1
|
Promila L, Sarkar K, Guleria S, Rakshit A, Rathore M, Singh NC, Khan S, Tomar MS, Ammanathan V, Barthwal MK, Kumaravelu J, Shrivastava A, Mitra K, Guha R, Aggarwal A, Lahiri A. Mitochondrial calcium uniporter regulates human fibroblast-like synoviocytes invasion via altering mitochondrial dynamics and dictates rheumatoid arthritis pathogenesis. Free Radic Biol Med 2025; 234:55-71. [PMID: 40188890 DOI: 10.1016/j.freeradbiomed.2025.04.008] [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: 10/21/2024] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease that currently has no cure. Fibroblast-like synoviocytes (FLS), present in the RA synovium, play a pivotal role in RA pathogenesis. Notably, FLS in the RA patients (RA-FLS) exhibit characteristics similar to cancer cells, like enhanced migration, invasiveness, uncontrolled proliferation, resistance to apoptosis, and metabolic reprogramming. RA-FLS invasiveness is linked to radiographic joint damage in the patients, whereas inhibiting the FLS migration mitigates disease pathology. However, the molecular mechanisms underlying the migration and invasion capabilities of RA-FLS are not entirely understood. In this work, we have explored the function of mitochondrial calcium uniporter (MCU) and calcium signaling in FLS invasion. Our findings demonstrate a positive correlation between MCU expression and RA disease score. Interestingly, mitochondrial size was reduced, and peripheral localization was more pronounced in the RA-FLS when compared to the control FLS. Mitochondrial calcium import inhibition in the FLS by specific MCU inhibitor, Ruthenium-360 restored these altered mitochondrial dynamics and reduced the invasive phenotype. Through unbiased transcriptome analysis, we identified that MCU-mediated calcium signaling in RA-FLS leads to the enriched actin cytoskeleton and focal adhesion pathways responsible for the invasion phenotype, which can be effectively suppressed by inhibiting MCU. Additionally, we found that mitochondrial transport facilitator Miro1 binds to MCU in a calcium-dependent manner and regulates MCU-mediated mitochondrial dynamics and RA-FLS invasion. Experiments utilizing mice xenograft model demonstrated that MCU silencing diminishes the migration of RA-FLS toward the sites of inflammation in the immunocompromised SCID mice. Altogether, our findings highlight MCU as a promising therapeutic target to inhibit RA-FLS migration and RA progression.
Collapse
Affiliation(s)
- Lakra Promila
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kabita Sarkar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shivika Guleria
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Adrija Rakshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Manisha Rathore
- Lab Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nishakumari C Singh
- Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shaziya Khan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | | | - Veena Ammanathan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jagavelu Kumaravelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | | | - Kalyan Mitra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajdeep Guha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Lab Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amita Aggarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Amit Lahiri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Liao H, Zheng J, Lu J, Shen HL. NF-κB Signaling Pathway in Rheumatoid Arthritis: Mechanisms and Therapeutic Potential. Mol Neurobiol 2025; 62:6998-7021. [PMID: 39560902 DOI: 10.1007/s12035-024-04634-2] [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: 03/11/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that imposes a heavy economic burden on patients and society. Bone and cartilage destruction is considered an important factor leading to RA, and inflammation, oxidative stress, and mitochondrial dysfunction are closely related to bone erosion and cartilage destruction in RA. Currently, there are limitations in the clinical treatment methods for RA, which urgently necessitates finding new effective treatments for patients. Nuclear transcription factor-κB (NF-κB) is a signaling transcription factor that is widely present in various cells. It plays an important role as a stress source in the cellular environment and regulates gene expression in processes such as immunity, inflammation, cell proliferation, and apoptosis. NF-κB has long been recognized as a pathogenic factor of RA, and its activation can exacerbate RA by promoting inflammation, oxidative stress, mitochondrial dysfunction, and bone destruction. Conversely, inhibiting the activity of the NF-κB pathway effectively inhibits these pathological processes, thereby alleviating RA. Therefore, NF-κB may be a potential therapeutic target for RA. This article describes the physiological structure of NF-κB and its important role in RA through the regulation of oxidative stress, inflammatory response, mitochondrial function, and bone destruction. Meanwhile, we also summarized the impact of NF-κB crosstalk with other signaling pathways on RA and the effect of related drugs or inhibitors targeting NF-κB on RA. The purpose of this article is to provide evidence for the role of NF-κB in RA and to emphasize its significant role in RA by elucidating the mechanisms, so as to provide a theoretical basis for targeting the NF-κB pathway as a treatment for RA.
Collapse
Affiliation(s)
- Haiyang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jianxiong Zheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jinyue Lu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Hai-Li Shen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
3
|
Wang H, Sun X, Cui K, Wang Y, Sun Y. Ubiquitin specific peptidase 13 protects against inflammation-associated joint injury in collagen-induced rheumatoid arthritis mice by targeting TRAF6. Int Immunopharmacol 2025; 156:114617. [PMID: 40252462 DOI: 10.1016/j.intimp.2025.114617] [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: 08/08/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease with complex pathogenesis. Ubiquitin-specific protease 13 (USP13), a member of the deubiquitinating enzyme (DUB) superfamily, plays diverse roles in cellular events. This study investigates the role of USP13 in RA, revealing its significant downregulation in peripheral blood mononuclear cells (PBMCs) from RA patients compared to healthy individuals. USP13 expression negatively correlates with RA characteristics, including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and rheumatoid factor (RF). Consistent with these findings, USP13 levels were reduced in knee joint synovial tissues of collagen-induced arthritis (CIA) mice and lipopolysaccharide (LPS)-stimulated bone marrow-derived macrophages (BMDMs). Mechanistically, USP13 directly interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), reducing its K63-linked polyubiquitination and thereby inhibiting TRAF6 expression. This interaction restrains nuclear factor κB (NF-κB) signaling, leading to a marked attenuation of LPS-induced inflammatory responses in BMDMs. Importantly, the anti-inflammatory effects of USP13 over-expression are largely dependent on TRAF6 suppression. In vitro, USP13 over-expression in BMDMs inhibits the proliferation of fibroblast-like synoviocytes (FLS) and osteoclastogenesis. In addition, USP13-overexpressing BMDMs in CIA mice significantly alleviates RA development, as evidenced by reduced synovial hyperplasia, inflammatory cell infiltration, cartilage destruction, and bone loss. These findings highlight the essential role of USP13 in macrophages during RA progression and reveal its therapeutic potential by targeting TRAF6 signaling.
Collapse
Affiliation(s)
- Haili Wang
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Xinyi Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Kai Cui
- Department of Oncology, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Yan Wang
- Geriatric Department, Jilin Integrated Traditional Chinese and Western Medicine Hospital of Jilin, Jilin 132011, China
| | - Yuman Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China.
| |
Collapse
|
4
|
Xu HY, Jiang MT, Yang YF, Huang Y, Yang WD, Li HY, Wang X. Microalgae-Based Fucoxanthin Attenuates Rheumatoid Arthritis by Targeting the JAK-STAT Signaling Pathway and Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11708-11719. [PMID: 40325616 DOI: 10.1021/acs.jafc.4c12474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Fucoxanthin, an abundant carotenoid in marine algae, has garnered attention for its diverse health benefits, including anti-inflammatory and anticancer properties. Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joint inflammation and damage. This study investigated the therapeutic potential of fucoxanthin extracted from Phaeodactylum tricornutum in collagen-induced RA. Our results demonstrated that fucoxanthin significantly alleviated RA symptoms, including weight loss, joint swelling, and decreased appetite. Histological analysis revealed that fucoxanthin mitigated synovial inflammation, cartilage damage, and bone erosion. Mechanistically, transcriptomic analysis and cell experiments indicated that fucoxanthin suppressed the JAK-STAT signaling pathway by downregulating the expression of inflammatory cytokines, such as IL-6 and IL-1β. Furthermore, metagenomic analysis suggested that fucoxanthin restored the altered gut microbiota composition associated with RA. These findings highlight the therapeutic potential of fucoxanthin from P. tricornutum in the management of RA by targeting multiple pathways, including inflammation and gut microbiota.
Collapse
Affiliation(s)
- Hui-Ying Xu
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Meng-Ting Jiang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yu-Feng Yang
- Biotechnology and Nuclear Technology Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China
| | - Yisha Huang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Wei-Dong Yang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hong-Ye Li
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiang Wang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
5
|
Qiu F, Xie D, Chen H, Wang Z, Huang J, Cao C, Liang Y, Yang X, He DY, Fu X, Lu A, Liang C. Generation of cytotoxic aptamers specifically targeting fibroblast-like synoviocytes by CSCT-SELEX for treatment of rheumatoid arthritis. Ann Rheum Dis 2025; 84:726-745. [PMID: 39237134 DOI: 10.1136/ard-2024-225565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an autoimmune disease characterised by aggressive fibroblast-like synoviocytes (FLSs). Very few RA patients-derived FLSs (RA-FLSs)-specific surface signatures have been identified, and there is currently no approved targeted therapy for RA-FLSs. This study aimed to screen therapeutic aptamers with cell-targeting and cytotoxic properties against RA-FLSs and to uncover the molecular targets and mechanism of action of the screened aptamers. METHODS We developed a cell-specific and cytotoxic systematic evolution of ligands by exponential enrichment (CSCT-SELEX) method to screen the therapeutic aptamers without prior knowledge of the surface signatures of RA-FLSs. The molecular targets and mechanisms of action of the screened aptamers were determined by pull-down assays and RNA sequencing. The therapeutic efficacy of the screened aptamers was examined in arthritic mouse models. RESULTS We obtained an aptamer SAPT8 that selectively recognised and killed RA-FLSs. The molecular target of SAPT8 was nucleolin (NCL), a shuttling protein overexpressed on the surface and involved in the tumor-like transformation of RA-FLSs. Mechanistically, SAPT8 interacted with the surface NCL and was internalised to achieve lysosomal degradation of NCL, leading to the upregulation of proapoptotic p53 and downregulation of antiapoptotic B-cell lymphoma 2 (Bcl-2) in RA-FLSs. When administrated systemically to arthritic mice, SAPT8 accumulated in the inflamed FLSs of joints. SAPT8 monotherapy or its combination with tumour necrosis factor (TNF)-targeted biologics was shown to relieve arthritis in mouse models. CONCLUSIONS CSCT-SELEX could be a promising strategy for developing cell-targeting and cytotoxic aptamers. SAPT8 aptamer selectively ablates RA-FLSs via modulating NCL-p53/Bcl-2 signalling, representing a potential alternative or complementary therapy for RA.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Xu Yang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong-Yi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China; Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
6
|
Luo X, Tang N, Ren Y, Li J, Zhu H, Wu S, Ding Z. Single-cell multi-dimensional data analysis decodes RNF19A-mediated drug resistance in rheumatoid arthritis fibroblast-like synoviocytes: mechanisms and biological insights. Cell Mol Life Sci 2025; 82:180. [PMID: 40293542 PMCID: PMC12037462 DOI: 10.1007/s00018-025-05707-8] [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: 11/10/2024] [Revised: 03/16/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Rheumatoid arthritis (RA) is a prevalent autoimmune disease, affecting approximately 1% of the global population. Methotrexate (MTX) is the most widely prescribed drug for RA treatment; however, its efficacy is often limited, with resistance frequently observed. Fibroblast-like synoviocytes (FLS) play a pivotal role in RA progression and are closely linked to drug resistance, although the underlying mechanisms remain poorly understood. In this study, we conducted a comprehensive analysis of public single-cell transcriptomics data from osteoarthritis and rheumatoid arthritis synovial tissues, identifying RNF19A as a gene potentially associated with RA resistance in FLS. Our findings indicate that RNF19A is significantly overexpressed in drug-resistant FLS and is closely associated with the dysregulation of FLS proliferation, migration, invasion, and apoptosis. Furthermore, we demonstrated that RNF19A promotes functional disruption in FLS by ubiquitinating and degrading MKP-1, thereby activating the MAPK signaling pathway. This activation also facilitates the nuclear translocation of ZBTB20, an upstream transcription factor of RNF19A, which further enhances RNF19A transcription. This biological process creates a positive feedback loop in FLS, contributing to RA resistance-a mechanism that was also validated in vivo. In summary, this study is the first to underscore the crucial role of RNF19A in mediating drug resistance in RA FLS, elucidating the underlying biological processes, and providing novel insights into RA pathogenesis, thereby offering a new experimental foundation for RA drug development.
Collapse
Affiliation(s)
- Xin Luo
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ning Tang
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yijun Ren
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingchen Li
- Department of Food Engineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Huancheng Zhu
- Lab of Research and Engineering of Cell Therapy Technology, Hangzhou Institute of Medicine Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Song Wu
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiyu Ding
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Lab of Research and Engineering of Cell Therapy Technology, Hangzhou Institute of Medicine Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Huang YH, Qiu L, Peng Y, Cai R, Xiao QQ, Li MC, Xu LJ, Fu Q. Ecdysteroid-enriched fraction of Cyathula officinalis suppresses synovial proliferation and inflammation to ameliorate RA by inhibiting the AKT/PI3K/mTOR signaling pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-23. [PMID: 40257367 DOI: 10.1080/10286020.2025.2492357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
This study investigated the chemical compositions isolated from Cyathula officinalis (CES) and elucidated its anti-RA effects and potential mechanisms. In vitro results revealed that CES suppressed interleukin (IL)-1β-stimulated proliferation, migration, and invasion of MH7A cells in a concentration-dependent manner. In vivo results indicated that CES could reduce the arthritis score, paw swelling, and improve histopathological deterioration. The levels of inflammatory factors, matrix metalloproteinases, and proteins expressed in rat synovial tissue were suppressed after treatment with CES. These findings illustrate that CES is involved in the PI3K/AKT/mTOR/SREBP1/SCD-1/GPX4 axis-mediated ferroptosis and anti-inflammation, exerting its protective effects against the progression of RA.
Collapse
Affiliation(s)
- Yue-Hui Huang
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
- School of Traditional Chinese Medicine and Nutritional Health Care, Meishan Pharmaceutical College, Meishan620000, China
| | - Lu Qiu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Yi Peng
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Rui Cai
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Qing-Qing Xiao
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Mei-Chen Li
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Li-Jia Xu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| | - Qiang Fu
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu610106, China
| |
Collapse
|
8
|
Haroon MM, Hegazy GA, Hassanien MA, Shaker OG, Labib S, Hussein WH. Expression of lncRNA NEAT1, miR-21, and IL17 in Rheumatoid Arthritis Patients. Biologics 2025; 19:201-211. [PMID: 40256764 PMCID: PMC12009040 DOI: 10.2147/btt.s519558] [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: 01/30/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Background Rheumatoid arthritis (RA) is a relatively frequent autoimmune disorder with individual and socioeconomic burden, particularly if diagnosed late. Therefore, identifying novel biomarkers for RA that assist in early diagnosis and managing plan is essential. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1), micro-RNA 21 (miR-21) and interleukin 17 (IL17) have emerging roles in the pathogenesis of numerous inflammatory conditions. The present research aims to evaluate NEAT1, miR-21 and IL17 roles in RA manifestations and activity and the possibility of utilizing them as biomarkers or therapeutic targets for the disease. Therefore, expression levels of NEAT1, miR-21 and IL17 in sera of 100 RA cases, and 100 age and sex-matched healthy controls were compared. A subsequent analysis was conducted to examine the correlation of their levels to various RA manifestations and disease activity. Results Both NEAT1 and IL17 were significantly up regulated, while miR-21 was significantly down regulated in cases compared to controls. NEAT1 demonstrated a significant positive correlation with tender and swollen joint counts and with the overall DAS-28 score. A significant negative correlation was noted between miR-21 and RA disease duration. Conclusion NEAT1, miR-21, and IL17 have differential levels in patients with RA where NEAT1 and IL17 have up regulation, while miR-21 has down regulation. NEAT1 has a significant correlation with RA disease activity. We recommend further research to determine if they could be useful as future biomarkers for RA.
Collapse
Affiliation(s)
- Maysa M Haroon
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Gehan A Hegazy
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Medical Biochemistry Department, Medical Research and clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Mohammed A Hassanien
- Pharmacy Practice Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Safa Labib
- Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Wafaa H Hussein
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Xiao C, Su Z, Zhao J, Tan S, He M, Li Y, Liu J, Xu J, Hu Y, Li Z, Fan C, Liu X. Novel regulation mechanism of histone methyltransferase SMYD5 in rheumatoid arthritis. Cell Mol Biol Lett 2025; 30:38. [PMID: 40165083 PMCID: PMC11959843 DOI: 10.1186/s11658-025-00707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Fibroblast-like synoviocytes (FLS) are crucial for maintaining synovial homeostasis. SMYD5, a member of the histone lysine methyltransferase subfamily SMYDs, is involved in many pathological processes. This study aimed to investigate the role of SMYD5 in regulating synovial fibroblast homeostasis and the pathogenesis of rheumatoid arthritis (RA). METHODS Proteomic screening was conducted to assess SMYD5 expression in the synovium of patients with osteoarthritis (OA) and RA. In vitro, interleukin-1 beta (IL-1β) was used to induce proliferation and inflammation in FLS. Further, we performed loss-of-function and gain-of-function experiments to investigate the biological function of SMYD5. In vivo, adeno-associated virus (AAV) vectors carrying SMYD5 short-hairpin RNA (AAV-shSMYD5) were injected into the knee joints to knock down SMYD5 in a collagen-induced arthritis (CIA) mouse model to evaluate its role in joint damage. RESULTS We observed a significant elevation of SMYD5 expression in the synovial tissues of patients with RA and IL-1β-induced FLS. SMYD5 facilitated posttranslational modifications and activated downstream signaling pathways, thereby promoting proliferation and inflammation in FLS. Mechanistically, SMYD5 mediated the methylation of Forkhead box protein O1 (FoxO1), which accelerated its degradation through ubiquitination, resulting in substantial FLS proliferation. Additionally, SMYD5 promoted lactate release to activate NF-κB signaling pathways by upregulating hexokinases-2 (HK2) expression, a key glycolytic enzyme, thereby intensifying the inflammatory response in FLS. Supporting these findings, intraarticular delivery of AAV-mediated SMYD5 knockdown in the CIA mice model effectively alleviated joint swelling, bone erosion, and overall arthritis severity. CONCLUSIONS Together, these findings suggest that SMYD5 is a dual target for regulating synovial fibroblast homeostasis and the pathogenesis of RA. Targeting SMYD5 through local treatment strategies may provide a novel therapeutic approach for RA, particularly when combined with immunotherapy.
Collapse
Affiliation(s)
- Chenxi Xiao
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Zhenghua Su
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Jialin Zhao
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Subei Tan
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Mengting He
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Yuhui Li
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Jiayao Liu
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Jie Xu
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Yajie Hu
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China
| | - Zhongzheng Li
- The 9th Hospital of Ningbo, 68, Xiangbei Road, Jiangbei District, Ningbo, 315020, Zhejiang, China.
| | - Chunxiang Fan
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China.
| | - Xinhua Liu
- Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Phenome Research Center of TCM, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China.
| |
Collapse
|
10
|
Wang H, Zhao YC, Xu L, Zhang TJ, Liu LH, Zhou MQ, Zhang H, Yang YN, Pan P, Jin L, Zhang ZW, Zhang XZ, Zhang LL. HIF-1α mediates mitochondrial damage by down-regulating ALKBH7 expression to promote the aberrant activation of FLS in rheumatoid arthritis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01520-y. [PMID: 40140527 DOI: 10.1038/s41401-025-01520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/23/2025] [Indexed: 03/28/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation and progressive joint destruction. Existing evidence indicates that hypoxia potentially contributes to the pathology of RA, though the specific mechanism remains unidentified. In this study, we explored the molecular mechanism through which the hypoxia-inducible factor (HIF-1α) contributed to the pathological process of RA. Our preliminary results suggested that hypoxia stimulates the activation of fibroblast-like synoviocytes (FLS) by inducing mitochondrial damage to activate cGAS-STING signaling, which can be effectively inhibited by silencing HIF-1α. In line with this, HIF-1α deficiency significantly alleviated the symptoms of collagen-induced arthritis (CIA) mice. RNA-Seq and CUT-Tag analysis revealed that HIF-1α down-regulated the expression of AlkB homologue 7 (ALKBH7) by acting on the ALKBH7 promoter site on chromosome 19 6372400-6372578. Using dual luciferase reporter analysis, we identified that ACCGTGGC as the motif to which HIF-1α bound directly. Subsequently, we demonstrated that knockdown of ALKBH7 induces mitochondrial damage and activates cGAS-STING signaling by downregulating the expression of UQCRC2. Conversely, overexpression of ALKBH7 could resist hypoxia-induced mitochondrial damage and FLS activation. In conclusion, HIF-1α triggers mitochondrial damage by downregulating the expression of ALKBH7 thereby promoting FLS activation, which may be the molecular mechanism by which hypoxia is involved in the pathological process of RA. Hypoxia promotes the activation of FLS through the induction of mitochondrial damage, which subsequently activates cGAS-STING signaling. Mechanistically, HIF-1α triggers mitochondrial damage by downregulating the expression of ALKBH7 in a target manner. Furthermore, the deletion of ALKBH7 leads to mitochondrial damage under hypoxic conditions, primarily through the downregulation of UQCRC2, as opposed to other complexes.
Collapse
Affiliation(s)
- Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Yu-Chen Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Li Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Tian-Jing Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Liang-Hu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Meng-Qi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Han Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Yin-Ning Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Pin Pan
- The Second People's Hospital of Hefei, Hefei, 230011, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Zi-Wei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Xian-Zheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China.
- Department of Oncology, The First Affiliated Hospital, Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China.
| | - Ling-Ling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China.
| |
Collapse
|
11
|
Chen L, Xu L, Zhang Y, Xia H. Dioscin alleviates the dysfunction of fibroblast-like synoviocytes by circ_0008267/miR-942-5p/FKBP5 axis during rheumatoid arthritis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03872-y. [PMID: 40116870 DOI: 10.1007/s00210-025-03872-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/31/2025] [Indexed: 03/23/2025]
Abstract
Dioscin is a natural, bioactive steroid saponin that has the antiarthritic activity. Circular RNAs (circRNAs) are stable noncoding RNAs involving in the pathogenesis of rheumatoid arthritis (RA). Here, this study aimed to probe the role and mechanism of dioscin and circ_0008267 in RA progression. Cell proliferation, apoptosis, invasive, and migratory abilities, as well as inflammatory response were evaluated by CCK-8 assay, EdU assay, flow cytometery, transwell assay, wound healing assay, and ELISA analysis, respectively. Levels of genes and protein were tested by qRT-PCR and western blotting. The interaction between miR-942-5p and circ_0008267 or FK506-binding protein 5 (FKBP5) was confirmed using dual-luciferase reporter and RNA pull-down assays. Dioscin treatment was demonstrated to suppress RA-FLS proliferation, invasion, migration, and inflammatory response, but induced cell apoptosis. Circ_0008267 is a stable circRNA, and was increased in RA samples. Moreover, its expression was reduced by dioscin in RA-FLS, overexpression of circ_0008267 reversed the effects of dioscin on RA-FLS. Mechanistically, circ_0008267 acted as a sponge for miR-942-5p, which targeted FKBP5. Dioscin reduced FKBP5 expression, but elevated miR-942-5p level in RA-FLS. MiR-942-5p inhibition or FKBP5 upregulation abolished the inhibitory effects of dioscin on RA-FLS dysfunction. Moreover, circ_0008267 deficiency impaired RA-FLS proliferation, invasion, migration, and inflammation through regulating FKBP5. Dioscin suppressed the proliferation, invasion, migration, and inflammatory response in RA-FLS via circ_0008267/miR-942-5p/FKBP5 axis, providing new insights for RA prevention.
Collapse
Affiliation(s)
- Lifeng Chen
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, 430070, Hubei, China.
| | - Li Xu
- Department of Cardiovascular Medicine, Guiqian International General Hospital, Guiyang City, 550024, Guizhou Province, China
| | - Yujing Zhang
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, 430070, Hubei, China
| | - Hao Xia
- Medical College of Wuhan University of Science and Technology, Wuhan, 430070, Hubei, China
| |
Collapse
|
12
|
Lin YY, Huang CC, Ko CY, Tsai CH, Chang JW, Achudhan D, Tang CH. Omentin-1 modulates interleukin expression and macrophage polarization: Implications for rheumatoid arthritis therapy. Int Immunopharmacol 2025; 149:114205. [PMID: 39908806 DOI: 10.1016/j.intimp.2025.114205] [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: 11/24/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic inflammatory and autoimmune disorder in which monocytes/macrophage infiltrate synovial membrane, differentiating into the pro- and anti-inflammatory M1 and M2 macrophage phenotypes. Omentin-1 is one of the adipokines that has anti-inflammatory and immunomodulatory effects; nevertheless, investigators have yet to elucidate the function of omentin-1 in RA development. It is still unclear how omentin-1 affects human autoimmune disease and what its beneficial role is. Thus, we show that omentin-1 exhibits a therapeutic effect on RA. METHODS Utilizing patient or animal tissue, MH7A cell-line, ELISA, and qPCR, we examined the expression of omentin-1 and inflammatory cytokines in the GEO databases. Omentin-1's effects on macrophage polarization were investigated using Immunofluorescence staining (IF) and qPCR. Additionally, the method by which omentin-1 regulates interleukins was discovered by IF labeling for STAT6 translocation, siRNA transfection, IPA software using several and pharmacological inhibitors. Omentin-1's effects were examined in an in vivo investigation using the type II collagen-induced arthritis model, micro-CT, and histological evaluation. RESULTS Results from the GSE97779 dataset and patients' tissues discovered that the level of omentin-1 and M2 macrophage markers are downregulated in human RA tissue samples compared to healthy tissue and negatively correlated with the expression of pro-inflammatory interleukins (ILs) and M1 macrophage. Stimulation of RA synovial fibroblasts with omentin-1 augmented IL-4 synthesis and subsequently enhanced anti-inflammatory ability as well as M2 polarization. The STAT6 transactivation through AMPK, PI3K, ERK, and JAK cascades regulates omentin-1-induced promotion of IL-4. Importantly, intra-articular injection of omentin-1 blocked collagen-induced arthritis-augmented pro-inflammatory response, cartilage degradation, and bone loss through upregulating IL-4 and M2 macrophages in vivo. CONCLUSION Our findings support a potential therapy goal for RA and a tenable mechanism to explain the relationship between omentin-1.
Collapse
Affiliation(s)
- Yen-You Lin
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung, Taiwan; Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Ko
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Jun-Way Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - David Achudhan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
13
|
Xia X, He C, Xue Z, Wang Y, Qin Y, Ren Z, Huang Y, Luo H, Chen HN, Zhang WH, Huang LB, Shi Y, Bai Y, Cai B, Wang L, Zhang F, Qian M, Zhang W, Shu Y, Yin G, Xu H, Xie Q. Single cell immunoprofile of synovial fluid in rheumatoid arthritis with TNF/JAK inhibitor treatment. Nat Commun 2025; 16:2152. [PMID: 40038288 PMCID: PMC11880340 DOI: 10.1038/s41467-025-57361-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Numerous patients with rheumatoid arthritis (RA) manifest severe syndromes, including elevated synovial fluid volumes (SF) with abundant immune cells, which can be controlled by TNF/JAK inhibitors. Here, we apply single-cell RNA sequencing (scRNA-seq) and subsequent validations in SF from RA patients. These analyses of synovial tissue show reduced density of SF-derived pathogenic cells (e.g., SPP1+ macrophages and CXCL13+CD4+ T cells), altered gene expression (e.g., SPP1 and STAT1), molecular pathway changes (e.g., JAK/STAT), and cell-cell communications in drug-specific manners in samples from patients pre-/post-treated with adalimumab/tofacitinib. Particularly, SPP1+ macrophages exhibit pronounced communication with CXCL13+CD4+ T cells, which are abolished after treatment and correlate with treatment efficacy. These pathogenic cell types alone or in combination can augment inflammation of fibroblast-like synoviocytes in vitro, while conditional Spp1 knocking-out reduces RA-related cytokine expression in collagen-induced arthritis mice models. Our study shows the functional role of SF-derived pathogenic cells in progression and drug-specific treatment outcomes in RA.
Collapse
Affiliation(s)
- Xuyang Xia
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenjia He
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhinan Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuelan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Qin
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixiang Ren
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yupeng Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Han Luo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Han Zhang
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunying Shi
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangjuan Bai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lanlan Wang
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Tianfu Jincheng Laboratory, Chengdu, Sichuan, China.
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Wu D, Deng Y, Huang Y, Zhao J, Long W, Peng Y, Xiong Z, Wu R. Refining synovial inflammation assessment: A modified General Synovitis Score for active rheumatoid arthritis. Exp Ther Med 2025; 29:58. [PMID: 39927277 PMCID: PMC11803189 DOI: 10.3892/etm.2025.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/19/2024] [Indexed: 02/11/2025] Open
Abstract
The General Synovitis Score (GSS) is a well-established method for scoring synovial inflammation. Despite its widespread use, the GSS does not fully capture the inflammatory manifestations characteristic of the synovium in rheumatoid arthritis (RA). To address this limitation, the modified GSS (mGSS) was developed. The present study compared the correlation of the mGSS and the GSS with clinical disease activity. The aim was to provide a more precise histopathological scoring system based on hematoxylin and eosin (H&E) staining for assessing synovial inflammation in patients with active RA. In this cross-sectional study, synovial tissues were obtained from 60 patients with RA using a novel synovial biopsy device. Sections from synovial tissues were stained with H&E, and were assessed using the GSS and the mGSS. Neovascularization was observed in 56 patients (93.3%) and was significantly correlated with disease activity score in 28 joints-C-reactive protein (DAS28-CRP) (ρ=0.49; P<0.001), erythrocyte sedimentation rate (ESR) (ρ=0.44; P<0.001) and CRP (ρ=0.51; P<0.001). In addition, patients with severe neovascularization had significantly higher DAS28-CRP, ESR and CRP levels than those with mild-to-moderate neovascularization (P<0.05). Synoviocyte detachment, which occurred in nine patients (15.0%), was associated with higher DAS28-CRP, ESR and CRP levels than in the patients with synoviocyte proliferation (P<0.05). Furthermore, the mGSS was more strongly correlated with DAS28-CRP (ρ=0.62; P<0.001) than the GSS (ρ=0.37; P=0.003). These findings indicated that neovascularization and synoviocyte detachment, which are critical yet often overlooked aspects in the traditional GSS, are important in RA. Incorporating these elements into the mGSS may enhance the assessment of disease activity, providing a more precise and accurate evaluation of the synovial histopathology in patients with RA.
Collapse
Affiliation(s)
- Dengfeng Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yihan Deng
- First Clinical Medical School, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yiping Huang
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wei Long
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yilin Peng
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhenfang Xiong
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Cai Y, Qiu R, Huang Q, Lai W, Han Y, Lu X, Qin J, Ouyang Q, Yang M. Circ_0088200 acts as a sponge for miR-127-5p to promote the migration and invasion of rheumatoid arthritis fibroblast-like synoviocytes. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2025; 6:7-20. [PMID: 40191465 PMCID: PMC11966198 DOI: 10.1515/rir-2025-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/04/2025] [Indexed: 04/09/2025]
Abstract
Background Circular RNAs (circRNAs) play a crucial role in the development of various diseases. However, few studies have investigated the role of circRNAs in rheumatoid arthritis (RA). Herein, we aimed to identified the novel circRNAs involved in the migration and invasion of RA fibroblast-like synoviocytes (RA-FLS). Methods The RA-FLS were isolated from the synovial membrane of patients with RA. The CircRNA profile was screened by CircRNA microarray analysis. Circ_0088200 and miR-127-5p expression levels were detected using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). The protein level of matrix metalloproteinase 1 (MMP1) was evaluated by western blotting. Wound healing and Transwell assays were performed to analyze the migration and invasion of RA-FLS. RNA immunoprecipitation (RIP) and dual-luciferase reporter assays were used to validate the interaction between Circ_0088200 and miR-127-5p. Collagen-induced arthritis (CIA) mouse models were established to evaluate the role of Circ_0088200 in the development of arthritis in vivo. Results Circ_0088200 was highly expressed in RA-FLS compared with osteoarthritis fibroblast-like synoviocytes (OAFLS) and correlated positively with the disease activity score in 28 joints. Inhibition of Circ_0088200 suppressed the migration and invasion of RA-FLS. Conversely, overexpression of Circ_0088200 significant promoted the migration and invasion of RA-FLS. Mechanistically, Circ_0088200 functions as a sponge for miR-127-5p and relieve its repressive effect on MMP1, thereby promoting the migration and invasion of RA-FLS. Importantly, intra-articular injection of Adenoassociated virus expressing Circ_0088200 significantly increased the severity of arthritis in mice with CIA. Conclusion Circ_0088200 promotes the migration and invasion of RA-FLS by sponging miR-127-5p. Thus Circ_0088200 is a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Yujie Cai
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Rong Qiu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Qin Huang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Weinan Lai
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Yipeng Han
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Xiaoxi Lu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Jiayu Qin
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Qingqing Ouyang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| | - Min Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510510, Guangdong Province, China
| |
Collapse
|
16
|
Ma Y, Lin H, Li Y, An Z. Amentoflavone Induces Ferroptosis to Alleviate Proliferation, Migration, Invasion and Inflammation in Rheumatoid Arthritis Fibroblast-like Synoviocytes by Inhibiting PIN1. Cell Biochem Biophys 2025; 83:1299-1312. [PMID: 39354278 DOI: 10.1007/s12013-024-01563-8] [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] [Accepted: 09/17/2024] [Indexed: 10/03/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease that is prevalent worldwide and seriously threatens human health. RA-fibroblast-like synoviocytes (FLS) play important roles in almost all aspects of RA progression. This study aimed to study the effect of Amentoflavone (AMF), a polyphenol compound derived from extracts of Selaginella tamariscina, on the abnormal biological behaviors of RA-FLS. The immortalized human RA-FLS cell line (MH7A) was treated with AMF or transfected with small interfering RNAs (siRNAs) targeting peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1). Then, cell viability was detected by CCK-8 assay. EDU staining, wound healing and transwell assays were employed to measure the capacities of MH7A cell proliferation, migration and invasion. The levels of inflammatory factors were assessed using ELISA kits. Additionally, ferroptosis was analyzed by detecting Fe2+ content, lipid reactive oxygen species (ROS) level and expression of ferroptosis-related proteins. Pull-down assay was employed to verify the targeted binding of AMF to PIN1. Further, PIN1 overexpression or ferroptosis inhibitor Ferrostatin-1 (Fer-1) addition was conducted to elucidate the regulatory mechanism of AMF on PIN1 and ferroptosis. Results revealed that AMF intervention or PIN1 knockdown inhibited the proliferation, migration, invasion and inflammation in MH7A cells. AMF facilitated lipid peroxidation and ferroptosis in MH7A cells. Moreover, AMF targeted inhibition of PIN1 expression, and PIN1 overexpression restored the promoting effect of AMF on lipid peroxidation and ferroptosis in MH7A cells. Besides, Fer-1 reversed the impacts of AMF on the abnormal biological behaviors of MH7A cells. In summary, AMF induced ferroptosis to inhibit the proliferation, migration, invasion and inflammation in RA-FLS by inhibiting PIN1, providing a promising candidate for RA treatment.
Collapse
Affiliation(s)
- Yan Ma
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, 100020, Beijing, People's Republic of China
| | - Hongjun Lin
- Henan Institute for Drug and Medical Device Inspection, Zhengzhou, 450003, Henan, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, Jiangsu, People's Republic of China.
| | - Zhuoling An
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, 100020, Beijing, People's Republic of China
| |
Collapse
|
17
|
Chen Q, Chen D, Wang S, Huang X, Liang L, Xie T, Lu J. RND1 Induces Ferroptosis to Alleviate Inflammatory Response, Proliferation, Invasion, and Migration of Rheumatoid Synoviocytes. J Inflamm Res 2025; 18:2647-2659. [PMID: 40008082 PMCID: PMC11853921 DOI: 10.2147/jir.s500630] [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: 10/12/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background Ferroptosis is involved in the occurrence and development of inflammatory arthritis. RND1 has been reported to possess pro-ferroptosis activity. Objective This study was designed to explore the role and the molecular mechanism of RND1 in rheumatoid arthritis (RA). Methods DBA/1 mice were exposed to type II collagen immunization. The pathological damage of the knee joints of mice was observed with H&E staining and RND1 expression in synovial tissues was detected using Western blot. In vitro, Western blot was used to measure RND1, ferroptosis-, migration- and inflammation-related proteins. The cell proliferation, migration and invasion were detected using CCK-8 method, EdU staining, wound healing and transwell assays. The levels of inflammatory factors were detected with ELISA and RT-qPCR. Relative iron level, GSH and MDA concentrations were detected with corresponding assay kits. BODIPY 581/591 C11 kit measured lipid ROS. 4-HNE and GPX4 expression were detected using immunofluorescence assay. Results This study found that RND1 expression was reduced in the synovial tissues of RA mice and human fibroblast-like MH7A synoviocytes. It was also found that the upregulation of RND1 inhibited the proliferation, migration, invasion and inflammatory response in rheumatoid synovial cells via ferroptosis. Conclusion Collectively, RND1 exerted protective impacts on RA, which might be mediated by ferroptosis.
Collapse
Affiliation(s)
- Qiuhua Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Donglan Chen
- Department of Infectious Diseases and Tropical Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Sijie Wang
- Biomedical Diagnostic Center of Ultrastructure, Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Xiaomei Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Liang Liang
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Tong Xie
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Jie Lu
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| |
Collapse
|
18
|
Batal A, Garousi S, Finnson KW, Philip A. CD109, a master regulator of inflammatory responses. Front Immunol 2025; 15:1505008. [PMID: 39990858 PMCID: PMC11842317 DOI: 10.3389/fimmu.2024.1505008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/23/2024] [Indexed: 02/25/2025] Open
Abstract
Inflammation is a complex response to harmful stimuli, crucial for immunity, and linked to chronic diseases and cancer, with TGF-β and NF-κB pathways as key regulators. CD109 is a glycosylphosphatidylinositol (GPI)-anchored protein, that our group has originally identified as a TGF-β co-receptor and inhibitor of TGF-β signaling. CD109 modulates TGF-β and NF-κB pathways, to influence immune responses and inflammation. CD109's multifaceted role in inflammation spans various tissue types, including the skin, lung, bone and bone-related tissues, and various types of cancers. CD109 exerts its effects by modulating processes such as cytokine secretion, immune cell recruitment, macrophage polarization, T helper cell function and cancer cell phenotype and function. Here, we review CD109's regulatory functions in inflammatory responses in these various tissues and cell types. Exploration of CD109's mechanisms of action will enhance our understanding of its contributions to disease pathology and its potential for therapeutic applications.
Collapse
Affiliation(s)
- Adel Batal
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Setareh Garousi
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Kenneth W. Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
19
|
Zhao H, Wang Y, Ren J. Helicobacter pylori and rheumatoid arthritis: Investigation of relation from traditional Chinese medicine. Microb Pathog 2025; 199:107239. [PMID: 39708982 DOI: 10.1016/j.micpath.2024.107239] [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: 08/28/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition that predominantly affects synovial joints, manifesting with joint swelling, pain, and stiffness. In advanced stages, unchecked inflammation can inflict damage on bone and cartilage, resulting in disabilities and deformities of the joints. Additionally, systemic and extra-articular complications may arise due to the consequences of uncontrolled inflammation. Helicobacter pylori (H. pylori) is one of the most prevalent chronic bacterial infections in humans. This microorganism is a spiral-shaped, flagellated, microaerophilic gram-negative bacterium. Prolonged exposure leads to the activation of the immune system, with infected gastric mucosa epithelial cells continuously producing cytokines. This production, in turn, triggers the generation of antibodies as well as T Helper 1 and T Helper 2 effector T cells. The persistent antigenic stimulation resulting from H. pylori infection could lead to the progression of autoimmune diseases. Numerous clinical and pharmacological trials have illustrated the efficacy of traditional Chinese medicine against H. pylori. This review aims to delve into the connection between H. pylori and rheumatoid arthritis so as understand the pathogenesis. The concluding section of this review explores the interplay of Chinese medicine and Helicobacter pylori concerning rheumatoid arthritis.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| | - Yige Wang
- Shandong University of Traditional Chinese Medicine, No.16369, Jingshi Road, Lixia District, Jinan, 250013, China
| | - Jiahui Ren
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| |
Collapse
|
20
|
Liu H, Li Q, Chen Y, Dong M, Liu H, Zhang J, Yang L, Yin G, Xie Q. Suberosin attenuates rheumatoid arthritis by repolarizing macrophages and inhibiting synovitis via the JAK/STAT signaling pathway. Arthritis Res Ther 2025; 27:12. [PMID: 39838477 PMCID: PMC11748358 DOI: 10.1186/s13075-025-03481-3] [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: 09/12/2024] [Accepted: 01/12/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic disease that primarily manifests as chronic synovitis of the symmetric small joints. Despite the availability of various targeted drugs for RA, these treatments are limited by adverse reactions, warranting new treatment approaches. Suberosin (SBR), isolated from Plumbago zeylanica-a medicinal plant traditionally used to treat RA in Asia-possesses notable biological activities. This study aimed to investigate the effects and potential underlying pathways of SBR on RA. METHODS Tumor necrosis factor-alpha (TNF-α) induced inflammation in RA-derived fibroblast-like synoviocytes (RA-FLS), and the expression of proinflammatory mediators was assessed using q-RT PCR and ELISA after treatment with various SBR concentrations. Bone marrow-derived macrophages (BMDMs) were induced to differentiate into M1 and M2 macrophages, followed by treatment with various SBR concentrations and macrophage polarization assessment. Low-dose (0.5 mg/kg/d) and high-dose (2 mg/kg/d) SBR regimens were administered to a collagen-induced arthritis (CIA) mouse model for 21 days, and the anti-arthritic effects of SBR were evaluated. Network pharmacology and molecular docking analyses were used to predict the anti-arthritic targets of SBR. The effect of SBR on the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway was evaluated. RESULTS SBR suppressed macrophage polarization toward the M1 phenotype while enhancing their polarization toward the M2 phenotype. SBR reduced the levels of proinflammatory mediators in TNF-α-induced RA-FLS. Mechanistically, SBR inhibited the phosphorylation of the JAK1/STAT3 signaling pathway in RA-FLS and M1 macrophages and promoted the phosphorylation of the JAK1/STAT6 pathway in M2 macrophages, enhancing M2 polarization. In vivo, prophylactic treatment of low-dose SBR reduced M1 macrophage infiltration into synovial tissue, increased the proportion of M2 macrophages, and decreased the expression of inflammatory mediators in the serum and synovial tissue, alleviating synovial inflammation. SBR significantly alleviated arthritis in CIA mice through macrophage repolarization and inhibition of inflammation. CONCLUSION SBR significantly reduced clinical symptoms, joint pathological damage, and expression inflammatory cytokine expression in CIA mice. SBR exhibited anti-arthritic effects via the JAK1/STAT3 and JAK1/STAT6 signaling pathways, inhibiting synovial tissue inflammation and M1 macrophage polarization while promoting M2 macrophage polarization. Therefore, SBR may be an effective candidate for RA treatment.
Collapse
Affiliation(s)
- Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianwei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuehong Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Dong
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaqian Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Leiyi Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Geng Yin
- Department of General Practice, West China Hospital, General Practice Medical Center, Sichuan University, Chengdu, 610041, China.
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
21
|
Zhang MY, Wang MQ, Huang Y, Gu SL, Zhou MY, Xu ZS, Li LL, Lv M, Cai L, Li R. Silencing aquaporin 1 inhibits autophagy to exert anti-rheumatoid arthritis effects in TNF-α-induced fibroblast-like synoviocytes and adjuvant-induced arthritis rats. Inflamm Res 2025; 74:12. [PMID: 39774992 DOI: 10.1007/s00011-024-01966-6] [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: 10/12/2024] [Revised: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) are key players in rheumatoid arthritis (RA) by resisting apoptosis via increased autophagy. Elevated synovial aquaporin 1 (AQP1) affects RA FLS behaviors, but its relationship with FLS autophagy is unclear. We aim to clarify that silencing AQP1 inhibits autophagy to exert its anti-RA effects. METHODS We studied the effects and mechanisms of AQP1 silencing on autophagy in TNF-α-induced RA FLS and examined the crucial role of autophagy inhibition in its impacts on RA FLS pathogenic behaviors. We explored whether silencing synovial AQP1 relieved rat adjuvant-induced arthritis (AIA) by reducing synovial autophagy. RESULTS TNF-α stimulation increased AQP1 expression and autophagy levels in RA FLS, with a positive correlation between them. AQP1 silencing inhibited autophagy in TNF-α-stimulated RA FLS, along with suppressing proliferation, promoting apoptosis, and mitigating inflammation. Notably, the inhibitory effects of AQP1 silencing on RA FLS pathogenic behaviors were cancelled by autophagy activation with rapamycin (Rapa) but enhanced by autophagy inhibition using 3-Methyladenine. Mechanistically, silencing AQP1 enhanced the binding of Bcl-2 to Beclin1 by decreasing Beclin1-K63 ubiquitination, thus inhibiting RA FLS autophagy. In vivo, silencing synovial AQP1 relieved the severity and development of rat AIA, alongside reducing Ki67 expression, promoting apoptosis, and decreasing autophagy within AIA rat synovium. Expectedly, the Rapa co-administration nullified the anti-AIA effects of silencing synovial AQP1. CONCLUSION These findings reveal that silencing AQP1 inhibits RA FLS pathogenic behaviors and attenuates rat AIA through autophagy inhibition. This study may help clarify the pathogenic role of AQP1 in enhancing autophagy during RA development.
Collapse
Affiliation(s)
- Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Qing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Min Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, China.
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
22
|
Ma L, Jiang X, Gao J. Revolutionizing rheumatoid arthritis therapy: harnessing cytomembrane biomimetic nanoparticles for novel treatment strategies. Drug Deliv Transl Res 2025; 15:66-83. [PMID: 38758497 DOI: 10.1007/s13346-024-01605-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] [Accepted: 04/12/2024] [Indexed: 05/18/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic immune disease with severe implications for joint health. The issue of non-specific drug distribution potentially limits the therapeutic efficacy and increases the risk associated with RA treatment. Researchers employed cytomembrane-coated biomimetic nanoparticles (NPs) to enhance the targeting delivery efficacy to meet the demand for drug accumulation within the affected joints. Furthermore, distinct cytomembranes offer unique functionalities, such as immune cell activation and augmented NP biocompatibility. In this review, the current strategies of RA treatments were summarized in detail, and then an overview of RA's pathogenesis and the methodologies for producing cytomembrane-coated biomimetic NPs was provided. The application of cytomembrane biomimetic NPs derived from various cell sources in RA therapy is explored, highlighting the distinctive attributes of individual cytomembranes as well as hybrid membrane configurations. Through this comprehensive assessment of cytomembrane biomimetic NPs, we elucidate the prospective applications and challenges in the realm of RA therapy, and the strategy of combined therapy is proposed. In the future, cytomembrane biomimetic NPs have a broad therapeutic prospect for RA.
Collapse
Affiliation(s)
- Lan Ma
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- College of Pharmacy, Inner Mongolia Medical University, Chilechuan dairy economic development zone, Hohhot, Inner Mongolia Autonomous Region, 010110, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
23
|
Cheng X, Su Y, Dong N, Liu M, Wang M, Zhou T, Zhou H. Gross saponins of Tribulus terrestris attenuate rheumatoid arthritis by promoting apoptosis of fibroblast-like synoviocytes and reducing inflammation by inhibiting MAPK signalling pathway. Clin Exp Pharmacol Physiol 2024; 51:e13925. [PMID: 39448092 DOI: 10.1111/1440-1681.13925] [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: 07/31/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Among the numerous treatment options for rheumatoid arthritis (RA), the promotion of synoviocyte apoptosis and inhibition of inflammation are considered the most effective. However, the potential pro-apoptotic effects of gross saponins of Tribulus terrestris (GSTT), which are natural saponins derived from the herb Tribulus terrestris L., on rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) and their essential molecular mechanisms remain unclear. The aim of the present study was to investigate the influence of different concentrations of GSTT on RA-FLSs using various assays, including cell counting kit-8 (CCK-8), reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) and western blot analysis. These assessments were conducted to evaluate the cell viability, changes in the levels of inflammatory cytokines, apoptosis rates and alterations in protein expression related to this process. In vivo, arthritis clinical score, haematoxylin and eosin (HE) staining and ELISA were used to assess paw inflammation, histopathology and serum inflammatory cytokine changes. Our findings demonstrated that GSTT substantially promotes the apoptosis of RA-FLSs and reduces pro-inflammatory cytokine levels. GSTT also reduced the Bcl-2/Bax ratio and inhibited JNK and p38 phosphorylation. Furthermore, GSTT exhibits positive effects on RA by improving clinical scores, reducing synovial inflammatory infiltration and lowering serum pro-inflammatory cytokine levels. Therefore, by promoting the apoptosis of RA-FLSs and suppressing inflammation through the inhibition of the MAPK signalling pathway, GSTT is a promising therapeutic intervention for RA.
Collapse
Affiliation(s)
- Xinghai Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopaedics, The Yancheng School of Clinical Medicine of Nanjing Medical University (Yancheng Third People's Hospital), Yancheng, China
| | - Yuantao Su
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Haibin Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Tang N, Luo X, Ding Z, Shi Y, Cao X, Wu S. Single-Cell Multi-Dimensional data analysis reveals the role of ARL4C in driving rheumatoid arthritis progression and Macrophage polarization dynamics. Int Immunopharmacol 2024; 141:112987. [PMID: 39182267 DOI: 10.1016/j.intimp.2024.112987] [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: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Rheumatoid arthritis (RA) is an enduring autoimmune inflammatory condition distinguished by continual joint inflammation, hyperplasia of the synovium, erosion of bone, and deterioration of cartilage.Fibroblast-like synoviocytes (FLSs) exhibiting "tumor-like" traits are central to this mechanism.ADP-ribosylation factor-like 4c (ARL4C) functions as a Ras-like small GTP-binding protein, significantly impacting tumor migration, invasion, and proliferation.However, it remains uncertain if ARL4C participates in the stimulation of RA FLSs exhibiting "tumor-like" features, thereby fostering the advancement of RA. In our investigation, we unveiled, for the inaugural instance, via the amalgamated scrutiny of single-cell RNA sequencing (scRNA-seq) and Bulk RNA sequencing (Bulk-seq) datasets, that activated fibroblast-like synoviocytes (FLSs) showcase high expression of ARL4C, and the ARL4C protein expression in FLSs derived from RA patients significantly surpasses that observed in individuals with osteoarthritis (OA) and traumatic injury (trauma).Silencing of the ARL4C gene markedly impeded the proliferation of RA FLSs by hindered the transition of cells from the G0/G1 phase to the S phase, and intensified cell apoptosis and diminished the migratory and invasive capabilities. Co-culture of ARL4C gene-silenced RA FLSs with monocytes/macrophages significantly inhibited the polarization of monocytes/macrophages toward M1 and the repolarization of M2 to M1.Furthermore, intra-articular injection of shARL4C significantly alleviated synovial inflammation and cartilage erosion in collagen-induced arthritis (CIA) rats. In conclusion, our discoveries propose that ARL4C assumes a central role in the synovial inflammation, cartilage degradation, and bone erosion associated with RA by triggering the PI3K/AKT and MAPK signaling pathways within RA FLSs.ARL4C holds promise as a prospective target for the development of pharmaceutical agents targeting FLSs, with the aim of addressing RA.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xin Luo
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Zhiyu Ding
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yanbin Shi
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xu Cao
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Song Wu
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
25
|
Li J, Wu Z, Wu Y, Hu X, Yang J, Zhu D, Wu M, Li X, Bentum-Ennin L, Wanglai H. IL-22, a vital cytokine in autoimmune diseases. Clin Exp Immunol 2024; 218:242-263. [PMID: 38651179 PMCID: PMC11557150 DOI: 10.1093/cei/uxae035] [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: 02/01/2024] [Revised: 03/05/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
Interleukin-22 (IL-22) is a vital cytokine that is dysregulated in various autoimmune conditions including rheumatoid arthritis (RA), multiple sclerosis (MS), and Alzheimer's disease (AD). As the starting point for the activation of numerous signaling pathways, IL-22 plays an important role in the initiation and development of autoimmune diseases. Specifically, imbalances in IL-22 signaling can interfere with other signaling pathways, causing cross-regulation of target genes which ultimately leads to the development of immune disorders. This review delineates the various connections between the IL-22 signaling pathway and autoimmune disease, focusing on the latest understanding of the cellular sources of IL-22 and its effects on various cell types. We further explore progress with pharmacological interventions related to targeting IL-22, describing how such therapeutic strategies promise to usher in a new era in the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Jiajin Li
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Zhen Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Yuxin Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - XinYu Hu
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Jun Yang
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Dacheng Zhu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Mingyue Wu
- The School of pharmacy, Anhui Medical University, Hefei, China
| | - Xin Li
- The School of pharmacy, Anhui Medical University, Hefei, China
| | | | - Hu Wanglai
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
26
|
Tsai SW, Cheng YC, Chao YH, Yang DH. Sinulariolide Suppresses Inflammation of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis and Mitigates Collagen-Induced Arthritis Symptoms in Mice. J Inflamm Res 2024; 17:8299-8311. [PMID: 39525321 PMCID: PMC11550694 DOI: 10.2147/jir.s476847] [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: 05/05/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a systemic inflammatory disease characterized by active polyarthritis, which leads to functional loss and joint deformities. Natural compounds derived from marine organisms are considered valuable immune-modulating agents. This study aimed to assess the anti-inflammatory effect of sinulariolide, a soft coral-derived compound, on RA fibroblast-like synoviocytes and its therapeutic efficacy against collagen-induced arthritis (CIA). Methods To determine the effects of sinulariolide on tumor necrosis factor-alpha (TNF-α)-induced inflammation, MH7A cells pre-treated with 10 ng/mL TNF-α for 24 h were treated with sinulariolide. The effect of sinulariolide on proinflammatory cytokine expressions at both the mRNA and protein levels in the MH7A cells was assessed using real-time-polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA). Further, we analyzed the effect of sinulariolide on the activation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways using Western blotting and the TransAM NF-κB p65 kit. To comprehensively evaluate the potential application of sinulariolide in the treatment of inflammatory diseases, we used a well-established collagen-induced arthritis (CIA) mouse model. We examined the tissue sections of the ankle joints of the mice, assessed synovial hyperplasia, inflammatory cell infiltration, and cartilage damage, and used ELISA to analyze changes in cytokine expression in the hind paw tissues. Results MH7A cells treated with sinulariolide showed a notable reduction in the expression of proinflammatory cytokines, which could be due to decreased activation of the MAPK and NF-kB pathways. Additionally, sinulariolide-treated mice showed significantly reduced joint swelling and lower clinical arthritis scores than those in the normal and control groups. Significant reductions in synovial hyperplasia, inflammatory cell infiltration, and cartilage damage were observed in the tissue sections of the ankle joints of the mice treated with sinulariolide. Furthermore, the expression of inflammatory cytokines in the hind paw tissue of the mice treated with sinulariolide was significantly decreased. Conclusion Sinulariolide inhibited the progression of inflammation in MH7A cells. Sinulariolide treatment significantly reduced clinical arthritis symptoms and histological inflammatory responses in mice with CIA. Sinulariolide may serve as a potential therapeutic agent for RA.
Collapse
Affiliation(s)
- Sen-Wei Tsai
- Department of Physical Medicine and Rehabilitation, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 402, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Yu-Chieh Cheng
- Department of Orthopaedics, Tungs’ Taichung Metro Harbor Hospital, Taichung, 433, Taiwan
| | - Ya-Hsuan Chao
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
| | - Deng-Ho Yang
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
- Department of Internal Medicine, Taichung Armed-Forces General Hospital, Taichung, 411, Taiwan
- Division of Rheumatology/Immunology/Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, 406, Taiwan
| |
Collapse
|
27
|
Kirdaite G, Denkovskij J, Mieliauskaite D, Pachaleva J, Bernotiene E. The Challenges of Local Intra-Articular Therapy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1819. [PMID: 39597004 PMCID: PMC11596802 DOI: 10.3390/medicina60111819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Fibroblast-like synoviocytes (FLSs) are among the main disease-driving players in most cases of monoarthritis (MonoA), oligoarthritis, and polyarthritis. In this review, we look at the characteristics and therapeutic challenges at the onset of arthritis and during follow-up management. In some cases, these forms of arthritis develop into autoimmune polyarthritis, such as rheumatoid arthritis (RA), whereas local eradication of the RA synovium could still be combined with systemic treatment using immunosuppressive agents. Currently, the outcomes of local synovectomies are well studied; however, there is still a lack of a comprehensive analysis of current local intra-articular treatments highlighting their advantages and disadvantages. Therefore, the aim of this study is to review local intra-articular therapy strategies. According to publications from the last decade on clinical studies focused on intra-articular treatment with anti-inflammatory molecules, a range of novel slow-acting forms of steroidal drugs for the local treatment of synovitis have been investigated. As pain is an essential symptom, caused by both inflammation and cartilage damage, various molecules acting on pain receptors are being investigated in clinical trials as potential targets for local intra-articular treatment. We also overview the new targets for local treatment, including surface markers and intracellular proteins, non-coding ribonucleic acids (RNAs), etc.
Collapse
Affiliation(s)
- Gailute Kirdaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Diana Mieliauskaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jolita Pachaleva
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
- Faculty of Fundamental Sciences, Vilnius Gediminas Technical University, VilniusTech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| |
Collapse
|
28
|
Chao M, Hua Z, Zhu J, Wu G, Fan L, Tang R, Chen H, Gao F. Hyaluronic acid modified prussian blue analogs/TiO₂ janus nanostructures through efficient charge separation to enhance photocatalytic-driven dual gas for achieve multimodal treatment of rheumatoid arthritis. Int J Biol Macromol 2024; 281:136567. [PMID: 39419160 DOI: 10.1016/j.ijbiomac.2024.136567] [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: 06/24/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by the abnormal proliferation of fibroblast-like synoviocytes and changes in the joint synovium, including elevated reactive oxygen species, decreased pH, and reduced oxygen content. In this study, we synthesized a novel nanocomposite material, namely HA-PBA-TiO2 Janus nanocomposite, by in situ etching in prussian blue analogs doped with Co and Ni, followed by the growth of TiO2 nano-flowers and encapsulation in hyaluronic acid. When these janus nanoparticles diffused to the inflammatory sites of RA, they exhibited outstanding photocatalytic water-splitting ability under 660 nm laser irradiation, generating H2 and O2. This capability helps ameliorate the hypoxic microenvironment at RA inflammatory sites by eliminating reactive oxygen species (ROS) and enhancing antioxidation and oxygenation. Furthermore, owing to the doping of Co and Ni, HA-PBA-TiO2 exhibits photothermal conversion capability, which significant damage to FLS upon exposure to 660 nm laser irradiation, thereby controlling their aberrant proliferation. Through a series of in vitro and in vivo experiments, we validated the significant therapeutic efficacy of HA-PBA-TiO2 in treating RA, highlighting its broad prospects for application.
Collapse
Affiliation(s)
- Minghao Chao
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang Province, China
| | - Zhiyuan Hua
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jun Zhu
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an 223002, China
| | - Guoquan Wu
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Liying Fan
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Rongze Tang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hongliang Chen
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
29
|
Bui VD, Jeon J, Duong VH, Shin S, Lee J, Ghahari F, Kim CH, Jo YJ, Jung WK, Um W, Park JH. Chondroitin sulfate-based microneedles for transdermal delivery of stem cell-derived extracellular vesicles to treat rheumatoid arthritis. J Control Release 2024; 375:105-115. [PMID: 39218160 DOI: 10.1016/j.jconrel.2024.08.050] [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: 06/09/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
For the non-invasive treatment of rheumatoid arthritis (RA), a chondroitin sulfate C (CSC)-based dissolving microneedles (cMN) was prepared to deliver human adipose stem cell-derived extracellular vesicles (hASC-EV) into inflamed joints. Owing to their anti-inflammatory function, the hASC-EV-bearing cMN (EV@cMN) significantly suppressed activated fibroblast-like synoviocytes (aFLS) and M1 macrophages (M1), which are responsible for the progression of RA. In addition, EV@cMN facilitated the chondrogenic differentiation of bone marrow-derived stem cells. In mice with collagen-induced arthritis, EV@cMN efficiently delivered both hASC-EV and CSC to inflamed joints. Interestingly, pro-inflammatory cytokines in the inflamed joints were remarkably downregulated by the synergistic effect of CSC and hASC-EV. Consequently, as judged from the overall clinical score and joint swelling, EV@cMN showed an outstanding therapeutic effect, even comparable to the wild-type mice, without significant adverse effects. Overall, EV@cMN might have therapeutic potential for RA by efficiently delivering CSC and hASC-EV into the inflamed joints in a non-invasive manner.
Collapse
Affiliation(s)
- Van Dat Bui
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jueun Jeon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Van Hieu Duong
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Sol Shin
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jungmi Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Farrokhroo Ghahari
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Yu Jin Jo
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
30
|
Zheng J, Xiong X, Li K, Wang G, Cao H, Huang H. SPHK2 Knockdown Inhibits the Proliferation and Migration of Fibroblast-Like Synoviocytes Through the IL-17 Signaling Pathway in Osteoarthritis. J Inflamm Res 2024; 17:7221-7234. [PMID: 39416266 PMCID: PMC11479950 DOI: 10.2147/jir.s476077] [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: 04/29/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Objective Synovial inflammation is vital for the progression of osteoarthritis (OA). The objective of this study was to explore the effects and potential molecular mechanisms of sphingosine kinase 2 (SPHK2) on the proliferation and migration of fibroblast-like synoviocytes (FLS). Methods A TNF-α-stimulated FLS model and a papain-induced OA rat model were constructed. The functions of SPHK2 knockdown in OA were explored by a series of in vivo and in vitro assays. Downstream target genes of SPHK2 were investigated using transcriptome sequencing and validated by reverse transcription quantitative PCR (RT-qPCR). The effects of the SPHK2/IL-17 signaling pathway on inflammation, proliferation, and migration of OA-FLS were investigated using the IL-17 pathway inhibitor (secukinumab) and the activator (rhIL-17A). Results TNF-α stimulation promoted SPHK2 expression at mRNA and protein levels in OA-FLS. SPHK2 knockdown reduced IL-1β, IL-6, MMP-2, MMP-9, cyclinD1, and PCNA levels and suppressed proliferation and migration of OA-FLS. SPHK2 knockdown alleviated cartilage damage and synovial inflammation in the OA rat model. LRRIQ3, H4C8, CXCL1, CABP4, COL23A1, and PROK2 expression levels were regulated by SPHK2. SPHK2 knockdown inhibited the protein levels of IL-17A, IL-17RA, and Act1. The IL-17 pathway inhibitor secukinumab enhanced the inhibitory effect of SPHK2 knockdown on the proliferation and migration of OA-FLS, while the IL-17 pathway activator rhIL-17A exerted the opposite effect. Conclusion SPHK2 knockdown inhibits proliferation and migration of OA-FLS by blocking the IL-17 pathway, which provides a novel approach to the OA treatment.
Collapse
Affiliation(s)
- Jiaxuan Zheng
- Department of Pathology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| | - Xiaolong Xiong
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| | - Ke Li
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| | - Guangji Wang
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| | - Huiyuan Cao
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| | - Hui Huang
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou City, Hainan Province, 570311, People’s Republic of China
| |
Collapse
|
31
|
Qin M, Chen L, Hou X, Wu W, Liu Y, Pan Y, Zhang M, Tan Z, Huang D. Ultra-High-Performance Liquid Chromatography-High-Definition Mass Spectrometry-Based Metabolomics to Reveal the Potential Anti-Arthritic Effects of Illicium verum in Cultured Fibroblast-like Synoviocytes Derived from Rheumatoid Arthritis. Metabolites 2024; 14:517. [PMID: 39452898 PMCID: PMC11509614 DOI: 10.3390/metabo14100517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease. The fruits of Illicium verum, which is a medicinal and edible resource, have been shown to have anti-inflammatory properties. METHODS In this study, we investigated the effects of I. verum extracts (IVEs) on human RA fibroblasts-like synoviocytes (RA-FLS) by using a sensitive and selective ultra-high-performance liquid chromatography with high-definition mass spectrometry (UPLC-HDMS) method. We subsequently analyzed the metabolites produced after the incubation of cultured RA-FLS with IVEs. RESULTS IVEs inhibited the proliferation and suppressed the migration of RA-FLS, and reduced the levels of inflammatory factors including TNF-α and IL-6. Twenty differential metabolites responsible for the effects of IVEs were screened and annotated based on the UPLC-HDMS data by using a cell metabolomics approach. DISCUSSION Our findings suggest that treating RA-FLS with IVEs can regulate lipid and amino acid metabolism, indicating that this extract has the potential to modify the metabolic pathways that cause inflammation in RA. CONCLUSIONS This might lead to novel therapeutic strategies for managing patients with RA.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Pan
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, 189 Changgang Road, Nanning 530023, China (W.W.); (Z.T.)
| | | | | | - Danna Huang
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, 189 Changgang Road, Nanning 530023, China (W.W.); (Z.T.)
| |
Collapse
|
32
|
Wang F, Liu J. Regulating the lncRNA DSCR9/RPLP2/PI3K/AKT axis: an important mechanism of Xinfeng capsules in improving rheumatoid arthritis. Front Immunol 2024; 15:1465442. [PMID: 39376558 PMCID: PMC11456487 DOI: 10.3389/fimmu.2024.1465442] [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: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic and symmetrical polyarthritis. RA patients often experience inflammatory reaction and hypercoagulable state, which together affect the self-perception of patient (SPP). Currently, inhibiting inflammation and hypercoagulable state are common treatment methods for alleviating RA symptoms. Xinfeng Capsules (XFC) has a long history of treating RA, and can effectively improve the inflammatory response and hypercoagulable state of RA. However, the potential mechanisms have not yet been determined. Purpose and study design This study elucidated the action mechanism of XFC in RA inflammation and hypercoagulability through the lncDSCR9/RPLP2/PI3K/AKT axis. Results Clinical observations indicated that there was a strong link between XFC therapy and improvements in inflammatory and coagulation biomarkers, as well as SPP among RA patients. The subsequent network pharmacology analysis results identified the PI3K/AKT signaling pathway as a potential mediator for XFC treatment of RA. Furthermore, clinical validation and sequencing results revealed that lncRNA DSCR9 expression (a gene implicated in inflammation and coagulation) was negatively correlated with clinical markers of inflammation and coagulation, while positively correlated with SF-36 indicators. Notably, XFC treatment remarkably upregulated lncRNA DSCR9 expression and downregulated PI3K and AKT expressions, showing opposite expression trends to the untreated cases.The regulatory effect of XFC on the lncRNA DSCR9/RPLP2/PI3K/AKT axis in RA was investigated using techniques such as RNA pull-down assay, Western blot analysis, RT-PCR, and EdU assay. Moreover, the administration of the PI3K/AKT agonist RMH can counteract the effects of XFC on p-PI3K, p-AKT, inflammation, and hypercoagulability, reinforcing the role of pathway. Finally, animal studies utilizing HE staining and transmission electron microscopy (TEM) demonstrated that XFC notably decreased PI3K and AKT expressions in adjuvant-induced arthritis (AA) rats, mitigated inflammation and hypercoagulability, and enhanced the ultrastructure of synovial cells. These findings underscored the potential mechanisms of XFC in the treatment of RA. Conclusion Regulating the lncRNA DSCR9/RPLP2/PI3K/AKT axis may be an important mechanism by which XFC improved RA inflammatory response and hypercoagulable state.
Collapse
Affiliation(s)
- Fanfan Wang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, First Clinical Medical College, Hefei, Anhui, China
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jian Liu
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
33
|
Liu D, Kuang Y, Chen S, Li R, Su F, Zhang S, Qiu Q, Lin S, Shen C, Liu Y, Liang L, Wang J, Xu H, Xiao Y. NAT10 promotes synovial aggression by increasing the stability and translation of N4-acetylated PTX3 mRNA in rheumatoid arthritis. Ann Rheum Dis 2024; 83:1118-1131. [PMID: 38724075 DOI: 10.1136/ard-2023-225343] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/22/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Recent studies indicate that N-acetyltransferase 10 (NAT10)-mediated ac4C modification plays unique roles in tumour metastasis and immune infiltration. This study aimed to uncover the role of NAT10-mediated ac4C in fibroblast-like synoviocytes (FLSs) functions and synovial immune cell infiltration in rheumatoid arthritis (RA). METHODS FLSs were obtained from active established patients with RA. Protein expression was determined by western blotting or immunohistochemistry or multiplexed immunohistochemistry. Cell migration was measured using a Boyden chamber. ac4C-RIP-seq combined with RNA-seq was performed to identify potential targets of NAT10. RNA immunoprecipitation was used to validate the interaction between protein and mRNA. NAT10 haploinsufficiency, inhibitor remodelin or intra-articular Adv-NAT10 was used to suppress arthritis in mice with delayed-type hypersensitivity arthritis (DYHA) and collagen II-induced arthritis (CIA) and rats with CIA. RESULTS We found elevated levels of NAT10 and ac4C in FLSs and synovium from patients with RA. NAT10 knockdown or specific inhibitor treatment reduced the migration and invasion of RA FLSs. Increased NAT10 level in the synovium was positively correlated with synovial infiltration of multiple types of immune cells. NAT10 inhibition in vivo attenuated the severity of arthritis in mice with CIA and DTHA, and rats with CIA. Mechanistically, we explored that NAT10 regulated RA FLS functions by promoting stability and translation efficiency of N4-acetylated PTX3 mRNA. PTX3 also regulated RA FLS aggression and is associated with synovial immune cell infiltration. CONCLUSION Our findings uncover the important roles of NAT10-mediated ac4C modification in promoting rheumatoid synovial aggression and inflammation, indicating that NAT10 may be a potential target for the treatment of RA, even other dysregulated FLSs-associated disorders.
Collapse
Affiliation(s)
- Di Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Kuang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Simin Chen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruiru Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fan Su
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuoyang Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qian Qiu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuyu Shen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingli Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liuqin Liang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingnan Wang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youjun Xiao
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Lee HR, Yoo SJ, Kim J, Ran Lee Y, Kyoung Joo H, Hwa Jeon B, Wook Kang S. Apurinic/apyrimidinic endonuclease 1 alleviates inflammation in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Cent Eur J Immunol 2024; 49:113-125. [PMID: 39381557 PMCID: PMC11457561 DOI: 10.5114/ceji.2024.141946] [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: 01/25/2024] [Accepted: 03/19/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Apurinic/apyrimidinic endonuclease 1 (APEX1) is a protein with elevated expression in synovial fluids from rheumatoid arthritis (RA) patients. However, its role in RA pathogenesis remains unexplored. This study investigated the influence of APEX1 on inflammatory pathways in fibroblast-like synoviocytes (FLS) isolated from RA patients. Material and methods FLS from RA patients (n = 5) were stimulated with recombinant tumor necrosis factor α (TNF-α) and interleukin (IL)-17. Subsequently, cells were treated with recombinant APEX1, and assessments were made on reactive oxygen species (ROS) production and mitochondrial membrane potential. Additionally, mRNA levels of IL-1 family members were quantified. Cell migration was evaluated through Transwell chamber assays, and levels of key secreted inflammatory cytokines were measured via enzyme-linked immunosorbent assay (ELISA). Results The results demonstrated that APEX1 significantly reduced mitochondrial-specific ROS expression and restored mitochondrial membrane potential in TNF-α/IL-17-stimulated RA FLS. Furthermore, APEX1 treatments attenuated TNF-α/IL-17-induced activation of p38 MAPK, NF-κB, and PI3K 110 δ signaling pathways. Similarly, APEX1 significantly diminished TNF-α/IL-17-induced expression of inflammatory cytokines, including IL-1 family members, IL-6, IL-8, and vascular endothelial growth factor (VEGF). Notably, APEX1 downregulated cell migration of TNF-α/IL-17-treated RA FLS via inhibition of matrix metalloproteinase 3 (MMP3). Conclusions These findings collectively underscore the role of APEX1 as a key mediator of cytokine-amplified migration, modulating ROS and MMP3 in RA FLS, thus supporting its potential as a therapeutic target in RA treatment.
Collapse
Affiliation(s)
- Ha-Reum Lee
- Chungnam National University, South Korea
- Chungnam National University Hospital, South Korea
| | - Su-Jin Yoo
- Chungnam National University, South Korea
- Chungnam National University Hospital, South Korea
| | - Jinhyun Kim
- Chungnam National University, South Korea
- Chungnam National University Hospital, South Korea
| | - Yu Ran Lee
- Chungnam National University, South Korea
| | | | | | - Seong Wook Kang
- Chungnam National University, South Korea
- Chungnam National University Hospital, South Korea
| |
Collapse
|
35
|
Lackner A, Cabral JE, Qiu Y, Zhou H, Leonidas L, Pham MA, Macapagal A, Lin S, Armanus E, McNulty R. Small molecule inhibitor binds to NOD-like receptor family pyrin domain containing 3 and prevents inflammasome activation. iScience 2024; 27:110459. [PMID: 39104412 PMCID: PMC11298654 DOI: 10.1016/j.isci.2024.110459] [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: 02/02/2024] [Revised: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Despite recent advances in the mechanism of oxidized DNA activating NLRP3, the molecular mechanism and consequence of oxidized DNA associating with NLRP3 remains unknown. Cytosolic NLRP3 binds oxidized DNA which has been released from the mitochondria, which subsequently triggers inflammasome activation. Human glycosylase (hOGG1) repairs oxidized DNA damage which inhibits inflammasome activation. The fold of NLRP3 pyrin domain contains amino acids and a protein fold similar to hOGG1. Amino acids that enable hOGG1 to bind and cleave oxidized DNA are conserved in NLRP3. We found NLRP3 could bind and cleave oxidized guanine within mitochondrial DNA. The binding of oxidized DNA to NLRP3 was prevented by small molecule drugs which also inhibit hOGG1. These same drugs also inhibited inflammasome activation. Elucidating this mechanism will enable the design of drug memetics that treat inflammasome pathologies, illustrated herein by NLRP3 pyrin domain inhibitors which suppressed interleukin-1β (IL-1β) production in macrophages.
Collapse
Affiliation(s)
- Angela Lackner
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Julia Elise Cabral
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Yanfei Qiu
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Haitian Zhou
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Lemuel Leonidas
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Minh Anh Pham
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Alijah Macapagal
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Sophia Lin
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Emy Armanus
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Reginald McNulty
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| |
Collapse
|
36
|
Sendo S, Machado CRL, Boyle DL, Benschop RJ, Perumal NB, Choi E, Wang W, Firestein GS. Dysregulated NUB1 and Neddylation Enhances Rheumatoid Arthritis Fibroblast-Like Synoviocyte Inflammatory Responses. Arthritis Rheumatol 2024; 76:1252-1262. [PMID: 38566346 DOI: 10.1002/art.42856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) contribute to the pathogenesis of rheumatoid arthritis (RA), in part due to activation of the proinflammatory transcription factor NF-κB. Neddylation is modulated by the negative regulator of ubiquitin-like protein (NUB) 1. We determined whether NUB1 and neddylation are aberrant in the models with RA FLS, thereby contributing to their aggressive phenotype. METHODS Models with RA or osteoarthritis (OA) FLS were obtained from arthroplasty synovia. Real-time quantitative polymerase chain reaction and Western blot analysis assessed gene and protein expression, respectively. NUB1 was overexpressed using an expression vector. NF-κB activation was assessed by stimulating FLS with interleukin (IL)-1β. Neddylation inhibitor (MLN4924) and proteasome inhibitor were used in migration and gene expression assays. MLN4924 was used in the model with K/BxN serum-transfer arthritis. RESULTS Enhanced H3K27ac and H3K27me3 peaks were observed in the NUB1 promoter in the OA FLS compared with the RA FLS. NUB1 was constitutively expressed by FLS, but induction by IL-1β was significantly greater in the OA FLS. The ratio of neddylated cullin (CUL) 1 to nonneddylated CUL1 was lower in the OA FLS than the RA FLS. NUB1 overexpression decreased NF-κB nuclear translocation and IL-6 messenger RNA (mRNA) in IL-1β-stimulated the RA FLS. MLN4924 decreased CUL1 neddylation, NF-κB nuclear translocation, and IL-6 mRNA in IL-1β-stimulated the RA FLS. MLN4924 significantly decreased arthritis severity in the model with K/BxN serum-transfer arthritis. CONCLUSION CUL1 neddylation and NUB1 induction is dysregulated in the models with RA, which increases FLS activation. Inhibition of neddylation is an effective therapy in an animal model of arthritis. These data suggest that the neddylation system contributes to the pathogenesis of RA and that regulation of neddylation could be a novel therapeutic approach.
Collapse
Affiliation(s)
- Sho Sendo
- University of California, San Diego, School of Medicine, La Jolla
| | | | - David L Boyle
- University of California, San Diego, School of Medicine, La Jolla
| | | | - Narayanan B Perumal
- Eli Lilly and Company, Indianapolis, Indiana, and Eli Lilly and Company, San Diego, California
| | - Eunice Choi
- University of California, San Diego, La Jolla
| | - Wei Wang
- University of California, San Diego, La Jolla
| | - Gary S Firestein
- University of California, San Diego, School of Medicine, La Jolla
| |
Collapse
|
37
|
Mastana S, Knight E, Hampson A, Akam L, Hunter DJ, Ghelani A, Samanta A, Singh P. Role of Selected Genetic Polymorphisms in the Development of Rheumatoid Arthritis in a British White Population. Genes (Basel) 2024; 15:1009. [PMID: 39202369 PMCID: PMC11354150 DOI: 10.3390/genes15081009] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a complex autoimmune disease that negatively affects synovial joints, leading to the deterioration of movement and mobility of patients. This chronic disease is considered to have a strong genetic inheritance, with genome-wide association studies (GWAS) highlighting many genetic loci associated with the disease. Moreover, numerous confounding and non-genetic factors also contribute to the risk of the disease. AIMS This study investigates the association of selected genetic polymorphisms with rheumatoid arthritis risk and develops a polygenic risk score (PRS) based on selected genes. METHODS A case-control study recruited fully consenting participants from the East Midlands region of the UK. DNA samples were genotyped for a range of polymorphisms and genetic associations were calculated under several inheritance models. PRS was calculated at crude (unweighted) and weighted levels, and its associations with clinical parameters were determined. RESULTS There were significant associations with the risk of RA at six genetic markers and their associated risk alleles (TNRF2*G, TRAF1*A, PTPN22*T, HLA-DRB1*G, TNFα*A, and IL4-590*T). The TTG haplotype at the VDR locus increased the risk of RA with an OR of 3.05 (CI 1.33-6.98, p = 0.009). The GA haplotype of HLADRB1-TNFα-308 was a significant contributor to the risk of RA in this population (OR = 2.77, CI 1.23-6.28, p = 0.01), although linkage disequilibrium was low. The polygenic risk score was significantly higher in cases over controls in both unweighted (mean difference = 1.48, t285 = 5.387, p < 0.001) and weighted (mean difference = 2.75, t285 = 6.437, p < 0.001) results. CONCLUSION Several genetic loci contribute to the increased risk of RA in the British White sample. The PRS is significantly higher in those with RA and can be used for clinical applications and personalised prevention of disease.
Collapse
Affiliation(s)
- Sarabjit Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Ella Knight
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Abigail Hampson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Liz Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - David John Hunter
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Anant Ghelani
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Ash Samanta
- Rheumatology, University Hospitals of Leicester NHS Trust, Leicester LE1 5WW, UK;
| | - Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India;
| |
Collapse
|
38
|
Jeong YJ, Park SA, Park YH, Kim LK, Lee HR, Kim HJ, Heo TH. Anti-inflammatory effect of the combined treatment of LMT-28 and kaempferol in a collagen-induced arthritis mouse model. PLoS One 2024; 19:e0302119. [PMID: 39083495 PMCID: PMC11290667 DOI: 10.1371/journal.pone.0302119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation and swelling. Several studies have demonstrated that RA fibroblast-like synovial cells (RA-FLS) play an important role in RA pathogenesis. Activated RA-FLS contribute to synovial inflammation by secreting inflammatory cytokines including interleukin (IL)-1β, IL-6 and tumor necrosis factor-α. LMT-28 is derivative of oxazolidone and exerts anti-inflammatory effects on RA via IL-6 signaling pathway regulation. LMT-28 also regulates T cell differentiation in RA condition. However, the effect of LMT-28 on the migration and invasion of RA-FLS remains unknown. Kaempferol has been reported to have pharmacological effects on various diseases, such as inflammatory diseases, autoimmune diseases, and cancer. Additionally, kaempferol has been reported to inhibit RA-FLS migration and invasion, but it is not known about the therapeutic mechanism including molecular mechanism such as receptor. The present study aimed to investigate the synergistic effects of the combined treatment of LMT-28 and kaempferol on RA-FLS activation and RA pathogenesis in mouse model. LMT-28 and kaempferol co-administration inhibited RA disease severity and histological collapse in the joint tissues of CIA mice, as well as downregulated the levels of pro-inflammatory cytokines in mouse serum. Additionally, the combined treatment inhibited excessive differentiation of T helper 17 cells and osteoclasts. Furthermore, compared with single treatments, combined treatment showed enhanced inhibitory effects on the hyperactivation of IL-6-induced signaling pathway in RA-FLS. Combined treatment also inhibited RA-FLS cell proliferation, migration, and invasion and suppressed the expression of matrix metalloproteinase in RA-FLS. Furthermore, we confirmed that the combined treatment inhibited chondrocyte proliferation, migration, and invasion. In conclusion, our results suggest that the combined treatment of LMT-28 and kaempferol exerts a synergistic effect on the RA development via the regulation of IL-6-induced hyperactivation of RA-FLS. Furthermore, this study suggests that combination therapies can be an effective therapeutic option for arthritis.
Collapse
Affiliation(s)
- Young-Jin Jeong
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Yeon-Hwa Park
- Biowave, Anyangcheon-ro, Yangcheon-gu, Seoul, Republic of Korea
| | - Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Hae-Ri Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
39
|
Gong X, Su L, Huang J, Liu J, Wang Q, Luo X, Yang G, Chi H. An overview of multi-omics technologies in rheumatoid arthritis: applications in biomarker and pathway discovery. Front Immunol 2024; 15:1381272. [PMID: 39139555 PMCID: PMC11319186 DOI: 10.3389/fimmu.2024.1381272] [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: 02/03/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease with a complex pathological mechanism involving autoimmune response, local inflammation and bone destruction. Metabolic pathways play an important role in immune-related diseases and their immune responses. The pathogenesis of rheumatoid arthritis may be related to its metabolic dysregulation. Moreover, histological techniques, including genomics, transcriptomics, proteomics and metabolomics, provide powerful tools for comprehensive analysis of molecular changes in biological systems. The present study explores the molecular and metabolic mechanisms of RA, emphasizing the central role of metabolic dysregulation in the RA disease process and highlighting the complexity of metabolic pathways, particularly metabolic remodeling in synovial tissues and its association with cytokine-mediated inflammation. This paper reveals the potential of histological techniques in identifying metabolically relevant therapeutic targets in RA; specifically, we summarize the genetic basis of RA and the dysregulated metabolic pathways, and explore their functional significance in the context of immune cell activation and differentiation. This study demonstrates the critical role of histological techniques in decoding the complex metabolic network of RA and discusses the integration of histological data with other types of biological data.
Collapse
Affiliation(s)
- Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jie Liu
- Department of Geriatric, Dazhou Central Hospital, Dazhou, China
| | - Qinglai Wang
- Orthopedics and Traumatology Department of TCM, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Xiufang Luo
- Department of Geriatric, Dazhou Central Hospital, Dazhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| |
Collapse
|
40
|
Hu S, Lin Y, Tang Y, Zhang J, He Y, Li G, Li L, Cai X. Targeting dysregulated intracellular immunometabolism within synovial microenvironment in rheumatoid arthritis with natural products. Front Pharmacol 2024; 15:1403823. [PMID: 39104392 PMCID: PMC11298361 DOI: 10.3389/fphar.2024.1403823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Immunometabolism has been an emerging hotspot in the fields of tumors, obesity, and atherosclerosis in recent decades, yet few studies have investigated its connection with rheumatoid arthritis (RA). In principle, intracellular metabolic pathways upstream regulated by nutrients and growth factors control the effector functions of immune cells. Dynamic communication and hypermetabolic lesions of immune cells within the inflammatory synovial microenvironment contributes to the development and progression of RA. Hence, targeting metabolic pathways within immune subpopulations and pathological cells may represent novel therapeutic strategies for RA. Natural products constitute a great potential treasury for the research and development of novel drugs targeting RA. Here, we aimed to delineate an atlas of glycolysis, lipid metabolism, amino acid biosynthesis, and nucleotide metabolism in the synovial microenvironment of RA that affect the pathological processes of synovial cells. Meanwhile, therapeutic potentials and pharmacological mechanisms of natural products that are demonstrated to inhibit related key enzymes in the metabolic pathways or reverse the metabolic microenvironment and communication signals were discussed and highlighted.
Collapse
Affiliation(s)
- Shengtao Hu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuanyuan Tang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junlan Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yini He
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gejing Li
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liqing Li
- The Central Research Laboratory, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Central Research Laboratory, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| |
Collapse
|
41
|
Zhang Y, He X, Yin D, Zhang Y. Redefinition of Synovial Fibroblasts in Rheumatoid Arthritis. Aging Dis 2024:AD.2024.0514. [PMID: 39122458 DOI: 10.14336/ad.2024.0514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
The breakdown of immune tolerance and the rise in autoimmunity contribute to the onset of rheumatoid arthritis (RA), driven by significant changes in immune components. Recent advances in single-cell and spatial transcriptome profiling have revealed shifts in cell distribution and composition, expanding our understanding beyond molecular-level changes in inflammatory cytokines, autoantibodies, and autoantigens in RA. Surprisingly, synovial fibroblasts (SFs) play an active immunopathogenic role rather than remaining passive bystanders in RA, with notable alterations in their subpopulation distribution and composition. This study examines these changes in SF heterogeneity, assesses their impact on RA progression, and elucidates the immune characteristics and functions of SF subsets in the RA autoimmunity, encompassing both intrinsic and adaptive immunity. Additionally, this review discusses therapeutic strategies targeting immune SF subsets, highlighting the potential of future interventions in SF phenotypic reprogramming. Overall, this review redefines the role of SFs in RA and suggests targeting SF phenotypic reprogramming and its upstream molecules as a promising therapeutic approach to restore immune balance and modulate immune tolerance in RA.
Collapse
Affiliation(s)
- Yinci Zhang
- First Affiliated Hospital of Medical School, Anhui University of Science and Technology, Huainan, China
| | - Xiong He
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Dongdong Yin
- First Affiliated Hospital of Medical School, Anhui University of Science and Technology, Huainan, China
| | - Yihao Zhang
- Department of health inspection and quarantine, School of Public Health, Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
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
|
43
|
Haloi P, Choudhary R, Lokesh BS, Konkimalla VB. Dual drug nanoparticle synergistically induced apoptosis, suppressed inflammation, and protected autophagic response in rheumatoid arthritis fibroblast-like synoviocytes. Immunol Lett 2024; 267:106854. [PMID: 38537719 DOI: 10.1016/j.imlet.2024.106854] [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: 08/07/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic immune-mediated joint inflammatory disorder associated with aberrant activation of fibroblast-like synoviocytes (FLS). Recently, FLS gained importance due to its crucial role in RA pathogenesis, and thus, targeting FLS is suggested as an attractive treatment strategy for RA. FLS-targeted approaches may be combined with disease-modifying antirheumatic drugs (DMARDs) and natural phytochemicals to improve efficacy in RA control and negate immunosuppression. In this study, we assessed the therapeutic effectiveness of DD NP HG in primary RA-FLS cells isolated from the synovial tissue of FCA-induced RA rats. We observed that DD NP HG had good biosafety for healthy FLS cells and, at higher concentrations, a mild inhibitory effect on RA-FLS. The combination therapy (DD NP HG) of MTX NP and PEITC NE in RA-FLS showed a higher rate of apoptosis with significantly reduced LPS-induced expression of pro-inflammatory cytokines (TNF-α, IL-17A, and IL-6) in arthritic FLS. Further, the gene expression studies showed that DD NP HG significantly down-regulated the mRNA expression of IL-1β, RANKL, NFATc1, DKK1, Bcl-xl, Mcl-1, Atg12, and ULK1, and up-regulated the mRNA expression of OPG, PUMA, NOXA and SQSTM1 in LPS-stimulated RA-FLS cells. Collectively, our results demonstrated that DD NP HG significantly inhibited the RA-FLS proliferation via inducing apoptosis, down-regulating pro-inflammatory cytokines, and further enhancing the expression of genes associated with bone destruction in RA pathogenesis. A nanotechnology approach is a promising strategy for the co-delivery of dual drugs to regulate the RA-FLS function and achieve synergistic treatment of RA.
Collapse
Affiliation(s)
- Prakash Haloi
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Rajat Choudhary
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - B Siva Lokesh
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - V Badireenath Konkimalla
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
44
|
Fang Y, Huang W, Zhu X, Wang X, Wu X, Wang H, Hong W, Yan S, Zhang L, Deng Y, Wei W, Tu J, Zhu C. Epigenetic Regulatory Axis MIR22-TET3-MTRNR2L2 Represses Fibroblast-Like Synoviocyte-Mediated Inflammation in Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:845-856. [PMID: 38221658 DOI: 10.1002/art.42795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
OBJECTIVE The specific role of fibroblast-like synoviocytes (FLSs) in the pathogenesis of rheumatoid arthritis (RA) is still not fully elucidated. This study aimed to explore the molecular mechanisms of epigenetic pathways, including three epigenetic factors, microRNA (miRNA)-22 (MIR22), ten-eleven translocation methylcytosine dioxygenase 3 (TET3), and MT-RNR2 like 2 (MTRNR2L2), in RA-FLSs. METHODS The expression of MIR22, TET3, and MTRNR2L2 in the synovium of patients with RA and arthritic mice were determined by fluorescence in situ hybridization, quantitative polymerase chain reaction (qPCR), immunohistochemistry, and Western blot. Mir22-/- and Tet3+/- mice were used to establish a collagen antibody-induced arthritis (CAIA) model. Mir22 angomir and Tet3 small interfering RNA (siRNA) were used to illustrate the therapeutic effects on arthritis using a collagen-induced (CIA) model. Bioinformatics, luciferase reporter assay, 5-hydroxymethylcytosine (5hmC) dot blotting, chromatin immunoprecipitation-qPCR, and hydroxymethylated DNA immunoprecipitation were conducted to show the direct repression of MIR22 on the TET3 and transcriptional activation of TET3 on MTRNR2L2. RESULTS The Mir22-/- CAIA model and RA-FLS-related in vitro experiments demonstrated the inhibitory effect of MIR22 on inflammation. MIR22 can directly inhibit the translation of TET3 in RA-FLSs by binding to its 3' untranslated region in TET3. The Tet3+/- mice-established CAIA model showed less severe symptoms of arthritis in vivo. In vitro experiments further confirmed the proinflammatory effect of TET3 in RA. In addition, the CIA model was used to validate the therapeutic effects of Mir22 angomir and Tet3 siRNA. Finally, TET3 exerts its proinflammatory effect by promoting 5hmC production in the promoter of its target MTRNR2L2 in RA-FLSs. CONCLUSION The key role of the MIR22-TET3-MTRNR2L2 pathway in RA-FLSs provided an experimental basis for further studies into the pathogenesis and related targets of RA from the perspective of FLSs.
Collapse
Affiliation(s)
- Yilong Fang
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Wei Huang
- The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, China
| | - Xiangling Zhu
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Xinming Wang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuming Wu
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Huihui Wang
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Wenming Hong
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shangxue Yan
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Lingling Zhang
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Yujie Deng
- Guangzhou National Laboratory, Guangzhou, China
| | - Wei Wei
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Jiajie Tu
- Anhui Medical University and Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Chen Zhu
- The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, China
| |
Collapse
|
45
|
Lu M, Zhu M, Wu Z, Liu W, Cao C, Shi J. The role of YAP/TAZ on joint and arthritis. FASEB J 2024; 38:e23636. [PMID: 38752683 DOI: 10.1096/fj.202302273rr] [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: 11/03/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are two common forms of arthritis with undefined etiology and pathogenesis. Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ), which act as sensors for cellular mechanical and inflammatory cues, have been identified as crucial players in the regulation of joint homeostasis. Current studies also reveal a significant association between YAP/TAZ and the pathogenesis of OA and RA. The objective of this review is to elucidate the impact of YAP/TAZ on different joint tissues and to provide inspiration for further studying the potential therapeutic implications of YAP/TAZ on arthritis. Databases, such as PubMed, Cochran Library, and Embase, were searched for all available studies during the past two decades, with keywords "YAP," "TAZ," "OA," and "RA."
Collapse
Affiliation(s)
- Mingcheng Lu
- Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Mengqi Zhu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Zuping Wu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Wei Liu
- Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Chuwen Cao
- Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Jiejun Shi
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang, Hangzhou, China
| |
Collapse
|
46
|
Qian H, Deng C, Chen S, Zhang X, He Y, Lan J, Wang A, Shi G, Liu Y. Targeting pathogenic fibroblast-like synoviocyte subsets in rheumatoid arthritis. Arthritis Res Ther 2024; 26:103. [PMID: 38783357 PMCID: PMC11112866 DOI: 10.1186/s13075-024-03343-4] [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: 12/21/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Fibroblast-like synoviocytes (FLSs) play a central role in RA pathogenesis and are the main cellular component in the inflamed synovium of patients with rheumatoid arthritis (RA). FLSs are emerging as promising new therapeutic targets in RA. However, fibroblasts perform many essential functions that are required for sustaining tissue homeostasis. Direct targeting of general fibroblast markers on FLSs is challenging because fibroblasts in other tissues might be altered and side effects such as reduced wound healing or fibrosis can occur. To date, no FLS-specific targeted therapies have been applied in the clinical management of RA. With the help of high-throughput technologies such as scRNA-seq in recent years, several specific pathogenic FLS subsets in RA have been identified. Understanding the characteristics of these pathogenic FLS clusters and the mechanisms that drive their differentiation can provide new insights into the development of novel FLS-targeting strategies for RA. Here, we discuss the pathogenic FLS subsets in RA that have been elucidated in recent years and potential strategies for targeting pathogenic FLSs.
Collapse
Affiliation(s)
- Hongyan Qian
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Chaoqiong Deng
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
| | - Shiju Chen
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Xinwei Zhang
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Yan He
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Jingying Lan
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
| | - Aodi Wang
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China
| | - Guixiu Shi
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China.
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China.
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China.
| | - Yuan Liu
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 55th, Zhenhai Road, Xiamen, XM, 361000, China.
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China.
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China.
| |
Collapse
|
47
|
Chen H, Fu X, Wu X, Zhao J, Qiu F, Wang Z, Wang Z, Chen X, Xie D, Huang J, Fan J, Yang X, Song Y, Li J, He D, Xiao G, Lu A, Liang C. Gut microbial metabolite targets HDAC3-FOXK1-interferon axis in fibroblast-like synoviocytes to ameliorate rheumatoid arthritis. Bone Res 2024; 12:31. [PMID: 38782893 PMCID: PMC11116389 DOI: 10.1038/s41413-024-00336-6] [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: 12/13/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease. Early studies hold an opinion that gut microbiota is environmentally acquired and associated with RA susceptibility. However, accumulating evidence demonstrates that genetics also shape the gut microbiota. It is known that some strains of inbred laboratory mice are highly susceptible to collagen-induced arthritis (CIA), while the others are resistant to CIA. Here, we show that transplantation of fecal microbiota of CIA-resistant C57BL/6J mice to CIA-susceptible DBA/1J mice confer CIA resistance in DBA/1J mice. C57BL/6J mice and healthy human individuals have enriched B. fragilis than DBA/1J mice and RA patients. Transplantation of B. fragilis prevents CIA in DBA/1J mice. We identify that B. fragilis mainly produces propionate and C57BL/6J mice and healthy human individuals have higher level of propionate. Fibroblast-like synoviocytes (FLSs) in RA are activated to undergo tumor-like transformation. Propionate disrupts HDAC3-FOXK1 interaction to increase acetylation of FOXK1, resulting in reduced FOXK1 stability, blocked interferon signaling and deactivation of RA-FLSs. We treat CIA mice with propionate and show that propionate attenuates CIA. Moreover, a combination of propionate with anti-TNF etanercept synergistically relieves CIA. These results suggest that B. fragilis or propionate could be an alternative or complementary approach to the current therapies.
Collapse
Affiliation(s)
- Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Junyi Zhao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Zhenghong Wang
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Junyu Fan
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yi Song
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jie Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dongyi He
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510006, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China.
| |
Collapse
|
48
|
Lin Y, Chen Y, Hu W, Liu X, Hao W, Xing J, Ding J, Xu Y, Yao F, Zhao Y, Wang K, Li S, Yu Q, Hu W, Zhou R. TRPM7 facilitates fibroblast-like synoviocyte proliferation, metastasis and inflammation through increasing IL-6 stability via the PKCα-HuR axis in rheumatoid arthritis. Int Immunopharmacol 2024; 132:111933. [PMID: 38581988 DOI: 10.1016/j.intimp.2024.111933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/08/2024]
Abstract
Transient receptor potential melastatin 7 (TRPM7) is a cation channel that plays a role in the progression of rheumatoid arthritis (RA), yet its involvement in synovial hyperplasia and inflammation has not been determined. We previously reported that TRPM7 affects the destruction of articular cartilage in RA. Herein, we further confirmed the involvement of TRPM7 in fibroblast-like synoviocyte (FLS) proliferation, metastasis and inflammation. We observed increased TRPM7 expression in FLSs derived from human RA patients. Pharmacological inhibition of TRPM7 protected primary RA-FLSs from proliferation, metastasis and inflammation. Furthermore, we found that TRPM7 contributes to RA-FLS proliferation, metastasis and inflammation by increasing the intracellular Ca2+ concentration. Mechanistically, the PKCα-HuR axis was demonstrated to respond to Ca2+ influx, leading to TRPM7-mediated RA-FLS proliferation, metastasis and inflammation. Moreover, HuR was shown to bind to IL-6 mRNA after nuclear translocation, which could be weakened by TRPM7 channel inhibition. Additionally, adeno-associated virus 9-mediated TRPM7 silencing is highly effective at alleviating synovial hyperplasia and inflammation in adjuvant-induced arthritis rats. In conclusion, our findings unveil a novel regulatory mechanism involved in the pathogenesis of RA and suggest that targeting TRPM7 might be a potential strategy for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Yi Lin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Weirong Hu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xingyu Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenjuan Hao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jing Xing
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yucai Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Qiuxia Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
49
|
Chang MH, Fuhlbrigge RC, Nigrovic PA. Joint-specific memory, resident memory T cells and the rolling window of opportunity in arthritis. Nat Rev Rheumatol 2024; 20:258-271. [PMID: 38600215 PMCID: PMC11295581 DOI: 10.1038/s41584-024-01107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
In rheumatoid arthritis, juvenile idiopathic arthritis and other forms of inflammatory arthritis, the immune system targets certain joints but not others. The pattern of joints affected varies by disease and by individual, with flares most commonly involving joints that were previously inflamed. This phenomenon, termed joint-specific memory, is difficult to explain by systemic immunity alone. Mechanisms of joint-specific memory include the involvement of synovial resident memory T cells that remain in the joint during remission and initiate localized disease recurrence. In addition, arthritis-induced durable changes in synovial fibroblasts and macrophages can amplify inflammation in a site-specific manner. Together with ongoing systemic processes that promote extension of arthritis to new joints, these local factors set the stage for a stepwise progression in disease severity, a paradigm for arthritis chronicity that we term the joint accumulation model. Although durable drug-free remission through early treatment remains elusive for most forms of arthritis, the joint accumulation paradigm defines new therapeutic targets, emphasizes the importance of sustained treatment to prevent disease extension to new joints, and identifies a rolling window of opportunity for altering the natural history of arthritis that extends well beyond the initiation phase of disease.
Collapse
Affiliation(s)
- Margaret H Chang
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Robert C Fuhlbrigge
- Department of Paediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA.
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
50
|
Wang H, Han J, Dmitrii G, Ning K, Zhang X. KLF transcription factors in bone diseases. J Cell Mol Med 2024; 28:e18278. [PMID: 38546623 PMCID: PMC10977429 DOI: 10.1111/jcmm.18278] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2025] Open
Abstract
Krüppel-like factors (KLFs) are crucial in the development of bone disease. They are a family of zinc finger transcription factors that are unusual in containing three highly conserved zinc finger structural domains interacting with DNA. It has been discovered that it engages in various cell functions, including proliferation, apoptosis, autophagy, stemness, invasion and migration, and is crucial for the development of human tissues. In recent years, the role of KLFs in bone physiology and pathology has received adequate attention. In addition to regulating the normal growth and development of the musculoskeletal system, KLFs participate in the pathological process of the bones and joints and are intimately linked to several skeletal illnesses, such as osteoarthritis (OA), rheumatoid arthritis (RA), osteoporosis (OP) and osteosarcoma (OS). Consequently, targeting KLFs has emerged as a promising therapeutic approach for an array of bone disorders. In this review, we summarize the current literature on the importance of KLFs in the emergence and regulation of bone illnesses, with a particular emphasis on the pertinent mechanisms by which KLFs regulate skeletal diseases. We also discuss the need for KLFs-based medication-targeted treatment. These endeavours offer new perspectives on the use of KLFs in bone disorders and provide prognostic biomarkers, therapeutic targets and possible drug candidates for bone diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and HealthShenyang Sport UniversityShenyangLiaoningChina
| | - Juanjuan Han
- College of Exercise and HealthShenyang Sport UniversityShenyangLiaoningChina
- Department of Sport RehabilitationShanghai University of SportShanghaiChina
| | - Gorbachev Dmitrii
- Head of General Hygiene DepartmentSamara State Medical UniversitySamaraRussia
| | - Ke Ning
- College of Exercise and HealthShenyang Sport UniversityShenyangLiaoningChina
| | - Xin‐an Zhang
- College of Exercise and HealthShenyang Sport UniversityShenyangLiaoningChina
| |
Collapse
|