patients in the JUVENTAS-trial (NCT00371371) and healthy controls. Blood levels of markers for endothelial injury (sE-selectin, sICAM-1, sVCAM-1, and thrombomodulin), and progenitor cell mobilizing and inflammatory factors were assessed by conventional and multiplex ELISA. BM levels and activity of the EPC mobilizing protease MMP-9 were assessed by ELISA and zymography. Circulating angiogenic cells (CAC) were cultured and their paracrine function was assessed. RESULTS Endothelial injury markers were higher in CLI (P<0.01). CLI patients had higher levels of VEGF, SDF-1α, SCF, G-CSF (P<0.05) and of IL-6, IL-8 and IP-10 (P<0.05). Circulating EPC and BM CD34(+) cells (P<0.05), lymphocytic expression of CXCR4 and CD26 in BM (P<0.05), and BM levels and activity of MMP-9 (P<0.01) were lower in CLI. Multivariate regression analysis showed an inverse association between IL-6 and BM CD34(+) cell levels (P = 0.007). CAC from CLI patients had reduced paracrine function (P<0.0001). CONCLUSION CLI patients have reduced levels of circulating EPC, despite profound endothelial injury and an EPC mobilizing response. Moreover, CLI patients have lower BM CD34(+)-cell levels, which were inversely associated with the inflammatory marker IL-6, and lower BM MMP-9 levels and activity. The results of this study suggest that inflammation-induced BM exhaustion and a disturbed progenitor cell mobilization response due to reduced levels and activity of MMP-9 in the BM and alterations in the SDF-1α/CXCR4 interaction contribute to the attenuated neovascularization in CLI patients.
Collapse
Affiliation(s)
- Martin Teraa
- Department of Nephrology & Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ralf W. Sprengers
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter E. Westerweel
- Department of Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Nephrology & Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Tom Teerlink
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Frans L. Moll
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology & Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Kawakami Y, Ii M, Alev C, Kawamoto A, Matsumoto T, Kuroda R, Shoji T, Fukui T, Masuda H, Akimaru H, Mifune Y, Kuroda T, Horii M, Yokoyama A, Kurosaka M, Asahara T. Local Transplantation of Ex Vivo Expanded Bone Marrow-Derived CD34-Positive Cells Accelerates Fracture Healing. Cell Transplant 2012; 21:2689-709. [DOI: 10.3727/096368912x654920] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transplantation of bone marrow (BM) CD34+ cells, an endothelial/hematopoietic progenitor-enriched cell population, has shown therapeutic efficiency in the treatment of ischemic diseases enhancing neovascularization. However, the number of CD34+ cells obtained from bone marrow is not sufficient for routine clinical application. To overcome this issue, we developed a more efficient and clinically applicable CD34+ cell expansion method. Seven-day ex vivo expansion culture of BM CD34+ cells with a cocktail of five growth factors containing VEGF, SCF, IL-6, Flt-3 ligand, and TPO resulted in reproducible more than 20-fold increase in cell number. The favorable effect of the local transplantation of culture expanded (cEx)-BM CD34+ cells on rat unhealing fractures was equivalent or higher than that of nonexpanded (fresh) BM CD34+ cells exhibiting sufficient therapeutic outcome with frequent vasculogenic/osteogenic differentiation of transplanted cEx-BM CD34+ cells and fresh BM CD34+ cells as well as intrinsic enhancement of angiogenesis/osteogenesis at the treated fracture sites. Specifically, cEx-BM CD34+ cell treatment demonstrated the best blood flow recovery at fracture sites compared with the nonexpanded BM CD34+ cells. In vitro, cEx-BM CD34+ cells showed higher colony/tube-forming capacity than nonexpanded BM CD34+ cells. Both cells demonstrated differentiation potential into osteoblasts. Since fresh BM CD34+ cells can be easily collected from fracture sites at the time of primary operation and stored for future use, autologous cEx-BM CD34+ cell transplantation would be not only a simple but also a promising therapeutic strategy for unhealing fractures in the field of orthopedic trauma surgery.
Collapse
Affiliation(s)
- Yohei Kawakami
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Masaaki Ii
- Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Cantas Alev
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Laboratory for Early Embryogenesis, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Atsuhiko Kawamoto
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Tomoyuki Matsumoto
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Taro Shoji
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Tomoaki Fukui
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Haruchika Masuda
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroshi Akimaru
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Yutaka Mifune
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Tomoya Kuroda
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Miki Horii
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Ayumi Yokoyama
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takayuki Asahara
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
23
|
Endothelial progenitor cells: the promise of cell-based therapies for acute lung injury. Inflamm Res 2012; 62:3-8. [PMID: 23138575 DOI: 10.1007/s00011-012-0570-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 10/03/2012] [Accepted: 10/22/2012] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are defined as a special type of stem cell that have been found to directly incorporate into injured vessels and that participate in angiogenesis and reconstruction by differentiation into endothelial cells. EPCs are widely used to therapeutically treat cardiovascular disease, limb ischemia and vascular repair. However, the role of EPCs in inflammatory diseases, especially in lung injury, is less studied. OBJECTIVE To investigate the application of EPCs to vascular repair, and the role of EPCs in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). METHODS A computer-based online search was performed in the PubMed database and Web of Science database for articles published, concerning EPCs, angiogenesis, ALI/ARDS and stem cell transplantation CONCLUSION EPCs have a therapeutic potential for vascular regeneration and may emerge as novel strategy for the diseases that are associated with ALI/ARDS.
Collapse
|
24
|
Janeczek Portalska K, Leferink A, Groen N, Fernandes H, Moroni L, van Blitterswijk C, de Boer J. Endothelial differentiation of mesenchymal stromal cells. PLoS One 2012; 7:e46842. [PMID: 23056481 PMCID: PMC3464214 DOI: 10.1371/journal.pone.0046842] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/06/2012] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stromal cells (hMSCs) are increasingly used in regenerative medicine for restoring worn-out or damaged tissue. Newly engineered tissues need to be properly vascularized and current candidates for in vitro tissue pre-vascularization are endothelial cells and endothelial progenitor cells. However, their use in therapy is hampered by their limited expansion capacity and lack of autologous sources. Our approach to engineering large grafts is to use hMSCs both as a source of cells for regeneration of targeted tissue and at the same time as the source of endothelial cells. Here we investigate how different stimuli influence endothelial differentiation of hMSCs. Although growth supplements together with shear force were not sufficient to differentiate hMSCs with respect to expression of endothelial markers such as CD31 and KDR, these conditions did prime the cells to differentiate into cells with an endothelial gene expression profile and morphology when seeded on Matrigel. In addition, we show that endothelial-like hMSCs are able to create a capillary network in 3D culture both in vitro and in vivo conditions. The expansion phase in the presence of growth supplements was crucial for the stability of the capillaries formed in vitro. To conclude, we established a robust protocol for endothelial differentiation of hMSCs, including an immortalized MSC line (iMSCs) which allows for reproducible in vitro analysis in further studies.
Collapse
Affiliation(s)
- Karolina Janeczek Portalska
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Anne Leferink
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Nathalie Groen
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Hugo Fernandes
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Lorenzo Moroni
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Clemens van Blitterswijk
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Jan de Boer
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| |
Collapse
|
25
|
Bennis Y, Sarlon-Bartoli G, Guillet B, Lucas L, Pellegrini L, Velly L, Blot-Chabaud M, Dignat-Georges F, Sabatier F, Pisano P. Priming of late endothelial progenitor cells with erythropoietin before transplantation requires the CD131 receptor subunit and enhances their angiogenic potential. J Thromb Haemost 2012; 10:1914-28. [PMID: 22738133 DOI: 10.1111/j.1538-7836.2012.04835.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Endothelial colony-forming cells (ECFCs) are promising candidates for cell therapy of ischemic diseases. Erythropoietin (EPO) is a cytokine that promotes angiogenesis after ischemic injury. EPO receptors (EPORs) classically include two EPOR subunits, but may also associate with the β-common chain (CD131) in a newly identified receptor involved in EPO cytoprotective effects. OBJECTIVE The aim was to take advantage of the proangiogenic properties of EPO to enhance ECFC graft efficiency. We postulated that priming ECFCs by adding epoietin α in culture medium prior to experiments might increase their angiogenic properties. We also explored the role of the CD131 subunit in EPO priming of ECFCs. METHODS AND RESULTS By western blotting on cord blood ECFC lysates, we showed that EPOR and CD131 expression increased significantly after EPO priming. These proteins coimmunoprecipitated and colocalized, suggesting that they are covalently bound in ECFCs. EPO at 5 IU mL(-1) significantly stimulated proliferation, wound healing, migration and tube formation of ECFCs. EPO priming also increased ECFC resistance to H2 O2-induced apoptosis and survival in vivo. Similarly, in vivo studies showed that, as compared with non-primed ECFC injection, 5 IU mL(-1) EPO-primed ECFCs, injected intravenously 24 h after hindlimb ischemia in athymic nude mice, increased the ischemic/non-ischemic ratios of hindlimb blood flow and capillary density. These effects were all prevented by CD131 small interfering RNA transfection, and involved the phosphoinositide 3-kinase-Akt pathway. CONCLUSION These results highlight the potential role of EPO-primed ECFCs for cell-based therapy in hindlimb ischemia, and underline the critical role of CD131 as an EPO coreceptor.
Collapse
Affiliation(s)
- Y Bennis
- Aix-Marseille Université, UMR INSERM1076, Faculté de Pharmacie, Marseille, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kaneko Y, Tajiri N, Yu S, Hayashi T, Stahl CE, Bae E, Mestre H, Franzese N, Rodrigues A, Rodrigues MC, Ishikawa H, Shinozuka K, Hethorn W, Weinbren N, Glover LE, Tan J, Achyuta AH, van Loveren H, Sanberg PR, Shivsankar S, Borlongan CV. Nestin overexpression precedes caspase-3 upregulation in rats exposed to controlled cortical impact traumatic brain injury. CELL MEDICINE 2012; 4:55-63. [PMID: 23101029 DOI: 10.3727/215517912x639306] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Our understanding of biological mechanisms and treatment options for traumatic brain injury (TBI) is limited. Here, we employed quantitative real-time PCR (QRT-PCR) and immunohistochemical analyses to determine the dynamic expression of cell proliferation and apoptosis in an effort to provide insights into the therapeutic window for developing regenerative strategies for TBI. For this purpose, young adult Sprague-Dawley rats were subjected to experimental TBI using a controlled cortical impactor, then euthanized 1-48 hours after TBI for QRT-PCR and immunohistochemistry. QRT-PCR revealed that brains from TBI exposed rats initially displayed nestin mRNA expression that modestly increased as early as 1-hour post-TBI, then significantly peaked at 8 hours, but thereafter reverted to pre-TBI levels. On the other hand, caspase-3 mRNA expression was slightly elevated at 8 hours post-TBI, which did not become significantly upregulated until 48 hours. Immunofluorescent microscopy revealed a significant surge in nestin immunoreactive cells in the cortex, corpus callosum, and subventricular zone at 24 hours post-TBI, whereas a significant increase in the number of active caspase-3 immunoreactive cells was only found in the cortex and not until 48 hours. These results suggest that the injured brain attempts to repair itself via cell proliferation immediately after TBI, but that this endogenous regenerative mechanism is not sufficient to abrogate the secondary apoptotic cell death. Treatment strategies designed to amplify cell proliferation and to prevent apoptosis are likely to exert maximal benefits when initiated at the acute phase of TBI.
Collapse
Affiliation(s)
- Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Xu DY, Davis BB, Wang ZH, Zhao SP, Wasti B, Liu ZL, Li N, Morisseau C, Chiamvimonvat N, Hammock BD. A potent soluble epoxide hydrolase inhibitor, t-AUCB, acts through PPARγ to modulate the function of endothelial progenitor cells from patients with acute myocardial infarction. Int J Cardiol 2012; 167:1298-304. [PMID: 22525341 DOI: 10.1016/j.ijcard.2012.03.167] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Revised: 03/21/2012] [Accepted: 03/30/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Epoxyeicosatrienoic acids (EETs) are natural angiogenic mediators regulated by soluble epoxide hydrolase (sEH). Inhibitors of sEH can stabilize EETs levels and were reported to reduce atherosclerosis and inhibit myocardial infarction in animal models. In this work, we investigated whether increasing EETs with the sEH inhibitor t-AUCB would increase angiogenesis related function in endothelial progenitor cells (EPCs) from patients with acute myocardial infarction (AMI). METHODS AND RESULTS EPCs were isolated from 50 AMI patients and 50 healthy subjects (control). EPCs were treated with different concentrations of t-AUCB for 24h with or without peroxisome proliferator activated receptor γ (PPARγ) inhibitor GW9662. Migration of EPCs was assayed in trans-well chambers. Angiogenesis assays were performed using a Matrigel-Matrix in vitro model. The expression of vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1α (HIF-1α) mRNA and protein in EPCs was measured by real-time PCR or Western blot, respectively. Also, the concentration of EETs in the culture supernatant was detected by ELISA. The activity of EPCs in the AMI patient group was reduced compared to healthy controls. Whereas increasing EET levels with t-AUCB promoted a dose dependent angiogenesis and migration in EPCs from AMI patients. Additionally, the t-AUCB dose dependently increased the expression of the angiogenic factors VEGF and HIF-α. Lastly, we provide evidence that these effects were PPARγ dependent. CONCLUSION The results demonstrate that the sEH inhibitor positively modulated the functions of EPCs in patients with AMI through the EETs-PPARγ pathway. The present study suggests the potential utility of sEHi in the therapy of ischemic heart disease.
Collapse
Affiliation(s)
- Dan-yan Xu
- Department of Cardiology, Second Xiangya Hospital, Central South University, Changsha 410011, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cantaluppi V, Biancone L, Figliolini F, Beltramo S, Medica D, Deregibus MC, Galimi F, Romagnoli R, Salizzoni M, Tetta C, Segoloni GP, Camussi G. Microvesicles derived from endothelial progenitor cells enhance neoangiogenesis of human pancreatic islets. Cell Transplant 2012; 21:1305-20. [PMID: 22455973 DOI: 10.3727/096368911x627534] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The efficacy of islet transplantation is limited by poor graft vascularization. We herein demonstrated that microvesicles (MVs) released from endothelial progenitor cells (EPCs) enhanced human islet vascularization. After incorporation into islet endothelium and β-cells, EPC-derived MVs favored insulin secretion, survival, and revascularization of islets transplanted in SCID mice. MVs induced in vitro islet endothelial cell proliferation, migration, resistance to apoptosis, and organization in vessel-like structures. Moreover, MVs partially overcame the antiangiogenic effect of rapamycin and inhibited endothelial-leukocyte interaction via L-selectin and CD40. MVs were previously shown to contain defined patterns of mRNAs. Here we demonstrated that MVs carried the proangiogenic miR-126 and miR-296 microRNAs (miRNAs). MVs pretreated with RNase or derived from Dicer knocked-down EPCs showed a reduced angiogenic effect. In addition, MVs overcame the antiangiogenic effect of the specific antagomiRs of miR-126 and miR-296, suggesting a relevant contribution of miRNAs delivered by MVs to islet endothelium. Microarray analysis of MV-stimulated islet endothelium indicated the upregulation of mRNAs coding for factors involved in endothelial proliferation, differentiation, and angiogenesis. In addition, MVs induced the activation of the PI3K-Akt and eNOS signaling pathways in islet endothelium. These results suggest that MVs activate an angiogenic program in islet endothelium that may sustain revascularization and β-cell function.
Collapse
Affiliation(s)
- Vincenzo Cantaluppi
- Research Center for Experimental Medicine (CeRMS), University of Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Vaughan EE, Liew A, Mashayekhi K, Dockery P, McDermott J, Kealy B, Flynn A, Duffy A, Coleman C, O'Regan A, Barry FP, O'Brien T. Pretreatment of endothelial progenitor cells with osteopontin enhances cell therapy for peripheral vascular disease. Cell Transplant 2012; 21:1095-107. [PMID: 22304991 DOI: 10.3727/096368911x623880] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tissue necrosis resulting from critical limb ischemia (CLI) leads to amputation in a significant number of patients. Autologous cell therapy using angiogenic cells such as endothelial progenitor cells (EPCs) holds promise as a treatment for CLI but a limitation of this treatment is that the underlying disease etiology that resulted in CLI may also contribute to dysfunction of the therapeutic EPCs. This study aimed to elucidate the mechanism of EPC dysfunction using diabetes mellitus as a model and to determine whether correction of this defect in dysfunctional EPCs ex vivo would improve the outcome after cell transplantation in the murine hind limb ischemia model. EPC dysfunction was confirmed in a homogenous population of patients with type 1 diabetes mellitus and a microarray study was preformed to identify dysregulated genes. Notably, the secreted proangiogenic protein osteopontin (OPN) was significantly downregulated in diabetic EPCs. Furthermore, OPN-deficient mice showed impaired recovery following hind limb ischemia, suggesting a critical role for OPN in postnatal neovascularization. EPCs isolated from OPN KO mice showed decreased ability to adhere to endothelial cells as well as impaired angiogenic potential. However, this dysfunction was reversed upon exposure to recombinant OPN, suggesting that OPN may act in an autocrine manner on EPCs. Indeed, exposure of OPN knockout (KO) EPCs to OPN was sufficient to induce the secretion of angiogenic proteins (IL-6, TGF-α, and FGF-α). We also demonstrated that vascular regeneration following hind limb ischemia in OPN KO mice was significantly improved upon injection of EPCs preexposed to OPN. We concluded that OPN acts in an autocrine manner on EPCs to induce the secretion of angiogenic proteins, thereby playing a critical role in EPC-mediated neovascularization. Modification of cells by exposure to OPN may improve the efficacy of autologous EPC transplantation via the enhanced secretion of angiogenic proteins.
Collapse
Affiliation(s)
- E E Vaughan
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science (NCBES), National University Ireland Galway (NUIG), Galway, Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Szöke K, Beckstrøm KJ, Brinchmann JE. Human Adipose Tissue as a Source of Cells with Angiogenic Potential. Cell Transplant 2012; 21:235-50. [DOI: 10.3727/096368911x580518] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) are involved in the process of angiogenesis, the outgrowth of new vessels from preexisting blood vessels. If available in sufficiently large numbers, ECs could be used therapeutically to establish blood flow through in vitro engineered tissues and tissues suffering from severe ischemia. Adipose tissue (AT) is an easily available source of large number of autologous ECs. Here we describe the isolation, in vitro expansion, and characterization of human AT derived ECs (AT-ECs). AT-ECs proliferated rapidly through 15–20 population doublings. The cultured cells showed cobblestone morphology and expressed EC markers including CD31, CD144, eNOS, CD309, CD105, von Willebrand factor, CD146, CD54, and CD102. They bound Ulex europaeus agglutinin I lectin and took up DiI-Ac-LDL. The AT-ECs formed capillary-like tubes in Matrigel in vitro and formed functional blood vessels in Matrigel following subcutaneous injection into immunodeficient mice. In conclusion, AT-ECs reach clinically significant cell numbers after few population doublings and are easily accessible from autologous AT, which also contains mesenchymal stem cells/pericytes. Thus, AT yields two cell populations that may be used together in the treatment of tissue ischemia and in clinical applications of tissue engineering.
Collapse
Affiliation(s)
- Krisztina Szöke
- Norwegian Center for Stem Cell Research, Institute of Immunology, Oslo University Hospital, Rikshospitalet and Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Karen Johanne Beckstrøm
- Norwegian Center for Stem Cell Research, Institute of Immunology, Oslo University Hospital, Rikshospitalet and Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jan E. Brinchmann
- Norwegian Center for Stem Cell Research, Institute of Immunology, Oslo University Hospital, Rikshospitalet and Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Bagno LLS, Werneck-de-Castro JPS, Oliveira PF, Cunha-Abreu MS, Rocha NN, Kasai-Brunswick TH, Lago VM, Goldenberg RCS, Campos-de-Carvalho AC. Adipose-derived stromal cell therapy improves cardiac function after coronary occlusion in rats. Cell Transplant 2012; 21:1985-96. [PMID: 22472303 DOI: 10.3727/096368912x636858] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recent studies have identified adipose tissue as a new source of mesenchymal stem cells for therapy. The purpose of this study was to investigate the therapy with adipose-derived stromal cells (ASCs) in a rat model of healed myocardial infarction (MI). ASCs from inguinal subcutaneous adipose tissue of male Wistar rats were isolated by enzymatic digestion and filtration. Cells were then cultured until passage 3. Four weeks after ligation of the left coronary artery of female rats, a suspension of either 100 µl with phosphate-buffered saline (PBS) + Matrigel + 2 × 10(6) ASCs labeled with Hoechst (n = 11) or 100 µl of PBS + Matrigel (n = 10) was injected along the borders of the ventricular wall scar tissue. A sham-operated group (n = 5) was submitted to the same surgical procedure except permanent ligation of left coronary artery. Cardiac performance was assessed by electro- and echocardiogram. Echo was performed prior to injections (baseline, BL) and 6 weeks after injections (follow-up, FU), and values after treatment were normalized by values obtained before treatment. Hemodynamic measurements were performed 6 weeks after injections. All infarcted animals exhibited cardiac function impairment. Ejection fraction (EF), shortening fractional area (SFA), and left ventricular akinesia (LVA) were similar between infarcted groups before treatment. Six weeks after therapy, ASC group showed significant improvement in all three ECHO indices in comparison to vehicle group. In anesthetized animals dp/dt(+) was also significantly higher in ASCs when compared to vehicle. In agreement with functional improvement, scar area was diminished in the ASC group. We conclude that ASCs improve cardiac function in infarcted rats when administered directly to the myocardium.
Collapse
Affiliation(s)
- Luiza L S Bagno
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gong W, Han Z, Zhao H, Wang Y, Wang J, Zhong J, Wang B, Wang S, Wang Y, Sun L, Han Z. Banking Human Umbilical Cord-Derived Mesenchymal Stromal Cells for Clinical Use. Cell Transplant 2012; 21:207-16. [DOI: 10.3727/096368911x586756] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A great deal of interest has arisen recently with respect to human mesenchymal stem cells (MSCs), due to their broad therapeutic potential. However, the safety and efficacy of MSCs expanded ex vivo for clinical applications remain a concern. In this article, we establish a standardized process for manufacture of human umbilical cord-derived MSCs (UC-MSCs), which encompasses donor screening and testing, recovery, two-stage expansion, and administration. The biological properties and safety of UC-MSCs were then characterized and tested. The safety data from use in human patients have also been reported. After clinical-scale expansion, a yield of 1.03–3.78 × 108 MSCs was achieved in 10 batch manufacturing runs. The biological properties, such as plastic adherence, morphology, specific surface antigen (CD105, CD73, CD90, positive ≥ 95%; CD45, CD34, CD31, CD11b, CD19, HLA-DR, negative ≤2%), and multipotent differentiation potential (osteogenesis and adipogenesis) were retained. Bacterial and mycoplasma tests were negative and endotoxin levels were lower than 2 EU/ml. No adverse events were noted in two patients treated with intravenously and/or intrathecally administered MSCs. The data obtained indicate that banking UC-MSCs for clinical use is feasible.
Collapse
Affiliation(s)
- Wei Gong
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Zhibo Han
- Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Hui Zhao
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Youwei Wang
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Jiming Wang
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Jian Zhong
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Bin Wang
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Shanshan Wang
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Yongjuan Wang
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhongchao Han
- National Engineering Research Center of Cell Products, AmCellGene Co. Ltd, TEDA, Tianjin, China
- Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| |
Collapse
|
33
|
Concise Review: Circulating Endothelial Progenitor Cells for Vascular Medicine. Stem Cells 2011; 29:1650-5. [DOI: 10.1002/stem.745] [Citation(s) in RCA: 331] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
34
|
Alev C, Ii M, Asahara T. Endothelial progenitor cells: a novel tool for the therapy of ischemic diseases. Antioxid Redox Signal 2011; 15:949-65. [PMID: 21254837 DOI: 10.1089/ars.2010.3872] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) are believed to home to sites of neovascularization, contributing to vascular regeneration either directly via incorporation into newly forming vascular structures or indirectly via the secretion of pro-angiogenic growth factors, thereby enhancing the overall vascular and hemodynamic recovery of ischemic tissues. The therapeutic application of EPCs has been shown to be effective in animal models of ischemia, and we as well as other groups involved in clinical trials have demonstrated that the use of EPCs was safe and feasible for the treatment of critical limb ischemia and cardiovascular diseases. However, many issues in the field of EPC biology, especially in regard to the proper and unambiguous molecular characterization of these cells, still remain unresolved, hampering not only basic research but also the effective therapeutic use and widespread application of these cells. Further, recent evidence suggests that several diseases and pathological conditions are correlated with a reduction in the number and biological activity of EPCs, making the development of novel strategies to overcome the current limitations and shortcomings of this promising but still limited therapeutic tool by refinement and improvement of EPC purification, expansion, and administration techniques, a rather pressing issue.
Collapse
Affiliation(s)
- Cantas Alev
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation/RIKEN Center for Developmental Biology, Kobe, Japan
| | | | | |
Collapse
|
35
|
Affiliation(s)
- Shinn-Zong Lin
- Center for Neuropsychiatry and Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
36
|
Tan Q, Tang H, Hu J, Hu Y, Zhou X, Tao Y, Wu Z. Controlled release of chitosan/heparin nanoparticle-delivered VEGF enhances regeneration of decellularized tissue-engineered scaffolds. Int J Nanomedicine 2011; 6:929-42. [PMID: 21720505 PMCID: PMC3124397 DOI: 10.2147/ijn.s18753] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Indexed: 12/13/2022] Open
Abstract
Regeneration deficiency is one of the main obstacles limiting the effectiveness of tissue-engineered scaffolds. To develop scaffolds that are capable of accelerating regeneration, we created a heparin/chitosan nanoparticle-immobilized decellularized bovine jugular vein scaffold to increase the loading capacity and allow for controlled release of vascular endothelial growth factor (VEGF). The vascularization of the scaffold was evaluated in vitro and in vivo. The functional nanoparticles were prepared by physical self-assembly with a diameter of 67–132 nm, positive charge, and a zeta potential of ∼30 mV and then the nanoparticles were successfully immobilized to the nanofibers of scaffolds by ethylcarbodiimide hydrochloride/hydroxysulfosuccinimide modification. The scaffolds immobilized with heparin/chitosan nanoparticles exhibited highly effective localization and sustained release of VEGF for several weeks in vitro. This modified scaffold significantly stimulated endothelial cells’ proliferation in vitro. Importantly, utilization of heparin/chitosan nanoparticles to localize VEGF significantly increased fibroblast infiltration, extracellular matrix production, and accelerated vascularization in mouse subcutaneous implantation model in vivo. This study provided a novel and promising system for accelerated regeneration of tissue-engineering scaffolds.
Collapse
Affiliation(s)
- Qi Tan
- Department of Cardiothoracic Surgery Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Tatsumi R, Suzuki Y, Sumi T, Sone M, Suemori H, Nakatsuji N. Simple and highly efficient method for production of endothelial cells from human embryonic stem cells. Cell Transplant 2010; 20:1423-30. [PMID: 21176397 DOI: 10.3727/096368910x547444] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Endothelial cells derived from human embryonic stem cells (hESC-ECs) hold much promise as a valuable tool for basic vascular research and for medical application such as cell transplantation or regenerative medicine. Here we have developed an efficient approach for the production of hESC-ECs. Using a differentiation method consisting of a stepwise combination of treatment with glycogen synthase kinase-3β (GSK-3β) inhibitor and culturing in vascular endothelial growth factor (VEGF)-supplemented medium, hESC-ECs are induced in 5 days with about 20% efficiency. These cells express vascular endothelial cadherin (VE-cadherin), VEGF receptor-2 (VEGFR-2), CD34, and platelet endothelial cell adhesion molecule-1 (PECAM-1). These hESC-ECs can then be isolated with 95% purity using a magnetic sorting system, and expanded to more than 100-fold within a month. The hESC-ECs thus produced exhibit the endothelial morphological characteristics and specific functions such as capillary tube formation and acetylated low-density lipoprotein uptake. We propose that our methodology is useful for efficient and large-scale production of hESC-ECs.
Collapse
Affiliation(s)
- Rie Tatsumi
- Stem Cell and Drug Discovery Institute, Kyoto Research Park, Shimogyo-ku, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Molnar M, Friberg P, Fu Y, Brisslert M, Adams M, Chen Y. Effects of Quantum Dot Labeling on Endothelial Progenitor Cell Function and Viability. CELL MEDICINE 2010; 1:105-12. [PMID: 26966634 DOI: 10.3727/215517910x451603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Endothelial progenitor cells (EPC) play an important role in repairing damaged endothelium. An effective imaging method for in vivo tracking of EPCs is essential for understanding EPC-based cell therapy. Fluorescent quantum dots (QDs) have attractive optical characteristics such as extreme brightness and photostability. QDs are currently being investigated as probes for stem cell labeling; however, there is concern about whether QDs can be used safely. We investigated whether quantum dot (QD) labeling would influence EPC viability and function. Rat bone marrow-derived EPCs were cultured and characterized. The cells were labeled with near-infrared-emitting, carboxyl-coated QDs (8 nM) for 24 h. QD labeling efficiency was higher than 97%. Using WST-1 assay, we showed that the viability of the QD-labeled EPCs was not different from that of the control EPCs. Moreover, QD labeling did not influence the ability of EPCs to form capillary tubes on Matrigel and to migrate. The percentage of QD-positive cells decreased with time, probably due to the rapid division of EPCs. These data suggest that the carboxyl-coated QD705 can be useful for labeling EPCs without interrupting their viability and functions.
Collapse
Affiliation(s)
- Matyas Molnar
- Department of Molecular and Clinical Medicine/Clinical Physiology, The Sahlgrenska Academy and University Hospital, University of Gothenburg, Gothenburg, Sweden; †Department of Theoretical Chemistry, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Peter Friberg
- Department of Molecular and Clinical Medicine/Clinical Physiology, The Sahlgrenska Academy and University Hospital, University of Gothenburg , Gothenburg , Sweden
| | - Ying Fu
- † Department of Theoretical Chemistry, School of Biotechnology, Royal Institute of Technology , Stockholm , Sweden
| | - Mikeal Brisslert
- ‡ Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy and University Hospital, University of Gothenburg , Gothenburg , Sweden
| | - Michael Adams
- § Department of Pharmacology and Toxicology, Queen's University , Kingston, Ontario , Canada
| | - Yun Chen
- Department of Molecular and Clinical Medicine/Clinical Physiology, The Sahlgrenska Academy and University Hospital, University of Gothenburg, Gothenburg, Sweden; †Department of Theoretical Chemistry, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
39
|
Eve DJ, Fillmore RW, Borlongan CV, Sanberg PR. Stem cell research in cell transplantation: sources, geopolitical influence, and transplantation. Cell Transplant 2010; 19:1493-509. [PMID: 21054954 DOI: 10.3727/096368910x540612] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
If the rapidly progressing field of stem cell research reaches its full potential, successful treatments and enhanced understanding of many diseases are the likely results. However, the full potential of stem cell science will only be reached if all possible avenues can be explored and on a worldwide scale. Until 2009, the US had a highly restrictive policy on obtaining cells from human embryos and fetal tissue, a policy that pushed research toward the use of adult-derived cells. Currently, US policy is still in flux, and retrospective analysis does show the US lagging behind the rest of the world in the proportional increase in embryonic/fetal stem cell research. The majority of US studies being on either a limited number of cell lines, or on cells derived elsewhere (or funded by other sources than Federal) rather than on freshly isolated embryonic or fetal material. Neural, mesenchymal, and the mixed stem cell mononuclear fraction are the most commonly investigated types, which can generally be classified as adult-derived stem cells, although roughly half of the neural stem cells are fetal derived. Other types, such as embryonic and fat-derived stem cells, are increasing in their prominence, suggesting that new types of stem cells are still being pursued. Sixty percent of the reported stem cell studies involved transplantation, of which over three quarters were allogeneic transplants. A high proportion of the cardiovascular systems articles were on allogeneic transplants in a number of different species, including several autologous studies. A number of pharmaceutical grade stem cell products have also recently been tested and reported on. Stem cell research shows considerable promise for the treatment of a number of disorders, some of which have entered clinical trials; over the next few years it will be interesting to see how these treatments progress in the clinic.
Collapse
Affiliation(s)
- David J Eve
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | | | |
Collapse
|
40
|
Kim JY, Song SH, Kim KL, Ko JJ, Im JE, Yie SW, Ahn YK, Kim DK, Suh W. Human cord blood-derived endothelial progenitor cells and their conditioned media exhibit therapeutic equivalence for diabetic wound healing. Cell Transplant 2010; 19:1635-44. [PMID: 20659357 DOI: 10.3727/096368910x516637] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Transplantation of human cord blood-derived endothelial progenitor cells (EPCs) is reported to contribute to neovascularization in various ischemic diseases. However, the possible beneficial role and underlying mechanisms in diabetes-impaired wound healing have been less well characterized. In this study, EPC transplantation stimulated keratinocyte and fibroblast proliferation substantially as early as 3 days after injury, leading to significantly accelerated wound closure in streptozotocin-induced diabetic nude mice, compared to PBS control. RT-PCR analysis showed that EPCs secreted various wound healing-related growth factors. Among them, keratinocyte growth factor and platelet-derived growth factor were highly expressed in the EPCs and were present at substantial levels in the EPC-injected dermal tissue. Using EPC-conditioned medium (CM), we found that paracrine factors from EPCs directly exerted mitogenic and chemotactic effects on keratinocytes and fibroblasts. Moreover, injection of EPC-CM alone into the same diabetic wound mice promoted wound healing and increased neovascularization to a similar extent as achieved with EPC transplantation. These results indicate that the beneficial effect of EPC transplantation on diabetic wounds was mainly achieved by their direct paracrine action on keratinocytes, fibroblasts, and endothelial cells, rather than through their physical engraftment into host tissues (vasculogenesis). In addition, EPC-CM was shown to be therapeutically equivalent to EPCs, at least for the treatment of diabetic dermal wounds, suggesting that conditioned medium may serve as a novel therapeutic option that is free from allograft-associated immune rejection concern.
Collapse
Affiliation(s)
- Ji Yeon Kim
- Department of Biomedical Science, College of Life Science, CHA University, Kangnam-gu, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bouchentouf M, Paradis P, Forner KA, Cuerquis J, Boivin MN, Zheng J, Boulassel MR, Routy JP, Schiffrin EL, Galipeau J. Monocyte Derivatives Promote Angiogenesis and Myocyte Survival in a Model of Myocardial Infarction. Cell Transplant 2010; 19:369-86. [DOI: 10.3727/096368909x484266] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In this study, we have investigated the hypothesis that previously reported beneficial effect of peripheral blood mononuclear cells cultured under angiogenic conditions on cardiovascular function following ischemia is not limited to EPCs but also to monocytes contained therein. We first purified and analyzed the phenotype and secretome of human and murine blood monocytes cultured under angiogenic conditions (named MDs for monocyte derivatives) and tested their effect in a mouse model of myocardial infarction (MI). FACS analysis of MDs shows that these cells express mature endothelial cell markers and that their proliferative capacity is virtually absent, consistent with their end-differentiated monocytic ontogeny. MDs secreted significant levels of HGF, IGF-1, MCP-1, and sTNFR-1 relative to their monocyte precursors. MDs were unable to form vascular networks in vitro when cultured on matrix coated flasks. Treatment of murine HL-1 cardiomyocyte cell line with MD-conditioned medium reduced their death induced by TNF-α, staurosporine, and oxidative stress, and this effect was dependent upon MD-derived sTNFR-1, HGF, and IGF-1. We further demonstrate that MD secretome promoted endothelial cell proliferation and capacity to form vessels in vitro and this was dependent upon MD-derived MCP-1, HGF, and IGF-1. Echocardiography analysis showed that MD myocardial implantation improved left ventricle fractional shortening of mouse hearts following MI and was associated with reduced myocardial fibrosis and enhancement of angiogenesis. Transplanted MDs and their secretome participate in preserving functional myocardium after ischemic insult and attenuate pathological remodeling.
Collapse
Affiliation(s)
- M. Bouchentouf
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - P. Paradis
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - K. A. Forner
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - J. Cuerquis
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - M. N. Boivin
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - J. Zheng
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - M. R. Boulassel
- Royal Victoria Hospital, Division of Hematology, McGill University Health Center, Montreal, Quebec, Canada
| | - J. P. Routy
- Royal Victoria Hospital, Division of Hematology, McGill University Health Center, Montreal, Quebec, Canada
| | - E. L. Schiffrin
- Royal Victoria Hospital, Division of Hematology, McGill University Health Center, Montreal, Quebec, Canada
| | - J. Galipeau
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Affiliation(s)
- Shinn-Zong Lin
- Scientific Committee Professor of Neurosurgery, China Medical University Superintendent, China Medical University Beigan Hospital
- China Medical University Hospital Taichung, Taiwan
| |
Collapse
|