1
|
Gao Y, Liu Y, Hao H. A 10-year knowledge mapping of T cells in rheumatoid arthritis: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2450855. [PMID: 39962980 PMCID: PMC11845019 DOI: 10.1080/21645515.2025.2450855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 02/23/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease of autoimmune origin. T cells play crucial roles in the initiation and progression of RA. Although bibliometric methods have been widely used to synthesize knowledge trajectories across different biomedical fields, it has hardly been used to underscore the knowledge trends in relation to T cell and RA. This study used bibliometric methods to delineate the evolution of research on T cells and RA. Data were sourced from the Web of Science™ Core Collection and were scientometrically analyzed using CiteSpace and VOSviewer. From 2014 to 2023, 7037 papers on T cells and RA were retrieved. The number of annual publications is stable between 600 and 800, and the citation frequency continues to rise. The United States, China, the United Kingdom and Japan were the most productive countries. Karolinska Institute, and Harvard Medical School were the institutions that published the most research papers. Wei W, Cho ML, and Park SH were the most prolific authors. Mclnnes IB and Smolen JS were the most frequently cited authors. The journals with the most articles are Frontiers in Immunology, Arthritis Research & Therapy, and Arthritis & Rheumatology. Current research hotspots include pathogenic factors and targeted biological therapy, immune mechanisms, inflammatory mechanisms, and bone destruction mechanisms. The current research frontiers in this field are gut microbiota, identification, fibroblast-like synoviocytes, biologic therapy, mesenchymal stem cells, and risk. This work provides new insights into the scientific research and clinical application of T cells to develop therapeutic targets for RA.
Collapse
Affiliation(s)
- Yanfang Gao
- Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yang Liu
- Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Huiqin Hao
- Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
2
|
Zhao X, Huang C, Liang X, Chang H, Zhang L. A sensitive fluorescent probe for monitoring hypochlorous acid levels in rheumatoid arthritis. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 336:126020. [PMID: 40081226 DOI: 10.1016/j.saa.2025.126020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease. Hypochlorous acid (HClO) is a signature reactive oxygen species (ROS) closely associated with the progression of RA. Here, we report a novel fluorescent probe, ZCP1, which exhibits high sensitivity to HClO. In the presence of HClO, ZCP1 demonstrates a rapid detection time of 20 s and a low detection limit of 19.1 nM, allowing for fast and sensitive reactions with HClO, with a 150-fold fluorescence enhancement. ZCP1 can be employed for fluorescent detection of both exogenous and endogenous HClO levels in live cells. Furthermore, ZCP1 has been utilized to detect endogenous HClO in a mouse model of RA. This work provides a reliable tool for monitoring endogenous HClO both in vivo and in vitro, offering significant potential for future biological and pathological studies related to HClO.
Collapse
Affiliation(s)
- Xin Zhao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Chi Huang
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Xiao Liang
- College of Pharmacy, Heze University, Heze, Shandong Province, China.
| | - Hao Chang
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China.
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| |
Collapse
|
3
|
Zhang J, Shi X, Wu Z. A Caspase-3 responsive nanoemulsion for targeted treatment of rheumatoid arthritis through dual modulation of inflammation and mitochondrial dysfunction. Colloids Surf B Biointerfaces 2025; 251:114629. [PMID: 40090172 DOI: 10.1016/j.colsurfb.2025.114629] [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: 01/02/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joint inflammation, pain, and progressive joint damage. Current treatments, while effective, are limited by their potential side effects, particularly in long-term use. This study introduces a novel nanoemulsion-based therapeutic approach combining rapamycin, an mTOR inhibitor, with SS31, a mitochondrial-targeting antioxidant peptide. The rapamycin-SS31 conjugate (RS31) is encapsulated within a nanoemulsion (RS31@NEs) designed to selectively release its components in response to elevated Caspase-3 levels, prevalent in inflamed joints. In vitro and in vivo studies using zymosan-induced arthritis (ZIA) and collagen-induced arthritis (CIA) mouse models demonstrated that RS31@NEs effectively reduced pro-inflammatory cytokines, mitigated oxidative stress, and improved immune modulation by enhancing regulatory T and B cell functions. These findings highlight RS31@NEs as a promising dual-action therapy for RA, combining anti-inflammatory and mitochondrial protective effects while minimizing systemic toxicity.
Collapse
Affiliation(s)
- Jia Zhang
- Department of hematology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhuo Wu
- Outpatient Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
4
|
Fehringer M, Vogl T. Molecular mimicry in the pathogenesis of autoimmune rheumatic diseases. J Transl Autoimmun 2025; 10:100269. [PMID: 39877080 PMCID: PMC11773492 DOI: 10.1016/j.jtauto.2025.100269] [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: 10/31/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Autoimmune rheumatic diseases (ARDs) are a heterogeneous group of conditions characterized by excessive and misdirected immune responses against the body's own musculoskeletal tissues. Their exact aetiology remains unclear, with genetic, demographic, behavioural and environmental factors implicated in disease onset. One prominent hypothesis for the initial breach of immune tolerance (leading to autoimmunity) is molecular mimicry, which describes structural or sequence similarities between human and microbial proteins (mimotopes). This similarity can lead to cross-reactive antibodies and T-cell receptors, resulting in an immune response against autoantigens. Both commensal microbes in the human microbiome and pathogens can trigger molecular mimicry, thereby potentially contributing to the onset of ARDs. In this review, we focus on the role of molecular mimicry in the onset of rheumatoid arthritis and systemic lupus erythematosus. Moreover, implications of molecular mimicry are also briefly discussed for ankylosing spondylitis, systemic sclerosis and myositis.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| |
Collapse
|
5
|
Basu N. Probing the synovium-brain axis. Semin Arthritis Rheum 2025; 72S:152673. [PMID: 40037997 DOI: 10.1016/j.semarthrit.2025.152673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Affiliation(s)
- Neil Basu
- INSTITUTE OF INFECTION & IMMUNITY, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK; The University of Glasgow, charity number SC004401,UK.
| |
Collapse
|
6
|
Ghouri M, Siddiqui NN, Lateef M, Avesi L, Khan R, Ghauri H, Asif E, Zehra S. Modified expression of JAK-STAT pathway genes in an in vivo rheumatoid arthritis model: A preclinical study to explore genetic insights. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167780. [PMID: 40073707 DOI: 10.1016/j.bbadis.2025.167780] [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: 11/06/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease characterised by inflammatory synovial tissue, joint deterioration, and effects on systems other than the joints. The biological process underlying the progression of the disease remains unknown, however cell-mediated immunity plays an important part in the onset of RA. The current study investigated the involvement of the JAK-STAT pathway genes (JAK-1, IL-6, and SOCS-2) in the pathogenesis of RA (Rheumatoid arthritis). METHODOLOGY The study was carried out on thirty male Albino Wistar rats categorised in to the three groups. The AIA (Adjuvant induced animal) model was utilised to study the disease pathogenesis. The haematoxylin and Eosin (H and E) was performed followed by ELISA and expression analyses by RT-q-PCR. The obtained data was analysed using one-way ANOVA (Analysis of Variance). RESULTS Histopathology confirmed that diseased group appeared to be severely impaired, demonstrating manifestations of inflammation with chronic as well as cartilage degenerative changes. Furthermore, chronic inflammation was also noticed in the intertrabecular area. The significant increased levels of JAK1, IL-6 and TYK-2 were recorded among RA group. The gene expression assessment indicated that higher expression of JAK-1 and IL-6 was linked to the further development of RA in the disease group. The SOSC2 (a negative regulator of the JAK-STAT pathway) was significantly (p < 0.01) downregulated. Moreover, SOCS2 may be unable to suppress the transcription of the related JAKs (IL-6 and JAK-1), resulting in the constant release of immune mediators and contributing to the pathophysiology of RA. CONCLUSIONS The JAK-STAT pathway may serve as the target for diagnosing and treating inflammatory and autoimmune disorders (RA). The findings may enhance therapeutic possibilities by investigating the possible implications of JAK-STAT pathway genes as candidates for progressive rheumatoid arthritis therapies.
Collapse
Affiliation(s)
- Maham Ghouri
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| | - Nadir Naveed Siddiqui
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| | - Mehreen Lateef
- Bahria University Medical and Dental College (BUMDC), Karachi, Sindh, Pakistan
| | - Lubna Avesi
- Department of Pathology, Dow University of Health Sciences, Karachi, Sindh, Pakistan.
| | - Rizma Khan
- Department of Biochemistry, Ziauddin University, Clifton, Karachi, Pakistan; Department of Molecular Genetics, Dr. Ziauddin Hospital, North Nazimabad, Karachi, Pakistan.
| | - Humaira Ghauri
- Bahria University Medical and Dental College (BUMDC), Karachi, Sindh, Pakistan.
| | - Ehtisham Asif
- Department of Biotechnology, University of Karachi, Karachi, Sindh, Pakistan
| | - Sitwat Zehra
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| |
Collapse
|
7
|
Gonçalves WA, de Sousa CDF, Teixeira MM, Souza DG. A brief overview of chikungunya-related pain. Eur J Pharmacol 2025; 994:177322. [PMID: 39892450 DOI: 10.1016/j.ejphar.2025.177322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Pain is an important symptom associated with the arboviral disease caused by the Chikungunya virus (CHIKV). For a significant number of patients, this symptom can persist for months or even years, negatively affecting their quality of life. Unfortunately, pharmacological options for this condition are limited and only partially effective, as the underlying mechanisms associated with CHIKV-induced pain are still poorly understood. The re-emergence of CHIKV has led to new outbreaks, and the expected high prevalence of pain in these global events requires new scientific advances to find more effective solutions. Here we review the main aspects of pain caused by CHIKV infection, such as the anatomy of the affected sites, the prevalence and management of this symptom, the diversity of possible cellular and molecular mechanisms, and finally highlight a promising meningeal pathway to elucidate the mechanisms involved in the unsolved problem of CHIKV-associated pain.
Collapse
Affiliation(s)
- William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Carla Daiane Ferreira de Sousa
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany.
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Daniele G Souza
- Laboratório Interação Microrganismo Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| |
Collapse
|
8
|
Li Y, Liu J, Sun Y, Hu Y, Cong C, Chen Y, Fang Y. Interdisciplinary integration strategy reveals the anti-inflammatory efficacy and potential mechanism of Jianpi Qingre Tongluo prescription in rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156625. [PMID: 40073777 DOI: 10.1016/j.phymed.2025.156625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic synovitis and associated with high rates of disability and systemic damage. Jianpi Qingre Tongluo prescription (Huangqin Qingre Chubi Capsule, HQC), an herbal formula with abundant clinical applications, has played a definite role in both clinical and experimental studies of RA. However, the specific mechanisms by which HQC relieves inflammation in RA have not been fully elucidated. OBJECTIVE This study aimed to elucidate the anti-inflammatory efficacy and potential molecular mechanisms of HQC in RA and provide new targets and strategies for its clinical treatment. METHODS An adjuvant-induced arthritis with damp-heat pattern rat model was established to observe the in vivo effects of HQC. Hematoxylin-eosin and toluidine blue staining, and enzyme-linked immunosorbent assay were used to assess potential efficacy. Bioinformatics methods and molecular docking were used to predict potential targets and intervention pathways in which HQC might act on RA. Clinical samples, overexpressed / silenced genes, and pathway agonists were selected to investigate the clinical relevance and regulatory relationships of the pathways. The regulatory mechanism of HQC was confirmed in an in vitro co-culture of neutrophils and fibroblast-like synoviocytes (FLSs) and an in vivo model. RESULTS HQC dose-dependently reversed synovial pathological injury and systemic inflammatory responses in rats in vivo. Integrated bioinformatics and molecular docking identified the p38 mitogen-activated protein kinase (MAPK) signaling pathway and neutrophil extracellular trap (NET) formation as the key mechanisms by which HQC exerts anti-inflammatory effects on RA. Subsequently, a high correlation between circ0005732, p38 MAPK, and clinical features of RA was confirmed in clinical samples. In vitro experiments demonstrated that HQC alleviated the proliferation and inflammatory response of FLSs by regulating circ0005732 expression to inhibit NET formation driven by the p38 MAPK signaling pathway. Finally, RT-qPCR and western blotting confirmed that HQC modulated circ0005732, p38 MAPK pathway, and NET formation to alleviate RA in vivo. CONCLUSION HQC exerts therapeutic effects against RA by modulating circ0005732 to inhibit p38 MAPK signaling pathway-mediated NET generation and inflammation progression.
Collapse
Affiliation(s)
- Yang Li
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Jian Liu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230031, China.
| | - Yue Sun
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yuedi Hu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Chengzhi Cong
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Yiming Chen
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Yanyan Fang
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230031, China; First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| |
Collapse
|
9
|
Mokhtar ER, Abd El-Fattah DA, Hussein NR, Elhakeem H, Gad LA, Mohamed EF, Mohammed RAE, Kasim SA, Elhadad SM, Ali Abd El Rahim MMM, Mohamed MS, Rezk MSM, Fathy D, Hamdy NM, El Magdoub HM. FCGR1A(CD64) expression on monocyte subsets and FIL1Z(IL-37) serum level as biomarkers of rheumatoid arthritis activity: A case controlled study and in silico analysis. Pathol Res Pract 2025; 269:155910. [PMID: 40088754 DOI: 10.1016/j.prp.2025.155910] [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: 11/30/2024] [Revised: 02/13/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Rheumatoid arthritis (RA) is one of the most common chronic autoimmune diseases. Chronic joint inflammation and bone destruction were shown to be caused by expanded monocytes in RA affected individuals. Interleukin-37 which known as FIL1Z(IL-37) is a well-known anti-inflammatory cytokine that plays a negative regulatory role of inflammation in RA. A total of 48 RA patients were divided equally into active RA group and stable RA group using the Disease Activity score (DAS)-28 score. Twenty-four age-and sex-matched healthy subjects were enrolled as controls. The expression level of Fc gamma receptor IA (FCGR1A(CD64)) on monocytes and their subsets in peripheral blood were assessed by flow cytometry (FC) and serum levels of FIL1Z(IL-37) were measured by ELISA. The mean fluorescence intensity (MFI) of FCGR1A(CD64) expressing classical and intermediate monocyte subsets and serum levels of FIL1Z(IL-37) were significantly elevated in RA patients compared to the control and positively correlated with erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), rheumatoid factor (RF), anti-cyclic citrullinated peptide (anti-CCP) and DAS-28 scores. The MFI of FCGR1A(CD64) expressing classical monocyte and serum levels of FIL1Z(IL-37) were significantly elevated in the active RA group compared to the stable RA group. The serum concentration of FIL1Z(IL-37) revealed very high specificity but limited sensitivity in discriminating between active and stable RA patients. Our results demonstrate a strong correlation between serum levels of FIL1Z(IL-37) and FCGR1A(CD64) expression on activated monocytes and their subsets in peripheral blood of RA patients. The results also depict that activated monocytes and their subsets may contribute to the elevated levels of FIL1Z(IL-37) during an active disease status to counter-act the inflammatory process.
Collapse
Affiliation(s)
- Entsar R Mokhtar
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Doaa Aly Abd El-Fattah
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Neama R Hussein
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Heba Elhakeem
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Lamia A Gad
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Eman Fekry Mohamed
- Internal Medicine Department, Faculty of Medicine (for Girls), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | | | - Sammar Ahmed Kasim
- Internal Medicine Department, Faculty of Medicine (for Girls), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Sara M Elhadad
- Internal Medicine Department, Faculty of Medicine (for Girls), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | | | - Maha S Mohamed
- Rheumatology and Rehabilitation Department, Faculty of Medicine (for Girls), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Mohamed Sobhy Mahmoud Rezk
- Rheumatology and Rehabilitation Department, Faculty of Medicine (for Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Doaa Fathy
- Biochemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt.
| | - Hekmat M El Magdoub
- Biochemistry Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt
| |
Collapse
|
10
|
Torregrossa M, Davies L, Hans-Günther M, Simon JC, Franz S, Rinkevich Y. Effects of embryonic origin, tissue cues and pathological signals on fibroblast diversity in humans. Nat Cell Biol 2025:10.1038/s41556-025-01638-5. [PMID: 40263573 DOI: 10.1038/s41556-025-01638-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/18/2025] [Indexed: 04/24/2025]
Abstract
Fibroblasts, once perceived as a uniform cell type, are now recognized as a mosaic of distinct populations with specialized roles in tissue homeostasis and pathology. Here we provide a global overview of the expanding compendium of fibroblast cell types and states, their diverse lineage origins and multifaceted functions across various human organs. By integrating insights from developmental biology, lineage tracing and single-cell technologies, we highlight the complex nature of fibroblasts. We delve into their origination from embryonic mesenchyme and tissue-resident populations, elucidating lineage-specific behaviours in response to physiological cues. Furthermore, we highlight the pivotal role of fibroblasts in orchestrating tissue repair, connective tissue remodelling and immune modulation across diverse pathologies. This knowledge is essential to develop novel fibroblast-targeted therapies to restore steady-state fibroblast function and advance regenerative medicine strategies across multiple diseases.
Collapse
Affiliation(s)
- Marta Torregrossa
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Lindsay Davies
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Machens Hans-Günther
- Department for Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany.
| | - Yuval Rinkevich
- Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Zheng X, Zhang Z, Zheng Y, Liu Y, Wu C, Liang X, Yang X, Tang J, He X, Li C, Wang G, Zhou M. Thermosensitive Light-Driven Smart Platform Induces Apoptosis of Fibroblast-like Synovial Cells and Macrophages for Enhanced Rheumatoid Arthritis Therapy. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40249836 DOI: 10.1021/acsami.5c01723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
Macrophage activation induces rapid proliferation and division of fibroblast-like synovial cells (FLSs), resulting in the degradation of cartilage matrix and bone destruction, which are the main pathological characteristics of rheumatoid arthritis (RA). Inducing apoptosis in these inflammatory cells to mitigate the inflammatory response and alleviate bone damage is a potential therapeutic strategy for RA. In this study, we developed a smart platform for synergistic photothermal therapy (PTT) and chemotherapy by utilizing hyaluronic acid (HA)-modified thermally sensitive liposomes loaded with celastrol (CEL) and gold nanorods (GNRs), termed HA/Lipo-CEL-GNRs, for application in a rat RA model. Under laser irradiation, GNRs exhibited excellent photothermal effects due to localized surface plasmon resonance. The resulting increase in temperature not only effectively eliminated hyperproliferative inflammatory cells in the joints but also triggered CEL release from the thermosensitive liposomes, significantly increasing its concentration in the synovium. The synergistic effect of PTT and chemotherapy significantly promoted the apoptosis of FLSs and macrophages and effectively suppressed the inflammatory response in the RA microenvironment. In summary, multifunctional thermosensitive HA/Lipo-CEL-GNRs represent promising nanotherapeutic platforms capable of achieving light-driven enrichment of heat and therapeutic agents, significantly preventing the progression of RA.
Collapse
Affiliation(s)
- Xiu Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Drug Laboratory, Department of Pharmacy, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, Sichuan 610000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chengxi Wu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Tang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Guan Wang
- Department of Orthopaedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
12
|
Hashimoto T, Tsuboi K, Abe T, Yoshikawa T, Noguchi K, Furukawa T, Tateishi K, Terashima Y, Sibanuma N, Azuma N, Yamane T, Matsui K, Hashiramoto A. Nocturnal baricitinib administration leads to rapid drug responses in rheumatoid arthritis: a multicenter non-randomized controlled study. Arthritis Res Ther 2025; 27:91. [PMID: 40247352 PMCID: PMC12004773 DOI: 10.1186/s13075-025-03555-2] [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: 01/27/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Inflammatory cytokine levels exhibit a circadian rhythm in sera, peaking from late night to early morning in patients with rheumatoid arthritis (RA). This cytokine kinetics is a recognized therapeutic target. This clinical study aimed to evaluate the effectiveness of night-time baricitinib administration based on cytokine secretion. METHODS In this 52-week multicenter non-randomized controlled study, 122 patients with RA were assigned to four groups: baricitinib 2 mg morning (BAR2MORN), 2 mg evening (BAR2EVE), 4 mg morning (BAR4MORN), or 4 mg evening (BAR4EVE). The primary endpoint was assessed using the 20% improvement in the American College of Rheumatology criteria (ACR20) at week 12. The secondary endpoints were ACR20/50/70 and changes in the clinical disease activity index (CDAI) through 52 weeks. The results were evaluated using the propensity score inverse probability of treatment weighted to reduce selection bias in patient background. RESULTS BAR4EVE resulted in better primary endpoint improvement than BAR4MORN (78.2 vs. 43.3%; p < 0.001). No difference in improvement was observed in the primary endpoint between BAR2EVE and BAR2MORN (75.5 vs. 60.6%; p = 0.10). However, BAR2EVE demonstrated higher ACR20 at weeks 4, 24, and 52 and ACR50 at weeks 4 and 12 than BAR2MORN. BAR4EVE demonstrated higher ACR20/50 at weeks 4, 8, and 12 and ACR70 at weeks 8, 12, and 24 than BAR4MORN. CDAI changes were significantly reduced in BAR4EVE than in BAR4MORN at weeks 4 and 8. CONCLUSION Chronotherapy targeting cytokine secretion resulted in rapid drug response, proposing a new potential application for JAK inhibitors. TRIAL REGISTRATION UMIN000040094, July 1, 2020.
Collapse
Affiliation(s)
- Teppei Hashimoto
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Kazuyuki Tsuboi
- Department of Rheumatology, Kobe City Medical Center West Hospital, Kobe, Japan
| | - Takeo Abe
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Takahiro Yoshikawa
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
- Department of Rheumatology, Amagasaki Central Hospital, Amagasaki, Japan
| | - Kazuteru Noguchi
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Tetsuya Furukawa
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Koji Tateishi
- Department of Orthopedics, Hakuhokai Kakogawa Hospital, Kakogawa, Japan
| | | | - Nao Sibanuma
- Department of Orthopedics, Kobe Kaisei Hospital, Kobe, Japan
| | - Naoto Azuma
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Takashi Yamane
- Department of Rheumatology, Kakogawa Central City Hospital, Kakogawa, Japan
| | - Kiyoshi Matsui
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
- Department of Rheumatology, Amagasaki Central Hospital, Amagasaki, Japan
| | - Akira Hashiramoto
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| |
Collapse
|
13
|
Farquhar HJ, Huria T, Beckert L, Frampton CMA, Haslett J, Stamp LK. Incidence, prevalence and mortality of rheumatoid arthritis in Canterbury, Aotearoa New Zealand. Intern Med J 2025. [PMID: 40235064 DOI: 10.1111/imj.70068] [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: 10/18/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND There is relatively little research regarding the frequency of rheumatoid arthritis (RA) in Canterbury and no previous studies comparing the mortality of RA in Aotearoa New Zealand to a general population. AIMS To determine the incidence, prevalence and mortality of RA in Canterbury, Aotearoa New Zealand. METHODS Clinical records of persons attending the Christchurch Hospital rheumatology service between 1 January 2006 and 31 December 2008 (Period 1) and between 1 January 2011 and 31 December 2013 (Period 2) were reviewed to identify individuals who met the 2010 American College of Rheumatology/European League Against Rheumatism RA classification criteria. Medical records for persons treated by private sector rheumatologists were also reviewed. The period prevalence and average annual incidence for each time period were calculated. Prevalence and incidence rates for the total population were age- and sex-adjusted based on the usually resident population from the 2013 national Census. Standardised mortality ratios (SMRs) compared to the general population were calculated. RESULTS The annual incidence was 24.40 per 100 000 (95% confidence interval (CI): 21.36-27.43) for Period 1 and 22.06 per 100 000 (95% CI: 19.32-24.81) for Period 2. The period prevalence was 442.65 per 100 000 (95% CI: 420.08-465.04) for Period 1 and 448.66 per 100 000 (95% CI: 427.31-470.01) for Period 2. The all-cause SMR for individuals with incident RA was 1.25 (95% CI: 0.95-1.60). The SMR for all prevalent RA during Period 1 was 2.01 (95% CI: 1.85-2.19) and for Period 2 was 1.87 (95% CI: 1.69-2.06). CONCLUSIONS The frequency of RA in the Canterbury population is comparable to other regions. Mortality for all individuals with prevalent RA was approximately two times higher than the general population.
Collapse
Affiliation(s)
- Hamish J Farquhar
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Tania Huria
- Māori Indigenous Health Innovation Department, University of Otago Christchurch, Christchurch, New Zealand
| | - Lutz Beckert
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | | | - Janine Haslett
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
14
|
Škorjanc A, Smrkolj V, Umek N. GOReverseLookup: A gene ontology reverse lookup tool. Comput Biol Med 2025; 191:110185. [PMID: 40239235 DOI: 10.1016/j.compbiomed.2025.110185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND AND OBJECTIVE The Gene Ontology (GO) project has been pivotal in providing a structured framework for characterizing genes and annotating them to specific biological concepts. While traditional gene annotation primarily focuses on mapping genes to GO terms, descriptors of biological concepts, there is a growing need for tools facilitating reverse querying. This paper introduces GOReverseLookup, a novel tool designed to identify over- or underrepresented genes in researcher-defined states of interest (phenotypes), described by sets of GO terms. GOReverseLookup supplements the existing power of Gene Ontology by the possibility of orthologous gene querying across several databases, such as Ensembl and UniProtKB. This combination allows for a more nuanced identification of significant genes across a range of cross-species research contexts. METHODS GOReverseLookup queries genes associated with input GO terms. Bundles of GO terms encapsulate user-defined states of interest, e.g., angiogenesis. In the second stage of the analysis, all GO terms associated with each gene are fetched, and finally, the statistical relevance of the genes being involved in one (or all) of the defined states of interests is computed. RESULTS The two presented use cases illustrate its utility in discovering genes related to rheumatoid arthritis and genes linked with chronic inflammation and tumorigenesis. In both cases, GOReverseLookup discovered a substantial number of genes significantly associated with the aforementioned states of interest. CONCLUSIONS GOReverseLookup proves to be a valuable resource for unraveling the genetic basis of phenotypes, with diverse practical potentials in functional genomics, systems biology, and drug discovery. We anticipate that GOReverseLookup will significantly aid in identifying potential gene targets during the initial research phases.
Collapse
Affiliation(s)
- Aljoša Škorjanc
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia
| | - Vladimir Smrkolj
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia; National Institute of Chemistry, Hajdrihova ulica 19, Ljubljana, Slovenia
| | - Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia.
| |
Collapse
|
15
|
Cho SK, Choi SR, Kim HW, Nam E, Lee SW, Lee SS, Lee HS, Park SH, Lee YA, Chang SH, Park MC, Kim HA, Kwok SK, Kim HS, Yoon BY, Kim YG, Kim HR, Kim JH, Lee J, Choi J, Uhm WS, Sung YK. Real-world comparative effectiveness study of Janus kinase inhibitors compared to biologic disease-modifying antirheumatic drugs in Korean patients with rheumatoid arthritis. Semin Arthritis Rheum 2025; 73:152720. [PMID: 40245585 DOI: 10.1016/j.semarthrit.2025.152720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVES To evaluate the real-world effectiveness and safety of Janus kinase inhibitors (JAKis) compared to biologic disease-modifying antirheumatic drugs (bDMARDs) in Korean patients with rheumatoid arthritis (RA) who have not previously been treated with either JAKis or bDMARDs. METHODS This prospective, multicenter, observational study was conducted at 17 centres in the Republic of Korea. Patients with an inadequate response to methotrexate were enrolled and started treatment with either JAKis or bDMARDs. The primary endpoint was the proportion of patients achieving low disease activity (LDA) at 24 weeks, measured by the disease activity score (DAS) 28-erythrocyte sedimentation rate (ESR). Secondary endpoints included the remission rate at 24 weeks, and LDA and remission rates at 48 weeks. Safety was assessed by the exposure-adjusted event rate (EAER) of adverse events (AEs), adjusted for length of the follow-up period and presented per 100 person-years. RESULTS A total of 506 patients were enrolled, with 253 patients in each group. Among bDMARD users, 60.1 % received tumour necrosis factor inhibitors (TNFis; n = 152) and 39.9 % received non-TNFis (n = 101). At 24 weeks, 48.2 % of the JAKi group achieved LDA, as did 42.7 % of the bDMARD group. Remission rates at 24 weeks were 28.9 % for the JAKi group and 27.3 % for the bDMARD group. At 48 weeks, there were no significant intergroup differences in the EAER of overall AEs. CONCLUSIONS In this observational real-world study of Korean patients with RA who were eligible for targeted therapy, JAKis demonstrated comparable effectiveness and safety to bDMARDs.
Collapse
Affiliation(s)
- Soo-Kyung Cho
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Se Rim Choi
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Hye Won Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Eunwoo Nam
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Sang Won Lee
- Department of Pharmacology, Hanyang University, College of Medicine, Seoul, South Korea
| | - Shin-Seok Lee
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Hye-Soon Lee
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University School of Medicine, Guri, South Korea
| | - Sung-Hoon Park
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, South Korea
| | - Yeon-Ah Lee
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Sung Hae Chang
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, South Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Suwon, South Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun-Sook Kim
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, South Korea
| | - Bo Young Yoon
- Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, South Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Jae Hoon Kim
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Jisoo Lee
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Jeongim Choi
- Division of Rheumatology, Department of Internal Medicine, Busan Saint Mary's Hospital, Busan, South Korea
| | | | - Yoon-Kyoung Sung
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea.
| |
Collapse
|
16
|
Yin X, Wei H, Guo L, Liu B, Peng Y, Zhou M, Qiu Y, Qu R, Gao Y, Wu Q, Jiang W, Bi H, Guo D. Therapeutic effect of miR-30b-5p-loaded lentivirus on experimental autoimmune uveitis via inhibiting Notch signaling activation. J Transl Med 2025; 23:426. [PMID: 40211315 PMCID: PMC11987260 DOI: 10.1186/s12967-025-06438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Uveitis is a common recurrent autoimmune disease that seriously endangers the visual health of patients. MicroRNAs (miRNAs) are closely related to a series of autoimmune diseases. METHODS The present study aimed to investigate the effect of miR-30b-5p on experimental autoimmune uveitis (EAU) and its role in Notch signal activation as well as T helper (Th) cell differentiation, the relationship between miR-30b-5p levels and Notch signal activation, as well as Th cell differentiation in uveitis was further explored through flow cytometry, Immunohistochemistry immunofluorescence staining, PCR Array, and Ingenuity Pathway Analysis, and other technical methods to determine the fidelity of miR-30b-5p strategies in treating uveitis in vivo and in vitro. RESULTS We demonstrated that ocular inflammation was significantly alleviated in EAU rats after miR-30b-5p intervention. miR-30b-5p could effectively inhibit Notch signaling activation and Th17 cell differentiation both in vitro and in vivo, and the Th17/Treg ratios were also notably decreased. Moreover, both Notch signaling and Th17 activation pathways were enriched and activated, in which Notch1 was the upstream regulatory molecule of Dll4 and IL-10 was an up-regulated upstream regulatory network molecule. Furthermore, miR-30b-5p could significantly reduce apoptosis in vitro, and clinical in vitro cell studies have shown that inactivating Notch pathway can improve the imbalance of Th17/Treg and cell apoptosis in T lymphocytes of patients with uveitis. CONCLUSIONS Together these studies identify that miR-30b-5p can significantly inhibit Notch signaling activation and Th17 cell differentiation, thereby restoring the Th17/Treg balance to treat uveitis, which may provide new insights into treating uveitis using miRNA interfering strategies.
Collapse
Affiliation(s)
- Xuewei Yin
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
| | - Huixia Wei
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China
| | - Lijie Guo
- Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 510000, China
| | - Bin Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Yuan Peng
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Mengxian Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Yan Qiu
- Shandong University of Traditional Chinese Medicine Second Affiliated Hospital, Jinan, 250002, China
| | - Ruyi Qu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Yane Gao
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China
| | - Qiuxin Wu
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China
| | - Wenjun Jiang
- Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China.
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China.
- Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, 250002, China.
| |
Collapse
|
17
|
Charneca S, Hernando A, Almada-Correia I, Polido-Pereira J, Vieira A, Sousa J, Almeida AS, Motta C, Barreto G, Eklund KK, Alonso-Pérez A, Gómez R, Cicci F, Mauro D, Pinho SS, Fonseca JE, Costa-Reis P, Guerreiro CS. TASTY trial: protocol for a study on the triad of nutrition, intestinal microbiota and rheumatoid arthritis. Nutr J 2025; 24:52. [PMID: 40189532 PMCID: PMC11974026 DOI: 10.1186/s12937-025-01089-6] [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: 12/08/2024] [Accepted: 02/06/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND The gut microbiota has been implicated in the onset and progression of Rheumatoid Arthritis (RA). It has been proposed that gut dysbiosis impairs gut barrier function, leading to alterations in mucosal integrity and immunity. This disruption allows bacterial translocation, contributing to the perpetuation of the inflammatory process. Since diet is recognised as a key environmental factor influencing the gut microbiota, nutritional interventions targeting RA activity are currently being explored. This study aims to investigate whether a dietary intervention based on a typical Mediterranean Diet enriched with fermented foods (MedDiet +) can impact the gut microbiota, intestinal permeability, and RA-related outcomes. METHODS One hundred RA patients are being recruited at Unidade Local de Saúde (ULS) Santa Maria in Lisbon, Portugal, and randomly assigned to either the intervention (MedDiet +) or the control group. The 12-week nutritional intervention includes a personalised dietary plan following the MedDiet + pattern, along with educational resources, food basket deliveries, and clinical culinary workshops, all developed and monitored weekly by registered dietitians. The control group receives standardised general healthy diet recommendations at baseline. The intervention's effects will be assessed by evaluating disease activity, functional status, quality of life, intestinal permeability, endotoxemia, inflammatory biomarkers, intestinal and oral microbiota, serum proteomics, and serum glycome profile characterisation. DISCUSSION We anticipate obtaining integrative insights into the interplay between diet, the gut, and RA, while also exploring the underlying mechanisms driving these changes. This study, conducted by a multidisciplinary research team of registered dietitians, rheumatologists, biologists, and immunologists, aims to bridge the current gap between nutrition-related knowledge and RA. TRIAL REGISTRATION Registered in ClinicalTrials.gov (NCT06758817; date of registry: January 6th 2025).
Collapse
Affiliation(s)
- Sofia Charneca
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Hernando
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Inês Almada-Correia
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joaquim Polido-Pereira
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Adriana Vieira
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joana Sousa
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Santos Almeida
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Carla Motta
- Departamento de Alimentação e Nutrição, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
| | - Gonçalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland.
| | - Kari K Eklund
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland
- Department of Rheumatology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Francesco Cicci
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Daniele Mauro
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3s), University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - João Eurico Fonseca
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Patrícia Costa-Reis
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Pediatric Rheumatology Unit, ULS Santa Maria, Lisbon, Portugal
| | - Catarina Sousa Guerreiro
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| |
Collapse
|
18
|
Rydland A, Heinicke F, Nyman TA, Trøseid AMS, Flåm ST, Stensland M, Gehin J, Eikeland J, Øvstebø R, Mjaavatten MD, Lie BA. Newly-diagnosed rheumatoid arthritis patients have elevated levels of plasma extracellular vesicles with protein cargo altered towards inflammatory processes. Sci Rep 2025; 15:11632. [PMID: 40185859 PMCID: PMC11971360 DOI: 10.1038/s41598-025-96325-8] [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: 12/16/2024] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Extracellular vesicles (EVs) are implicated in rheumatoid arthritis (RA) but have mainly been assessed in RA patients taking disease modifying anti-rheumatic drugs. EVs are nanoparticles important in cell-cell communication and their molecular cargo are biomarker candidates. We characterized the protein profiles of EVs from blood plasma from newly diagnosed, treatment naïve RA patients (N = 32) and compared them to healthy controls (N = 20), by size exclusion chromatography-based EV enrichment coupled with high-resolution quantitative proteomics. The RA patients had higher EV concentration and larger EVs than controls. A total of 682 EV proteins were reliably quantified, and the overall profiles were distinctly different between patients and controls. Specifically, 26 proteins were significantly upregulated and 31 downregulated in RA patients, with several proteins acting in inflammatory networks and with immunologically important upstream regulators. The RA associated EVs appear, based on the tissue expression of their cargo proteins, to originate mainly from hepatocytes or immune cells, like neutrophils. Interestingly, the strongest RA associated EV proteins were inflammatory molecules, like SAA1 and S100A9, already suggested as biomarkers in RA. Furthermore, the RA associated EV proteins were generally not correlated with total serum protein levels, stressing the importance of EV transport of inflammatory proteins in RA pathogenesis.
Collapse
Affiliation(s)
- Anne Rydland
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
| | - Fatima Heinicke
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Department of Biostatistics, Institute of Basic Medical Sciences, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anne-Marie Siebke Trøseid
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Siri T Flåm
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Maria Stensland
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Johanna Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Joakim Eikeland
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Reidun Øvstebø
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maria Dahl Mjaavatten
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Division of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
19
|
Tanaka Y, Horiuchi N, Sasajima C, Matsumoto R, Kawanishi M, Uchida S, Takeuchi T. Effect of Ozoralizumab Administration with or without Methotrexate in Patients with Rheumatoid Arthritis: A Post-Hoc Analysis. Rheumatol Ther 2025; 12:283-296. [PMID: 39869270 PMCID: PMC11920534 DOI: 10.1007/s40744-024-00737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
INTRODUCTION Ozoralizumab (OZR) is a novel tumor necrosis factor (TNF) inhibitor that was launched in Japan for treating patients with rheumatoid arthritis (RA) who have had an inadequate response to existing therapies. This post-hoc analysis aimed to compare the efficacy of OZR administered without methotrexate (MTX) with placebo or OZR administration in combination with MTX. METHODS We analyzed the OZR group (30 mg) in the NATSUZORA trial (non-MTX, open trial) (OZR group; n = 94) and the placebo group (MTX group; n = 75) and the 30-mg OZR group (OZR + MTX group; n = 152) in the OHZORA trial (combined MTX, double-blind trial), and the covariates were adjusted by propensity score matching. Subsequently, the American College of Rheumatology (ACR) 20/50/70 response rates from baseline to 24 or 52 weeks were compared. Furthermore, to compare longitudinal data on disease activity indicators, a mixed-effects model for repeated-measures analyses was used. RESULTS Comparing the OZR and MTX groups, 52 patients were matched in each group. The OZR group showed improvements in the ACR20 (OZR group, 67.3% vs. MTX group, 34.6%, p = 0.001), ACR50 (51.9% vs. 17.3%, p < 0.001), and ACR70 (26.9% vs. 11.5%, p = 0.047) response rates compared to those in the MTX group. Comparing the OZR and OZR + MTX groups, 77 patients were matched in each group. No significant difference was observed in the ACR20 response rate (OZR group, 58.4% vs. OZR + MTX group, 70.1%, p = 0.130). However, the OZR + MTX group showed higher ACR50 (44.2% vs. 62.3%, p = 0.024) and ACR70 (29.9% vs. 45.5%, p = 0.046) response rates. CONCLUSION OZR administration without MTX was associated with an improvement in the signs and symptoms of RA compared to placebo administration (continuation of MTX monotherapy). OZR and MTX administration showed better efficacy than OZR administration alone.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Kitakyushu, Fukuoka, 807-8555, Japan.
| | - Nobuko Horiuchi
- Taisho Pharmaceutical Co., Ltd., 3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Cosmo Sasajima
- Taisho Pharmaceutical Co., Ltd., 3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Rumiko Matsumoto
- Taisho Pharmaceutical Co., Ltd., 3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Masafumi Kawanishi
- Taisho Pharmaceutical Co., Ltd., 3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Saeko Uchida
- Taisho Pharmaceutical Co., Ltd., 3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Tsutomu Takeuchi
- Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
- Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
20
|
Miyazaki Y, Nakayamada S, Tanaka H, Hanami K, Fukuyo S, Kubo S, Miyagawa I, Yamaguchi A, Todoroki Y, Inoue Y, Ueno M, Tanaka Y. Comparison of anti-IL-6 receptor and JAK inhibitors in patients with rheumatoid arthritis from the real-world practice FIRST registry. Rheumatology (Oxford) 2025; 64:1627-1636. [PMID: 38889301 DOI: 10.1093/rheumatology/keae334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVES A molecular-targeted drug that is suitable as the second choice for patients with rheumatoid arthritis (RA) who show an inadequate response to the first biological disease-modifying antirheumatic drug (bDMARD) is unknown. This study aimed to analyse the efficacy and safety of interleukin-6 receptor (IL-6Ri) and Janus kinase inhibitors (JAKis), often selected as molecular-targeted drugs for second or subsequent treatments. METHODS The efficacy and safety of JAKis and IL-6Ri were compared using propensity score-based inverse probability of treatment weighting (PS-IPTW) using propensity scores after 26 weeks of therapy in patients with RA. RESULTS The remission rate at week 26, determined by the clinical disease activity index (CDAI), and the incidence of infection were higher in the JAKis than in the IL-6Ri group. The CDAI trajectories were divided into four according to the growth mixture modeling. IL-6Ri demonstrated greater efficacy in RA patients with ineffective to single bDMARD therapy compared with those with multiple ineffective bDMARDs. In patients who failed to respond to one bDMARD, there was no significant difference in the CDAI remission rate at week 26 between the JAKis (29.1%) and IL-6Ri (21.8%) groups (P = 0.21). However, for patients who did not respond to at least two bDMARDs, the CDAI remission rate at week 26 was higher in the JAKis than in the IL-6Ri group. CONCLUSIONS IL-6Ri offers a superior balance of efficacy and safety compared with JAKis for RA patients unresponsive to one bDMARD. However, JAKis may suit patients who do not respond to multiple bDMARDs.
Collapse
Affiliation(s)
- Yusuke Miyazaki
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroaki Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Kentaro Hanami
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Shunsuke Fukuyo
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Satoshi Kubo
- Department of Internal Medicine (Molecular Targeted Therapy), University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Ippei Miyagawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Ayako Yamaguchi
- Department of Laboratory and Transfusion Medicine, Hospital of the University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- Department of Internal Medicine (Molecular Targeted Therapy), University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshino Inoue
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Masanobu Ueno
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
21
|
Chen L, Zhao J, Meng Q. From genetic variants to therapeutic targets: insights into understanding rheumatoid arthritis. Front Immunol 2025; 16:1556971. [PMID: 40236704 PMCID: PMC11996834 DOI: 10.3389/fimmu.2025.1556971] [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: 01/07/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects multiple systems and is driven by various factors, including interactions between genetic and environmental elements. Over the past few decades, genome-wide association studies (GWAS) have been instrumental in regard to identifying genetic and environmental risk factors associated with RA susceptibility and pathogenesis. The recent discoveries of novel genetic susceptibility loci and pathways offer promising therapeutic targets for RA and precision medicine. More than 100 genetic loci have been identified in RA patients. In this review, we have focused on more than 40 genes that have been supported by evidence to be closely associated with the development of RA. These include genes involved in various mechanisms, such as loss of self-tolerance, autoimmune antibody production (e.g., HLA-DRB1, HLA-DPB1), inflammatory signaling and bone destruction (e.g., PTPN22, CCR6), complication (e.g., HLA-DQB1, IRF5), and differential drug responses (e.g., HLA-E, NKG2D). These novel players and mechanisms enhance our understanding of the RA pathogenesis and also provide a reference for personalized and precise medicine, including diagnosis and treatment.
Collapse
Affiliation(s)
- Lu Chen
- Department of Traditional Chinese Medicine, Aviation General Hospital, Beijing, China
| | - Jianan Zhao
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingliang Meng
- Department of Rheumatism, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
22
|
Jin L, Wang Z, Peng C, He M, Wang F, He H, Liu C. Microneedle Assisted Melittin-Chondroitin Sulfate Administration for the Transdermal Therapy of Rheumatoid Arthritis. Adv Healthc Mater 2025; 14:e2400543. [PMID: 39972637 PMCID: PMC11973948 DOI: 10.1002/adhm.202400543] [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: 02/26/2024] [Revised: 06/09/2024] [Indexed: 02/21/2025]
Abstract
Rheumatoid arthritis (RA), a persistent and debilitating chronic disease characterize by joint damage and deformity, significantly impairs the life quality of patients and presents challenges for conventional drug administration due to organ damage and unsatisfactory therapeutic outcomes. To address these challenges, this study introduces an innovative hydrogel cryo-microneedle patch (CMNP)-mediated local administration system, primarily composed of chondroitin sulfate (CS), to deliver the potent anti-inflammatory drug melittin (MEL). This innovative approach not only circumvents organ impairment but also enhances patient compliance. The acute toxicity of MEL is effectively mitigated by electrostatic binding with CS molecules, forming MC complexes that induce apoptosis in fibroblast-like synoviocytes (FLS). The MC-loaded CMNPs (MC@CCMNPs) exhibit remarkable therapeutic capacity with a notably meliorated joint damage and suppressed arthritis severity in the RA rat model. Therefore, MC@CCMNPs emerge as a promising anti-inflammatory and safe therapy for RA treatment, as well as for other inflammation related chronic diseases.
Collapse
Affiliation(s)
- Lili Jin
- Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and TechnologyShanghai200237P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
- School of Material Science and EngineeringEast China University of Science and TechnologyShanghai200237P. R. China
| | - Zhenhui Wang
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
- School of Material Science and EngineeringEast China University of Science and TechnologyShanghai200237P. R. China
| | - Cheng Peng
- The Education Ministry Key Lab of Resource ChemistryJoint International Research Laboratory of Resource Chemistry of Ministry of EducationShanghai Key Laboratory of Rare Earth Functional MaterialsShanghai Frontiers Science Center of Biomimetic CatalysisShanghai Normal UniversityShanghai200234P. R. China
| | - Miao He
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Feng Wang
- The Education Ministry Key Lab of Resource ChemistryJoint International Research Laboratory of Resource Chemistry of Ministry of EducationShanghai Key Laboratory of Rare Earth Functional MaterialsShanghai Frontiers Science Center of Biomimetic CatalysisShanghai Normal UniversityShanghai200234P. R. China
| | - Hongyan He
- Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and TechnologyShanghai200237P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
- School of Material Science and EngineeringEast China University of Science and TechnologyShanghai200237P. R. China
| | - Changsheng Liu
- Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and TechnologyShanghai200237P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| |
Collapse
|
23
|
Li F, Xian D, Yang K. Mendelian randomization and mediation analysis reveal the role of immune cell subsets in the causal pathways between blood cell perturbation responses and rheumatoid arthritis. Clin Rheumatol 2025; 44:1537-1548. [PMID: 40072781 DOI: 10.1007/s10067-025-07387-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/11/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by complex immune interactions. Elucidating the causal relationships between blood cell perturbations, immune cell subsets, and RA can provide valuable insights into its pathogenesis. METHODS This study employed bidirectional two-sample Mendelian Randomization (MR) to explore the causal effects of blood cell perturbations on RA risk, with a focus on immune cell mediation. Genetic data from large-scale Genome-Wide Association Studies (GWAS) were utilized to select instrumental variables (IVs) for exposure, mediator, and outcome. Inverse Variance Weighted (IVW) analysis was applied, supplemented by sensitivity tests. Mediation analysis was conducted to assess the indirect effects mediated by immune cells. RESULTS Significant causal associations were identified between perturbations in reticulocytes, monocytes, and lymphocytes and specific immune cell subsets, including CD3 + CD39 + regulatory T cells (Tregs) and CD45RA + terminally differentiated CD8 + T cells (CD45RA + TD CD8 + cells). Erythropoiesis perturbation was associated with a reduced RA risk, while perturbations in monocytes and lymphocytes were found to facilitate RA progression through immune-mediated mechanisms. CONCLUSION This study underscores the pivotal role of immune cell subsets in mediating the effects of blood cell perturbations on RA development. These findings suggest that targeting immune cell-mediated pathways, particularly those involving Tregs and CD8 + T cells, can provide new therapeutic strategies for RA management. Key Points • Causal Relationships: Mendelian randomization (MR) analysis identified significant causal relationships between specific blood cell disturbances (e.g., reticulocytes, monocytes, and lymphocytes) and rheumatoid arthritis (RA). • Role of Immune Cells: CD3 + CD39 + regulatory T cells (Tregs) and CD45RA + Terminally Differentiated CD8 + T cells (CD45RA + TD CD8 + cells) mediate the association between blood cell disturbances and RA. • Protective Role of Reticulocytes: Reticulocyte disturbances under potassium chloride (KCl) conditions are negatively associated with RA, potentially protecting joints from inflammatory damage by reducing oxidative stress. • Protective Role of Non-Classical Monocytes: Baseline disturbances in monocyte median side scatter are negatively associated with RA, suggesting non-classical monocytes may reduce RA-related inflammation. • Positive Association of Lymphocyte Disturbances with RA: Lymphocyte side scatter standard deviation under colchicine disturbances shows a significant positive association with RA, indicating abnormal T cell activation may exacerbate RA progression.AQ.
Collapse
Affiliation(s)
- Feng Li
- Spinal Orthopedics Department I, Neijiang Hospital of Traditional Chinese Medicine, Neijiang City, Sichuan Province, China
| | - Dehai Xian
- Laboratory of Human Anatomy, School of Basic Medicine Anatomy , Southwest Medical University, Xianglin Road, Longmatan District, Luzhou City, Sichuan Province, China.
| | - Kaiwen Yang
- Laboratory of Human Anatomy, School of Basic Medicine Anatomy , Southwest Medical University, Xianglin Road, Longmatan District, Luzhou City, Sichuan Province, China.
| |
Collapse
|
24
|
Wu H, Yin F, Wang Y, Tang Z, Hong H, Jiang T, Guo J, Ma X, Yao G, Zhang H. Increased macrophage migration inhibitory factor is associated with inflammation in patients with rheumatoid arthritis. Clin Rheumatol 2025; 44:1475-1484. [PMID: 39953337 DOI: 10.1007/s10067-025-07361-8] [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] [Received: 08/22/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE The macrophage migration inhibitory factor (MIF) in the plasma, hydrops articuli, and synovium, and its relationship with laboratory indexes in patients with rheumatoid arthritis (RA) were determined, for the purpose to reveal the role of MIF on the pathogenesis of RA. METHODS MIF mRNA expression in PBMCs was detected by qPCR. Plasma MIF was measured by enzyme linked immunosorbent assay (ELISA). MIF in hydrops articuli and synovium from RA patients and OA patients was evaluated by immunofluorescence (IF) and immunohistochemistry (IHC). The relationship between MIF and laboratory indexes of RA patients was analyzed. Human fibroblast-like synoviocytes (FLS) were treated with recombinant human MIF, and expression of inflammatory factors was determined by qPCR. The matrix metalloproteinase (MMP) 9 and extracellular regulated protein kinases (ERK)1/2 in FLS with MIF treatment were detected. RESULTS MIF is significantly increased in plasma and hydrops articuli in RA patients. The expression of multiple inflammatory factors and MMPs was increased in RA patients and in FLS with rhMIF treatment. MIF was correlated with laboratory indexes in RA patients. Mechanistically, MIF promoted production of MMP9 by FLS through the ERK1/2 pathway. CONCLUSION Our results indicated that increased MIF was correlated with disease activity of RA patients. These findings also suggested that MIF induced multiple inflammatory factors and MMP 9 in FLS via ERK 1/2 pathway. Key Points • MIF plays a key role in the initiation of RA by promoting the expression of various inflammatory factors in FLS and MMPs. • This study provides a basis for MIF-targeted RA clinical therapy and for exploring the feasibility of MIF as a therapeutic target for RA. • Increased MIF correlates with disease activity in RA patients.
Collapse
Affiliation(s)
- Haolin Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Fanzhang Yin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yue Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zhicheng Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Huiming Hong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Tingting Jiang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Junqiao Guo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xiaolei Ma
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Huayong Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
25
|
Qian C, Wang Q, Qiao Y, Xu Z, Zhang L, Xiao H, Lin Z, Wu M, Xia W, Yang H, Bai J, Geng D. Arachidonic acid in aging: New roles for old players. J Adv Res 2025; 70:79-101. [PMID: 38710468 PMCID: PMC11976421 DOI: 10.1016/j.jare.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Arachidonic acid (AA), one of the most ubiquitous polyunsaturated fatty acids (PUFAs), provides fluidity to mammalian cell membranes. It is derived from linoleic acid (LA) and can be transformed into various bioactive metabolites, including prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs), hydroxy-eicosatetraenoic acids (HETEs), leukotrienes (LTs), and epoxyeicosatrienoic acids (EETs), by different pathways. All these processes are involved in AA metabolism. Currently, in the context of an increasingly visible aging world population, several scholars have revealed the essential role of AA metabolism in osteoporosis, chronic obstructive pulmonary disease, and many other aging diseases. AIM OF REVIEW Although there are some reviews describing the role of AA in some specific diseases, there seems to be no or little information on the role of AA metabolism in aging tissues or organs. This review scrutinizes and highlights the role of AA metabolism in aging and provides a new idea for strategies for treating aging-related diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW As a member of lipid metabolism, AA metabolism regulates the important lipids that interfere with the aging in several ways. We present a comprehensivereviewofthe role ofAA metabolism in aging, with the aim of relieving the extreme suffering of families and the heavy economic burden on society caused by age-related diseases. We also collected and summarized data on anti-aging therapies associated with AA metabolism, with the expectation of identifying a novel and efficient way to protect against aging.
Collapse
Affiliation(s)
- Chen Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Ze Xu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Linlin Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Zhixiang Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
26
|
Kubo S, Tanaka Y. Pursuing Precision Medicine in Managing Rheumatoid Arthritis. Int J Rheum Dis 2025; 28:e70239. [PMID: 40269471 PMCID: PMC12018724 DOI: 10.1111/1756-185x.70239] [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: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Rheumatoid arthritis, characterized by immune dysregulation and joint destruction, is managed through a stepwise algorithm that combines methotrexate with biological and targeted synthetic disease-modifying antirheumatic drugs. Despite considerable advances, the lack of reliable biomarkers for selecting the most effective medication, especially in Phase II and beyond, remains a significant obstacle. As a result, achieving early clinical remission in all patients continues to be challenging. Rheumatoid arthritis demonstrates considerable clinical and molecular diversity, influenced by both genetic predispositions and environmental factors. Recent scientific and technological advances have shed light on the pathogenesis of rheumatoid arthritis, facilitating the stratification of patients into distinct phenotypic subgroups and potentially optimizing the choice of targeted therapies. However, persistent challenges include the high costs and logistical demands of these methodologies, as well as the complexities of conducting large-scale clinical trials. This review highlights the intricate pathogenesis of rheumatoid arthritis and underscores the need to address the disease's heterogeneity through precision medicine. Moving forward, a deeper investigation into rheumatoid arthritis pathogenesis, encompassing both genetic and environmental factors, is crucial. Pursuing precision medicine, grounded in accurate patient stratification, should be embraced as a "moonshot" objective in rheumatoid arthritis treatment, aiming to achieve transformative breakthroughs in management.
Collapse
Affiliation(s)
- Satoshi Kubo
- Department of Molecular Targeted TherapeuticsUniversity of Occupational and Environmental HealthKitakyushuJapan
- The First Department of Internal MedicineUniversity of Occupational and Environmental HealthKitakyushuJapan
| | - Yoshiya Tanaka
- Department of Molecular Targeted TherapeuticsUniversity of Occupational and Environmental HealthKitakyushuJapan
- The First Department of Internal MedicineUniversity of Occupational and Environmental HealthKitakyushuJapan
| |
Collapse
|
27
|
Shao Y, Yang Y, Yang X, Xu Z, Zhang H, Li N, Xu H, Zhao Y, Wang Y, Shi Q, Liang Q. A diagnostic model for assessing the risk of osteoporosis in patients with rheumatoid arthritis based on bone turnover markers. Arthritis Res Ther 2025; 27:75. [PMID: 40170112 PMCID: PMC11959775 DOI: 10.1186/s13075-025-03544-5] [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: 09/01/2024] [Accepted: 03/22/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND The risk of developing osteoporosis (OP) is increased in patients with rheumatoid arthritis (RA), which is associated with poorer prognosis and higher mortality. Many patients with RA may experience bone loss early in the disease course. Therefore, timely assessment of the risk of OP in RA patients is essential. METHODS This is a retrospective study in which we collected information from 500 RA patients who underwent bone mineral density assessments at Longhua Hospital, Shanghai University of Traditional Chinese Medicine, from January 2018 to December 2022. Based on the data collection timeline, the first 70% of patients were assigned to the training set, while the remaining 30% were included in the validation set. The model was established using the training set and evaluated through plotting of the receiver operating characteristic curves, calibration curves, and clinical decision curves. Internal validation was performed by resampling the training set data 1,000 times using the bootstrap method, while internal hold-out validation was conducted using the validation dataset. RESULTS Ultimately, six variables were identified as independently associated with RA combined with OP (RA-OP): female sex, age, beta C-terminal cross-linked peptide (β-CTX), anti-cyclic citrullinated peptide antibody (ACPA), triglycerides (TG), and N-terminal propeptide of type I procollagen (PINP). The regression equation for the model is as follows: Logistic (RA-OP) = -8.703 + 0.946*female + 0.053*age + 0.004*β-CTX + 0.001*ACPA + 0.6*TG-0.008*PINP. The model demonstrated good discrimination (AUC = 0.819, 95% CI: 0.775-0.863) and calibration. In both internal and internal hold-out validation, the model also performed well, with AUC values of 0.814 (95% CI: 0.772-0.864) and 0.772 (95% CI: 0.697-0.847), respectively. Clinical decision curves indicated that the model outperformed both extreme curves, suggesting good clinical utility. CONCLUSIONS Our model is user-friendly and has shown good predictive performance in both internal and internal hold-out validation, offering new insights for the early screening and treatment of OP risk in RA patients.
Collapse
Affiliation(s)
- Yubo Shao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- School of Graduate, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yazhu Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- School of Graduate, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - XiaoYu Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- School of Graduate, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zihang Xu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- School of Graduate, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Key laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
28
|
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] [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
|
29
|
Leon L, Freites-Nuñez D, Toledano E, Candelas G, Martinez C, Rodríguez-Laguna M, Fernández-Gutiérrez B, Abasolo L. Primary non-response in inflammatory arthritis treated with biologics and targeted therapies in daily clinical practice. Ther Adv Musculoskelet Dis 2025; 17:1759720X251325665. [PMID: 40166776 PMCID: PMC11956518 DOI: 10.1177/1759720x251325665] [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: 11/05/2024] [Accepted: 02/19/2025] [Indexed: 04/02/2025] Open
Abstract
Background Switching between therapies is a recommended strategy for rheumatoid arthritis (RA) and psoriatic arthritis (PsA) patients who experience treatment failure; moreover, data on switching due to primary non-response and subsequent failures are limited. Objectives To obtain information from clinical practice regarding failures due to primary non-response in patients on biologic and target synthetic disease-modifying antirheumatic drugs (ts/bDMARDs), assessing the incidence rate (IR) of switching due to primary non-response and risk of subsequent treatment failure by cycling compared to swapping. Design A longitudinal retrospective study, spanning from 2007 to 2022, was conducted on patients with RA or PsA treated with ts/bDMARDs at an outpatient rheumatology clinic. Methods The main outcomes were as follows: (1) ts/bDMARD failure due to primary non-response and (2) subsequent discontinuation of prescribed ts/bDMARD due to lack of efficacy. The independent variable was switching between classes compared to switching within class. As covariates, clinical, sociodemographic, clinical, and treatments were considered. To estimate ts/bDMARDs switching rates, survival techniques were used, expressing the IR per 100 patients * year with their 95% confidence interval. Cox multivariate regression analyses were run to assess the role of switching between/within class in the subsequent treatment failure. Results In total, 327 patients were included. Of these, 50 patients in 77 treatment courses developed primary non-response with an IR of 4.25 (3.4-5.3). The IR was quite similar between RA and PsA, higher in women, and in those who started ts/bDMARDs after 2018. In those with primary non-response, there were 42 subsequent treatment failures with an IR of 26.38 (19.49-35.69). The multivariate model showed that cycling increased the risk of subsequent failure compared to swapping (hazard ratio: 2 (1.1-3.77), p = 0.03). Conclusion This study provides support to consider swapping a better alternative rather than cycling after primary non-response.
Collapse
Affiliation(s)
- Leticia Leon
- Musculoskeletal Pathology Group, Rheumatology Department, IdISSC, Hospital Clínico San Carlos, c\Prof, Martín Lagos s/n, Madrid 28040, Spain
- Faculty of Health Sciences—HM Hospitals, University Camilo José Cela, Madrid, Spain
| | | | - Esther Toledano
- Rheumatology Department and IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| | - Gloria Candelas
- Rheumatology Department and IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| | - Cristina Martinez
- Rheumatology Department and IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Benjamín Fernández-Gutiérrez
- Rheumatology Department and IdISSC, Hospital Clínico San Carlos, Madrid, Spain Medicine Department, Universidad Complutense de Madrid, Madrid, Spain
| | - Lydia Abasolo
- Musculoskeletal Pathology Group, Rheumatology Department, IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
30
|
Fujii T, Murata K, Kohjitani H, Onishi A, Murakami K, Tanaka M, Yamamoto W, Nagai K, Yoshikawa A, Etani Y, Okita Y, Yoshida N, Amuro H, Okano T, Ueda Y, Okano T, Hara R, Hashimoto M, Morinobu A, Matsuda S. Predicting rheumatoid arthritis progression from seronegative undifferentiated arthritis using machine learning: a deep learning model trained on the KURAMA cohort and externally validated with the ANSWER cohort. Arthritis Res Ther 2025; 27:65. [PMID: 40140918 PMCID: PMC11938622 DOI: 10.1186/s13075-025-03541-8] [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: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Undifferentiated arthritis (UA) often develops into rheumatoid arthritis (RA), but predicting disease progression from seronegative UA remains challenging because seronegative RA often does not meet the classification criteria. This study aims to build a machine learning (ML) model to predict the progression from seronegative UA to RA using clinical and laboratory parameters. METHODS KURAMA cohort (training dataset) and ANSWER cohort (validation dataset) were utilized. Patients with seronegative UA were selected based on specific inclusion and exclusion criteria. Clinical and laboratory parameters, including demographic data, acute phase reactants, autoantibodies, and physical examination findings, were collected. Various ML models, including a Feedforward Neural Network (FNN), were developed and compared. Model performance was evaluated using the area under the receiver operating characteristic curve (AUC), sensitivity, and other metrics. SHapley Additive exPlanations (SHAP) values were computed to interpret the importance of variables. RESULTS KURAMA cohort included 210 patients with seronegative UA, of whom 57 (27.1%) progressed to RA. The FNN model demonstrated the highest predictive performance with an AUC of 0.924 and a sensitivity of 80.7% in the training dataset. Validation with ANSWER cohort (140 patients; 32.1% progressed to RA) showed an AUC of 0.777, sensitivity of 77.8%. MMP-3 had the highest impact on the model. CONCLUSIONS The FNN model exhibited robust performance in predicting the progression of RA from seronegative UA and maintained substantial sensitivity in an independent validation cohort. This model using only clinical and laboratory parameters has potential for predicting RA progression in patients with seronegative UA.
Collapse
Affiliation(s)
- Takayuki Fujii
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo, Kyoto, Japan.
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirohiko Kohjitani
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Onishi
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo, Kyoto, Japan
| | - Kosaku Murakami
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo, Kyoto, Japan
| | - Wataru Yamamoto
- Department of Health Information Management, Kurashiki Sweet Hospital, Kurashiki, Japan
| | - Koji Nagai
- Department of Internal Medicine (IV), Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Ayaka Yoshikawa
- Department of Internal Medicine (IV), Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Yuki Etani
- Department of Sports Medical Biomechanics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasutaka Okita
- Department of Respiratory Medicine and Clinical Immunology, Osaka University, Suita, Japan
| | - Naofumi Yoshida
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Hideki Amuro
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Tadashi Okano
- Center for Senile Degenerative Disorders (CSDD), Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yo Ueda
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takaichi Okano
- Department of Clinical Laboratory, Kobe University Hospital, Kobe, Japan
| | - Ryota Hara
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Japan
| | - Motomu Hashimoto
- Department of Clinical Immunology, Osaka Metropolitan University, Osaka, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Ye X, Ren D, Chen Q, Shen J, Wang B, Wu S, Zhang H. Resolution of inflammation during rheumatoid arthritis. Front Cell Dev Biol 2025; 13:1556359. [PMID: 40206402 PMCID: PMC11979130 DOI: 10.3389/fcell.2025.1556359] [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: 01/06/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes synovial joint inflammation as well as bone destruction and erosion, typically characterized by joint pain, swelling, and stiffness, with complications and persistent pain after remission posing a significant health burden for RA patients. The etiology of RA has not yet been fully elucidated, but a large number of studies have shown that the initiation of inflammation in RA is closely related to T-cell activation, the production of a variety of pro-inflammatory cytokines, macrophage M1/M2 imbalance, homeostatic imbalance of the intestinal flora, fibroblast-like synoviocytes (FLSs) and synovial tissue macrophages (STMs) in the synovial lumen of joints that exhibit an aggressive phenotype. While the resolution of RA is less discussed, therefore, we provided a systematic review of the relevant remission mechanisms including blocking T cell activation, regulating macrophage polarization status, modulating the signaling pathway of FLSs, modulating the subpopulation of STMs, and inhibiting the relevant inflammatory factors, as well as the probable causes of persistent arthritis pain after the remission of RA and its pain management methods. Achieving resolution in RA is crucial for improving the quality of life and long-term prognosis of patients. Thus, understanding these mechanisms provide novel potential for further drug development and treatment of RA.
Collapse
Affiliation(s)
- Xiaoou Ye
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Dan Ren
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Qingyuan Chen
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Jiquan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Bo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| |
Collapse
|
32
|
Tao M, Guo X, Ji X, Xu L, Yuan H. Trajectories of health status and their association with rheumatoid arthritis risk: insights from a national prospective cohort study. BMC Public Health 2025; 25:1132. [PMID: 40133953 PMCID: PMC11934476 DOI: 10.1186/s12889-025-22303-4] [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] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The association between trajectories of different health states and rheumatoid arthritis (RA) is unknown. Our cohort study aimed to investigate the impact of various trajectories (including depressive symptoms, physical activity and multimorbidity status) on subsequent RA risk. METHODS A prospective cohort study was conducted using seven waves of national data from the Health and Retirement Study (HRS 2004-2018) involving 9,795 US adults. A growth mixture model identified 6-year trajectories from 2004 to 2010, and participants were screened for RA by self-reported physician diagnosis in the subsequent four waves (2010-2018). Cox proportional hazards model calculated hazard ratios (HR). RESULTS Trajectories of depressive symptoms, physical activity, and multimorbidity status were all associated with the risk of RA. Specifically, keeping a low trajectories (HR = 0.649, 95%CI = 0.533-0.790) or maintaining a moderate rating trajectories (HR = 0.798, 95%CI = 0.644-0.988) for depressive reduced the risk of RA. For physical activity, both high and descending trajectories (HR = 1.456, 95%CI = 1.170-1.812) and high and rising trajectories (HR = 1.244, 95%CI = 1.016-1.522) increased the risk. High multimorbidity trajectories (HR = 1.305, 95%CI = 1.094-1.556) and highest multimorbidity trajectories (HR = 1.393, 95%CI = 1.131-1.715) increased the risk. CONCLUSION The results suggest that tracking trajectories of depressive symptoms, physical activity, and multiple disease states may be a potential and feasible screening method for identifying those at risk for RA.
Collapse
Affiliation(s)
- Mengjun Tao
- Department of Health Management Center, The First Affiliated Hospital of Wannan Medical College, No. 2, Zheshan West Road, Jinghu District, Wuhu, Anhui Province, China
| | - Xin Guo
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu City, Anhui Province, China
| | - Xiancan Ji
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu City, Anhui Province, China
| | - Liang Xu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wannan Medical College, No. 2, Zheshan West Road, Jinghu District, Wuhu, Anhui Province, China
| | - Hui Yuan
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu City, Anhui Province, China.
| |
Collapse
|
33
|
Tarjányi O, Olasz K, Rátky F, Sétáló G, Boldizsár F. Proteasome Inhibitors: Potential in Rheumatoid Arthritis Therapy? Int J Mol Sci 2025; 26:2943. [PMID: 40243560 PMCID: PMC11988683 DOI: 10.3390/ijms26072943] [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: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to the destruction of peripheral joint cartilage and bone tissue. Despite the advent of biological therapies in the past decades, the complete remission of RA patients is still out of reach. Therefore, the search for novel therapeutic approaches is still open in the field of RA. Proteasome inhibitors (PIs) were originally designed to be used in hematological malignancies like multiple myeloma. However, evidence has shown that they are potent inhibitors of the NF-κB pathway, which plays a pivotal role in inflammatory processes and RA. Furthermore, inhibition of cell activation and induction of apoptosis was also reported about PIs. In the present review, we summarize the current knowledge about the potential effects of PIs in RA based on reports from animal and human studies. We believe that there is substantial potential in the use of PIs in RA therapy either alone or in combination with the medications already used.
Collapse
Affiliation(s)
- Oktávia Tarjányi
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Katalin Olasz
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| | - Fanni Rátky
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - György Sétáló
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| |
Collapse
|
34
|
Suda Y, Ikuta K, Hayashi S, Wada K, Anjiki K, Kamenaga T, Tsubosaka M, Kuroda Y, Nakano N, Maeda T, Tsumiyama K, Matsumoto T, Kuroda R, Matsubara T. Comparison of anti-inflammatory and anti-angiogenic effects of JAK inhibitors in IL-6 and TNFα-stimulated fibroblast-like synoviocytes derived from patients with RA. Sci Rep 2025; 15:9736. [PMID: 40118969 PMCID: PMC11928453 DOI: 10.1038/s41598-025-94894-2] [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: 08/07/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Rheumatoid arthritis (RA) involves synovial tissue proliferation, inflammation, and angiogenesis, and contributes to joint destruction. Angiogenesis is a key therapeutic target for the treatment of RA, and Janus kinase (JAK) inhibitors have emerged as a promising therapy. In this study, we compared the inhibitory effects of five JAK inhibitors, including tofacitinib (TOF), baricitinib, peficitinib, upadacitinib, and filgotinib, on interleukin (IL)-6-induced inflammation in RA synovial tissues. All five inhibitors effectively suppressed IL-6-induced inflammatory and angiogenic factors, including vascular endothelial growth factor, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, by inhibiting the phosphorylation of signal transducer and activator of transcription (STAT)1 and STAT3. Overall, the results suggest that while all five JAK inhibitors are effective in reducing IL-6-induced inflammatory and angiogenic factors, their efficacy may differ owing to specific molecular mechanisms and pharmacological properties.
Collapse
Affiliation(s)
- Yoshihito Suda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, 944-25, Japan
| | - Kemmei Ikuta
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan.
| | - Kensuke Wada
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kensuke Anjiki
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yuichi Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Naoki Nakano
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Toshihisa Maeda
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, 944-25, Japan
| | - Ken Tsumiyama
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, 944-25, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki- Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Tsukasa Matsubara
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, 944-25, Japan
| |
Collapse
|
35
|
Calvier L, Wasser CR, Solow EB, Wu S, Evers BM, Karp DS, Kounnas MZ, Herz J. Genetic or therapeutic disruption of the Reelin/Apoer2 signaling pathway improves inflammatory arthritis outcomes. Proc Natl Acad Sci U S A 2025; 122:e2418642122. [PMID: 40073057 PMCID: PMC11929474 DOI: 10.1073/pnas.2418642122] [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: 09/18/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation, pannus formation, and progressive joint destruction. The inflammatory milieu in RA drives endothelial cell activation and upregulation of adhesion molecules, thus facilitating leukocyte infiltration into the synovium. Reelin, a circulating glycoprotein previously implicated in endothelial activation and leukocyte recruitment in diseases such as atherosclerosis and multiple sclerosis, has emerged as a potential upstream regulator of these processes. However, its role in RA pathogenesis remains poorly understood. Here, we demonstrate that Reelin levels are markedly elevated in the plasma of both RA patients and mouse models of arthritis, with higher concentrations correlating with greater disease severity. Genetic deletion of the Reelin receptor Apoer2 conferred significant protection against serum transfer arthritis (STA), underscoring the relevance of this pathway in disease progression. Furthermore, therapeutic inhibition of Reelin using the CR-50 antibody yielded robust anti-inflammatory effects in multiple preclinical arthritis models, including STA, K/BxN, and collagen-induced arthritis. Notably, CR-50 treatment not only reduced leukocyte infiltration and synovial inflammation but also mitigated pannus formation. Importantly, these benefits were achieved without the gastrointestinal side effects commonly associated with nonsteroidal anti-inflammatory drugs like diclofenac. Our findings position Reelin as a proinflammatory endothelial biomarker and therapeutic target in RA. By modulating endothelial activation and leukocyte recruitment, anti-Reelin strategies offer an alternative approach to attenuate synovial inflammation and joint damage. These results provide a compelling rationale for further exploration of Reelin-targeted therapies as alternatives to conventional immunosuppressive treatments in RA and other chronic inflammatory diseases.
Collapse
MESH Headings
- Reelin Protein
- Animals
- Mice
- Humans
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Adhesion Molecules, Neuronal/genetics
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Signal Transduction
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- LDL-Receptor Related Proteins/metabolism
- LDL-Receptor Related Proteins/genetics
- Serine Endopeptidases/metabolism
- Serine Endopeptidases/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/drug therapy
- Mice, Knockout
- Male
- Female
- Disease Models, Animal
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Mice, Inbred C57BL
- Inflammation/metabolism
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Catherine R Wasser
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - E Blair Solow
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Sharon Wu
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - David S Karp
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | | | - Joachim Herz
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| |
Collapse
|
36
|
Basu S, Ulbricht Y, Rossol M. Healthy and premature aging of monocytes and macrophages. Front Immunol 2025; 16:1506165. [PMID: 40165963 PMCID: PMC11955604 DOI: 10.3389/fimmu.2025.1506165] [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: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Aging is associated with immunosenescence, a decline in immune functions, but also with inflammaging, a chronic, low-grade inflammation, contributing to immunosenescence. Monocytes and macrophages belong to the innate immune system and aging has a profound impact on these cells, leading to functional changes and most importantly, to the secretion of pro-inflammatory cytokines and thereby contributing to inflammaging. Rheumatoid arthritis (RA) is an autoimmune disease and age is an important risk factor for developing RA. RA is associated with the early development of age-related co-morbidities like cardiovascular manifestations and osteoporosis. The immune system of RA patients shows signs of premature aging like age-inappropriate increased production of myeloid cells, accelerated telomeric erosion, and the uncontrolled production of pro-inflammatory cytokines. In this review we discuss the influence of aging on monocytes and macrophages during healthy aging and premature aging in rheumatoid arthritis.
Collapse
Affiliation(s)
- Syamantak Basu
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| | - Ying Ulbricht
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| | - Manuela Rossol
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Environment and Natural Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| |
Collapse
|
37
|
Qiu D, Yan B, Xue H, Xu Z, Tan G, Liu Y. Perspectives of exosomal ncRNAs in the treatment of bone metabolic diseases: Focusing on osteoporosis, osteoarthritis, and rheumatoid arthritis. Exp Cell Res 2025; 446:114457. [PMID: 39986599 DOI: 10.1016/j.yexcr.2025.114457] [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: 10/23/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Bone metabolic disorders, constituting a group of prevalent and grave conditions, currently have a scarcity of therapeutic alternatives. Over the recent past, exosomes have been at the forefront of research interest, owing to their nanoparticulate nature and potential for therapeutic intervention. ncRNAs are a class of heterogeneous transcripts that they lack protein-encoding capacity, yet they can modulate the expression of other genes through multiple mechanisms. Mounting evidence underscores the intricate role of exosomes as ncRNAs couriers implicated in the pathogenesis of bone metabolic disorders. In this review, we endeavor to elucidate recent insights into the roles of three ncRNAs - miRNAs, lncRNAs, and circRNAs - in bone metabolic ailments such as osteoporosis, osteoarthritis, and rheumatoid arthritis. Additionally, we examine the viability of exosomal ncRNAs as innovative, cell-free modalities in the diagnosis and therapeutic management of bone metabolic disorders. We aim to uncover the critical function of exosomal ncRNAs within the context of bone metabolic diseases.
Collapse
Affiliation(s)
- Daodi Qiu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Binghan Yan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Haipeng Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhanwang Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guoqing Tan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yajuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China.
| |
Collapse
|
38
|
Tran LS, Chia J, Le Guezennec X, Tham KM, Nguyen AT, Sandrin V, Chen WC, Leng TT, Sechachalam S, Leong KP, Bard FA. ER O-glycosylation in synovial fibroblasts drives cartilage degradation. Nat Commun 2025; 16:2535. [PMID: 40087276 PMCID: PMC11909126 DOI: 10.1038/s41467-025-57401-9] [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: 06/11/2021] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
How arthritic synovial fibroblasts (SFs) activate cartilage ECM degradation remains unclear. GALNT enzymes initiate O-glycosylation in the Golgi; when relocated to the ER, their activity stimulates ECM degradation. Here, we show that in human rheumatoid and osteoarthritic synovial SFs, GALNTs are relocated to the ER. In an RA mouse model, GALNTs relocation occurs shortly before arthritis symptoms and abates as the animal recovers. An ER GALNTs inhibitor prevents cartilage ECM degradation in vitro and expression of this chimeric protein in SFs results in the protection of cartilage. One of the ER targets of GALNTs is the resident protein Calnexin, which is exported to the cell surface of arthritic SFs. Calnexin participates in matrix degradation by reducing ECM disulfide bonds. Anti-Calnexin antibodies block ECM degradation and protect animals from RA. In sum, ER O-glycosylation is a key switch in arthritic SFs and glycosylated surface Calnexin could be a therapeutic target.
Collapse
Affiliation(s)
- Le Son Tran
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Joanne Chia
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Albatroz Therapeutics Pte Ltd, Singapore, Singapore
| | - Xavier Le Guezennec
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Albatroz Therapeutics Pte Ltd, Singapore, Singapore
| | - Keit Min Tham
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Albatroz Therapeutics Pte Ltd, Singapore, Singapore
| | - Anh Tuan Nguyen
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Albatroz Therapeutics Pte Ltd, Singapore, Singapore
| | - Virginie Sandrin
- Roche Pharma Research & Early Development, Innovation Center Basel, Basel, Switzerland
| | | | - Tan Tong Leng
- Department of Orthopaedic Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Sreedharan Sechachalam
- Department of Hand and Reconstructive Microsurgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Khai Pang Leong
- Department of Rheumatology, Allergy & Immunology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Frederic A Bard
- Institute of Molecular and Cell Biology, Singapore, Singapore.
- Albatroz Therapeutics Pte Ltd, Singapore, Singapore.
- Cancer Research Center of Marseille (CRCM), Marseille, France.
| |
Collapse
|
39
|
Snoeck Henkemans SVJ, van den Biggelaar AF, van Mulligen E, Bergstra SA, Luime JJ, Kok MR, Tchetverikov I, van Oosterhout M, van der Kaap JH, van der Helm-van Mil AHM, Vis M, de Jong PHP. Rheumatoid arthritis and psoriatic arthritis: is the disease impact different? A large matching study at diagnosis and after 1 year of treatment. RMD Open 2025; 11:e005143. [PMID: 40081914 DOI: 10.1136/rmdopen-2024-005143] [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: 10/16/2024] [Accepted: 01/29/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are currently treated similarly. However, it is unclear which patient-reported outcome (PRO) domains need specific attention in the management of RA and PsA. Therefore, we aimed to determine the difference in disease impact between matched RA and PsA patients at diagnosis and after 1 year in two different regions. METHODS RA patients from the treatment in the Rotterdam Early Arthritis CoHort trial (tREACH), PsA patients from the Dutch southwest Early PsA cohoRt (DEPAR) and RA and PsA patients from the Leiden Early Arthritis Clinic (EAC) were included. The difference in disease impact between RA and PsA was measured with the following PROs: pain (Visual Analogue Scale (VAS), 0-100), fatigue (VAS), activity limitation (Health Assessment Questionnaire-Disability Index) and health impact (general health (VAS) and 36-item Short-Form Health Survey (SF-36)). Propensity scores were used to match RA and PsA patients, after which inverse probability weights (IPWs) were calculated. IPW-weighted linear regression models were used to measure PRO differences. RESULTS 391 RA patients from tREACH, 416 PsA patients from DEPAR, 702 RA and 99 PsA patients from the EAC were included. At diagnosis, PsA-DEPAR patients scored 5.04 units worse (95% CI 2.21 to 7.87) on SF-36 mental health compared with RA-tREACH patients. This difference still existed after 1 year of treatment (3.88 (95% CI 1.90 to 5.86)). PsA-EAC patients had more activity limitations after 1 year of treatment compared with RA-EAC patients (-0.30 (95% CI -0.50 to -0.10)). No significant differences were present in the other PRO domains. CONCLUSION The disease impact of early RA patients is similar to matched early PsA patients on most PRO domains, except for mental health and functional limitations, which were worse in PsA after 1 year of treatment.
Collapse
Affiliation(s)
| | | | - Elise van Mulligen
- Department of Rheumatology, Erasmus MC, Rotterdam, The Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sytske Anne Bergstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda J Luime
- Department of Rheumatology, Erasmus MC, Rotterdam, The Netherlands
| | - Marc R Kok
- Department of Rheumatology and Clinical Immunology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Ilja Tchetverikov
- Department of Rheumatology, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | | | - Jos H van der Kaap
- Department of Rheumatology, Admiraal De Ruyter Hospital, Goes, The Netherlands
| | - Annette H M van der Helm-van Mil
- Department of Rheumatology, Erasmus MC, Rotterdam, The Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marijn Vis
- Department of Rheumatology, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
40
|
Šimek M, Rubanová D, Nešporová K, Skoroplyas S, Lehká K, Raptová P, Velebný V, Kubala L. Pharmacokinetics of the systemic application of hyaluronic acid for joint arthritis treatment. Int J Biol Macromol 2025; 307:141937. [PMID: 40074120 DOI: 10.1016/j.ijbiomac.2025.141937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Frequent degenerative joint diseases, known as arthritis, are characterized by joint inflammation and cartilage breakdown. Various arthritis types are traditionally managed with intraarticular injections of hyaluronan or its derivatives. However, intravenous administration of hyaluronan is emerging as a vital alternative, particularly because intraarticular injections can be challenging for clinicians when targeting small or swollen joints. Pharmacokinetics of intravenously and intraperitoneally administered middle-Mw hyaluronan were studied in an adjuvant-induced arthritis mouse model alongside therapeutic effects. Using 13C-, biotin- and fluoresce-labeling, we found hyaluronan accumulated in inflamed joint tissues while distribution in other organs remained similar to healthy controls. Repeated administrations significantly reduced arthritis symptoms like swelling and redness, RANKL, inducible nitric oxide synthase, COMP and prostaglandin E2 levels. Moreover, hyaluronan treatment prevented dextran-FITC penetration into inflamed paws suggesting reduced vascular permeability at the site of inflammation. These findings support systemic hyaluronan administration as a promising arthritis treatment strategy.
Collapse
Affiliation(s)
- Matěj Šimek
- Contipro a.s., Dolní Dobrouč, Czech Republic.
| | - Daniela Rubanová
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Svitlana Skoroplyas
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | | | - Petra Raptová
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | | | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
41
|
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
|
42
|
Kim MJ, Yoo HM, Lee YJ, Jang HH, Shim SC, Won EJ, Kim TJ. Clonorchis sinensis excretory/secretory proteins ameliorate inflammation in rheumatoid arthritis and ankylosing spondylitis. Parasit Vectors 2025; 18:85. [PMID: 40038824 DOI: 10.1186/s13071-025-06677-3] [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: 09/26/2024] [Accepted: 01/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND We aimed to investigate whether substances secreted by Clonorchis sinensis excretory/secretory protein (CS-ESP) have an effect on the inflammation of rheumatoid arthritis (RA) and ankylosing spondylitis (AS) and to identify specific peptides through related proteomic analysis to determine which proteins exhibit anti-inflammatory effects more specifically. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from healthy controls (HCs), RA and AS patients. Cytotoxicity of CS-ESP at different doses was assessed by MTS and flow cytometry before performing experiments. Inflammatory cytokine producing cells were analyzed using flow cytometry. To determine the effect of CS-ESP in an arthritis mouse model, 8-week-old SKG mice were injected intraperitoneally with curdlan and treated with CS-ESP; body weight and paw swelling were checked twice a week. Inflammation was evaluated using immunohistochemistry. We conducted proteomic analysis on CS-ESP and identified specific Cs-GT and Cs-Severin proteins. In vitro effect of coculture with Cs-GT and Cs-Severin was determined by inflammatory cytokine measurements. RESULT Treatment with CS-ESP resulted in no reduced cell viability of PBMCs. In experiments culturing PBMCs, the frequencies of IL-17A and GM-CSF producing cells were significantly reduced after CS-ESP treatment. In the SKG mouse model, CS-ESP treatment significantly suppressed clinical score, arthritis and enthesitis. Treatment with Cs-GT and Cs-Severin resulted in no reduced cell viability of HC PBMCs. After Cs-GT and Cs-Severin treatment of HC PBMC, the frequencies of IL-17A and GM-CSF producing cells were significantly reduced. CONCLUSIONS We provide evidence showing that CS-ESP, Cs-GT and Cs-Severin can ameliorate clinical signs and cytokine derangements in AS.
Collapse
Affiliation(s)
- Moon-Ju Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
- Department of Precision Measurement, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Yu Jeong Lee
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
- Department of Biomedical Sciences, Graduate School of Chonnam National University, Gwangju, 61469, Republic of Korea
| | - Hyun Hee Jang
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Eun Jeong Won
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
43
|
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 DOI: 10.1038/s41467-025-57361-0] [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: 07/13/2023] [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
|
44
|
Chen M, Wang Z, Chen H, Li J, Guo X, Zhou S. Biomimetic Nanoparticles Inhibit the HIF-1α/iNOS/NLRP3 Pathway to Alleviate Rheumatoid Arthritis. NANO LETTERS 2025. [PMID: 40033154 DOI: 10.1021/acs.nanolett.4c05782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease distinguished by inflammatory synovitis. Chrysin can alleviate the inflammatory response and inhibit the progression of RA. However, unfavorable physicochemical properties and nonselective biodistribution of chrysin make it difficult to achieve good therapeutic efficacy. To address these challenges, we developed a biomimetic nanocarrier to enhance the targeted delivery of chrysin to synoviocytes, a key cellular component in RA pathology. Our nanodrug, FMPlipo@C, was engineered by integrating fibroblast-like synoviocyte (FLS) membrane proteins into chrysin-loaded liposomes. This innovative approach harnesses homologous targeting mediated by FLS membrane proteins to direct liposomes to inflamed joints, facilitating cargo release within synoviocytes. We showed that FMPlipo@C reduces inflammation in collagen-induced rheumatoid arthritis (CIA) model mice by inhibiting the HIF-1α/iNOS/NLRP3 pathway, protecting cartilage, and preventing bone erosion, thus reducing swelling and stiffness. This study offers valuable insights into the development of novel therapeutic strategies for the treatment of RA.
Collapse
Affiliation(s)
- Mo Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Zhenhua Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Haolong Chen
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jin Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Xing Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
45
|
Nash P, Kerschbaumer A, Konzett V, Aletaha D, Dörner T, Fleischmann R, McInnes I, Primdahl J, Sattar N, Tanaka Y, Trauner M, Winthrop K, de Wit M, Askling J, Baraliakos X, Boehncke WH, Emery P, Gossec L, Isaacs JD, Krauth M, Lee EB, Maksymowych W, Pope J, Scholte-Voshaar M, Schreiber K, Schreiber S, Stamm T, Taylor PC, Takeuchi T, Tam LS, Van den Bosch F, Westhovens R, Zeitlinger M, Smolen JS. Expert consensus statement on the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: 2024 update. Ann Rheum Dis 2025:S0003-4967(25)00181-5. [PMID: 40037995 DOI: 10.1016/j.ard.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 03/06/2025]
Abstract
In light of the introduction of new Janus kinase inhibitors (JAKi), new indications for JAKi and recent safety considerations that have arisen since the preceding consensus statement on JAKi therapy, a multidisciplinary taskforce was assembled, encompassing patients, health care professionals, and clinicians with expertise in JAKi therapy across specialties. This taskforce, informed by two comprehensive systematic literature reviews, undertook the objective to update the previous expert consensus for using JAKi developed in 2019. The taskforce deliberated on overarching principles, indications, dosage and comedication strategies, warnings and contraindications, screening protocols, monitoring recommendations, and adverse effect profiles. The methodology was based on the European Alliance of Associations for Rheumatology standard operating procedures, with voting on these important elements. Furthermore, an updated research agenda was proposed. The task force did not address when a JAKi should be prescribed but rather considerations once this decision has been made. This update aimed to equip clinicians with the necessary knowledge and guidance for the efficient and safe administration of this expanding and significant class of drugs.
Collapse
Affiliation(s)
- Peter Nash
- Griffith University School of Medicine, Nathan, Brisbane, QLD, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Victoria Konzett
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Thomas Dörner
- Department of Medicine/Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Roy Fleischmann
- Metroplex Clinical Research Center and University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jette Primdahl
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Kevin Winthrop
- Division of Infectious Diseases and School of Public Health, Oregon Health and Science University, Portland, OR, USA
| | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Johan Askling
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Wolf-Henning Boehncke
- Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Paul Emery
- Leeds NIHR Biomedical Research Centre, LTHT, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Laure Gossec
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris France, AP-HP, Pitié-Salpêtrière Hospital, Rheumatology Department, Paris, France
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UKNIHR Newcastle Biomedical Research Centre and Musculoskeletal Unit, Newcastle upon Tyne Hospitals, Newcastle upon Tyne, UK
| | - Maria Krauth
- Division of Haematology and Haemostaseology, Department of Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Eun Bong Lee
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Walter Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, AB, Canada
| | - Janet Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, ON, Canada
| | - Marieke Scholte-Voshaar
- Department of Pharmacy, Sint Maartenskliniek, Netherlands; Department of Pharmacy, Radboudumc, Nijmegen, Netherlands
| | - Karen Schreiber
- Danish Centre for Expertise in Rheumatology (CeViG), Danish Hospital for Rheumatic Diseases, Sønderborg, Denmark; Department of Regional Health Research (IRS), University of Southern Denmark, Odense, Denmark; Thrombosis and Haemostasis, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Stefan Schreiber
- Department Internal Medicine I, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Tanja Stamm
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, and Saitama Medical University, Saitama, Japan
| | - Lai-Shan Tam
- Rheumatology, Department of Medicine and Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Filip Van den Bosch
- VIB-UGent Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Ghent University Hospital, Department of Rheumatology, Ghent, Belgium
| | - Rene Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
46
|
Sami MM, Jamai MJ, Alkareem TAA, Ayram NB. Low progesterone levels and their role in the co-existence of polycystic ovary syndrome and rheumatoid arthritis: A comprehensive analysis among Iraqi patient. J Steroid Biochem Mol Biol 2025; 247:106680. [PMID: 39870327 DOI: 10.1016/j.jsbmb.2025.106680] [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: 10/06/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a low-grade and chronic inflammation defined by irregular hormonal status that primarily triggers females in their reproductive age. Multi cysts are a primary manifestation of PCOS; a high level of androgen production characterizes the condition via ovaries. Rheumatoid arthritis (RA) is a chronic, systemic, and symmetrical inflammatory autoimmune disease that affects 1-2 % of adults. Females are more likely to generate RA. During the inflammatory activity, immune cells attack the synovium and the synovial space. This invasion is essential in releasing many cytokines in the synovial and joint spaces, leading to joint damage and pain, stiffens, heat, and tenderness in the joint. To evaluate the strength of the link between PCOS and RA, the cross-sectional study examined hormonal, metabolic, and autoantibodies in PCOS, RA as a positive control and the study groups. Statistical analysis Shapiro-Wilk test, student t-test, one-way ANOVA, and multi-linear regression analysis were used to evaluate the results. The data highlights significant values for the BMI, WHR, and hirsutism of PCOS and RA groups in comparison to the negative control. The ANOVA results of these parameters also showed a significant p < 0.05 among the groups. According to the negative control, the levels of insulin, HOMA-IR, testosterone, LH, estradiol, and CRP showed a substantial increase in the PCOS group. Also, the RA group showed a significant p < 0.05 rise in CRP, RF, and Ani-CCP, and the ANOVA results showed significant value among the groups under investigation. Progesterone D as a model showed a correlation with Anti-CCP B, RF C, Anti-CCP C, CRP D, RF D, and Anti-CCP D with the highest level of f2 between other models. In addition, statistical tests show that progesterone D with R2= 0.565 and RMSE equal to 0.996 have heteroscedasticity, which means that low levels of progesterone are associated inversely with high levels of RF and Anit-CCP. There is a relative association between the progesterone D model and corresponding predictions. Regardless of solid f2, only 56 % of the sample shows an association between the model and predictors; this relation may differ if we consider the study's limitations.
Collapse
Affiliation(s)
- Mohammed Mahdi Sami
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Mataz J Jamai
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Tamara Ahmed Abd Alkareem
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Nabeel Bunyan Ayram
- Department of chemistry-College of Science, Mustansiriyah University, Baghdad, Iraq.
| |
Collapse
|
47
|
Uke P, Maharaj A, Adebajo A. A review on the epidemiology of rheumatoid arthritis: An update and trends from current literature. Best Pract Res Clin Rheumatol 2025; 39:102036. [PMID: 39939219 DOI: 10.1016/j.berh.2025.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic, chronic autoimmune disease affecting mainly the joints, often with extra articular manifestations. This review provides an update on RA epidemiological trends and management. PubMed and EMBASE were searched from 2014 to 2024 using rheumatoid arthritis as keyword, combined with incidence, prevalence, diagnosis, classification, and management. Emphasis was on papers published in the past 5 years. Globally, the age-standardised prevalence and incidence rate (ASPR and ASIR) of RA increased with varying figures. The ASPR increased by 0.37%, 14.1%, and 6.4% from 1990 to 2019, 2020 and 2017 respectively; and 9% from 1980 to 2019. The ASIR increased by 0.3% and 8.2% from 1990 to 2019 and 2017 respectively; the disability-adjusted life years (DALY) figures increased 0.12% and decreased 0.36% in the same period from different authors. Reduction in ASIR were reported while ASPR varies. Disease modifying anti-rheumatic drugs (DMARDs) remain the cornerstone of treatment.
Collapse
Affiliation(s)
- Perpetual Uke
- Department of Rheumatology, Birmingham City Hospital, United Kingdom; Department of Inflammation and Ageing, College of Medicine and Health, University of Birmingham, United Kingdom
| | - Ajesh Maharaj
- Department of Internal Medicine and Pharmacology, Faculty of Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adewale Adebajo
- School of Medicine and Population Health, University of Sheffield, United Kingdom.
| |
Collapse
|
48
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 PMCID: PMC11875835 DOI: 10.1016/j.ultsonch.2025.107270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
49
|
Wu S, Xie Y, Jiang Y, Zhang X, Zhou Y, Zuo X, Li T. GTS-21 modulates rheumatoid arthritis Th17 and Th2 lymphocyte subset differentiation through the ɑ7nAch receptor. Clin Rheumatol 2025; 44:989-998. [PMID: 39812970 DOI: 10.1007/s10067-025-07320-3] [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: 05/17/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Previous research has demonstrated ɑ7nAch receptor (ɑ7nAchR) agonists to provide benefit for rheumatoid arthritis (RA) patients. However, the immunological mechanism of action for these ɑ7nAchR agonists has not been elucidated. Herein, the effect of GTS-21, a selective ɑ7nAchR agonist, on the differentiation of Th17 and Th2 cells was assessed. CD4 + T cells were obtained from the peripheral blood mononuclear cells (PBMCs) of RA patients and healthy donors. CD4 + T cells were separately differentiated into Th2 or Th17 cells with or without GTS-21 and with or without alpha-bungarotoxin (ɑBgt) (a ɑ7nAchR antagonist). The proportions of Th17 and Th2 cells were assessed by flow cytometry. Levels of the T cell cytokines, IL-17A and IL-4, were assessed by ELISA. Specific transcription factors, retinoic orphan receptor c (RORc), and GATA Binding Protein 3 (GATA-3) were detected by western blot. GTS-21 reduced IL-17A and increased IL-4 production by RA PBMCs. GTS-21 reduced the percentage of Th17 cells and increased the percentage of Th2 cells during Th17 and Th2 differentiation, respectively. GTS-21 downregulated RA CD4 + T cells RORc levels and reduced the secretion of IL-17A during Th17 differentiation. GTS-21 upregulated RA CD4 + T cells GATA3 and promoted IL-4 production during Th2 differentiation. ɑ-Bgt blocked the effects of GTS-21 during Th17 and Th2 differentiation. These results demonstrated that GTS-21 suppressed RA Th17 differentiation and promoted Th2 differentiation. As such, the use of GTS-21 may be a new therapeutic approach by which to treat RA patients. Key Points • GTS-21 suppressed RA Th17 differentiation and promoted Th2 differentiation via acting on ɑ7nAchR. • The protective effect of GTS-21 on RA may be related to its regulation of Th cell subsets.
Collapse
Affiliation(s)
- Shiyao Wu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanli Xie
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Jiang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoli Zhang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaou Zhou
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zuo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tong Li
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
50
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|