101
|
Taechangam N, Iyer SS, Walker NJ, Arzi B, Borjesson DL. Mechanisms utilized by feline adipose-derived mesenchymal stem cells to inhibit T lymphocyte proliferation. Stem Cell Res Ther 2019; 10:188. [PMID: 31238978 PMCID: PMC6593543 DOI: 10.1186/s13287-019-1300-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/20/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Feline adipose-derived mesenchymal stem cells (ASCs) have been successfully used in clinical trials for the treatment of immune-mediated diseases with T cell dysregulation. However, the immunomodulatory pathways utilized by feline ASCs to suppress T cell activation have not been fully determined. We investigated the mechanisms used by feline ASCs to inhibit T cell proliferation, including the soluble factors and the cell-cell contact ligands responsible for ASC-T cell interaction. METHODS The immunomodulatory activity of feline ASCs was evaluated via cell cycle analysis and in vitro mixed leukocyte reaction using specific immunomodulatory inhibitors. Cell-cell interactions were assessed with static adhesion assays, also with inhibitors. RESULTS Feline ASCs decrease T cell proliferation by causing cell cycle arrest in G0-G1. Blocking prostaglandin (PGE2), but not IDO, partially restored lymphocyte proliferation. Although PDL-1 and CD137L are both expressed on activated feline ASCs, only the interaction of intercellular adhesion molecule 1 (ICAM-1, CD54) with its ligand, lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), was responsible for ASC-T cell adhesion. Blocking this interaction reduced cell-cell adhesion and mediator (IFN-γ) secretion and signaling. CONCLUSIONS Feline ASCs utilize PGE2 and ICAM-1/LFA-1 ligand interaction to inhibit T cell proliferation with a resultant cell cycle arrest in G0-G1. These data further elucidate the mechanisms by which feline ASCs interact with T cells, help define appropriate T cell-mediated disease targets in cats that may be amenable to ASC therapy, and may also inform potential translational models for human diseases.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Smita S Iyer
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, University of California, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA. .,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
102
|
Weiss ARR, Dahlke MH. Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front Immunol 2019; 10:1191. [PMID: 31214172 PMCID: PMC6557979 DOI: 10.3389/fimmu.2019.01191] [Citation(s) in RCA: 477] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022] Open
Abstract
Expectations on mesenchymal stem cell (MSC) treatment are high, especially in the fields of sepsis, transplant medicine, and autoimmune diseases. Various pre-clinical studies have been conducted with encouraging results, although the mechanisms of action behind the observed immunomodulatory capacity of mesenchymal stem cells have not been fully understood. Previous studies have demonstrated that the immunomodulatory effect of MSCs is communicated via MSC-secreted cytokines and has been proven to rely on the local microenvironment as some of the observed effects depend on a pre-treatment of MSCs with inflammatory cytokines. Nonetheless, recent findings indicate that the cytokine-mediated effects are only one part of the equation as apoptotic, metabolically inactivated, or even fragmented MSCs have been shown to possess an immunomodulatory potential as well. Both cytokine-dependent and cytokine-independent mechanisms suggest a key role for regulatory T cells and monocytes in the overall pattern, but the principle as to why viable and non-viable MSCs have similar immunomodulatory capacities remains elusive. Here we review the current knowledge on cellular and molecular mechanisms involved in MSC-mediated immunomodulation and focus on the viability of MSCs, as there is still uncertainty concerning the tumorigenic potential of living MSCs.
Collapse
|
103
|
Zhou W, Lin J, Zhao K, Jin K, He Q, Hu Y, Feng G, Cai Y, Xia C, Liu H, Shen W, Hu X, Ouyang H. Single-Cell Profiles and Clinically Useful Properties of Human Mesenchymal Stem Cells of Adipose and Bone Marrow Origin. Am J Sports Med 2019; 47:1722-1733. [PMID: 31100005 DOI: 10.1177/0363546519848678] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) can be isolated from various tissues and can present themselves as a promising cell source for cell-based therapies. Although adipose- and bone marrow-derived mesenchymal stem cells have already been used in a considerable number of clinical trials for osteoarthritis treatment, systematic analyses from single- to bulk-cell resolution as well as clinical outcomes of these 2 MSCs are still insufficient. PURPOSE To explore the characteristics and differences of adipose-derived stem cells (ADSCs) and bone marrow MSCs (BMSCs) at single- and bulk-cell levels, to study the clinical outcomes of these 2 cells on the treatment of osteoarthritis, and to provide potential guidance on the more precise clinical application of these MSCs. STUDY DESIGN Controlled laboratory study and meta-analysis. METHODS Same donor-derived ADSCs and BMSCs were isolated and cultured. Single- and bulk-cell assays were used to identify the characteristics of these 2 cells. Meta-analysis of clinical trials was done to compare the clinical therapeutic effects in osteoarthritis treatment with ADSCs and BMSCs. RESULTS Single-cell RNA sequencing analysis showed that the population of ADSCs showed lower transcriptomic heterogeneity when compared with BMSCs. Additionally, as compared with BMSCs, ADSCs were less dependent on mitochondrial respiration for energy production. Furthermore, ADSCs had a lower expression level of human leukocyte antigen class I antigen and higher immunosuppression capacity when compared with the BMSC population. Meta-analysis of current clinical trials of osteoarthritis treatment with MSCs consistently showed that ADSCs are more stable than BMSCs in their therapeutic effect. CONCLUSION These results provide basic biological insights into human ADSCs and BMSCs at the single-cell resolution. Findings indicated that ADSCs may be a more controllable stem cell source, may be more adaptable to surviving in the hypoxic articular cavity niche, and may exhibit superiority in regulating inflammation. Based on the meta-analysis results of the different characteristics of ADSCs and BMSCs, ADSCs were implicated as being a better cell source for osteoarthritis treatment. CLINICAL RELEVANCE These results guide a more precise clinical application of adipose and bone marrow mesenchymal stem cells.
Collapse
Affiliation(s)
- Wenyan Zhou
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Junxin Lin
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Zhao
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaixiu Jin
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiulin He
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Yejun Hu
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Gang Feng
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Youzhi Cai
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Xia
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Hua Liu
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiliang Shen
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Xueqing Hu
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongwei Ouyang
- Investigation performed at the School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
104
|
Romano B, Elangovan S, Erreni M, Sala E, Petti L, Kunderfranco P, Massimino L, Restelli S, Sinha S, Lucchetti D, Anselmo A, Colombo FS, Stravalaci M, Arena V, D'Alessio S, Ungaro F, Inforzato A, Izzo AA, Sgambato A, Day AJ, Vetrano S. TNF-Stimulated Gene-6 Is a Key Regulator in Switching Stemness and Biological Properties of Mesenchymal Stem Cells. Stem Cells 2019; 37:973-987. [PMID: 30942926 DOI: 10.1002/stem.3010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/22/2019] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are well established to have promising therapeutic properties. TNF-stimulated gene-6 (TSG-6), a potent tissue-protective and anti-inflammatory factor, has been demonstrated to be responsible for a significant part of the tissue-protecting properties mediated by MSCs. Nevertheless, current knowledge about the biological function of TSG-6 in MSCs is limited. Here, we demonstrated that TSG-6 is a crucial factor that influences many functional properties of MSCs. The transcriptomic sequencing analysis of wild-type (WT) and TSG-6-/- -MSCs shows that the loss of TSG-6 expression leads to the perturbation of several transcription factors, cytokines, and other key biological pathways. TSG-6-/- -MSCs appeared morphologically different with dissimilar cytoskeleton organization, significantly reduced size of extracellular vesicles, decreased cell proliferative rate, and loss of differentiation abilities compared with the WT cells. These cellular effects may be due to TSG-6-mediated changes in the extracellular matrix (ECM) environment. The supplementation of ECM with exogenous TSG-6, in fact, rescued cell proliferation and changes in morphology. Importantly, TSG-6-deficient MSCs displayed an increased capacity to release interleukin-6 conferring pro-inflammatory and pro-tumorigenic properties to the MSCs. Overall, our data provide strong evidence that TSG-6 is crucial for the maintenance of stemness and other biological properties of murine MSCs.
Collapse
Affiliation(s)
- Barbara Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Sudharshan Elangovan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Emanuela Sala
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Luciana Petti
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Restelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Shruti Sinha
- Genome Biology Unit, Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" (INGM), Milan, Italy
| | - Donatella Lucchetti
- Institute of General Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS-Catholic University, Rome, Italy
| | - Achille Anselmo
- Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Matteo Stravalaci
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Vincenzo Arena
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS
| | - Silvia D'Alessio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Federica Ungaro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Antonio Inforzato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Angelo A Izzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Alessandro Sgambato
- Institute of General Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS-Catholic University, Rome, Italy
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine, & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
105
|
Lee SJ, Kim J, Choi WH, Park SR, Choi BH, Min BH. Immunophenotype and Immune-Modulatory Activities of Human Fetal Cartilage-Derived Progenitor Cells. Cell Transplant 2019; 28:932-942. [PMID: 30983392 PMCID: PMC6719489 DOI: 10.1177/0963689719842166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We have previously reported human fetal cartilage progenitor cells (hFCPCs) as a novel source of therapeutic cells showing high proliferation and stem cell properties superior to those of adult mesenchymal stem cells (MSCs). In this study, we investigated the immunophenotype and immune-modulatory activities of hFCPCs. With institutional review board approval, hFCPCs were isolated from fetuses at 11–13 weeks of gestation. hFCPCs showed strong expression of HLA class I molecules but low or no expression of HLA class II and co-stimulatory molecules, which was not changed significantly after 4 days of IFN-γ treatment. In a mixed lymphocyte reaction (MLR), hFCPCs showed no allogeneic immune response to peripheral blood lymphocytes (PBLs) and suppressed concanavalin A (Con A)-mediated proliferation of PBLs in a dose-dependent manner. In addition, hFCPCs inhibited Con A-induced secretion of pro-inflammatory cytokines TNF-α and IFN-γ from PBLs but showed no significant decrease of secretion of IL-10, anti-inflammatory cytokine. Co-culture of hFCPCs with stimulated PBLs for 4 days resulted in a significant increase in CD4+CD25+FoxP3+ T regulatory cells (Tregs). hFCPCs expressed LIF, TGF-β1, TSG-6, and sHLA-G5 but did not express IDO and HGF. Stimulation of hFCPCs with TNF-α for 12 h showed slight induction in the expression of LIF, TSG-6, IDO, and HGF, whereas stimulation with IFN-γ did not affect expression of any of these factors. These results suggest that hFCPCs have low allogeneic immunogenicity and immune-modulatory activity in vitro, comparable to those of MSCs. However, compared with MSCs, hFCPCs were less responsive to TNF-α and IFN-γ, and the mechanisms underlying responses to these two cell types appeared distinct.
Collapse
Affiliation(s)
- Su Jeong Lee
- 1 Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Jiyoung Kim
- 2 Department of Physiology, Inha University College of Medicine, Incheon, Korea
| | - Woo Hee Choi
- 1 Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - So Ra Park
- 2 Department of Physiology, Inha University College of Medicine, Incheon, Korea
| | - Byung Hyune Choi
- 3 Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Byoung-Hyun Min
- 1 Department of Molecular Science and Technology, Ajou University, Suwon, Korea.,4 Cell Therapy Center, Ajou University Medical Center, Suwon, Republic of Korea.,5 Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, Korea
| |
Collapse
|
106
|
Wang Y, Huang J, Gong L, Yu D, An C, Bunpetch V, Dai J, Huang H, Zou X, Ouyang H, Liu H. The Plasticity of Mesenchymal Stem Cells in Regulating Surface HLA-I. iScience 2019; 15:66-78. [PMID: 31030183 PMCID: PMC6487373 DOI: 10.1016/j.isci.2019.04.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/27/2019] [Accepted: 04/06/2019] [Indexed: 02/07/2023] Open
Abstract
A low surface expression level of human leukocyte antigen class I (HLA-I) ensures that the mesenchymal stem cells (MSCs) escape from the allogeneic recipients' immunological surveillance. Here, we discovered that both transcriptional and synthesis levels of HLA-I in MSCs increased continuously after interferon (IFN)-γ treatment, whereas interestingly, their surface HLA-I expression was downregulated after reaching an HLA-I surface expression peak. Microarray data indicated that the post-transcriptional process plays an important role in the downregulation of surface HLA-I. Further studies identified that IFN-γ-treated MSCs accelerated HLA-I endocytosis through a clathrin-independent dynamin-dependent endocytosis pathway. Furthermore, cells that have self-downregulated surface HLA-I expression elicit a weaker immune response than they previously could. Thus uncovering the plasticity of MSCs in the regulation of HLA-I surface expression would reveal insights into the membrane transportation events leading to the maintenance of low surface HLA-I expression, providing more evidence for selecting and optimizing low-immunogenic MSCs to improve the therapeutic efficiency. hESC-MSCs have the plasticity of maintaining low HLA-I expression on cell surface hESC-MSCs downregulate the surface HLA-I expression through endocytosis of HLA-I hESC-MSCs with lower HLA-I surface expression induce weaker MLR and slighter DTH
Collapse
Affiliation(s)
- Yafei Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China
| | - Jiayun Huang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Department of Orthopedic Surgery, 2nd Affiliated Hospital, Zhejiang University, School of Medicine, Zhejiang 310009, P.R.China; Orthopaedics Research Institute of Zhejiang University, Zhejiang 310009, P.R.China
| | - Lin Gong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China
| | - Dongsheng Yu
- Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P.R.China
| | - Chenrui An
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China
| | - Jun Dai
- Department of Medical Genetics, Medicum, University of Helsinki, Helsinki 00290, Finland
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310003, P.R. China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang 310003, P.R.China
| | - Xiaohui Zou
- Central Laboratory, the First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310003, P.R.China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Department of Sports Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University, School of Medicine, Hangzhou 310003, P. R. China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310003, P.R. China
| | - Hua Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, School of Medicine, Hangzhou 310058, P.R.China.
| |
Collapse
|
107
|
Yan F, Liu O, Zhang H, Zhou Y, Zhou D, Zhou Z, He Y, Tang Z, Wang S. Human dental pulp stem cells regulate allogeneic NK cells' function via induction of anti-inflammatory purinergic signalling in activated NK cells. Cell Prolif 2019; 52:e12595. [PMID: 30953394 PMCID: PMC6536423 DOI: 10.1111/cpr.12595] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/17/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Objectives Mesenchymal stem cells (MSCs) could regulate the function of various immune cells. It remains unclear whether MSCs additionally possess immunostimulatory properties. We investigated the impact of human MSCs on the responsiveness of primary natural killer (NK) cells in terms of induction of anti‐inflammatory purinergic signalling. Material and Methods We obtained human bone marrow mesenchymal stem cells (BMMSCs) and dental pulp stem cells (DPSCs). NK cells were isolated from peripheral blood of healthy volunteers. Activated NK cells were cultured with MSCs. Proliferation assay, apoptosis analysis, activating or inhibitory receptor expression and degranulation assay were used to explore NK cells’ function. High‐performance liquid chromatography was used to investigate the purinergic signalling in activated NK cells. Results Both DPSCs and BMMSCs could impair proliferation and promote apoptosis of activated NK cells. Also, activated NK cells could cause DPSCs to lyse. Furthermore, the expression of activating NK cells’ receptors was decreased, but inhibitory receptors of NK cells were elevated following co‐cultivation. NK cells acquired CD73 expression, while MSCs could release ATP into the extracellular space where nucleotides were converted into adenosine (ADO) following co‐culture system. Under the existence of exogenous 2‐chloroadenosine (CADO), the cytotoxic capacity of NK cells was remarkably depressed in a concentration‐dependent manner. Conclusions DPSCs and BMMSCs could depress NK cells’ function by hydrolysing ATP to ADO using CD39 and CD73 enzymatic activity. Our data suggested that DPSCs might represent a new strategy for treating immune‐related diseases by regulating previously unrecognized functions in innate immune responses.
Collapse
Affiliation(s)
- Fei Yan
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China.,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Ousheng Liu
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China.,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Haixia Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yueying Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Dian Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zekun Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuhong He
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zhangui Tang
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Songlin Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
108
|
Huaman O, Bahamonde J, Cahuascanco B, Jervis M, Palomino J, Torres CG, Peralta OA. Immunomodulatory and immunogenic properties of mesenchymal stem cells derived from bovine fetal bone marrow and adipose tissue. Res Vet Sci 2019; 124:212-222. [PMID: 30925336 DOI: 10.1016/j.rvsc.2019.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 01/24/2019] [Accepted: 03/20/2019] [Indexed: 02/07/2023]
Abstract
Little information is currently available on therapeutic features of bovine mesenchymal stem cells (MSCs), despite the development of large animal experimental models including cattle may open alternative strategies for investigating MSC physiology and eventual applications for regenerative therapy. The aim of the present study was to compare in vitro immunomodulatory and immunogenic potentials of bovine fetal MSCs (bfMSCs) derived from bovine fetal bone marrow (BM-MSCs) and adipose tissue (AT-MSCs). Immunomodulatory analyses in bfMSCs were performed by determination of the effect of interferon-γ (IFNγ) on mRNA levels of indoleamine 2, 3-dioxygenase (IDO), transforming growth factor β1 (TGFβ1), prostaglandin E receptor 2 (PTGER2), interleukin-6 and -10 (IL-6 and IL-10), and IDO enzymatic activity. The effect of conditioned medium from IFNγ-stimulated bfMSCs on the proliferation of alloantigen-activated peripheral blood lymphocytes (PBLs) was assessed. Immunogenicity of bfMSCs was determined by quantification of mRNA levels of major histocompatibility complex I and II (MHC-I and -II), CD80 and CD86, and the proportion of cells positive for MHC-I and -II by flowcytometry (FACS) analyses. IFNγ treatment increased IL-6, PTGER2 and IDO gene expression and activity in bfMSCs but did not affect suppressive effect on proliferation of PBLs. Lower proportion of AT-MSCs expressed MHC-I and MHC-II in comparison to BM-MSCs. In conclusion, BM-MSCs and AT-MSCs upregulated expression of immunomodulatory genes in a similar way after IFNγ stimuli. BM-MSCs and AT-MSCs in basal condition and treated with IFNγ displayed similar in vitro immunomodulatory ability. Lower expression of MHC-I and MHC-II suggest that AT-MSCs might be less immunogenic compared to BM-MSCs.
Collapse
Affiliation(s)
- Olger Huaman
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile
| | - Javiera Bahamonde
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061-0442, USA
| | - Berly Cahuascanco
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile
| | - Miguel Jervis
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile
| | - Jaime Palomino
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile
| | - Cristian G Torres
- Department of Clinical Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile
| | - Oscar A Peralta
- Department of Animal Production Sciences, Faculty of Animal and Veterinary Sciences, University of Chile, Santiago 8820808, Chile; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061-0442, USA.
| |
Collapse
|
109
|
Kong CM, Lin HD, Biswas A, Bongso A, Fong CY. Manufacturing of human Wharton's jelly stem cells for clinical use: selection of serum is important. Cytotherapy 2019; 21:483-495. [PMID: 30879965 DOI: 10.1016/j.jcyt.2019.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Human Wharton's jelly-derived mesenchymal stromal cells (hWJSCs) have gained considerable attention for their use in cell therapy. Many of these applications would require manufacturing of millions of hWJSCs. It is, therefore, necessary to develop a Good Manufacturing Practice (GMP)-compliant hWJSC expansion protocol, allowing the generation of a large quantity of cells to meet both clinical and regulatory requirements. Here, we compared human platelet lysate (HPL) and human serum (HS) in supporting clinical-grade hWJSC expansion. METHODS hWJSCs were successfully isolated from six different umbilical cords using GMP-compliant dissociation enzymes. Freshly isolated hWJSCs were cultured in media supplemented with 10% of one of the following sera: fetal bovine serum (FBS), HPL and HS. Properties of the expanded hWJSCs were analyzed. RESULTS We showed that GMP-compliant dissociation enzymes were as efficient as research-grade dissociation enzymes in isolating hWJSCs. hWJSC fresh cell yield and cell viability using HPL and HS supplementations were at greater advantages than FBS. Moreover, hWJSCs expanded in HPL and HS supplementations not only preserved classical MSCs phenotypes and differentiation potential to adipocytes, osteocytes and chondrocytes, they also enhanced the migration of skin fibroblasts. However, HS, unlike HPL, did not alter immunogenicity properties of hWJSCs. hWJSCs expanded in HS supplementation also exerted greater immunosuppressive action in inhibiting T-cell proliferation and increased extracellular matrix (ECM) gene expression, making them useful in tissue repair clinical application. CONCLUSION Our findings indicate that HS can be considered as a promising and safer alternative to FBS, and should be recommended for clinical-grade expansion of hWJSCs.
Collapse
Affiliation(s)
- Chiou Mee Kong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Hao Daniel Lin
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Arijit Biswas
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Ariff Bongso
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore.
| | - Chui-Yee Fong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore.
| |
Collapse
|
110
|
Hakim R, Covacu R, Zachariadis V, Frostell A, Sankavaram S, Svensson M, Brundin L. Syngeneic, in contrast to allogeneic, mesenchymal stem cells have superior therapeutic potential following spinal cord injury. J Neuroimmunol 2019; 328:5-19. [PMID: 30551037 DOI: 10.1016/j.jneuroim.2018.11.005] [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] [Received: 10/11/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 01/11/2023]
Abstract
We evaluated the importance of histocompatibility of transplanted MSCs in terms of therapeutic potential. Mouse syngeneic MSCs or allogeneic MSCs were transplanted following SCI in mouse. In this study we found that syngeneic, but not allogeneic, MSCs alternatively activated macrophages resulting in a down-regulation of pro-inflammation. Syngeneic MSCs also had a general suppressive effect on the immune response as compared to allogeneic MSCs. Additionally, syngeneic, but not allogeneic, MSCs significantly enhanced the recovery of hind limb function. In this study we show that the histocompatibility of transplanted MSCs is of importance for their therapeutic potential.
Collapse
Affiliation(s)
- Ramil Hakim
- Center for Molecular Medicine, Karolinska Institutet, Solna 17176, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Ruxandra Covacu
- Center for Molecular Medicine, Karolinska Institutet, Solna 17176, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Vasilios Zachariadis
- Department of Oncology and Pathology, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Arvid Frostell
- Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Sreenivasa Sankavaram
- Center for Molecular Medicine, Karolinska Institutet, Solna 17176, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Mikael Svensson
- Department of Neurology and Neurosurgery, Karolinska University Hospital, BioClinicum, Solnavägen 30, Solna, Stockholm 17176, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| | - Lou Brundin
- Department of Neurology and Neurosurgery, Karolinska University Hospital, BioClinicum, Solnavägen 30, Solna, Stockholm 17176, Sweden; Center for Molecular Medicine, Karolinska Institutet, Solna 17176, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Solna 17176, Stockholm, Sweden.
| |
Collapse
|
111
|
Liao S, Zhang Y, Ting S, Zhen Z, Luo F, Zhu Z, Jiang Y, Sun S, Lai WH, Lian Q, Tse HF. Potent immunomodulation and angiogenic effects of mesenchymal stem cells versus cardiomyocytes derived from pluripotent stem cells for treatment of heart failure. Stem Cell Res Ther 2019; 10:78. [PMID: 30845990 PMCID: PMC6407247 DOI: 10.1186/s13287-019-1183-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Optimal cell type as cell-based therapies for heart failure (HF) remains unclear. We sought to compare the safety and efficacy of direct intramyocardial transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) in a porcine model of HF. METHODS Eight weeks after induction of HF with myocardial infarction (MI) and rapid pacing, animals with impaired left ventricular ejection fraction (LVEF) were randomly assigned to receive direct intramyocardial injection of saline (MI group), 2 × 108 hESC-CMs (hESC-CM group), or 2 × 108 hiPSC-MSCs (hiPSC-MSC group). The hearts were harvested for immunohistochemical evaluation after serial echocardiography and hemodynamic evaluation and ventricular tachyarrhythmia (VT) induction by in vivo programmed electrical stimulation. RESULTS At 8 weeks post-transplantation, LVEF, left ventricular maximal positive pressure derivative, and end systolic pressure-volume relationship were significantly higher in the hiPSC-MSC group but not in the hESC-CM group compared with the MI group. The incidence of early spontaneous ventricular tachyarrhythmia (VT) episodes was higher in the hESC-CM group but the incidence of inducible VT was similar among the different groups. Histological examination showed no tumor formation but hiPSC-MSCs exhibited a stronger survival capacity by activating regulatory T cells and reducing the inflammatory cells. In vitro study showed that hiPSC-MSCs were insensitive to pro-inflammatory interferon-gamma-induced human leukocyte antigen class II expression compared with hESC-CMs. Moreover, hiPSC-MSCs also significantly enhanced angiogenesis compared with other groups via increasing expression of distinct angiogenic factors. CONCLUSIONS Our results demonstrate that transplantation of hiPSC-MSCs is safe and does not increase proarrhythmia or tumor formation and superior to hESC-CMs for the improvement of cardiac function in HF. This is due to their immunomodulation that improves in vivo survival and enhanced angiogenesis via paracrine effects.
Collapse
Affiliation(s)
- Songyan Liao
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Yuelin Zhang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China.,Department of Emergency, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Sherwin Ting
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, 138668, Singapore
| | - Zhe Zhen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Fan Luo
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China
| | - Ziyi Zhu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China
| | - Yu Jiang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Sijia Sun
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Qizhou Lian
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China. .,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China.
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China. .,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China.
| |
Collapse
|
112
|
Magatti M, Vertua E, Cargnoni A, Silini A, Parolini O. The Immunomodulatory Properties of Amniotic Cells: The Two Sides of the Coin. Cell Transplant 2019; 27:31-44. [PMID: 29562786 PMCID: PMC6434482 DOI: 10.1177/0963689717742819] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Among the many cell types useful in developing therapeutic treatments, human amniotic cells from placenta have been proposed as valid candidates. Both human amniotic epithelial and mesenchymal stromal cells, and the conditioned medium generated from their culture, exert multiple immunosuppressive activities. Indeed, they inhibit T and B cell proliferation, suppress inflammatory properties of monocytes, macrophages, dendritic cells, neutrophils, and natural killer cells, while promoting induction of cells with regulatory functions such as regulatory T cells and anti-inflammatory M2 macrophages. These properties have laid the foundation for their use for the treatment of inflammatory-based diseases, and encouraging results have been obtained in different preclinical disease models where exacerbated inflammation is present. Moreover, an immune-privileged status of amniotic cells has been often highlighted. However, even if long-term engraftment of amniotic cells has been reported into immunocompetent animals, only few cells survive after infusion. Furthermore, amniotic cells have been shown to be able to induce immune responses in vivo and, under specific culture conditions, they can stimulate T cell proliferation in vitro. Although immunosuppressive properties are a widely recognized characteristic of amniotic cells, immunogenic and stimulatory activities appear to be less reported, sporadic events. In order to improve therapeutic outcome, the mechanisms responsible for the suppressive versus stimulatory activity need to be carefully addressed. In this review, both the immunosuppressive and immunostimulatory activity of amniotic cells will be discussed.
Collapse
Affiliation(s)
- Marta Magatti
- 1 Centro di Ricerca "E. Menni", Fondazione Poliambulanza- Istituto Ospedaliero, Brescia, Italy
| | - Elsa Vertua
- 1 Centro di Ricerca "E. Menni", Fondazione Poliambulanza- Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- 1 Centro di Ricerca "E. Menni", Fondazione Poliambulanza- Istituto Ospedaliero, Brescia, Italy
| | - Antonietta Silini
- 1 Centro di Ricerca "E. Menni", Fondazione Poliambulanza- Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- 1 Centro di Ricerca "E. Menni", Fondazione Poliambulanza- Istituto Ospedaliero, Brescia, Italy.,2 Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| |
Collapse
|
113
|
Sharma A, Mawrie D, Magdalene D, Jaganathan BG. Isolation of Multipotent Mesenchymal Stem Cells from Human Extraocular Muscle Tissue. Bio Protoc 2019; 9:e3167. [PMID: 33654973 DOI: 10.21769/bioprotoc.3167] [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: 12/12/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 11/02/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted significant attention as potential therapeutic cells to treat various diseases ranging from tissue injuries, graft versus host disease, degenerative diseases and cancer. Since the initial discovery of MSCs in the bone marrow cells, MSCs have been successfully isolated from various adult and neo-natal tissues, albeit the procedures are often coupled with difficulties in harvesting tissue and produce low yield of cells, requiring extensive expansion in vitro. Here, we explored extra-ocular muscle tissues obtained from patients as a novel source of MSCs which express characteristic cell surface markers of MSCs and show multilineage differentiation potential with high proliferation capacity.
Collapse
Affiliation(s)
- Amit Sharma
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| | - Darilang Mawrie
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| | - Damaris Magdalene
- Department of Pediatric Ophthalmology, Sri Sankaradeva Nethralaya, Guwahati, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| |
Collapse
|
114
|
Mesenchymal stem cells for inflammatory airway disorders: promises and challenges. Biosci Rep 2019; 39:BSR20182160. [PMID: 30610158 PMCID: PMC6356012 DOI: 10.1042/bsr20182160] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
The regenerative and immunomodulatory characteristics of mesenchymal stem cells (MSCs) make them attractive in the treatment of many diseases. Although they have shown promising preclinical studies of immunomodulation and paracrine effects in inflammatory airway disorders and other lung diseases, there are still challenges that have to be overcome before MSCs can be safely, effectively, and routinely applied in the clinical setting. A good understanding of the roles and mechanisms of the MSC immunomodulatory effects will benefit the application of MSC-based clinical therapy. In this review, we summarize the promises and challenges of the preclinical and clinical trials of MSC therapies, aiming to better understand the role that MSCs play in attempt to treat inflammatory airway disorders.
Collapse
|
115
|
Mahmoud M, Abu-Shahba N, Azmy O, El-Badri N. Impact of Diabetes Mellitus on Human Mesenchymal Stromal Cell Biology and Functionality: Implications for Autologous Transplantation. Stem Cell Rev Rep 2019; 15:194-217. [DOI: 10.1007/s12015-018-9869-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
116
|
Immunomodulatory Functions of Mesenchymal Stem Cells in Tissue Engineering. Stem Cells Int 2019; 2019:9671206. [PMID: 30766609 PMCID: PMC6350611 DOI: 10.1155/2019/9671206] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/26/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response to chronic injury affects tissue regeneration and has become an important factor influencing the prognosis of patients. In previous stem cell treatments, it was revealed that stem cells not only have the ability for direct differentiation or regeneration in chronic tissue damage but also have a regulatory effect on the immune microenvironment. Stem cells can regulate the immune microenvironment during tissue repair and provide a good "soil" for tissue regeneration. In the current study, the regulation of immune cells by mesenchymal stem cells (MSCs) in the local tissue microenvironment and the tissue damage repair mechanisms are revealed. The application of the concepts of "seed" and "soil" has opened up new research avenues for regenerative medicine. Tissue engineering (TE) technology has been used in multiple tissues and organs using its biomimetic and cellular cell abilities, and scaffolds are now seen as an important part of building seed cell microenvironments. The effect of tissue engineering techniques on stem cell immune regulation is related to the shape and structure of the scaffold, the preinflammatory microenvironment constructed by the implanted scaffold, and the material selection of the scaffold. In the application of scaffold, stem cell technology has important applications in cartilage, bone, heart, and liver and other research fields. In this review, we separately explore the mechanism of MSCs in different tissue and organs through immunoregulation for tissue regeneration and MSC combined with 3D scaffolds to promote MSC immunoregulation to repair damaged tissues.
Collapse
|
117
|
Mesenchymal stem cell-based therapy for autoimmune diseases: emerging roles of extracellular vesicles. Mol Biol Rep 2019; 46:1533-1549. [PMID: 30623280 DOI: 10.1007/s11033-019-04588-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2019] [Indexed: 02/07/2023]
Abstract
In autoimmune disease body's own immune system knows healthy cells as undesired and foreign cells. Over 80 types of autoimmune diseases have been recognized. Currently, at clinical practice, treatment strategies for autoimmune disorders are based on relieving symptoms and preventing difficulties. In other words, there is no effective and useful therapy up to now. It has been well-known that mesenchymal stem cells (MSCs) possess immunomodulatory effects. This strongly suggests that MSCs might be as a novel modality for treatment of autoimmune diseases. Supporting this notion a few preclinical and clinical studies indicate that MSCs ameliorate autoimmune disorders. Interestingly, it has been found that the beneficial effects of MSCs in autoimmune disorders are not relying only on direct cell-to-cell communication but on their capability to produce a broad range of paracrine factors including growth factors, cytokines and extracellular vehicles (EVs). EVs are multi-signal messengers that play a serious role in intercellular signaling through carrying cargo such as mRNA, miRNA, and proteins. Numerous studies have shown that MSC-derived EVs are able to mimic the effects of the cell of origin on immune cells. In this review, we discuss the current studies dealing with MSC-based therapies in autoimmune diseases and provide a vision and highlight in order to introduce MSC-derived EVs as an alternative and emerging modality for autoimmune disorders.
Collapse
|
118
|
In Vitro Mesenchymal Progenitor Cell Expansion is a Predictor of Transplant-related Mortality and acute GvHD III-IV After Bone Marrow Transplantation in Univariate Analysis: A Large Single-Center Experience. J Pediatr Hematol Oncol 2019; 41:42-46. [PMID: 30113355 DOI: 10.1097/mph.0000000000001281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent stem cells able to differentiate into mesenchymal origin tissue and support the growth of hematopoietic stem cells. In order to understand the role of MSCs infused in bone marrow grafts, 53 consecutive patients were analyzed for engraftment, acute and chronic graft-versus-host disease (GvHD), transplant-related mortality (TRM), relapse incidence, and overall survival. The MSC content was measured as MSC expansion at the second passage. When in vitro-expanded MSC (cumulative population doubling at second passage, cPDp2) values were stratified according to the median value (2.2-fold increase), the univariate analysis showed a significant difference in TRM (23% vs. 3.8%, P=0.05.) and in acute GvHD III-IV incidence (12% vs. 4%, P=0.04), while the multivariate analysis did not confirm its independent role. No clinical parameters in donors and recipients were identified as predictors of cPDp2 expansion. Our study suggests a role for short-term ex vivo-expanded MSCs in reduced aGVHD III-IV incidence and TRM in univariate analysis. A multicenter, larger study is warranted to confirm these data.
Collapse
|
119
|
Mesenchymal stromal cells prevent progression of liver fibrosis in a novel zebrafish embryo model. Sci Rep 2018; 8:16005. [PMID: 30375438 PMCID: PMC6207680 DOI: 10.1038/s41598-018-34351-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/12/2018] [Indexed: 12/22/2022] Open
Abstract
Chronic liver damage leads to the onset of fibrogenesis. Rodent models for liver fibrosis have been widely used, but are less suitable for screening purposes. Therefore the aim of our study was to design a novel model for liver fibrosis in zebrafish embryos, suitable for high throughput screening. Furthermore, we evaluated the efficacy of mesenchymal stromal cells (MSCs) to inhibit the fibrotic process and thereby the applicability of this model to evaluate therapeutic responses. Zebrafish embryos were exposed to TAA or CCL4 and mRNA levels of fibrosis-related genes (Collagen-1α1, Hand-2, and Acta-2) and tissue damage-related genes (TGF-β and SDF-1a, SDF-1b) were determined, while Sirius-red staining was used to estimate collagen deposition. Three days after start of TAA exposure, MSCs were injected after which the fibrotic response was determined. In contrast to CCL4, TAA resulted in an upregulation of the fibrosis-related genes, increased extracellular matrix deposition and decreased liver sizes suggesting the onset of fibrosis. The applicability of this model to evaluate therapeutic responses was shown by local treatment with MSCs which resulted in decreased expression of the fibrosis-related RNA markers. In conclusion, TAA induces liver fibrosis in zebrafish embryos, thereby providing a promising model for future mechanistic and therapeutic studies.
Collapse
|
120
|
Siniscalco D, Kannan S, Semprún-Hernández N, Eshraghi AA, Brigida AL, Antonucci N. Stem cell therapy in autism: recent insights. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:55-67. [PMID: 30425534 PMCID: PMC6204871 DOI: 10.2147/sccaa.s155410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorders (ASDs) are characterized by core domains: persistent deficits in social communication and interaction; restricted, repetitive patterns of behavior, interests, or activities. ASDs comprise heterogeneous and complex neurodevelopmental pathologies with well-defined inflammatory conditions and immune system dysfunction. Due to neurobiologic changes underlying ASD development, cell-based therapies have been proposed and applied to ASDs. Indeed, stem cells show specific immunologic properties, which make them promising candidates in ASD treatment. This comprehensive up-to-date review focuses on ASD cellular/molecular abnormalities, potentially useful stem cell types, animal models, and current clinical trials on the use of stem cells in treating autism. Limitations are also discussed.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, University of Campania, Napoli, Italy,
| | - Suresh Kannan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Neomar Semprún-Hernández
- Research Division, Autism Immunology Unit of Maracaibo, Catedra libre de Autismo, Universidad del Zulia, Maracaibo, Venezuela
| | - Adrien A Eshraghi
- Department of Otolaryngology, Hearing Research and Cochlear Implant Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Nicola Antonucci
- Biomedical Centre for Autism Research and Treatment, Bari, Italy
| |
Collapse
|
121
|
Chaput CD, Shar A, Jupiter D, Hubert Z, Clough B, Krause U, Gregory CA. How stem cell composition in bone marrow aspirate relates to clinical outcomes when used for cervical spine fusion. PLoS One 2018; 13:e0203714. [PMID: 30248138 PMCID: PMC6152872 DOI: 10.1371/journal.pone.0203714] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/24/2018] [Indexed: 01/14/2023] Open
Abstract
Anterior cervical discectomy and fusion (ACDF) is performed to relieve pain caused by degenerative disk disease and nerve obstruction. As an alternative to bone graft, autologous concentrated bone marrow aspirate (CBMA) is used to achieve vertebral fusion with a satisfactory success rate. This has been attributed in part to bone marrow-resident mesenchymal stromal cells (MSCs) with the capacity to differentiate into osteoblasts and generate bone tissue. To date, there has been no study comparing cellular yields, MSC frequencies and their osteogenic potential with ACDF outcome. Patients (n = 24) received ACDF with CBMA and allograft bone matrix. Colony forming unit fibroblast (CFU-F) and CFU-osteoblasts (CFU-O) assays were performed on CBMA samples to enumerate MSCs (CFU-F) and osteogenic MSCs (CFU-O). CFUs were normalized to CBMA volume to define yield and also to mononuclear cells (MNC) to define frequency. After 1-year, fusion rates were good (86.7%) with pain and disability improved. There was a negative relationship between MNC and CFU-F measurements with age of patient and CFU-Os negatively correlated with age in females but not males. Tobacco use did not affect CBMA but was associated with poorer clinical outcome. Surprisingly, we found that while high-grade fusion was not associated with CFU-O, it correlated strongly (p<0.0067) with CBMA containing the lowest frequencies of CFU-F (3.0x10-6–5.83x10-5 CFU-F/MNC). MNC levels alone were not responsible for the results. These observations suggest that osteogenesis by human bone marrow is controlled by homeostatic ratio of MSCs to other cellular bone marrow components rather than absolute level of osteogenic MSCs, and that a lower ratio of MSCs to other cellular components in marrow tends to predict effective osteogenesis during ACDF. The results presented herein challenge the current dogma surrounding the proposed mechanism of MSCs in bone healing.
Collapse
Affiliation(s)
- Christopher D. Chaput
- Department of Orthopedics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
- * E-mail: (CAG); (CC)
| | - Adam Shar
- Department of Orthopedics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
- Medical Education Building, Texas A&M Health Science Center, Temple Campus, Temple, Texas, United States of America
| | - Daniel Jupiter
- Department of Preventive Medicine and Community Health, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Zach Hubert
- Medical Education Building, Texas A&M Health Science Center, Temple Campus, Temple, Texas, United States of America
| | - Bret Clough
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, Texas, United States of America
| | - Ulf Krause
- Institute for Transfusion Medicine and Transplant Immunology, University Hospital Muenster, Muenster, Germany
| | - Carl A. Gregory
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, Texas, United States of America
- * E-mail: (CAG); (CC)
| |
Collapse
|
122
|
Evaluation of platelet lysate as a substitute for FBS in explant and enzymatic isolation methods of human umbilical cord MSCs. Sci Rep 2018; 8:12439. [PMID: 30127445 PMCID: PMC6102222 DOI: 10.1038/s41598-018-30772-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have immense potential for cell-based therapy of acute and chronic pathological conditions. MSC transplantation for cell-based therapy requires a substantial number of cells in the range of 0.5–2.5 × 106 cells/kg body weight of an individual. A prolific source of MSCs followed by in vitro propagation is therefore an absolute prerequisite for clinical applications. Umbilical cord tissue (UCT) is an abundantly available prolific source of MSC that are fetal in nature and have higher potential for ex-vivo expansion. However, the ex-vivo expansion of MSCs using a xenogeneic supplement such as fetal bovine serum (FBS) carries the risk of transmission of zoonotic infections and immunological reactions. We used platelet lysate (PL) as a xeno-free, allogeneic replacement for FBS and compared the biological and functional characteristics of MSC processed and expanded with PL and FBS by explant and enzymatic method. UCT-MSCs expanded using PL displayed typical immunophenotype, plasticity, immunomodulatory property and chromosomal stability. PL supplementation also showed 2-fold increase in MSC yield from explant culture with improved immunomodulatory activity as compared to enzymatically dissociated cultures. In conclusion, PL from expired platelets is a viable alternative to FBS for generating clinically relevant numbers of MSC from explant cultures over enzymatic method.
Collapse
|
123
|
Bor R, Fábián A, Farkas K, Molnár T, Szepes Z. Human mesenchymal stem cell therapy in the management of luminal and perianal fistulizing Crohn’s disease – review of pathomechanism and existing clinical data. Expert Opin Biol Ther 2018; 18:737-745. [DOI: 10.1080/14712598.2018.1492543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Renáta Bor
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Anna Fábián
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Klaudia Farkas
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Molnár
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán Szepes
- First Department of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
124
|
Bolli R, Hare JM, Henry TD, Lenneman CG, March KL, Miller K, Pepine CJ, Perin EC, Traverse JH, Willerson JT, Yang PC, Gee AP, Lima JA, Moyé L, Vojvodic RW, Sayre SL, Bettencourt J, Cohen M, Ebert RF, Simari RD. Rationale and Design of the SENECA (StEm cell iNjECtion in cAncer survivors) Trial. Am Heart J 2018; 201:54-62. [PMID: 29910056 PMCID: PMC7282462 DOI: 10.1016/j.ahj.2018.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/07/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVES SENECA (StEm cell iNjECtion in cAncer survivors) is a phase I, randomized, double-blind, placebo-controlled study to evaluate the safety and feasibility of delivering allogeneic mesenchymal stromal cells (allo-MSCs) transendocardially in subjects with anthracycline-induced cardiomyopathy (AIC). BACKGROUND AIC is an incurable and often fatal syndrome, with a prognosis worse than that of ischemic or nonischemic cardiomyopathy. Recently, cell therapy with MSCs has emerged as a promising new approach to repair damaged myocardium. METHODS The study population is 36 cancer survivors with a diagnosis of AIC, left ventricular (LV) ejection fraction ≤40%, and symptoms of heart failure (NYHA class II-III) on optimally-tolerated medical therapy. Subjects must be clinically free of cancer for at least two years with a ≤ 30% estimated five-year risk of recurrence. The first six subjects participated in an open-label, lead-in phase and received 100 million allo-MSCs; the remaining 30 will be randomized 1:1 to receive allo-MSCs or vehicle via 20 transendocardial injections. Efficacy measures (obtained at baseline, 6 months, and 12 months) include MRI evaluation of LV function, LV volumes, fibrosis, and scar burden; assessment of exercise tolerance (six-minute walk test) and quality of life (Minnesota Living with Heart Failure Questionnaire); clinical outcomes (MACE and cumulative days alive and out of hospital); and biomarkers of heart failure (NT-proBNP). CONCLUSIONS This is the first clinical trial using direct cardiac injection of cells for the treatment of AIC. If administration of allo-MSCs is found feasible and safe, SENECA will pave the way for larger phase II/III studies with therapeutic efficacy as the primary outcome.
Collapse
Affiliation(s)
| | - Joshua M Hare
- University of Miami Miller School of Medicine, Miami, Florida
| | | | | | - Keith L March
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Kathy Miller
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Carl J Pepine
- University of Florida School of Medicine, Gainesville, Florida
| | | | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN
| | | | - Phillip C Yang
- Stanford University School of Medicine, Stanford, California
| | | | | | - Lem Moyé
- UT Health School of Public Health, Houston, TX.
| | | | | | | | | | - Ray F Ebert
- NIH, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Robert D Simari
- University of Kansas School of Medicine, Kansas City, Kansas
| | | |
Collapse
|
125
|
Wang L, Zhu CY, Ma DX, Gu ZY, Xu CC, Wang FY, Chen JG, Liu CJ, Guan LX, Gao R, Gao Z, Fang S, Zhuo DJ, Liu SF, Gao CJ. Efficacy and safety of mesenchymal stromal cells for the prophylaxis of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: a meta-analysis of randomized controlled trials. Ann Hematol 2018; 97:1941-1950. [PMID: 29947972 DOI: 10.1007/s00277-018-3384-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
Abstract
A meta-analysis of randomized controlled trials (RCTs) was conducted to evaluate the efficacy and safety of mesenchymal stromal cells (MSCs) for the prophylaxis of chronic graft-versus-host disease (cGVHD) in patients with hematological malignancies undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Six studies involving 365 patients were included. The pooled results showed that MSCs significantly reduced the incidence of cGVHD (risk ratio [RR] 0.63, 95% confidence interval [CI] 0.46 to 0.86, P = 0.004). Favorable prophylactic effects of MSCs on cGVHD were observed with umbilical cord-derived, high-dose, and late-infusion MSCs, while bone marrow-derived, low-dose, and coinfused MSCs did not confer beneficial prophylactic effects. In addition, MSC infusion did not increase the risk of primary disease relapse and infection (RR 1.02, 95% CI 0.70 to 1.50, P = 0.913; RR 0.89, 95% CI 0.44 to 1.81, P = 0.752; respectively). Moreover, there was an apparent trend toward increased overall survival (OS) in the MSC group compared with that in the control group (RR 1.13, 95% CI 0.98 to 1.29, P = 0.084). In conclusion, this meta-analysis demonstrated that MSC infusion is an effective and safe prophylactic strategy for cGVHD in patients with hematological malignancies undergoing allo-HSCT.
Collapse
Affiliation(s)
- Li Wang
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Cheng-Ying Zhu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - De-Xun Ma
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Zhen-Yang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Chang-Chun Xu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Fei-Yan Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Ji-Gang Chen
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Cheng-Jun Liu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Li-Xun Guan
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Rui Gao
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Zhe Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Shu Fang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Du-Jun Zhuo
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Shu-Feng Liu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China.
| | - Chun-Ji Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
126
|
Alfaifi M, Eom YW, Newsome PN, Baik SK. Mesenchymal stromal cell therapy for liver diseases. J Hepatol 2018; 68:1272-1285. [PMID: 29425678 DOI: 10.1016/j.jhep.2018.01.030] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/16/2018] [Accepted: 01/30/2018] [Indexed: 02/08/2023]
Abstract
The therapeutic potential of mesenchymal stromal cells (MSCs) in the treatment of liver fibrosis is predominantly based on their immunosuppressive properties, and their ability to secrete various trophic factors. This potential has been investigated in clinical and preclinical studies. Although the therapeutic mechanisms of MSC transplantation are still not fully characterised, accumulating evidence has revealed that various trophic factors secreted by MSCs play key therapeutic roles in regeneration by alleviating inflammation, apoptosis, and fibrosis as well as stimulating angiogenesis and tissue regeneration in damaged liver. In this review, we summarise the safety, efficacy, potential transplantation routes and therapeutic effects of MSCs in patients with liver fibrosis. We also discuss some of the key strategies to enhance the functionality of MSCs, which include sorting and/or priming with factors such as cytokines, as well as genetic engineering.
Collapse
Affiliation(s)
- Mohammed Alfaifi
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Philip N Newsome
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, UK; National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, UK; Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Soon Koo Baik
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea.
| |
Collapse
|
127
|
Iohara K, Utsunomiya S, Kohara S, Nakashima M. Allogeneic transplantation of mobilized dental pulp stem cells with the mismatched dog leukocyte antigen type is safe and efficacious for total pulp regeneration. Stem Cell Res Ther 2018; 9:116. [PMID: 29703239 PMCID: PMC5921747 DOI: 10.1186/s13287-018-0855-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
Background We recently demonstrated that autologous transplantation of mobilized dental pulp stem cells (MDPSCs) was a safe and efficacious potential therapy for total pulp regeneration in a clinical study. The autologous MDPSCs, however, have some limitations to overcome, such as limited availability of discarded teeth from older patients. In the present study, we investigated whether MDPSCs can be used for allogeneic applications to expand their therapeutic use. Methods Analysis of dog leukocyte antigen (DLA) was performed using polymerase chain reaction from blood. Canine allogeneic MDPSCs with the matched and mismatched DLA were transplanted with granulocyte-colony stimulating factor in collagen into pulpectomized teeth respectively (n = 7, each). Results were evaluated by hematoxylin and eosin staining, Masson trichrome staining, PGP9.5 immunostaining, and BS-1 lectin immunostaining performed 12 weeks after transplantation. The MDPSCs of the same DLA used in the first transplantation were further transplanted into another pulpectomized tooth and evaluated 12 weeks after transplantation. Results There was no evidence of toxicity or adverse events of the allogeneic transplantation of the MDPSCs with the mismatched DLA. No adverse event of dual transplantation of the MDPSCs with the matched and mismatched DLA was observed. Regenerated pulp tissues including neovascularization and neuronal extension were quantitatively and qualitatively similar at 12 weeks in both matched and mismatched DLA transplants. Regenerated pulp tissue was similarly observed in the dual transplantation as in the single transplantation of MDPSCs both with the matched and mismatched DLA. Conclusions Dual allogeneic transplantation of MDPSCs with the mismatched DLA is a safe and efficacious method for total pulp regeneration. Electronic supplementary material The online version of this article (10.1186/s13287-018-0855-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Koichiro Iohara
- Department of Stem Cell Biology and Regenerative Medicine, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| | - Shinji Utsunomiya
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories Ltd, Kagoshima, Japan
| | - Sakae Kohara
- Preclinical Research Support Division, Shin Nippon Biomedical Laboratories Ltd, Kainan, Japan
| | - Misako Nakashima
- Department of Stem Cell Biology and Regenerative Medicine, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka, Obu, Aichi, 474-8511, Japan.
| |
Collapse
|
128
|
Layek B, Sadhukha T, Panyam J, Prabha S. Nano-Engineered Mesenchymal Stem Cells Increase Therapeutic Efficacy of Anticancer Drug Through True Active Tumor Targeting. Mol Cancer Ther 2018; 17:1196-1206. [PMID: 29592881 DOI: 10.1158/1535-7163.mct-17-0682] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/10/2018] [Accepted: 03/09/2018] [Indexed: 12/18/2022]
Abstract
Tumor-targeted drug delivery has the potential to improve therapeutic efficacy and mitigate non-specific toxicity of anticancer drugs. However, current drug delivery approaches rely on inefficient passive accumulation of the drug carrier in the tumor. We have developed a unique, truly active tumor-targeting strategy that relies on engineering mesenchymal stem cells (MSC) with drug-loaded nanoparticles. Our studies using the A549 orthotopic lung tumor model show that nano-engineered MSCs carrying the anticancer drug paclitaxel (PTX) home to tumors and create cellular drug depots that release the drug payload over several days. Despite significantly lower doses of PTX, nano-engineered MSCs resulted in significant inhibition of tumor growth and superior survival. Anticancer efficacy of nano-engineered MSCs was confirmed in immunocompetent C57BL/6 albino female mice bearing orthotopic Lewis Lung Carcinoma (LL/2-luc) tumors. Furthermore, at doses that resulted in equivalent therapeutic efficacy, nano-engineered MSCs had no effect on white blood cell count, whereas PTX solution and PTX nanoparticle treatments caused leukopenia. Biodistribution studies showed that nano-engineered MSCs resulted in greater than 9-fold higher AUClung of PTX (1.5 μg.day/g) than PTX solution and nanoparticles (0.2 and 0.1 μg.day/g tissue, respectively) in the target lung tumors. Furthermore, the lung-to-liver and the lung-to-spleen ratios of PTX were several folds higher for nano-engineered MSCs relative to those for PTX solution and nanoparticle groups, suggesting that nano-engineered MSCs demonstrate significantly less off-target deposition. In summary, our results demonstrate that nano-engineered MSCs can serve as an efficient carrier for tumor-specific drug delivery and significantly improved anti-cancer efficacy of conventional chemotherapeutic drugs. Mol Cancer Ther; 17(6); 1196-206. ©2018 AACR.
Collapse
Affiliation(s)
- Buddhadev Layek
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Tanmoy Sadhukha
- Albany Molecular Research Inc., Albany, New York.,Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Jayanth Panyam
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Swayam Prabha
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota. .,Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
129
|
Stem cell therapy in Asherman syndrome and thin endometrium: Stem cell- based therapy. Biomed Pharmacother 2018; 102:333-343. [PMID: 29571018 DOI: 10.1016/j.biopha.2018.03.091] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/06/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
The endometrium is one of the essential components of the uterus. The endometrium of human is a complex and dynamic tissue, which undergoes periods of growth and turn over during any menstrual cycle. Stem cells are initially undifferentiated cells that display a wide range of differentiation potential with no distinct morphological features. Stem cell therapy method recently has become a novel procedure for treatment of tissue injury and fibrosis in response to damage. Currently, there is massive interest in stem cells as a novel treatment method for regenerative medicine and more specifically for the regeneration of human endometrium disorder like Asherman syndrome (AS) and thin endometrium. AS also known as intrauterine adhesion (IUA) is a uterine disorder with the aberrant creation of adhesions within the uterus and/or cervix. Patients with IUA are significantly associated with menstrual abnormalities and suffer from pelvic pain. In addition, IUA might prevent implantation of the blastocyst, impair the blood supply to the uterus and early fetus, and finally result in the recurrent miscarriage or infertility in the AS patients. It has been evidenced that the transplantation of different stem cells with a diverse source in the endometrial zone had effects on endometrium such as declined the fibrotic area, an elevated number of glands, stimulated angiogenesis, the enhanced thickness of the endometrium, better formed tissue construction, protected gestation, and improved pregnancy rate. This study presents a summary of the investigations that indicate the key role of stem cell therapy in regeneration and renovation of defective parts.
Collapse
|
130
|
Roser-Page S, Vikulina T, Weiss D, Habib MM, Beck GR, Pacifici R, Lane TF, Weitzmann MN. CTLA-4Ig (abatacept) balances bone anabolic effects of T cells and Wnt-10b with antianabolic effects of osteoblastic sclerostin. Ann N Y Acad Sci 2018; 1415:21-33. [PMID: 29500936 DOI: 10.1111/nyas.13643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/12/2022]
Abstract
Activated lymphocytes promote inflammation and bone destruction in rheumatoid arthritis (RA), making T cells and B cells therapeutic targets. Indeed, pharmacological blockade of CD28 costimulation using CTLA-4Ig (abatacept), approved for amelioration of RA, renders T cells dormant (anergic). CTLA-4Ig also promotes bone accretion in healthy mice; surprisingly, however, this effect is driven exclusively through upregulation of bone formation, rather than anti-inflammatory effects on resorption. In the study presented here, we utilized T cell receptor β gene and Wnt-10b gene knockout mice to investigate the roles of T cells and Wnt-10b in CTLA-4Ig-induced bone anabolism. Ablation of either T cells or Wnt-10b not only abolished CTLA-4Ig-induced bone anabolism but also, paradoxically, suppressed bone formation leading to bone loss. Stalled bone formation was accompanied by bone marrow stromal cell expression of the Wnt pathway inhibitor sclerostin. Our data suggest that an immunoskeletal pivot may promote or suppress bone formation, depending on the net outcome of CTLA-4Ig action directed independently on T cells and osteoblast-linage cells that counter Wnt-10b-induced bone anabolism, by secretion of sclerostin. While CTLA-4Ig action is tipped in favor of bone formation under physiological conditions, pathological immunodeficiency may lead to suppressed bone formation and skeletal damage.
Collapse
Affiliation(s)
| | - Tatyana Vikulina
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Daiana Weiss
- Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mark M Habib
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - George R Beck
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Emory Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Roberto Pacifici
- Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia
| | - Timothy F Lane
- Departments of Obstetrics and Gynecology and Biological Chemistry, and Orthopedic Hospital Research Center, University of California Los Angeles, Los Angeles, California
| | - M Neale Weitzmann
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Emory Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
131
|
Repair of maxillary cystic bone defects with mesenchymal stem cells seeded on a cross-linked serum scaffold. J Craniomaxillofac Surg 2018; 46:222-229. [DOI: 10.1016/j.jcms.2017.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/20/2017] [Accepted: 11/02/2017] [Indexed: 12/22/2022] Open
|
132
|
Feng Z, Zhai Y, Zheng Z, Yang L, Luo X, Dong X, Han Q, Jin J, Chen ZN, Zhu P. Loss of A20 in BM-MSCs regulates the Th17/Treg balance in Rheumatoid Arthritis. Sci Rep 2018; 8:427. [PMID: 29323140 PMCID: PMC5765124 DOI: 10.1038/s41598-017-18693-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multi-potent cells that are self-renewable and possess the potential to differentiate into multiple lineages. Several studies demonstrated that MSCs could regulate a Th17/Treg balance and could be a potential therapeutic target for Rheumatoid Arthritis (RA). A20 is highly expressed in many cell types after the stimulation of TNF-α, where it may inhibit pro-inflammatory cytokine secretion. However, the expression of A20 in BM-MSCs in RA is not fully understood. In our study, we found that A20 was decreased in RA patients’ bone marrow MSCs (BM-MSCs), and with more IL-6 secretion, the balance of Th17/Treg was broken. In CIA mice, we found a moderate A20 decrease in mice MSCs as compared with those of control group in mRNA and protein levels. However, the IL-6 expression was increased. After umbilical cord MSCs treatment, A20 and IL-6 expressions were equal to the control group. Thus, our study indicates that loss of A20 in MSCs regulates the Th17/Treg balance in RA and the regulatory role of A20 in pro-inflammatory IL-6 production could be a potential target for the transfer of MSCs in RA adoptive therapy.
Collapse
Affiliation(s)
- Zhuan Feng
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,Department of Cell Biology, Fourth Military Medical University, Xi'an, China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Yue Zhai
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,Department of Cell Biology, Fourth Military Medical University, Xi'an, China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Zhaohui Zheng
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Lijie Yang
- Department of hematology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China
| | - Xing Luo
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Xiwen Dong
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,Department of Cell Biology, Fourth Military Medical University, Xi'an, China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Qing Han
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Jin Jin
- Department of Cell Biology, Fourth Military Medical University, Xi'an, China.,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China
| | - Zhi-Nan Chen
- Department of Cell Biology, Fourth Military Medical University, Xi'an, China. .,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China.
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, No. 127 West Changle Road, Xi'an, Shaanxi Province, People's Republic of China. .,National Translational Science Center for Molecular Medicine, Xi'an, 710032, China.
| |
Collapse
|
133
|
Krishnan R, Ko D, Foster CE, Liu W, Smink AM, de Haan B, De Vos P, Lakey JRT. Immunological Challenges Facing Translation of Alginate Encapsulated Porcine Islet Xenotransplantation to Human Clinical Trials. Methods Mol Biol 2017; 1479:305-333. [PMID: 27738946 DOI: 10.1007/978-1-4939-6364-5_24] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transplantation of alginate-encapsulated islets has the potential to treat patients suffering from type I diabetes, a condition characterized by an autoimmune attack against insulin-secreting beta cells. However, there are multiple immunological challenges associated with this procedure, all of which must be adequately addressed prior to translation from trials in small animal and nonhuman primate models to human clinical trials. Principal threats to graft viability include immune-mediated destruction triggered by immunogenic alginate impurities, unfavorable polymer composition and surface characteristics, and release of membrane-permeable antigens, as well as damage associated molecular patterns (DAMPs) by the encapsulated islets themselves. The lack of standardization of significant parameters of bioencapsulation device design and manufacture (i.e., purification protocols, surface-modification grafting techniques, alginate composition modifications) between labs is yet another obstacle that must be overcome before a clinically effective and applicable protocol for encapsulating islets can be implemented. Nonetheless, substantial progress is being made, as is evident from prolonged graft survival times and improved protection from immune-mediated graft destruction reported by various research groups, but also with regard to discoveries of specific pathways involved in explaining observed outcomes. Progress in the latter is essential for a comprehensive understanding of the mechanisms responsible for the varying levels of immunogenicity of certain alginate devices. Successful translation of encapsulated islet transplantation from in vitro and animal model testing to human clinical trials hinges on application of this knowledge of the pathways and interactions which comprise immune-mediated rejection. Thus, this review not only focuses on the different factors contributing to provocation of the immune reaction by encapsulated islets, but also on the defining characteristics of the response itself.
Collapse
Affiliation(s)
- Rahul Krishnan
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA
| | - David Ko
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA
| | - Clarence E Foster
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA.,Department of Transplantation, University of California Irvine, Orange, CA, USA
| | - Wendy Liu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - A M Smink
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Bart de Haan
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Paul De Vos
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Jonathan R T Lakey
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA. .,Department of Transplantation, University of California Irvine, Orange, CA, USA. .,Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
134
|
Kay AG, Long G, Tyler G, Stefan A, Broadfoot SJ, Piccinini AM, Middleton J, Kehoe O. Mesenchymal Stem Cell-Conditioned Medium Reduces Disease Severity and Immune Responses in Inflammatory Arthritis. Sci Rep 2017; 7:18019. [PMID: 29269885 PMCID: PMC5740178 DOI: 10.1038/s41598-017-18144-w] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/05/2017] [Indexed: 12/29/2022] Open
Abstract
We evaluated the therapeutic potential of mesenchymal stem cell-conditioned medium (CM-MSC) as an alternative to cell therapy in an antigen-induced model of arthritis (AIA). Disease severity and cartilage loss were evaluated by histopathological analysis of arthritic knee joints and immunostaining of aggrecan neoepitopes. Cell proliferation was assessed for activated and naïve CD4+ T cells from healthy mice following culture with CM-MSC or co-culture with MSCs. T cell polarization was analysed in CD4+ T cells isolated from spleens and lymph nodes of arthritic mice treated with CM-MSC or MSCs. CM-MSC treatment significantly reduced knee-joint swelling, histopathological signs of AIA, cartilage loss and suppressed TNFα induction. Proliferation of CD4+ cells from spleens of healthy mice was not affected by CM-MSC but reduced when cells were co-cultured with MSCs. In the presence of CM-MSC or MSCs, increases in IL-10 concentration were observed in culture medium. Finally, CD4+ T cells from arthritic mice treated with CM-MSC showed increases in FOXP3 and IL-4 expression and positively affected the Treg:Th17 balance in the tissue. CM-MSC treatment reduces cartilage damage and suppresses immune responses by reducing aggrecan cleavage, enhancing Treg function and adjusting the Treg:Th17 ratio. CM-MSC may provide an effective cell-free therapy for inflammatory arthritis.
Collapse
Affiliation(s)
- Alasdair G Kay
- Biology Department, University of York, Wentworth Way, York, UK.,ISTM at RJAH Orthopaedic Hospital, Keele University, Oswestry, UK
| | - Grace Long
- School of Medicine, Keele University, Staffordshire, UK
| | - George Tyler
- School of Medicine, Keele University, Staffordshire, UK
| | - Andrei Stefan
- ISTM at RJAH Orthopaedic Hospital, Keele University, Oswestry, UK
| | | | | | - Jim Middleton
- Faculty of Health Sciences, School of Oral and Dental Science, University of Bristol, Bristol, UK
| | - Oksana Kehoe
- ISTM at RJAH Orthopaedic Hospital, Keele University, Oswestry, UK.
| |
Collapse
|
135
|
The future of mesenchymal stem cell-based therapeutic approaches for cancer - From cells to ghosts. Cancer Lett 2017; 414:239-249. [PMID: 29175461 DOI: 10.1016/j.canlet.2017.11.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/19/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells which can differentiate into a variety of cell types including osteoblasts, adipocytes and chondrocytes. They are normally resident in adipose tissue, bone marrow and the umbilical cord, but can also be found in other tissues and are known to be recruited to sites of wound healing as well as growing tumours. The therapeutic potential of MSCs has been explored in a number of phase I/II and III clinical trials, of which several were targeted against graft-versus-host disease and to support engraftment of haematopoietic stem cells (HSCs), but currently only very few in the oncology field. There are now three clinical trials either ongoing or recruiting patients that use MSCs to treat tumour disease. In these, MSCs target gastrointestinal, lung and ovarian cancer, respectively. The first study uses MSCs loaded with a HSV-TK expression construct under the control of the CCL5 promoter, and has recently reported successful completion of Phase I/II. While no adverse side effects were seen during this study, no outcomes with respect to therapeutic benefits have been published. The other clinical trials targeting lung and ovarian cancer will be using MSCs expressing cytokines as therapeutic payload. Despite these encouraging early steps towards their clinical use, many questions are still unanswered regarding the biology of MSCs in normal and pathophysiological settings. In this review, in addition to summarising the current state of MSC-based therapeutic approaches for cancer, we will describe the remaining questions, obstacles and risks, as well as novel developments such as MSC-derived nanoghosts.
Collapse
|
136
|
Guo H, Zhao N, Gao H, He X. Mesenchymal Stem Cells Overexpressing Interleukin-35 Propagate Immunosuppressive Effects in Mice. Scand J Immunol 2017; 86:389-395. [PMID: 28888053 DOI: 10.1111/sji.12613] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022]
Abstract
To explore generation of interleukin (IL)-35-expressing mouse adipocyte-derived mesenchymal stem cells (Ad-MSCs) using lentiviral vector and their potential immunosuppressive effects in mice. Ad-MSCs were isolated and cultured in vitro and transfected with a lentivirus vector for overexpression of the therapeutic murine IL-35 gene. IL-35 expression in transfected MSCs (IL-35-MSCs) was quantified by enzyme-linked immunosorbent assay (ELISA). The lymphocytes subsets after one-way mixed lymphocyte culture and in vivo intravenous transplantation were analysed by flow cytometry to evaluate the immunosuppressive effects of IL-35-MSCs. ELISA was performed to examine IL-10, IL-17A and IL-35 expression in lymphocyte culture. Mouse Ad-MSCs were isolated and cultured. IL-35 was expressed in the MSC supernatant and serum after IL-35 transduction into Ad-MSCs by lentiviral vector transfection in vitro and in vivo. The percentage of CD4+ CD25+ T regulatory (Treg) cells in mice treated with IL-35-MSCs significantly increased. IL-35-MSCs upregulated the CD4+ CD25+ Treg cells in the allogeneic mixed lymphocyte reaction system, and lowered the percentage of CD4+ T cells compared with the other two control groups (P < 0.01). IL-17A expression significantly decreased and IL-10 expression significantly increased in IL-35-MSCs and MSCs when compared by ELISA to the control groups (P < 0.01). IL-35-transduced Ad-MSCs in vivo can enhance proliferation of CD4+ CD25+ Treg cells and suppress the function of effector T cells such as T helper (Th) 1, Th2 and Th17 cells and may reduce the development of allograft rejection. Our data suggest that transduced Ad-MSCs overexpressing IL-35 may provide a useful approach for basic research on cell-based immunotolerance therapy for inducing transplantation tolerance.
Collapse
Affiliation(s)
- H Guo
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - N Zhao
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - H Gao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - X He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
137
|
The Immunomodulatory Effects of Mesenchymal Stem Cell Polarization within the Tumor Microenvironment Niche. Stem Cells Int 2017; 2017:4015039. [PMID: 29181035 PMCID: PMC5664329 DOI: 10.1155/2017/4015039] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent a promising tool for cell therapy, particularly for their antitumor effects. This cell population can be isolated from multiple tissue sources and also display an innate ability to home to areas of inflammation, such as tumors. Upon entry into the tumor microenvironment niche, MSCs promote or inhibit tumor progression by various mechanisms, largely through the release of soluble factors. These factors can be immunomodulatory by activating or inhibiting both the adaptive and innate immune responses. The mechanisms by which MSCs modulate the immune response are not well understood. Because of this, the relationship between MSCs and immune cells within the tumor microenvironment niche continues to be an active area of research in order to help explain the apparent contradictory findings currently available in the literature. The ongoing research aims to enhance the potential of MSCs in future therapeutic applications.
Collapse
|
138
|
Liu J, Ren J, Su L, Cheng S, Zhou J, Ye X, Dong Y, Sun S, Qi F, Liu Z, Pleat J, Zhai H, Zhu N. Human adipose tissue-derived stem cells inhibit the activity of keloid fibroblasts and fibrosis in a keloid model by paracrine signaling. Burns 2017; 44:370-385. [PMID: 29029852 DOI: 10.1016/j.burns.2017.08.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Human adipose tissue-derived mesenchymal stem cells (ASCs) have potential utility as modulators of the regeneration of tissue that is inflamed or scarred secondary to injuries such as burns or trauma. However, the effect of ASCs on one particular type of scarring, keloidal disease, remains unknown. The absence of an optimal model for investigation has hindered the development of an effective therapy using ASCs for keloids. OBJECTIVE To investigate the influence of ASCs on angiogenesis, extracellular matrix deposition, and inflammatory cell influx in keloids. METHODS We analyzed the proliferation, migration, and apoptosis of human keloid-derived fibroblasts treated with a starvation-induced, conditioned medium from ASCs (ASCs-CM). This was achieved by Brdu proliferation assay, a validated co-culture migration assay, and flow cytometry, respectively. To assess the change in phenotype to a pro-fibrotic state, fibroblasts were analyzed by real-time PCR and contraction assay. A keloid implantation animal model was used to assess the paracrine effect of ASCs histochemically and immunohistochemically on scar morphology, collagen deposition, inflammatory cell composition, and blood vessel density. In tandem, an antibody-based array was used to identify protein concentration in the presence of ASCs-CM at time point 0, 24, and 48h. RESULTS ASCs-CM inhibited the proliferation and collagen synthesis of human keloid-derived fibroblasts. ASCs-CM was associated with reduced inflammation and fibrosis in the keloid implantation model. Thirty-four cytokines were differentially regulated by ASCs-CM at 24h. These included molecules associated with apoptosis, matrix metalloproteases, and their inhibitors. The same molecules were present at relatively higher concentrations at the 48h timepoint. CONCLUSION These results suggest that ASCs are associated with the inhibition of fibrosis in keloids by a paracrine effect. This phenomenon may have utility as a therapeutic approach in the clinical environment.
Collapse
Affiliation(s)
- Jianlan Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Ren
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lina Su
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shimeng Cheng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Zhou
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaolu Ye
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yabin Dong
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Silei Sun
- Department of Emergency Intensive Care Unit, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fazhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Shanghai, China
| | - Zhifei Liu
- Plastic Surgery Department, Peking Union Medical Hospital, Chinese Academy of Medical Sciences, Beijing 100032, China
| | - Jonathon Pleat
- Department of Plastic, Reconstructive and Burns Surgery, Southmead Hospital & University of Bristol, Bristol, UK
| | - Hongjun Zhai
- Anshan Hospital of The First Hospital of China Medical University, China.
| | - Ningwen Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
139
|
Retinoic acid-mediated anti-inflammatory responses in equine immune cells stimulated by LPS and allogeneic mesenchymal stem cells. Res Vet Sci 2017; 114:225-232. [DOI: 10.1016/j.rvsc.2017.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/05/2017] [Indexed: 01/08/2023]
|
140
|
Uder C, Brückner S, Winkler S, Tautenhahn HM, Christ B. Mammalian MSC from selected species: Features and applications. Cytometry A 2017; 93:32-49. [PMID: 28906582 DOI: 10.1002/cyto.a.23239] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy of different diseases in humans and in animals. Following the guidelines of the International Society for Cell Therapy, human MSC may be identified by expression of a specific panel of cell surface markers (CD105+, CD73+, CD90+, CD34-, CD14-, or CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion for MSC definition. Human MSC and MSC of a variety of mammals isolated from different tissues meet these criteria. In addition to the abovementioned, they express many more cell surface markers. Yet, these are not uniquely expressed by MSC. The gross phenotypic appearance like marker expression and differentiation potential is similar albeit not identical for MSC from different tissues and species. Similarly, MSC may feature different biological characteristics depending on the tissue source and the isolation and culture procedures. Their versatile biological qualities comprising immunomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion and secrete factors to promote self-repair of the injured tissue. This is a big perspective for clinical MSC applications in both veterinary and human medicine. Phase I/II clinical trials have been initiated to assess safety and feasibility of MSC therapies in acute and chronic disease settings. Yet, since the mode of MSC action in a specific disease environment is still unknown at large, it is mandatory to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models prior to clinical applications. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Christiane Uder
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Hans-Michael Tautenhahn
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | | |
Collapse
|
141
|
Andreeva E, Bobyleva P, Gornostaeva A, Buravkova L. Interaction of multipotent mesenchymal stromal and immune cells: Bidirectional effects. Cytotherapy 2017; 19:1152-1166. [PMID: 28823421 DOI: 10.1016/j.jcyt.2017.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 05/24/2017] [Accepted: 07/02/2017] [Indexed: 12/11/2022]
Abstract
Adult multipotent mesenchymal stromal cells (MSCs) are considered one of the key players in physiological remodeling and tissue reparation. Elucidation of MSC functions is one of the most intriguing issues in modern cell physiology. In the present review, the interaction of MSCs and immune cells is discussed in terms of reciprocal effects, which modifies the properties of "partner" cells with special focus on the contribution of direct cell-to-cell contacts, soluble mediators and local microenvironmental factors, the most important of which is oxygen tension. The immunosuppressive phenomenon of MSCs is considered as the integral part of the response-to-injury mechanism.
Collapse
Affiliation(s)
- Elena Andreeva
- Institute of Biomedical Problems, the Russian Academy of Sciences, Moscow, Russia
| | - Polina Bobyleva
- Institute of Biomedical Problems, the Russian Academy of Sciences, Moscow, Russia
| | | | - Ludmila Buravkova
- Institute of Biomedical Problems, the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
142
|
Henao Agudelo JS, Braga TT, Amano MT, Cenedeze MA, Cavinato RA, Peixoto-Santos AR, Muscará MN, Teixeira SA, Cruz MC, Castoldi A, Sinigaglia-Coimbra R, Pacheco-Silva A, de Almeida DC, Camara NOS. Mesenchymal Stromal Cell-Derived Microvesicles Regulate an Internal Pro-Inflammatory Program in Activated Macrophages. Front Immunol 2017; 8:881. [PMID: 28824619 PMCID: PMC5535070 DOI: 10.3389/fimmu.2017.00881] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells with abilities to exert immunosuppressive response promoting tissue repair. Studies have shown that MSCs can secrete extracellular vesicles (MVs-MSCs) with similar regulatory functions to the parental cells. Furthermore, strong evidence suggesting that MVs-MSCs can modulate several immune cells (i.e., Th1, Th17, and Foxp3+ T cells). However, their precise effect on macrophages (Mϕs) remains unexplored. We investigated the immunoregulatory effect of MVs-MSCs on activated M1-Mϕs in vitro and in vivo using differentiated bone marrow Mϕs and an acute experimental model of thioglycollate-induced peritonitis, respectively. We observed that MVs-MSCs shared surface molecules with MSCs (CD44, CD105, CD90, CD73) and expressed classical microvesicle markers (Annexin V and CD9). The in vitro treatment with MVs-MSCs exerted a regulatory-like phenotype in M1-Mϕs, which showed higher CD206 level and reduced CCR7 expression. This was associated with decreased levels of inflammatory molecules (IL-1β, IL-6, nitric oxide) and increased immunoregulatory markers (IL-10 and Arginase) in M1-Mϕs. In addition, we detected that MVs-MSCs promoted the downregulation of inflammatory miRNAs (miR-155 and miR-21), as well as, upregulated its predicted target gene SOCS3 in activated M1-Mϕs. In vivo MVs-MSCs treatment reduced the Mϕs infiltrate in the peritoneal cavity inducing a M2-like regulatory phenotype in peritoneal Mϕs (higher arginase activity and reduced expression of CD86, iNOS, IFN-γ, IL-1β, TNF-α, IL-1α, and IL-6 molecules). This in vivo immunomodulatory effect of MVs-MSCs on M1-Mϕs was partially associated with the upregulation of CX3CR1 in F4/80+/Ly6C+/CCR2+ Mϕs subsets. In summary, our findings indicate that MVs-MSCs can modulate an internal program in activated Mϕs establishing an alternative regulatory-like phenotype.
Collapse
Affiliation(s)
- Juan S Henao Agudelo
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil
| | - Tarcio T Braga
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Mariane T Amano
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Marcos A Cenedeze
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil
| | - Regiane A Cavinato
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil
| | - Amandda R Peixoto-Santos
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil
| | - Marcelo N Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Simone A Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Mario C Cruz
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil
| | - Angela Castoldi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | | | - Alvaro Pacheco-Silva
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil.,IEP, Albert Einstein Hospital, Sao Paulo, Brazil
| | - Danilo C de Almeida
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Sao Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil.,Laboratory of Renal Pathophysiology, Department of Medicine, School of Medicine, University of São Paulo, Sao Paulo, Brazil
| |
Collapse
|
143
|
Yin ML, Song HL, Yang Y, Zheng WP, Liu T, Shen ZY. Effect of CXCR3/HO-1 genes modified bone marrow mesenchymal stem cells on small bowel transplant rejection. World J Gastroenterol 2017; 23:4016-4038. [PMID: 28652655 PMCID: PMC5473121 DOI: 10.3748/wjg.v23.i22.4016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/20/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether bone marrow mesenchymal stem cells (BMMSCs) modified with the HO-1 and CXCR3 genes can augment the inhibitory effect of BMMSCs on small bowel transplant rejection. METHODS Lewis rat BMMSCs were cultured in vitro. Third-passage BMMSCs were transduced with the CXCR3/HO-1 genes or the HO-1 gene alone. The rats were divided into six groups and rats in the experimental group were pretreated with BMMSCs 7 d prior to small bowel transplant. Six time points (instant, 1 d, 3 d, 7 d, 10 d, and 14 d) (n = 6) were chosen for each group. Hematoxylin-eosin staining was used to observe pathologic rejection, while immunohistochemistry and Western blot were used to detect protein expression. Flow cytometry was used to detect T lymphocytes and enzyme linked immunosorbent assay was used to detect cytokines. RESULTS The median survival time of BMMSCs from the CXCR3/HO-1 modified group (53 d) was significantly longer than that of the HO-1 modified BMMSCs group (39 d), the BMMSCs group (26 d), and the NS group (control group) (16 d) (P < 0.05). Compared with BMMSCs from the HO-1 modified BMMSCs, BMMSCs, and NS groups, rejection of the small bowel in the CXCR3/HO-1 modified group was significantly reduced, while the weight of transplant recipients was also significantly decreased (P < 0.05). Furthermore, IL-2, IL-6, IL-17, IFN-γ, and TNF-α levels were significantly decreased and the levels of IL-10 and TGF-β were significantly increased (P < 0.05). CONCLUSION BMMSCs modified with the CXCR3 and HO-1 genes can abrogate the rejection of transplanted small bowel more effectively and significantly increase the survival time of rats that receive a small bowel transplant.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Survival
- Cells, Cultured
- Cytokines/blood
- Graft Rejection/enzymology
- Graft Rejection/immunology
- Graft Rejection/pathology
- Graft Rejection/prevention & control
- Graft Survival
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Intestine, Small/enzymology
- Intestine, Small/immunology
- Intestine, Small/pathology
- Intestine, Small/transplantation
- Male
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/enzymology
- Mesenchymal Stem Cells/immunology
- Phenotype
- Rats, Inbred BN
- Rats, Inbred Lew
- Receptors, CXCR3/genetics
- Receptors, CXCR3/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Time Factors
- Transfection
Collapse
|
144
|
Liang C, Jiang E, Yao J, Wang M, Chen S, Zhou Z, Zhai W, Ma Q, Feng S, Han M. Interferon-γ mediates the immunosuppression of bone marrow mesenchymal stem cells on T-lymphocytes in vitro. Hematology 2017; 23:44-49. [PMID: 28581352 DOI: 10.1080/10245332.2017.1333245] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Chen Liang
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Erlie Jiang
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jianfeng Yao
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mei Wang
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shulian Chen
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zheng Zhou
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Weihua Zhai
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qiaoling Ma
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Sizhou Feng
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mingzhe Han
- Transplant Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
145
|
Phermthai T, Thongbopit S, Pokathikorn P, Wichitwiengrat S, Julavijitphong S, Tirawanchai N. Carcinogenicity, efficiency and biosafety analysis in xeno-free human amniotic stem cells for regenerative medical therapies. Cytotherapy 2017; 19:990-1001. [PMID: 28566211 DOI: 10.1016/j.jcyt.2017.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND AIMS Human amniotic mesenchymal stromal cells (hAMSCs) are a potent and attractive stem cell source for use in regenerative medicine. However, the safe uses of therapeutic-grade MSCs are equally as important as the efficiency of MSCs. To provide efficient, clinic-compliant (safe for therapeutic use) MSCs, hAMSC lines that completely eliminate the use of animal products and have been characterized for carcinogenicity and biosafety are required. METHODS Here, we have efficiently generated 10 hAMSC lines under human umbilical cord blood serum (hUCS)-supplemented medium (xeno-free culture) and fetal bovine serum (FBS)-supplemented medium (standard culture) and investigated carcinogenicity and immunosuppressive properties in the resultant hAMSC lines. All hAMSC lines were examined for efficiency (growth kinetics, cryopreservation, telomere length, phenotypic characterization, differentiation potential), carcinogenicity (proto-oncogene and tumor suppressor gene and epigenomic stability) and safety (immunosuppressive properties). RESULTS Stem cell characteristics between the xeno-free hAMSC lines and the cell lines generated using the standard culture system showed no differences. Xeno-free hAMSC lines displayed normal growth proliferation potential, morphological, karyotypic, phenotypic differentiation properties and telomere lengths. Additionally, they retained normal immunosuppressive effects. As a marker of carcinogenicity and biosafety, proto-oncogenes expression levels showed no differences in xeno-free hAMSCs, and we detected no SNP mutations on hotspot codons of the P53 tumor suppressor gene and stable epigenomic imprinting in xeno-free hAMSC lines. CONCLUSIONS Xeno-free hAMSC lines retain essential stem cell characteristics, with a high degree of certainty for meeting biosafety and carcinogenicity standards for a xeno-free system supplemented with allogenic hUCS. The cell lines are suitable and valuable for therapeutic purposes.
Collapse
Affiliation(s)
- Tatsanee Phermthai
- Stem Cell Research and Development Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Sasiprapa Thongbopit
- Stem Cell Research and Development Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Puttachart Pokathikorn
- Stem Cell Research and Development Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suparat Wichitwiengrat
- Stem Cell Research and Development Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suphakde Julavijitphong
- Stem Cell Research and Development Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Infertility Unit, Department of Obstetrics & Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nednapis Tirawanchai
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
146
|
Pang Y, Xiao HW, Zhang H, Liu ZH, Li L, Gao Y, Li HB, Jiang ZJ, Tan H, Lin JR, Du X, Weng JY, Nie DN, Lin DJ, Zhang XZ, Liu QF, Xu DR, Chen HJ, Ge XH, Wang XY, Xiao Y. Allogeneic Bone Marrow-Derived Mesenchymal Stromal Cells Expanded In Vitro for Treatment of Aplastic Anemia: A Multicenter Phase II Trial. Stem Cells Transl Med 2017; 6:1569-1575. [PMID: 28504860 PMCID: PMC5689769 DOI: 10.1002/sctm.16-0227] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 10/20/2016] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
We conducted a phase II, noncomparative, multicenter study to assess the efficacy and safety of allogeneic bone marrow‐derived mesenchymal stromal cells (BM‐MSCs) expanded in vitro for patients with aplastic anemia (AA) refractory to immunosuppressive therapy. Seventy‐four patients from seven centers received allogeneic BM‐MSCs at a dose of 1–2 × 106 cells/kg per week for 4 weeks. Responses were assessed at 0.5, 1, 2, 3, 6, 9, and 12 months after the first cells infusion. Patients with response at 1 month continued to receive four infusions. All patients were evaluable. The overall response rate was 28.4% (95% confidence interval, 19%–40%), with 6.8% complete response and 21.6% partial response. The median times to response of leukocytic, erythrocytic, and megakaryocytic linages were 19 (range, 11–29), 17 (range, 12–25), and 31 (range, 26–84) days, respectively. After median follow‐up of 17 months, overall survival was 87.8%. Seven patients developed transitory and mild headache and fever, but no other adverse events were observed. Antithymocyte globulin used in previous treatment and no activated infection throughout treatment were predictors for response. Allogeneic BM‐MSCs infusion is a feasible and effective treatment option for refractory AA. The trial was registered at www.clinicaltrials.gov as NCT00195624. Stem Cells Translational Medicine2017;6:1569–1575
Collapse
Affiliation(s)
- Yan Pang
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Hao-Wen Xiao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Hang Zhang
- Center of Cell-biological Therapy & Research Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Zeng-Hui Liu
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Li Li
- Center of Cell-biological Therapy & Research Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Yang Gao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Hong-Bo Li
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Zu-Jun Jiang
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| | - Huo Tan
- Department of Hematology, Guangzhou Medical University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Jing-Ren Lin
- Department of Hematology, Guangzhou Medical University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Xin Du
- Department of Haematology, Guangdong General Hospital, Guangzhou, People's Republic of China
| | - Jian-Yu Weng
- Department of Haematology, Guangdong General Hospital, Guangzhou, People's Republic of China
| | - Da-Nian Nie
- Department of Hematology, Sun Yat-Sen University Sun Yat-Sen Memorial Hospital, Guangzhou, People's Republic of China
| | - Dong-Jun Lin
- Department of Hematology, Sun Yat-Sen University Third Affiliated Hospital, Guangzhou, People's Republic of China
| | - Xiang-Zhong Zhang
- Department of Hematology, Sun Yat-Sen University Third Affiliated Hospital, Guangzhou, People's Republic of China
| | - Qi-Fa Liu
- Department of Hematology, Southern Medical University Nanfang Hospital, Guangzhou, People's Republic of China
| | - Duo-Rong Xu
- Department of Hematology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Hai-Jia Chen
- Guangzhou Saliai Stem Cell Science and Technology Co. Ltd, Guangzhou, People's Republic of China
| | - Xiao-Hu Ge
- Guangzhou Saliai Stem Cell Science and Technology Co. Ltd, Guangzhou, People's Republic of China
| | - Xiao-Yan Wang
- Guangzhou Saliai Stem Cell Science and Technology Co. Ltd, Guangzhou, People's Republic of China
| | - Yang Xiao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, People's Republic of China
| |
Collapse
|
147
|
Nwabo Kamdje AH, Kamga PT, Simo RT, Vecchio L, Seke Etet PF, Muller JM, Bassi G, Lukong E, Goel RK, Amvene JM, Krampera M. Mesenchymal stromal cells' role in tumor microenvironment: involvement of signaling pathways. Cancer Biol Med 2017; 14:129-141. [PMID: 28607804 PMCID: PMC5444925 DOI: 10.20892/j.issn.2095-3941.2016.0033] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are adult multipotent stem cells residing as pericytes in various tissues and organs where they can differentiate into specialized cells to replace dying cells and damaged tissues. These cells are commonly found at injury sites and in tumors that are known to behave like " wounds that do not heal." In this article, we discuss the mechanisms of MSCs in migrating, homing, and repairing injured tissues. We also review a number of reports showing that tumor microenvironment triggers plasticity mechanisms in MSCs to induce malignant neoplastic tissue formation, maintenance, and chemoresistance, as well as tumor growth. The antitumor properties and therapeutic potential of MSCs are also discussed.
Collapse
Affiliation(s)
| | - Paul Takam Kamga
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | | | - Jean Marc Muller
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Giulio Bassi
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Erique Lukong
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Raghuveera Kumar Goel
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Jeremie Mbo Amvene
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Mauro Krampera
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| |
Collapse
|
148
|
Mohamed SA, Shalaby SM, Abdelaziz M, Brakta S, Hill WD, Ismail N, Al-Hendy A. Human Mesenchymal Stem Cells Partially Reverse Infertility in Chemotherapy-Induced Ovarian Failure. Reprod Sci 2017; 25:51-63. [PMID: 28460567 DOI: 10.1177/1933719117699705] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Chemotherapy is the most commonly used modality to treat human cancers; however, in many cases it causes irreversible ovarian failure. In this work, we plan to evaluate the restorative function of human bone marrow mesenchymal stem cells (BMSCs) in a chemotherapy-induced ovarian failure mouse model. METHODS Acclimatized 4 to 6 week-old female mice (C57BL/6) were assigned randomly to a vehicle-treated control group (group 1), chemotherapy-treated group followed by vehicle alone (group 2), or chemotherapy-treated group followed by stem cell intraovarian injection (group 3). Outcomes were evaluated using immunohistochemistry (IHC), serum hormonal assays, and estrous cycle monitoring and breeding potential. RESULTS Post BMSCs administration, group 3 promptly showed detectable vaginal smears with estrogenic changes. Increase in total body weight, ovarian weight, and weight of estrogen-responsive organs (uterus and liver) was observed at 2 weeks and continued to end of the experiment. Hematoxylin and Eosin histological evaluation of the ovaries demonstrated a higher mean follicle count in group 3 than in group 2. Group 3 had lower follicle-stimulating hormone (FSH) levels ( P = .03) and higher anti-Müllerian hormone serum (AMH) levels ( P = .0005) than group 2. The IHC analysis demonstrated higher expression of AMH, FSH receptor, inhibin A, and inhibin B in growing follicles of group 3 versus group 2. Tracking studies demonstrated that human BMSCs evenly repopulated the growing follicles in treated ovaries. Importantly, breeding data showed significant increases in the pregnancies numbers, 2 pregnancies in group 1 and 12 in group 3 ( P = .02). CONCLUSIONS Intraovarian administered BMSCs are able to restore ovarian hormone production and reactivate folliculogenesis in chemotherapy-induced ovarian failure mouse model.
Collapse
Affiliation(s)
- Sara A Mohamed
- 1 Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia Augusta University, Augusta, GA, USA.,2 Department of Obstetrics and Gynecology, Mansoura Faculty of Medicine, Mansoura University Hospital, Mansoura, Egypt
| | - Shahinaz M Shalaby
- 1 Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia Augusta University, Augusta, GA, USA.,3 Department of Pharmacology, Tanta Faculty of Medicine, Tanta, Egypt
| | - Mohamed Abdelaziz
- 1 Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia Augusta University, Augusta, GA, USA.,2 Department of Obstetrics and Gynecology, Mansoura Faculty of Medicine, Mansoura University Hospital, Mansoura, Egypt
| | - Soumia Brakta
- 1 Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia Augusta University, Augusta, GA, USA
| | - William D Hill
- 4 Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nahed Ismail
- 5 Division of Clinical Microbiology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ayman Al-Hendy
- 1 Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia Augusta University, Augusta, GA, USA
| |
Collapse
|
149
|
Li L, Xu G, Duan C. TLR2 affects CD86 expression and inflammatory response in burn injury mice through regulation of p38. Biochem Cell Biol 2017; 95:549-555. [PMID: 28460187 DOI: 10.1139/bcb-2016-0210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to assess the effects of TLR2-p38-CD86 signaling pathways on the inflammatory response in a mouse model of burn injury. Wild-type (TLR2+/+) and mutant-type (TLR2-/-) mice were obtained, and a mouse burn injury model was constructed. Tissue samples were examined with hematoxylin and eosin staining and the transferase mediated nick end labeling (TUNEL) method. Macrophages were treated with TLR2 agonist and p38 inhibitor. The expression levels of TLR2, p38, CD86, IL-1β, and TNF-α were quantified by RT-qPCR, Western blot, and ELISA. When compared with the sham group, the burn group had a significantly higher rate of apoptosis as well as higher expressions of TLR2, p38, CD86, IL-1β, and TNF-α. Inhibiting TLR2 was shown to significantly reduce the expressions of p-p38, CD86, IL-1β, and TNF-α. In the results of in-vitro experiments, TLR2 agonist increased the expression of p-p38, CD86, IL-1β, and TNF-α, whereas a p38 inhibitor was shown to reduce the expression of CD86, IL-1β, and TNF-α. Our results suggest that the TLR2-p38-CD86 signaling pathway plays a vital role in inflammation associated with burn injury.
Collapse
Affiliation(s)
- Li Li
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| | - Gang Xu
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| | - Chenwang Duan
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| |
Collapse
|
150
|
Lee HJ, Kang KS, Kang SY, Kim HS, Park SJ, Lee SY, Kim KD, Lee HC, Park JK, Paik WY, Lee L, Yeon SC. Immunologic properties of differentiated and undifferentiated mesenchymal stem cells derived from umbilical cord blood. J Vet Sci 2017; 17:289-97. [PMID: 26726028 PMCID: PMC5037295 DOI: 10.4142/jvs.2016.17.3.289] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/21/2015] [Accepted: 12/30/2015] [Indexed: 12/30/2022] Open
Abstract
The expression of immunogenic markers after differentiation of umbilical cord blood (UCB)-derived mesenchymal stem cells (MSC) has been poorly investigated and requires extensive in vitro and in vivo testing for clinical application. The expression of human leukocyte antigen (HLA) classes on UCB-derived MSC was tested by Fluorescence-activated cell sorting analysis and immunocytochemical staining. The undifferentiated MSC were moderately positive for HLA-ABC, but almost completely negative for HLA-DR. The MSC differentiated to chondrocytes expressed neither HLA-ABC nor HLA-DR. The proliferation of MSC was not significantly affected by the allogeneic lymphocytes stimulated with concanavalin A. The responder lymphocytes showed no significant decrease in proliferation in the presence of the MSC, but the apoptosis rate of the lymphocytes was increased in the presence of MSC. Taken together, these findings indicate that UCB-derived MSC differentiated to chondrocytes expressed less HLA class I and no class II antigens. The MSC showed an immunomodulatory effect on the proliferation and apoptosis of allogeneic lymphocytes. These data suggest that the differentiated and undifferentiated allogeneic MSC derived from umbilical cord blood can be a useful candidate for allogeneic cell therapy and transplantation without a major risk of rejection.
Collapse
Affiliation(s)
- Hyo-Jong Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.,Human Biotech Co. Ltd., Jinju 52839, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Sun-Young Kang
- Gyeongnam Wildlife Center, Gyeongsang National University, Jinju 52828, Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Se-Jin Park
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Seung-Yong Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Kwang-Dong Kim
- Division of Applied Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Hee-Chun Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Ji-Kwon Park
- School of Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Won-Young Paik
- School of Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Lyon Lee
- College of Veterinary Medicine, Western University, Pomona, CA 91766-1854, USA
| | - Seong-Chan Yeon
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.,Gyeongnam Wildlife Center, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|