1
|
Duan L, Lin W, Zhang Y, Jin L, Xiao J, Wang H, Pang S, Wang H, Sun D, Gong Y, Li H. Exosomes in Autoimmune Diseases: A Review of Mechanisms and Diagnostic Applications. Clin Rev Allergy Immunol 2025; 68:5. [PMID: 39820756 DOI: 10.1007/s12016-024-09013-2] [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: 12/13/2024] [Indexed: 01/19/2025]
Abstract
Exosomes, small extracellular vesicles secreted by various cell types, have emerged as key players in the pathophysiology of autoimmune diseases. These vesicles serve as mediators of intercellular communication, facilitating the transfer of bioactive molecules such as proteins, lipids, and nucleotide. In autoimmune diseases, exosomes have been implicated in modulating immune responses, oxidative stress, autophagy, gut microbes, and the cell cycle, contributing to disease initiation, progression, and immune dysregulation. Recent advancements in exosome isolation techniques and their molecular characterization have paved the way for exploring their clinical potential as biomarkers and therapeutic targets. This review focuses on the mechanisms by which exosomes influence autoimmune disease development and their potential clinical applications, particularly in diagnosis. The role of exosomes in autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), type 1 diabetes mellitus (T1DM), inflammatory bowel disease (IBD), and Sjögren's syndrome (SS), is discussed in relation to their involvements in antigen presentation, T-cell activation, and the induction of inflammatory pathways. Additionally, exosome-based biomarkers offer promising non-invasive diagnostic tools for early diagnostic, disease monitoring, and therapeutic response assessment. However, challenges such as standardization of exosome isolation protocols and validation of their clinical significance remain. This review highlights the potential of exosomes as both diagnostic biomarkers and therapeutic targets in autoimmune diseases, emphasizing the need for further research to overcome current limitations and fully harness their clinical value.
Collapse
Affiliation(s)
- Lina Duan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wanying Lin
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yi Zhang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Lingyue Jin
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jie Xiao
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Haifang Wang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuyin Pang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Hongxia Wang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Dehua Sun
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Ying Gong
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Haixia Li
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
2
|
Pang X, Xu F, Fan C, Jiang H. Global research trends of MicroRNAs in rheumatoid arthritis: bibliometrics and visualization analysis. Clin Rheumatol 2025; 44:53-66. [PMID: 39604743 DOI: 10.1007/s10067-024-07250-6] [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: 10/13/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder, and a growing body of research has demonstrated the involvement and regulatory roles of microRNAs (miRNAs) in the pathogenesis of RA. Consequently, this study aims to evaluate the current status and emerging trends in miRNA research related to RA by employing bibliometric analyses from a comprehensive perspective. METHODS Relevant literature pertaining to miRNA-related RA was systematically searched and screened from the Web of Science Core Collection (WOSCC) database, covering the period from January 1, 2010 to June 30, 2024. Subsequently, the data were subjected to visual analysis using CiteSpace, VOSviewer, the R package, and Bibliometric. This comprehensive approach aimed to elucidate the global state of research, as well as to identify prevailing trends and emerging hotspots within the field. RESULTS A comprehensive analysis included 843 publications authored by 4451 researchers from 1096 institutions across 47 countries/regions, with relevant articles disseminated through 297 journals. China emerged as the leading contributor in terms of research output. Specifically, China Medical University and the researcher Chih-Hsin Tang were identified as the most influential institution and author, respectively. "Frontiers in Immunology" was recognized as the most prolific journal, whereas "Arthritis Research & Therapy" garnered the highest number of citations. Notably, Stanczyk's 2008 study published in "Arthritis & Rheumatism" received the highest citation count. Additionally, related research areas such as circular RNAs (circRNAs) and extracellular vesicles (EVs) have become new hotspots in the field. CONCLUSIONS MiRNAs have garnered significant interest among researchers studying RA, as they show considerable potential for improving RA diagnosis and treatment. As research in this area advances, it is anticipated that scholars will allocate more resources to investigating circRNAs and EVs associated with miRNAs. This focus is expected to provide valuable insights for identifying novel therapeutic targets and drug delivery vectors for RA.
Collapse
Affiliation(s)
- Xue Pang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China
| | - Fengxia Xu
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China
| | - Chang Fan
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Hui Jiang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China.
| |
Collapse
|
3
|
Bostan ZZ, Şare Bulut M, Özen Ünaldı B, Albayrak Buhurcu C, Akbulut G. Effect of Plant-Based Diets on Rheumatoid Arthritis: A Systematic Review. Nutr Rev 2024:nuae181. [PMID: 39786551 DOI: 10.1093/nutrit/nuae181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
CONTEXT Rheumatoid arthritis (RA) is an autoimmune disease that leads to chronic inflammation and joint damage. Various plant-based diets are thought to have effects on RA symptoms and disease activity. OBJECTIVE Relevant literature on the effect of different types of plant-based diets on RA was reviewed. DATA SOURCES A systematic search was conducted using the Web of Science, PubMed, Scopus, and Cochrane databases and following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. DATA EXTRACTION Articles on observational and interventional human studies carried out in the adult population and published in English between February 2013 and April 2023 were eligible for inclusion. Articles reporting on studies performed in populations under 18 years of age, with unclear methods and results, or for which the full text was unavailable were excluded. The risk of bias in the selected studies was assessed using the Joanna Briggs Institute checklists. DATA ANALYSIS Of 547 screened articles, 15 were included, comprising 4 cross-sectional, 2 cohort, and 9 randomized controlled trials. These studies examined the effects of various diets, including the Mediterranean, low-fat high-carbohydrate, anti-inflammatory, and vegan, on RA. Results indicate adherence to plant-based diets generally reduced disease severity and improved dietary intake, physical activity, body weight, and key clinical markers such as the Disease Activity Score-28, Visual Analog Scale score, Health Assessment Questionnaire Disability Index, erythrocyte sedimentation rate, and C-reactive protein level in patients with in RA. CONCLUSIONS Most of the studies suggested the Mediterranean diet may positively affect the severity of RA. However, the included studies show heterogeneity. Therefore, more randomized controlled studies are needed in this area to increase understanding of the effect of diet on RA and facilitate the implementation of strategies to prevent RA. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023420577.
Collapse
Affiliation(s)
- Zekiye Zeynep Bostan
- Department of Nutrition and Dietetics, İstanbul Gedik University, 34906 İstanbul, Türkiye
| | - Melike Şare Bulut
- Department of Nutrition and Dietetics, Biruni University, 34015 İstanbul, Türkiye
| | - Buket Özen Ünaldı
- Department of Nutrition and Dietetics, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye
| | | | - Gamze Akbulut
- Department of Nutrition and Dietetics, Kent University, 34406 İstanbul, Türkiye
| |
Collapse
|
4
|
Jia M, Ren W, Wang M, Liu Y, Wang C, Zhang Z, Xu M, Ding N, Li C, Yang H. Surface saturation of drug-loaded hollow manganese dioxide nanoparticles with human serum albumin for treating rheumatoid arthritis. Drug Deliv 2024; 31:2380538. [PMID: 39044468 PMCID: PMC11271085 DOI: 10.1080/10717544.2024.2380538] [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: 09/20/2023] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nanchong Institute for Food and Drug Control, Nanchong, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Minrui Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Nianhui Ding
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Hong Yang
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
- The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Abebaw D, Akelew Y, Adugna A, Teffera ZH, Tegegne BA, Fenta A, Selabat B, Amare GA, Getinet M, Jemal M, Baylie T, Atnaf A. Extracellular vesicles: immunomodulation, diagnosis, and promising therapeutic roles for rheumatoid arthritis. Front Immunol 2024; 15:1499929. [PMID: 39624102 PMCID: PMC11609219 DOI: 10.3389/fimmu.2024.1499929] [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: 09/22/2024] [Accepted: 10/30/2024] [Indexed: 01/03/2025] Open
Abstract
Extracellular vesicles (EV) can be produced as part of pathology and physiology with increased amounts in pathological conditions. EVs can carry and transfer cargo such as proteins, nucleic acids, and lipids to target cells and mediate intercellular communication resulting in modulation of gene expression, signaling pathways, and phenotype of recipient cells. EVs greatly influence the extracellular environment and the immune response. Their immunomodulatory properties are crucial in rheumatoid arthritis (RA), a condition marked by dysregulated immune response. EVs can modulate the functions of innate and adaptive immune cells in RA pathogenesis. Differentially expressed EV-associated molecules in RA, such as microRNAs (miRNAs), long-noncoding RNAs (lncRNAs), messenger RNAs (mRNAs) and proteins are promising markers to diagnose the disease. miRNA, lncRNA, and circular RNA (circRNA) cargos in EV regulate inflammation and the pathogenic functions of RA fibroblast-like synoviocytes (RA-FLS). Downregulated molecules in RA tissue and drugs can be encapsulated in EVs for RA therapy. This review provides an updated overview of EVs' immunomodulatory, diagnostic, and therapeutic roles, particularly emphasizing mesenchymal stem cell-derived EVs (MSC-EVs).
Collapse
Affiliation(s)
- Desalegn Abebaw
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Akelew
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Adane Adugna
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantegize Selabat
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Temesgen Baylie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Aytenew Atnaf
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
6
|
Wang Z, Yang C, Yan S, Sun J, Zhang J, Qu Z, Sun W, Zang J, Xu D. Emerging Role and Mechanism of Mesenchymal Stem Cells-Derived Extracellular Vesicles in Rheumatic Disease. J Inflamm Res 2024; 17:6827-6846. [PMID: 39372581 PMCID: PMC11451471 DOI: 10.2147/jir.s488201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from mesoderm. Through cell-to-cell contact or paracrine effects, they carry out biological tasks like immunomodulatory, anti-inflammatory, regeneration, and repair. Extracellular vesicles (EVs) are the primary mechanism for the paracrine regulation of MSCs. They deliver proteins, nucleic acids, lipids, and other active compounds to various tissues and organs, thus facilitating intercellular communication. Rheumatic diseases may be treated using MSCs and MSC-derived EVs (MSC-EVs) due to their immunomodulatory capabilities, according to mounting data. Since MSC-EVs have low immunogenicity, high stability, and similar biological effects as to MSCs themselves, they are advantageous over cell therapy for potential therapeutic applications in rheumatoid arthritis, systemic erythematosus lupus, systemic sclerosis, Sjogren's syndrome, and other rheumatoid diseases. This review integrates recent advances in the characteristics, functions, and potential molecular mechanisms of MSC-EVs in rheumatic diseases and provides a new understanding of the pathogenesis of rheumatic diseases and MSC-EV-based treatment strategies.
Collapse
Affiliation(s)
- Zhangxue Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
- Department of Rheumatology and Immunology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chunjuan Yang
- Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Jiamei Sun
- Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Jin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
- Department of Rheumatology and Immunology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Zhuojian Qu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Wenchang Sun
- Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Jie Zang
- Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Donghua Xu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
- Department of Rheumatology and Immunology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
- Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
7
|
Wu J, Wu J, Liu Z, Gong Y, Feng D, Xiang W, Fang S, Chen R, Wu Y, Huang S, Zhou Y, Liu N, Xu H, Zhou S, Liu B, Ni Z. Mesenchymal stem cell-derived extracellular vesicles in joint diseases: Therapeutic effects and underlying mechanisms. J Orthop Translat 2024; 48:53-69. [PMID: 39170747 PMCID: PMC11338158 DOI: 10.1016/j.jot.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/09/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Joint diseases greatly impact the daily lives and occupational functioning of patients globally. However, conventional treatments for joint diseases have several limitations, such as unsatisfatory efficacy and side effects, necessitating the exploration of more efficacious therapeutic strategies. Mesenchymal stem cell (MSC)-derived EVs (MSC-EVs) have demonstrated high therapeutic efficacyin tissue repair and regeneration, with low immunogenicity and tumorigenicity. Recent studies have reported that EVs-based therapy has considerable therapeutic effects against joint diseases, including osteoarthritis, tendon and ligament injuries, femoral head osteonecrosis, and rheumatoid arthritis. Herein, we review the therapeutic potential of various types of MSC-EVs in the aforementioned joint diseases, summarise the mechanisms underlying specific biological effects of MSC-EVs, and discuss future prospects for basic research on MSC-EV-based therapeutic modalities and their clinical translation. In general, this review provides an in-depth understanding of the therapeutic effects of MSC-EVs in joint diseases, as well as the underlying mechanisms, which may be beneficial to the clinical translation of MSC-EV-based treatment. The translational potential of this article: MSC-EV-based cell-free therapy can effectively promote regeneration and tissue repair. When used to treat joint diseases, MSC-EVs have demonstrated desirable therapeutic effects in preclinical research. This review may supplement further research on MSC-EV-based treatment of joint diseases and its clinical translation.
Collapse
Affiliation(s)
- Jinhui Wu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Jiangyi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Zheng Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yunquan Gong
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Daibo Feng
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Wei Xiang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Shunzheng Fang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Shu Huang
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yizhao Zhou
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Ningning Liu
- Department of Laboratory Medicine, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450003, China
| | - Hao Xu
- Department of Laboratory Medicine, the Third Affiliated Hospital of Zhengzhou University Zhengzhou, 450003, China
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Baorong Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| |
Collapse
|
8
|
Wang Y, Zhang J, Yang Y, Liu Z, Sun S, Li R, Zhu H, Li T, Zheng J, Li J, Ma L. Circular RNAs in human diseases. MedComm (Beijing) 2024; 5:e699. [PMID: 39239069 PMCID: PMC11374765 DOI: 10.1002/mco2.699] [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: 06/20/2023] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Circular RNAs (circRNAs) are a unique class of RNA molecules formed through back-splicing rather than linear splicing. As an emerging field in molecular biology, circRNAs have garnered significant attention due to their distinct structure and potential functional implications. A comprehensive understanding of circRNAs' functions and potential clinical applications remains elusive despite accumulating evidence of their involvement in disease pathogenesis. Recent research highlights their significant roles in various human diseases, but comprehensive reviews on their functions and applications remain scarce. This review provides an in-depth examination of circRNAs, focusing first on their involvement in non-neoplastic diseases such as respiratory, endocrine, metabolic, musculoskeletal, cardiovascular, and renal disorders. We then explore their roles in tumors, with particular emphasis on exosomal circular RNAs, which are crucial for cancer initiation, progression, and resistance to treatment. By detailing their biogenesis, functions, and impact on disease mechanisms, this review underscores the potential of circRNAs as diagnostic biomarkers and therapeutic targets. The review not only enhances our understanding of circRNAs' roles in specific diseases and tumor types but also highlights their potential as novel diagnostic and therapeutic tools, thereby paving the way for future clinical investigations and potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuanyong Wang
- Department of Thoracic Surgery Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) The First Department of Thoracic Surgery Peking University Cancer Hospital and Institute Peking University School of Oncology Beijing China
| | - Jin Zhang
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Yuchen Yang
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Zhuofeng Liu
- Department of Traditional Chinese Medicine The Third Affiliated Hospital of Xi'an Medical University Xi'an China
| | - Sijia Sun
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Rui Li
- Department of Epidemiology School of Public Health Air Force Medical University Xi'an China
| | - Hui Zhu
- Department of Anatomy Medical College of Yan'an University Yan'an China
- Institute of Medical Research Northwestern Polytechnical University Xi'an China
| | - Tian Li
- School of Basic Medicine Fourth Military Medical University Xi'an China
| | - Jin Zheng
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Jie Li
- Department of Endocrine Xijing 986 Hospital Air Force Medical University Xi'an China
| | - Litian Ma
- Department of Thoracic Surgery Tangdu Hospital Air Force Medical University Xi'an China
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
- Department of Gastroenterology Tangdu Hospital Air Force Medical University Xi'an China
- School of Medicine Northwest University Xi'an China
| |
Collapse
|
9
|
Xu S, Zhang Y, Zheng Z, Sun J, Wei Y, Ding G. Mesenchymal stem cells and their extracellular vesicles in bone and joint diseases: targeting the NLRP3 inflammasome. Hum Cell 2024; 37:1276-1289. [PMID: 38985391 DOI: 10.1007/s13577-024-01101-x] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024]
Abstract
The nucleotide-binding oligomerization domain-like-receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a cytosolic multi-subunit protein complex, and recent studies have demonstrated the vital role of the NLRP3 inflammasome in the pathological and physiological conditions, which cleaves gasdermin D to induce inflammatory cell death called pyroptosis and mediates the release of interleukin-1 beta and interleukin-18 in response to microbial infection or cellular injury. Over-activation of the NLRP3 inflammasome is associated with the pathogenesis of many disorders affecting bone and joints, including gouty arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, and periodontitis. Moreover, mesenchymal stem cells (MSCs) have been discovered to facilitate the inhibition of NLRP3 and maybe ideal for treating bone and joint diseases. In this review, we implicate the structure and activation of the NLRP3 inflammasome along with the detail on the involvement of NLRP3 inflammasome in bone and joint diseases pathology. In addition, we focused on MSCs and MSC-extracellular vesicles targeting NLRP3 inflammasomes in bone and joint diseases. Finally, the existing problems and future direction are also discussed.
Collapse
Affiliation(s)
- Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Yanan Wei
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China.
| |
Collapse
|
10
|
Liu YR, Wang JQ, Fang L, Xia Q. Diagnostic and Therapeutic Roles of Extracellular Vesicles and Their Enwrapped ncRNAs in Rheumatoid Arthritis. J Inflamm Res 2024; 17:5475-5494. [PMID: 39165320 PMCID: PMC11334919 DOI: 10.2147/jir.s469032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory disease whose precise pathogenesis remains mysterious. The involvement of epigenetic regulation in the pathogenesis of RA is one of the most anticipated findings, among which non-coding RNAs (ncRNAs) hold great application promise as diagnostic and therapeutic biomarkers for RA. Extracellular vesicles (EVs) are a heterogeneous group of nano-sized, membrane-enclosed vesicles that mediate intercellular communication and substance exchange, especially the transfer of ncRNAs from donor cells, thereby regulating the functional activities and biological processes of recipient cells. In light of the significant correlation between EVs, ncRNAs, and RA, we first documented expression levels of EVs and their-encapsulated ncRNAs in RA individuals, and methodically discussed their-implicated signaling pathways and phenotypic changes. The last but not least, we paied special attention to the therapeutic benefits of gene therapy reagents specifically imitating or silencing candidate ncRNAs with exosomes as carriers on RA animal models, and briefly highlighted their clinical application advantage and foreground. In conclusion, the present review may be conducive to a deeper comprehension of the diagnostic and therapeutic roles of EVs-enwrapped ncRNAs in RA, with special emphasis on exosomal ncRNAs, which may offer hints for the monitoring and treatment of RA.
Collapse
Affiliation(s)
- Ya-ru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, 230022, People’s Republic of China
| | - Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, 230000, People’s Republic of China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, 230000, People’s Republic of China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, 230000, People’s Republic of China
| | - Ling Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, 230022, People’s Republic of China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, 230022, People’s Republic of China
| |
Collapse
|
11
|
Yan Q, Liu H, Sun S, Yang Y, Fan D, Yang Y, Zhao Y, Song Z, Chen Y, Zhu R, Zhang Z. Adipose-derived stem cell exosomes loaded with icariin alleviates rheumatoid arthritis by modulating macrophage polarization in rats. J Nanobiotechnology 2024; 22:423. [PMID: 39026367 PMCID: PMC11256651 DOI: 10.1186/s12951-024-02711-1] [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: 06/04/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease marked by synovitis and cartilage destruction. The active compound, icariin (ICA), derived from the herb Epimedium, exhibits potent anti-inflammatory properties. However, its clinical utility is limited by its water insolubility, poor permeability, and low bioavailability. To address these challenges, we developed a multifunctional drug delivery system-adipose-derived stem cells-exosomes (ADSCs-EXO)-ICA to target active macrophages in synovial tissue and modulate macrophage polarization from M1 to M2. High-performance liquid chromatography analysis confirmed a 92.4 ± 0.008% loading efficiency for ADSCs-EXO-ICA. In vitro studies utilizing cellular immunofluorescence (IF) and flow cytometry demonstrated significant inhibition of M1 macrophage proliferation by ADSCs-EXO-ICA. Enzyme-linked immunosorbent assay, cellular transcriptomics, and real-time quantitative PCR indicated that ADSCs-EXO-ICA promotes an M1-to-M2 phenotypic transition by reducing glycolysis through the inhibition of the ERK/HIF-1α/GLUT1 pathway. In vivo, ADSCs-EXO-ICA effectively accumulated in the joints. Pharmacodynamic assessments revealed that ADSCs-EXO-ICA decreased cytokine levels and mitigated arthritis symptoms in collagen-induced arthritis (CIA) rats. Histological analysis and micro computed tomography confirmed that ADSCs-EXO-ICA markedly ameliorated synovitis and preserved cartilage. Further in vivo studies indicated that ADSCs-EXO-ICA suppresses arthritis by promoting an M1-to-M2 switch and suppressing glycolysis. Western blotting supported the therapeutic efficacy of ADSCs-EXO-ICA in RA, confirming its role in modulating macrophage function through energy metabolism regulation. Thus, this study not only introduces a drug delivery system that significantly enhances the anti-RA efficacy of ADSCs-EXO-ICA but also elucidates its mechanism of action in macrophage function inhibition.
Collapse
Affiliation(s)
- Qiqi Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiyue Sun
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongsheng Yang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - DanPing Fan
- Institute of Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuqin Yang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yukun Zhao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiqian Song
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruyuan Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
12
|
Yang X, Zhang S, Lu J, Chen X, Zheng T, He R, Ye C, Xu J. Therapeutic potential of mesenchymal stem cell-derived exosomes in skeletal diseases. Front Mol Biosci 2024; 11:1268019. [PMID: 38903180 PMCID: PMC11187108 DOI: 10.3389/fmolb.2024.1268019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Shaodian Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Xiaoling Chen
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Tian Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
13
|
Liao HJ, Hsu PN. Immunomodulatory effects of extracellular vesicles from mesenchymal stromal cells: Implication for therapeutic approach in autoimmune diseases. Kaohsiung J Med Sci 2024; 40:520-529. [PMID: 38712483 DOI: 10.1002/kjm2.12841] [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: 03/24/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Autoimmune disease is characterized by the proliferation of harmful immune cells, inducing tissue inflammation and ultimately causing organ damage. Current treatments often lack specificity, necessitating high doses, prolonged usage, and high recurrence rates. Therefore, the identification of innovative and safe therapeutic strategies is urgently required. Recent preclinical studies and clinical trials on inflammatory and autoimmune diseases have evidenced the immunosuppressive properties of mesenchymal stromal cells (MSCs). Studies have demonstrated that extracellular vesicles (EV) derived from MSCs can mitigate abnormal autoinflammation while maintaining safety within the diseased microenvironment. This study conducted a systematic review to elucidate the crucial role of MSC-EVs in alleviating autoimmune diseases, particularly focusing on their impact on the underlying mechanisms of autoimmune conditions such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). By specifically examining the regulatory functions of microRNAs (miRNAs) derived from MSC-EVs, the comprehensive study aimed to enhance the understanding related to disease mechanisms and identify potential diagnostic markers and therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Ping-Ning Hsu
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Liao HJ, Yang YP, Liu YH, Tseng HC, Huo TI, Chiou SH, Chang CH. Harnessing the potential of mesenchymal stem cells-derived exosomes in degenerative diseases. Regen Ther 2024; 26:599-610. [PMID: 39253597 PMCID: PMC11382214 DOI: 10.1016/j.reth.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained attention as a promising therapeutic approach in both preclinical and clinical osteoarthritis (OA) settings. Various joint cell types, such as chondrocytes, synovial fibroblasts, osteoblasts, and tenocytes, can produce and release extracellular vesicles (EVs), which subsequently influence the biological activities of recipient cells. Recently, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have shown the potential to modulate various physiological and pathological processes through the modulation of cellular differentiation, immune responses, and tissue repair. This review explores the roles and therapeutic potential of MSC-EVs in OA and rheumatoid arthritis, cardiovascular disease, age-related macular degeneration, Alzheimer's disease, and other degenerative diseases. Notably, we provide a comprehensive summary of exosome biogenesis, microRNA composition, mechanisms of intercellular transfer, and their evolving role in the highlight of exosome-based treatments in both preclinical and clinical avenues.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Huan-Chin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ia Huo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, Taiwan
| |
Collapse
|
15
|
Liu XM, Yang L, Yang QB. Advanced Progress of Histone Deacetylases in Rheumatic Diseases. J Inflamm Res 2024; 17:947-955. [PMID: 38370467 PMCID: PMC10870932 DOI: 10.2147/jir.s447811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
Rheumatic disease is a disease which is not yet fully clarified to etiology and also involved in a local pathological injury or systemic disease. With the continuous improvement of clinical medical research in recent years, the development process of rheumatic diseases has been gradually elucidated; with the intensely study of epigenetics, it is realized that environmental changes can affect genetics, among which histone acetylation is one of the essential mechanisms in epigenetics. Histone deacetylases (HDACs) play an important role in regulating gene expression in various biological processes, including differentiation, development, stress response, and injury. HDACs are involved in a variety of physiological processes and are promising drug targets in various pathological conditions, such as cancer, cardiac and neurodegenerative diseases, inflammation, metabolic and immune disorders, and viral and parasitic infections. In this paper, we reviewed the roles of HDACs in rheumatic diseases in terms of their classification and function.
Collapse
Affiliation(s)
- Xue-Mei Liu
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People’s Republic of China
- Department of Clinical Medicine, Graduate School of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People’s Republic of China
| | - Liu Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People’s Republic of China
- Department of Clinical Medicine, Graduate School of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People’s Republic of China
| | - Qi-Bin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People’s Republic of China
| |
Collapse
|
16
|
Wei Z, Li H, Lv S, Yang J. Current situation and trend of non-coding RNA in rheumatoid arthritis: a review and bibliometric analysis. Front Immunol 2024; 14:1301545. [PMID: 38292492 PMCID: PMC10824985 DOI: 10.3389/fimmu.2023.1301545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease that affects multiple joints and has adverse effects on various organs throughout the body, often leading to a poor prognosis. Recent studies have shown significant progress in the research of non-coding RNAs (ncRNAs) in RA. Therefore, this study aims to comprehensively assess the current status and research trends of ncRNAs in RA through a bibliometric analysis. Methods This study retrieved articles relevant to ncRNAs and RA from the Science Citation Index Expanded Database of the Web of Science Core Collection between January 1st, 2003, and July 31st, 2023. The relevant articles were screened based on the inclusion criteria. VOSviewer and CiteSpace are utilized for bibliometric and visual analysis. Results A total of 1697 publications were included in this study, and there was a noticeable increase in annual publications from January 1st, 2003, to July 31st, 2023. China, the United States, and the United Kingdom were the most productive countries in this field, contributing to 43.81%, 13.09%, and 3.87% of the publications. Anhui Medical University and Lu Qianjin were identified as the most influential institution and author. Frontiers In Immunology stood out as the most prolific journal, while Arthritis & Rheumatology was the most co-cited journal. Additionally, the research related to "circular RNA", "oxidative stress", "proliferation", and "migration" have emerged as new hotspots in the field. Conclusion In this study, we have summarized the publication characteristics related to ncRNA and RA and identified the most productive countries, institutions, authors, journals, hot topics, and trends.
Collapse
Affiliation(s)
- Zehong Wei
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Huaiyu Li
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Senhao Lv
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Junping Yang
- Clinical Laboratory, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
17
|
Zhang B, Gu J, Wang Y, Guo L, Xie J, Yang M. TNF-α stimulated exosome derived from fibroblast-like synoviocytes isolated from rheumatoid arthritis patients promotes HUVEC migration, invasion and angiogenesis by targeting the miR-200a-3p/KLF6/VEGFA axis. Autoimmunity 2023; 56:2282939. [PMID: 37975481 DOI: 10.1080/08916934.2023.2282939] [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: 08/16/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
The pathogenesis of rheumatoid arthritis (RA) is heavily impacted by the inflammation and activation of fibroblast-like synoviocytes (FLS). The objective of this investigation is to clarify the involvement of exosomes derived from FLS stimulated by tumour necrosis factor α (TNF-α) in angiogenesis and the underlying mechanisms. FLS cells were obtained from synovial fluid of RA patients and exosomes were obtained from FLS cell supernatant with TNF-α stimulation by ultracentrifugation. Exosomes were subsequently analysed using transmission electron microscopy, nanoparticle tracking analysis, and western blotting. The functional effects of exosomes with TNF-α stimulation on human umbilical vein endothelial cells (HUVEC) migration, invasion, and angiogenesis was evaluated using wound scratch healing test, transwell invasion assay, and tube formation assay. DNA nanoball-seq (DNBSEQ) sequencing platform was utilised to analysis different expression miRNA from exosomes, miRNA and mRNA from HUVEC. The expression level of miR-200a-3p was determined through quantitative real-time polymerase chain reaction (qRT-PCR). The quantification of KLF6 and VEGFA expression levels were performed by qRT-PCR and western blot analysis. The validation of the association between miR-200a-3p and KLF6 was established through a fluorescence enzyme reporting assay. In comparison to exosome induced by PBS, exosome induced by TNF-α exhibited a substantial exacerbation of invasion, migration, and angiogenesis in HUVEC. 4 miRNAs in exosomes and HUVEC cells, namely miR-1246, miR-200a-3p, miR-30a-3p, and miR-99b-3p was obtained. MiR-200a-3p maintained high consistency with the sequencing results. We obtained 5 gene symbols, and KLF6 was chose for further investigation. The expression of miR-200a-3p in exosomes induced by TNF-α and in HUVEC treated with these exosomes demonstrated a significantly increase. Additionally, HUVEC cells displayed a notable decrease in KLF6 expression and a significant elevation in VEGFA expression. This was further confirmed by the fluorescence enzyme report assay, which provided evidence of the direct targeting of KLF6 by miR-200a-3p. Exosomes induced by TNF-α have the ability to enhance the migration, invasion, and angiogenesis of HUVEC cells via the miR-200a-3p/KLF6/VEGFA axis.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
- Jiaxing Key Laboratory of Osteoporosis and Bone Metabolism, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Juanfang Gu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
- Jiaxing Key Laboratory of Osteoporosis and Bone Metabolism, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Yiwen Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
- Jiaxing Key Laboratory of Osteoporosis and Bone Metabolism, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Linfeng Guo
- Zhejiang Chinese Medicine University and Jiaxing university Master degree cultivation base, Jiaxing, Zhejiang, China
| | | | - Mingfeng Yang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
- Jiaxing Key Laboratory of Osteoporosis and Bone Metabolism, The Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| |
Collapse
|
18
|
Ghafouri-Fard S, Shoorei H, Sabernia T, Hussen BM, Taheri M, Pourmoshtagh H. Circular RNAs and inflammation: Epigenetic regulators with diagnostic role. Pathol Res Pract 2023; 251:154912. [PMID: 38238072 DOI: 10.1016/j.prp.2023.154912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024]
Abstract
Circular RNAs (circRNAs) are a group of transcripts generally known to be non-coding transcripts, but occasionally producing short peptides. Circ_Ttc3/miR-148a, circ_TLK1/miR-106a-5p, circ_VMA21/miR-9-3p, circ_0068,888/miR-21-5p, circ_VMA21/miR-199a-5p, circ_AFF2/miR-375, circ_0008360/miR-135b-5p and circ-FBXW7/miR-216a-3p are examples of circRNA/miRNA pairs that contribute in the pathogenesis of immune-related conditions. CircRNAs have been found to regulate function of immune system and participate in the pathophysiology of immune-related disorders. In the current study, we searched PubMed and Google Scholar databases until July 2022 with the key words "circRNA" OR "circular RNA" AND "inflammation". Then, we assessed the abstract of retrieved articles to include original articles that assessed contribution of circRNAs in the pathoetiology of inflammation and related disorders. Finally, we went through the main texts of the articles and tabulated the available information. Therefore, the current study summarizes the role of circRNAs in the pathoetiology of sepsis, atherosclerosis, rheumatoid arthritis and osteoarthritis, immune-related cardiovascular, pulmonary, gastrointestinal and nervous system disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Toofan Sabernia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq; Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hasan Pourmoshtagh
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Bertolino GM, Maumus M, Jorgensen C, Noël D. Therapeutic potential in rheumatic diseases of extracellular vesicles derived from mesenchymal stromal cells. Nat Rev Rheumatol 2023; 19:682-694. [PMID: 37666995 DOI: 10.1038/s41584-023-01010-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 09/06/2023]
Abstract
The incidence of rheumatic diseases such as rheumatoid arthritis and osteoarthritis and injuries to articular cartilage that lead to osteochondral defects is predicted to rise as a result of population ageing and the increase in high-intensity physical activities among young and middle-aged people. Current treatments focus on the management of pain and joint functionality to improve the patient's quality of life, but curative strategies are greatly desired. In the past two decades, the therapeutic value of mesenchymal stromal cells (MSCs) has been evaluated because of their regenerative potential, which is mainly attributed to the secretion of paracrine factors. Many of these factors are enclosed in extracellular vesicles (EVs) that reproduce the main functions of parental cells. MSC-derived EVs have anti-inflammatory, anti-apoptotic as well as pro-regenerative activities. Research on EVs has gained considerable attention as they are a potential cell-free therapy with lower immunogenicity and easier management than whole cells. MSC-derived EVs can rescue the pathogenetic phenotypes of chondrocytes and exert a protective effect in animal models of rheumatic disease. To facilitate the therapeutic use of EVs, appropriate cell sources for the production of EVs with the desired biological effects in each disease should be identified. Production and isolation of EVs should be optimized, and pre-isolation and post-isolation modifications should be considered to maximize the disease-modifying potential of the EVs.
Collapse
Affiliation(s)
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France.
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France.
| |
Collapse
|
20
|
Wang Y, Huang Y, Cheng C, Xue Q, Chang J, Wang X, Duan Q, Miao C. Dysregulation of circRNAs in rheumatoid arthritis, with special emphasis on circRNAs secreted by exosomes and the crosstalk between circRNAs and RNA methylations. Int Immunopharmacol 2023; 122:110549. [PMID: 37421778 DOI: 10.1016/j.intimp.2023.110549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/09/2023] [Accepted: 06/18/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease caused by a variety of unknown factors. It mainly occurs in the small joints of hands and feet, leading to cartilage destruction and bone erosion. Various pathologic mechanisms such as exosomes and RNA methylations are involved in the pathogenesis of RA. METHODS This work searches PubMed, Web of Science (SCIE) and Science Direct Online (SDOL) databases, it role of abnormally expressed circulating RNAs (circRNAs) in the pathogenesis of RA was summarized. And the relationship between circRNAs and exosomes and methylations. RESULTS Both the abnormal expression of circRNAs and the sponge effect of circRNAs on microRNAs (miRNAs) affect the pathogenesis of RA by regulating target genes. CircRNAs affect the proliferation, migration and inflammatory reaction of RA-fibroblast-like synovial cells (FLSs), circRNAs in peripheral blood mononuclear cells (PBMCs) and macrophages also participate in the pathological mechanism of RA (Fig. 1). CircRNAs in exosomes are closely related to the pathogenesis of RA. In addition, exosomal circRNAs and the relationship between circRNAs and RNA methylations are closely related to the pathogenesis of RA. CONCLUSION CircRNAs play an important role in the pathogenesis of RA and have the potential to be a new target for the diagnosis and treatment of RA. However, the development of mature circRNAs for clinical application is not a small challenge.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chenglong Cheng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital, Anhui Medical University, Hefei 230032, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Qiangjun Duan
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
21
|
Zhang S, Duan Z, Liu F, Wu Q, Sun X, Ma H. The impact of exosomes derived from distinct sources on rheumatoid arthritis. Front Immunol 2023; 14:1240747. [PMID: 37575235 PMCID: PMC10414108 DOI: 10.3389/fimmu.2023.1240747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that can induce joint deformities and functional impairment, significantly impacting the overall well-being of individuals. Exosomes, which are cellularly secreted vesicles, possess favorable biological traits such as biocompatibility, stability, and minimal toxicity. Additionally, they contain nucleic acids, lipids, proteins, amino acids, and metabolites, serving as mediators in cellular communication and information exchange. Recent studies have demonstrated the association between exosomes and the pathogenesis of RA. Exosomes derived from mesenchymal stem cells, dendritic cells, and neutrophils exert influence on the biological functions of immune cells and joint cells, however, the precise mechanism remains largely unclarified. This comprehensive review systematically analyzes and summarizes the biological characteristics and functionalities of exosomes derived from diverse cellular sources, thus establishing a scientific foundation for the utilization of exosomes as diagnostic targets and therapeutic modalities in the context of RA.
Collapse
Affiliation(s)
- Sicheng Zhang
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Zhen Duan
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Fang Liu
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Qingjie Wu
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Xiwei Sun
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Hailong Ma
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
22
|
He X, He H, Zhang Y, Wu T, Chen Y, Tang C, Xia T, Zhang X, Xie C. Role of ceRNA network in inflammatory cells of rheumatoid arthritis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:750-759. [PMID: 37539578 PMCID: PMC10930406 DOI: 10.11817/j.issn.1672-7347.2023.220621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Indexed: 08/05/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease caused by inflammatory cells. Various inflammatory cells involved in RA include fibroblast-like synoviocytes, macrophages, CD4+T-lymphocytes, B lymphocytes, osteoclasts and chondrocytes. The close interaction between various inflammatory cells leads to imbalance of immune response and disorder of the expression of mRNA in inflammatory cells. It helps to drive production of pro-inflammatory cytokines and stimulate specific antigen-specific T- and B-lymphocytes to produce autoantibodies which is an important pathogenic factor for RA. Competing endogenous RNA (ceRNA) can regulate the expression of mRNA by competitively binding to miRNA. The related ceRNA network is a new regulatory mechanism for RNA interaction. It has been found to be involved in the regulation of abnormal biological processes such as proliferation, apoptosis, invasion and release of inflammatory factors of RA inflammatory cells. Understanding the ceRNA network in 6 kinds of RA common inflammatory cells provides a new idea for further elucidating the pathogenesis of RA, and provides a theoretical basis for the discovery of new biomarkers and effective therapeutic targets.
Collapse
Affiliation(s)
- Xiaoyu He
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004.
| | - Haohua He
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004
| | - Yan Zhang
- Department of Clinical Medicine, Bengbu Medical College, Bengbu Anhui 233030
| | - Tianyu Wu
- School of Public Health, Bengbu Medical College, Bengbu Anhui 233030
| | - Yongjie Chen
- School of Public Health, Bengbu Medical College, Bengbu Anhui 233030
| | - Chengzhi Tang
- School of Public Health, Bengbu Medical College, Bengbu Anhui 233030
| | - Tian Xia
- Department of Clinical Medicine, Bengbu Medical College, Bengbu Anhui 233030
| | - Xiaonan Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu Anhui 233030.
| | - Changhao Xie
- Department of Rheumatology and Immunology, First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004.
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu Anhui 233030, China.
| |
Collapse
|
23
|
Heydari R, Koohi F, Rasouli M, Rezaei K, Abbasgholinejad E, Bekeschus S, Doroudian M. Exosomes as Rheumatoid Arthritis Diagnostic Biomarkers and Therapeutic Agents. Vaccines (Basel) 2023; 11:vaccines11030687. [PMID: 36992270 DOI: 10.3390/vaccines11030687] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disorder that causes systemic inflammation, autoimmunity, and joint abnormalities that result in permanent disability. Exosomes are nanosized extracellular particles found in mammals (40–100 nm). They are a transporter of lipids, proteins, and genetic material involved in mammalian cell–cell signaling, biological processes, and cell signaling. Exosomes have been identified as playing a role in rheumatoid arthritis-related joint inflammation (RA). Uniquely functioning extracellular vesicles (EVs) are responsible for the transport of autoantigens and mediators between distant cells. In addition, paracrine factors, such as exosomes, modulate the immunomodulatory function of mesenchymal stem cells (MSCs). In addition to transporting genetic information, exosomes convey miRNAs between cells and have been studied as drug delivery vehicles. In animal models, it has been observed that MSCs secrete EVs with immunomodulatory properties, and promising results have been observed in this area. By understanding the diversity of exosomal contents and their corresponding targets, it may be possible to diagnose autoimmune diseases. Exosomes can be employed as diagnostic biomarkers for immunological disorders. We here discuss the most recent findings regarding the diagnostic, prognostic, and therapeutic potential of these nanoparticles in rheumatoid arthritis and provide an overview of the evidence pertaining to the biology of exosomes in RA.
Collapse
Affiliation(s)
- Romina Heydari
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
| | - Fatemeh Koohi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
| | - Milad Rasouli
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Jalale-Al-Ahmad Ave, 1411713137 Tehran, Iran
- Department of Physics, Kharazmi University, 49 Dr. Mofatteh Ave, Tehran 15614, Iran
| | - Kimia Rezaei
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
| | - Elham Abbasgholinejad
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str 2, 17489 Greifswald, Germany
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
| |
Collapse
|
24
|
Huang Y, Xue Q, Cheng C, Wang Y, Wang X, Chang J, Miao C. Circular RNA in autoimmune diseases: special emphasis on regulation mechanism in RA and SLE. J Pharm Pharmacol 2023; 75:370-384. [PMID: 36583516 DOI: 10.1093/jpp/rgac096] [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: 08/01/2022] [Accepted: 11/26/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Autoimmune diseases are diseases caused by tissue damage caused by the body's immune response to autoantibodies. Circular RNAs (CircRNAs) are a kind of special endogenous non-coding RNA that play a biological role by regulating gene transcription. METHODS In this work, we searched the PubMed, Web of Science (SCIE), National Science and Technology Library (NSTL), and ScienceDirect Online (SDOL) databases to summarize the impact of circRNAs on autoimmune diseases, especially the results of circRNAs in rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). RESULTS The study on the function of circRNAs and autoimmune diseases further deepened our understanding of the development and pathogenesis of autoimmune diseases. CircRNAs may act as miRNA sponges to regulate biological processes and affect the occurrence and development of autoimmune diseases. CircRNAs are closely related to the pathogenesis of RA and SLE and may become potential biomarkers for the diagnosis and treatment of RA and SLE. CONCLUSION CircRNAs play an important role in the pathogenesis of RA, SLE and other autoimmune diseases, and are expected to provide new biomarkers for the diagnosis and treatment of autoimmune diseases. However, the function and mechanism of circRNAs in autoimmune diseases need more comprehensive research.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chenglong Cheng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei 230032, China.,Anhui Public Health Clinical Center, Hefei, China
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
25
|
Fang Y, Ni J, Wang YS, Zhao Y, Jiang LQ, Chen C, Zhang RD, Fang X, Wang P, Pan HF. Exosomes as biomarkers and therapeutic delivery for autoimmune diseases: Opportunities and challenges. Autoimmun Rev 2023; 22:103260. [PMID: 36565798 DOI: 10.1016/j.autrev.2022.103260] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Exosomes are spherical lipid bilayer vesicles composed of lipids, proteins and nucleic acids that deliver signaling molecules through a vesicular transport system to regulate the function and morphology of target cells, thereby involving in a variety of biological processes, such as cell apoptosis or proliferation, and cytokine production. In the past decades, there are emerging evidence that exosomes play pivotal roles in the pathological mechanisms of several autoimmune diseases (ADs), including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes mellitus (T1DM), Sjogren's syndrome (SS), multiple sclerosis (MS), inflammatory bowel disease (IBD). systemic sclerosis (SSc), etc. Several publications have shown that exosomes are involved in the pathogenesis of ADs mainly through intercellular communication and by influencing the response of immune cells. The level of exosomes and the expression of nucleic acids can reflect the degree of disease progression and are excellent biomarkers for ADs. In addition, exosomes have the potential to be used as drug carriers thanks to their biocompatibility and stability. In this review, we briefly summarized the current researches regarding the biological functions of exosomes in ADs, and provided an insight into the potential of exosomes as biomarkers and therapeutic delivery for these diseases.
Collapse
Affiliation(s)
- Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Yun-Sheng Wang
- Department of Endocrinology, the Second People's Hospital of Hefei, the Affiliated Hefei Hospital of Anhui Medical University, Hefei 230011, Anhui, China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Peng Wang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China; Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China.
| |
Collapse
|
26
|
Ni R, Liu H, Song G, Fu X, Deng B, Xu Z, Dai S, Huang G. MiR-216a-3p inhibits the proliferation and invasion of fibroblast-like synoviocytes by targeting dual-specificity phosphatase 5. Int J Rheum Dis 2023; 26:699-709. [PMID: 36843205 DOI: 10.1111/1756-185x.14622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/28/2023]
Abstract
Dual-specificity phosphatase 5 (DUSP5) is a novel anti-inflammatory modulator in many inflammatory diseases. However, the role of DUSP5 in fibroblast-like synoviocytes (FLS) of rheumatoid arthritis (RA) remains unknown. In this study, we aimed to explore the biological function and regulation of DUSP5 in FLS. We found that lower DUSP5 expression level was detected in collagen-induced arthritis (CIA) and synoviocyte MH7A. Overexpression of DUSP5 markedly decreased the proliferation, migration, and invasion of MH7A, which correlated with suppressing the phosphorylation of extracellular signal-regulated kinase (ERK). Moreover, DUSP5 was identified as a novel target gene of miR-216a-3p, which was upregulated in FLS. Therefore, DUSP5 expression was negatively regulated by miR-216a-3p, and the effect of DUSP5 overexpression on FLS was reversed by miR-216a-3p mimics. Overall, our study demonstrates that DUSP5 is a miR-216a-3p target gene and its anti-inflammatory function in FLS via inactivation of ERK. These results revealed that the miR-216a-3p/DUSP5 pathway may play a crucial role in the malignant behavior of FLS, which may serve as a new target for the treatment of RA.
Collapse
Affiliation(s)
- Rongrong Ni
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Heting Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Guojing Song
- Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaohong Fu
- Office of Academic Research, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Bingqian Deng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Shuangshuang Dai
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, Chongqing, China
| |
Collapse
|
27
|
Gerami MH, Khorram R, Rasoolzadegan S, Mardpour S, Nakhaei P, Hashemi S, Al-Naqeeb BZT, Aminian A, Samimi S. Emerging role of mesenchymal stem/stromal cells (MSCs) and MSCs-derived exosomes in bone- and joint-associated musculoskeletal disorders: a new frontier. Eur J Med Res 2023; 28:86. [PMID: 36803566 PMCID: PMC9939872 DOI: 10.1186/s40001-023-01034-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023] Open
Abstract
Exosomes are membranous vesicles with a 30 to 150 nm diameter secreted by mesenchymal stem/stromal cells (MSCs) and other cells, such as immune cells and cancer cells. Exosomes convey proteins, bioactive lipids, and genetic components to recipient cells, such as microRNAs (miRNAs). Consequently, they have been implicated in regulating intercellular communication mediators under physiological and pathological circumstances. Exosomes therapy as a cell-free approach bypasses many concerns regarding the therapeutic application of stem/stromal cells, including undesirable proliferation, heterogeneity, and immunogenic effects. Indeed, exosomes have become a promising strategy to treat human diseases, particularly bone- and joint-associated musculoskeletal disorders, because of their characteristics, such as potentiated stability in circulation, biocompatibility, low immunogenicity, and toxicity. In this light, a diversity of studies have indicated that inhibiting inflammation, inducing angiogenesis, provoking osteoblast and chondrocyte proliferation and migration, and negative regulation of matrix-degrading enzymes result in bone and cartilage recovery upon administration of MSCs-derived exosomes. Notwithstanding, insufficient quantity of isolated exosomes, lack of reliable potency test, and exosomes heterogeneity hurdle their application in clinics. Herein, we will deliver an outline respecting the advantages of MSCs-derived exosomes-based therapy in common bone- and joint-associated musculoskeletal disorders. Moreover, we will have a glimpse the underlying mechanism behind the MSCs-elicited therapeutic merits in these conditions.
Collapse
Affiliation(s)
- Mohammad Hadi Gerami
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Khorram
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheil Rasoolzadegan
- grid.411600.2Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Mardpour
- grid.411705.60000 0001 0166 0922Department of Radiology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooria Nakhaei
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheyla Hashemi
- grid.411036.10000 0001 1498 685XObstetrician, Gynaecology & Infertility Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Amir Aminian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Zhao J, Zhang B, Meng W, Hu J. Elucidating a fresh perspective on the interplay between exosomes and rheumatoid arthritis. Front Cell Dev Biol 2023; 11:1177303. [PMID: 37187619 PMCID: PMC10175795 DOI: 10.3389/fcell.2023.1177303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by chronic synovitis and the destruction of bones and joints. Exosomes are nanoscale lipid membrane vesicles originating from multivesicular bodies and are used as a vital means of intercellular communication. Both exosomes and the microbial community are essential in RA pathogenesis. Multiple types of exosomes from different origins have been demonstrated to have effects on various immune cells through distinct mechanisms in RA, which depend on the specific cargo carried by the exosomes. Tens of thousands of microorganisms exist in the human intestinal system. Microorganisms exert various physiological and pathological effects on the host directly or through their metabolites. Gut microbe-derived exosomes are being studied in the field of liver disease; however, information on their role in the context of RA is still limited. Gut microbe-derived exosomes may enhance autoimmunity by altering intestinal permeability and transporting cargo to the extraintestinal system. Therefore, we performed a comprehensive literature review on the latest progress on exosomes in RA and provided an outlook on the potential role of microbe-derived exosomes as emerging players in clinical and translational research on RA. This review aimed to provide a theoretical basis for developing new clinical targets for RA therapy.
Collapse
Affiliation(s)
- Jianan Zhao
- Department of Nephropathy, The Seventh People’s Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Binbin Zhang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
- Department of Translational Medicine Platform, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Wanting Meng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Hu
- Department of Nephropathy, The Seventh People’s Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Jing Hu,
| |
Collapse
|
29
|
Alcaraz MJ, Guillén MI. Cellular and Molecular Targets of Extracellular Vesicles from Mesenchymal Stem/Stromal Cells in Rheumatoid Arthritis. Stem Cells Transl Med 2022; 11:1177-1185. [PMID: 36318277 PMCID: PMC9801303 DOI: 10.1093/stcltm/szac075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes progressive joint destruction. Despite the advances in the treatment of this condition there remains a clinical need for safe therapies leading to clinical remission. Mesenchymal stem/stromal cells (MSCs) play immunomodulatory and regenerative roles which can be partly mediated by their secretome. In recent years, the important contribution of extracellular vesicles (EVs) to MSC actions has received an increasing interest as a new therapeutic approach. We provide an extensive overview of the immunomodulatory properties of MSC EVs and their effects on articular cells such as fibroblast-like synoviocytes that play a central role in joint destruction. This review discusses the anti-arthritic effects of MSC EVs in vitro and in animal models of RA as well as their potential mechanisms. Recent preclinical data suggest that transfer of non-coding RNAs by MSC EVs regulates key signaling pathways involved in the pathogenesis of RA. We also examine a number of EV modifications for improving their anti-arthritic efficacy and carrier ability for drug delivery.
Collapse
Affiliation(s)
- María José Alcaraz
- Corresponding author: María José Alcaraz, PhD, Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Burjassot, Valencia, Spain. E-mail:
| | - María Isabel Guillén
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Av. Vicent A. Estellés s/n, Burjassot, Valencia, Spain,Department of Pharmacy, Faculty of Health Sciences, Cardenal Herrera-CEU University, Alfara del Patriarca, Valencia, Spain
| |
Collapse
|
30
|
MSC-EV therapy for bone/cartilage diseases. Bone Rep 2022; 17:101636. [DOI: 10.1016/j.bonr.2022.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
|
31
|
Pang H, Fan W, Shi X, Luo S, Wang Y, Lin J, Xiao Y, Li X, Huang G, Xie Z, Zhou Z. Differential Expression and Bioinformatics Analysis of Plasma-Derived Exosomal circRNA in Type 1 Diabetes Mellitus. J Immunol Res 2022; 2022:3625052. [PMID: 36339941 PMCID: PMC9634467 DOI: 10.1155/2022/3625052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/29/2022] [Accepted: 10/08/2022] [Indexed: 12/02/2022] Open
Abstract
Backgrounds Both exosome and circular RNA (circRNA) have been reported to participate in the pathogenesis of type 1 diabetes mellitus (T1DM). However, the exact role of exosomal circRNA in T1DM is largely unknown. Here, we identified the exosomal circRNA expression profiles in the plasma of T1DM patients and explored their potential function using bioinformatics analysis. Material and Methods . Exosomes were extracted by the size exclusion chromatography method from plasma of 10 T1DM patients and 10 age- and sex- matched control subjects. Illumina Novaseq6000 platform was used to detect the exosomal circRNA expression profiles. Multiple bioinformatics analysis was applied to investigate the potential biological functions of exosomal circRNAs. Results A total of 784 differentially expressed exosomal circRNAs have been identified in T1DM patients, of which 528 were upregulated and 256 were downregulated. Gene Ontology analysis enriched terms such as protein ubiquitination involved in ubiquitin-dependent protein catabolic protein (GO:0042787), membrane (GO:0016020), and GTPase activator activity (GO:0005096). The most enriched pathway in Kyoto Encyclopedia of Genes and Genomes was ubiquitin-mediated proteolysis (ko04120). The miRNA-targeting prediction method was used to identify the miRNAs that bind to circRNAs, and circRNA-miRNA-mRNA pathways were constructed, indicating that interactions between circRNA, miRNA, and gene might be involved in the disease progression. Conclusions The present study identified the exosomal circRNA expression profiles in T1DM for the first time. Our results threw novel insights into the molecular mechanisms of T1DM.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Wenqi Fan
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Xiajie Shi
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Yimeng Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Jian Lin
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011 Hunan, China
| |
Collapse
|
32
|
Műzes G, Sipos F. Mesenchymal Stem Cell-Derived Secretome: A Potential Therapeutic Option for Autoimmune and Immune-Mediated Inflammatory Diseases. Cells 2022; 11:cells11152300. [PMID: 35892597 PMCID: PMC9367576 DOI: 10.3390/cells11152300] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/16/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) encompass several entities such as "classic" autoimmune disorders or immune-mediated diseases with autoinflammatory characteristics. Adult stem cells including mesenchymal stem cells (MSCs) are by far the most commonly used type in clinical practice. However, due to the possible side effects of MSC-based treatments, there is an increase in interest in the MSC-secretome (containing large extracellular vesicles, microvesicles, and exosomes) as an alternative therapeutic option in IMIDs. A wide spectrum of MSC-secretome-related biological activities has been proven thus far including anti-inflammatory, anti-apoptotic, and immunomodulatory properties. In comparison with MSCs, the secretome is less immunogenic but exerts similar biological actions, so it can be considered as an ideal cell-free therapeutic alternative. Additionally, since the composition of the MSC-secretome can be engineered, for a future perspective, it could also be viewed as part of a potential delivery system within nanomedicine, allowing us to specifically target dysfunctional cells or tissues. Although many encouraging results from pre-clinical studies have recently been obtained that strongly support the application of the MSC-secretome in IMIDs, human studies with MSC-secretome administration are still in their infancy. This article reviews the immunomodulatory effects of the MSC-secretome in IMIDs and provides insight into the interpretation of its beneficial biological actions.
Collapse
|
33
|
Adipose-Derived Stem Cell Exosomes as a Novel Anti-Inflammatory Agent and the Current Therapeutic Targets for Rheumatoid Arthritis. Biomedicines 2022; 10:biomedicines10071725. [PMID: 35885030 PMCID: PMC9312519 DOI: 10.3390/biomedicines10071725] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Patients with rheumatoid arthritis (RA), a chronic inflammatory joint disorder, may not respond adequately to current RA treatments. Mesenchymal stem cells (MSCs) elicit several immunomodulatory and anti-inflammatory effects and, thus, have therapeutic potential. Specifically, adipose-derived stem cell (ADSC)-based RA therapy may have considerable potency in modulating the immune response, and human adipose tissue is abundant and easy to obtain. Paracrine factors, such as exosomes (Exos), contribute to ADSCs’ immunomodulatory function. ADSC-Exo-based treatment can reproduce ADSCs’ immunomodulatory function and overcome the limitations of traditional cell therapy. ADSC-Exos combined with current drug therapies may provide improved therapeutic effects. Using ADSC-Exos, instead of ADSCs, to treat RA may be a promising cell-free treatment strategy. This review summarizes the current knowledge of medical therapies, ADSC-based therapy, and ADSC-Exos for RA and discusses the anti-inflammatory properties of ADSCs and ADSC-Exos. Finally, this review highlights the expanding role and potential immunomodulatory activity of ADSC-Exos in patients with RA.
Collapse
|
34
|
Mou X, Jin Y, Jin D, Guan J, Zhang Q. Serum
HDAC4
level in rheumatoid arthritis: Longitudinal change during treatment and correlation with clinical outcomes. J Clin Lab Anal 2022; 36:e24594. [PMID: 35792020 PMCID: PMC9396184 DOI: 10.1002/jcla.24594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/09/2022] [Accepted: 06/26/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Histone deacetylase 4 (HDAC4) modulates immunity, inflammation, and osteoblast differentiation to engage in rheumatoid arthritis (RA) etiology. This study aimed to evaluate the HDAC4 longitudinal change and its relationship with clinical features and outcomes in RA patients. Methods Eighty‐three RA patients were enrolled. Their serum HDAC4 level was detected by ELISA at baseline (W0), week (W) 4, W12, and W24 after treatment. RA patients were divided into response or non‐response, low disease activity (LDA) or non‐LDA, remission or non‐remission patients according to their treatment outcomes at W24. Meanwhile, serum HDAC4 was detected by ELISA in 20 osteoarthritis patients and 20 healthy controls (HCs). Results HDAC4 level was reduced in RA patients compared with HCs (p < 0.001) and osteoarthritis patients (p = 0.009). HDAC4 was negatively related to some of the disease activity indexes such as C‐reactive protein (p = 0.003), tender joint count (p = 0.025), and disease activity score based on 28 joints (p = 0.013) in RA patients; it was also negatively correlated with TNF‐α (p = 0.003), IL‐6 (p = 0.022), and IL‐17A (p = 0.015). However, the HDAC4 level was not related to different treatment histories or current initiating treatment regimens (all p < 0.05). After treatment, HDAC4 was gradually elevated along with the time (p < 0.001). Interestingly, HDAC4 level at W12 (p = 0.041) and W24 (p = 0.012) was higher in response patients versus non‐response patients, and its level at W24 was higher in LDA patients versus non‐LDA patients (p = 0.019), and in remission patients versus non‐remission patients (p = 0.039). Conclusion HDAC4 gradually increases during treatment and its elevation estimates good treatment outcomes in RA patients.
Collapse
Affiliation(s)
- Xiaoyue Mou
- Department of RheumatologyFirst People's Hospital of TaizhouTaizhouChina
| | - Yi Jin
- Department of OphthalmologyFirst People's Hospital of TaizhouTaizhouChina
| | - Du Jin
- Department of RheumatologyFirst People's Hospital of TaizhouTaizhouChina
| | - Jintao Guan
- Department of RheumatologyFirst People's Hospital of TaizhouTaizhouChina
| | - Qian Zhang
- Department of RheumatologyFirst People's Hospital of TaizhouTaizhouChina
| |
Collapse
|
35
|
Dou B, Ma F, Jiang Z, Zhao L. Blood HDAC4 Variation Links With Disease Activity and Response to Tumor Necrosis Factor Inhibitor and Regulates CD4+ T Cell Differentiation in Ankylosing Spondylitis. Front Med (Lausanne) 2022; 9:875341. [PMID: 35602496 PMCID: PMC9121817 DOI: 10.3389/fmed.2022.875341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 12/17/2022] Open
Abstract
Purpose Histone deacetylase 4 (HDAC4) regulates the progression of autoimmune diseases. This study aimed to further investigate the correlation between HDAC4 and Th cells, inflammation, disease activity, and treatment response in patients with ankylosing spondylitis (AS). Methods A total of 132 active patients with AS were enrolled, of whom 54 patients received TNF inhibitor (TNFi) and 78 patients received NSAID. Serum HDAC4 was measured by ELISA in patients with AS before treatment (W0) and at week (W)4, W8, and W12 after treatment. Meanwhile, serum HDAC4 was detected in 30 patients with osteoarthritis and in 30 healthy controls (HCs) by ELISA. Besides, naïve CD4+ T cells from patients with AS were isolated, followed by modulation of HDAC4 and then polarization toward Th1, Th2, and Th17. Results Histone deacetylase 4 was reduced in patients with AS compared with HCs and patients with osteoarthritis (both P < 0.01). In patients with AS, HDAC4 was negatively correlated with TNF (P < 0.001), IL-1β (P = 0.003), Th17 proportion (P = 0.008), C-reactive protein (P < 0.001), and ASDAS (P = 0.038), but not with IL-6, Th1 proportion, or other characteristics. Meanwhile, HDAC4 increased from W0 to W12 (P < 0.001); HDAC4 at W8 (P = 0.014) and W12 (P = 0.006) was raised in ASAS40-response patients than ASAS40-non-response patients; further subgroup analysis showed that HDAC4 at W12 was higher in ASAS40-response patients than ASAS40-non-response patients (P = 0.016) in the TNFi-treated group, but not in the NSAID-treated group. In addition, HDAC4 negatively regulated the polarization of naïve CD4+ T cells toward Th17 (P < 0.01), but not Th1 or Th2. Conclusion Histone deacetylase 4 is associated with lower inflammation, and the disease activity negatively regulates Th17 polarization, whose increment after treatment reflects favorable outcomes in patients with AS.
Collapse
Affiliation(s)
- Bin Dou
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, China
| | - Fuzhe Ma
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Zhenyu Jiang
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zhenyu Jiang
| | - Ling Zhao
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, China
- Ling Zhao
| |
Collapse
|
36
|
Liu X, Hu L, Liu F. Mesenchymal stem cell-derived extracellular vesicles for cell-free therapy of ocular diseases. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:102-117. [PMID: 39698446 PMCID: PMC11648472 DOI: 10.20517/evcna.2022.08] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/31/2022] [Accepted: 04/18/2022] [Indexed: 12/20/2024]
Abstract
Mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) have noticeably attracted clinicians' attention in treating ocular diseases. As the paracrine factor of MSCs and an alternative for cell-free therapies, MSC-EVs can be conveniently dropped over the ocular surface or diffused through the retina upon intravitreal injection, without increasing the risks of cellular rejection and tumor formation. For clinical translation, a standardized and scalable production, as well as reprogramming the MSC-EVs, are highly encouraged. This review aims to assess the potential approaches for EV production and functional modification, in addition to summarizing the worldwide clinical trials initiated for various physiological systems and the specific biochemical effects of MSC-EVs on the therapy of eye diseases. Recent advances in the therapy of ocular diseases based on MSC-EVs are reviewed, and the associated challenges and prospects are discussed as well.
Collapse
Affiliation(s)
- Xiaoling Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Liang Hu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Fei Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Science, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
37
|
Zhao Y, Yao J. The Biological Functions and Clinical Values of Exosomal Circular RNAs in Hepatocellular Carcinoma. Front Oncol 2022; 12:885214. [PMID: 35530313 PMCID: PMC9069097 DOI: 10.3389/fonc.2022.885214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/23/2022] [Indexed: 11/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) exacts a heavy disease burden and is currently the second most common cause of cancer-related deaths worldwide. HCC usually lacks obvious symptoms in the early stage, and most HCC patients are diagnosed at advanced stages with poor prognosis. Circular RNAs (circRNAs) are single-stranded RNAs that form covalently closed loops and are stable in exosomes. Exosomes are known as important messengers of the cross-talk between tumor and immune cells. Accumulating studies have demonstrated the promoter or suppressor roles of exosomal circRNAs in the carcinogenesis, progression, and metastasis of HCC. In this review, we summarized the current studies on the biological functions and diagnostic and prognostic values of exosomal circRNAs in HCC progression.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, Hangzhou, China
| | | |
Collapse
|
38
|
Martinez-Arroyo O, Ortega A, Forner MJ, Cortes R. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Non-Coding RNA Therapeutic Vehicles in Autoimmune Diseases. Pharmaceutics 2022; 14:pharmaceutics14040733. [PMID: 35456567 PMCID: PMC9028692 DOI: 10.3390/pharmaceutics14040733] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/17/2022] [Accepted: 03/26/2022] [Indexed: 02/07/2023] Open
Abstract
Autoimmune diseases (ADs) are characterized by the activation of the immune system against self-antigens. More common in women than in men and with an early onset, their incidence is increasing worldwide, and this, combined with their chronic nature, is contributing to an enlarged medical and economic burden. Conventional immunosuppressive agents are designed to alleviate symptoms but do not constitute an effective therapy, highlighting a need to develop new alternatives. In this regard, mesenchymal stem cells (MSCs) have demonstrated powerful immunosuppressive and regenerative effects. MSC-derived extracellular vesicles (MSC-EVs) have shown some advantages, such as less immunogenicity, and are proposed as novel therapies for ADs. In this review, we summarize current perspectives on therapeutic options for ADs based on MSCs and MSC-EVs, focusing particularly on their mechanism of action exerted through their non-coding RNA (ncRNA) cargo. A complete state-of-the-art review was performed, centralized on some of the most severe ADs (rheumatoid arthritis, autoimmune type 1 diabetes mellitus, and systemic lupus erythematosus), giving evidence that a promising field is evolving to overcome the current knowledge and provide new therapeutic possibilities centered on MSC-EVs and their role as ncRNA delivery vehicles for AD gene therapy.
Collapse
Affiliation(s)
- Olga Martinez-Arroyo
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (O.M.-A.); (M.J.F.)
| | - Ana Ortega
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (O.M.-A.); (M.J.F.)
- Correspondence: (A.O.); (R.C.); Tel.: +34-96398-3916 (R.C.); Fax: +34-96398-7860 (R.C.)
| | - Maria J. Forner
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (O.M.-A.); (M.J.F.)
- Internal Medicine Unit, Hospital Clinico Universitario, 46010 Valencia, Spain
| | - Raquel Cortes
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (O.M.-A.); (M.J.F.)
- Correspondence: (A.O.); (R.C.); Tel.: +34-96398-3916 (R.C.); Fax: +34-96398-7860 (R.C.)
| |
Collapse
|
39
|
Zhao J, Wei K, Chang C, Xu L, Jiang P, Guo S, Schrodi SJ, He D. DNA Methylation of T Lymphocytes as a Therapeutic Target: Implications for Rheumatoid Arthritis Etiology. Front Immunol 2022; 13:863703. [PMID: 35309322 PMCID: PMC8927780 DOI: 10.3389/fimmu.2022.863703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that can cause joint damage and disability. Epigenetic variation, especially DNA methylation, has been shown to be involved in almost all the stages of the pathology of RA, from autoantibody production to various self-effector T cells and the defects of protective T cells that can lead to chronic inflammation and erosion of bones and joints. Given the critical role of T cells in the pathology of RA, the regulatory functions of DNA methylation in T cell biology remain unclear. In this review, we elaborate on the relationship between RA pathogenesis and DNA methylation in the context of different T cell populations. We summarize the relevant methylation events in T cell development, differentiation, and T cell-related genes in disease prediction and drug efficacy. Understanding the epigenetic regulation of T cells has the potential to profoundly translate preclinical results into clinical practice and provide a framework for the development of novel, individualized RA therapeutics.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Steven J Schrodi
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China.,Arthritis Institute of Integrated Traditional and Western medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
40
|
王 杰, 刘 健, 文 建, 王 馨. [Triptolide inhibits inflammatory response and migration of fibroblast like synovial cells in rheumatoid arthritis through the circRNA 0003353/JAK2/STAT3 signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:367-374. [PMID: 35426800 PMCID: PMC9010992 DOI: 10.12122/j.issn.1673-4254.2022.03.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effect of triptolide (TPL) on inflammatory response and migration of fibroblast like synovial cells (FLS) in rheumatoid arthritis (RA-FLS) and the mechanism of circular noncoding RNA (circRNA) 0003353 for mediating this effect. METHODS We collected peripheral blood mononuclear cells (PBMCs) and serum samples from 50 hospitalized RA patients and 30 healthy individuals for detecting the expression of circRNA 0003353, immune and inflammatory indexes (ESR, CRP, RF, anti-CCP, IgA, IgG, IgM, C3, and C4) and DAS28 score. Cultured RA-FLS was treated with 10 ng/mL TPL and transfected with a circRNA 0003353 overexpression plasmid, and cell counting kit-8 (CCK-8) assay and Transwell assay were used to detect the changes in the viability and migration of the cells. Enzyme-linked immunosorbent assay (ELISA) was used to examine the cytokines IL-4, IL-6, and IL-17, and real-time fluorescence quantitative PCR (RT-qPCR) was performed to detect the expression of circRNA 003353; Western blotting was used to detect the expressions of p-JAK2, pSTAT3, JAK2 and STAT3 proteins in the treated cells. RESULTS The expression of circRNA 0003353 was significantly increased in PBMCs from RA patients and showed a good performance in assisting the diagnosis of RA (AUC=90.5%, P < 0.001, 95% CI: 0.83-0.98). CircRNA 0003353 expression was positively correlated with ESR, RF and DAS28 (P < 0.05). Treatment with TPL significantly decreased the expression of circRNA 0003353, suppressed the viability and migration ability, decreased the expressions of IL-6 and IL-17, and increased the expression IL-4 in cultured RA-FLS in a time-dependent manner (P < 0.01). TNF-α stimulation of RA-FLS significantly increased the ratios of p-JAK2/JAK2 and p-STAT3/STAT3, which were obviously lowered by TPL treatment (P < 0.01). TPL-treated RA-FLS overexpressing circRNA 0003353 showed significantly increased cell viability and migration ability with decreased IL-4 expression and increased IL-6 and IL-17 expressions and ratios of p-JAK2/ JAK2 and p-STAT3/STAT3 (P < 0.01). CONCLUSION The expression of circRNA 0003353 is increased in PBMCs in RA patients and in RA-FLS. TPL treatment can regulate JAK2/STAT3 signal pathway and inhibit the inflammatory response and migration of RA-FLS through circRNA 0003353.
Collapse
Affiliation(s)
- 杰 王
- />安徽中医药大学第一附属医院风湿科,安徽 合肥 230031Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - 健 刘
- />安徽中医药大学第一附属医院风湿科,安徽 合肥 230031Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - 建庭 文
- />安徽中医药大学第一附属医院风湿科,安徽 合肥 230031Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - 馨 王
- />安徽中医药大学第一附属医院风湿科,安徽 合肥 230031Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| |
Collapse
|