1
|
Guo W, Pan L, Yang R, Sun J, Hu Q, Huang P. Acupoint transplantation versus non-acupoint transplantation using autologous peripheral blood mononuclear cells in treating peripheral arterial disease. BLOOD SCIENCE 2024; 6:e00175. [PMID: 38226019 PMCID: PMC10789451 DOI: 10.1097/bs9.0000000000000175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/13/2023] [Indexed: 01/17/2024] Open
Abstract
Numerous studies have discussed the therapeutic outcomes of using cell therapy or acupuncture to treat peripheral artery disease (PAD). However, there are no long-term studies on the safety and efficacy of transplanting peripheral blood mononuclear cells (PBMNCs) via acupoints to treat PAD. We first reviewed the short-term and long-term clinical results of PAD patients treated with PBMNCs through intramuscular non-acupoint transplantation (control group; n = 45) or intramuscular acupoint transplantation (acupoint group; n = 45) at a single university hospital general medical center between December 2002 and September 2022. Pain intensity (assessed with the verbal rating scale [VRS] score) in the acupoint group was considerably lower than that in the control group at month 1 (mean ± standard deviation [SD]: 1.29 ± 0.96 vs 1.76 ± 0.82; P = 0.016) and month 3 (mean ± SD: 1.27 ± 0.90 vs 1.61 ± 0.86; P = 0.042). We observed significant improvement of VRS score (P < .001 for all) and ankle-brachial index (ABI; P < .001 for all) from baseline in both groups at months 1, 3, 6, 12, 36, and 60. The 10-year cumulative rate of major amputation-free survival (MAFS) was higher in the acupoint group as compared to the control group (81.9%, 95% confidence interval [CI]: 71.3%-94.1% vs 78.5%, 95% CI: 66.7%-92.3%; P = 0.768). Compared with the routine injection method, intramuscular transplantation of PBMNCs via selected acupoints could significantly decrease the short-term pain intensity in patients with PAD, which remains an option for consideration.
Collapse
Affiliation(s)
- Wenjing Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ling Pan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ruiyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiali Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qinglin Hu
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Pingping Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
2
|
Zhong T, Gao N, Guan Y, Liu Z, Guan J. Co-Delivery of Bioengineered Exosomes and Oxygen for Treating Critical Limb Ischemia in Diabetic Mice. ACS NANO 2023; 17:25157-25174. [PMID: 38063490 PMCID: PMC10790628 DOI: 10.1021/acsnano.3c08088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Diabetic patients with critical limb ischemia face a high rate of limb amputation. Regeneration of the vasculature and skeletal muscles can salvage diseased limbs. Therapy using stem cell-derived exosomes that contain multiple proangiogenic and promyogenic factors represents a promising strategy. Yet the therapeutic efficacy is not optimal because exosomes alone cannot efficiently rescue and recruit endothelial and skeletal muscle cells and restore their functions under hyperglycemic and ischemic conditions. To address these limitations, we fabricated ischemic-limb-targeting stem cell-derived exosomes and oxygen-releasing nanoparticles and codelivered them in order to recruit endothelial and skeletal muscle cells, improve cell survival under ischemia before vasculature is established, and restore cell morphogenic function under high glucose and ischemic conditions. The exosomes and oxygen-releasing nanoparticles, delivered by intravenous injection, specifically accumulated in the ischemic limbs. Following 4 weeks of delivery, the exosomes and released oxygen synergistically stimulated angiogenesis and muscle regeneration without inducing substantial inflammation and reactive oxygen species overproduction. Our work demonstrates that codelivery of exosomes and oxygen is a promising treatment solution for saving diabetic ischemic limbs.
Collapse
Affiliation(s)
- Ting Zhong
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ning Gao
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ya Guan
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Zhongting Liu
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Jianjun Guan
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
3
|
Al-nakhle HH, Yagoub HS, Anbarkhan SH, Alamri GA, Alsubaie NM. In Silico Evaluation of Coding and Non-Coding nsSNPs in the Thrombopoietin Receptor ( MPL) Proto-Oncogene: Assessing Their Influence on Protein Stability, Structure, and Function. Curr Issues Mol Biol 2023; 45:9390-9412. [PMID: 38132435 PMCID: PMC10742084 DOI: 10.3390/cimb45120589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
The thrombopoietin receptor (MPL) gene is a critical regulator of hematopoiesis, and any alterations in its structure or function can result in a range of hematological disorders. Non-synonymous single nucleotide polymorphisms (nsSNPs) in MPL have the potential to disrupt normal protein function, prompting our investigation into the most deleterious MPL SNPs and the associated structural changes affecting protein-protein interactions. We employed a comprehensive suite of bioinformatics tools, including PredictSNP, InterPro, ConSurf, I-Mutant2.0, MUpro, Musitedeep, Project HOPE, STRING, RegulomeDB, Mutpred2, CScape, and CScape Somatic, to analyze 635 nsSNPs within the MPL gene. Among the analyzed nsSNPs, PredictSNP identified 28 as significantly pathogenic, revealing three critical functional domains within MPL. Ten of these nsSNPs exhibited high conservation scores, indicating potential effects on protein structure and function, while 14 were found to compromise MPL protein stability. Although the most harmful nsSNPs did not directly impact post-translational modification sites, 13 had the capacity to substantially alter the protein's physicochemical properties. Some mutations posed a risk to vital protein-protein interactions crucial for hematological functions, and three non-coding region nsSNPs displayed significant regulatory potential with potential implications for hematopoiesis. Furthermore, 13 out of 21 nsSNPs evaluated were classified as high-risk pathogenic variants by Mutpred2. Notably, amino acid alterations such as C291S, T293N, D295G, and W435C, while impactful on protein stability and function, were deemed non-oncogenic "passenger" mutations. Our study underscores the substantial impact of missense nsSNPs on MPL protein structure and function. Given MPL's central role in hematopoiesis, these mutations can significantly disrupt hematological processes, potentially leading to a variety of disorders. The identified high-risk pathogenic nsSNPs may hold promise as potential biomarkers or therapeutic targets for hematological diseases. This research lays the foundation for future investigations into the MPL gene's role in the realm of hematological health and diseases.
Collapse
Affiliation(s)
- Hakeemah H. Al-nakhle
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia; (H.S.Y.); (S.H.A.); (N.M.A.)
| | - Hind S. Yagoub
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia; (H.S.Y.); (S.H.A.); (N.M.A.)
- Faculty of Medical Laboratory Sciences, Omdurman Islamic University, Omdurman 14415, Sudan
| | - Sadin H. Anbarkhan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia; (H.S.Y.); (S.H.A.); (N.M.A.)
| | - Ghadah A. Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia; (H.S.Y.); (S.H.A.); (N.M.A.)
| | - Norah M. Alsubaie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia; (H.S.Y.); (S.H.A.); (N.M.A.)
| |
Collapse
|
4
|
Yazdanpanah A, Madjd Z, Pezeshki-Modaress M, Khosrowpour Z, Farshi P, Eini L, Kiani J, Seifi M, Kundu SC, Ghods R, Gholipourmalekabadi M. Bioengineering of fibroblast-conditioned Polycaprolactone/Gelatin electrospun scaffold for skin tissue engineering. Artif Organs 2022; 46:1040-1054. [PMID: 35006608 DOI: 10.1111/aor.14169] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Synthetic tissue engineering scaffolds has poor biocompatiblity with very low angiogenic properties. Conditioning the scaffolds with functional groups, coating with biological components, especially extracellular matrix (ECM), is an excellent strategy for improving their biomechanical and biological properties. METHODS In the current study, a composite of polycaprolactone and gelatin (PCL/Gel) was electrospun in the ratio of 70/30 and surface modified with 1% gelatin-coating (G-PCL/Gel) or plasma treatment (P-PCL/Gel). The surface modification was determined by SEM and ATR-FTIR spectroscopy, respectively. The scaffolds were cultured with fibroblast 3T3, then decellularized during freeze-thawing process to fabricate a fibroblast ECM-conditioned PCL/Gel scaffold (FC-PCL/Gel). The swelling and degaradtion as well as in vitro and in vivo biocompatibility and angiogenic properties of the scaffolds were evaluated. RESULTS The structure of the surface-modified G-PCL/Gel and P-PCL/Gel were unique and not changed compared to the PCL/Gel scaffolds. ATR-FTIR analysis admitted the formation of oxygen-containing groups, hydroxyl and carboxyl, on the surface of the P-PCL/Gel scaffold. The SEM micrographs and DAPI staining confirmed the cell attachment and the ECM deposition on the platform and successful removal of the cells after decellularization. P-PCL/Gel showed better cell attachment, ECM secretion and deposition after decellularization compared with G-PCL/Gel. The FC-PCL/Gel was considered as an optimized scaffold for further assays in this study. The FC-PCL/Gel showed increased hydrophilic behavior and cytobiocompatibility compared with P-PCL/Gel. The ECM on the FC-PCL/Gel scaffold showed a gradual degradation during 30 days degradation time, as a small amount of ECM remained over the FC-PCL/Gel scaffold at day 30. The FC-PCL/Gel showed significant biocompatibility and improved angiogenic property compared with P-PCL/Gel when subcutaneously implanted in a mouse animal model for 7 and 28 days. CONCLUSIONS Our findings suggest FC-PCL/Gel as an excellent biomimetic construct with high angiogenic properties. This bioengineered construct can serve as a possible application in our future pre-clinical and clinical studies for skin regeneration.
Collapse
Affiliation(s)
- Ayna Yazdanpanah
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences, Tehran, Iran
| | | | - Zahra Khosrowpour
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Paniz Farshi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran.,Department of Biomaterials, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Leila Eini
- Department of Basic Science, Faculty of Veterinary, Science and Research Branch of Islamic, Azad University, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences, Tehran, Iran
| | - Morteza Seifi
- Dept of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Subhas C Kundu
- 3B's Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimarães, Portugal
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Zhang J, Le THV, Rethineswaran VK, Kim YJ, Jang WB, Ji ST, Ly TTG, Ha JS, Yun J, Cheong JH, Jung J, Kwon SM. Dronedarone hydrochloride enhances the bioactivity of endothelial progenitor cells via regulation of the AKT signaling pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:459-466. [PMID: 34448463 PMCID: PMC8405444 DOI: 10.4196/kjpp.2021.25.5.459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 11/15/2022]
Abstract
Cardiovascular disease (CVD) and its complications are the leading cause of morbidity and mortality in the world. Because of the side effects and incomplete recovery from current therapy, stem cell therapy emerges as a potential therapy for CVD treatment, and endothelial progenitor cell (EPC) is one of the key stem cells used for therapeutic applications. The effect of this therapy required the expansion of EPC function. To enhance the EPC activation, proliferation, and angiogenesis using dronedarone hydrochloride (DH) is the purpose of this study. DH received approval for atrial fibrillation treatment and its cardiovascular protective effects were already reported. In this study, DH significantly increased EPC proliferation, tube formation, migration, and maintained EPCs surface marker expression. In addition, DH treatment up-regulated the phosphorylation of AKT and reduced the reactive oxygen species production. In summary, the cell priming by DH considerably improved the functional activity of EPCs, and the use of which might be a novel strategy for CVD treatment.
Collapse
Affiliation(s)
- Jian Zhang
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Thi Hong Van Le
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Vinoth Kumar Rethineswaran
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Yeon-Ju Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Seung Taek Ji
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Thanh Truong Giang Ly
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Jong Seong Ha
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Jae Hun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Jinsup Jung
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea.,Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
6
|
Xu H, Wang C, Liu C, Peng Z, Li J, Jin Y, Wang Y, Guo J, Zhu L. Cotransplantation of mesenchymal stem cells and endothelial progenitor cells for treating steroid-induced osteonecrosis of the femoral head. Stem Cells Transl Med 2021; 10:781-796. [PMID: 33438370 PMCID: PMC8046137 DOI: 10.1002/sctm.20-0346] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/14/2020] [Accepted: 12/06/2020] [Indexed: 11/20/2022] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (ONFH) is characterized by decreased osteogenesis, angiogenesis, and increased adipogenesis. While bone tissue engineering has been widely investigated to treat ONFH, its therapeutic effects remain unsatisfactory. Therefore, further studies are required to determine optimal osteogenesis, angiogenesis and adipogenesis in the necrotic area of the femoral head. In our study, we developed a carboxymethyl chitosan/alginate/bone marrow mesenchymal stem cell/endothelial progenitor cell (CMC/ALG/BMSC/EPC) composite implant, and evaluated its ability to repair steroid-induced ONFH. Our in vitro studies showed that BMSC and EPC coculture displayed enhanced osteogenic and angiogenic differentiation. When compared with single BMSC cultures, adipogenic differentiation in coculture systems was reduced. We also fabricated a three-dimensional (3D) CMC/ALG scaffold for loading cells, using a lyophilization approach, and confirmed its good cell compatibility characteristics, that is, high porosity, low cytotoxicity and favorable cell adhesion. 3D coculture of BMSCs and EPCs also promoted secretion of osteogenic and angiogenic factors. Then, we established an rabbit model of steroid-induced ONFH. The CMC/ALG/BMSC/EPC composite implant was transplanted into the bone tunnel of the rabbit femoral head after core decompression (CD) surgery. Twelve weeks later, radiographical and histological analyses revealed CMC/ALG/BMSC/EPC composite implants had facilitated the repair of steroid-induced ONFH, by promoting osteogenesis and angiogenesis, and reducing adipogenesis when compared with CD, CMC/ALG, CMC/ALG/BMSC and CMC/ALG/EPC groups. Thus, our data show that cotransplantation of BMSCs and EPCs in 3D scaffolds is beneficial in treating steroid-induced ONFH.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Chengqiang Wang
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Chun Liu
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Ziyue Peng
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Jianjun Li
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Yanglei Jin
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Yihan Wang
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Jiasong Guo
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
- Department of Histology and EmbryologySouthern Medical UniversityGuangzhouPeople's Republic of China
- Key Laboratory of Tissue Construction and Detection of Guangdong ProvinceGuangzhouPeople's Republic of China
- Institute of Bone BiologyAcademy of Orthopaedics, Guangdong ProvinceGuangzhouPeople's Republic of China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical CenterZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
7
|
Park IS. Enhancement of Wound Healing by Conditioned Medium of Adipose-Derived Stromal Cell with Photobiomodulation in Skin Wound. Int J Stem Cells 2021; 14:212-220. [PMID: 33632992 PMCID: PMC8138655 DOI: 10.15283/ijsc20175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/24/2020] [Accepted: 12/31/2020] [Indexed: 01/04/2023] Open
Abstract
Background and Objectives The objective of this study was to investigate whether conditioned medium from photobiomodulation (PBM) irradiated adipose-derived stromal cell (ASC) spheroids prior to implanting could stimulate angiogenesis and tissue regeneration to improve functional recovery of skin tissue in an animal skin wound model. Methods and Results ASC were split and seeded on chitosan-coated 24 well plate at a density of 7.5×104 cells/cm2, and allowed to adhere at 37℃. Within 3 days of culture, ASC formed spheroids by PBM irradiation. Conditioned medium (CM) fractions were collected from the PBM-ASC to yield nor adipose-derived stromal cell spheroid (spheroid) and PBM-spheroid, respectively, centrifuged at 13,000 g at 4℃ for 10 min, and stored prior to use for ELISA, protein assay, or in vivo wound-healing assays. Phosphate-buffered saline, cultured CM from ASCs, PBM irradiation prior to implanting conditioned medium from ASC, cultured CM from ASC spheroid, and PBM–spheroid-CM (PSC) were transplanted into a wound bed in athymic mice to evaluate therapeutic effects of PSC in vivo. PSC enhanced wound closure in a skin injury model compared to PBS, CM, PBM–CM, and spheroid-CM. The density of vascular formations increased as a result of angiogenic factors released by the wound bed and enhanced tissue regeneration at the lesion site. Conclusions These results indicate that implant of PSC can significantly improve functional recovery compared to PBS, CM, PBM–CM, or spheroid-CM treatment. Implant of PSC may be an effective form of paracrine mediated therapy for treating a wound bed.
Collapse
Affiliation(s)
- In-Su Park
- Cell Therapy Center, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
8
|
Dang Y, Gao N, Niu H, Guan Y, Fan Z, Guan J. Targeted Delivery of a Matrix Metalloproteinases-2 Specific Inhibitor Using Multifunctional Nanogels to Attenuate Ischemic Skeletal Muscle Degeneration and Promote Revascularization. ACS APPLIED MATERIALS & INTERFACES 2021; 13:5907-5918. [PMID: 33506676 PMCID: PMC8007230 DOI: 10.1021/acsami.0c19271] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Critical limb ischemia (CLI) is a severe form of peripheral artery disease (PAD). It is featured by degenerated skeletal muscle and poor vascularization. During the development of CLI, the upregulated matrix metalloproteinase-2 (MMP-2) degrades muscle extracellular matrix to initiate the degeneration. Meanwhile, MMP-2 is necessary for blood vessel formation. It is thus hypothesized that appropriate MMP-2 bioactivity in ischemic limbs will not only attenuate muscle degeneration but also promote blood vessel formation. Herein, we developed ischemia-targeting poly(N-isopropylacrylamide)-based nanogels to specifically deliver an MMP-2 inhibitor CTTHWGFTLC (CTT) into ischemic limbs to tailor MMP-2 bioactivity. Besides acting as an MMP-2 inhibitor, CTT promoted endothelial cell migration under conditions mimicking the ischemic limbs. The nanogels were sensitive to the pH of ischemic tissues, allowing them to largely aggregate in the injured area. To help reduce nanogel uptake by macrophages and increase circulation time, the nanogels were cloaked with a platelet membrane. An ischemia-targeting peptide CSTSMLKA (CST) was further conjugated on the platelet membrane for targeted delivery of nanogels into the ischemic area. CTT gradually released from the nanogels for 4 weeks. The nanogels mostly accumulated in the ischemic area for 28 days. The released CTT preserved collagen in the muscle and promoted its regeneration. In addition, CTT stimulated angiogenesis. Four weeks after CLI, the blood flow and vessel density of the ischemic limbs treated with the nanogels were remarkably higher than the control groups without CTT release. These results demonstrate that the developed nanogel-based CTT release system has the potential to stimulate ischemic limb regeneration.
Collapse
Affiliation(s)
- Yu Dang
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ning Gao
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Hong Niu
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ya Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jianjun Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
9
|
Fang G, Jiang X, Fang Y, Pan T, Liu H, Ren B, Wei Z, Gu S, Chen B, Jiang J, Shi Y, Guo D, Liu P, Fu W, Dong Z. Autologous peripheral blood-derived stem cells transplantation for treatment of no-option angiitis-induced critical limb ischemia: 10-year management experience. Stem Cell Res Ther 2020; 11:458. [PMID: 33115517 PMCID: PMC7594448 DOI: 10.1186/s13287-020-01981-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Background Previous studies have demonstrated that no-option angiitis-induced critical limb ischemia (NO-AICLI) could be significantly improved by transplantation of peripheral blood-derived stem cells (PBDSCs). Additionally, a randomized controlled trial (RCT) recently conducted by us suggested that peripheral blood-derived purified CD34+ cells (PCCs) were not inferior to non-purified peripheral blood mononuclear cells (PBMNCs) at limb salvage in treatment of NO-AICLI. However, most of these clinical trials whether RCT or single-arm studies were characterized with a small sample size and absence of long-term outcomes. Methods To analyze long-term clinical outcomes of PBDSCs transplantation for NO-AICLI, we reviewed clinical data of patients with NO-AICLI receiving PBDSCs transplantation at our center during the past decade. Meanwhile, we first compared the long-term safety and efficacy of intramuscular transplantation of PCCs versus PBMNCs in a sizable number of patients with NO-AICLI. Results From May 2009 to December 2019, a total of 160 patients with NO-AICLI patients were treated by PBDSCs transplantation (82 with PCCs, 78 with PBMNCs) at our center. Baseline characteristics between two groups were similar. Up to June 2020, the mean follow-up period was 46.6 ± 35.3 months. No critical adverse events were observed in either group. There was one death during the follow-up period. A total of eight major amputations occurred. The cumulative major amputation-free survival (MAFS) rate at 5 years after PBDSCs transplantation was 94.4%, without difference between two groups (P = .855). Wound healing, rest pain, pain-free walking time, ankle-brachial index, transcutaneous oxygen pressure, and quality of life (QoL) also significantly improved after PBDSCs transplantation. Conclusions Autologous PBDSCs intramuscular transplantation could significantly decrease the major amputation rates and improve the QoL in patients with NO-AICLI. Long-term observation of a large sample of patients confirmed that the clinical benefits of PBDSCs transplantation were durable, without difference between the PCCs and PBMNCs groups.
Collapse
Affiliation(s)
- Gang Fang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolang Jiang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Fang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianyue Pan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Liu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bichen Ren
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wei
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyang Gu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Chen
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junhao Jiang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shi
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China. .,Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Zhihui Dong
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China. .,Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
10
|
Atherosclerotic Pre-Conditioning Affects the Paracrine Role of Circulating Angiogenic Cells Ex-Vivo. Int J Mol Sci 2020; 21:ijms21155256. [PMID: 32722151 PMCID: PMC7432497 DOI: 10.3390/ijms21155256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
In atherosclerosis, circulating angiogenic cells (CAC), also known as early endothelial progenitor cells (eEPC), are thought to participate mainly in a paracrine fashion by promoting the recruitment of other cell populations such as late EPC, or endothelial colony-forming cells (ECFC), to the injured areas. There, ECFC replace the damaged endothelium, promoting neovascularization. However, despite their regenerative role, the number and function of EPC are severely affected under pathological conditions, being essential to further understand how these cells react to such environments in order to implement their use in regenerative cell therapies. Herein, we evaluated the effect of direct incubation ex vivo of healthy CAC with the secretome of atherosclerotic arteries. By using a quantitative proteomics approach, 194 altered proteins were identified in the secretome of pre-conditioned CAC, many of them related to inhibition of angiogenesis (e.g., endostatin, thrombospondin-1, fibulins) and cell migration. Functional assays corroborated that healthy CAC released factors enhanced ECFC angiogenesis, but, after atherosclerotic pre-conditioning, the secretome of pre-stimulated CAC negatively affected ECFC migration, as well as their ability to form tubules on a basement membrane matrix assay. Overall, we have shown here, for the first time, the effect of atherosclerotic factors over the paracrine role of CAC ex vivo. The increased release of angiogenic inhibitors by CAC in response to atherosclerotic factors induced an angiogenic switch, by blocking ECFC ability to form tubules in response to pre-conditioned CAC. Thus, we confirmed here that the angiogenic role of CAC is highly affected by the atherosclerotic environment.
Collapse
|
11
|
Lee NG, Jeung IC, Heo SC, Song J, Kim W, Hwang B, Kwon MG, Kim YG, Lee J, Park JG, Shin MG, Cho YL, Son MY, Bae KH, Lee SH, Kim JH, Min JK. Ischemia-induced Netrin-4 promotes neovascularization through endothelial progenitor cell activation via Unc-5 Netrin receptor B. FASEB J 2019; 34:1231-1246. [PMID: 31914695 DOI: 10.1096/fj.201900866rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 11/11/2022]
Abstract
Endothelial progenitor cells (EPCs) promote neovascularization and tissue repair by migrating to vascular injury sites; therefore, factors that enhance EPC homing to damaged tissues are of interest. Here, we provide evidence of the prominent role of the Netrin-4 (NTN4)-Unc-5 Netrin receptor B (UNC5B) axis in EPC-specific promotion of ischemic neovascularization. Our results showed that NTN4 promoted the proliferation, chemotactic migration, and paracrine effects of small EPCs (SEPCs) and significantly increased the incorporation of large EPCs (LEPCs) into tubule networks. Additionally, NTN4 prominently augmented neovascularization in mice with hindlimb ischemia by increasing the homing of exogenously transplanted EPCs to the ischemic limb and incorporating EPCs into vessels. Moreover, silencing of UNC5B, an NTN4 receptor, abrogated the NTN4-induced cellular activities of SEPCs in vitro and blood-flow recovery and neovascularization in vivo in ischemic muscle by reducing EPC homing and incorporation. These findings suggest NTN4 as an EPC-based therapy for treating angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Na Geum Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - In Cheul Jeung
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Soon Chul Heo
- Department of Physiology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jinhoi Song
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Wooil Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Byungtae Hwang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Min-Gi Kwon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Min-Gyeong Shin
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young-Lai Cho
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Mi-Young Son
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Kwang-Hee Bae
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
12
|
Imaging VEGF Receptors and α vβ 3 Integrins in a Mouse Hindlimb Ischemia Model of Peripheral Arterial Disease. Mol Imaging Biol 2019; 20:963-972. [PMID: 29687324 DOI: 10.1007/s11307-018-1191-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To compare targeted imaging of vascular endothelial growth factor (VEGF) receptors vs. αvβ3 integrins in a mouse hindlimb ischemia model of peripheral artery disease. PROCEDURES Male wild-type (WT) C57BL/6 mice (8- to 10-week old) (n = 24) underwent left femoral artery ligation. The right leg served as control. Five days later, mice were injected with either VEGF receptor targeting [99mTc]DOTA-PEG-scVEGF ([99mTc]scV) (n = 8) or with αvβ3-targeting tracer [99mTc]HYNIC-cycloRGD ([99mTc]RGD) (n = 8) and underwent single photon emission computed tomography (SPECT) x-ray computed tomography imaging. To assess non-specific [99mTc]scV uptake, six additional mice received a mixture of [99mTc]scV and 30-fold excess of targeting protein, scVEGF. Tracer uptake as %ID was measured using volumetric regions encompassing the hindlimb muscles and as %ID/g from harvested limb muscles. Double and triple immunofluorescent analysis on tissue sections established localization of αvβ3, VEGFR-1, VEGFR-2, as well as certain cell lineage markers. RESULTS Tracer uptake, as %ID/g, was higher in ligated limbs of mice injected with [99mTc]scV compared to ligated hindlimbs in mice injected with [99mTc]RGD (p = 0.02). The ratio of tracer uptake for ligated/control hindlimb was borderline higher for [99mTc]scV than for [99mTc]RGD (p = 0.06). Immunofluorescent analysis showed higher prevalence of VEGFR-1, VEGFR-2, and αvβ3, in damaged vs. undamaged hindlimb tissue, but with little co-localization of these markers. Double immunofluorescent staining showed partial co-localization of VEGFR-1, VEGFR-2, and αvβ3, with endothelial cell marker FVIII, but not with CD31. Immunostaining for VEGFR-1 and VEGFR-2 additionally co-localized with lineage markers for endothelial progenitor cell and monocytes/macrophages, with a more diverse pattern of co-localization for VEGFR-2. CONCLUSION In a mouse hindlimb ischemia model of peripheral artery disease, [99mTc]scV SPECT tracer-targeting VEGF receptors showed a more robust signal than [99mTc]RGD tracer-targeting αvβ3. Immunofluorescent analysis suggests that uptake of [99mTc]scV and [99mTc]RGD in damaged tissue is due to non-overlapping cell populations and reflects different dynamic processes and that enhanced uptake of [99mTc]scV may be due to the presence of VEGF receptors on additional cell types.
Collapse
|
13
|
Shen WC, Chou YH, Huang HP, Sheen JF, Hung SC, Chen HF. Induced pluripotent stem cell-derived endothelial progenitor cells attenuate ischemic acute kidney injury and cardiac dysfunction. Stem Cell Res Ther 2018; 9:344. [PMID: 30526689 PMCID: PMC6288873 DOI: 10.1186/s13287-018-1092-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/31/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022] Open
Abstract
Background Renal ischemia–reperfusion (I/R) injury is a major cause of acute kidney injury (AKI), which is associated with high morbidity and mortality. AKI is a serious and costly medical condition. Effective therapy for AKI is an unmet clinical need, and molecular mechanisms underlying the interactions between an injured kidney and distant organs remain unclear. Therefore, novel therapeutic strategies should be developed. Methods We directed the differentiation of human induced pluripotent stem (iPS) cells into endothelial progenitor cells (iEPCs), which were then applied for treating mouse AKI. The mouse model of AKI was induced by I/R injury. Results We discovered that intravenously infused iEPCs were recruited to the injured kidney, expressed the mature endothelial cell marker CD31, and replaced injured endothelial cells. Moreover, infused iEPCs produced abundant proangiogenic proteins, which entered into circulation. In AKI mice, blood urea nitrogen and plasma creatinine levels increased 2 days after I/R injury and reduced after the infusion of iEPCs. Tubular injury, cell apoptosis, and peritubular capillary rarefaction in injured kidneys were attenuated accordingly. In the AKI mice, iEPC therapy also ameliorated apoptosis of cardiomyocytes and cardiac dysfunction, as indicated by echocardiography. The therapy also ameliorated an increase in serum brain natriuretic peptide. Regarding the relevant mechanisms, indoxyl sulfate and interleukin-1β synergistically induced apoptosis of cardiomyocytes. Systemic iEPC therapy downregulated the proapoptotic protein caspase-3 and upregulated the anti-apoptotic protein Bcl-2 in the hearts of the AKI mice, possibly through the reduction of indoxyl sulfate and interleukin-1β. Conclusions Therapy using human iPS cell-derived iEPCs provided a protective effect against ischemic AKI and remote cardiac dysfunction through the repair of endothelial cells and the attenuation of cardiomyocyte apoptosis. Electronic supplementary material The online version of this article (10.1186/s13287-018-1092-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Ching Shen
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Hsiang Chou
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jenn-Feng Sheen
- Department of Biotechnology, National Formosa University, Yun-Lin, Taiwan
| | - Shih-Chieh Hung
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.,Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung, 404, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 105, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan. .,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
14
|
Li WD, Zhou DM, Sun LL, Xiao L, Liu Z, Zhou M, Wang WB, Li XQ. LncRNA WTAPP1 Promotes Migration and Angiogenesis of Endothelial Progenitor Cells via MMP1 Through MicroRNA 3120 and Akt/PI3K/Autophagy Pathways. Stem Cells 2018; 36:1863-1874. [PMID: 30171660 DOI: 10.1002/stem.2904] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/22/2018] [Accepted: 07/27/2018] [Indexed: 01/17/2023]
Affiliation(s)
- Wen-Dong Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Dong-Ming Zhou
- Department of Hematology, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Li-Li Sun
- Department of Vascular Surgery; The Second Affiliated Hospital of Soochow University; Suzhou JiangSu People's Republic of China
| | - Lun Xiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Zhao Liu
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Min Zhou
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Wen-Bin Wang
- Department of General Surgery; The Fourth Affiliated Hospital of Anhui Medical University; HeFei People's Republic of China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| |
Collapse
|
15
|
Yuan KM, Zhang PH, Qi SS, Zhu QZ, Li P. Emerging Role for Exosomes in the Progress of Stem Cell Research. Am J Med Sci 2018; 356:481-486. [DOI: 10.1016/j.amjms.2018.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 06/27/2018] [Accepted: 07/13/2018] [Indexed: 01/08/2023]
|
16
|
Peng X, Li C, Bai Y, Wang X, Zhang Y, An Y, Teng GJ, Ju S. Noninvasive evaluation of the migration effect of transplanted endothelial progenitor cells in ischemic muscle using a multimodal imaging agent. Int J Nanomedicine 2018; 13:1819-1829. [PMID: 29606873 PMCID: PMC5868615 DOI: 10.2147/ijn.s152976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Endothelial progenitor cells (EPCs) play an important role in repairing ischemia tissues. However, the survival, migration and therapeutic efficacy of EPCs after transplantation need to be better understood for further cell therapy. Purpose This study investigated the migration effect of EPCs labeled with a multimodal imaging agent in a murine ischemic hindlimb model, using magnetic resonance imaging (MRI) and optical imaging after transplantation. Methods EPCs derived from mouse bone marrow were labeled with a multimodal imaging agent and were administered through intracardiac delivery to mice with ischemic hindlimbs. The injected EPCs and their migration effect were observed via MRI and optical imaging in vivo, and then compared to a reference standard based on histological data. The quantification of gadolinium in tissue samples was done using inductively coupled plasma mass spectrometry (ICP-MS). Results Using in vivo MRI and optical imaging, the labeled EPCs were observed to migrate to ischemic muscle on days 3-5 after injection, while ex vivo, the EPCs were observed in the capillary vessels of the injured tissue. There were significant linear correlations between the Gd contents measured using ICP-MS in samples from the ischemic hindlimbs and livers and T1 relaxation times calculated using MRI, as well as the average fluorescence signal intensities recorded in optical images (T1 relaxation time: r=0.491; average signal from optical imaging: r=0.704, P<0.01). EPC treatment upregulated the levels of C-X-C chemokine receptor 4 and vascular endothelial growth factor (VEGF) receptor 2 and enhanced the expression of stromal cell-derived factor-1 and VEGF. Conclusion Transplanted EPCs can be monitored with noninvasive MRI and optical imaging in vivo and were found to enhance the paracrine secretion of angiogenic factors.
Collapse
Affiliation(s)
- Xingui Peng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yingying Bai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Xinyi Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Yanli An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| |
Collapse
|
17
|
Kim JH, Kim KA, Shin YJ, Kim H, Majid A, Bae ON. Methylglyoxal induced advanced glycation end products (AGE)/receptor for AGE (RAGE)-mediated angiogenic impairment in bone marrow-derived endothelial progenitor cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2018; 81:266-277. [PMID: 29473788 DOI: 10.1080/15287394.2018.1440185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Endothelial cells (ECs) maintain the structure and function of blood vessels and are readily exposed to exogenous and endogenous toxic substances in the circulatory system. Bone marrow-derived endothelial progenitor cells (EPCs) circulate in the blood and differentiate to EC, which are known to participate in angiogenesis and regeneration of injured vessels. Dysfunction in EPC contributes to cardiovascular complications in patients with diabetes, but the precise molecular mechanisms underlying diabetic EPC abnormalities are not completely understood. The aim of this study was to investigate the mechanisms underlying diabetic EPC dysfunction using methylglyoxal (MG), an endogenous toxic diabetic metabolite. Data demonstrated that MG decreased cell viability and protein expression of vascular endothelial growth factor receptor (VEGFR)-2 associated with functional impairment of tube formation in EPC. The generation of advanced glycation end (AGE) products was increased in EPC following exposure to MG. Blockage of receptor for AGE (RAGE) by FPS-ZM1, a specific antagonist for RAGE, significantly reversed the decrease of VEGFR-2 protein expression and angiogenic dysfunction in MG-incubated EPC. Taken together, data demonstrated that MG induced angiogenic impairment in EPC via alterations in the AGE/RAGE-VEGFR-2 pathway which may be utilized in the development of potential therapeutic and preventive targets for diabetic vascular complications.
Collapse
Affiliation(s)
- Jeong-Hyeon Kim
- a College of Pharmacy Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Republic of Korea
| | - Kyeong-A Kim
- a College of Pharmacy Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Republic of Korea
| | - Young-Jun Shin
- a College of Pharmacy Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Republic of Korea
| | - Haram Kim
- a College of Pharmacy Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Republic of Korea
| | - Arshad Majid
- b Sheffield Institute for Translational Neuroscience , University of Sheffield , Sheffield , England
| | - Ok-Nam Bae
- a College of Pharmacy Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Republic of Korea
| |
Collapse
|
18
|
Jeong GJ, Oh JY, Kim YJ, Bhang SH, Jang HK, Han J, Yoon JK, Kwon SM, Lee TI, Kim BS. Therapeutic Angiogenesis via Solar Cell-Facilitated Electrical Stimulation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38344-38355. [PMID: 29043772 DOI: 10.1021/acsami.7b13322] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cell therapy has been suggested as a treatment modality for ischemic diseases, but the poor survival and engraftment of implanted cells limit its therapeutic efficacy. To overcome such limitation, we used electrical stimulation (ES) derived from a wearable solar cell for inducing angiogenesis in ischemic tissue. ES enhanced the secretion of angiogenic growth factors and the migration of mesenchymal stem cells (MSCs), myoblasts, endothelial progenitor cells, and endothelial cells in vitro. In a mouse ischemic hindlimb model, ES generated by a solar cell and applied to the ischemic region promoted migration of MSCs toward the ischemic site and upregulated expression of angiogenic paracrine factors (vascular endothelial, basic fibroblast, and hepatocyte growth factors; and stromal cell-derived factor-1α). Importantly, solar cell-generated ES promoted the formation of capillaries and arterioles at the ischemic region, attenuated muscle necrosis and fibrosis, and eventually prevented loss of the ischemic limb. Solar cell ES therapy showed higher angiogenic efficacy than conventional MSC therapy. This study shows the feasibility of using solar cell ES as a novel treatment for therapeutic angiogenesis.
Collapse
Affiliation(s)
| | - Jin Young Oh
- Department of Materials Science and Engineering, Yonsei University , Seoul 03722, Republic of Korea
| | - Yeon-Ju Kim
- Department of Physiology, School of Medicine, Pusan National University , Yangsan, 50612 Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University , Suwon 16419, Republic of Korea
| | | | | | | | - Sang-Mo Kwon
- Department of Physiology, School of Medicine, Pusan National University , Yangsan, 50612 Republic of Korea
| | - Tae Il Lee
- Department of BioNano Technology, Gachon University , Seongnam 13120, Republic of Korea
| | | |
Collapse
|
19
|
Adipose Extracellular Matrix/Stromal Vascular Fraction Gel Secretes Angiogenic Factors and Enhances Skin Wound Healing in a Murine Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3105780. [PMID: 28835892 PMCID: PMC5556995 DOI: 10.1155/2017/3105780] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/13/2017] [Accepted: 06/15/2017] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells are an attractive cell type for cytotherapy in wound healing. The authors recently developed a novel, adipose-tissue-derived, injectable extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel) for stem cell therapy. This study was designed to assess the therapeutic effects of ECM/SVF-gel on wound healing and potential mechanisms. ECM/SVF-gel was prepared for use in nude mouse excisional wound healing model. An SVF cell suspension and phosphate-buffered saline injection served as the control. The expression levels of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and monocyte chemotactic protein-1 (MCP-1) in ECM/SVF-gel were analyzed at different time points. Angiogenesis (tube formation) assays of ECM/SVF-gel extracts were evaluated, and vessels density in skin was determined. The ECM/SVF-gel extract promoted tube formation in vitro and increased the expression of the angiogenic factors VEGF and bFGF compared with those in the control. The expression of the inflammatory chemoattractant MCP-1 was high in ECM/SVF-gel at the early stage and decreased sharply during the late stage of wound healing. The potent angiogenic effects exerted by ECM/SVF-gel may contribute to the improvement of wound healing, and these effects could be related to the enhanced inflammatory response in ECM/SVF-gel during the early stage of wound healing.
Collapse
|
20
|
Microlens topography combined with vascular endothelial growth factor induces endothelial differentiation of human mesenchymal stem cells into vasculogenic progenitors. Biomaterials 2017; 131:68-85. [DOI: 10.1016/j.biomaterials.2017.03.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/09/2017] [Accepted: 03/23/2017] [Indexed: 02/06/2023]
|
21
|
Berezin AE, Samura TA, Kremzer AA, Berezina TA, Martovitskaya YV, Gromenko EA. An association of serum vistafin level and number of circulating endothelial progenitor cells in type 2 diabetes mellitus patients. Diabetes Metab Syndr 2016; 10:205-212. [PMID: 27377688 DOI: 10.1016/j.dsx.2016.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/06/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND The decreased number and impaired functions of endothelial progenitor cells (EPCs) may associate with cardiovascular disease (CV) including atherosclerosis. However, the role of vistafin in regulation of angiogenic EPC subset maturation in T2DM patients without known atherosclerosis is still not fully understood. THE AIM OF THE STUDY To investigate an association of serum vistafin level and number of circulating EPCs in T2DM patients beyond known CV disease. METHODS This case-control observational investigation was evolved 54 subjects with T2DM and 35 healthy volunteers. The flow cytometry was used for predictably distinguishing cell subsets, which depend on expression of CD45, CD34, CD14, Tie-2, and VEGFR2. Biomarkers were measured at baseline of the study. RESULTS All T2DM patients were divided depending median of vistafin level (5.88ng/mL) in to two cohorts with low vistafin level (<5.88ng/mL; n=29) and high vistafin level (≥5.88ng/mL; n=25) respectively. Logistic regression analysis has shown that visfatin, hs-CRP, age and BMI were the best variables in the prediction of EPC number labeled as CD14+CD309+ and CD14+CD309+Tie2+ cells. After adjustment of the model to age and BMI elevated visfatin level remained the best predictor for both CD14+CD309+ and CD14+CD309+Tie2+ EPCs (OR 0.92, 95% CI: 0.88-0.95; P=0.001 and OR 0.90, 95% CI: 0.87-0.96; P=0.001 respectively). CONCLUSION We found that elevated level of vistafin was an independent predictor for declined numerous of non-classical EPCs labeled as CD14+CD309+ and CD14+CD309+Tie2+, whereas CD34+ subsets of EPCs did not associate with vistafin level in T2DM individuals.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, State Medical University, 26, Mayakovsky Av., Zaporozhye, Postcode 69035, Ukraine.
| | - Tatyana A Samura
- Clinical Pharmacology Department, State Medical University, Zaporozhye, Ukraine
| | - Alexander A Kremzer
- Clinical Pharmacology Department, State Medical University, Zaporozhye, Ukraine
| | | | | | | |
Collapse
|
22
|
Transplanted Endothelial Progenitor Cells Improve Ischemia Muscle Regeneration in Mice by Diffusion Tensor MR Imaging. Stem Cells Int 2016; 2016:3641401. [PMID: 27656214 PMCID: PMC5021888 DOI: 10.1155/2016/3641401] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 12/24/2022] Open
Abstract
Endothelial progenitor cells (EPCs) play an important role in repairing ischemia tissues. Diffusion tensor imaging (DTI) was applied to detect the architectural organization of skeletal muscle. This study investigated the feasibility and accuracy of using the DTI to evaluate effectiveness of EPCs treatment. Mouse bone marrow-derived EPCs were isolated, cultured, characterized, and transplanted to hindlimb ischemia mice model. DTI was performed on the hindlimb at postischemia time points. The edema regions of diffusion restriction (high signal in diffusion weighted imaging) were decreased in the ischemic muscle of EPCs treated mice after 14 days compared with the controls. These results from DTI show the lower apparent diffusion coefficient and eigenvalues (λ1, λ2, and λ3) and the higher fractional anisotropy and fiber counts of ischemic muscle on 7 and 14 days after EPCs treatment compared to the controls. There was a significant correlation between fiber counts calculated by DTI and survival fibers evaluated by histological section (r = 0.873, P < 0.01). Our study demonstrated that the time frame for muscle fiber regeneration after EPCs transplantation was significantly shortened in vivo. DTI could be a useful tool for noninvasive evaluation of muscle tissue damage and repair in animal models and patient with ischemic diseases.
Collapse
|
23
|
Stress-Induced Premature Senescence of Endothelial and Endothelial Progenitor Cells. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 77:281-306. [PMID: 27451101 DOI: 10.1016/bs.apha.2016.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This brief overview of premature senescence of dysfunctional endothelial and endothelial progenitor cells provides information on endothelial cell differentiation and specialization, their ontogeny, and controversies related to endothelial stem and progenitor cells. Stressors responsible for the dysfunction of endothelial and endothelial progenitor cells, as well as cellular mechanisms and consequences of endothelial cell dysfunction are presented. Metabolic signatures of dysfunctional endothelial cells and senescence pathways are described. Emerging strategies to rejuvenate endothelial and endothelial progenitor cells conclude the review.
Collapse
|
24
|
Choi YH, Jang IH, Heo SC, Kim JH, Hwang NS. Biomedical therapy using synthetic WKYMVm hexapeptide. Organogenesis 2016; 12:53-60. [PMID: 27077939 DOI: 10.1080/15476278.2016.1172155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
WKYMVm hexapeptide has been identified as a strong FPR2 agonist through a library screening of synthetic peptides. The FPR2 has been reported to play a crucial role in inflammation and angiogenic responses via stimulation of chemotaxis, migration, cell proliferation, wound healing and vessel growth. Recently, the therapeutic effects of WKYMVm have been reported in various disease models. In cutaneous wound model in diabetic mice, WKYMVm facilitated wound healing processes by stimulating the formation of capillary and arteriole and re-epithelialization. In coronary artery stenosis model, WKYMVm coating on stent promoted re-endothelialization and lowered restenosis rate. In hindlimb ischemia mouse model, intramuscular injection of WKYMVm promoted homing of exogenously transplanted endothelial colony-forming cells and neovascularization, resulting in salvaging hindlimb. Furthermore, a single injection of WKYMVm encapsulated in poly (lactide-co-glycolide) microspheres was demonstrated to be as efficient as multiple injections of WKYMVm in restoring blood flow in hindlimb ischemia model. These observations may open up promising biomedical applications of WKYMVm for tissue repairs and regenerations.
Collapse
Affiliation(s)
- Young Hwan Choi
- a School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University , Seoul , Republic of Korea
| | - Il Ho Jang
- b Department of Physiology , School of Medicine, Pusan National University Yangsan Hospital , Yangsan , Republic of Korea
| | - Soon Chul Heo
- b Department of Physiology , School of Medicine, Pusan National University Yangsan Hospital , Yangsan , Republic of Korea
| | - Jae Ho Kim
- b Department of Physiology , School of Medicine, Pusan National University Yangsan Hospital , Yangsan , Republic of Korea.,c Research Institute of Convergence Biomedical Science and Technology , Yangsan , Gyeongsangnam-do , Republic of Korea
| | - Nathaniel S Hwang
- a School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University , Seoul , Republic of Korea.,d N-Bio Institute , Seoul National University , Seoul , Republic of Korea
| |
Collapse
|
25
|
Xu Y, Fu M, Li Z, Fan Z, Li X, Liu Y, Anderson PM, Xie X, Liu Z, Guan J. A prosurvival and proangiogenic stem cell delivery system to promote ischemic limb regeneration. Acta Biomater 2016; 31:99-113. [PMID: 26689466 DOI: 10.1016/j.actbio.2015.12.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/17/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
Stem cell therapy is one of the most promising strategies to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the low oxygen and nutrient environment of the injured limbs. To increase therapeutic efficacy, high rates of both short- and long-term cell survival are essential, which current approaches do not support. In this work, we hypothesized that a high rate of short-term cell survival can be achieved by introducing a prosurvival environment into the stem cell delivery system to enhance cell survival before vascularization is established; and that a high rate of long-term cell survival can be attained by building a proangiogenic environment in the system to quickly vascularize the limbs. The system was based on a biodegradable and thermosensitive poly(N-Isopropylacrylamide)-based hydrogel, a prosurvival and proangiogenic growth factor bFGF, and bone marrow-derived mesenchymal stem cells (MSCs). bFGF can be continuously released from the system for 4weeks. The released bFGF significantly improved MSC survival and paracrine effects under low nutrient and oxygen conditions (0% FBS and 1% O2) in vitro. The prosurvival effect of the bFGF on MSCs was resulted from activating cell Kruppel-like factor 4 (KLF4) pathway. When transplanted into the ischemic limbs, the system dramatically improved MSC survival. Some of the engrafted cells were differentiated into skeletal muscle and endothelial cells, respectively. The system also promoted the proliferation of host cells. After only 2weeks of implantation, tissue blood perfusion was completely recovered; and after 4weeks, the muscle fiber diameter was restored similarly to that of the normal limbs. These pronounced results demonstrate that the developed stem cell delivery system has a potential for ischemic limb regeneration. STATEMENT OF SIGNIFICANCE Stem cell therapy is a promising strategy to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the ischemic environment of the injured limbs. To increase therapeutic efficacy, high rate of cell survival is essential, which current approaches do not support. In this work, we tested the hypothesis that a stem cell delivery system that can continuously release a prosurvival and proangiogenic growth factor will promote high rates of cell survival in the ischemic limbs. The prosurvival effect could augment cell survival before vascularization is established, while the proangiogenic effect could stimulate quick angiogenesis to achieve long-term cell survival. Meanwhile, the differentiation of stem cells into endothelial and myogenic lineages, and cell paracrine effects will enhance vascularization and muscle regeneration.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Minghuan Fu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Department of Gerontology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Zhihong Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Division of General Surgery, Shanghai Pudong New District Zhoupu Hospital, Shanghai 201200, China
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaofei Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Ying Liu
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Peter M Anderson
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaoyun Xie
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Zhenguo Liu
- Davis Heart and Lung Research Institute, The Ohio State University, OH 43210, United States
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Tongji Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
26
|
Tu TC, Nagano M, Yamashita T, Hamada H, Ohneda K, Kimura K, Ohneda O. A Chemokine Receptor, CXCR4, Which Is Regulated by Hypoxia-Inducible Factor 2α, Is Crucial for Functional Endothelial Progenitor Cells Migration to Ischemic Tissue and Wound Repair. Stem Cells Dev 2015; 25:266-76. [PMID: 26620723 PMCID: PMC4742989 DOI: 10.1089/scd.2015.0290] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Endothelial progenitor cells (EPCs) have the ability to form new blood vessels and protect ischemic tissues from damage. We previously reported that EPCs with low activity of aldehyde dehydrogenase (Alde-Low EPCs) possess the greater ability to treat ischemic tissues compared with Alde-High EPCs. The expression level of the hypoxia-inducible factors (HIFs), HIF-1α and HIF-2α, was found to be greater in Alde-Low EPCs than in Alde-High EPCs. However, the precise role of the HIF factors in the regulation of EPC activity remains obscure. In this study, we demonstrate a critical role of HIF-2α and its target gene CXCR4 for controlling the migratory activity of EPC to ischemic tissue. We found that coculture of Alde-High EPCs with microvesicles derived from Alde-Low EPCs improved their ability to repair an ischemic skin flap, and the expression of CXCR4 and its ligand SDF1 was significantly increased following the coculture. In Alde-Low EPCs, the expression of CXCR4 was suppressed by short hairpin RNA (shRNA)-mediated HIF-2α, but not HIF-1α downregulation. Chromatin immunoprecipitation assays showed that HIF-2α, but not HIF-1α, binds to the promoter region of CXCR4 gene. The CXCR4 shRNA treatment in Alde-Low EPCs almost completely abrogated their migratory activity to ischemic tissues, whereas the reduction of vascular endothelial growth factor (VEGF) showed much less effect. The CXCR4 overexpression in Alde-High EPCs resulted in a partial, but significant improvement in their repairing ability in an ischemic skin flap. Collectively, these findings indicate that the CXCR4/SDF-1 axis, which is specifically regulated by HIF-2α, plays a crucial role in the regulation of EPC migration to ischemic tissues.
Collapse
Affiliation(s)
- Tran Cam Tu
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Masumi Nagano
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Toshiharu Yamashita
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Hiromi Hamada
- 2 Department of Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Kinuko Ohneda
- 3 Laboratory of Molecular Pathophysiology, Faculty of Pharmacy, Takasaki University of Health and Welfare , Takasaki, Japan
| | - Kenichi Kimura
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Osamu Ohneda
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| |
Collapse
|
27
|
Diabetes-Induced Oxidative Stress in Endothelial Progenitor Cells May Be Sustained by a Positive Feedback Loop Involving High Mobility Group Box-1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:1943918. [PMID: 26798412 PMCID: PMC4698939 DOI: 10.1155/2016/1943918] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/21/2022]
Abstract
Oxidative stress is considered to be a critical factor in diabetes-induced endothelial progenitor cell (EPC) dysfunction, although the underlying mechanisms are not fully understood. In this study, we investigated the role of high mobility group box-1 (HMGB-1) in diabetes-induced oxidative stress. HMGB-1 was upregulated in both serum and bone marrow-derived monocytes from diabetic mice compared with control mice. In vitro, advanced glycation end productions (AGEs) induced, expression of HMGB-1 in EPCs and in cell culture supernatants in a dose-dependent manner. However, inhibition of oxidative stress with N-acetylcysteine (NAC) partially inhibited the induction of HMGB-1 induced by AGEs. Furthermore, p66shc expression in EPCs induced by AGEs was abrogated by incubation with glycyrrhizin (Gly), while increased superoxide dismutase (SOD) activity in cell culture supernatants was observed in the Gly treated group. Thus, HMGB-1 may play an important role in diabetes-induced oxidative stress in EPCs via a positive feedback loop involving the AGE/reactive oxygen species/HMGB-1 pathway.
Collapse
|
28
|
Wei S, Huang J, Li Y, Zhao J, Luo Y, Meng X, Sun H, Zhou X, Zhang M, Zhang W. Novel zinc finger transcription factor ZFP580 promotes differentiation of bone marrow-derived endothelial progenitor cells into endothelial cells via eNOS/NO pathway. J Mol Cell Cardiol 2015; 87:17-26. [DOI: 10.1016/j.yjmcc.2015.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 06/30/2015] [Accepted: 08/05/2015] [Indexed: 01/01/2023]
|
29
|
Xu Y, Li Z, Li X, Fan Z, Liu Z, Xie X, Guan J. Regulating myogenic differentiation of mesenchymal stem cells using thermosensitive hydrogels. Acta Biomater 2015; 26:23-33. [PMID: 26277379 DOI: 10.1016/j.actbio.2015.08.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/30/2015] [Accepted: 08/11/2015] [Indexed: 01/02/2023]
Abstract
Stem cell therapy has potential to regenerate skeletal muscle tissue in ischemic limb. However, the delivered stem cells experience low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as their modulus may be tailored to induce the differentiation. Yet current approaches used to manipulate hydrogel modulus often simultaneously vary other properties that also affect stem cell differentiation, such as chemical structure, composition and water content. Thus it is challenging to demonstrate the decoupled effect of hydrogel modulus on stem cell differentiation. In this report, we decoupled the hydrogel modulus from chemical structure, composition, and water content using injectable and thermosensitive hydrogels. The hydrogels were synthesized from N-isopropylacrylamide (NIPAAm), acrylic acid (AAc), and degradable macromer 2-hydroxyethyl methacrylate-oligomer [oligolatide, oligohydroxybutyrate, or oligo(trimethylene carbonate)]. We found that using the same monomer composition and oligomer chemical structure but different oligomer length can independently vary hydrogel modulus. Rat bone marrow mesenchymal stem cells (MSCs) were encapsulated in the hydrogels with elastic expansion moduli of 11, 20, and 40 kPa, respectively. After 14 days of culture, significant myogenic differentiation was achieved for the hydrogel with elastic expansion modulus of 20 kPa, as judged from both the gene and protein expression. In addition, MSCs exhibited an elastic expansion modulus-dependent proliferation rate. The most significant proliferation was observed in the hydrogel with elastic expansion modulus of 40 kPa. These results demonstrate that the developed injectable and thermosensitive hydrogels with suitable modulus has the potential to deliver stem cells into ischemic limb for enhanced myogenic differentiation and muscle regeneration. STATEMENT OF SIGNIFICANCE Stem cell therapy for skeletal muscle regeneration in ischemic limb experiences low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as hydrogel modulus may be modulated to induce the differentiation. Yet current approaches used to modulate hydrogel modulus may simultaneously vary other properties that also affect stem cell myogenic differentiation, such as chemistry, composition and water content. In this report, we decoupled the hydrogel modulus from chemistry, composition, and water content using injectable and thermosensitive hydrogels. We found that mesenchymal stem cells best differentiated into myogenic lineage in the hydrogel with elastic modulus of 20 kPa.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenqing Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaofei Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenguo Liu
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaoyun Xie
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Tongji Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
30
|
Ali M, Mehmood A, Anjum MS, Tarrar MN, Khan SN, Riazuddin S. Diazoxide preconditioning of endothelial progenitor cells from streptozotocin-induced type 1 diabetic rats improves their ability to repair diabetic cardiomyopathy. Mol Cell Biochem 2015; 410:267-79. [PMID: 26359087 DOI: 10.1007/s11010-015-2560-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023]
Abstract
Type 1 diabetes mellitus (DM) is a strong risk factor for the development of diabetic cardiomyopathy (DCM) which is the leading cause of morbidity and mortality in the type 1 diabetic patients. Stem cells may act as a therapeutic agent for the repair of DCM. However, deteriorated functional abilities and survival of stem cells derived from type 1 diabetic subjects need to be overcome for obtaining potential outcome of the stem cell therapy. Diazoxide (DZ) a highly selective mitochondrial ATP-sensitive K(+) channel opener has been previously shown to improve the ability of mesenchymal stem cells for the repair of heart failure. In the present study, we evaluated the effects of DZ preconditioning in improving the ability of streptozotocin-induced type 1 diabetes affected bone marrow-derived endothelial progenitor cells (DM-EPCs) for the repair of DCM in the type 1 diabetic rats. DM-EPCs were characterized by immunocytochemistry, flow cytometry, and reverse transcriptase PCR for endothelial cell-specific markers like vWF, VE cadherin, VEGFR2, PECAM, CD34, and eNOS. In vitro studies included preconditioning of DM-EPCs with 200 μM DZ for 30 min followed by exposure to either 200 μM H2O2 for 2 h (for oxidative stress induction) or 30 mM glucose media (for induction of hyperglycemic stress) for 48 h. Non-preconditioned EPCs with and without exposure to H2O2 and 30 mM high glucose served as controls. These cells were then evaluated for survival (by MTT and XTT cell viability assays), senescence, paracrine potential (by ELISA for VEGF), and alteration in gene expression [VEGF, stromal derived factor-1α (SDF-1α), HGF, bFGF, Bcl2, and Caspase-3]. DZ preconditioned DM-EPCs demonstrated significantly increased survival and VEGF release while reduced cell injury and senescence. Furthermore, DZ preconditioned DM-EPCs exhibited up-regulated expression of prosurvival genes (VEGF, SDF-1α, HGF, bFGF, and Bcl2) on exposure to H2O2, and VEGF and Bcl2 on exposure to hyperglycemia while down regulation of Caspase-3 gene. Eight weeks after type 1 diabetes induction, DZ preconditioned, and non-preconditioned DM-EPCs were transplanted into left ventricle of diabetic rats (at a dose of 2 × 10(6) DM-EPCs/70 μl serum free medium). After 4 weeks, DZ preconditioned DM-EPCs transplantation improved cardiac function as assessed by Millar's apparatus. There was decrease in collagen content estimated by Masson's trichrome and sirius red staining. Furthermore, reduced cell injury was observed as evidenced by decreased expression of Caspase-3 and increased expression of prosurvival genes Bcl2, VEGF, and bFGF by semi-quantitative real-time PCR. In conclusion, the present study demonstrated that DZ preconditioning enhanced EPCs survival under oxidative and hyperglycemic stress and their ability to treat DCM.
Collapse
Affiliation(s)
- Muhammad Ali
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan.
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan.
| | - Muhammad Sohail Anjum
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan.
| | | | - Shaheen N Khan
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan.
| | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, University of Punjab, 87-West Canal Bank Road, Lahore, Pakistan. .,Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan.
| |
Collapse
|
31
|
Isolation of Foreign Material-Free Endothelial Progenitor Cells Using CD31 Aptamer and Therapeutic Application for Ischemic Injury. PLoS One 2015; 10:e0131785. [PMID: 26148001 PMCID: PMC4493074 DOI: 10.1371/journal.pone.0131785] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/05/2015] [Indexed: 01/09/2023] Open
Abstract
Endothelial progenitor cells (EPCs) can be isolated from human bone marrow or peripheral blood and reportedly contribute to neovascularization. Aptamers are 40-120-mer nucleotides that bind to a specific target molecule, as antibodies do. To utilize apatmers for isolation of EPCs, in the present study, we successfully generated aptamers that recognize human CD31, an endothelial cell marker. CD31 aptamers bound to human umbilical cord blood-derived EPCs and showed specific interaction with human CD31, but not with mouse CD31. However, CD31 aptamers showed non-specific interaction with CD31-negative 293FT cells and addition of polyanionic competitor dextran sulfate eliminated non-specific interaction without affecting cell viability. From the mixture of EPCs and 293FT cells, CD31 aptamers successfully isolated EPCs with 97.6% purity and 94.2% yield, comparable to those from antibody isolation. In addition, isolated EPCs were decoupled from CD31 aptamers with a brief treatment of high concentration dextran sulfate. EPCs isolated with CD31 aptamers and subsequently decoupled from CD31 aptamers were functional and enhanced the restoration of blood flow when transplanted into a murine hindlimb ischemia model. In this study, we demonstrated isolation of foreign material-free EPCs, which can be utilized as a universal protocol in preparation of cells for therapeutic transplantation.
Collapse
|
32
|
Adipose-derived stromal cell cluster with light therapy enhance angiogenesis and skin wound healing in mice. Biochem Biophys Res Commun 2015; 462:171-7. [DOI: 10.1016/j.bbrc.2015.04.059] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 01/03/2023]
|
33
|
Park IS, Chung PS, Ahn JC. Enhancement of Ischemic Wound Healing by Spheroid Grafting of Human Adipose-Derived Stem Cells Treated with Low-Level Light Irradiation. PLoS One 2015; 10:e0122776. [PMID: 26065900 PMCID: PMC4465903 DOI: 10.1371/journal.pone.0122776] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/12/2015] [Indexed: 12/31/2022] Open
Abstract
We investigated whether low-level light irradiation prior to transplantation of adipose-derived stromal cell (ASC) spheroids in an animal skin wound model stimulated angiogenesis and tissue regeneration to improve functional recovery of skin tissue. The spheroid, composed of hASCs, was irradiated with low-level light and expressed angiogenic factors, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF), and hepatocyte growth factor (HGF). Immunochemical staining analysis revealed that the spheroid of the hASCs was CD31+, KDR+, and CD34+. On the other hand, monolayer-cultured hASCs were negative for these markers. PBS, human adipose tissue-derived stromal cells, and the ASC spheroid were transplanted into a wound bed in athymic mice to evaluate the therapeutic effects of the ASC spheroid in vivo. The ASC spheroid transplanted into the wound bed differentiated into endothelial cells and remained differentiated. The density of vascular formations increased as a result of the angiogenic factors released by the wound bed and enhanced tissue regeneration at the lesion site. These results indicate that the transplantation of the ASC spheroid significantly improved functional recovery relative to both ASC transplantation and PBS treatment. These findings suggest that transplantation of an ASC spheroid treated with low-level light may be an effective form of stem cell therapy for treatment of a wound bed.
Collapse
Affiliation(s)
- In-Su Park
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam, 330–714, Korea
| | - Phil-Sang Chung
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam, 330–714, Korea
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, 119 Dandae-ro, Cheonan, Chungnam, 330–714, Korea
| | - Jin Chul Ahn
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam, 330–714, Korea
- Department of Biomedical Science, Dankook University, Cheonan, Chungnam, 330–714, Korea
- Biomedical Translational Research Institute, Dankook University, Cheonan, Chungnam, 330–714, Korea
- * E-mail:
| |
Collapse
|
34
|
Melanoma cell therapy: Endothelial progenitor cells as shuttle of the MMP12 uPAR-degrading enzyme. Oncotarget 2015; 5:3711-27. [PMID: 25003596 PMCID: PMC4116515 DOI: 10.18632/oncotarget.1987] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The receptor for the urokinase-type plasminogen activator (uPAR) accounts for many features of cancer progression, and is therefore considered a target for anti-tumoral therapy. Only full length uPAR mediates tumor progression. Matrix-metallo-proteinase-12 (MMP12)-dependent uPAR cleavage results into the loss of invasion properties and angiogenesis. MMP12 can be employed in the field of “targeted therapies” as a biological drug to be delivered directly in patient's tumor mass. Endothelial Progenitor Cells (EPCs) are selectively recruited within the tumor and could be used as cellular vehicles for delivering anti-cancer molecules. The aim of our study is to inhibit cancer progression by engeneering ECFCs, a subset of EPC, with a lentivirus encoding the anti-tumor uPAR-degrading enzyme MMP12. Ex vivo manipulated ECFCs lost the capacity to perform capillary morphogenesis and acquired the anti-tumor and anti-angiogenetic activity. In vivo MMP12-engineered ECFCs cleaved uPAR within the tumor mass and strongly inhibited tumor growth, tumor angiogenesis and development of lung metastasis. The possibility to exploit tumor homing and activity of autologous MMP12-engineered ECFCs represents a novel way to combat melanoma by a “personalized therapy”, without rejection risk. The i.v. injection of radiolabelled MMP12-ECFCs can thus provide a new theranostic approach to control melanoma progression and metastasis.
Collapse
|
35
|
Karcher JR, Hoffmann BR, Liu P, Liu Y, Liang M, Greene AS. Genome-wide epigenetic and proteomic analysis reveals altered Notch signaling in EPC dysfunction. Physiol Rep 2015; 3:e12358. [PMID: 25921777 PMCID: PMC4425964 DOI: 10.14814/phy2.12358] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/27/2015] [Accepted: 03/07/2015] [Indexed: 12/13/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are bone-marrow-derived mononuclear cells that participate in tube formation in vitro and vessel formation in vivo. EPC transplantation, as a therapeutic approach in cardiovascular diseases, has produced mixed results likely due to underlying disease states and environmental factors affecting EPC function. In this study, we investigated the mechanisms by which a high-salt diet impairs EPC function. The number of endothelial progenitor cells (CD34(+), VEGFR2(+), CD133(+), and c-Kit(+)) was decreased in the bone marrow of Sprague-Dawley (SD) rats fed a high-salt diet (HSD; 4% NaCl) as compared to SD rats on a normal-salt diet (NSD; 0.4% NaCl). NSD EPCs augmented endothelial cell tube formation in vitro, whereas HSD EPCs did not. NSD EPCs were a potent therapeutic restoring electrical stimulation-induced angiogenesis in vivo. HSD EPCs were not able to restore angiogenesis in vivo. EPC DNA methylation was analyzed by reduced representative bisulfite sequencing and membrane proteins were analyzed using high accuracy liquid chromatography mass spectrometry. Differentially methylated genes and differentially abundant membrane proteins measured between the NSD and HSD EPCs, revealed a total of 886 gene-protein sets where reciprocal methylation and expression occurred. Based on stringent criteria, Notch4 was found to be hypermethylated in HSD EPCs and had corresponding decrease in protein expression. Suppression of Notch4 protein expression in EPCs using siRNA confirmed a role for Notch4 in EPC-mediated angiogenesis, suggesting Notch4 suppression as a mechanism by which high-salt diet inhibits EPC-mediated angiogenesis.
Collapse
Affiliation(s)
- Jamie R Karcher
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Brian R Hoffmann
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yong Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Andrew S Greene
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
36
|
Dragoni S, Guerra G, Fiorio Pla A, Bertoni G, Rappa A, Poletto V, Bottino C, Aronica A, Lodola F, Cinelli MP, Laforenza U, Rosti V, Tanzi F, Munaron L, Moccia F. A functional transient receptor potential vanilloid 4 (TRPV4) channel is expressed in human endothelial progenitor cells. J Cell Physiol 2015; 230:95-104. [PMID: 24911002 DOI: 10.1002/jcp.24686] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Abstract
Endothelial progenitor cells (EPCs) are mobilized into circulation to replace damaged endothelial cells and recapitulate the vascular network of injured tissues. Intracellular Ca(2+) signals are key to EPC activation, but it is yet to be elucidated whether they are endowed with the same blend of Ca(2+) -permeable channels expressed by mature endothelial cells. For instance, endothelial colony forming cells (ECFCs), the only EPC subset truly committed to acquire a mature endothelial phenotype, lack canonical transient receptor potential channels 3, 5 and 6 (TRPC3, 5 and 6), which are widely distributed in vascular endothelium; on the other hand, they express a functional store-operated Ca(2+) entry (SOCE). The present study was undertaken to assess whether human circulating EPCs possess TRP vanilloid channel 4 (TRPV4), which plays a master signalling role in mature endothelium, by controlling both vascular remodelling and arterial pressure. We found that EPCs express both TRPV4 mRNA and protein. Moreover, both GSK1016790A (GSK) and phorbol myristate acetate and, two widely employed TRPV4 agonists, induced intracellular Ca(2+) signals uniquely in presence of extracellular Ca(2+). GSK- and PMA-induced Ca(2+) elevations were inhibited by RN-1734 and ruthenium red, which selectively target TRPV4 in mature endothelium. However, TRPV4 stimulation with GSK did not cause EPC proliferation, while the pharmacological blockade of TRPV4 only modestly affected EPC growth in the presence of a growth factor-enriched culture medium. Conversely, SOCE inhibition with BTP-2, La(3+) and Gd(3+) dramatically decreased cell proliferation. These data indicate that human circulating EPCs possess a functional TRPV4 protein before their engraftment into nascent vessels.
Collapse
Affiliation(s)
- Silvia Dragoni
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lu A, Wang L, Qian L. The role of eNOS in the migration and proliferation of bone-marrow derived endothelial progenitor cells and in vitro angiogenesis. Cell Biol Int 2015; 39:484-90. [PMID: 25492215 DOI: 10.1002/cbin.10405] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/28/2014] [Indexed: 12/23/2022]
Abstract
The role of endothelial nitric oxide synthase (eNOS) in the activities of endothelial progenitor cells (EPCs) including migration, proliferation, and tube formation in vitro was investigated. EPCs were obtained from rat bone mononuclear cells by culturing for 7-10 days in EGM-2MV and identified by their capacity for FITC-UEA-1 binding and acetylated low-density lipoprotein (Dil-ac-LDL) intake using fluorescence microscopy. Migration, proliferation and tube formation activities were assessed in the presence or absence of N(ω)-nitro-L-argininemethylester (L-NAME), an eNOS inhibitor. mRNA and protein expression of CXCR4, CXCR7, VEGFR2, and eNOS were detected by real-time PCR and western blotting in the presence or absence of L-NAME. Nitric oxide production was detected by nitrate reductase in the presence or absence of L-NAME. Typical spindle-shaped cells appeared on the 7(th)-10(th) day and confluence reached about 80%. The percentage of FITC-UEA-1 and Dil-ac-LDL double-stained cells was about 85%. Cell migration, proliferation, and tube formation were significantly weakened after eNOS was inhibited (P < 0.05), and the expressions of CXCR4 and eNOS were significantly reduced (P < 0.05, respectively), but there was little change in CXCR7 and VEGFR2. NO production was dramatically decreased after eNOS was inhibited (P < 0.05). In summary, L-NAME significantly reduced the expression of eNOS and NO production by EPCs and inhibited migration, proliferation and tube formation by these cells, suggesting that eNOS affects EPC activities; CXCR4 may be implicated in the action of eNOS.
Collapse
Affiliation(s)
- Aizhen Lu
- Departments of Pediatrics, Children's Hospital of Fudan University, Shanghai, 201102, P. R. China
| | | | | |
Collapse
|
38
|
Jang IH, Heo SC, Kwon YW, Choi EJ, Kim JH. Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis. Adv Biol Regul 2014; 57:162-72. [PMID: 25304660 DOI: 10.1016/j.jbior.2014.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/03/2014] [Indexed: 12/30/2022]
Abstract
Endothelial progenitor cells (EPCs) hold a great promise as a therapeutic mediator in treatment of ischemic disease conditions. The discovery of EPCs in adult blood has been a cause of significant enthusiasm in the field of endothelial cell research and numerous clinical trials have been expedited. After more than a decade of research in basic science and clinical applications, limitations and new strategies of EPC therapeutics have emerged. With various phenotypes, vague definitions, and uncertain distinction from hematopoietic cells, understanding EPC biology remains challenging. However, EPCs, still hold great hope for treatment of critical ischemic injury as low concern regarding safety can accelerate the clinical applications from basic findings. This review provides an introduction to EPC as cellular therapeutics, which highlights a recent finding that EPC homing was promoted through FPR2 signaling.
Collapse
Affiliation(s)
- Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Soon Chul Heo
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Yang Woo Kwon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Eun Jung Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, 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
|
39
|
Park IS, Chung PS, Ahn JC. Enhanced angiogenic effect of adipose-derived stromal cell spheroid with low-level light therapy in hind limb ischemia mice. Biomaterials 2014; 35:9280-9. [PMID: 25132605 DOI: 10.1016/j.biomaterials.2014.07.061] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 11/27/2022]
Abstract
The aim of this study was to investigate the effects of low-level laser therapy (LLLT) on transplanted human adipose-derived mesenchymal stem cells (hASCs) spheroid in a hind limb ischemia animal model. LLLT, hASCs spheroid and hASCs spheroid transplantation with LLLT (spheroid + LLLT) were applied to the ischemic hind limbs in athymic mice. The survival, differentiation and secretion of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF), and hepatocyte growth factor (HGF) of the spheroid ASCs were evaluated by immunohistochemistry and western blots. Spheroid + LLLT group had enhanced the tissue regeneration, including angiogenesis, compared with the ASC group. The spheroid ASCs contributed to tissue regeneration via differentiation and secretion of growth factors. In the spheroid + LLLT group, the survival of spheroid hASCs increased with a concomitant decrease in apoptosis of spheroid hASCs in the ischemic hind limb. The secretion of growth factors was stimulated in the spheroid + LLLT group compared with the ASCs and spheroid group. These data suggested that LLLT is an effective biostimulator of spheroid hASCs in tissue regeneration that enhanced the survival of ASCs and stimulated the secretion of growth factors in the ischemic hind limb.
Collapse
Affiliation(s)
- In-Su Park
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam 330-714, South Korea
| | - Phil-Sang Chung
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam 330-714, South Korea; Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, 119 Dandae-ro, Cheonan, Chungnam 330-714, South Korea
| | - Jin Chul Ahn
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan, Chungnam 330-714, South Korea; Department of Biomedical Science, Dankook University, Cheonan, Chungnam 330-714, South Korea; Biomedical Translational Research Institute, Dankook University, Cheonan, Chungnam 330-714, South Korea.
| |
Collapse
|
40
|
The effect of autologous endothelial progenitor cell transplantation combined with extracorporeal shock-wave therapy on ischemic skin flaps in rats. Cytotherapy 2014; 16:1098-109. [DOI: 10.1016/j.jcyt.2014.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 02/17/2014] [Accepted: 02/24/2014] [Indexed: 11/19/2022]
|
41
|
Park IS, Kang JA, Kang J, Rhie JW, Kim SH. Therapeutic Effect of Human Adipose-Derived Stromal Cells Cluster in Rat Hind-Limb Ischemia. Anat Rec (Hoboken) 2014; 297:2289-98. [DOI: 10.1002/ar.22961] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 11/10/2022]
Affiliation(s)
- In-Su Park
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
| | - Jo A. Kang
- Department of Plastic Surgery, College of Medicine; The Catholic University of Korea; Seoul Korea
| | - Jungmi Kang
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
- Department of Biomedical Engineering; University of Science and Technology; Seoul Korea
| | - Jong-Won Rhie
- Department of Plastic Surgery, College of Medicine; The Catholic University of Korea; Seoul Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
- Department of Biomedical Engineering; University of Science and Technology; Seoul Korea
| |
Collapse
|
42
|
Yin C, Liang Y, Guo S, Zhou X, Pan X. CCN1 enhances angiogenic potency of bone marrow transplantation in a rat model of hindlimb ischemia. Mol Biol Rep 2014; 41:5813-8. [DOI: 10.1007/s11033-014-3455-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
|
43
|
Zhang S, Ma X, Yao K, Zhu H, Huang Z, Shen L, Qian J, Zou Y, Sun A, Ge J. Combination of CD34-positive cell subsets with infarcted myocardium-like matrix stiffness: a potential solution to cell-based cardiac repair. J Cell Mol Med 2014; 18:1236-8. [PMID: 24945435 PMCID: PMC4508162 DOI: 10.1111/jcmm.12301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/21/2014] [Indexed: 02/04/2023] Open
Abstract
Detection of the optimal cell transplantation strategy for myocardial infarction (MI) has attracted a great deal of attention. Commitment of engrafted cells to angiogenesis within damaged myocardium is regarded as one of the major targets in cell-based cardiac repair. Bone marrow–derived CD34-positive cells, a well-characterized population of stem cells, might represent highly functional endothelial progenitor cells and result in the formation of new blood vessels. Recently, physical microenvironment (extracellular matrix stiffness) around the engrafted cells was found to exert an essential impact on their fate. Stem cells are able to feel and respond to the tissue-like matrix stiffness to commit to a relevant lineage. Notably, the infarct area after MI experiences a time-dependent stiffness change from flexible to rigid. Our previous observations demonstrated myocardial stiffness-dependent differentiation of the unselected bone marrow–derived mononuclear cells (BMMNCs) along endothelial lineage cells. Myocardial stiffness (∽42 kPa) within the optimal time domain of cell engraftment (at week 1 to 2) after MI provided a more favourable physical microenvironment for cell specification and cell-based cardiac repair. However, the difference in tissue stiffness-dependent cell differentiation between the specific cell subsets expressing and no expressing CD34 phenotype remains uncertain. We presumed that CD34-positive cell subsets facilitated angiogenesis and subsequently resulted in cardiac repair under induction of infarcted myocardium-like matrix stiffness compared with CD34-negative cells. If the hypothesis were true, it would contribute greatly to detect the optimal cell subsets for cell therapy and to establish an optimized therapy strategy for cell-based cardiac repair.
Collapse
Affiliation(s)
- Shuning Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Liang CJ, Shen WC, Chang FB, Wu VC, Wang SH, Young GH, Tsai JS, Tseng YC, Peng YS, Chen YL. Endothelial Progenitor Cells Derived From Wharton's Jelly of Human Umbilical Cord Attenuate Ischemic Acute Kidney Injury by Increasing Vascularization and Decreasing Apoptosis, Inflammation, and Fibrosis. Cell Transplant 2014; 24:1363-77. [PMID: 24819279 DOI: 10.3727/096368914x681720] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury to the kidney, a major cause of acute renal failure in humans, is associated with a high mortality, and the development of a new therapeutic strategy is therefore highly desirable. In this study, we examined the therapeutic potential of implantation of endothelial progenitor cells (EPCs) isolated from Wharton's jelly of human umbilical cords in the treatment of renal I/R injury in mice. To visualize the localization of the transplanted EPCs, the cells were labeled with Q-tracker before injection into the renal capsule. Mice with renal I/R injury showed a significant increase in blood urea nitrogen and creatinine levels, and these effects were decreased by EPC transplantation. The kidney injury score in the mice with I/R injury was also significantly decreased by EPC transplantation. EPC transplantation increased the microvascular density, and some of the EPCs surrounded and were incorporated into microvessels. In addition, EPC transplantation inhibited the I/R-induced cell apoptosis of endothelial, glomerular, and renal tubular cells, as demonstrated by TUNEL staining, and significantly reduced reactive oxygen species production and the expression of the inflammatory chemokines macrophage inflammatory protein-2 and keratinocyte-derived cytokine, as shown by immunostaining and ELISA. Moreover, EPC transplantation reduced I/R-induced fibrosis, as demonstrated by immunostaining for S100A4, a fibroblast marker, and by Jones silver staining. To our knowledge, this is the first report that transplantation of EPCs from Wharton's jelly of human umbilical cords might provide a novel therapy for ischemic acute kidney injury by promoting angiogenesis and inhibiting apoptosis, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Chan-Jung Liang
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Xiao M, Lu X, Li J, Li L, Li Y. Physiologic ischaemic training induces endothelial progenitor cell mobilization and myocardial angiogenesis via endothelial nitric oxide synthase related pathway in rabbits. J Cardiovasc Med (Hagerstown) 2014; 15:280-7. [DOI: 10.2459/jcm.0b013e32836009fe] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Wang J, Fu X, Jiang C, Yu L, Wang M, Han W, Liu L, Wang J. Bone marrow mononuclear cell transplantation promotes therapeutic angiogenesis via upregulation of the VEGF-VEGFR2 signaling pathway in a rat model of vascular dementia. Behav Brain Res 2014; 265:171-80. [PMID: 24589546 DOI: 10.1016/j.bbr.2014.02.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 01/07/2023]
Abstract
Bone marrow mononuclear cells (BMMNCs) are important for angiogenesis after stroke. We investigated the effects of BMMNCs on cognitive function, angiogenesis, and the vascular endothelial growth factor (VEGF)-VEGF receptor 2 (VEGFR2) signaling pathway in a rat model of vascular dementia. We transplanted BMMNCs into rats that had undergone permanent bilateral occlusion of the common carotid arteries (2VO) and observed their migration in vivo. On day 28, we assessed cognitive function with the Morris Water Maze test and examined vascular density and white matter damage within the corpus striatum by staining with fluorescein lycopersicon esculentum (tomato) lectin or Luxol fast blue. We evaluated expression of VEGF, rapidly accelerated fibrosarcoma 1 (Raf1), and extracellular-signal-regulated kinases 1 and 2 (ERK1/2) in the ischemic hemisphere by Western blot analysis on day 7 after cell transplantation. Contribution of the VEGF-VEGFR2 signaling pathway was confirmed by using VEGFR2 inhibitor SU5416. BMMNCs penetrated the blood-brain barrier and reached the ischemic cortex and white matter or incorporated into vascular walls of 2VO rats. BMMNC-treated 2VO rats had better learning and memory, higher vascular density, and less white matter damage than did vehicle-treated rats. The beneficial effects of BMMNCs were abolished by pretreatment of rats with SU5416. Protein expression of VEGF and phosphorylated Raf1 and ERK1/2 was also significantly increased by BMMNC treatment, but this upregulation was reversed by SU5416. BMMNCs can enhance angiogenesis, reduce white matter damage, and promote cognitive recovery in 2VO rats. The angiogenic effect may result from upregulation of the VEGF-VEGFR2 signaling pathway.
Collapse
Affiliation(s)
- Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Xiaojie Fu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Lie Yu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Menghan Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wei Han
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Liu Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Kizuka-Shibuya F, Tokuda N, Takagi K, Adachi Y, Lee L, Tamura I, Maekawa R, Tamura H, Suzuki T, Owada Y, Sugino N. Locally existing endothelial cells and pericytes in ovarian stroma, but not bone marrow-derived vascular progenitor cells, play a central role in neovascularization during follicular development in mice. J Ovarian Res 2014; 7:10. [PMID: 24444000 PMCID: PMC3905919 DOI: 10.1186/1757-2215-7-10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/17/2014] [Indexed: 01/12/2023] Open
Abstract
Background Neovascularization is necessary for follicular growth. Vascularization is first observed in preantral follicles, and thereafter the vasculature markedly increases in follicles undergoing development. Neovascularization includes angiogenesis and vasculogenesis. Vasculogenesis is the formation of new blood vessels by bone marrow-derived endothelial progenitor cells. It is unclear whether vasculogenesis occurs during follicular growth. Blood vessels must be mature to be functional blood vessels. Mature blood vessels are characterized by the recruitment of pericytes. However, it is unclear where pericytes come from and whether they contribute to neovascularization in the follicle during follicular growth. In this study, we investigated whether bone marrow-derived progenitor cells that differentiate into vascular endothelial cells or pericytes contribute to neovascularization during follicular growth. Methods A parabiosis model was used in this study. Six-week-old wild-type and transgenic female mice expressing green fluorescent protein (GFP) were conjoined between the lateral abdominal regions to create a shared circulatory system. After 6 weeks, the ovaries were obtained and immunostained for CD31/CD34 (a vascular endothelial cell marker), platelet-derived growth factor receptor-β (PDGFR-β) (a pericyte marker), and GFP (a bone marrow-derived cell marker). Results Cells that were positive for CD34 and PDGFR-β were observed in the stroma adjacent to the primary or early preantral follicles and in the theca cell layer of the follicles from the late preantral stage to the preovulatory stage. CD31/CD34 and GFP double-positive cells were observed in the theca cell layer of the follicle from the antral stage to the preovulatory stage while the number of double-positive cells in the preovulatory follicles did not increase. PDGFR-β and GFP double-positive cells were observed in the theca cell layer of the preovulatory follicle but not in the smaller follicle. Conclusions Locally existing endothelial cells and pericytes in the stroma play a central role in the neovascularization during follicular growth, while bone marrow-derived endothelial cells and pericytes partially contribute to this process.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan.
| |
Collapse
|
48
|
Espinosa-Heidmann DG, Malek G, Mettu PS, Caicedo A, Saloupis P, Gach S, Dunnon AK, Hu P, Spiga MG, Cousins SW. Bone marrow transplantation transfers age-related susceptibility to neovascular remodeling in murine laser-induced choroidal neovascularization. Invest Ophthalmol Vis Sci 2013; 54:7439-49. [PMID: 24135751 DOI: 10.1167/iovs.13-12546] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Neovascular remodeling (NVR), the progression of small capillaries into large-caliber arterioles with perivascular fibrosis, represents a major therapeutic challenge in neovascular age-related macular degeneration (AMD). Neovascular remodeling occurs after laser-induced choroidal neovascularization (CNV) in aged but not young mice. Additionally, bone marrow-derived cells, including macrophages, endothelial precursor cells, and mesenchymal precursor cells, contribute to CNV severity. In this study, we investigated the impact of aged bone marrow transplantation (BMT) on the degree of fibrosis, size, and vascular morphology of CNV lesions in a mouse model of laser-induced CNV. METHODS Young (2 months) and old (16 months) mice were transplanted with green fluorescent protein (GFP)-labeled bone marrow isolated from either young or old donors. Laser CNV was induced 1 month following transplant, and eyes were analyzed via choroidal flat mounts and immunohistochemistry 1 month postlaser. The identity of cells infiltrating CNV lesions was determined using specific markers for the labeled transplanted cells (GFP+), macrophages (F4/80+), perivascular mesenchymal-derived cells (smooth muscle actin, SMA+), and endothelial cells (CD31+). RESULTS Bone marrow transplantation from aged mice transferred susceptibility to NVR into young recipients. Inversely, transplantation of young marrow into old mice prevented NVR, preserving small size and minimal fibrosis. Mice with NVR demonstrated a greater relative contribution of marrow-derived SMA+ perivascular mesenchymal cells as compared to other cells. CONCLUSIONS Our findings indicate that the status of bone marrow is an important determining factor of neovascular severity. Furthermore, we find that perivascular mesenchymal cells, rather than endothelial cells, derived from aged bone marrow may contribute to increased CNV severity in this murine model of experimental neovascularization.
Collapse
|
49
|
Moccia F, Dragoni S, Cinelli M, Montagnani S, Amato B, Rosti V, Guerra G, Tanzi F. How to utilize Ca²⁺ signals to rejuvenate the repairative phenotype of senescent endothelial progenitor cells in elderly patients affected by cardiovascular diseases: a useful therapeutic support of surgical approach? BMC Surg 2013; 13 Suppl 2:S46. [PMID: 24267290 PMCID: PMC3851045 DOI: 10.1186/1471-2482-13-s2-s46] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Endothelial dysfunction or loss is the early event that leads to a host of severe cardiovascular diseases, such as atherosclerosis, hypertension, brain stroke, myocardial infarction, and peripheral artery disease. Ageing is regarded among the most detrimental risk factor for vascular endothelium and predisposes the subject to atheroscleorosis and inflammatory states even in absence of traditional comorbid conditions. Standard treatment to restore blood perfusion through stenotic arteries are surgical or endovascular revascularization. Unfortunately, ageing patients are not the most amenable candidates for such interventions, due to high operative risk or unfavourable vascular involvement. It has recently been suggested that the transplantation of autologous bone marrow-derived endothelial progenitor cells (EPCs) might constitute an alternative and viable therapeutic option for these individuals. Albeit pre-clinical studies demonstrated the feasibility of EPC-based therapy to recapitulate the diseased vasculature of young and healthy animals, clinical studies provided less impressive results in old ischemic human patients. One hurdle associated to this kind of approach is the senescence of autologous EPCs, which are less abundant in peripheral blood and display a reduced pro-angiogenic activity. Conversely, umbilical cord blood (UCB)-derived EPCs are more suitable for cellular therapeutics due to their higher frequency and sensitivity to growth factors, such as vascular endothelial growth factor (VEGF). An increase in intracellular Ca2+ concentration is central to EPC activation by VEGF. We have recently demonstrated that the Ca2+ signalling machinery driving the oscillatory Ca2+ response to this important growth factor is different in UCB-derived EPCs as compared to their peripheral counterparts. In particular, we focussed on the so-called endothelial colony forming cells (ECFCs), which are the only EPC population belonging to the endothelial lineage and able to form capillary-like structures in vitro and stably integrate with host vasculature in vivo. The present review provides a brief description of how exploiting the Ca2+ toolkit of juvenile EPCs to restore the repairative phenotype of senescent EPCs to enhance their regenerative outcome in therapeutic settings.
Collapse
|
50
|
Zhang X, Yan X, Wang C, Tang T, Chai Y. The dose-effect relationship in extracorporeal shock wave therapy: the optimal parameter for extracorporeal shock wave therapy. J Surg Res 2013; 186:484-92. [PMID: 24035231 DOI: 10.1016/j.jss.2013.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 07/09/2013] [Accepted: 08/12/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Extracorporeal shock wave therapy (ESWT) has been demonstrated to have the angiogenic effect on ischemic tissue. We hypothesize that ESWT exerts the proangiogenesis effect with an energy density-dependent mode on the target cells. MATERIALS AND METHODS Endothelial progenitor cells (EPCs) of rats were obtained by cultivation of bone marrow-derived mononuclear cells. EPCs were divided into five groups of different energy densities, and each group was furthermore subdivided into four groups of different shock numbers. Thus, there were 20 subgroups in total. The expressions of angiogenic factors, apoptotic factors, inflammation mediators, and chemotactic factors were examined, and the proliferation activity was measured after ESWT. RESULTS When EPCs were treated with low-energy (0.04-0.13 mJ/mm(2)) shock wave, the expressions of endothelial nitric oxide synthase, angiopoietin (Ang) 1, Ang-2, and B-cell lymphoma 2 increased and those of interleukin 6, fibroblast growth factor 2, C-X-C chemokine receptor type 4, vascular endothelial growth factor a, Bcl-2-associated X protein, and caspase 3 decreased. stromal cell-derived factor 1 changed without statistical significance. When cells were treated with high-energy (0.16 mJ/mm(2)) shock wave, most of the expressions of cytokines declined except the apoptotic factors and fibroblast growth factor 2, and cells lead to apoptosis. The proliferation activity and the ratio of Ang-1/Ang-2 reached their peak values, when cells were treated with ESWT with the intensity ranging from 0.10-0.13 mJ/mm(2) and shock number ranging from 200-300 impulses. Meanwhile, a minimal value of the ratio of Bax/Bcl-2 was observed. CONCLUSIONS There is a dose-effect relationship in ESWT. The shock intensity ranging from 0.10-0.13 mJ/mm(2) and shock number ranging from 200-300 impulses were the optimal parameters for ESWT to treat cells in vitro.
Collapse
Affiliation(s)
- Xiongliang Zhang
- Department of Orthopedics, Sixth People's Hospital, Shanghai JiaoTong University, Shanghai, China
| | | | | | | | | |
Collapse
|