1
|
Ishina IA, Zhiyanov AP, Kurbatskaia IN, Mamedov AE, Nersisyan SA, Ziganshin RH, Eliseev IE, Petrusenko YS, Nikonova AV, Zhbanova ES, Salnikova MA, Ovchinnikova LA, Mamedov IZ, Davydov AN, Nurbaeva KS, Lisitsyna TA, Reshetnyak TM, Lila AM, Nasonov EL, Lomakin YA, Belogurov AA, Zhang H, Tonevitskiy AG, Rubtsov YP, Gabibov AG, Zakharova MY. Autoantigenic peptide landscape of rheumatoid arthritis-associated HLA class II. Genes Dis 2025; 12:101469. [PMID: 40236665 PMCID: PMC11999199 DOI: 10.1016/j.gendis.2024.101469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/02/2024] [Indexed: 04/17/2025] Open
Affiliation(s)
- Irina A. Ishina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Anton P. Zhiyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Faculty of Biology and Biotechnology, Higher School of Economics, University, Moscow 117418, Russia
| | - Inna N. Kurbatskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Azad E. Mamedov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Stepan A. Nersisyan
- Faculty of Biology and Biotechnology, Higher School of Economics, University, Moscow 117418, Russia
| | - Rustam H. Ziganshin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Igor E. Eliseev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Yunna S. Petrusenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Biotech Campus Ltd, Moscow 117437, Russia
| | - Anastasia V. Nikonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Elizaveta S. Zhbanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Maria A. Salnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117513, Russia
| | - Leyla A. Ovchinnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Ilgar Z. Mamedov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexey N. Davydov
- Central European Institute of Technology, Brno 60177, Czech Republic
- MiLaboratories Inc, San Francisco, CA 94114, United States
| | | | | | | | - Alexander M. Lila
- V.A. Nasonova Research Institute of Rheumatology, Moscow 115522, Russia
| | | | - Yakov A. Lomakin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexey A. Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China
| | - Alexander G. Tonevitskiy
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Faculty of Biology and Biotechnology, Higher School of Economics, University, Moscow 117418, Russia
| | - Yury P. Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexander G. Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Faculty of Biology and Biotechnology, Higher School of Economics, University, Moscow 117418, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Maria Y. Zakharova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
2
|
Hansen B, Sánchez-Castro M, Schintgen L, Khakdan A, Schneider JG, Wilmes P. The impact of fasting and caloric restriction on rheumatoid arthritis in humans: A narrative review. Clin Nutr 2025; 49:178-186. [PMID: 40328175 DOI: 10.1016/j.clnu.2025.04.025] [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: 01/27/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease affecting approximately 1 % of the global population. It is characterized by swollen and painful joints eventually evolving into bone erosion, cartilage degradation and systemic inflammation, that significantly reduce patients' quality of life. While modern pharmacological treatments often lead to symptom improvement, they are also accompanied by substantial side effects, which can further impair patient wellbeing. Dietary interventions, particularly fasting and caloric restriction (CR), have gained increasing attention as adjunctive strategies for RA prevention and treatment. Their anti-inflammatory potential and ability to modulate the gut microbiome render them an attractive option to accompany or modify medical treatment. However, high-quality research on fasting and CR interventions in humans with RA remains limited, and the underlying mechanisms are not yet fully understood. The present narrative review reflects our current knowledge regarding fasting and CR, emphasising their impact on clinical outcomes, potential underlying mechanism and the sustainability of their effects. Evidence suggests that fasting and CR may lead to short-term improvements in RA disease activity, including reductions in inflammatory markers such as C-reactive protein (CRP) and interleukin-6 (IL-6). However, their long-term efficacy remains uncertain due to the limited duration of most studies. Future research should focus on identifying optimal fasting and CR protocols and their feasibility in long-term disease management, along with investigating patient adherence and potential risks associated with fasting interventions.
Collapse
Affiliation(s)
- Bérénice Hansen
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marta Sánchez-Castro
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lynn Schintgen
- Department of Microbiome Research and Applied Bioinformatics, Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Arefeh Khakdan
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jochen G Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Internal Medicine II, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany.
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
3
|
Gao QW, Liu WY, Jawad M, Ci L, Cao YY, Xi J, Wu JY, Lei YY, Hu YS, You XY, Zhang XY, Fei J, Luan Y. Aristolochic acid IVa ameliorates arthritis in SKG Mice by regulating macrophage polarization and Th17/Treg balance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156557. [PMID: 40043543 DOI: 10.1016/j.phymed.2025.156557] [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: 10/31/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Aristolochic acids (AAs)-containing herbs have been used as medicinal remedies for thousands of years. However, exposure to AAI and AAII increases the risk of nephropathy and cancers. Our previous study identified AAIVa, an analogue without carcinogenicity or nephrotoxicity, exerted anti-inflammatory effects. PURPOSE To explore AAIVa's anti-inflammatory mechanisms and assess its therapeutic potential in arthritis. METHODS AND RESULTS In this study, we employed in vitro assays on RAW 264.7 cells and explored the underlying mechanisms of AAIVa's anti-inflammatory effect through transcriptome analysis, identifying macrophage polarization-associated genes, IL-17 signaling, and Rheumatoid Arthritis (RA) pathway. Also, we used BALB/cAnSmoc-Zap70em(W163C)Smoc (SKG) mice, a model that spontaneously develops chronic arthritis closely resembling human RA, and revealed AAIVa's therapeutic potential in arthritis. AAIVa-treatment (10 mg/kg, i.g.) for 4 weeks protected SKG mice from mannan-accelerated arthritis symptoms, reducing inflammation and improving bone microstructure. We further isolated bone marrow-derived macrophages (BMDMs) and spleen primary cells from SKG and BALB/c mice to evaluate the impact of AAIVa on macrophage polarization and T cell differentiation. We found that AAIVa induced M2 macrophage polarization in BMDMs, and mitigated lipopolysaccharide-stimulated inflammation by increasing Tregs and decreasing Th17 cells. Subsequently, the elevation of M2 macrophages, increased Tregs expression and decreased Th17 cells in the ankle joints of SKG mice supported our in vitro observation. CONCLUSION We provide first evidence that AAIVa exerts anti-arthritis effects, likely through modulation of macrophage polarization and restoration of the Th17/Treg balance. Our findings highlight AAIVa's mechanism of action and AAIVa's potential as a therapeutic candidate for autoimmune arthritis.
Collapse
Affiliation(s)
- Qin-Wen Gao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei-Ying Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Mirza Jawad
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai 201203, China
| | - Yi-Yi Cao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Xi
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia-Ying Wu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Yang Lei
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Shi Hu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin-Yue You
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin-Yu Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Fei
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai 201203, China; School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Yang Luan
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Scheffler JM, Drevinge C, Lindholm C, Gjertsson I, Lend K, Lund Hetland M, Østergaard M, Uhlig T, Schrumpf Heiberg M, Haavardsholm EA, Nurmohamed MT, Lampa J, Sokka‐Isler T, Nordström D, Hørslev‐Petersen K, Gudbjornsson B, Gröndal G, van Vollenhoven R, Carlsten H, Lorentzon M, Hultgård Ekwall A, Rudin A, Islander U. Circulating Baseline CXCR3 +Th2 and Th17 Cell Proportions Correlate With Trabecular Bone Loss After 48 Weeks of Biological Treatment in Early Rheumatoid Arthritis. ACR Open Rheumatol 2025; 7:e11742. [PMID: 39411912 PMCID: PMC11667770 DOI: 10.1002/acr2.11742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 12/25/2024] Open
Abstract
OBJECTIVE The high prevalence of osteoporosis in rheumatoid arthritis (RA) is due to inflammation that stimulates differentiation of osteoclasts, a process involving circulating monocytes and T cell-derived factors. The aim of this study was to evaluate relations between circulating monocytes, T cell subsets, and changes in bone characteristics before and after treatment with biological disease-modifying antirheumatic drugs (bDMARDs) in RA. METHODS Thirty patients with untreated early RA who met the American College of Rheumatology/EULAR 2010 criteria were included. Data were collected before and 48 weeks after treatment with methotrexate (MTX) together with one of three bDMARDs (abatacept, tocilizumab, or certolizumab pegol). Disease activity was measured using the Clinical Disease Activity Index, swollen or tender joint counts, C-reactive protein levels, and erythrocyte sedimentation rates. Proportions of monocyte and CD4+ T cell subsets in blood samples were analyzed by flow cytometry. Bone densitometry was performed using high-resolution peripheral quantitative computed tomography (HR-pQCT). RESULTS HR-pQCT revealed an overall decrease in cortical (P = 0.009) and trabecular (P = 0.034) bone mineral density, although a subset of patients showed no bone loss after 48 weeks of treatment. The overall bone loss was not associated with age, body mass index, sex, intraarticular glucocorticoid injections, or baseline disease activity. Loss of trabecular bone volume fraction correlated with high proportions of circulating CXCR3+Th2 cells (r = -0.38, P = 0.04) and CXCR3+Th17 cells (r = -0.36, P = 0.05) at baseline. Similarly, no loss of trabecular bone volume fraction correlated with high proportions of regulatory T cells (r = 0.4, P = 0.03) at baseline. However, the associations were not significant when corrected for confounders and multiple testing. CONCLUSION MTX together with bDMARDs efficiently reduce disease activity but only prevent bone loss in a subset of patients with RA after 48 weeks of treatment. The correlations of circulating baseline T helper cell and regulatory T cell populations with trabecular bone changes suggest a potential novel role for these cells in systemic bone homeostasis during early RA.
Collapse
Affiliation(s)
| | | | - Catharina Lindholm
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Inger Gjertsson
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Kristina Lend
- Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Merete Lund Hetland
- Rigshospitalet, Glostrup, Denmark, and University of CopenhagenCopenhagenDenmark
| | - Mikkel Østergaard
- Rigshospitalet, Glostrup, Denmark, and University of CopenhagenCopenhagenDenmark
| | | | | | | | - Michael T. Nurmohamed
- Amsterdam Rheumatology and Immunology Center, Reade, the Netherlands, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jon Lampa
- Karolinska Institute, Karolinska University HospitalStockholmSweden
| | | | - Dan Nordström
- Helsinki University and University HospitalHelsinkiFinland
| | - Kim Hørslev‐Petersen
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark, and University of Southern DenmarkOdenseDenmark
| | - Bjorn Gudbjornsson
- Landspitali National University Hospital of Iceland and University of IcelandReykjavikIceland
| | - Gerdur Gröndal
- Landspitali National University Hospital of Iceland and University of IcelandReykjavikIceland
| | - Ronald van Vollenhoven
- Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Hans Carlsten
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Mattias Lorentzon
- University of Gothenburg, Gothenburg, Sweden and Australian Catholic UniversityMelbourneAustralia
| | | | - Anna Rudin
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | | |
Collapse
|
5
|
Balasundaram D, Veerasamy V, Sylvia Singarayar M, Neethirajan V, Ananth Devanesan A, Thilagar S. Therapeutic potential of probiotics in gut microbial homeostasis and Rheumatoid arthritis. Int Immunopharmacol 2024; 137:112501. [PMID: 38885604 DOI: 10.1016/j.intimp.2024.112501] [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: 02/29/2024] [Revised: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation and joint damage. Existing treatment options primarily focus on managing symptoms and slowing disease progression, often with side effects and limitations. The gut microbiome, a vast community of microorganisms present in the gastrointestinal tract, plays a crucial role in health and disease. Recent research suggests a bidirectional relationship between the gut microbiome and RA, highlighting its potential as a therapeutic option. This review focuses on the interaction between the gut microbiome and RA development, by discussing how dysbiosis, an imbalance in gut bacteria, can contribute to RA through multiple mechanisms such as molecular mimicry, leaky gut, and metabolic dysregulation. Probiotics, live microorganisms with health benefits, are emerging as promising tools for managing RA. They can prevent the negative effects of dysbiosis by displacing harmful bacteria, producing anti-inflammatory metabolites like short-chain fatty acids (SCFA), Directly influencing immune cells, and modifying host metabolism. animal and clinical studies demonstrate the potential of probiotics in improving RA symptoms and disease outcomes. However, further research is needed to optimize probiotic strains, dosages, and treatment protocols for personalized and effective management of RA. This review summarizes the current understanding of the gut microbiome and its role in RA and discusses future research directions. In addition to the established role of gut dysbiosis in RA, emerging strategies like fecal microbiota transplantation, prebiotics, and postbiotics offer exciting possibilities. However, individual variations in gut composition necessitate personalized treatment plans. Long-term effects and clear regulations need to be established. Future research focusing on metagenomic analysis, combination therapies, and mechanistic understanding will unlock the full potential of gut microbiome modulation for effective RA management.
Collapse
Affiliation(s)
| | - Veeramurugan Veerasamy
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Magdalin Sylvia Singarayar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Vivek Neethirajan
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | | | - Sivasudha Thilagar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India.
| |
Collapse
|
6
|
Ding Q, Xu Q, Hong Y, Zhou H, He X, Niu C, Tian Z, Li H, Zeng P, Liu J. Integrated analysis of single-cell RNA-seq, bulk RNA-seq, Mendelian randomization, and eQTL reveals T cell-related nomogram model and subtype classification in rheumatoid arthritis. Front Immunol 2024; 15:1399856. [PMID: 38962008 PMCID: PMC11219584 DOI: 10.3389/fimmu.2024.1399856] [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: 03/12/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Objective Rheumatoid arthritis (RA) is a systemic disease that attacks the joints and causes a heavy economic burden on humans worldwide. T cells regulate RA progression and are considered crucial targets for therapy. Therefore, we aimed to integrate multiple datasets to explore the mechanisms of RA. Moreover, we established a T cell-related diagnostic model to provide a new method for RA immunotherapy. Methods scRNA-seq and bulk-seq datasets for RA were obtained from the Gene Expression Omnibus (GEO) database. Various methods were used to analyze and characterize the T cell heterogeneity of RA. Using Mendelian randomization (MR) and expression quantitative trait loci (eQTL), we screened for potential pathogenic T cell marker genes in RA. Subsequently, we selected an optimal machine learning approach by comparing the nine types of machine learning in predicting RA to identify T cell-related diagnostic features to construct a nomogram model. Patients with RA were divided into different T cell-related clusters using the consensus clustering method. Finally, we performed immune cell infiltration and clinical correlation analyses of T cell-related diagnostic features. Results By analyzing the scRNA-seq dataset, we obtained 10,211 cells that were annotated into 7 different subtypes based on specific marker genes. By integrating the eQTL from blood and RA GWAS, combined with XGB machine learning, we identified a total of 8 T cell-related diagnostic features (MIER1, PPP1CB, ICOS, GADD45A, CD3D, SLFN5, PIP4K2A, and IL6ST). Consensus clustering analysis showed that RA could be classified into two different T-cell patterns (Cluster 1 and Cluster 2), with Cluster 2 having a higher T-cell score than Cluster 1. The two clusters involved different pathways and had different immune cell infiltration states. There was no difference in age or sex between the two different T cell patterns. In addition, ICOS and IL6ST were negatively correlated with age in RA patients. Conclusion Our findings elucidate the heterogeneity of T cells in RA and the communication role of these cells in an RA immune microenvironment. The construction of T cell-related diagnostic models provides a resource for guiding RA immunotherapeutic strategies.
Collapse
Affiliation(s)
- Qiang Ding
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Qingyuan Xu
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Yini Hong
- Gynecology Department, The First People’s Hospital of Guangzhou, Guangzhou, China
| | - Honghai Zhou
- Faculty of Orthopedics and Traumatology, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyu He
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Chicheng Niu
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Zhao Tian
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Hao Li
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Ping Zeng
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| | - Jinfu Liu
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| |
Collapse
|
7
|
Vergouwen DPC, van Beek AA, de Hoog J, de Boer JH, Los LI, Gijs M, Erckens RJ, Verdijk RM, Haasnoot GW, Roelen DL, Rothova A, Rönnelid J, Ten Berge JC, Schreurs MWJ. The enigma of sclera-specific autoimmunity in scleritis. J Autoimmun 2024; 144:103178. [PMID: 38368769 DOI: 10.1016/j.jaut.2024.103178] [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: 07/05/2023] [Revised: 12/29/2023] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Scleritis is a severe and painful ophthalmic disorder, in which a pathogenic role for collagen-directed autoimmunity was repeatedly suggested. We evaluated the presence of sclera-specific antibodies in a large cohort of patients with non-infectious scleritis. Therefore, we prospectively collected serum samples from 121 patients with non-infectious scleritis in a multicenter cohort study in the Netherlands. In addition, healthy (n = 39) and uveitis controls (n = 48) were included. Serum samples were tested for anti-native human type II collagen antibodies using a validated enzyme-linked immunosorbent assay (ELISA). Further, sclera-specific antibodies were determined using indirect immunofluorescence (IIF) on primate retinal/scleral cryosections. Lastly, human leukocyte antigen (HLA) typing was performed in 111 patients with scleritis. Anti-type II collagen antibodies were found in 13% of scleritis patients, in 10% of healthy controls and in 11% of uveitis controls (p = 0.91). A specific reaction to scleral nerve tissue on IIF was observed in 33% of patients with scleritis, which was higher than in healthy controls (11%; p = 0.01), but similar to uveitis controls (25%; p = 0.36). Reactivity to the scleral nerve tissue was significantly associated with earlier onset of scleritis (48 versus 56 years; p < 0.001), bilateral involvement (65% versus 42%; p = 0.01), and less frequent development of scleral necrosis (5% versus 22%; p = 0.02). HLA-B27 was found to be twice as prevalent in patients with scleritis (15.3%) compared to a healthy population (7.2%). In conclusion, scleral nerve autoantibody reactivity was more common in scleritis and uveitis patients in contrast to healthy controls. Further research is needed to characterize these scleral-nerve directed antibodies and assess their clinical value.
Collapse
Affiliation(s)
- Daphne P C Vergouwen
- Department of Ophthalmology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Adriaan A van Beek
- Department of Immunology, HLA laboratory, Leiden University Medical Center, Leiden, the Netherlands
| | - Joeri de Hoog
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Joke H de Boer
- Department of Ophthalmology, UMC Utrecht, Utrecht, the Netherlands
| | - Leonoor I Los
- Department of Ophthalmology, UMC Groningen, Groningen, the Netherlands
| | - Marlies Gijs
- University Eye Clinic, Maastricht UMC+, Maastricht, the Netherlands
| | - Roel J Erckens
- University Eye Clinic, Maastricht UMC+, Maastricht, the Netherlands
| | - Rob M Verdijk
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Geert W Haasnoot
- Eurotransplant Reference Laboratory, Leiden University Medical Center, Leiden, the Netherlands
| | - Dave L Roelen
- Department of Immunology, HLA laboratory, Leiden University Medical Center, Leiden, the Netherlands
| | - Aniki Rothova
- Department of Ophthalmology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Johan Rönnelid
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Josianne C Ten Berge
- Department of Ophthalmology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marco W J Schreurs
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Arleevskaya MI, Larionova RV, Shagimardanova EI, Gogoleva NE, Kravtsova OA, Novikov AA, Kazarian GG, Carlé C, Renaudineau Y. Predictive risk factors before the onset of familial rheumatoid arthritis: the Tatarstan cohort study. Front Med (Lausanne) 2023; 10:1227786. [PMID: 37877020 PMCID: PMC10593450 DOI: 10.3389/fmed.2023.1227786] [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: 05/23/2023] [Accepted: 08/14/2023] [Indexed: 10/26/2023] Open
Abstract
Background A familial history of rheumatoid arthritis (RA) predisposes an individual to develop RA. This study aimed at investigating factors associated with this conversion from the Tatarstan cohort. Methods A total of 144 individuals, referred to as pre-RA and at risk for familial RA, were selected 2 years (range: 2-21 years) before conversion to RA and compared to non-converted 328 first-degree relatives (FDR) from RA as assessed after ≥2 years follow-up, and 355 healthy controls were also selected (HC). Preclinical parameters and socio-demographic/individual/HLA genetic factors were analyzed when data were available at the time of enrollment. Results As compared to FDR and HC groups, pre-RA individuals were characterized before conversion to RA by the presence of arthralgia, severe morning symptoms, a lower educational level, and rural location. An association with the HLA-DRB1 SE risk factor was also retrieved with symmetrical arthralgia and passive smoking. On the contrary, alcohol consumption and childlessness in women were protective and associated with the HLA-DRB1*07:01 locus. Conclusion Before RA onset, a combination of individual and genetic factors characterized those who are at risk of progressing to RA among those with familial RA relatives.
Collapse
Affiliation(s)
- Marina I. Arleevskaya
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Regina V. Larionova
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Elena I. Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Natalia E. Gogoleva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Olga A. Kravtsova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Andrej A. Novikov
- Innovation Department, Sobolev Institute of Mathematics, Siberian Brunch of Russian Academy of Science, Novosibirsk, Russia
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital Center, Toulouse, France
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | | | - Caroline Carlé
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital Center, Toulouse, France
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Yves Renaudineau
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital Center, Toulouse, France
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| |
Collapse
|