1
|
Yang Y, Xia C, Yao C, Ma X, Shen Z, Chen P, Jiang Q, Gong X. Mucosal immunity and rheumatoid arthritis: An update on mechanisms and therapeutic potential. Autoimmun Rev 2025; 24:103775. [PMID: 39954755 DOI: 10.1016/j.autrev.2025.103775] [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: 12/12/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Rheumatoid Arthritis (RA) is a persistent autoimmune inflammatory disorder that arises from the intricate interaction between genetic predisposition and environmental influences. The progression of RA can be delineated into four distinct phases: initially, the influence of genetic and environmental risk factors; followed by the emergence of systemic autoimmunity; subsequently, an asymptomatic inflammatory phase; and ultimately, the manifestation of clinical arthritis. Recently, the role of mucosal immunity in RA has gained significant attention in research. Evidence from published studies suggests that mucosal immunity not only influences the onset of RA but also plays a crucial role in its progression. Scholars have begun to unravel the intricate links between RA and the mucosal barriers of the gastrointestinal tract, respiratory system, and oral cavity. Specifically, shifts in the mucosal microbiota, dysfunction of mucosal barriers, and the abnormal activation of mucosal immune tissues are all implicated in the pathogenesis of RA.Despite this growing body of knowledge, a comprehensive review of the abnormal mucosal immunity in RA and its therapeutic implications is yet to be conducted. This review emphasizes the driving role of mucosal immune abnormalities in the development of systemic autoimmunity in rheumatoid arthritis (RA). It further explores the therapeutic potential of mucosal immunity in RA, as well as the issues and challenges that need to be addressed in the current research field, providing a new perspective and potential therapeutic targets for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Yuchen Yang
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Congmin Xia
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chuanhui Yao
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xieli Ma
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zhengyao Shen
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Peng Chen
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Quan Jiang
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xun Gong
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
2
|
Duo R, Wang Y, Ma Q, Wang X, Zhang Y, Shen H. MTX-induced gastrointestinal reactions in RA: Prevotella enrichment, gut dysbiosis, and PI3K/Akt/Ras/AMPK pathways. Clin Rheumatol 2025:10.1007/s10067-025-07406-y. [PMID: 40198451 DOI: 10.1007/s10067-025-07406-y] [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: 11/03/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
OBJECTIVES To investigate the role of gut microbiota in methotrexate (MTX)-induced gastrointestinal reactions (MRGR) in patients with rheumatoid arthritis (RA). METHODS As a prospective, single-center, convenience sampling study, stool samples were obtained from 28 RA patients (male: female = 10:18) at Lanzhou University Second Hospital who were undergoing MTX treatment for analysis of their gut microbiota using 16S rRNA gene sequencing. Clinical disease activity (CDAI) and MRGR were assessed after two months of MTX therapy. All data collection periods exceeded one year. Intestinal germ-free mice, generated through antibiotic treatment, received fecal microbiota transplantation (FMT) from the patients, followed by varying doses of MTX to observe MRGR. Intestinal transcriptomics and markers related to intestinal barrier function were subsequently examined. RESULTS Females (84.6%) and high disease activity (CDAI scores, 39.6 ± 11.2 vs 26.3 ± 9.2) were prone to have MRGR in RA patients. Patients with MRGR (PT-GR) showed lower gut microbial diversity versus non-MRGR (PT-noGR). Prevotella abundance, positively correlated with CDAI and MRGR (p < 0.05), was elevated in PT-GR. Administering 10 mg/kg MTX to mice caused intestinal damage. FMT-GR-MTX mice exhibited weight loss (95.2%), morphological deterioration (86.4%), and reduced tight junction proteins (Claudin-1:72.4%; ZO-1:81.2%). Transcriptomics linked upregulated Gβγ/CREB/Atp4b to PI3K/Akt/Ras pathways and downregulated PFK2/PP2 to AMPK signaling in MRGR. CONCLUSION Our study identified notable gut microbiota alterations in RA patients prone to MRGR, with changes in intestinal gene expression and reduced intestinal barrier function potentially contributing to MRGR. These findings suggest potential strategies to mitigate MRGR in RA patients undergoing MTX treatment. Key Points • The RA-related MRGR is correlated with the intestinal microbiota. • Females, low gut diversity, and Prevotella enrichment are MRGR risks in RA. • Upregulated DEGs in MRGR linked to PI3K/Akt, Ras pathways. • Downregulated DEGs in MRGR focus on the AMPK pathway.
Collapse
Affiliation(s)
- Ruixue Duo
- Department of Rheumatology and Immunology, Lanzhou University Second Hospital, 82 Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yining Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Quanzhi Ma
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Xiaoyuan Wang
- Department of Rheumatology and Immunology, Lanzhou University Second Hospital, 82 Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yan Zhang
- Department of Rheumatology and Immunology, Lanzhou University Second Hospital, 82 Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Haili Shen
- Department of Rheumatology and Immunology, Lanzhou University Second Hospital, 82 Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, China.
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
3
|
Park SJ, Kim M, Jeong J, Park YJ, Jeong S, Kim M, Kim HJ, Song J, Kim SM, Chang J, Kim KH, Ko A, Park SM. Association between antibiotic use and the risk of rheumatoid arthritis: a retrospective cohort study in South Korea. Rheumatology (Oxford) 2025; 64:1732-1740. [PMID: 39340800 DOI: 10.1093/rheumatology/keae483] [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: 03/07/2024] [Revised: 07/28/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Certain studies propose that antibiotic use may influence RA incidence, but the clear association between antibiotics and RA remains unclear. Therefore, this study aimed to examine the relationship between antibiotics and RA risk to provide additional epidemiological evidence. METHODS This population-based retrospective cohort study was conducted with adults aged 40 years or older using the Korean National Health Insurance Service database. Antibiotic exposure was measured from 2003 to 2007. Study participants were followed up from 1 January 2008 to 31 December 2019. Multivariable Cox hazard regression was utilized to evaluate adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs) for the risk of RA according to accumulative days of antibiotic use and the number of antibiotic classes used, respectively. RESULTS During 3 395 590 person-years of follow-up, 29 274 cases of RA were identified. Participants who used antibiotics for 91 or more days had a higher risk of RA (aHR, 1.79; 95% CI, 1.67-1.92) than antibiotic non-users. Additionally, individuals who used four or more kinds of antibiotic classes had a higher risk of RA (aHR, 1.61; 95% CI, 1.51-1.71) than those who did not prescribe antibiotics. The risk of RA was positively associated with both higher cumulative days of antibiotic exposure and a larger number of drug classes. These trends were maintained in sensitivity analyses, including variations in antibiotic exposure periods. CONCLUSION Our findings suggest a possible association between the long-term use of antibiotics and RA incidence. Further studies are necessary for a clearer understanding of this association.
Collapse
Affiliation(s)
- Sun Jae Park
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Minkyung Kim
- College of Nursing, Korea University, Seoul, South Korea
| | - Jihui Jeong
- Department of Medicine, Inje University, Busan, South Korea
| | - Young Jun Park
- Medical Research Center, Genomic Medicine Institute, Seoul National University, Seoul, South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, South Korea
| | - Minseo Kim
- College of Medicine, Jeonbuk National University, Jeonju, South Korea
| | - Hye Jun Kim
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jihun Song
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Sung Min Kim
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jooyoung Chang
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyae Hyung Kim
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
- Public Healthcare Center, Seoul National University Hospital, Seoul, South Korea
| | - Ahryoung Ko
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
Kragsnaes MS, Risbo N, Pedersen JK, Obel N, Finckh A, Pedersen AB, Ellingsen T. Antibiotics in inflammatory arthritis and background population one year before and after diagnosis: a nationwide drug utilization study. Rheumatology (Oxford) 2025; 64:1705-1714. [PMID: 39189999 DOI: 10.1093/rheumatology/keae396] [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: 02/06/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
OBJECTIVES To describe antibiotic use in patients with inflammatory arthritis (IA) and in the background population (BP) within one year before and after IA diagnosis. METHODS Using data from Danish nationwide registries, we identified all adults with a first-time diagnosis of RA, PsA, or AS/spondyloarthritis (AS/SpA) from 2010 through 2018. For each IA patient, we randomly sampled 10 persons from the BP, matched on sex and birthdate. We calculated the prevalence (n [%]) of any antibiotic dispensing and the total antibiotic dispensing in the year before and after diagnosis. RESULTS We identified 28 504 new-onset IA patients (RA, n = 16 130; PsA, n = 5988; AS/SpA, n = 6386) and 285 040 BP individuals. Within one year before diagnosis, the total amount of dispensed antibiotics was higher in both RA, PsA and As/SpA compared with the BP (prevalence rate ratios [PRR], 1.48 [1.46; 1.51]; 1.67 [1.62; 1.72]; 1.52 [1.47; 1.56], respectively). The amount increased with 22% in IA patients three months before diagnosis compared with the preceding three-month period. Although the prevalence of any antibiotic dispensing in IA patients decreased in the year following the diagnosis (IA; 40.6%), the total one-year antibiotic dispensing remained constant in RA (PRR 0.99 [0.97; 1.01]), decreased in PsA (0.91 [0.87; 0.94]) and increased in AS/SpA (1.08 [1.04; 1.12]) patients after diagnosis compared with before. CONCLUSION Antibiotics are more frequently dispensed to individuals developing IA compared with the BP. Antibiotic utilization patterns change after IA diagnosis with marked differences among IA subgroups.
Collapse
Affiliation(s)
- Maja S Kragsnaes
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Nickolaj Risbo
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Kristian Pedersen
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Niels Obel
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Axel Finckh
- Division of Rheumatology, Department of Internal Medicine and Department of Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Alma B Pedersen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Torkell Ellingsen
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Gao A, Wu R, Mu Y, Jin R, Jiang S, Gao C, Li X, Wang C. Restoring immune tolerance in pre-RA: immunometabolic dialogue between gut microbiota and regulatory T cells. Front Immunol 2025; 16:1565133. [PMID: 40181974 PMCID: PMC11965651 DOI: 10.3389/fimmu.2025.1565133] [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: 01/22/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Rheumatoid arthritis (RA) is a complex chronic autoimmune disease that remains incurable for most patients. With advances in our understanding of the disease's natural history, the concept of pre-RA has emerged as a window of opportunity to intervene before irreversible joint damage occurs. Numerous studies have indicated that the key step driving autoimmunity in early pre-RA lies at an extra-articular site, which is closely related to the regulatory T (Treg) cell-established immune tolerance to the gut microbiota. The intricate immunometabolic crosstalk between Treg cells and the gut microbiota is beginning to be understood, with the re-recognition of Treg cells as metabolic sensors in recent years. In the future, deciphering their immunometabolic dialogue may help to elucidate the underlying mechanisms of pre-RA. Identifying novel biological pathways in the pre-RA stage will bring insights into restoring immune tolerance, thereby potentially curing or preventing the onset of RA.
Collapse
Affiliation(s)
- Anqi Gao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Yanfei Mu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Ruqing Jin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Saixin Jiang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Wu T, Li Y, Liu Y, Chu CQ. Preclinical RA: How to halt its progression. Best Pract Res Clin Rheumatol 2025; 39:102030. [PMID: 39721896 DOI: 10.1016/j.berh.2024.102030] [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: 12/04/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder with a complex pathogenesis that evolves through various stages before clinical symptoms emerge. This review outlines the natural history of RA, starting from genetic predisposition and environmental triggers to preclinical autoimmunity and subsequent joint inflammation. Key genetic factors interact with environmental elements like smoking and infections, producing autoantibodies such as anti-citrullinated protein antibodies (ACPA) and rheumatoid factor, which precede clinical manifestations by several years. The preclinical phases offer critical opportunities for intervention aiming at halting disease progression. Preventive strategies including lifestyle modifications, dietary interventions, and targeted immune modulation may halt the progression to clinical RA in those at-risk individuals.
Collapse
Affiliation(s)
- Tong Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Lecheng Hospital, Sichuan University, Boao, Hainan, 571435, China.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
7
|
Jin J, Cai X, Rao P, Xu J, Li J. Microbiota and immune dynamics in rheumatoid arthritis: Mechanisms and therapeutic potential. Best Pract Res Clin Rheumatol 2025; 39:102035. [PMID: 39863438 DOI: 10.1016/j.berh.2025.102035] [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: 12/12/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease with growing evidence implicating the microbiota as a critical contributor to its pathogenesis. This review explores the multifaceted roles of microbial dysbiosis in RA, emphasizing its impact on immune cell modulation, autoantibody production, gut barrier integrity, and joint inflammation. Animal models reveal how genetic predisposition and environmental factors interact with specific microbial taxa to influence disease susceptibility. Dysbiosis-driven metabolic disruptions, including alterations in short-chain fatty acids and bile acids, further exacerbate immune dysregulation and systemic inflammation. Emerging therapeutic strategies-probiotics, microbial metabolites, fecal microbiota transplantation, and antibiotics-offer innovative avenues for restoring microbial balance and mitigating disease progression. By integrating microbiota-targeted approaches with existing treatments, this review highlights the potential to revolutionize RA management through precision medicine and underscores the need for further research to harness the microbiota's therapeutic potential.
Collapse
Affiliation(s)
- Jiayang Jin
- Department of Rheumatology and Immunology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xuanlin Cai
- Department of Rheumatology and Immunology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Peishi Rao
- Department of Rheumatology and Immunology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Jing Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China; Division of Rheumatology, Department of Medicine, University of Colorado, No. 11, Xizhimen South Street, Xicheng District, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Zhu X, Lu H, Li W, Niu S, Xue J, Sun H, Zhang J, Zhang Z. Ferroptosis Induces gut microbiota and metabolic dysbiosis in Collagen-Induced arthritis mice via PAD4 enzyme. Gene 2025; 936:149106. [PMID: 39561902 DOI: 10.1016/j.gene.2024.149106] [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: 09/18/2024] [Revised: 10/17/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation and joint destruction, with emerging evidence implicating gut microbiota dysbiosis in its pathogenesis. The current study explores the role of ferroptosis, a form of regulated cell death driven by iron-dependent lipid peroxidation, in modulating gut microbiota and metabolic dysregulation through the enzyme peptidyl arginine deiminase 4 (PAD4) in collagen-induced arthritis (CIA) mouse model. Our findings demonstrate that ferroptosis exacerbates RA-related inflammatory responses and joint damage by upregulating PAD4 expression, which, in turn, influences the gut microbial composition and associated metabolite profiles. Erastin, a known ferroptosis agonist, significantly increased the relative abundance of pro-inflammatory bacteria such as Proteobacteria while reducing beneficial taxa like Firmicutes and Bacteroidetes. This microbial shift was associated with heightened oxidative stress and an imbalance in key metabolites, such as lysophosphatidyl ethanolamine 14:0 (LysoPE 14:0), further exacerbated by ferroptosis. Co-treatment with GSK484, a PAD4 inhibitor, reversed these effects, restoring microbial homeostasis and reducing joint inflammation. This study suggests that ferroptosis-mediated PAD4 activity contributes to RA pathogenesis by disrupting the gut-joint axis, providing novel insights into potential therapeutic targets for RA. Our results highlight the intricate interplay between immune-mediated cell death, gut microbiota, and systemic inflammation, emphasizing the importance of ferroptosis as a therapeutic target in mitigating RA progression.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hanya Lu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wenjing Li
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Sijia Niu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jiawei Xue
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Haoyuan Sun
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin 150001, China
| | - Juan Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
9
|
Ng BCK, Lassere M. The role of the gastrointestinal microbiome on rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis and reactive arthritis: A systematic review. Semin Arthritis Rheum 2025; 70:152574. [PMID: 39644691 DOI: 10.1016/j.semarthrit.2024.152574] [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: 01/23/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND There is an increasing body of literature observing a state of dysbiosis in the gut microbiome in different autoimmune conditions including inflammatory arthritis. It is unknown whether the microbiome can be a biomarker for prognostication purposes or for stratification of treatment strategies. This review aims to evaluate the existing evidence on the association between the microbiome and inflammatory arthritis, including rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ankylosing spondylitis (AS) and reactive arthritis (ReA) population groups. METHODS This systematic review was performed based on methods from the Cochrane guidelines and reported based on PRISMA criteria. Studies exploring the microbiome of patients with RA, AS, PsA or ReA compared with controls via 16s rRNA or shotgun sequencing were evaluated. The outcomes of interest include alpha and beta diversity, abundance or depletion of organisms and functional analysis. Literature up to August 2024 was retrieved searching the databases PubMed, Medline, ScienceDirect, Scopus, Web of Science, Cochrane, EMBASE and CINAHL. All references were systematically evaluated by two reviewers. Quality of the studies were evaluated by the Newcastle-Ottawa Scale. FINDINGS The review yielded 25,794 search results, of which 53 studies were included for the RA group, 34 studies for the AS group, 6 studies for the PsA group and 2 studies for the ReA group. Reduced diversity has been observed in disease groups and in patients with higher disease activity. INTERPRETATION There are limited longitudinal studies on the role of the microbiome in inflammatory arthritis, in particular PsA. Existing cross-sectional studies suggest altered microbiome in disease states compared with controls. Further studies are required to understand the utility of the microbiome as a biomarker to better understand prognosis and tailor treatments.
Collapse
Affiliation(s)
- Beverly Cheok Kuan Ng
- Department of Rheumatology, St George Hospital, Australia; University of New South Wales, School of Public Health and Community Medicine, Australia.
| | - Marissa Lassere
- Department of Rheumatology, St George Hospital, Australia; University of New South Wales, School of Public Health and Community Medicine, Australia
| |
Collapse
|
10
|
Yoneda K, Sendo S, Okano T, Shimizu H, Yamada H, Nishimura K, Ueda Y, Saegusa J. Impact of dysregulated microbiota-derived C18 polyunsaturated fatty acid metabolites on arthritis severity in mice with collagen-induced arthritis. Front Immunol 2025; 15:1444892. [PMID: 39850876 PMCID: PMC11754244 DOI: 10.3389/fimmu.2024.1444892] [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: 06/06/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Objective We aimed to evaluate microbiome and microbiota-derived C18 dietary polyunsaturated fatty acids (PUFAs), such as conjugated linoleic acid (CLA), and to investigate their differences that correlate with arthritis severity in collagen-induced arthritis (CIA) mice. Methods On day 84 after induction, during the chronic phase of arthritis, cecal samples were analyzed using 16S rRNA sequencing, and plasma and cecal digesta were evaluated using liquid chromatography-tandem mass spectrometry. Differences in microbial composition between 10 control (Ctrl) and 29 CIA mice or between the mild and severe subgroups based on arthritis scores were identified. The cecal metabolite profile and its correlation with the microbiome were evaluated with respect to arthritis severity. Results The hydroxy and oxo metabolite levels were higher in CIA mice than in Ctrl mice, some of which, including 10-hydroxy-cis-6-18:1, were positively correlated with arthritis scores. The 9-trans,11-trans CLA levels in CIA mice had a negative linear correlation with arthritis scores. Microbial diversity was lower in severe CIA mice than in mild CIA or Ctrl mice. The abundance of Lactobacillus relatively increased in the severe subgroup of CIA mice compared with that in the mild subgroup and was positively correlated with arthritis severity. Conclusion Alterations in gut microbiota and microbiota-derived C18 PUFA metabolites are associated in CIA mice and correlated with arthritis scores, indicating that plasma or fecal C18 PUFA metabolites can be potential biomarkers for arthritis severity and dysbiosis.
Collapse
Affiliation(s)
- Katsuhiko Yoneda
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sho Sendo
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takaichi Okano
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Hirotaka Yamada
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Nishimura
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yo Ueda
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
11
|
Zhang D, Zhang Z, Liao L, Dong B, Xiong X, Qin X, Fan X. Impact of fecal microbiota transplantation on lung function and gut microbiome in an ARDS rat model: A multi-omics analysis including 16S rRNA sequencing, metabolomics, and transcriptomics. Int J Immunopathol Pharmacol 2025; 39:3946320251333982. [PMID: 40265594 DOI: 10.1177/03946320251333982] [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] [Indexed: 04/24/2025] Open
Abstract
OBJECTIVE Acute respiratory distress syndrome (ARDS) is a severe pulmonary condition characterized by inflammation and lung damage, frequently resulting in poor clinical outcomes. Recent studies suggest that the gut-lung axis, mediated by gut microbiota, is critical in ARDS progression. This study investigates the therapeutic potential of fecal microbiota transplantation (FMT) in an ARDS rat model (n = 6). INTRODUCTION The pathogenesis of ARDS involves complex interactions between the lungs and gut, with microbiota playing a key role. Understanding the effects of FMT on lung function and gut microbiota may provide new therapeutic strategies for ARDS management. METHODS Sprague-Dawley rats were pre-treated with a broad-spectrum antibiotic cocktail to create a germ-free state and subsequently exposed to intranasal lipopolysaccharide to induce ARDS. The rats then received FMT treatment. Lung samples were analyzed using histopathology and transcriptomics. Fecal samples were analyzed using 16S rRNA sequencing and metabolomics. RESULTS FMT treatment significantly reduced lung injury and improved pulmonary function, as evidenced by increased partial pressure of arterial oxygen (PaO2) and decreased partial pressure of arterial carbon dioxide (PaCO2). FMT also significantly altered in gut microbiota composition by regulating the gut microbiota composition of Akkermansia and Lactobacillus, restoring the abundance of genera such as Muribaculaceae, Clostridia_UCG-014, Prevotella, and Adlercreutzia, while reducing Romboutsia. FMT restored key metabolic pathways involved in lipid metabolism, amino acid biosynthesis, and immune regulation, including the modulation of immune pathways like mTOR signaling. These alterations contribute to reduced lung injury and improved pulmonary function. CONCLUSION These findings indicate that FMT may exert its beneficial effects in ARDS by modulating the gut microbiota and enhancing metabolic and immune responses. However, given that this study remains in the preclinical stage, further validation in clinical studies is necessary before considering clinical application.
Collapse
Affiliation(s)
- Dongwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Zhenqiang Zhang
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Longxiong Liao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Biying Dong
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xuejun Qin
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
12
|
Geng Q, Wang Z, Shi T, Wen C, Xu J, Jiao Y, Diao W, Gu J, Wang Z, Zhao L, Deng T, Xiao C. Cannabidiol regulates L-carnitine and butyric acid metabolism by modulating the gut microbiota to ameliorate collagen-induced arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156270. [PMID: 39591767 DOI: 10.1016/j.phymed.2024.156270] [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: 09/08/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is one of the most common autoimmune diseases, affecting multiple systems in the body. Cannabidiol (CBD) is one of the most medically valuable active ingredients in cannabis. At present, CBD has been shown to alleviate the progression of RA; however, owing to its multiple targets, the mechanism of CBD is not clear. METHODS On the basis of the gut microbiota, we explored the mechanism by which CBD inhibits RA progression. Metagenomic and nontargeted metabolomic analyses were used to determine the changes in the intestinal ecology and plasma metabolites of collagen-induced arthritis (CIA) rats after CBD treatment. RESULTS CBD reversed gut dysbiosis in CIA rats, notably altering the abundances of Allobaculum_unclassified, Allobaculum_fili, and Prevotella_unclassified. In addition, metabolomic analysis confirmed that CBD increased the contents of butyric acid and L-carnitine. Allobaculum could produce butyric acid and Prevotella could accelerate the metabolism of L-carnitine. In addition, in vitro experiments demonstrated that L-carnitine participated in the regulation of neutrophils, macrophages and RA-fibroblast-like synoviocytes (RA-FLSs), which was consistent with the synovial changes in CIA rats caused by CBD. CONCLUSION In summary, CBD increased the plasma contents of butyric acid and L-carnitine by altering the abundances of gut microbiota, thereby inhibiting inflammation in neutrophils, macrophages and RA-FLSs. Our study is the first to explain the mechanism by which CBD alleviates progression in CIA rats from the perspective of gut microbes and metabolites, providing new views into CBD mechanisms, which warrants clinical attention.
Collapse
Affiliation(s)
- Qishun Geng
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhaoran Wang
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Tong Shi
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Chaoying Wen
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiahe Xu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Yi Jiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China; Beijing University of Chinese Medicine, China-Japan Friendship Clinical Medical College, Beijing, 100029, China
| | - Wenya Diao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China; Beijing University of Chinese Medicine, China-Japan Friendship Clinical Medical College, Beijing, 100029, China
| | - Jienan Gu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China; Beijing University of Chinese Medicine, China-Japan Friendship Clinical Medical College, Beijing, 100029, China
| | - Zihan Wang
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical Medical College, Beijing, 100029, China; Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Lu Zhao
- China-Japan Friendship Hospital, Capital Medical University, Beijing, 100029, China
| | - Tingting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Cheng Xiao
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China; Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
13
|
Scher JU, Nayak R, Clemente JC. Microbiome research in autoimmune and immune-mediated inflammatory diseases: lessons, advances and unmet needs. Ann Rheum Dis 2025; 84:9-13. [PMID: 39874238 DOI: 10.1136/ard-2024-225735] [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: 09/19/2024] [Indexed: 10/19/2024]
Abstract
The increasing prevalence of autoimmune and immune-mediated diseases (AIMDs) underscores the need to understand environmental factors that contribute to their pathogenesis, with the microbiome emerging as a key player. Despite significant advancements in understanding how the microbiome influences physiological and inflammatory responses, translating these findings into clinical practice remains challenging. This viewpoint reviews the progress and obstacles in microbiome research related to AIMDs, examining molecular techniques that enhance our understanding of microbial contributions to disease. We discuss significant discoveries linking specific taxa and metabolites to diseases such as rheumatoid arthritis, systemic lupus erythematosus and spondyloarthritis, highlighting the role of gut dysbiosis and host-microbiome interactions. Furthermore, we explore the potential of microbiome-based therapeutics, including faecal microbiota transplantation and pharmacomicrobiomics, while addressing the challenges of identifying robust microbial targets. We advocate for integrative, transdisease studies and emphasise the need for diverse cohort research to generalise findings across populations. Understanding the microbiome's role in AIMDs will pave the way for personalised medicine and innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, NYU Psoriatic Arthritis Center, and NYU Colton Center for Autoimmunity, New York University School of Medicine, New York, New York, USA.
| | - Renuka Nayak
- University of California San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, San Francisco, California, USA. https://x.com/RNayak
| | - Jose C Clemente
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
Li J, Fan R, Zhang Z, Zhao L, Han Y, Zhu Y, Duan JA, Su S. Role of gut microbiota in rheumatoid arthritis: Potential cellular mechanisms regulated by prebiotic, probiotic, and pharmacological interventions. Microbiol Res 2025; 290:127973. [PMID: 39541714 DOI: 10.1016/j.micres.2024.127973] [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: 08/13/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that primarily affects joints and multiple organs and systems, which is long-lasting and challenging to cure and significantly impacting patients' quality of life. Alterations in the composition of intestinal flora in both preclinical and confirmed RA patients indicate that intestinal bacteria play a vital role in RA immune function. However, the mechanism by which the intestinal flora is regulated to improve the condition of RA is not fully understood. This paper reviews the methods of regulating gut microbiota and its metabolites through prebiotics, probiotics, and pharmacological interventions, and discusses their effects on RA. Additionally, it explores the potential predictive role of cellular therapy mechanisms of intestinal flora in treating RA. These findings suggest that restoring the ecological balance of intestinal flora and regulating intestinal barrier function may enhance immune system function, thereby improving rheumatoid arthritis. This offers new insights into its treatment.
Collapse
Affiliation(s)
- Jiashang Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruoying Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhe Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lihui Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Han
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yue Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shulan Su
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
15
|
Remalante-Rayco P, Nakamura A. Year in Review: Novel Insights in the Pathogenesis of Spondyloarthritis - SPARTAN 2024 Annual Meeting Proceedings. Curr Rheumatol Rep 2024; 27:9. [PMID: 39731620 DOI: 10.1007/s11926-024-01176-3] [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] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE OF REVIEW The canonical pathogenesis of spondyloarthritis (SpA) involves inflammation driven by HLA-B27, type 3 immunity, and gut microbial dysregulation. This review based on information presented at the SPARTAN meeting highlights studies on the pathogenesis of SpA from the past year, focusing on emerging mechanisms such as the roles of microbe-derived metabolites, microRNAs (miRNAs) and cytokines in plasma exosomes, specific T cell subsets, and neutrophils. RECENT FINDINGS The induction of arthritis in a preclinical model through microbiota-driven alterations in tryptophan catabolism provides new insights as to how intestinal dysbiosis may activate disease via the gut-joint axis. Immune activation may likewise be modulated by dysregulated miRNAs and cytokines contained in plasma exosomes, which appear to influence the homeostasis of both effector T cells and regulatory T cells (Tregs). Closer examination of T cells in animal models has uncovered distinct transcriptional and functional profiles between gut and joint Tregs, as well as highly specific T cell subsets that can be targeted to induce disease modification. Newer studies including both SpA patients and preclinical models have focused on the key role of neutrophils as drivers of inflammation and new bone formation in hypoxic, inflammation-driven tissue environments, potentially through interactions with adipocytes and mesenchymal stem cells. Functional studies and high-throughput techniques using samples from SpA patients and preclinical models have significantly enhanced our understanding of SpA pathogenesis, offering new insights into the specific mechanisms of immune regulation and identifying promising therapeutic targets.
Collapse
Affiliation(s)
- Patricia Remalante-Rayco
- Schroeder Arthritis Institute, Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, ON, Canada.
- Translational Institute of Medicine, School of Medicine, Department of Medicine, Queen's University, 94 Stuart Street, Kingston, ON, K7L 3N6, Canada.
- Rheumatology Clinic, Kingston Health Science Centre, Kingston, ON, Canada.
| |
Collapse
|
16
|
Lin YH, Lian WS, Wu RW, Chen YS, Wu SL, Ko JY, Wang SY, Jahr H, Wang FS. Trimethylamine-N-oxide accelerates osteoporosis by PERK activation of ATF5 unfolding. Cell Mol Life Sci 2024; 82:13. [PMID: 39719538 DOI: 10.1007/s00018-024-05501-y] [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: 02/06/2024] [Revised: 08/21/2024] [Accepted: 11/05/2024] [Indexed: 12/26/2024]
Abstract
Imbalances in gut microbiota and their metabolites have been implicated in osteoporotic disorders. Trimethylamine-n-oxide (TMAO), a metabolite of L-carnitine produced by gut microorganisms and flavin-containing monooxygenase-3, is known to accelerate tissue metabolism and remodeling; however, its role in bone loss remained unexplored. This study investigates the relationship between gut microbiota dysbiosis, TMAO production, and osteoporosis development. We further demonstrate that the loss of beneficial gut microbiota is associated with the development of murine osteoporosis and alterations in the serum metabolome, particularly affecting L-carnitine metabolism. TMAO emerges as a functional metabolite detrimental to bone homeostasis. Notably, transplantation of mouse gut microbiota counteracts obesity- or estrogen deficiency-induced TMAO overproduction and mitigates key features of osteoporosis. Mechanistically, excessive TMAO intake augments bone mass loss by inhibiting bone mineral acquisition and osteogenic differentiation. TMAO activates the PERK and ATF4-dependent disruption of endoplasmic reticulum autophagy and suppresses the folding of ATF5, hindering mitochondrial unfolding protein response (UPRmt) in osteoblasts. Importantly, UPRmt activation by nicotinamide riboside mitigates TMAO-induced inhibition of mineralized matrix biosynthesis by preserving mitochondrial oxidative phosphorylation and mitophagy. Collectively, our findings revealed that gut microbiota dysbiosis leads to TMAO overproduction, impairing ER homeostasis and UPRmt, thereby aggravating osteoblast dysfunction and development of osteoporosis. Our study elucidates the catabolic role of gut microflora-derived TMAO in bone integrity and highlights the therapeutic potential of healthy donor gut microbiota transplantation to alter the progression of osteoporosis.
Collapse
Affiliation(s)
- Yu-Han Lin
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wei-Shiung Lian
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung, 833, Taiwan
| | - Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung, 833, Taiwan
| | - Shin-Long Wu
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung, 833, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung, 833, Taiwan
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH, Aachen, Germany
- Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Feng-Sheng Wang
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung, 833, Taiwan.
| |
Collapse
|
17
|
Yang D, Lv G, Wu Y, Guo W, Wang Y, Hu J, Li N, Zheng F, Dai Y, Pi Z, Yue H. Licorice-regulated gut-joint axis for alleviating collagen-induced rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156203. [PMID: 39510013 DOI: 10.1016/j.phymed.2024.156203] [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: 05/25/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is partially affected by the integrity of the intestinal barrier. Licorice (GC), a medicinal and food-related herb, exhibits potent anti-inflammatory activity; however, studies on its mechanisms of action in RA are limited. METHOD Using a bovine type-II collagen-induced arthritis rat model, this study examined how GC influences the gut-joint axis to decrease RA. The Th17/Treg cell ratios in the blood, colon, and joints were also measured. Metabolomics and 16S rRNA sequencing were applied to explore the effects of variations in gut flora and metabolites. RESULTS The arthropathological slices, inflammation markers, and joint inflammation index scores in the GC treatment group significantly differed from those in the CIA group. Studies on the effect of GC on the gut-joint axis showed changes in the levels of lipopolysaccharide and diamine oxidase, both directly associated with intestinal permeability. ZO-1, occludin, and claudin-1, three intestinal tight-junction proteins, may express themselves more when exposed to GC. By maintaining an appropriate Th17/Treg cell ratio in the blood, colon, and joints, GC may reduce impaired to the intestinal barrier. An imbalance in the intestinal microenvironment, caused by modifications in gut flora and endogenous substances, can damage the intestinal barrier. GC may modify the relative abundances of Papillibacter, Clostridium, Eubacterium, Helicobacter, Provotella, and Barnesiella during RA treatment by repairing the intestinal barrier. The metabolic differences were mainly related to primary bile acid biosynthesis, pyrimidine metabolism, steroid biosynthesis, biotin metabolism, and sphingolipid metabolism. A fecal microbiota transplantation experiment confirmed the involvement of the gut microbiota and its metabolites in GC-mediated RA therapy. CONCLUSION The results demonstrated that GC repairs the intestinal barrier and adjusts the gut-joint axis to manage immunological imbalance in RA.
Collapse
Affiliation(s)
- Di Yang
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Guangfu Lv
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Yongxi Wu
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Wentao Guo
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Yuchen Wang
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Jiannan Hu
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Nian Li
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Fei Zheng
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Yulin Dai
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China
| | - Zifeng Pi
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China.
| | - Hao Yue
- Changchun University of Chinese Medicine, No. 1035 Boshuo Rd, Nanguan District, Changchun 130117, China.
| |
Collapse
|
18
|
Wang H, Cai Y, Wu W, Zhang M, Dai Y, Wang Q. Exploring the role of gut microbiome in autoimmune diseases: A comprehensive review. Autoimmun Rev 2024; 23:103654. [PMID: 39384149 DOI: 10.1016/j.autrev.2024.103654] [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: 07/07/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 10/11/2024]
Abstract
As the industrialized society advances, there has been a gradual increase in the prevalence of autoimmune disorders. A probe into the fundamental causes has disclosed several factors in modern society that have an influence on the gut microbiome. These dramatic shifts in the gut microbiome are likely to be one of the reasons for the disarray in the immune system, and the relationship between the immune system and the gut microbiome emerging as a perennial hot topic of research. This review enumerates the findings from sequencing studies of gut microbiota on seven autoimmune diseases (ADs): Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), Ankylosing Spondylitis (AS), Systemic Sclerosis (SSc), Sjögren's Syndrome (SjS), Juvenile Idiopathic Arthritis (JIA), and Behçet's Disease (BD). It aims to identify commonalities in changes in the gut microbiome within the autoimmune disease cohort and characteristics specific to each disease. The dysregulation of the gut microbiome involves a disruption of the internal balance and the balance between the external environment and the host. This dysregulation impacts the host's immune system, potentially playing a role in the development of ADs. Damage to the gut epithelial barrier allows potential pathogens to translocate to the mucosal layer, contacting epithelial cells, disrupting tight junctions, and being recognized by antigen-presenting cells, which triggers an immune response. Primed T-cells assist B-cells in producing antibodies against pathogens; if antigen mimicry occurs, an immune response is generated in extraintestinal organs during immune cell circulation, clinically manifesting as ADs. However, current research is limited; advancements in sequencing technology, large-scale cohort studies, and fecal microbiota transplantation (FMT) research are expected to propel this field to new peaks.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yueshu Cai
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Wenqi Wu
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Miaomiao Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yong Dai
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China.
| |
Collapse
|
19
|
Guo Y, Feng H, Du L, Yu Z. Patterns of antibiotic resistance genes and virulence factor genes in the gut microbiome of patients with osteoarthritis and rheumatoid arthritis. Front Microbiol 2024; 15:1427313. [PMID: 39633808 PMCID: PMC11615078 DOI: 10.3389/fmicb.2024.1427313] [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: 05/03/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Background The gut microbiome compositions of osteoarthritis (OA) and rheumatoid arthritis (RA) patients have been revealed; however, the functional genomics, particularly antibiotic resistance genes (ARGs) and virulence factor genes (VFGs), have not yet been explored. Methods We used gut metagenomic data to elucidate the distribution of ARGs and VFGs. Building on these differences in gut microbiome, we developed a diagnostic model using a random forest classifier based on ARG and VFG abundances. Results Our results indicated that both OA and RA patients exhibit significantly higher alpha diversity in ARGs, as measured by observed genes, the Shannon index, and the Simpson index, compared to healthy controls. However, this increased diversity is not significantly different between OA and RA patients. In contrast, VFGs showed higher diversity in RA patients than in healthy individuals, which was not as pronounced in OA patients. An analysis of the top 20 ARGs and VFGs revealed a largely similar composition between the three groups, with notable exceptions of certain genes that were uniquely enriched in either OA or RA patients. This suggests unique microbial patterns associated with each condition. Our beta diversity analysis further demonstrated distinct distributions of ARG and VFG profiles across the three groups, with several genes significantly enriched in both OA and RA patients, indicating potential markers for these diseases. The model achieved high accuracy (74.7-83.6%) when distinguishing both OA and RA from healthy controls using ARG profiles and substantial accuracy using VFG profiles. Conclusion These results support the potential of ARGs and VFGs as reliable biomarkers for diagnosing OA and RA.
Collapse
Affiliation(s)
| | | | | | - Zhenghong Yu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
20
|
Ma F, Li Z, Liu H, Chen S, Zheng S, Zhu J, Shi H, Ye H, Qiu Z, Gao L, Han B, Yang Q, Wang X, Zhang Y, Cheng L, Fan H, Lv S, Zhao X, Zhou H, Li J, Hong M. Dietary-timing-induced gut microbiota diurnal oscillations modulate inflammatory rhythms in rheumatoid arthritis. Cell Metab 2024; 36:2367-2382.e5. [PMID: 39260371 DOI: 10.1016/j.cmet.2024.08.007] [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: 02/07/2024] [Revised: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune condition characterized by inflammatory activity with distinct rhythmic fluctuations. However, the precise mechanisms governing these inflammatory rhythms remain elusive. Here, we explore the interaction between dietary patterns, gut microbiota diurnal oscillations, and the rhythmicity of RA in both collagen-induced arthritis (CIA) mice and patients with RA and highlight the significance of dietary timing in modulating RA inflammatory rhythms linked to gut microbiota. Specifically, we discovered that Parabacteroides distasonis (P. distasonis) uses β-glucosidase (β-GC) to release glycitein (GLY) from the diet in response to daily nutritional cues, influencing RA inflammatory rhythms dependent on the sirtuin 5-nuclear factor-κB (SIRT5-NF-κB) axis. Notably, we validated the daily fluctuations of P. distasonis-β-GC-GLY in patients with RA through continuous sampling across day-night cycles. These findings underscore the crucial role of dietary timing in RA rhythmicity and propose potential clinical implications for novel therapeutic strategies to alleviate arthritis.
Collapse
Affiliation(s)
- Fopei Ma
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Zhuang Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Haihua Liu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shixian Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Songyuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Junqing Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Hao Shi
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Haixin Ye
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Zhantu Qiu
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Lei Gao
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Bingqi Han
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Qian Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xing Wang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yang Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Lifang Cheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huijie Fan
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Shuaijun Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China.
| | - Mukeng Hong
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
21
|
Zhang X, Yin M, Zhang D, Cao D, Hou X, Xu Z, Wen C, Zhou J. Metabolomics Reveals Disturbed Amino Acid Metabolism During Different Stages of RA in Collagen-Induced Arthritis Mice. Inflammation 2024; 47:1853-1867. [PMID: 39212888 DOI: 10.1007/s10753-024-02123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease featured by chronic synovitis and progressive joint damage. Early treatment before the onset of clinical symptoms (also known as the pre-RA stage) may slow or stop the progression of the disease. We sought to discover the dynamic metabolic changes during the evolution of collagen-induced arthritis (CIA) to better characterize the disease stages. Untargeted metabolomics analysis using gas chromatography-mass spectrometry revealed that the metabolic profiles of CIA mice gradually differed from that of the control group with the progression of the disease. During the induction phase, the CIA group showed some metabolic alterations in galactose metabolism, arginine biosynthesis, tricarboxylic acid cycle (TCA cycle), pyruvate metabolism, and starch/sucrose metabolism. During the early inflammatory phase, no joint swelling was observed in CIA mice, and metabolites changed mainly involving amino acid metabolism (arginine biosynthesis, arginine/proline metabolism, phenylalanine/tyrosine/tryptophan biosynthesis), and glutathione metabolism. During the peak inflammatory phase, severe arthritis symptoms were observed in CIA mice, and there were more extensive metabolic alterations in valine/leucine/isoleucine biosynthesis, phenylalanine/tyrosine/tryptophan biosynthesis, TCA cycle, galactose metabolism, and arginine biosynthesis. Moreover, the reduction of specific amino acids, such as glycine, serine, and proline, during the early stages may result in an imbalance in macrophage polarization and enhance the inflammatory response in CIA mice. Our study confirmed that specific perturbations in amino acid metabolism have occurred in CIA mice prior to the onset of joint symptoms, which may be related to autoimmune disorders. The findings could provide insights into the metabolic mechanism and the diagnosis of pre-RA.
Collapse
Affiliation(s)
- Xiafeng Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengdi Yin
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dingyi Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dandan Cao
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoxiao Hou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhenghao Xu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Academy of Chinese Medical Science, Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Academy of Chinese Medical Science, Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jia Zhou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Academy of Chinese Medical Science, Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
22
|
Holers VM, Demoruelle KM, Buckner JH, James EA, Firestein GS, Robinson WH, Steere AC, Zhang F, Norris JM, Kuhn KA, Deane KD. Distinct mucosal endotypes as initiators and drivers of rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:601-613. [PMID: 39251771 DOI: 10.1038/s41584-024-01154-0] [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] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Rheumatoid arthritis (RA) is a potentially devastating autoimmune disease. The great majority of patients with RA are seropositive for anti-citrullinated protein antibodies (ACPAs), rheumatoid factors, or other autoantibodies. The onset of clinically apparent inflammatory arthritis meeting classification criteria (clinical RA) is preceded by ACPA seropositivity for an average of 3-5 years, a period that is designated as 'at-risk' of RA for ACPA-positive individuals who do not display signs of arthritis, or 'pre-RA' for individuals who are known to have progressed to developing clinical RA. Prior studies of individuals at-risk of RA have associated pulmonary mucosal inflammation with local production of ACPAs and rheumatoid factors, leading to development of the 'mucosal origins hypothesis'. Recent work now suggests the presence of multiple distinct mucosal site-specific mechanisms that drive RA evolution. Indicatively, subsets of individuals at-risk of RA and patients with RA harbour a faecal bacterial strain that has exhibited arthritogenic activity in animal models and that favours T helper 17 (TH17) cell responses in patients. Periodontal inflammation and oral microbiota have also been suggested to promote the development of arthritis through breaches in the mucosal barrier. Herein, we argue that mucosal sites and their associated microbial strains can contribute to RA evolution via distinct pathogenic mechanisms, which can be considered causal mucosal endotypes. Future therapies instituted for prevention in the at-risk period, or, perhaps, during clinical RA as therapeutics for active arthritis, will possibly have to address these individual mechanisms as part of precision medicine approaches.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA.
| | | | | | | | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
23
|
Bakinowska E, Stańska W, Kiełbowski K, Szwedkowicz A, Boboryko D, Pawlik A. Gut Dysbiosis and Dietary Interventions in Rheumatoid Arthritis-A Narrative Review. Nutrients 2024; 16:3215. [PMID: 39339815 PMCID: PMC11435214 DOI: 10.3390/nu16183215] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease. The pathogenesis of RA is complex and involves interactions between articular cells, such as fibroblast-like synoviocytes, and immune cells. These cells secrete pro-inflammatory cytokines, chemokines, metalloproteinases and other molecules that together participate in joint degradation. The current evidence suggests the important immunoregulatory role of the gut microbiome, which can affect susceptibility to diseases and infections. An altered microbiome, a phenomenon known as gut dysbiosis, is associated with the development of inflammatory diseases. Importantly, the profile of the gut microbiome depends on dietary habits. Therefore, dietary elements and interventions can indirectly impact the progression of diseases. This review summarises the evidence on the involvement of gut dysbiosis and diet in the pathogenesis of RA.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Agata Szwedkowicz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
24
|
Zhuang R, Yan Z, Gao Y, Nurmamat A, Zhang S, Xiu M, Zhou Y, Pang Y, Li D, Zhao L, Liu X, Han Y. Evolutionary and functional analysis of metabotropic glutamate receptors in lampreys. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1861-1877. [PMID: 38951427 DOI: 10.1007/s10695-024-01374-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024]
Abstract
The metabotropic glutamate receptor (mGluR, GRM) family is involved in multiple signaling pathways and regulates neurotransmitter release. However, the evolutionary history, distribution, and function of the mGluRs family in lampreys have not been determined. Therefore, we identified the mGluRs gene family in the genome of Lethenteron reissneri, which has been conserved throughout vertebrate evolution. We confirmed that Lr-GRM3, Lr-GRM5, and Lr-GRM7 encode three types of mGluRs in lamprey. Additionally, we investigated the distribution of Lr-GRM3 within this species by qPCR and Western blotting. Furthermore, we conducted RNA sequencing to investigate the molecular function of Lr-GRM3 in lamprey. Our gene expression profile revealed that, similar to that in jawed vertebrates, Lr-GRM3 participates in multiple signal transduction pathways and influences synaptic excitability in lampreys. Moreover, it also affects intestinal motility and the inflammatory response in lampreys. This study not only enhances the understanding of mGluRs' gene evolution but also highlights the conservation of GRM3's role in signal transduction while expanding our knowledge of its functions specifically within lampreys. In summary, our experimental findings provide valuable insights for studying both the evolution and functionality of the mGluRs family.
Collapse
Affiliation(s)
- Ruyu Zhuang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Zihao Yan
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Yicheng Gao
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Ayqeqan Nurmamat
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Shuyuan Zhang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Min Xiu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Yuesi Zhou
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Ya Pang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Ding Li
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Liang Zhao
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Xin Liu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Yinglun Han
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China.
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|
25
|
Seida I, Al Shawaf M, Mahroum N. Fecal microbiota transplantation in autoimmune diseases - An extensive paper on a pathogenetic therapy. Autoimmun Rev 2024; 23:103541. [PMID: 38593970 DOI: 10.1016/j.autrev.2024.103541] [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: 01/11/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
The role of infections in the pathogenesis of autoimmune diseases has long been recognized and reported. In addition to infectious agents, the internal composition of the "friendly" living bacteria, (microbiome) and its correlation to immune balance and dysregulation have drawn the attention of researchers for decades. Nevertheless, only recently, scientific papers regarding the potential role of transferring microbiome from healthy donor subjects to patients with autoimmune diseases has been proposed. Fecal microbiota transplantation or FMT, carries the logic of transferring microorganisms responsible for immune balance from healthy donors to individuals with immune dysregulation or more accurately for our paper, autoimmune diseases. Viewing the microbiome as a pathogenetic player allows us to consider FMT as a pathogenetic-based treatment. Promising results alongside improved outcomes have been demonstrated in patients with different autoimmune diseases following FMT. Therefore, in our current extensive review, we aimed to highlight the implication of FMT in various autoimmune diseases, such as inflammatory bowel disease, autoimmune thyroid and liver diseases, systemic lupus erythematosus, and type 1 diabetes mellitus, among others. Presenting all the aspects of FMT in more than 12 autoimmune diseases in one paper, to the best of our knowledge, is the first time presented in medical literature. Viewing FMT as such could contribute to better understanding and newer application of the model in the therapy of autoimmune diseases, indeed.
Collapse
Affiliation(s)
- Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Maisam Al Shawaf
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
26
|
Ma Y, Wei X, Peng J, Wei F, Wen Y, Liu M, Song B, Wang Y, Zhang Y, Peng T. Ephedra sinica polysaccharide regulate the anti-inflammatory immunity of intestinal microecology and bacterial metabolites in rheumatoid arthritis. Front Pharmacol 2024; 15:1414675. [PMID: 38846095 PMCID: PMC11153800 DOI: 10.3389/fphar.2024.1414675] [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: 04/09/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Ephedra sinica polysaccharide (ESP) exerts substantial therapeutic effects on rheumatoid arthritis (RA). However, the mechanism through which ESP intervenes in RA remains unclear. A close correlation has been observed between enzymes and derivatives in the gut microbiota and the inflammatory immune response in RA. Methods A type II collagen-induced arthritis (CIA) mice model was treated with Ephedra sinica polysaccharide. The therapeutic effect of ESP on collagen-induced arthritis mice was evaluated. The anti-inflammatory and cartilage-protective effects of ESP were also evaluated. Additionally, metagenomic sequencing was performed to identify changes in carbohydrate-active enzymes and resistance genes in the gut microbiota of the ESP-treated CIA mice. Liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry were performed to observe the levels of serum metabolites and short-chain fatty acids in the gut. Spearman's correlational analysis revealed a correlation among the gut microbiota, antibiotic-resistance genes, and microbiota-derived metabolites. Results ESP treatment significantly reduced inflammation levels and cartilage damage in the CIA mice. It also decreased the levels of pro-inflammatory cytokines interleukin (IL)-6, and IL-1-β and protected the intestinal mucosal epithelial barrier, inhibiting inflammatory cell infiltration and mucosal damage. Here, ESP reduced the TLR4, MyD88, and TRAF6 levels in the synovium, inhibited the p65 expression and pp65 phosphorylation in the NF-κB signaling pathway, and blocked histone deacetylase (HDAC1 and HDAC2) signals. ESP influenced the gut microbiota structure, microbial carbohydrate-active enzymes, and microbial resistance related to resistance genes. ESP increased the serum levels of L-tyrosine, sn-glycero-3-phosphocholine, octadecanoic acid, N-oleoyl taurine, and decreased N-palmitoyl taurine in the CIA mice. Conclusion ESP exhibited an inhibitory effect on RA. Its action mechanism may be related to the ability of ESP to effectively reduce pro-inflammatory cytokines levels, protect the intestinal barrier, and regulate the interaction between mucosal immune systems and abnormal local microbiota. Accordingly, immune homeostasis was maintained and the inhibition of fibroblast-like synoviocyte (FLS) proliferation through the HDAC/TLR4/NF-κB pathway was mediated, thereby contributing to its anti-inflammatory and immune-modulating effects.
Collapse
Affiliation(s)
- Yanmiao Ma
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiuhong Wei
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiehao Peng
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Fuxia Wei
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ya Wen
- Department of First Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Mingran Liu
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Bo Song
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yonghui Wang
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yumin Zhang
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Tao Peng
- Famous Chinese Medicine Studio, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan, China
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine Taiyuan, Taiyuan, China
| |
Collapse
|
27
|
Seymour BJ, Allen BE, Kuhn KA. Microbial Mechanisms of Rheumatoid Arthritis Pathogenesis. Curr Rheumatol Rep 2024; 26:124-132. [PMID: 38300467 PMCID: PMC11141067 DOI: 10.1007/s11926-024-01135-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE OF REVIEW Host-microbiome interactions have been implicated in the pathophysiology of rheumatoid arthritis (RA), but the data linking specific microbes to RA is largely associative. Here, we review recent studies that have interrogated specific mechanistic links between microbes and host in the setting of RA. RECENT FINDINGS Several candidate bacterial species and antigens that may trigger the conversion of an anti-bacterial to an autoimmune response have been recently identified. Additional studies have identified microbial metabolic pathways that are altered in RA. Some of these microbial species and metabolic pathways have been validated in mouse models to induce RA-like immune responses, providing initial evidence of specific mechanisms by which the microbiota contributes to the development of RA. Several microbial species, antigens, and metabolites have been identified as potential contributors to RA pathophysiology. Further interrogation and validation of these pathways may identify novel biomarkers of or therapeutic avenues for RA.
Collapse
Affiliation(s)
- Brenda J Seymour
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brendan E Allen
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
28
|
Pascual-García S, Martínez-Peinado P, Pujalte-Satorre C, Navarro-Sempere A, Esteve-Girbés J, López-Jaén AB, Javaloyes-Antón J, Cobo-Velacoracho R, Navarro-Blasco FJ, Sempere-Ortells JM. Exosomal Osteoclast-Derived miRNA in Rheumatoid Arthritis: From Their Pathogenesis in Bone Erosion to New Therapeutic Approaches. Int J Mol Sci 2024; 25:1506. [PMID: 38338785 PMCID: PMC10855630 DOI: 10.3390/ijms25031506] [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: 12/20/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that causes inflammation, pain, and ultimately, bone erosion of the joints. The causes of this disease are multifactorial, including genetic factors, such as the presence of the human leukocyte antigen (HLA)-DRB1*04 variant, alterations in the microbiota, or immune factors including increased cytotoxic T lymphocytes (CTLs), neutrophils, or elevated M1 macrophages which, taken together, produce high levels of pro-inflammatory cytokines. In this review, we focused on the function exerted by osteoclasts on osteoblasts and other osteoclasts by means of the release of exosomal microRNAs (miRNAs). Based on a thorough revision, we classified these molecules into three categories according to their function: osteoclast inhibitors (miR-23a, miR-29b, and miR-214), osteoblast inhibitors (miR-22-3p, miR-26a, miR-27a, miR-29a, miR-125b, and miR-146a), and osteoblast enhancers (miR-20a, miR-34a, miR-96, miR-106a, miR-142, miR-199a, miR-324, and miR-486b). Finally, we analyzed potential therapeutic targets of these exosomal miRNAs, such as the use of antagomiRs, blockmiRs, agomiRs and competitive endogenous RNAs (ceRNAs), which are already being tested in murine and ex vivo models of RA. These strategies might have an important role in reestablishing the regulation of osteoclast and osteoblast differentiation making progress in the development of personalized medicine.
Collapse
Affiliation(s)
- Sandra Pascual-García
- Department of Biotechnology, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | | | | | - Alicia Navarro-Sempere
- Department of Biotechnology, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | - Jorge Esteve-Girbés
- Department of Legal Studies of the State, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | - Ana B. López-Jaén
- Department of Biotechnology, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | - Juan Javaloyes-Antón
- Department of Physics, Systems Engineering and Signal Theory, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | - Raúl Cobo-Velacoracho
- Department of Biotechnology, University of Alicante, 03690 San Vicente del Raspeig, Spain
| | - Francisco J. Navarro-Blasco
- Department of Biotechnology, University of Alicante, 03690 San Vicente del Raspeig, Spain
- Rheumatology Unit, University General Hospital of Elche, 03203 Elche, Spain
| | | |
Collapse
|
29
|
Wu R, Wang D, Cheng L, Su R, Li B, Fan C, Gao C, Wang C. Impaired immune tolerance mediated by reduced Tfr cells in rheumatoid arthritis linked to gut microbiota dysbiosis and altered metabolites. Arthritis Res Ther 2024; 26:21. [PMID: 38218985 PMCID: PMC10787489 DOI: 10.1186/s13075-023-03260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) showed impaired immune tolerance characterized by reduced follicular regulatory T (Tfr) cells, and they also exhibited altered gut microbiotas and their metabolites in RA. However, the association of gut microbiotas and their metabolites with the immune tolerance mediated by Tfr cells in RA remains unclear. METHODS Peripheral blood and stool samples were collected from 32 new-onset RA patients and 17 healthy controls (HCs) in the Second Hospital of Shanxi Medical University between January 2022 and June 2022. The peripheral blood was used to detect the circulating regulatory T (Treg), helper T(Th)17, Tfr, and follicular helper T (Tfh) cells by modified flow cytometry. The stool samples were used to analyze the gut microbiotas and their metabolites via 16S rDNA sequencing and metabolomic profiling. We aimed to characterize the gut microbiotas and their metabolites in RA and identified their association with Tfr cell-mediated immune tolerance. RESULTS The new-onset RA demonstrated reduced Treg and Tfr cells, associated with the disease activity and autoantibodies. There were significant differences in gut microbiotas between the two groups as the results of β diversity analysis (P = 0.039) including 21 differential gut microbiotas from the phylum to genus levels. In which, Ruminococcus 2 was associated with the disease activity and autoantibodies of RA, and it was identified as the potential biomarker of RA [area under curve (AUC) = 0.782, 95% confidence interval (CI) = 0.636-0.929, P = 0.001]. Eleven differential metabolites were identified and participated in four main pathways related to RA. Arachidonic acid might be the potential biomarker of RA (AUC = 0.724, 95% CI = 0.595-0.909, P = 0.038), and it was the core metabolite as the positive association with six gut microbiotas enriched in RA. The reduced Tfr cells were associated with the altered gut microbiotas and their metabolites including the Ruminococcus 2, the arachidonic acid involved in the biosynthesis of unsaturated fatty acid pathway and the 3-methyldioxyindole involved in the tryptophan metabolism pathway. CONCLUSION The breakdown of immune tolerance mediated by reduced Tfr cells was associated with the altered gut microbiotas and their metabolites implying the possible mechanism of RA pathogenesis from the perspective of microecology-metabolism-immune.
Collapse
Affiliation(s)
- Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Dongming Wang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Liyun Cheng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chunxue Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
30
|
Seymour BJ, Trent B, Allen BE, Berlinberg AJ, Tangchittsumran J, Jubair WK, Chriswell ME, Liu S, Ornelas A, Stahly A, Alexeev EE, Dowdell AS, Sneed SL, Fechtner S, Kofonow JM, Robertson CE, Dillon SM, Wilson CC, Anthony RM, Frank DN, Colgan SP, Kuhn KA. Microbiota-dependent indole production stimulates the development of collagen-induced arthritis in mice. J Clin Invest 2023; 134:e167671. [PMID: 38113112 PMCID: PMC10866668 DOI: 10.1172/jci167671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Altered tryptophan catabolism has been identified in inflammatory diseases like rheumatoid arthritis (RA) and spondyloarthritis (SpA), but the causal mechanisms linking tryptophan metabolites to disease are unknown. Using the collagen-induced arthritis (CIA) model, we identified alterations in tryptophan metabolism, and specifically indole, that correlated with disease. We demonstrated that both bacteria and dietary tryptophan were required for disease and that indole supplementation was sufficient to induce disease in their absence. When mice with CIA on a low-tryptophan diet were supplemented with indole, we observed significant increases in serum IL-6, TNF, and IL-1β; splenic RORγt+CD4+ T cells and ex vivo collagen-stimulated IL-17 production; and a pattern of anti-collagen antibody isotype switching and glycosylation that corresponded with increased complement fixation. IL-23 neutralization reduced disease severity in indole-induced CIA. Finally, exposure of human colonic lymphocytes to indole increased the expression of genes involved in IL-17 signaling and plasma cell activation. Altogether, we propose a mechanism by which intestinal dysbiosis during inflammatory arthritis results in altered tryptophan catabolism, leading to indole stimulation of arthritis development. Blockade of indole generation may present a unique therapeutic pathway for RA and SpA.
Collapse
Affiliation(s)
| | - Brandon Trent
- Division of Rheumatology, Department of Medicine, and
| | | | | | | | | | | | - Sucai Liu
- Division of Rheumatology, Department of Medicine, and
| | - Alfredo Ornelas
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, and
| | - Erica E. Alexeev
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alexander S. Dowdell
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sunny L. Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jennifer M. Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charles E. Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Stephanie M. Dillon
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cara C. Wilson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Robert M. Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
31
|
Liu Q, Shen J, Wang J, Xia J, Yin J, Cheng G, Qian X, Jiang Y, Ge X, Wang Q. PR-957 retards rheumatoid arthritis progression and inflammation by inhibiting LMP7-mediated CD4 + T cell imbalance. Int Immunopharmacol 2023; 124:110860. [PMID: 37716163 DOI: 10.1016/j.intimp.2023.110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Low molecular mass polypeptide 7 (LMP7) is an immunoproteasome subunit that regulates T cell amplification, differentiation, and inflammation and is involved in rheumatoid arthritis (RA) progression. This study intended to apply PR-957 (an anti-LMP7 agent) for RA treatment in vitro and in vivo and evaluate its interaction with LMP7-mediated CD4+ T cell imbalance. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from 30 RA patients and 30 healthy controls. RA fibroblast-like synoviocytes (RA-FLSs) and CD4+ T cells were isolated from RA patients and then cocultured with PR-957 and/or LMP7 overexpression adenovirus (Ad-LMP7). Collagen-induced arthritis (CIA) mice were constructed and then treated with PR-957 and/or Ad-LMP7. RESULTS LMP7 was higher in RA patients (versus healthy controls) and positively correlated with T helper (Th)1 cells, the Th1/Th2 ratio, Th17 cells, and the Th17/Treg ratio but not with Th2 or T regulatory (Treg) cells. PR-957 reduced Th1 and Th17 cells but increased Th2 and Treg cells in RA-CD4+ T cells, and this effect was partially reversed by Ad-LMP7 transfection. Interestingly, when cocultured with RA-CD4+ T cells, PR-957 increased RA-FLS apoptosis and decreased its invasive ability, viability, and inflammation, as suggested by IL-6, CCL2, MMP1, and MMP3; however, these phenomena were weakened in RA-FLSs without RA-CD4+ T cell coculture. In addition, Ad-LMP7 transfection attenuated the above effects of PR-957. In CIA mice, PR-957 decreased the arthritis score, synovial hyperproliferation and articular injury, inflammation in the synovium and serum, and the imbalance of Th1/Th2 and Th17/Treg in the spleen, and these effects were attenuated by Ad-LMP7. CONCLUSION PR-957 ameliorates RA progression and inflammation by repressing LMP7-mediated CD4+ T cell imbalance.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Jin Shen
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Jian Wang
- Department of Joint Surgery, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Jinjun Xia
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Jian Yin
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Guowei Cheng
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Ximing Qian
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Yun Jiang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China.
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China.
| |
Collapse
|
32
|
Seymour BJ, Trent B, Allen B, Berlinberg AJ, Tangchittsumran J, Jubair WK, Chriswell ME, Liu S, Ornelas A, Stahly A, Alexeev EE, Dowdell AS, Sneed SL, Fechtner S, Kofonow JM, Robertson CE, Dillon SM, Wilson CC, Anthony RM, Frank DN, Colgan SP, Kuhn KA. Microbiota-dependent indole production is required for the development of collagen-induced arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.561693. [PMID: 37873395 PMCID: PMC10592798 DOI: 10.1101/2023.10.13.561693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Altered tryptophan catabolism has been identified in inflammatory diseases like rheumatoid arthritis (RA) and spondyloarthritis (SpA), but the causal mechanisms linking tryptophan metabolites to disease are unknown. Using the collagen-induced arthritis (CIA) model we identify alterations in tryptophan metabolism, and specifically indole, that correlate with disease. We demonstrate that both bacteria and dietary tryptophan are required for disease, and indole supplementation is sufficient to induce disease in their absence. When mice with CIA on a low-tryptophan diet were supplemented with indole, we observed significant increases in serum IL-6, TNF, and IL-1β; splenic RORγt+CD4+ T cells and ex vivo collagen-stimulated IL-17 production; and a pattern of anti-collagen antibody isotype switching and glycosylation that corresponded with increased complement fixation. IL-23 neutralization reduced disease severity in indole-induced CIA. Finally, exposure of human colon lymphocytes to indole increased expression of genes involved in IL-17 signaling and plasma cell activation. Altogether, we propose a mechanism by which intestinal dysbiosis during inflammatory arthritis results in altered tryptophan catabolism, leading to indole stimulation of arthritis development. Blockade of indole generation may present a novel therapeutic pathway for RA and SpA.
Collapse
Affiliation(s)
- Brenda J. Seymour
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon Trent
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brendan Allen
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Adam J. Berlinberg
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jimmy Tangchittsumran
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Widian K. Jubair
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alfredo Ornelas
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erica E. Alexeev
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander S. Dowdell
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sunny L. Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sabrina Fechtner
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer M. Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles E. Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie M. Dillon
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cara C. Wilson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert M. Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
33
|
Berryman MA, Ilonen J, Triplett EW, Ludvigsson J. Important denominator between autoimmune comorbidities: a review of class II HLA, autoimmune disease, and the gut. Front Immunol 2023; 14:1270488. [PMID: 37828987 PMCID: PMC10566625 DOI: 10.3389/fimmu.2023.1270488] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Human leukocyte antigen (HLA) genes are associated with more diseases than any other region of the genome. Highly polymorphic HLA genes produce variable haplotypes that are specifically correlated with pathogenically different autoimmunities. Despite differing etiologies, however, many autoimmune disorders share the same risk-associated HLA haplotypes often resulting in comorbidity. This shared risk remains an unanswered question in the field. Yet, several groups have revealed links between gut microbial community composition and autoimmune diseases. Autoimmunity is frequently associated with dysbiosis, resulting in loss of barrier function and permeability of tight junctions, which increases HLA class II expression levels and thus further influences the composition of the gut microbiome. However, autoimmune-risk-associated HLA haplotypes are connected to gut dysbiosis long before autoimmunity even begins. This review evaluates current research on the HLA-microbiome-autoimmunity triplex and proposes that pre-autoimmune bacterial dysbiosis in the gut is an important determinant between autoimmune comorbidities with systemic inflammation as a common denominator.
Collapse
Affiliation(s)
- Meghan A. Berryman
- Triplett Laboratory, Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eric W. Triplett
- Triplett Laboratory, Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Johnny Ludvigsson
- Crown Princess Victoria’s Children’s Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
34
|
Berthelot JM, Lioté F, Sibilia J. Methotrexate also improves rheumatoid arthritis through correction of microbiota dysbiosis. Joint Bone Spine 2023; 90:105602. [PMID: 37352969 DOI: 10.1016/j.jbspin.2023.105602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Department, Nantes University Hospital, Hôtel-Dieu, place Alexis-Ricordeau, 44093 Nantes cedex 01, France.
| | - Frédéric Lioté
- Rheumatology Department & Inserm UMR 1132 (Centre Viggo Petersen), hôpital Lariboisière, 2, rue Ambroise-Paré, 75010 Paris, France; Université Paris Cité, faculté de Santé, UFR de médecine, 75010 Paris, France
| | - Jean Sibilia
- Service de rhumatologie, hôpitaux universitaires de Strasbourg, 67000 Strasbourg, France; RESO: Centre de référence des maladies autoimmunes systémiques rares Est Sud-Ouest, Inserm UMR_S1109, université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|