1
|
Ye H, Wu X, Shen Y, Zhao L, Zhang H, Yang J, Li F, Zhang F, Zhang K, Chen J, Shui X. Exosomal lncRNA TUG1 derived from BMSC ameliorate collagen-induced arthritis via BLIMP1-mediated Th17/Treg balance. Int Immunopharmacol 2024; 142:113072. [PMID: 39241514 DOI: 10.1016/j.intimp.2024.113072] [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] [Received: 02/27/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Aberrant differentiation of Th17 cells has been identified as a critical factor in the development of rheumatoid arthritis (RA). BLIMP1 plays a key role in regulating plasma cell differentiation, T helper cell differentiation and Treg cell differentiation. Treatment with exosome injection or bone marrow mesenchymal stem cell (BMSC) transplantation reduce joint damage in RA. But the precise regulatory mechanisms remain unclear. METHODS We injected BMSC-derived exosomes into RA mice, and then performed histological analysis on mouse ankle joints. We cultured CD4+ T cells in vitro, then added exosomes with or without si-TUG1 and induced the differentiation of Th17 cells and Treg cells, and then we used flow cytometry to detect the ratio of Th17 cells and Treg cells. Furthermore, we injected exosomes into sh-NC or sh-BLIMP1-treated RA mice, and then performed histological analysis on the ankle joints. RESULT The results of our study demonstrate that exosome treatment decreased the proportion of differentiated Th17 cells, while increasing the proportion of Treg cells. And we observed that the Exo si-TUG1 group had an increased proportion of Th17 cells and a decreased proportion of Treg cells. We observed an increase in BLIMP1 expression in both the peripheral blood of mice and in CD4+ T cells cultured in vitro in the Exo group. Conversely, the Exo si-TUG1 group showed a decrease in BLIMP1 expression. Notably, inhibiting BLIMP1 expression led to the reversal of the therapeutic effects of exosomes. CONCLUSION Our findings suggest that BMSC-derived exosomes promote the expression of BLIMP1 through Lnc TUG1-carrying exosomes, which may modulate the balance between Th17 cells and Treg cells. This mechanism ultimately alleviates damage caused by RA, suggesting that BMSC-derived exosomes enriched in Lnc TUG1 hold promise as a potential therapeutic approach for treating RA.
Collapse
Affiliation(s)
- Hantao Ye
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuanzhang Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yang Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lin Zhao
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haojie Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jianxin Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Feida Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Fengyu Zhang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Kaiying Zhang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jiaoxiang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China.
| | - Xiaolong Shui
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Xu Q, Hou W, Zhao B, Fan P, Wang S, Wang L, Gao J. Mesenchymal stem cells lineage and their role in disease development. Mol Med 2024; 30:207. [PMID: 39523306 PMCID: PMC11552129 DOI: 10.1186/s10020-024-00967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are widely dispersed in vivo and are isolated from several tissues, including bone marrow, heart, body fluids, skin, and perinatal tissues. Bone marrow MSCs have a multidirectional differentiation potential, which can be induced to differentiate the medium in a specific direction or by adding specific regulatory factors. MSCs repair damaged tissues through lineage differentiation, and the ex vivo transplantation of bone marrow MSCs can heal injured sites. MSCs have different propensities for lineage differentiation and pathological evolution for different diseases, which are crucial in disease progression. In this study, we describe various lineage analysis methods to explore lineage ontology in vitro and in vivo, elucidate the impact of MSC lineage differentiation on diseases, advance our understanding of the role of MSC differentiation in physiological and pathological states, and explore new targets and ideas associated with disease diagnosis and treatment.
Collapse
Affiliation(s)
- Qi Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenrun Hou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Baorui Zhao
- Stem cell Translational laboratory, Shanxi Technological Innovation Center for Clinical Diagnosis and Treatment of Immune and Rheumatic Diseases, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Peixin Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Sheng Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lei Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
3
|
Zhao T, Mu Y, Deng H, Liang K, Zhou F, Lin Q, Cao F, Zhou F, Yang Z. Research hotspots and trends of mesenchymal stem cell-derived extracellular vesicles for drug delivery: a bibliometric and visualization analysis from 2013 to 2023. Front Cell Dev Biol 2024; 12:1412363. [PMID: 39539963 PMCID: PMC11557358 DOI: 10.3389/fcell.2024.1412363] [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: 04/04/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Our study aims to provide a comprehensive overview of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in drug delivery research, focusing on the period between 2013 and 2023. Given the increasing global interest in this field, we utilized bibliometric tools to explore publication trends, key contributors, and thematic research clusters. Methods Data was collected from the Web of Science (WoS) database, and an in-depth bibliometric analysis was conducted using VOSviewer. The analysis encompassed bibliographic coupling, co-citation, co-authorship, and co-occurrence trends, offering a structured insight into global research activity. We also employed Citespace to further analyze thematic clusters in this domain. Results Our analysis revealed a total of 1,045 publications related to MSC-EVs in drug delivery over the past decade, showing a steady increase in research output. China led in publication count, H-index, prolific authors, and research funding, while the United States ranked highest in total citations, average citation counts, and H-index performance. Pharmaceutics emerged as the leading journal by publication volume, with the Journal of Controlled Release having the strongest total link strength. Top institutions driving research included Shanghai Jiao Tong University, Zhejiang University, and Harvard University. VOSviewer analysis identified four major research clusters: tissue engineering, cancer, neurological diseases, and targeted delivery. Citespace analysis refined this further into ten thematic areas, including differentiation, tissue regeneration, and drug resistance. Discussion This bibliometric assessment provides a holistic visualization of the research landscape for MSC-EVs in drug delivery, underlining the significant contributions of China and the United States. Our findings underscore the increasing global importance of MSC-EV research and highlight emerging themes that will likely guide future research directions. The insights from this study offer a foundational framework for identifying nascent frontiers in MSC-EV-based drug delivery.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yuhao Mu
- School of Medicine, Nankai University, Tianjin, China
| | - Haobin Deng
- Department of Oncology, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Kaini Liang
- School of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Fanfan Zhou
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Qiyuan Lin
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Fuyang Cao
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Feifei Zhou
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Zhen Yang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
4
|
Ding X, Liu J, Chen X, Zhang X, Fang Y, Huang D. Application of methylation in the diagnosis of ankylosing spondylitis. Clin Rheumatol 2024; 43:3073-3082. [PMID: 39167325 DOI: 10.1007/s10067-024-07113-0] [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] [Received: 06/28/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory autoimmune disease, mainly characterized by perifibrocartilage osteitis of the sacroiliac joints and spinal enthesitis. To date, the exact pathogenesis of AS remains elusive. It is generally believed that AS is a multifactorial disease involving genetics, infection, environment, and immunity. Among them, genetic factors are the primary determinants of disease risk and severity. In recent years, epigenetic mechanisms such as DNA methylation have been extensively surveyed with respect to the pathogenesis of AS. This review summarizes the latest research progress of methylation in AS, from whole-genome sequencing to individual differentially methylated gene. And finally, the role of methylase in AS inflammation, autophagy, and osteogenic differentiation was explored. In summary, the results of this review attempt to explain the role of methylation in the occurrence and development of AS and point out the shortcomings of current methylation research, providing directions for subsequent methylation research in AS.
Collapse
Affiliation(s)
- Xiang Ding
- Anhui University of Traditional Chinese Medicine, Hefei, 230031, Anhui, China
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China.
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Xiaolu Chen
- Anhui University of Traditional Chinese Medicine, Hefei, 230031, Anhui, China
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China
| | - Xianheng Zhang
- Anhui University of Traditional Chinese Medicine, Hefei, 230031, Anhui, China
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China
| | - Yanyan Fang
- Anhui University of Traditional Chinese Medicine, Hefei, 230031, Anhui, China
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China
| | - Dan Huang
- Anhui University of Traditional Chinese Medicine, Hefei, 230031, Anhui, China
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Shushan, Hefei, 230038, Anhui, China
| |
Collapse
|
5
|
Li C, Sun Y, Xu W, Chang F, Wang Y, Ding J. Mesenchymal Stem Cells-Involved Strategies for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305116. [PMID: 38477559 PMCID: PMC11200100 DOI: 10.1002/advs.202305116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/13/2023] [Indexed: 03/14/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the joints and bone destruction. Because of systemic administration and poor targeting, traditional anti-rheumatic drugs have unsatisfactory treatment efficacy and strong side effects, including myelosuppression, liver or kidney function damage, and malignant tumors. Consequently, mesenchymal stem cells (MSCs)-involved therapy is proposed for RA therapy as a benefit of their immunosuppressive and tissue-repairing effects. This review summarizes the progress of MSCs-involved RA therapy through suppressing inflammation and promoting tissue regeneration and predicts their potential clinical application.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yifu Sun
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Fei Chang
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Yinan Wang
- Department of BiobankDivision of Clinical ResearchThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of EducationThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
6
|
Guo Y, Tian T, Yang S, Cai Y. Ginsenoside Rg1/ADSCs supplemented with hyaluronic acid as the matrix improves rabbit temporomandibular joint osteoarthrosis. Biotechnol Genet Eng Rev 2024; 40:253-274. [PMID: 36892223 DOI: 10.1080/02648725.2023.2183575] [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] [Received: 01/07/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
OBJECTIVE To investigate whether and how ginsenoside Rg1/ADSCs supplemented with hyaluronic acid as the matrix can improve rabbit temporomandibular joint osteoarthrosis. METHOD Isolate and culture adipose stem cells, measure the activity of differentiated chondrocytes by MTT assay and expression of type II collagen in these cells by immunohistochemistry, in order to evaluate the effect of ginsenoside Rg1 on adipose stem cell proliferation and differentiation into chondrocytes.32 New Zealand white rabbits were randomly divided into four groups: blank group, model group, control group and experimental group, 8 in each group. Osteoarthritis model was established by intra-articular injection of papain. Two weeks after successful model building, medication was given for the rabbits in control group and experimental group. 0.6 mL ginsenoside Rg1/ ADSCs suspension was injected into superior joint space for the rabbits in control group, once a week; 0.6 mL ginsenoside Rg1/ ADSCs complex was injected for the rabbits in experimental group, once a week. RESULTS Ginsenoside Rg1 can promote ADSCs-derived chondrocytes' activity and expression of type II collagen. Scanning electron microscopy histology images showed cartilage lesions of the experimental group was significantly improved in comparison with control group. CONCLUSION Ginsenoside Rg1 can promote ADSCs differentiate into chondrocytes, and Ginsenoside Rg1/ADSCs supplemented with hyaluronic acid as the matrix can significantly improve rabbit temporomandibular joint osteoarthrosis.
Collapse
Affiliation(s)
- Yanwei Guo
- Department of Oral and Maxillofacial Surgery, Jining Stomatology Hospital, Jining City, Shandong Province, China
| | - Tingyu Tian
- The second Department of Pediatric Stomatology, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Shimao Yang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Yuping Cai
- Department of prosthodontics, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| |
Collapse
|
7
|
Wu Y, Gong Y, Liu Y, Chen F, Chen S, Zhang F, Wang C, Li S, Hu M, Huang R, Guo X, Wang X, Ning Y, Yang L. Comparative Analysis of Differentially Expressed Genes in Chondrocytes from Rats Exposed to Low Selenium and T-2 Toxin. Biol Trace Elem Res 2024; 202:1020-1030. [PMID: 37326932 DOI: 10.1007/s12011-023-03725-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
The aim of this study was to construct rat models of environmental risk factors for Kashin-Beck disease (KBD) with low selenium and T-2 toxin levels and to screen the differentially expressed genes (DEGs) between the rat models exposed to environmental risk factors. The Se-deficient (SD) group and T-2 toxin exposure (T-2) group were constructed. Knee joint samples were stained with hematoxylin-eosin, and cartilage tissue damage was observed. Illumina high-throughput sequencing technology was used to detect the gene expression profiles of the rat models in each group. Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway enrichment analysis were performed and five differential gene expression results were verified by quantitative real-time polymerase chain reaction (qRT‒PCR). A total of 124 DEGs were identified from the SD group, including 56 upregulated genes and 68 downregulated genes. A total of 135 DEGs were identified in the T-2 group, including 68 upregulated genes and 67 downregulated genes. The DEGs were significantly enriched in 4 KEGG pathways in the SD group and 9 KEGG pathways in the T-2 group. The expression levels of Dbp, Pc, Selenow, Rpl30, and Mt2A were consistent with the results of transcriptome sequencing by qRT‒PCR. The results of this study confirmed that there were some differences in DEGs between the SD group and the T-2 group and provided new evidence for further exploration of the etiology and pathogenesis of KBD.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yi Gong
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yanli Liu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Sijie Chen
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feiyu Zhang
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chaowei Wang
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Shujin Li
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Minhan Hu
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Ruitian Huang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xiong Guo
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
- Clinical Research Center for Endemic Disease of Shaanxi Province, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 Xi Wu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Xi Wang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China.
| | - Yujie Ning
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| | - Lei Yang
- School of Nursing, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
8
|
Zhang Y, Jiao Z, Wang S. Bone Marrow Mesenchymal Stem Cells Release miR-378a-5p-Carried Extracellular Vesicles to Alleviate Rheumatoid Arthritis. J Innate Immun 2023; 15:893-910. [PMID: 37926093 PMCID: PMC10715757 DOI: 10.1159/000534830] [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] [Received: 07/20/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
This study investigates whether bone marrow mesenchymal stem cell (BMSC)-derived extracellular vesicles (EVs) can affect rheumatoid arthritis (RA) by delivering microRNA (miR)-378a-5p to regulate the interferon regulatory factor 1/signal transducer and transcription 1 (IRF1/STAT1) axis. We identified RA-associated miRNAs using the GEO microarray dataset GSE121894. We found the most important miRNAs in RA synovial tissues using RT-qPCR. BMSC-derived EVs were ultracentrifuged and cocultured with human synovial microvascular endothelial cells (HSMECs) in vitro. Dual-luciferase and RNA immunoprecipitation studies examined miR-378a-5p's specific binding to IRF1. We also measured angiogenesis, migration, and proliferation using CCK-8, Transwell, and tube formation assays. Collagen-induced arthritis (CIA) mice models were created by inducing arthritis and scoring it. RA synovial tissues had low miR-378a-5p expression, whereas BMSC-derived EVs had high levels. The transfer of miR-378a-5p by BMSC-derived EVs to HSMECs boosted proliferation, migration, and angiogenesis. miR-378a-5p inhibited IRF1. MiR-378a-5p-containing BMSC-derived EVs decreased STAT1 phosphorylation and HSMEC IRF1 expression. EVs with miR-378a-5p mimic promoted HSMEC proliferation, migration, and angiogenesis, whereas dexmedetomidine inhibited STAT1 phosphorylation. In CIA mice, BMSC-derived EVs containing miR-378a-5p enhanced synovial vascular remodeling and histopathology. Thus, miR-378a-5p from BMSC-derived EVs promotes HSMEC proliferation, migration, and angiogenesis, inactivating the IRF1/STAT1 axis and preventing RA.
Collapse
Affiliation(s)
- Yaqin Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Ziying Jiao
- Department of Physiology, School of Basic Medicine of Anhui Medical University, Hefei, PR China
| | - Shanshan Wang
- Department of Endocrinology, Anhui No.2 Provincial People’s Hospital, Hefei, PR China
| |
Collapse
|
9
|
Dehnavi S, Sadeghi M, Tavakol Afshari J, Mohammadi M. Interactions of mesenchymal stromal/stem cells and immune cells following MSC-based therapeutic approaches in rheumatoid arthritis. Cell Immunol 2023; 393-394:104771. [PMID: 37783061 DOI: 10.1016/j.cellimm.2023.104771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
Rheumatoid arthritis (RA) is considered to be a degenerative and progressive autoimmune disorder. Although several medicinal regimens are used to treat RA, potential adverse events such as metabolic disorders and increased risk of infection, as well as drug resistance in some patients, make it essential to find an effective and safe therapeutic approach. Mesenchymal stromal/stem cells (MSCs) are a group of non-hematopoietic stromal cells with immunomodulatory and inhibitory potential. These cells exert their regulatory properties through direct cell-to-cell interactions and paracrine effects on various immune and non-immune cells. As conventional therapeutic approaches for RA are limited due to their side effects, and some patients became refractory to the treatment, MSCs are considered as a promising alternative treatment for RA. In this review, we introduced various experimental and clinical studies conducted to evaluate the therapeutic effects of MSCs on animal models of arthritis and RA patients. Then, possible modulatory and suppressive effects of MSCs on different innate and adaptive immune cells, including dendritic cells, neutrophils, macrophages, natural killer cells, B lymphocytes, and various subtypes of T cells, were categorized and summarized. Finally, limitations and future considerations for the efficient application of MSCs as a therapeutic approach in RA patients were presented.
Collapse
Affiliation(s)
- Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Shimizu Y, Ntege EH, Azuma C, Uehara F, Toma T, Higa K, Yabiku H, Matsuura N, Inoue Y, Sunami H. Management of Rheumatoid Arthritis: Possibilities and Challenges of Mesenchymal Stromal/Stem Cell-Based Therapies. Cells 2023; 12:1905. [PMID: 37508569 PMCID: PMC10378234 DOI: 10.3390/cells12141905] [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: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent, chronic, and progressive autoimmune disorder primarily affecting joints and muscles. The associated inflammation, pain, and motor restriction negatively impact patient quality of life (QOL) and can even contribute to premature mortality. Further, conventional treatments such as antiinflammatory drugs are only symptomatic. Substantial progress has been made on elucidating the etiopathology of overt RA, in particular the contributions of innate and adaptive immune system dysfunction to chronic inflammation. Although the precise mechanisms underlying onset and progression remain elusive, the discovery of new drug targets, early diagnosis, and new targeted treatments have greatly improved the prognosis and QOL of patients with RA. However, a sizable proportion of patients develop severe adverse effects, exhibit poor responses, or cannot tolerate long-term use of these drugs, necessitating more effective and safer therapeutic alternatives. Mounting preclinical and clinical evidence suggests that the transplantation of multipotent adult stem cells such as mesenchymal stromal/stem cells is a safe and effective treatment strategy for controlling chronic inflammation and promoting tissue regeneration in patients with intractable diseases, including RA. This review describes the current status of MSC-based therapies for RA as well as the opportunities and challenges to broader clinical application.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Chinatsu Azuma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Fuminari Uehara
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Takashi Toma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Kotaro Higa
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Hiroki Yabiku
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| |
Collapse
|
11
|
Guan Y, Zhang Y, Zhu Y, Wang Y. CXCL10 as a shared specific marker in rheumatoid arthritis and inflammatory bowel disease and a clue involved in the mechanism of intestinal flora in rheumatoid arthritis. Sci Rep 2023; 13:9754. [PMID: 37328529 PMCID: PMC10276029 DOI: 10.1038/s41598-023-36833-7] [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] [Received: 01/04/2023] [Accepted: 06/10/2023] [Indexed: 06/18/2023] Open
Abstract
This study aimed to identify shared specific genes associated with rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) through bioinformatic analysis and to examine the role of the gut microbiome in RA. The data were extracted from the 3 RA and 1 IBD gene expression datasets and 1 RA gut microbiome metagenomic dataset. Weighted correlation network analysis (WGCNA) and machine learnings was performed to identify candidate genes associated with RA and IBD. Differential analysis and two different machine learning algorithms were used to investigate RA's gut microbiome characteristics. Subsequently, the shared specific genes related to the gut microbiome in RA were identified, and an interaction network was constructed utilizing the gutMGene, STITCH, and STRING databases. We identified 15 candidates shared genes through a joint analysis of the WGCNA for RA and IBD. The candidate gene CXCL10 was identified as the shared hub gene by the interaction network analysis of the corresponding WGCNA module gene to each disease, and CXCL10 was further identified as the shared specific gene by two machine learning algorithms. Additionally, we identified 3 RA-associated characteristic intestinal flora (Prevotella, Ruminococcus, and Ruminococcus bromii) and built a network of interactions between the microbiomes, genes, and pathways. Finally, it was discovered that the gene CXCL10 shared between IBD and RA was associated with the three gut microbiomes mentioned above. This study demonstrates the relationship between RA and IBD and provides a reference for research into the role of the gut microbiome in RA.
Collapse
Affiliation(s)
- Yin Guan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Yue Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Yifan Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Yue Wang
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Qinhuai, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
12
|
Mao X, Li Z, Gu S, Song W, Zhang M, Tan X, Mao Z. MicroRNA-211-5p in extracellular vesicles derived from BMSCs facilitates the repair of rat frozen shoulder via regulating KDM2B/LACC1 axis. Tissue Cell 2023; 81:102006. [PMID: 36610229 DOI: 10.1016/j.tice.2022.102006] [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] [Received: 07/06/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study aims to explore the mechanism of miR-211-5p in extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (BMSCs) in improving frozen shoulder (FS) in rat models. METHODS Rat BMSCs and EVs derived from rat BMSCs were isolated, identified, and then injected into rats to assess the expression of TGF-β, MMP1, MMP3, MMP12, GAP43, and PGP9.5 in shoulder capsule tissues. The range of motion of bilateral glenohumeral joints was assessed and pathological changes of shoulder capsule tissues were observed after hematoxylin-eosin staining. The binding sites of miR-211-5p to KDM2B and LACC1 to H3K4me3 were measured. FS rat models with LACC1 highly expressed were established to assess the motion of bilateral glenohumeral joints and expression of arthritis related factors in rats. RESULTS EVs were successfully extracted from BMSCs. Injection of BMSCs-EVs could improve the activity of bilateral glenohumeral joints and the pathological condition of joint capsule in rats. Elevated expression of miR-211-5p was found in rats injected with BMSCs-EVs. Dual luciferase assay showed that miR-211-5p had a binding site with KDM2B. ChIP, qRT-PCR, and western blot experiments showed BMSCs-EVs injection resulted in elevated enrichment of LACC1 promoter in shoulder capsule tissues of FS rats, and decreased mRNA and protein expression of KDM2B and increased H3K4me3 methylation. Overexpression of LACC1 could also improve the pathological condition of joint capsule tissue. CONCLUSION miR-211-5p in EVs derived from BMSCs increased H3K4me3 methylation in shoulder capsule tissue of rats by binding KDM2B, resulting in up-regulated transcription level of LACC1 and improving FS. AVAILABILITY OF DATA AND MATERIALS The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Xiaodong Mao
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Zhi Li
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Shaofang Gu
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Wei Song
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Mimi Zhang
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Xiao Tan
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China
| | - Ziqing Mao
- Department of Orthopedics & Traumatology, Department of Joint Surgery, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410008, PR China.
| |
Collapse
|
13
|
Arshad M, Jalil F, Jaleel H, Ghafoor F. Bone marrow derived mesenchymal stem cells therapy for rheumatoid arthritis - a concise review of past ten years. Mol Biol Rep 2023; 50:4619-4629. [PMID: 36929285 DOI: 10.1007/s11033-023-08277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/11/2023] [Indexed: 03/18/2023]
Abstract
Rheumatoid arthritis is an autoimmune disorder characterized by swelling in synovial joints and erosion of bones. The disease is normally treated with conventional drugs which provide only temporary relief to the symptoms. Over the past few years, mesenchymal stromal cells have become the center of attention for treating this disease due to their immuno-modulatory and anti-inflammatory characteristics. Various studies on treatment of rheumatoid arthritis by using these cells have shown positive outcomes in terms of reduction in the level of pain as well as improvement of the function and structure of joints. Mesenchymal stromal cells can be derived from multiple sources, however, the ones derived from bone marrow are considered most beneficial for treating several disorders including rheumatoid arthritis on account of being safer and more effective. This review summarizes all the preclinical and clinical studies which were conducted over the last ten years for therapy of rheumatoid arthritis utilizing these cells. The literature was reviewed using the terms "mesenchymal stem/stromal cells and rheumatoid arthritis'' and "bone marrow derived mesenchymal stromal cells and therapy of rheumatoid arthritis''. Data was extracted to enable the readers to have access to the most relevant information regarding advancement in therapeutic potential of these stromal cells. Additionally, this review will also help in fulfilling any gap in current knowledge of readers about the outcome of using these cells in animal models, cell line and in patients suffering from rheumatoid arthritis and other autoimmune disorders as well.
Collapse
Affiliation(s)
- Maria Arshad
- Department of Research & Innovation, Shalamar Institute of Health Sciences, Lahore, Pakistan.
| | - Fazal Jalil
- Department of Biotechnology, Abdul Wali Khan University, Mardan, Pakistan
| | - Hadiqa Jaleel
- Department of Research & Innovation, Shalamar Institute of Health Sciences, Lahore, Pakistan
| | - Farkhanda Ghafoor
- Department of Research & Innovation, Shalamar Institute of Health Sciences, Lahore, Pakistan
| |
Collapse
|
14
|
Hu J, Shi ZS, Liu XZ, Cai HT, Yang AF, Sun DM, Xu LL, Yang Y, Li ZH. Exosomes Derived from Runx2-Overexpressing BMSCs Enhance Cartilage Tissue Regeneration and Prevent Osteoarthritis of the Knee in a Rabbit Model. Stem Cells Int 2022; 2022:6865041. [PMID: 39282499 PMCID: PMC11401735 DOI: 10.1155/2022/6865041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/18/2022] [Accepted: 11/08/2022] [Indexed: 09/19/2024] Open
Abstract
Objectives Osteoarthritis is the leading disease of joints worldwide. Osteoarthritis may be treated by exosomes derived from Runx2-overexpressed bone marrow mesenchymal stem cells (R-BMSCs-Exos). R-BMSCs-Exos would promote the proliferation, migration, and phenotypic maintenance of articular chondrocytes. Methods BMSCs were transfected with and without Runx2. Exosomes derived from BMSCs and R-BMSCs (BMSCs-Exos and R-BMSCs-Exos) were isolated and identified. Proliferation, migration, and phenotypic maintenance were determined in vitro and compared between groups. The mechanism for activation of Yes-associated protein (YAP) was investigated using small interfering RNA (siRNA). The exosomes' preventive role was determined in vivo using Masson trichrome and immunohistochemical staining. Results R-BMSCs-Exos enhance the proliferation, migration, and phenotypic maintenance of articular chondrocytes based on the YAP being activated. R-BMSCs-Exos prevent knee osteoarthritis as studied in vivo through a rabbit model. Conclusions Findings emphasize the efficacy of R-BMSCs-Exos in preventing osteoarthritis. Potential source of exosomes is sorted out for the advantages and shortcomings. The exosomes are then modified based on the molecular mechanisms to address their limitations. Such exosomes derived from modified cells have the role in future therapeutics.
Collapse
Affiliation(s)
- Jing Hu
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430015, China
| | - Zheng-Shuai Shi
- Huanggang Hospital of TCM Affiliated to Hubei University of Chinese Medicine, Huanggang 438000, China
| | - Xiang-Zhong Liu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, China
| | - Han-Tao Cai
- Hubei University of Chinese Medicine, Wuhan 430061, China
| | - Ao-Fei Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
| | - Da-Ming Sun
- Wuhan Sports University, Wuhan 430079, China
| | | | - Yi Yang
- Wuhan Sports University, Wuhan 430079, China
| | - Zhang-Hua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
15
|
Liu J, Gao J, Niu Q, Wu F, Wu Z, Zhang L. Bibliometric and visualization analysis of mesenchymal stem cells and rheumatoid arthritis (from 2012 to 2021). Front Immunol 2022; 13:1001598. [PMID: 36311707 PMCID: PMC9606664 DOI: 10.3389/fimmu.2022.1001598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease that can lead to joint deformity and loss of function. Recent studies have shown great progress in the research of mesenchymal stem cells (MSCs) in RA. However, thus far, there have been no bibliometric or visualization analyses in this field. This bibliometric analysis provides a comprehensive overview of the general information and research hotspots of MSCs and RA. Methods Articles relevant to MSCs and RA, published between 2012 and 2021, were searched using the Web of Science Core Collection database. Irrelevant publications were excluded from the analysis. Bibliometric and visualization analyses were conducted using VOSviewer, CiteSpace, and Scimago Graphica. Results A total of 577 articles were analyzed. The annual number of publications increased from 2012 to 2017 and plateaued from 2017 to 2021. China and the USA had the largest number of publications. Collaboration among different organizations mainly occurs between institutes of the same country. Stem Cell Research and Therapy and Frontiers in Immunology were the most popular journals in this field. All the top 20 co-cited authors had a positive co-citation relationship. The top references indicate that MSCs can contribute to RA research and treatment mainly via immunomodulation. From 2012 to 2021, “collagen-induced arthritis,” “immunomodulation,” and “therapy” were some of the keywords associated with MSCs and RA, while “extracellular vesicles” showed a strong keyword burst from 2019 to 2021. Conclusion MSCs and RA have been widely studied in different countries and institutions and by different authors over the last ten years. China and the USA had the largest number of publications. Different types of journals provide admirable sources for researchers. Some keywords, including immunomodulation and extracellular vesicles, may be hot spots in the near future. There will be more basic research and clinical translation of MSCs and RA, and substantial new treatments for RA will soon be developed.
Collapse
Affiliation(s)
- Jiaxi Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Qing Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Fengping Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- *Correspondence: Liyun Zhang,
| |
Collapse
|
16
|
Li YJ, Chen Z. Cell-based therapies for rheumatoid arthritis: opportunities and challenges. Ther Adv Musculoskelet Dis 2022; 14:1759720X221100294. [PMID: 35634355 PMCID: PMC9131381 DOI: 10.1177/1759720x221100294] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common immune-mediated inflammatory disease characterized by chronic synovitis that hardly resolves spontaneously. The current treatment of RA consists of nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, conventional disease-modifying antirheumatic drugs (cDMARDs), biologic and targeted synthetic DMARDs. Although the treat-to-target strategy has been intensively applied in the past decade, clinical unmet needs still exist since a substantial proportion of patients are refractory or even develop severe adverse effects to current therapies. In recent years, with the deeper understanding of immunopathogenesis of the disease, cell-based therapies have exhibited effective and promising interventions to RA. Several cell-based therapies, such as mesenchymal stem cells (MSC), adoptive transfer of regulatory T cells (Treg), and chimeric antigen receptor (CAR)-T cell therapy as well as their beneficial effects have been documented and verified so far. In this review, we summarize the current evidence and discuss the prospect as well as challenges for these three types of cellular therapies in RA.
Collapse
Affiliation(s)
- Yu-Jing Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Second Clinical Medical School, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | | |
Collapse
|
17
|
Shin MJ, Park JY, Lee DH, Khang D. Stem Cell Mimicking Nanoencapsulation for Targeting Arthritis. Int J Nanomedicine 2022; 16:8485-8507. [PMID: 35002240 PMCID: PMC8725870 DOI: 10.2147/ijn.s334298] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered a promising regenerative therapy due to their ability to migrate toward damaged tissues. The homing ability of MSCs is unique compared with that of non-migrating cells and MSCs are considered promising therapeutic vectors for targeting major cells in many pathophysiological sites. MSCs have many advantages in the treatment of malignant diseases, particularly rheumatoid arthritis (RA). RA is a representative autoimmune disease that primarily affects joints, and secreted chemokines in the joints are well recognized by MSCs following their migration to the joints. Furthermore, MSCs can regulate the inflammatory process and repair damaged cells in the joints. However, the functionality and migration ability of MSCs injected in vivo still show insufficient. The targeting ability and migration efficiency of MSCs can be enhanced by genetic engineering or modification, eg, overexpressing chemokine receptors or migration-related genes, thus maximizing their therapeutic effect. However, there are concerns about genetic changes due to the increased probability of oncogenesis resulting from genome integration of the viral vector, and thus, clinical application is limited. Furthermore, it is suspected that administering MSCs can promote tumor growth and metastasis in xenograft and orthotopic models. For this reason, MSC mimicking nanoencapsulations are an alternative strategy that does not involve using MSCs or bioengineered MSCs. MSC mimicking nanoencapsulations consist of MSC membrane-coated nanoparticles, MSC-derived exosomes and artificial ectosomes, and MSC membrane-fused liposomes with natural or genetically engineered MSC membranes. MSC mimicking nanoencapsulations not only retain the targeting ability of MSCs but also have many advantages in terms of targeted drug delivery. Specifically, MSC mimicking nanoencapsulations are capable of encapsulating drugs with various components, including chemotherapeutic agents, nucleic acids, and proteins. Furthermore, there are fewer concerns over safety issues on MSC mimicking nanoencapsulations associated with mutagenesis even when using genetically engineered MSCs, because MSC mimicking nanoencapsulations use only the membrane fraction of MSCs. Genetic engineering is a promising route in clinical settings, where nano-encapsulated technology strategies are combined. In this review, the mechanism underlying MSC homing and the advantages of MSC mimicking nanoencapsulations are discussed. In addition, genetic engineering of MSCs and MSC mimicking nanoencapsulation is described as a promising strategy for the treatment of immune-related diseases.
Collapse
Affiliation(s)
- Min Jun Shin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jun Young Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, 21999, South Korea.,Department of Internal Medicine, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
18
|
Huang F, Thokerunga E, He F, Zhu X, Wang Z, Tu J. Research progress of the application of mesenchymal stem cells in chronic inflammatory systemic diseases. Stem Cell Res Ther 2022; 13:1. [PMID: 34998430 PMCID: PMC8742935 DOI: 10.1186/s13287-021-02613-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/04/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic inflammatory systemic diseases are the result of the body's immune imbalance, with a long course and recurring episodes. Immunosuppressants are the main treatment, but not all patients respond well to it. Being capable of both self-renewal and differentiation into multiple tissue cells and low immunogenicity, mesenchymal stem cell is a promising treatment for chronic inflammatory systemic diseases. In this article, we describe the research progress and clinical application of mesenchymal stem cells in chronic inflammatory systemic diseases and look for influencing factors and biomarkers that can predict the outcome of patient with mesenchymal stem cell transplantation.
Collapse
Affiliation(s)
- Fangfang Huang
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Erick Thokerunga
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fajian He
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinyu Zhu
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zi Wang
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiancheng Tu
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
19
|
He T, Sun S. Evaluation of the therapeutic efficacy of human bone marrow mesenchymal stem cells with COX-2 silence and TGF-β3 overexpression in rabbits with antigen-induced arthritis. Exp Cell Res 2022; 410:112945. [PMID: 34838812 DOI: 10.1016/j.yexcr.2021.112945] [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: 07/16/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs), especially genetically modified MSCs, have become a promising therapeutic approach for the treatment of rheumatoid arthritis (RA) through modulating immune responses. However, most MSCs used in the treatment of RA are modified based on a single gene. In this study, we evaluated the therapeutic effects of human BMSCs (hBMSCs) with COX-2 silence and TGF-β3 overexpression in the treatment of RA in a rabbit model. MATERIALS AND METHODS hBMSCs were cotransfected with shCOX-2 and TGF-β3 through lentiviral vector delivery. After SPIO-Molday ION Rhodamine-B™ (MIRB) labeling, lenti-shCOX2-TGF-β3 hBMSCs, lenti-shCOX2 hBMSCs, lenti-TGF-β3 hBMSCs, hBMSCs without genetic modification, or phosphate-buffered saline (PBS) were injected into the knee joint of rabbits with antigen-induced arthritis (AIA). The diameter of the knee joint and soft-tissue swelling score (STS) were recorded, and the levels of inflammatory mediators, including interleukin-1β (IL-1β), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and prostaglandin E2 (PGE2) were evaluated by ELISA. Clinical 3.0T MR imaging (MRI) was used to track the distribution and dynamic migration of hBMSCs in the joint. Histopathological and immunohistochemical assays were conducted to localize labeled hBMSCs and assess the alteration of synovial hyperplasia, inflammatory cell infiltration, and cartilage damage. RESULTS COX-2 silencing and TGF-β3 overexpression in hBMSCs were confirmed through real-time PCR and Western blot analyses. Reduced joint diameter, soft-tissue swelling (STS) score, and PGE2, IL-1β, and TNF-α expression were detected 4 weeks after injection of MIRB-labeled lenti-shCOX2-TGF-β3 hBMSCs into the joint in rabbits with AIA. Eight weeks after hBMSC injection, reduced inflammatory cell infiltration, improved hyperplasia of the synovial lining, recovered cartilage damage, and increased matrix staining were observed in joints injected with lenti-shCOX2-TGF-β3 hBMSCs and lenti-shCOX2 hBMSCs. Slight synovial hyperplasia, no surface fibrillation, and strong positive expression of collagen II staining in chondrocytes and cartilage matrix were detected in the joints 12 weeks after injection of lenti-shCOX2-TGF-β3 hBMSCs. In addition, hBMSCs were detected by MRI imaging throughout the process of hBMSC treatment. CONCLUSION Intra-articular injection of hBMSCs with COX-2 silence and TGFβ3 overexpression not only significantly inhibited joint inflammation and synovium hyperplasia, but also protected articular cartilage at the early stage. In addition, intra-articular injection of hBMSCs with COX-2 silence and TGFβ3 overexpression promoted chondrocyte and matrix proliferation. This study provides an alternative therapeutic strategy for the treatment of RA using genetically modified hBMSCs.
Collapse
Affiliation(s)
- Tian He
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China; Department of Orthopedics Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong, 264400, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
20
|
Tang S, Chen P, Zhang H, Weng H, Fang Z, Chen C, Peng G, Gao H, Hu K, Chen J, Chen L, Chen X. Comparison of Curative Effect of Human Umbilical Cord-Derived Mesenchymal Stem Cells and Their Small Extracellular Vesicles in Treating Osteoarthritis. Int J Nanomedicine 2021; 16:8185-8202. [PMID: 34938076 PMCID: PMC8687685 DOI: 10.2147/ijn.s336062] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/04/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) and their small extracellular vesicles (hUC-MSC-sEVs) have shown attractive prospects applying in regenerative medicine. This study aimed to compare the therapeutic effects of two agents on osteoarthritis (OA) and investigate underlying mechanism using proteomics. Methods In vitro, the proliferation and migration abilities of chondrocytes treated with hUC-MSCs or hUC-MSC-sEVs were detected by Cell Counting Kit-8 assay and scratch wound assay. In vivo, hUC-MSCs (a single dose of 5 × 105) or hUC-MSC-sEVs (30 μg/time) were injected into the knee joints of anterior cruciate ligament transection-induced OA model. Hematoxylin and eosin, Safranin O/Fast Green staining were used to observe cartilage degeneration. The levels of cartilage matrix metabolic molecules (Collagen II, MMP13 and ADAMTS5) and macrophage polarization markers (CD14, IL-1β, IL-10 and CD206) were assessed by immunohistochemistry. Finally, proteomics analysis was performed to characterize the proteinaceous contents of two agents. Results In vitro data showed that hUC-MSC-sEVs were taken up by chondrocytes. A total of 15 μg/mL of sEVs show the greatest proliferative and migratory capacities among all groups. In the animal study, hUC-MSCs and hUC-MSC-sEVs alleviated cartilage damage. This effect was mediated via maintaining cartilage homeostasis, as was confirmed by upregulation of the COL II and downregulation of the MMP13 and ADAMTS5. Moreover, the M1 macrophage markers (CD14) were significantly reduced, while the M2 macrophage markers (CD206 and IL-10) were increased in the hUC-MSCs and hUC-MSC-sEVs relative to the untreated group. Mechanistically, we found that many proteins connected to cartilage repair were more abundant in sEVs. Notably, compared to hUC-MSCs, the upregulated proteins in sEVs were mostly involved in the regulation of immune effector process, extracellular matrix organization, PI3K-AKT signaling pathways, and Rap1 signaling pathway. Conclusion Our study indicated that hUC-MSC-sEVs protect cartilage from damage and many cartilage repair-related proteins are probably involved in the restoration process. These data suggest the promising potential of hUC-MSC-sEVs as a therapeutic agent for OA.
Collapse
Affiliation(s)
- Shijie Tang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Penghong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Haoruo Zhang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Haiyan Weng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Zhuoqun Fang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Caixiang Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Guohao Peng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Hangqi Gao
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Kailun Hu
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Jinghua Chen
- Department of Pharmaceutical Analysis, the School of Pharmacy, Fujian Medical University, Fuzhou, 350100, People's Republic of China
| | - Liangwan Chen
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Xiaosong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China
| |
Collapse
|
21
|
Tang W, Zhang H, Liu D, Jiao F. Icariin accelerates cartilage defect repair by promoting chondrogenic differentiation of BMSCs under conditions of oxygen-glucose deprivation. J Cell Mol Med 2021; 26:202-215. [PMID: 34859578 PMCID: PMC8742234 DOI: 10.1111/jcmm.17073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/17/2021] [Accepted: 11/14/2021] [Indexed: 12/16/2022] Open
Abstract
This study explored the role played by combined ICA and bone mesenchymal stem cells (BMSCs) in repairing rabbit knee cartilage defects. Firstly, rabbit BMSCs were isolated and used to construct an in vitro cellular model of oxygen‐glucose deprivation/reoxygenation (OGD/R). Subsequently, ICA processing, Alcian blue staining, immunofluorescence and Western blot studies were performed to evaluate the ability of BMSCs to display signs of chondrogenic differentiation. Furthermore, a rabbit knee cartilage injury model was established in vivo. International Cartilage Repair Society (ICRS) macroscopic evaluations, H&E, Alcian blue and EdU staining, as well as immunohistochemistry, were analysed cartilage repair and pathological condition of the knee cartilage tissue. Our in vitro results showed that ICA promoted the chondrogenic differentiation of BMSCs, as well as aggrecan (AGR), bone morphogenetic protein 2 (BMP2) and COL2A1 protein expression in BMSCs. In vivo experiments showed that rabbits in the BMSCs or ICA treatment group had higher ICRS scores and displayed a better restoration of cartilage‐like tissue and chondrocyte expression on the surface of their cartilage defects. In conclusion, ICA or BMSCs alone could repair rabbit knee cartilage damage, and combined treatment with ICA and BMSCs showed a better ability to repair rabbit knee cartilage damage.
Collapse
Affiliation(s)
- Wang Tang
- Spinal Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Hongyi Zhang
- Joint Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Donghua Liu
- Spinal Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Feng Jiao
- Joint Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| |
Collapse
|
22
|
Zheng HL, Xu WN, Zhou WS, Yang RZ, Chen PB, Liu T, Jiang LS, Jiang SD. Beraprost ameliorates postmenopausal osteoporosis by regulating Nedd4-induced Runx2 ubiquitination. Cell Death Dis 2021; 12:497. [PMID: 33993186 PMCID: PMC8124066 DOI: 10.1038/s41419-021-03784-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
Bone health requires adequate bone mass, which is maintained by a critical balance between bone resorption and formation. In our study, we identified beraprost as a pivotal regulator of bone formation and resorption. The administration of beraprost promoted differentiation of mouse bone mesenchymal stem cells (M-BMSCs) through the PI3K–AKT pathway. In co-culture, osteoblasts stimulated with beraprost inhibited osteoclastogenesis in a rankl-dependent manner. Bone mass of p53 knockout mice remained stable, regardless of the administration of beraprost, indicating that p53 plays a vital role in the bone mass regulation by beraprost. Mechanistic in vitro studies showed that p53 binds to the promoter region of neuronal precursor cell-expressed developmentally downregulated 4 (Nedd4) to promote its transcription. As a ubiquitinating enzyme, Nedd4 binds to runt-related transcription factor 2 (Runx2), which results in its ubiquitination and subsequent degradation. These data indicate that the p53–Nedd4–Runx2 axis is an effective regulator of bone formation and highlight the potential of beraprost as a therapeutic drug for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Huo-Liang Zheng
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Wen-Ning Xu
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Wen-Sheng Zhou
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Run-Ze Yang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Peng-Bo Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Tao Liu
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China
| | - Lei-Sheng Jiang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China.
| | - Sheng-Dan Jiang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 200082, Shanghai, China.
| |
Collapse
|
23
|
Ma Y, Ran D, Shi X, Zhao H, Liu Z. Cadmium toxicity: A role in bone cell function and teeth development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 769:144646. [PMID: 33485206 DOI: 10.1016/j.scitotenv.2020.144646] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) is a widespread environmental contaminant that causes severe bone metabolism disease, such as osteoporosis, osteoarthritis, and osteomalacia. The present review aimed to explore the molecular mechanisms of Cd-induced bone injury starting from bone cell function and teeth development. Cd inhibits the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts, and directly causes BMSC apoptosis. In the case of osteoporosis, Cd mainly affects the activation of osteoclasts and promotes bone resorption. Cd-induces osteoblast injury and oxidative stress, which causes DNA damage, mitochondrial dysfunction, and endoplasmic reticulum stress, resulting in apoptosis. In addition, the development of osteoarthritis (OA) might be related to Cd-induced chondrocyte damage. The high expression of metallothionein (MT) might reduce Cd toxicity toward osteocytes. The toxicity of Cd toward teeth mainly focuses on enamel development and dental caries. Understanding the effect of Cd on bone cell function and teeth development could contribute to revealing the mechanisms of Cd-induced bone damage. This review explores Cd-induced bone disease from cellular and molecular levels, and provides new directions for removing this heavy metal from the environment.
Collapse
Affiliation(s)
- Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Di Ran
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Xueni Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Hongyan Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
24
|
Rusch RM, Ogawa Y, Sato S, Morikawa S, Inagaki E, Shimizu E, Tsubota K, Shimmura S. MSCs Become Collagen-Type I Producing Cells with Different Phenotype in Allogeneic and Syngeneic Bone Marrow Transplantation. Int J Mol Sci 2021; 22:4895. [PMID: 34063118 PMCID: PMC8125797 DOI: 10.3390/ijms22094895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used in therapeutic applications for many decades. However, more and more evidence suggests that factors such as the site of origin and pre-implantation treatment have a crucial impact on the result. This study investigates the role of freshly isolated MSCs in the lacrimal gland after allogeneic transplantation. For this purpose, MSCs from transgenic GFP mice were isolated and transplanted into allogeneic and syngeneic recipients. While the syngeneic MSCs maintained a spherical shape, allogeneic MSCs engrafted into the tissue as spindle-shaped cells in the interstitial stroma. Furthermore, the MSCs produced collagen type I in more than 85% to 95% of the detected GFP+ MSCs in the recipients of both models, supposedly contributing to pathogenic fibrosis in allogeneic recipients compared to syngeneic models. These findings indicate that allogeneic MSCs act completely differently from syngeneic MSCs, highlighting the importance of understanding the exact mechanisms behind MSCs.
Collapse
Affiliation(s)
- Robert Maximilian Rusch
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Satoru Morikawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan;
| | - Emi Inagaki
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; (R.M.R.); (S.S.); (E.I.); (E.S.); (K.T.)
| |
Collapse
|
25
|
Mesenchymal Stem Cells Enhance Therapeutic Effect and Prevent Adverse Gastrointestinal Reaction of Methotrexate Treatment in Collagen-Induced Arthritis. Stem Cells Int 2021; 2021:8850820. [PMID: 33505476 PMCID: PMC7814936 DOI: 10.1155/2021/8850820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/04/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by articular destruction and functional loss. Methotrexate (MTX) is effective in RA treatment. However, MTX induces several adverse events and 20%-30% of patients do not respond to MTX. Thus, it is urgent to enhance the therapeutic effects and reduce the side effects of MTX. Recent studies showed that mesenchymal stem cells (MSCs) were participants in anti-inflammation, immunoregulation, and tissue regeneration. However, whether the combined application of MSCs and MTX promotes the therapeutic effects and reduces the side effects of MTX has not been studied. In this study, we used bovine type II collagen to induce rheumatoid arthritis in mice (collagen-induced arthritis, CIA). Then, CIA mice were subjected to MTX or MSC treatment, or both. The therapeutic effect and adverse events of different treatments on RA were evaluated with micro-CT, HE staining, and immunohistochemistry in vivo. Apoptosis and proliferation of MODE-K cells were measured after treated with MTX or/and cocultured with UCs. To test M2 polarization, Raw264.7 macrophages were stimulated by MTX with different concentrations or cocultured with UCs. We found that the combined application of MSCs and MTX increased the therapeutic effects on RA, as evidenced by decreased arthritis score, inflammatory responses, and mortality. Moreover, in this combination remedy, MTX prefers to suppress inflammation by facilitating macrophage polarization to M2 type while UCs prefer to eliminate gastrointestinal side effects of MTX via mitigating the apoptosis of intestinal epithelial cells. Thus, a combination of MTX and UCs is a promising strategy for RA treatment.
Collapse
|
26
|
Mesenchymal Stem/Stromal Cells for Rheumatoid Arthritis Treatment: An Update on Clinical Applications. Cells 2020; 9:cells9081852. [PMID: 32784608 PMCID: PMC7465092 DOI: 10.3390/cells9081852] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/29/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that affects the lining of the synovial joints leading to stiffness, pain, inflammation, loss of mobility, and erosion of joints. Its pathogenesis is related to aberrant immune responses against the synovium. Dysfunction of innate and adaptive immunity, including dysregulated cytokine networks and immune complex-mediated complement activation, are involved in the progression of RA. At present, drug treatments, including corticosteroids, antirheumatic drugs, and biological agents, are used in order to modulate the altered immune responses. Chronic use of these drugs may cause adverse effects to a significant number of RA patients. Additionally, some RA patients are resistant to these therapies. In recent years, mesenchymal stem/stromal cell (MSCs)-based therapies have been largely proposed as a novel and promising stem cell therapeutic approach in the treatment of RA. MSCs are multipotent progenitor cells that have immunomodulatory properties and can be obtained and expanded easily. Today, nearly one hundred studies in preclinical models of RA have shown promising trends for clinical application. Proof-of-concept clinical studies have demonstrated satisfactory safety profile of MSC therapy in RA patients. The present review discusses MSC-based therapy approaches with a focus on published clinical data, as well as on clinical trials, for treatment of RA that are currently underway.
Collapse
|