1
|
Wen S, Dooner M, Pereira M, Del Tatto M, Quesenberry P. Mesenchymal Stem Cell-Derived Extracellular Vesicles Improve Survival and Enhance Hematopoietic Recovery in Mice Exposed to High-Dose Irradiation. Stem Cells Dev 2025; 34:189-200. [PMID: 40135580 DOI: 10.1089/scd.2025.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
Exposure to high-dose radiation often results in hematopoietic acute radiation syndromes, leading to early mortality, while current therapies for patients exposed to lethal radiation doses are limited. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown promise in tissue repair and regeneration but have not been well investigated for mitigating high-dose radiation damage. We previously demonstrated that human or murine MSC-EVs can reverse bone marrow injury caused by mild or moderate radiation. The current study evaluated the therapeutic potential of human MSC-EVs in mice exposed to high-dose total body irradiation (TBI). Mice were exposed to 0, 700, or 950 cGy TBI and subsequently received daily intravenous MSC-EV injections (1 × 109 particles) for 3 days postirradiation. We evaluated survival rates, peripheral blood recovery, bone marrow engraftment, and bone marrow gene expression profiles at various intervals following treatment. MSC-EV administration significantly enhanced survival, with 70% of treated mice surviving 120 days after 950 cGy TBI exposure, compared with 0% survival in untreated controls by day 30. Although early peripheral blood recovery was not observed within 14 days, MSC-EV treatment facilitated substantial recovery at 3 months postirradiation, with significant increases in red blood cell, platelet, white blood cell, and hemoglobin levels, despite white blood cell and hemoglobin levels remaining slightly below normal. Furthermore, the engraftment capacity of bone marrow stem cells was significantly improved. The changes in hematopoietic-related gene expression presented at 14 days postirradiation returned to normal levels by 120 days in MSC-EV-treated mice. These results highlight the potential of MSC-EVs as a therapeutic strategy for high-dose radiation injuries by promoting hematopoietic recovery and improving survival. Our future research will focus on elucidating the radioprotective mechanisms and investigating their integration with existing therapies.
Collapse
Affiliation(s)
- Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mark Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mandy Pereira
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Michael Del Tatto
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Peter Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
2
|
Wei X, Sui K, Peng Y, Li S, Fang Y, Chen Z, Du X, Xie X, Tang H, Wen Q, Li J, He M, Cheng Q, Zhang W. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Loaded Mir-29-3p Targets AhR to Improve Juvenile Idiopathic Arthritis via Inhibiting the Expression of IL-22 in CD4 + T Cell. Stem Cell Rev Rep 2025; 21:536-553. [PMID: 39621151 DOI: 10.1007/s12015-024-10827-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is one of the most common chronic inflammatory rheumatic diseases in children. Human umbilical cord mesenchymal stem cells (HUCMSCs)-derived exosomes (HUCMSCs-Exos) are involved in autoimmune diseases. This study investigates the mechanism of HUCMSC-Exos in improving JIA by targeting AhR through delivery of miR-29-3p to inhibit IL-22 expression in CD4+ T cells. METHODS Collagen induced arthritis (CIA) mouse model was established, and mice were treated with HUCMSCs-Exos and miR-29-3p antagomir, respectively. CD4+ T cells from JIA patients were used for cell experiments. The mechanism was elucidated by histopathological staining, transmission electron microscopy (TEM), immunohistochemistry, CCK-8 assay, flow cytometry, Western blotting, real-time PCR, and enzyme-linked immunosorbent assay (ELISA), laser confocal microscopy, and luciferase assay. RESULT JIA-CD4+ T cells showed higher expression of IL-22 and lower the levels of miR-29-3p, while HUCMSCs-Exos significantly inhibited the expression of IL-22 and increased the levels of miR-29a-3p, miR-29b-3p, and miR-29c-3p in CD4+ T cells from JIA patients. The expression of miR-29a-3p, miR-29b-3p, miR-29c-3p, AhR, and IL-22 in CD4+ T cells was significantly reversed when co-cultured with HUCMSCs transfected with miR-29-3p mimic or miR-29-3p inhibitor. In vivo experiment, HUCMSCs-Exos ameliorated CIA mice by delivering miR-29-3p to inhibit AhR, IL-22, IL-22R1, MMP3, and MMP13 expression. Furthermore, HUCMSCs-Exos also deliver miR-29-3p targeting AhR expression to inhibit IL-22 in JIA-CD4 + T cells through alleviating arthritic synovial fibroblast activation. CONCLUSION HUCMSCs-Exos loaded miR-29-3p targets AhR to improve JIA via inhibiting the expression of IL-22 in CD4+ T cell, which provides a scientific basis for the treatment of JIA.
Collapse
Affiliation(s)
- Xinyi Wei
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Kunpeng Sui
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuanyuan Peng
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Sha Li
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yu Fang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Zhi Chen
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xiao Du
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xue Xie
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Haiming Tang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - QiuYue Wen
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - JingWei Li
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Meilin He
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qin Cheng
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Wei Zhang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chief Physician, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No.1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Synovial fluid mesenchymal progenitor cells from patients with juvenile idiopathic arthritis demonstrate limited self-renewal and chondrogenesis. Sci Rep 2022; 12:16530. [PMID: 36192450 PMCID: PMC9530167 DOI: 10.1038/s41598-022-20880-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is a heterogeneous group of inflammatory diseases affecting joints with a prevalence of one in a thousand children. There is a growing body of literature examining the use of mesenchymal stem/progenitor cells (MPCs) for the treatment of adult and childhood arthritis, however, we still lack a clear understanding of how these MPC populations are impacted by arthritic disease states and how this could influence treatment efficacy. In the current study we examined the immunophenotyping, self-renewal ability and chondrogenic capacity (in vitro and in vivo) of synovial derived MPCs from normal, JIA and RA joints. Synovial MPCs from JIA patients demonstrated reduced self-renewal ability and chondrogenic differentiation capacity. Furthermore, they did not induce cartilage regeneration when xenotransplanted in a mouse cartilage injury model. Synovial MPCs from JIA patients are functionally compromised compared to MPCs from normal and/or RA joints. The molecular mechanisms behind this loss of function remain elusive. Further study is required to see if these cells can be re-functionalized and used in cell therapy strategies for these JIA patients, or if allogenic approaches should be considered.
Collapse
|
4
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
5
|
Wong SC, Medrano LC, Hoftman AD, Jones OY, McCurdy DK. Uncharted waters: mesenchymal stem cell treatment for pediatric refractory rheumatic diseases; a single center case series. Pediatr Rheumatol Online J 2021; 19:87. [PMID: 34112214 PMCID: PMC8194100 DOI: 10.1186/s12969-021-00575-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND With the advent of innovative therapies including biologics and Janus kinase inhibitors, children with rheumatic diseases are more likely to have improved outcomes. Despite these advances, some children do not respond, or they, or their parents fear adverse events and seek other alternatives. Increasingly, private companies are offering mesenchymal stem cells (MSC) as an alternative, which are described as natural therapies for rheumatic diseases, often insinuating them as a cure. MSC have immunomodulatory properties, and transplantation of these stem cells have been used to successfully treat immunologic conditions like graft-versus-host disease. Lately, MSC research in adult lupus has been encouraging, but the clinical trials are still underway and in most, MSC therapy is not a standalone treatment. This retrospective case series will highlight three cases of pediatric refractory autoimmune disease whose parents sought out and received MSC therapy as a self-decision without first seeking medical advice from our specialty. The three families felt that their children were improved and in two believed that their child was cured. MSC have the potential of beneficial immunomodulation and may be a powerful tool in the therapy of rheumatic disease, but well controlled clinical trials are necessary and should be designed and monitored by experts in childhood rheumatic disease. CASE PRESENTATION Three children with three different rheumatic diseases; systemic lupus erythematosus, mixed connective tissue disease and juvenile idiopathic arthritis were under the care of pediatric rheumatology at a large, tertiary-care, teaching institution. Multiple non-biologic and biologic disease-modifying anti-rheumatic drugs failed to significantly decrease disease activity, and as a result, the families chose to undergo MSC therapy. After transplantation, all children improved per patient and parent report and tapered off conventional immunosuppressive drugs. No serious adverse events occurred in these three patients. CONCLUSION The three cases presented in this report reflect comparable beneficial outcomes and minimal risks published in adult studies. These were not controlled studies, however, and benefit was reported rather than documented. These cases suggest that MSC transplantation may prove a promising adjunctive treatment option; however, further research, development of standardized infusion therapy protocols, and well-designed monitored clinical trials are essential.
Collapse
Affiliation(s)
- Stephen C. Wong
- grid.19006.3e0000 0000 9632 6718Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90095 USA ,grid.34477.330000000122986657Division of Rheumatology, Department of Pediatrics, University of Washington/Seattle Children’s Hospital, Seattle, 98105 USA
| | - Leah C. Medrano
- grid.19006.3e0000 0000 9632 6718Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Alice D. Hoftman
- grid.19006.3e0000 0000 9632 6718Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Olcay Y. Jones
- grid.414467.40000 0001 0560 6544Division Pediatric Rheumatology, Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD 20889 USA
| | - Deborah K. McCurdy
- grid.19006.3e0000 0000 9632 6718Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
6
|
Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trial. Signal Transduct Target Ther 2020; 5:172. [PMID: 32855385 PMCID: PMC7450163 DOI: 10.1038/s41392-020-00286-5] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
No effective drug treatments are available for coronavirus disease 2019 (COVID-19). Host-directed therapies targeting the underlying aberrant immune responses leading to pulmonary tissue damage, death, or long-term functional disability in survivors require clinical evaluation. We performed a parallel assigned controlled, non-randomized, phase 1 clinical trial to evaluate the safety of human umbilical cord-derived mesenchymal stem cells (UC-MSCs) infusions in the treatment of patients with moderate and severe COVID-19 pulmonary disease. The study enrolled 18 hospitalized patients with COVID-19 (n = 9 for each group). The treatment group received three cycles of intravenous infusion of UC-MSCs (3 × 107 cells per infusion) on days 0, 3, and 6. Both groups received standard COVID-treatment regimens. Adverse events, duration of clinical symptoms, laboratory parameters, length of hospitalization, serial chest computed tomography (CT) images, the PaO2/FiO2 ratio, dynamics of cytokines, and IgG and IgM anti-SARS-CoV-2 antibodies were analyzed. No serious UC-MSCs infusion-associated adverse events were observed. Two patients receiving UC-MSCs developed transient facial flushing and fever, and one patient developed transient hypoxia at 12 h post UC-MSCs transfusion. Mechanical ventilation was required in one patient in the treatment group compared with four in the control group. All patients recovered and were discharged. Our data show that intravenous UC-MSCs infusion in patients with moderate and severe COVID-19 is safe and well tolerated. Phase 2/3 randomized, controlled, double-blinded trials with long-term follow-up are needed to evaluate the therapeutic use of UC-MSCs to reduce deaths and improve long-term treatment outcomes in patients with serious COVID-19.
Collapse
|
7
|
Qi T, Gao H, Dang Y, Huang S, Peng M. Cervus and cucumis peptides combined umbilical cord mesenchymal stem cells therapy for rheumatoid arthritis. Medicine (Baltimore) 2020; 99:e21222. [PMID: 32664175 PMCID: PMC7360298 DOI: 10.1097/md.0000000000021222] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cervus and cucumis peptides (Lugua polypeptides, LG) are traditional Chinese medicine, which are active components of polypeptide extracted from Sika deer bone and melon seed, and they contain bone induced polypeptide biological factors. Umbilical cord mesenchymal stem cell, (UC-MSC) have tissue repair multiple effects, anti-inflammatory, and immune regulation function, which become a very promising start in rheumatoid arthritis (RA) treatment. Hence, LG combined UC-MSC can significantly enhance the UC-MSC treatment of rheumatoid arthritis (RA).To explore the clinical curative effect and therapeutic mechanism of LG combined UC-MSC for treating RA.119 patients were divided into control and treatment groups, and both groups were treated with methotrexate tablets, leflunomide, and UC-MSC. But, LG were added to the treatment group. In vitro, the effects of LG on UC-MSC cell secretion of anti-inflammatory factors were also performed.The Health Assessment Questionnaire; the 28 joint disease activity score; C reactive protein; the erythrocyte sedimentation rate; rheumatoid factor; and anti-cyclic citrullinated peptide antibody were significantly reduced in treatment group 1 year after treatment (P < .05). In vitro, compared with the control group, the number of hepatocyte growth factor (HGF), the secretion of prostaglandin E2 (PGE2) and tumor necrosis factor-inducible gene 6 protein (TSG6) increased significantly (P < .05).LG combined UC-MSCs can significantly improve the curative effect of RA patients, while LG may reduce inflammatory cytokines, regulate immunity, improve microcirculation, and are conducive to UC-MSCs migration and the repair of damaged tissue.
Collapse
Affiliation(s)
- Tao Qi
- Department of Radiation Oncology, 986 Hospital of People's Liberation Army Air Force
| | - Hongxiang Gao
- Department of Radiotherapy Oncology, Chang An Hospital, Xi’an, Shaan Xi
| | - Yazheng Dang
- Department of Radiation Oncology, 986 Hospital of People's Liberation Army Air Force
| | - Shigao Huang
- Cancer Centre
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR
| | - Minfei Peng
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province, Taizhou Enze Medical Center (Group), Linhai, Zhejiang Province, China
| |
Collapse
|
8
|
Swart JF, de Roock S, Nievelstein RAJ, Slaper-Cortenbach ICM, Boelens JJ, Wulffraat NM. Bone-marrow derived mesenchymal stromal cells infusion in therapy refractory juvenile idiopathic arthritis patients. Rheumatology (Oxford) 2020; 58:1812-1817. [PMID: 31070229 PMCID: PMC6758577 DOI: 10.1093/rheumatology/kez157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/25/2019] [Indexed: 12/29/2022] Open
Abstract
Objectives To compare the total number of adverse events (AEs) before and after mesenchymal stromal cell (MSC) infusion in refractory JIA and to evaluate its effectiveness. Methods Single-centre Proof of Mechanism Phase Ib, open label intervention study in JIA patients previously failing all biologicals registered for their diagnosis. Six patients received 2 million/kg intravenous infusions of allogeneic bone-marrow derived MSC. In case of ACR-Ped30-response but subsequent loss of response one and maximal two repeated infusions are allowed. Results Six JIA patients with 9.2 years median disease duration, still active arthritis and damage were included. All had failed methotrexate, corticosteroids and median five different biologicals. MSC were administered twice in three patients. No acute infusion reactions were observed and a lower post-treatment than pre-treatment incidence in AEs was found. The one systemic onset JIA (sJIA) patient had again an evolving macrophage activation syndrome, 9 weeks after tocilizumab discontinuation and 7 weeks post-MSC infusion. Statistically significant decreases were found 8 weeks after one MSC infusion in VAS well-being (75–56), the JADAS-71 (24.5–11.0) and the cJADAS10 (18.0–10.6). Conclusion MSC infusions in six refractory JIA patients were safe, although in sJIA stopping the ‘failing’ biologic treatment carries a risk of a MAS flare, as the drug might still suppress the systemic features. Trial registration Trial register.nl, http://https://www.trialregister.nl, NTR4146.
Collapse
Affiliation(s)
- Joost F Swart
- Department of Pediatric Immunology and Rheumatology, UMC Utrecht, Wilhelmina Children's Hospital.,Faculty of Medicine, Utrecht University
| | - Sytze de Roock
- Department of Pediatric Immunology and Rheumatology, UMC Utrecht, Wilhelmina Children's Hospital.,Faculty of Medicine, Utrecht University
| | - Rutger A J Nievelstein
- Faculty of Medicine, Utrecht University.,Department of Pediatric Radiology, Division Imaging, UMC Utrecht, Utrecht, The Netherlands
| | | | - Jaap J Boelens
- Department of Pediatric Immunology and Rheumatology, UMC Utrecht, Wilhelmina Children's Hospital.,Faculty of Medicine, Utrecht University
| | - Nico M Wulffraat
- Department of Pediatric Immunology and Rheumatology, UMC Utrecht, Wilhelmina Children's Hospital.,Faculty of Medicine, Utrecht University
| |
Collapse
|
9
|
Liu L, Farhoodi HP, Han M, Liu G, Yu J, Nguyen L, Nguyen B, Nguyen A, Liao W, Zhao W. Preclinical Evaluation of a Single Intravenous Infusion of hUC-MSC (BX-U001) in Rheumatoid Arthritis. Cell Transplant 2020; 29:963689720965896. [PMID: 33054324 PMCID: PMC7784581 DOI: 10.1177/0963689720965896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease of the joints, which causes severe pain and excessive systemic circulation of harmful inflammatory cytokines. Current treatments are limited, with some patients not responding well, and some experiencing severe and detrimental side effects. Mesenchymal stem cells (MSC) are cell-based therapeutics being evaluated as potent immunomodulators in RA and may provide relief to patients not responding well to drug-based treatments. We evaluated the safety and efficacy of BX-U001 human umbilical cord tissue-derived mesenchymal stem cells (hUC-MSC) to treat RA, in support of a successful investigational new drug application. A collagen-induced arthritis (CIA) mouse model of RA was established in DBA/1 J mice. Mice from the treatment assessment group were given a tail vein infusion of hUC-MSC 24 days after primary RA induction, while control assessment (CA) group mice were given cell-free carrier solution. All animals were evaluated daily for RA symptoms via clinical scoring, blood was taken periodically for cytokine analysis, and mice were dissected at end point for histological analysis. A linear mixed model was used to compare the rate of change among groups. The clinical scores of TA group were significantly reduced compared with CA group (P < 0.01), indicating therapeutic effects. The histological scores of the joints in TA group were significantly lower than those in the CA group (P < 0.05), but had no significant difference compared with Healthy groups (P > 0.05). The concentration of (interleukin) IL-6 in TA group was significantly reduced by 80.0% (P < 0.0001) 2 days after treatment and by 93.4% at the experimental endpoint compared with levels prior to hUC-MSC injection. A single intravenous infusion of hUC-MSC (2 × 106 cells/mouse), to CIA-induced DBA/1 J mice, resulted in significant alleviation of RA symptoms and may provide significant therapeutic benefits in humans.
Collapse
Affiliation(s)
| | - Henry P. Farhoodi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | - Menglu Han
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | - Guangyang Liu
- Department of Surgery, University of California, Irvine, Orange, CA, USA
| | - Jingxia Yu
- Department of Surgery, University of California, Irvine, Orange, CA, USA
| | - Lily Nguyen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | - Brenda Nguyen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | - Agnes Nguyen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
| | | | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA
- Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| |
Collapse
|
10
|
Liu L, Wong CW, Han M, Farhoodi HP, Liu G, Liu Y, Liao W, Zhao W. Meta-analysis of preclinical studies of mesenchymal stromal cells to treat rheumatoid arthritis. EBioMedicine 2019; 47:563-577. [PMID: 31501076 PMCID: PMC6796567 DOI: 10.1016/j.ebiom.2019.08.073] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND This study aims to evaluate the quality of preclinical data, determine the effect sizes, and identify experimental measures that inform efficacy using mesenchymal stromal (or stem) cells (MSC) therapy in animal models of rheumatoid arthritis (RA). METHODS Literature searches were performed on MSC preclinical studies to treat RA. MSC treatment effect sizes were determined by the most commonly used outcome measures, including paw thickness, clinical score, and histological score. FINDINGS A total of 48 studies and 94 treatment arms were included, among which 42 studies and 79 treatment arms reported that MSC improved outcomes. The effect sizes of RA treatments using MSC, when compared to the controls, were: paw thickness was ameliorated by 53.6% (95% confidence interval (CI): 26.7% -80.4%), histological score was decreased by 44.9% (95% CI: 33.3% -56.6%), and clinical score was decreased by 29.9% (95% CI: 16.7% -43.0%). Specifically, our results indicated that human umbilical cord derived MSC led to large improvements of the clinical score (-42.1%) and histological score (-51.4%). INTERPRETATION To the best of our knowledge, this meta-analysis is to quantitatively answer whether MSC represent a robust RA treatment in animal models. It suggests that in preclinical studies, MSC have consistently exhibited therapeutic benefits. The findings demonstrate a need for considering variations in different animal models and treatment protocols in future studies using MSC to treat RA in humans to maximise the therapeutic gains in the era of precision medicine. FUNDS NIH [1DP2CA195763], Baylx Inc.: BI-206512, NINDS/NIH Training Grant [Award# NS082174].
Collapse
Affiliation(s)
- Linan Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Chi W Wong
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Menglu Han
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Henry P Farhoodi
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Guangyang Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Surgery, University of California-Irvine, Irvine, CA 92697, USA
| | - Yongjun Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Wenbin Liao
- Baylx, Inc., 1 Technology Dr, C511, Irvine, CA 92618, USA.
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA; Departments of Biomedical Engineering and Biological Chemistry, University of California-Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
11
|
Wang S, Zhu R, Li H, Li J, Han Q, Zhao RC. Mesenchymal stem cells and immune disorders: from basic science to clinical transition. Front Med 2019; 13:138-151. [PMID: 30062557 DOI: 10.1007/s11684-018-0627-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
As a promising candidate seed cell type in regenerative medicine, mesenchymal stem cells (MSCs) have attracted considerable attention. The unique capacity of MSCs to exert a regulatory effect on immunity in an autologous/allergenic manner makes them an attractive therapeutic cell type for immune disorders. In this review, we discussed the current knowledge of and advances in MSCs, including its basic biological properties, i.e., multilineage differentiation, secretome, and immunomodulation. Specifically, on the basis of our previous work, we proposed three new concepts of MSCs, i.e., "subtotipotent stem cell" hypothesis, MSC system, and "Yin and Yang" balance of MSC regulation, which may bring new insights into our understanding of MSCs. Furthermore, we analyzed data from the Clinical Trials database ( http://clinicaltrials.gov ) on registered clinical trials using MSCs to treat a variety of immune diseases, such as graft-versus-host disease, systemic lupus erythematosus, and multiple sclerosis. In addition, we highlighted MSC clinical trials in China and discussed the challenges and future directions in the field of MSC clinical application.
Collapse
Affiliation(s)
- Shihua Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongjia Zhu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
- Peking Union Medical College Hospital, Beijing, 100005, China.
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
12
|
The use of stem cells in aesthetic dermatology and plastic surgery procedures. A compact review of experimental and clinical applications. Postepy Dermatol Alergol 2017; 34:526-534. [PMID: 29422816 PMCID: PMC5799755 DOI: 10.5114/ada.2017.72456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/04/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of this paper was to collect currently available data related to the use of stem cells in aesthetic dermatology and plastic surgery based on a systemic review of experimental and clinical applications. We found that the use of stem cells is very promising but the current state of art is still not effective. This situation is connected with not fully known mechanisms of cell interactions, possible risks and side effects. We think that there is a big need to create and conduct different studies which could resolve problems of stem cells use for implementation into aesthetic dermatology and plastic surgery.
Collapse
|
13
|
Are We Right to Consider Mesenchymal Stem Cells to Be a New Perspective for Patients with Juvenile Idiopathic Arthritis? Arch Immunol Ther Exp (Warsz) 2017; 66:267-271. [PMID: 29027570 DOI: 10.1007/s00005-017-0493-3] [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: 02/08/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
Abstract
Juvenile idiopathic arthritis (JIA) is the most common cause of chronic arthritis in childhood. Up to 50% of patients are resistant to standard therapy, which includes non-steroid anti-inflammatory drugs, corticosteroids, disease-modifying anti-rheumatic drugs and biologic therapies. Intra-articular injection of mesenchymal stem cells (MSCs) is proposed as a new approach to JIA treatment. MSCs can modulate inflammation via mechanisms of both adaptive and innate immune response. They are able to inhibit T and B cell proliferation, promote regulatory T cells, suppress the maturation of dendritic cells, stimulate macrophage differentiation into M2 phenotype and reduce effectiveness of natural killer cells. They also secrete plethora of soluble factors which influence joint inflammation. Recent clinical studies reviewed in the article provide promising results which may suggest including intra-articular injection of MSCs in therapy of patients with oligoarticular JIA.
Collapse
|
14
|
Abstract
Systemic juvenile idiopathic arthritis (sJIA) and its most significant complication, macrophage activation syndrome (MAS), have traditionally been treated with steroids and non-steroidal anti-inflammatory medications. However, the introduction of biologic medications that inhibit specific cytokines, such interleukins 1 and 6, has changed the treatment paradigm for sJIA patients. In this review, we discuss the therapies currently used in the treatment of sJIA as well as novel targets and approaches under consideration, including mesenchymal stromal cell therapy and JAK inhibitors. We also discuss targeting cytokines that have been implicated in MAS, such as interferon gamma and interleukin 18.
Collapse
Affiliation(s)
- Susan Canny
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Elizabeth Mellins
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Zhao K, Liu Q. The clinical application of mesenchymal stromal cells in hematopoietic stem cell transplantation. J Hematol Oncol 2016; 9:46. [PMID: 27193054 PMCID: PMC4870746 DOI: 10.1186/s13045-016-0276-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent stem cells well known for repairing tissue, supporting hematopoiesis, and modulating immune and inflammation response. These outstanding properties make MSCs as an attractive candidate for cellular therapy in immune-based disorders, especially hematopoietic stem cell transplantation (HSCT). In this review, we outline the progress of MSCs in preventing and treating engraftment failure (EF), graft-versus-host disease (GVHD) following HSCT and critically discuss unsolved issues in clinical applications.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Hematology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Blvd North, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Blvd North, Guangzhou, China.
| |
Collapse
|