1
|
Ziyadullaev SK, Khudaiberdiev SS, Aripova TU, Chirumbolo S, Kamalov ZS, Bjørklund G, Rizaev JA, Tashkenbaeva EN, Khamidov OA, Gaffarov UB. Synovial Fluid as a Crucial Component of the Joint Microenvironment in Rheumatoid Arthritis. Immune Netw 2025; 25:e2. [PMID: 40342839 PMCID: PMC12056296 DOI: 10.4110/in.2025.25.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 05/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease closely associated with synovial tissue proliferation, pannus formation in small joints such as the hands, wrists, and feet, cartilage destruction, and systemic complications such as pulmonary, cardiovascular, neurological, and skeletal muscle lesions, glucocorticoid-induced osteoporosis and infections. The importance of confirming the diagnosis and determining local activity is given to the study of synovial fluid. A deep understanding of the pathological process in the joint in RA, characterized by changes in autoreactive CD4+ T cells, B cells, macrophages, inflammatory cytokines, chemokines, and autoantibodies, has now been achieved, although much remains to be explored. This article provides an updated overview of the pathogenesis of RA, revealing even more therapeutic targets for the intra-articular pathological process.
Collapse
Affiliation(s)
| | | | | | | | | | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana 8622, Norway
| | | | | | - Obid Abdurakhmanovich Khamidov
- Rehabilitology and Sports Medicine Research Institute of the Samarkand State Medical University, Samarkand 140100, Uzbekistan
| | | |
Collapse
|
2
|
Neto M, Mendes B, Albuquerque F, da Silva JAP. Novel biomarkers in RA: Implication for diagnosis, prognosis, and personalised treatment. Best Pract Res Clin Rheumatol 2025; 39:102021. [PMID: 39550250 DOI: 10.1016/j.berh.2024.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
Over the past decades our understanding of rheumatoid arthritis (RA) pathogenesis has improved remarkably and major breakthroughs in the treatment of RA were made with the advent of numerous targeted therapies and new treatment strategies. Despite these advances, several unmet needs remain, namely in achieving earlier and more accurate diagnosis, monitoring disease activity, predicting disease prognosis and optimizing treatment. To address these gaps, recent research has focused on identifying biomarkers that may enhance diagnostic precision, predict disease prognosis, and optimize treatment strategies. In this narrative review we will describe recent developments in RA biomarkers with demonstrated or promising clinical relevance.
Collapse
Affiliation(s)
- Marcelo Neto
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal.
| | - Beatriz Mendes
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal.
| | | | - José António P da Silva
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal.
| |
Collapse
|
3
|
Peng W, Merlo LMF, Grabler S, Montgomery JD, Mandik-Nayak L. IDO2 Drives Autoantibody Production and Joint Inflammation in a Preclinical Model of Arthritis by Repressing Runx1 Function in B Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1595-1604. [PMID: 39400244 DOI: 10.4049/jimmunol.2400445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
The immunomodulatory enzyme IDO2 is an essential mediator of autoantibody production and joint inflammation in preclinical models of autoimmune arthritis. Although originally identified as a tryptophan-catabolizing enzyme, we recently discovered a previously unknown nonenzymatic pathway is essential for the proarthritic function of IDO2. We subsequently identified Runx1 (Runt-related transcription factor 1) as a potential component of the nonenzymatic pathway IDO2 uses to drive arthritis. In this study, we find that IDO2 directly binds Runx1 and inhibits its localization to the nucleus, implicating Runx1 as a downstream component of IDO2 function. To directly test whether Runx1 mediates the downstream pathway driving B cell activation in arthritis, we bred B cell conditional Runx1-deficient (CD19cre Runx1flox/flox) mice onto the KRN.g7 arthritis model in the presence or absence of IDO2. Runx1 loss did not affect arthritis in the presence of IDO2; however, deleting Runx1 reversed the antiarthritic effect of IDO2 loss in this model. Further studies demonstrated that the IDO2-Runx1 interaction could be blocked with a therapeutic anti-IDO2 mAb in vitro and that Runx1 was required for IDO2 Ig's therapeutic effect in vivo. Taken together, these data demonstrate that IDO2 mediates autoantibody production and joint inflammation by acting as a repressor of Runx1 function in B cells and implicate therapeutic targeting of IDO2-Runx1 binding as a strategy to inhibit autoimmune arthritis and other autoantibody-mediated diseases.
Collapse
Affiliation(s)
- Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA
| | | | | | | | | |
Collapse
|
4
|
Ohara D, Takeuchi Y, Hirota K. Type 17 immunity: novel insights into intestinal homeostasis and autoimmune pathogenesis driven by gut-primed T cells. Cell Mol Immunol 2024; 21:1183-1200. [PMID: 39379604 PMCID: PMC11528014 DOI: 10.1038/s41423-024-01218-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
The IL-23 signaling pathway in both innate and adaptive immune cells is vital for orchestrating type 17 immunity, which is marked by the secretion of signature cytokines such as IL-17, IL-22, and GM-CSF. These proinflammatory mediators play indispensable roles in maintaining intestinal immune equilibrium and mucosal host defense; however, their involvement has also been implicated in the pathogenesis of chronic inflammatory disorders, such as inflammatory bowel diseases and autoimmunity. However, the implications of type 17 immunity across diverse inflammation models are complex. This review provides a comprehensive overview of the multifaceted roles of these cytokines in maintaining gut homeostasis and in perturbing gut barrier integrity, leading to acute and chronic inflammation in various models of gut infection and colitis. Additionally, this review focuses on type 17 immunity interconnecting multiple organs in autoimmune conditions, with a particular emphasis on the pathogenesis of autoimmune arthritis and neuroinflammation driven by T cells primed within the gut microenvironment.
Collapse
Affiliation(s)
- Daiya Ohara
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- ImmunoSensation Cluster of Excellence, University of Bonn, Bonn, Germany.
| |
Collapse
|
5
|
Nishiyama T, Ohyama A, Miki H, Asashima H, Kondo Y, Tsuboi H, Ohno H, Shimano H, Matsumoto I. Mechanisms of age-related Treg dysfunction in an arthritic environment. Clin Immunol 2024; 266:110337. [PMID: 39111562 DOI: 10.1016/j.clim.2024.110337] [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: 04/24/2024] [Revised: 07/18/2024] [Accepted: 08/01/2024] [Indexed: 08/11/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by a polyarticular synovitis. In recent years, elderly onset rheumatoid arthritis (EORA) has been increasing. Treg cells in RA have been reported to be dysfunctional, but the relationship between aging and their functional changes is unclear. Here, we found that Treg cells from EORA patients had increased percentages, but decreased activity compared to those from younger onset RA (YORA) patients. In experiments using arthritis model mice, decreased suppressive function and oxygen consumption rate (OCR) were observed in Treg cells only from old arthritic mice. Furthermore, type I interferon (IFN) signaling was upregulated in Treg cells from old GIA mice, and IFN-β decreased the suppressive function of Treg cells. Our findings demonstrate that increased type I IFN signaling in old Treg cells is induced only in the arthritic environment and relates to decreased suppressive function of Treg cells, gets involved in EORA.
Collapse
Affiliation(s)
- Taihei Nishiyama
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ayako Ohyama
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruka Miki
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiromitsu Asashima
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuya Kondo
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroto Tsuboi
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ohno
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Isao Matsumoto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
6
|
Xu J, He C, Cai Y, Wang X, Yan J, Zhang J, Zhang F, Urbonaviciute V, Cheng Y, Lu S, Holmdahl R. NCF4 regulates antigen presentation of cysteine peptides by intracellular oxidative response and restricts activation of autoreactive and arthritogenic T cells. Redox Biol 2024; 72:103132. [PMID: 38547647 PMCID: PMC11096609 DOI: 10.1016/j.redox.2024.103132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Autoimmune diseases, such as rheumatoid arthritis (RA) and systemic lupus erythematous, are regulated by polymorphisms in genes contributing to the NOX2 complex. Mutations in both Ncf1 and Ncf4 affect development of arthritis in experimental models of RA, but the different regulatory pathways mediated by NOX2-derived reactive oxygen species (ROS) have not yet been clarified. Here we address the possibility that intracellular ROS, regulated by the NCF4 protein (earlier often denoted p40phox) which interacts with endosomal membranes, could play an important role in the oxidation of cysteine peptides in mononuclear phagocytic cells, thereby regulating antigen presentation and activation of arthritogenic T cells. To study the role of NCF4 we used mice with an amino acid replacing mutation (NCF4R58A), which is known to affect interaction with endosomal membranes, leading to decreased intracellular ROS production. To study the impact of NCF4 on T cell activation, we used the glucose phosphate isomerase peptide GPI325-339, which contains two cysteine residues (325-339c-c). Macrophages from mice with the NCF458A mutation efficiently presented the peptide when the two cysteines were intact and not crosslinked, leading to a strong arthritogenic T cell response. T cell priming occurred in the draining lymph nodes (LNs) within 8 days after immunization. Clodronate treatment, which depletes antigen-presenting mononuclear phagocytes, ameliorated arthritis severity, whereas treatment with FYT720, which traps activated T cells in LNs, prohibited arthritis. We conclude that NCF4-dependent intracellular ROS maintains cysteine peptides in an oxidized crosslinked state, which prevents presentation of peptides recognized by non-tolerized T cells and thereby protects against autoimmune arthritis.
Collapse
Affiliation(s)
- Jing Xu
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, and Department of Rheumatology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China; Medical Inflammation Research, Division of Immunology, Dept. of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Chang He
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China; Medical Inflammation Research, Division of Immunology, Dept. of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Cardiology, The Second Affiliated Hospital, Zhejiang University Schoole of Medicine, Zhejiang, Hangzhou, PR China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China
| | - Xipeng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Jidong Yan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, PR China
| | - Jing Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Vilma Urbonaviciute
- Medical Inflammation Research, Division of Immunology, Dept. of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Yuanyuan Cheng
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, and Department of Rheumatology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Rikard Holmdahl
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, and Department of Rheumatology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710061, PR China; Medical Inflammation Research, Division of Immunology, Dept. of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
7
|
Escal J, Neel T, Hodin S, Boussoualim K, Amouzougan A, Coassy A, Locrelle H, Thomas T, Delavenne X, Marotte H. Proteomics analyses of human plasma reveal triosephosphate isomerase as a potential blood marker of methotrexate resistance in rheumatoid arthritis. Rheumatology (Oxford) 2024; 63:1368-1376. [PMID: 37527020 DOI: 10.1093/rheumatology/kead390] [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: 04/26/2023] [Revised: 07/02/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023] Open
Abstract
OBJECTIVE The objective of this study was to assess differentially expressed blood proteins between patients with active RA and patients in remission after MTX treatment, with the aim of identifying a biomarker of MTX resistance (MTXR). METHODS Two populations of RA patients treated with a stable dose of s.c. MTX for at least 3 months were constituted according to the DAS28: remission (DAS28 < 2.6; n = 24) and active disease (DAS28 > 3.2; n = 32). The two groups of RA patients were homogeneous regarding their epidemiological characteristics, except for the duration of treatment, which was longer in the remission group. After collection of a blood sample, plasma protein digestion was performed, followed by untargeted proteomics analysis. Then, a targeted analysis was performed to confirm the results of the untargeted approach. RESULTS Untargeted proteomics analysis revealed eight plasma proteins that were differentially expressed between the two groups of patients. Among them, triosephosphate isomerase (TPI-1) and glucose-6-phosphate isomerase (GPI), which are main actors in glycolysis, were found down-regulated in the active group. This result was confirmed for TPI-1 in the targeted proteomics analysis. CONCLUSION A first step was achieved in the search for biomarkers of MTXR, with the identification of two actors in glycolysis (TPI-1 and GPI). The next step will be to confirm these results in a larger cohort, including samples from treatment-naive patients, to assess the predictive potential of these protein markers.
Collapse
Affiliation(s)
- Jean Escal
- Université Jean Monnet Saint-Étienne, CHU Saint-Etienne, Laboratoire de Pharmacologie et Toxicologie, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Tiphany Neel
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Sophie Hodin
- Université Jean Monnet Saint-Étienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | | | | | - Astrid Coassy
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Hervé Locrelle
- CHU Saint-Étienne, Service de Rhumatologie, Saint-Etienne, France
| | - Thierry Thomas
- Université Jean Monnet Saint-Étienne, CHU Saint-Étienne, Service de Rhumatologie, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Xavier Delavenne
- Université Jean Monnet Saint-Étienne, CHU Saint-Etienne, Laboratoire de Pharmacologie et Toxicologie, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Hubert Marotte
- Université Jean Monnet Saint-Étienne, CHU Saint-Étienne, Service de Rhumatologie, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| |
Collapse
|
8
|
Osinski V, Yellamilli A, Firulyova MM, Zhang MJ, Peck A, Auger JL, Faragher JL, Marath A, Voeller RK, O’Connell TD, Zaitsev K, Binstadt BA. Profibrotic VEGFR3-Dependent Lymphatic Vessel Growth in Autoimmune Valvular Carditis. Arterioscler Thromb Vasc Biol 2024; 44:807-821. [PMID: 38269589 PMCID: PMC10978259 DOI: 10.1161/atvbaha.123.320326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.
Collapse
Affiliation(s)
- Victoria Osinski
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Amritha Yellamilli
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alyssa Peck
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jennifer L. Auger
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jessica L. Faragher
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | | | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Bryce A. Binstadt
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
9
|
Kaieda S, Kinoshita T, Chiba A, Miyake S, Hoshino T. IL-18 receptor-α signalling pathway contributes to autoantibody-induced arthritis via neutrophil recruitment and mast cell activation. Mod Rheumatol 2024; 34:500-508. [PMID: 37285315 DOI: 10.1093/mr/road043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/08/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVES The interleukin (IL)-18 signalling pathway is involved in animal models of collagen-induced arthritis, but the role of this pathway in autoantibody-induced arthritis is poorly understood. An autoantibody-induced arthritis model, K/BxN serum transfer arthritis, reflects the effector phase of arthritis and is important in innate immunity including neutrophils and mast cells. This study aimed to investigate the role of the IL-18 signalling pathway in autoantibody-induced arthritis using IL-18 receptor (IL-18R) α-deficient mice. METHODS K/BxN serum transfer arthritis was induced in IL-18Rα-/- and wild-type B6 (controls) mice. The severity of arthritis was graded, and histological and immunohistochemical examinations were performed on paraffin-embedded ankle sections. Total Ribonucleic acid (RNA) isolated from mouse ankle joints was analysed by real-time reverse transcriptase-polymerase chain reaction. RESULTS IL-18 Rα-/- mice had significantly lower arthritis clinical scores, neutrophil infiltration, and numbers of activated, degranulated mast cells in the arthritic synovium than in controls. IL-1β, which is indispensable for the progression of arthritis, was significantly downregulated in inflamed ankle tissue in IL-18 Rα-/- mice. CONCLUSIONS IL-18/IL-18Rα signalling contributes to the development of autoantibody-induced arthritis by enhancing synovial tissue expression of IL-1β and inducing neutrophil recruitment and mast cell activation. Therefore, inhibition of the IL-18Rα signalling pathway might be a new therapeutic strategy for rheumatoid arthritis.
Collapse
Affiliation(s)
- Shinjiro Kaieda
- Department of Medicine, Division of Respirology, Neurology and Rheumatology, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Kinoshita
- Department of Medicine, Division of Respirology, Neurology and Rheumatology, Kurume University School of Medicine, Kurume, Japan
| | - Asako Chiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoaki Hoshino
- Department of Medicine, Division of Respirology, Neurology and Rheumatology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
10
|
Iwai T, Ohyama A, Osada A, Nishiyama T, Shimizu M, Miki H, Asashima H, Kondo Y, Tsuboi H, Mizuno S, Takahashi S, Ishigami A, Matsumoto I. Role of inter-alpha-trypsin inhibitor heavy chain 4 and its citrullinated form in experimental arthritis murine models. Clin Exp Immunol 2024; 215:302-312. [PMID: 38190323 PMCID: PMC10876112 DOI: 10.1093/cei/uxae001] [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: 05/25/2023] [Revised: 11/12/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Inter-α-trypsin inhibitor heavy chain 4 (ITIH4) is a major protein in serum and reported to be upregulated at the onset of rheumatoid arthritis (RA). Its citrullinated form, cit-ITIH4, is specifically found in the serum and synovial fluid of patients with RA. However, the detailed function of ITIH4 in arthritis remains unknown. The aim of this study was to clarify the role of ITIH4 and cit-ITIH4 using experimental arthritis models. ITIH4 and cit-ITIH4 expression was examined in steady-state mice and two different arthritis models, and their pathological effects were examined in Itih4-deficient mice. In naïve C57BL/6 (WT) mice, ITIH4 was expressed as mRNA in the liver and the lung and was expressed as protein in serum and hepatocytes. In K/BxN serum transferred arthritis (K/BxN-STA) and collagen-induced arthritis (CIA), ITIH4 and cit-ITIH4 in sera were increased before the onset of arthritis, and cit-ITIH4 was further increased at the peak of arthritis. In Itih4-deficient mice, citrullinated proteins in serum and joints, especially 120 kDa protein, were clearly diminished; however, there was no significant difference in arthritis severity between WT and itih-/- mice either in the K/BxN-STA or CIA model. CIA mice also exhibited pulmonary lesions and itih4-/- mice tended to show enhanced inflammatory cell aggregation compared to WT mice. Neutrophils in the lungs of itih4-/- mice were significantly increased compared to WT mice. In summary, ITIH4 itself did not alter the severity of arthritis but may inhibit autoimmune inflammation via suppression of neutrophil recruitment.
Collapse
Affiliation(s)
- Tamaki Iwai
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ayako Ohyama
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsumu Osada
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Taihei Nishiyama
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaru Shimizu
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruka Miki
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiromitsu Asashima
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuya Kondo
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroto Tsuboi
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Seiya Mizuno
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Japan
| | - Isao Matsumoto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
11
|
Titcombe PJ, Silva Morales M, Zhang N, Mueller DL. BATF represses BIM to sustain tolerant T cells in the periphery. J Exp Med 2023; 220:e20230183. [PMID: 37862030 PMCID: PMC10588758 DOI: 10.1084/jem.20230183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/13/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
T cells that encounter self-antigens after exiting the thymus avert autoimmunity through peripheral tolerance. Pathways for this include an unresponsive state known as anergy, clonal deletion, and T regulatory (Treg) cell induction. The transcription factor cues and kinetics that guide distinct peripheral tolerance outcomes remain unclear. Here, we found that anergic T cells are epigenetically primed for regulation by the non-classical AP-1 family member BATF. Tolerized BATF-deficient CD4+ T cells were resistant to anergy induction and instead underwent clonal deletion due to proapoptotic BIM (Bcl2l11) upregulation. During prolonged antigen exposure, BIM derepression resulted in fewer PD-1+ conventional T cells as well as loss of peripherally induced FOXP3+ Treg cells. Simultaneous Batf and Bcl2l11 knockdown meanwhile restored anergic T cell survival and Treg cell maintenance. The data identify the AP-1 nuclear factor BATF as a dominant driver of sustained T cell anergy and illustrate a mechanism for divergent peripheral tolerance fates.
Collapse
Affiliation(s)
- Philip J. Titcombe
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Milagros Silva Morales
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Na Zhang
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Daniel L. Mueller
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
12
|
Rani R, Raina N, Sharma A, Kumar P, Tulli HS, Gupta M. Advancement in nanotechnology for treatment of rheumatoid arthritis: scope and potential applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2287-2310. [PMID: 37166463 DOI: 10.1007/s00210-023-02514-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Rheumatoid arthritis is a hyperactive immune disorder that results in severe inflammation in synovial joints, cartilage, and bone deterioration, resulting in immobilization of joints. Traditional approaches for the treatment of rheumatoid arthritis are associated with some limiting factors such as suboptimal patient compliance, inability to control the progression of disorder, and safety concerns. Therefore, innovative drug delivery carriers for efficient therapeutic delivery at inflamed synovial sites with better safety assessment are urgently needed to address these issues. From this perspective, nanotechnology is an outstanding alternative to traditional drug delivery approaches, and it has shown great promise in developing novel carriers to treat rheumatoid arthritis. Considering the current research and future application of nanocarriers, it is believed that nanocarriers can be a crucial element in rheumatoid arthritis treatment. This paper covers all currently available pathophysiological aspects of rheumatoid arthritis and treatment options. Future research for the reduction of synovial inflammation should focus on developing multifunction nanoparticles capable of delivering therapeutic agents with improved safety, efficacy, and cost-effectiveness to be commercialized.
Collapse
Affiliation(s)
- Radha Rani
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Neha Raina
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Ajay Sharma
- Institute of Nuclear Medicine & Allied Sciences (INMAS-DRDO), Ministry of Defence, Brig. SK Mazumdar Marg, Lucknow Road, Timarpur, Delhi-110054, India
| | - Pramod Kumar
- Institute of Lung Health and Immunity, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Hardeep Singh Tulli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, 133207, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
13
|
Lee S, Choi E, Chae S, Koh JH, Choi Y, Kim JG, Yoo SA, Hwang D, Kim WU. Identification of MYH9 as a key regulator for synoviocyte migration and invasion through secretome profiling. Ann Rheum Dis 2023; 82:1035-1048. [PMID: 37188496 PMCID: PMC10359537 DOI: 10.1136/ard-2022-223625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/30/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVES 'Invasive pannus' is a pathological hallmark of rheumatoid arthritis (RA). This study aimed to investigate secretome profile of synovial fibroblasts of patients with RA (RA-FLSs), a major cell type comprising the invasive pannus. METHODS Secreted proteins from RA-FLSs were first identified using liquid chromatography-tandem mass spectrometry analysis. Ultrasonography was performed for affected joints to define synovitis severity at the time of arthrocentesis. Expression levels of myosin heavy chain 9 (MYH9) in RA-FLSs and synovial tissues were determined by ELISA, western blot analysis and immunostaining. A humanised synovitis model was induced in immuno-deficient mice. RESULTS We first identified 843 proteins secreted from RA-FLSs; 48.5% of the secretome was associated with pannus-driven pathologies. Parallel reaction monitoring analysis of the secretome facilitated discovery of 16 key proteins related to 'invasive pannus', including MYH9, in the synovial fluids, which represented synovial pathology based on ultrasonography and inflammatory activity in the joints. Particularly, MYH9, a key protein in actin-based cell motility, showed a strong correlation with fibroblastic activity in the transcriptome profile of RA synovia. Moreover, MYH9 expression was elevated in cultured RA-FLSs and RA synovium, and its secretion was induced by interleukin-1β, tumour necrosis factor α, toll-like receptor ligation and endoplasmic reticulum stimuli. Functional experiments demonstrated that MYH9 promoted migration and invasion of RA-FLSs in vitro and in a humanised synovitis model, which was substantially inhibited by blebbistatin, a specific MYH9 inhibitor. CONCLUSIONS This study provides a comprehensive resource of the RA-FLS-derived secretome and suggests that MYH9 represents a promising target for retarding abnormal migration and invasion of RA-FLSs.
Collapse
Affiliation(s)
- Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
| | - Eunbyeol Choi
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, The Republic of Korea
| | - Sehyun Chae
- Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, The Republic of Korea
| | - Jung Hee Koh
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
- Department of Internal Medicine, The Catholic University of Korea, School of Medicine, Seoul, The Republic of Korea
| | - Yoolim Choi
- Department of Biological Sciences, Seoul National University, Seoul, The Republic of Korea
| | - Jung Gon Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, The Republic of Korea
| | - Seung-Ah Yoo
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
- Department of Medical Life Sciences, The Catholic University of Korea, Seoul, The Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, The Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, The Republic of Korea
- Department of Internal Medicine, The Catholic University of Korea, School of Medicine, Seoul, The Republic of Korea
| |
Collapse
|
14
|
Zong M, Cheng Y, Ye B, Chen S, Yu S, Ding M, Lu L, Fan L. Preparation of mesoporous silica nanocarriers targeting glucose-6-phosphate isomerase inhibition and application in the treatment of rheumatoid arthritis. Clin Exp Immunol 2023; 211:46-56. [PMID: 36610698 PMCID: PMC9993455 DOI: 10.1093/cei/uxac116] [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: 05/26/2022] [Revised: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Glucose 6-phosphate isomerase (G6PI) is an indicator to assist in diagnosis of rheumatoid arthritis (RA) and monitor the disease. It also plays a key role in proliferating RA synovial tissues, pannus formation, and invasion and destruction of articular cartilage. In this study, we synthesized nanoparticles targeting G6PI (siG6PI-MSN) using mesoporous silica nanocarriers (MSN) and small interfering RNA (siRNA), followed by identifying the characteristics and functions, and preliminarily exploring their application in the treatment of RA in vivo with a type II collagen-induced arthritis (CIA) rat model. It showed that the synthetic functionalized carrier had a regular pore structure and a specific volume and surface area. No obvious hemolysis or toxicity of the carrier was found when its concentration was below 100 µg/ml. Cytological results in vitro suggested that siG6PI-MSN significantly inhibited G6PI expression and reduced the ability of proliferation, migration, and invasion of FLSs, compared with the siNC-MSN group. In vivo results in the CIA rat model showed that the arthritis index and degree of joint swelling among rats in the siG6PI-MSN-treatment group were significantly lower than those in the control group. Moreover, the number of FLSs in Synovium and the levels of TNF α and IL-1 β were also significantly decreased in the siG6PI-MSN group. Histopathology of the synovial tissue and cartilage revealed siG6PI-MSN treatment significantly reduced the pathological manifestations of arthritis. In conclusion, siG6PI-MSN effectively suppresses the proliferation and invasive growth of synovial tissue and improve joint swelling and inflammatory infiltration, thereby preventing joint damage in RA. This carrier may be a new therapeutic measure for RA, with potential social and economic benefits.
Collapse
Affiliation(s)
- Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Yu Cheng
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Bei Ye
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Saige Chen
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Shanshan Yu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Menglei Ding
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Liu Lu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| |
Collapse
|
15
|
Giovannini D, Belbezier A, Baillet A, Bouillet L, Kawano M, Dumestre-Perard C, Clavarino G, Noble J, Pers JO, Sturm N, Huard B. Heterogeneity of antibody-secreting cells infiltrating autoimmune tissues. Front Immunol 2023; 14:1111366. [PMID: 36895558 PMCID: PMC9989216 DOI: 10.3389/fimmu.2023.1111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
The humoral response is frequently dysfunctioning in autoimmunity with a frequent rise in total serum immunoglobulins, among which are found autoantibodies that may be pathogenic by themselves and/or propagate the inflammatory reaction. The infiltration of autoimmune tissues by antibody-secreting cells (ASCs) constitutes another dysfunction. The known high dependency of ASCs on the microenvironment to survive combined to the high diversity of infiltrated tissues implies that ASCs must adapt. Some tissues even within a single clinical autoimmune entity are devoid of infiltration. The latter means that either the tissue is not permissive or ASCs fail to adapt. The origin of infiltrated ASCs is also variable. Indeed, ASCs may be commonly generated in the secondary lymphoid organ draining the autoimmune tissue, and home at the inflammation site under the guidance of specific chemokines. Alternatively, ASCs may be generated locally, when ectopic germinal centers are formed in the autoimmune tissue. Alloimmune tissues with the example of kidney transplantation will also be discussed own to their high similarity with autoimmune tissues. It should also be noted that antibody production is not the only function of ASCs, since cells with regulatory functions have also been described. This article will review all the phenotypic variations indicative of tissue adaptation described so for at the level of ASC-infiltrating auto/alloimmune tissues. The aim is to potentially define tissue-specific molecular targets in ASCs to improve the specificity of future autoimmune treatments.
Collapse
Affiliation(s)
- Diane Giovannini
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Aude Belbezier
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Athan Baillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Rheumatology, Grenoble University Hospital, Grenoble, France
| | - Laurence Bouillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Mitsuhiro Kawano
- Department of Rheumatology, Kanazawa University Hospital, Kanazawa, Japan
| | | | | | - Johan Noble
- Department of Nephrology, Grenoble University Hospital, Grenoble, France
| | - Jacques-Olivier Pers
- B Lymphocytes, Autoimmunity and Immunotherapies, Brest University, INSERM, UMR1227, Brest, France.,Odontology Unit, Brest University Hospital, Brest, France
| | - Nathalie Sturm
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Bertrand Huard
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| |
Collapse
|
16
|
Wang S, Zhou Y, Huang J, Li H, Pang H, Niu D, Li G, Wang F, Zhou Z, Liu Z. Advances in experimental models of rheumatoid arthritis. Eur J Immunol 2023; 53:e2249962. [PMID: 36330559 DOI: 10.1002/eji.202249962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/16/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by persistent articular inflammation and joint damage. RA was first described over 200 years ago; however, its etiology and pathophysiology remain insufficiently understood. The current treatment of RA is mainly empirical or based on the current understanding of etiology with limited efficacy and/or substantial side effects. Thus, the development of safer and more potent therapeutics, validated and optimized in experimental models, is urgently required. To improve the transition from bench to bedside, researchers must carefully select the appropriate experimental models as well as draw the right conclusions. Here, we summarize the establishment, pathological features, potential mechanisms, advantages, and limitations of the currently available RA models. The aim of the review is to help researchers better understand available RA models; discuss future trends in RA model development, which can help highlight new translational and human-based avenues in RA research.
Collapse
Affiliation(s)
- Siwei Wang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Yanhua Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Jiangrong Huang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huilin Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huidan Pang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Dandan Niu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Guangyao Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Fei Wang
- Department of Experiment and Training, Hubei College of Chinese Medicine, Hubei Province, China
| | - Zushan Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Zhenzhen Liu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
17
|
Paradoxical Duel Role of Collagen in Rheumatoid Arthritis: Cause of Inflammation and Treatment. Bioengineering (Basel) 2022; 9:bioengineering9070321. [PMID: 35877372 PMCID: PMC9311863 DOI: 10.3390/bioengineering9070321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
In biology, collagen-biomaterial regulates several signaling mechanisms of bone and immune cells involved in tissue repair and any imbalance in collagen turnover may affect the homeostasis of cells, becoming a major cause of several complications. In this case, the administration of oral collagen may play a potential role in returning cells to their normal function. For several decades, the beneficial effects of collagen have been explored widely, and thus many commercial products are available in cosmetics, food, and biomedical fields. For instance, collagen-based-products have been widely used to treat the complications of cartilage-related-disorders. Many researchers are reporting the anti-arthritogenic properties of collagen-based materials. In contrast, collagen, especially type-II collagen (CII), has been widely used to induce arthritis by immunization in an animal-model with or without adjuvants, and the potentially immunogenic-properties of collagen have been continuously reported for a long time. Additionally, the immune tolerance of collagen is mainly regulated by the T-lymphocytes and B-cells. This controversial hypothesis is getting more and more evidence nowadays from both sides to support its mechanism. Therefore, this review links the gap between the arthritogenic and anti-arthritogenic effects of collagen and explored the actual mechanism to understand the fundamental concept of collagen in arthritis. Accordingly, this review opens-up several unrevealed scientific knots of collagen and arthritis and helps the researchers understand the potential use of collagen in therapeutic applications.
Collapse
|
18
|
Old but New: Group IIA Phospholipase A 2 as a Modulator of Gut Microbiota. Metabolites 2022; 12:metabo12040352. [PMID: 35448539 PMCID: PMC9029192 DOI: 10.3390/metabo12040352] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Among the phospholipase A2 (PLA2) superfamily, the secreted PLA2 (sPLA2) family contains 11 mammalian isoforms that exhibit unique tissue or cellular distributions and enzymatic properties. Current studies using sPLA2-deficient or -overexpressed mouse strains, along with mass spectrometric lipidomics to determine sPLA2-driven lipid pathways, have revealed the diverse pathophysiological roles of sPLA2s in various biological events. In general, individual sPLA2s exert their specific functions within tissue microenvironments, where they are intrinsically expressed through hydrolysis of extracellular phospholipids. Recent studies have uncovered a new aspect of group IIA sPLA2 (sPLA2-IIA), a prototypic sPLA2 with the oldest research history among the mammalian PLA2s, as a modulator of the gut microbiota. In the intestine, Paneth cell-derived sPLA2-IIA acts as an antimicrobial protein to shape the gut microbiota, thereby secondarily affecting inflammation, allergy, and cancer in proximal and distal tissues. Knockout of intestinal sPLA2-IIA in BALB/c mice leads to alterations in skin cancer, psoriasis, and anaphylaxis, while overexpression of sPLA2-IIA in Pla2g2a-null C57BL/6 mice induces systemic inflammation and exacerbates arthritis. These phenotypes are associated with notable changes in gut microbiota and fecal metabolites, are variable in different animal facilities, and are abrogated after antibiotic treatment, co-housing, or fecal transfer. These studies open a new mechanistic action of this old sPLA2 and add the sPLA2 family to the growing list of endogenous factors capable of affecting the microbe–host interaction and thereby systemic homeostasis and diseases.
Collapse
|
19
|
Merlo LM, Peng W, DuHadaway JB, Montgomery JD, Prendergast GC, Muller AJ, Mandik-Nayak L. The Immunomodulatory Enzyme IDO2 Mediates Autoimmune Arthritis through a Nonenzymatic Mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:571-581. [PMID: 34965962 PMCID: PMC8770583 DOI: 10.4049/jimmunol.2100705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
IDO2 is one of two closely related tryptophan catabolizing enzymes induced under inflammatory conditions. In contrast to the immunoregulatory role defined for IDO1 in cancer models, IDO2 has a proinflammatory function in models of autoimmunity and contact hypersensitivity. In humans, two common single-nucleotide polymorphisms have been identified that severely impair IDO2 enzymatic function, such that <25% of individuals express IDO2 with full catalytic potential. This, together with IDO2's relatively weak enzymatic activity, suggests that IDO2 may have a role outside of its function in tryptophan catabolism. To determine whether the enzymatic activity of IDO2 is required for its proinflammatory function, we used newly generated catalytically inactive IDO2 knock-in mice together with established models of contact hypersensitivity and autoimmune arthritis. Contact hypersensitivity was attenuated in catalytically inactive IDO2 knock-in mice. In contrast, induction of autoimmune arthritis was unaffected by the absence of IDO2 enzymatic activity. In pursuing this nonenzymatic IDO2 function, we identified GAPDH, Runx1, RANbp10, and Mgea5 as IDO2-binding proteins that do not interact with IDO1, implicating them as potential mediators of IDO2-specific function. Taken together, our findings identify a novel function for IDO2, independent of its tryptophan catabolizing activity, and suggest that this nonenzymatic function could involve multiple signaling pathways. These data show that the enzymatic activity of IDO2 is required only for some inflammatory immune responses and provide, to our knowledge, the first evidence of a nonenzymatic role for IDO2 in mediating autoimmune disease.
Collapse
Affiliation(s)
| | - Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA
| | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA,Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | | |
Collapse
|
20
|
Doré E, Joly-Beauparlant C, Morozumi S, Mathieu A, Lévesque T, Allaeys I, Duchez AC, Cloutier N, Leclercq M, Bodein A, Payré C, Martin C, Petit-Paitel A, Gelb MH, Rangachari M, Murakami M, Davidovic L, Flamand N, Arita M, Lambeau G, Droit A, Boilard E. The interaction of secreted phospholipase A2-IIA with the microbiota alters its lipidome and promotes inflammation. JCI Insight 2022; 7:152638. [PMID: 35076027 PMCID: PMC8855825 DOI: 10.1172/jci.insight.152638] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
Secreted phospholipase A2-IIA (sPLA2-IIA) hydrolyzes phospholipids to liberate lysophospholipids and fatty acids. Given its poor activity toward eukaryotic cell membranes, its role in the generation of proinflammatory lipid mediators is unclear. Conversely, sPLA2-IIA efficiently hydrolyzes bacterial membranes. Here, we show that sPLA2-IIA affects the immune system by acting on the intestinal microbial flora. Using mice overexpressing transgene-driven human sPLA2-IIA, we found that the intestinal microbiota was critical for both induction of an immune phenotype and promotion of inflammatory arthritis. The expression of sPLA2-IIA led to alterations of the intestinal microbiota composition, but housing in a more stringent pathogen-free facility revealed that its expression could affect the immune system in the absence of changes to the composition of this flora. In contrast, untargeted lipidomic analysis focusing on bacteria-derived lipid mediators revealed that sPLA2-IIA could profoundly alter the fecal lipidome. The data suggest that a singular protein, sPLA2-IIA, produces systemic effects on the immune system through its activity on the microbiota and its lipidome.
Collapse
Affiliation(s)
- Etienne Doré
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Charles Joly-Beauparlant
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Satoshi Morozumi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Alban Mathieu
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Tania Lévesque
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Isabelle Allaeys
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| | - Anne-Claire Duchez
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Nathalie Cloutier
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
| | - Mickaël Leclercq
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Antoine Bodein
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Christine Payré
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Cyril Martin
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Agnes Petit-Paitel
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Manu Rangachari
- CHU de Québec-Université Laval Research Center, Neurosciences Axis, Quebec City, Quebec, Canada
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Laetitia Davidovic
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Nicolas Flamand
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
- The Research Center of the University Institute of Cardiology and Pneumology of Quebec, Quebec City, Quebec, Canada
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama-City University, Yokohama, Japan
| | - Gérard Lambeau
- Côte d’Azur University, The French National Centre for Scientific Research, Institute of Molecular and Cellular Pharmacology, UMR7275, Valbonne Sophia Antipolis, France
| | - Arnaud Droit
- CHU de Québec-Université Laval Research Center, Endocrinology and Nephrology Axis, Quebec City, Quebec, Canada
| | - Eric Boilard
- CHU de Québec-Université Laval Research Center, Department of Microbiology, Infectiology and Immunology, Quebec City, Quebec, Canada
- ARThrite Research Center, University Laval, Quebec City, Quebec, Canada
| |
Collapse
|
21
|
Ohyama A, Osada A, Kawaguchi H, Kurata I, Nishiyama T, Iwai T, Ishigami A, Kondo Y, Tsuboi H, Sumida T, Matsumoto I. Specific Increase in Joint Neutrophil Extracellular Traps and Its Relation to Interleukin 6 in Autoimmune Arthritis. Int J Mol Sci 2021; 22:ijms22147633. [PMID: 34299252 PMCID: PMC8303722 DOI: 10.3390/ijms22147633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/10/2021] [Accepted: 07/15/2021] [Indexed: 12/25/2022] Open
Abstract
Neutrophils and their extracellular traps have been shown to play an important role in the pathogenesis of rheumatoid arthritis (RA), but the detailed mechanisms in joints are still unclear, and their regulation remains to be solved. Here, we explored neutrophil extracellular trap (NET)osis in experimental models of arthritis and further investigated the effects of interleukin-6 (IL-6) inhibition in neutrophils and NETosis. In skins of peptide GPI-induced arthritis (pGIA), citrullinated protein was detected as well as citrullinated histone expression in immunized skin but this was not specific to pGIA. Citrullinated histone expression in pGIA joints was specific to pGIA and was merged with neutrophil elastase, suggesting NETosis. Neutrophils in joints tend to upregulate IL-6 receptors when compared with bone marrow neutrophils. Administration of mouse anti-IL-6 receptor antibodies in pGIA suppressed arthritis in association with a decrease in neutrophil infiltration and NETosis in joints. In the plasma of RA patients, citrullinated protein was significantly reduced after tocilizumab treatment. Our results suggest that IL-6 enhances neutrophil chemotaxis and NETosis in inflammatory joints and could be the source of citrullinated proteins.
Collapse
Affiliation(s)
- Ayako Ohyama
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Atsumu Osada
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Hoshimi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
- Ichihara Hospital, Tsukuba, Ibaraki 300-3295, Japan
| | - Izumi Kurata
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Taihei Nishiyama
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Tamaki Iwai
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan;
| | - Yuya Kondo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Hiroto Tsuboi
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Takayuki Sumida
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
| | - Isao Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; (A.O.); (A.O.); (H.K.); (I.K.); (T.N.); (T.I.); (Y.K.); (H.T.); (T.S.)
- Correspondence: ; Tel.: +81-29-853-3186
| |
Collapse
|
22
|
Leceta J, Garin MI, Conde C. Mechanism of Immunoregulatory Properties of Vasoactive Intestinal Peptide in the K/BxN Mice Model of Autoimmune Arthritis. Front Immunol 2021; 12:701862. [PMID: 34335612 PMCID: PMC8322839 DOI: 10.3389/fimmu.2021.701862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
The K/BxN mouse model of rheumatoid arthritis (RA) closely resembles the human disease. In this model, arthritis results from activation of autoreactive KRN T cells recognizing the glycolytic enzyme glucose-6-phosphate isomerase (GPI) autoantigen, which provides help to GPI-specific B cells, resulting in the production of pathogenic anti-GPI antibodies that ultimately leads to arthritis symptoms from 4 weeks of age. Vasoactive intestinal peptide (VIP) is a neuropeptide broadly distributed in the central and peripheral nervous system that is also expressed in lymphocytes and other immune cell types. VIP is a modulator of innate and adaptive immunity, showing anti-inflammatory and immunoregulatory properties. Basically, this neuropeptide promotes a shift in the Th1/Th2 balance and enhances dedifferentiation of T regulatory cells (Treg). It has demonstrated its therapeutic effects on the collagen-induced arthritis (CIA) mouse model of RA. In the present hypothesis and theory article, we propose that the immunoregulatory properties of VIP may be due likely to the inhibition of T cell plasticity toward non-classic Th1 cells and an enhanced follicular regulatory T cells (Tfr) activity. The consequences of these regulatory properties are the reduction of systemic pathogenic antibody titers.
Collapse
Affiliation(s)
- Javier Leceta
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| | - Carmen Conde
- Laboratorio de Reumatología Experimental y Observacional, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| |
Collapse
|
23
|
Role of Intestinal Microbiota on Gut Homeostasis and Rheumatoid Arthritis. J Immunol Res 2021; 2021:8167283. [PMID: 34195296 PMCID: PMC8203374 DOI: 10.1155/2021/8167283] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/29/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that is immune mediated. Patients typically present with synovial inflammation, which gradually deteriorates to investigate severe cartilage and bone damage, affecting an individual's ability to perform basic tasks and impairing the quality of life. When evaluated against healthy controls, patients with RA have notable variations within the constituents of the gut microbiota. The human gastrointestinal tract mucosa is colonized by trillions of commensal microbacteria, which are key actors in the initiation, upkeep, and operation of the host immune system. Gut microbiota dysbiosis can adversely influence the immune system both locally and throughout the host, thus predisposing the host to a number of pathologies, including RA. Proximal intestinal immunomodulatory cells, situated in specific locales within the intestine, are a promising intermediary through which the gastrointestinal microbiota can influence the pathogenesis and progression of RA. In the early stages of the disease, the microbiota appear to differ from those present in healthy controls. This difference may reflect potential autoimmune mechanisms. Research studies evaluating intestinal microbiota have demonstrated that RA is associated with a bacterial population growth or with a decline when judged against control groups. The aim of this review is to examine the studies that connect intestinal dysbiosis with the autoimmune pathways implicated in the pathogenesis of RA.
Collapse
|
24
|
Osada A, Matsumoto I, Mikami N, Ohyama A, Kurata I, Kondo Y, Tsuboi H, Ishigami A, Sano Y, Arai T, Ise N, Sumida T. Citrullinated inter-alpha-trypsin inhibitor heavy chain 4 in arthritic joints and its potential effect in the neutrophil migration. Clin Exp Immunol 2021; 203:385-399. [PMID: 33238047 PMCID: PMC7874842 DOI: 10.1111/cei.13556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/31/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
The citrullinated inter-alpha-trypsin inhibitor heavy chain 4 (cit-ITIH4) was identified as its blood level was associated with the arthritis score in peptide glucose-6-phosphate-isomerase-induced arthritis (pGIA) mice and the disease activity in patients with rheumatoid arthritis (RA). This study aimed to clarify its citrullination pathway and function as related to neutrophils. In pGIA-afflicted joints, ITIH4 and cit-ITIH4 levels were examined by immunohistochemistry (IHC), immunoprecipitation (IP) and Western blotting (WB), while peptidylarginine deiminase (PAD) expression was measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), IHC and immunofluorescent methods. The pGIA mice received anti-lymphocyte antigen 6 complex locus G6D (Ly6G) antibodies to deplete neutrophils and the expression of cit-ITIH4 was investigated by WB. The amounts of ITIH4 and cit-ITIH4 in synovial fluid (SF) from RA and osteoarthritis (OA) patients were examined by I.P. and W.B. Recombinant ITIH4 and cit-ITIH4 were incubated with sera from healthy volunteers before its chemotactic ability and C5a level were evaluated using Boyden's chamber assay and enzyme-linked immunosorbent assay (ELISA). During peak arthritic phase, ITIH4 and cit-ITIH4 were increased in joints while PAD4 was over-expressed, especially in the infiltrating neutrophils of pGIA mice. Levels of cit-ITIH4 in plasma and joints significantly decreased upon neutrophil depletion. ITIH4 was specifically citrullinated in SF from RA patients compared with OA patients. Native ITIH4 inhibited neutrophilic migration and decreased C5a levels, while cit-ITIH4 increased its migration and C5a levels significantly. Cit-ITIH4 is generated mainly in inflamed joints by neutrophils via PAD4. Citrullination of ITIH4 may change its function to up-regulate neutrophilic migration by activating the complement cascade, exacerbating arthritis.
Collapse
Affiliation(s)
- A. Osada
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - I. Matsumoto
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - N. Mikami
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - A. Ohyama
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - I. Kurata
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Y. Kondo
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - H. Tsuboi
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - A. Ishigami
- Molecular Regulation of AgingTokyo Metropolitan Institute of GerontologyItabashi‐kuJapan
| | - Y. Sano
- Faculty of Arts and Sciences at FujiyoshidaShowa universityFujiyoshidaJapan
| | | | - N. Ise
- Fujirebio IncHachiojiJapan
| | - T. Sumida
- Division of RheumatologyDepartment of Internal MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| |
Collapse
|
25
|
Zhao Y, Urbonaviciute V, Xu B, Cai W, Sener Z, Ge C, Holmdahl R. Cartilage Oligomeric Matrix Protein Induced Arthritis-A New Model for Rheumatoid Arthritis in the C57BL/6 Mouse. Front Immunol 2021; 12:631249. [PMID: 33708221 PMCID: PMC7940517 DOI: 10.3389/fimmu.2021.631249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/07/2021] [Indexed: 12/29/2022] Open
Abstract
The most commonly used strains in experimental research, including genetically modified strains, are C57BL/6 mice. However, so far, no reliable model for rheumatoid arthritis is available, mainly due to the restriction by the MHC class II haplotype H-2b. Collagen-induced arthritis (CIA) is the most widely used animal model of rheumatoid arthritis, but C57BL/6 strain is resistant to CIA because there is no collagen II peptide associated with H-2b. To establish a rheumatoid arthritis model in C57BL/6 mice, we immunized C57BL/6NJ (B6N) mice with human cartilage oligomeric matrix protein (COMP), which induced severe arthritis with high incidence, accompanied by a strong auto-antibody response. Native COMP was required, as denatured COMP lost its ability to induce arthritis in B6N mice. An immunodominant COMP peptide was identified as the key T cell epitope, with a perfect fit into the Ab class II peptide binding pocket. A critical amino acid in this peptide was found to be phenylalanine at position 95. Recombinant COMP mutated at position 95 (COMP_F95S) lost its ability to induce arthritis or a strong immune response in the B6N mice. In conclusion, A new model for RA has been established using C57BL/6 mice through immunization with COMP, which is dependent on a COMP specific peptide binding Ab, thus in similarity with CIA in Aq expressing strains.
Collapse
Affiliation(s)
- Yunjuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Vilma Urbonaviciute
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Bingze Xu
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Weiwei Cai
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Zeynep Sener
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Changrong Ge
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
26
|
Galindo-Izquierdo M, Pablos Alvarez JL. Complement as a Therapeutic Target in Systemic Autoimmune Diseases. Cells 2021; 10:cells10010148. [PMID: 33451011 PMCID: PMC7828564 DOI: 10.3390/cells10010148] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
The complement system (CS) includes more than 50 proteins and its main function is to recognize and protect against foreign or damaged molecular components. Other homeostatic functions of CS are the elimination of apoptotic debris, neurological development, and the control of adaptive immune responses. Pathological activation plays prominent roles in the pathogenesis of most autoimmune diseases such as systemic lupus erythematosus, antiphospholipid syndrome, rheumatoid arthritis, dermatomyositis, and ANCA-associated vasculitis. In this review, we will review the main rheumatologic autoimmune processes in which complement plays a pathogenic role and its potential relevance as a therapeutic target.
Collapse
|
27
|
Bonami RH, Nyhoff LE, McNitt DH, Hulbert C, Felton JL, Kendall PL, Thomas JW. T-B Lymphocyte Interactions Promote Type 1 Diabetes Independently of SLAM-Associated Protein. THE JOURNAL OF IMMUNOLOGY 2020; 205:3263-3276. [PMID: 33199538 DOI: 10.4049/jimmunol.1900464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/15/2020] [Indexed: 01/05/2023]
Abstract
Signaling lymphocytic activation molecule-associated protein (SAP), a critical intracellular signaling molecule for T-B lymphocyte interactions, drives T follicular helper (Tfh) cell development in germinal centers (GCs). High-affinity islet autoantibodies predict type 1 diabetes (T1D) but do not cause β cell destruction. This paradox intimates Tfh cells as key pathologic effectors, consistent with an observed Tfh signature in T1D. To understand how fully developed Tfh (GC Tfh) contribute to different autoimmune processes, we investigated the role of SAP in T1D and autoantibody-mediated arthritis. Whereas spontaneous arthritis depended on SAP in the autoantibody-mediated K/BxN model, organized insulitis and diabetes onset were unabated, despite a blocked anti-insulin vaccine response in SAP-deficient NOD mice. GC Tfh and GC B cell development were blocked by loss of SAP in K/BxN mice. In contrast, although GC B cell formation was markedly reduced in SAP-deficient NOD mice, T cells with a GC Tfh phenotype were found at disease sites. CXCR3+ CCR6- (Tfh1) subset bias was observed among GC Tfh cells infiltrating the pancreas of NOD mice, which was enhanced by loss of SAP NOD T cells override SAP requirement to undergo activation and proliferation in response to Ag presentation, demonstrating the potential for productive cognate T-B lymphocyte interactions in T1D-prone mice. We find that SAP is essential when autoantibody-driven immune complexes promote inflammation but is not required for effective organ-specific autoimmune attack. Thus, Tfh induced in classic GC reactions are dispensable for T1D, but the autoimmune process in the NOD model retains pathogenic Tfh without SAP.
Collapse
Affiliation(s)
- Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; .,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Lindsay E Nyhoff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.,Division of Allergy, Pulmonary, and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Dudley H McNitt
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jamie L Felton
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Peggy L Kendall
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.,Division of Allergy, Pulmonary, and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - James W Thomas
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; .,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
28
|
Hubbard JJ, Pyzik M, Rath T, Kozicky LK, Sand KM, Gandhi AK, Grevys A, Foss S, Menzies SC, Glickman JN, Fiebiger E, Roopenian DC, Sandlie I, Andersen JT, Sly LM, Baker K, Blumberg RS. FcRn is a CD32a coreceptor that determines susceptibility to IgG immune complex-driven autoimmunity. J Exp Med 2020; 217:e20200359. [PMID: 32658257 PMCID: PMC7537387 DOI: 10.1084/jem.20200359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
IgG immune complexes (ICs) promote autoimmunity through binding fragment crystallizable (Fc) γ-receptors (FcγRs). Of these, the highly prevalent FcγRIIa (CD32a) histidine (H)-131 variant (CD32aH) is strongly linked to human autoimmune diseases through unclear mechanisms. We show that, relative to the CD32a arginine (R)-131 (CD32aR) variant, CD32aH more avidly bound human (h) IgG1 IC and formed a ternary complex with the neonatal Fc receptor (FcRn) under acidic conditions. In primary human and mouse cells, both CD32a variants required FcRn to induce innate and adaptive immune responses to hIgG1 ICs, which were augmented in the setting of CD32aH. Conversely, FcRn induced responses to IgG IC independently of classical FcγR, but optimal responses required FcRn and FcγR. Finally, FcRn blockade decreased inflammation in a rheumatoid arthritis model without reducing circulating autoantibody levels, providing support for FcRn's direct role in IgG IC-associated inflammation. Thus, CD32a and FcRn coregulate IgG IC-mediated immunity in a manner favoring the CD32aH variant, providing a novel mechanism for its disease association.
Collapse
Affiliation(s)
- Jonathan J. Hubbard
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Michal Pyzik
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Timo Rath
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa K. Kozicky
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kine M.K. Sand
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Amit K. Gandhi
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Algirdas Grevys
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stian Foss
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Susan C. Menzies
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan N. Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Edda Fiebiger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | | | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Laura M. Sly
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristi Baker
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Harvard Digestive Diseases Center, Boston, MA
| |
Collapse
|
29
|
The neuropathic phenotype of the K/BxN transgenic mouse with spontaneous arthritis: pain, nerve sprouting and joint remodeling. Sci Rep 2020; 10:15596. [PMID: 32973194 PMCID: PMC7515905 DOI: 10.1038/s41598-020-72441-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/13/2020] [Indexed: 01/11/2023] Open
Abstract
The adult K/BxN transgenic mouse develops spontaneous autoimmune arthritis with joint remodeling and profound bone loss. We report that both males and females display a severe sustained tactile allodynia which is reduced by gabapentin but not the potent cyclooxygenase inhibitor ketorolac. In dorsal horn, males and females show increased GFAP+ astrocytic cells; however, only males demonstrate an increase in Iba1+ microglia. In dorsal root ganglia (DRG), there is an increase in CGRP+, TH+, and Iba1+ (macrophage) labeling, but no increase in ATF3+ cells. At the ankle there is increased CGRP+, TH+, and GAP-43+ fiber synovial innervation. Thus, based on the changes in dorsal horn, DRG and peripheral innervation, we suggest that the adult K/BxN transgenic arthritic mice display a neuropathic phenotype, an assertion consistent with the analgesic pharmacology seen in this animal. These results indicate the relevance of this model to our understanding of the nociceptive processing which underlies the chronic pain state that evolves secondary to persistent joint inflammation.
Collapse
|
30
|
Autocrine motility factor and its receptor expression in musculoskeletal tumors. J Bone Oncol 2020; 24:100318. [PMID: 33101887 PMCID: PMC7574284 DOI: 10.1016/j.jbo.2020.100318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022] Open
Abstract
Discovery of Autocrine Motility factor (AMF) and its receptor (AMFR), both triggering tumor invasion and metastasis, may alter the therapeutic concept. Here, in this review, we show a novel outlook suggesting a cross-talking between musculoskeletal tumors and the skeletal milieu regulated by AMF-AMFR signaling. This review will highlight the pharmacological need for AMF and AMFR inhibitors for patients with malignant musculoskeletal tumors.
Management of aggressive malignant musculoskeletal tumors is clinically challenging and awaits the identification of regulator(s) that can be therapeutically used to improve patient outcome. Autocrine motility factor (AMF), a secreted cytokine, is known to alter the bone microenvironment by linking to its receptor AMFR (AMF Receptor), leading to tumor progression. It was noted that both the ligand and its receptor belong to the moonlighting family of proteins, as they contribute to intracellular metabolic function such as glycolysis and gluconeogenesis by expressing glucose-6-phosphate isomerase AMF/GPI and higher protein degradation by expressing AMFR/gp78 functioning as ubiquitin ligase activity. Thus, AMF/GPI and AMFR/gp78 contribute to higher metabolic turnover of protein and glucose. Recently, a large-scale cohort study including 23 different histological types of musculoskeletal tumors revealed that patients with osteosarcoma, multiple myeloma, rhabdomyosarcoma, and angiosarcoma tend to express higher levels of AMF, whereas multiple myeloma patients expressed high levels of AMFR. Consistently, the cellular data showed that a variety of musculoskeletal tumors express AMF and components of bone microenvironment express AMFR. Thus, a novel outlook suggests a cellular link and cross-talk between musculoskeletal tumors and the skeletal milieu are regulated by AMF-AMFR signaling. This review will highlight the pharmacological need for AMF and AMFR inhibitors as unmet medical needs for patients with malignant musculoskeletal tumors.
Collapse
|
31
|
Paranjape A, Haque TT, Kiwanuka KN, Qayum AA, Barnstein BO, Finkelman FD, Nigrovic PA, Ryan JJ. The Fyn-Stat5 cascade is required for Fcγ receptor-mediated mast cell function. Cell Immunol 2020; 356:104134. [PMID: 32862025 DOI: 10.1016/j.cellimm.2020.104134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023]
Abstract
Mast cells, well established effectors in allergic disease, can be activated by numerous stimuli. We previously found that the Fyn-Stat5B pathway is critical for FcεRI-stimulated mast cell function. Because IgG receptors employ similar signaling pathways, we investigated Fyn-Stat5B function downstream of FcγR. We report that FcγR elicits Fyn-dependent Stat5B tyrosine phosphorylation in mast cells. As we previously found for Fyn kinase, Stat5B is indispensable for IgG-mediated mast cell cytokine expression and secretion. However, Stat5B KO macrophages responded normally to FcγR signaling, indicating a lineage-restricted role for Stat5B. This was consistent in vivo, since passive FcγR activation induced anaphylaxis in a macrophage-dominated response even when Stat5B was deleted. We further investigated this lineage restriction using the K/BxN model of inflammatory arthritis. This model exhibits a rapid and transient mast cell-dependent joint inflammation followed days later by a macrophage- and neutrophil-dependent response. Consistent with our hypothesis, Fyn or Stat5B deficiency did not protect mice from late joint swelling, but greatly reduced the early mast cell-dependent response. This was associated with decreased joint and plasma histamine. We conclude that Fyn-Stat5B is a linage-restricted pathway critical for IgG-mediated mast cell responses.
Collapse
Affiliation(s)
- Anuya Paranjape
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Tamara T Haque
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Kasalina N Kiwanuka
- Department of Biochemistry, Virginia Commonwealth University, Box 980614, Richmond, VA 23298-0614, USA
| | - Amina Abdul Qayum
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Brian O Barnstein
- Department of Biology, Virginia Commonwealth University, Box 842012, Richmond, VA 23284-2012, USA
| | - Fred D Finkelman
- Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Peter A Nigrovic
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Box 842012, Richmond, VA 23284-2012, USA.
| |
Collapse
|
32
|
Merlo LMF, DuHadaway JB, Montgomery JD, Peng WD, Murray PJ, Prendergast GC, Caton AJ, Muller AJ, Mandik-Nayak L. Differential Roles of IDO1 and IDO2 in T and B Cell Inflammatory Immune Responses. Front Immunol 2020; 11:1861. [PMID: 32973768 PMCID: PMC7461966 DOI: 10.3389/fimmu.2020.01861] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Indoleamine-2,3-dioxygenase (IDO)1 and IDO2 are two closely related tryptophan catabolizing enzymes encoded by linked genes. The IDO pathway is also immunomodulatory, with IDO1 well-characterized as a mediator of tumor immune evasion. Due to its homology with IDO1, IDO2 has been proposed to have a similar immunoregulatory function. Indeed, IDO2, like IDO1, is necessary for the differentiation of regulatory T cells in vitro. However, compared to IDO1, in vivo studies demonstrated a contrasting role for IDO2, with experiments in preclinical models of autoimmune arthritis establishing a proinflammatory role for IDO2 in mediating B and T cell activation driving autoimmune disease. Given their potentially opposing roles in inflammatory responses, interpretation of results obtained using IDO1 or IDO2 single knockout mice could be complicated by the expression of the other enzyme. Here we use IDO1 and IDO2 single and double knockout (dko) mice to define the differential roles of IDO1 and IDO2 in B cell-mediated immune responses. Autoreactive T and B cell responses and severity of joint inflammation were decreased in IDO2 ko, but not IDO1 ko arthritic mice. Dko mice had a reduction in the number of autoantibody secreting cells and severity of arthritis: however, percentages of differentiated T cells and their associated cytokines were not reduced compared to IDO1 ko or wild-type mice. These data suggest that autoreactive B cell responses are mediated by IDO2, while autoreactive T cell responses are indirectly affected by IDO1 expression in the IDO2 ko mice. IDO2 also influenced antibody responses in models of influenza infection and immunization with T cell-independent type II antigens. Taken together, these studies provide evidence for the contrasting roles IDO1 and IDO2 play in immune responses, with IDO1 mediating T cell suppressive effects and IDO2 working directly in B cells as a proinflammatory mediator of B cell responses.
Collapse
Affiliation(s)
- Lauren M F Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - James B DuHadaway
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | | | - Wei-Dan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Peter J Murray
- Immunoregulation Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States.,Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | | | | | | |
Collapse
|
33
|
Ahmad L, Plancqueel S, Lazar N, Korri-Youssoufi H, Li de la Sierra-Gallay I, van Tilbeurgh H, Salmon L. Novel N-substituted 5-phosphate-d-arabinonamide derivatives as strong inhibitors of phosphoglucose isomerases: Synthesis, structure-activity relationship and crystallographic studies. Bioorg Chem 2020; 102:104048. [PMID: 32682158 DOI: 10.1016/j.bioorg.2020.104048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/26/2020] [Accepted: 06/24/2020] [Indexed: 10/24/2022]
Abstract
Phosphoglucose isomerase (PGI) is a cytosolic enzyme that catalyzes the reversible interconversion of d-glucose 6-phosphate and d-fructose 6-phosphate in glycolysis. Outside the cell, PGI is also known as autocrine motility factor (AMF), a cytokine secreted by a large variety of tumor cells that stimulates motility of cancer cells in vitro and metastases development in vivo. Human PGI and AMF are strictly identical proteins both in terms of sequence and 3D structure, and AMF activity is known to involve, at least in part, the enzymatic active site. Hence, with the purpose of finding new strong AMF-PGI inhibitors that could be potentially used as anticancer agents and/or as bioreceptors for carbohydrate-based electrochemical biosensors, we report in this study the synthesis and kinetic evaluation of several new human PGI inhibitors derived from the synthon 5-phospho-d-arabinono-1,4-lactone. Although not designed as high-energy intermediate analogue inhibitors of the enzyme catalyzed isomerization reaction, several of these N-substituted 5-phosphate-d-arabinonamide derivatives appears as new strong PGI inhibitors. For one of them, we report its crystal structure in complex with human PGI at 2.38 Å. Detailed analysis of its interactions at the active site reveals a new binding mode and shows that human PGI is relatively tolerant for modified inhibitors at the "head" C-1 part, offering promising perspectives for the future design of carbohydrate-based biosensors.
Collapse
Affiliation(s)
- Lama Ahmad
- Institut de Chimie Moléculaire et des Matériaux d'Orsay (ICMMO), Equipe de Chimie Bioorganique et Bioinorganique, CNRS UMR8182, LabEx LERMIT, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 420, 91405 Orsay Cedex, France
| | - Stéphane Plancqueel
- Institut de Biologie Intégrative de la Cellule (I2BC), CNRS UMR9198, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 430, 91405 Orsay Cedex, France
| | - Noureddine Lazar
- Institut de Biologie Intégrative de la Cellule (I2BC), CNRS UMR9198, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 430, 91405 Orsay Cedex, France
| | - Hafsa Korri-Youssoufi
- Institut de Chimie Moléculaire et des Matériaux d'Orsay (ICMMO), Equipe de Chimie Bioorganique et Bioinorganique, CNRS UMR8182, LabEx LERMIT, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 420, 91405 Orsay Cedex, France
| | - Inès Li de la Sierra-Gallay
- Institut de Biologie Intégrative de la Cellule (I2BC), CNRS UMR9198, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 430, 91405 Orsay Cedex, France
| | - Herman van Tilbeurgh
- Institut de Biologie Intégrative de la Cellule (I2BC), CNRS UMR9198, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 430, 91405 Orsay Cedex, France
| | - Laurent Salmon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay (ICMMO), Equipe de Chimie Bioorganique et Bioinorganique, CNRS UMR8182, LabEx LERMIT, Université Paris-Saclay, Rue du Doyen Georges Poitou, bât. 420, 91405 Orsay Cedex, France.
| |
Collapse
|
34
|
Yuan G, Yang S, Ng A, Fu C, Oursler MJ, Xing L, Yang S. RGS12 Is a Novel Critical NF-κB Activator in Inflammatory Arthritis. iScience 2020; 23:101172. [PMID: 32512384 PMCID: PMC7281782 DOI: 10.1016/j.isci.2020.101172] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/15/2019] [Accepted: 05/12/2020] [Indexed: 11/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common inflammatory disease, which currently lacks effective treatment. Here, we discovered that the Regulator of G Protein Signaling 12 (RGS12) plays a key role in regulating inflammation. Transcriptional and protein analysis revealed that RGS12 was upregulated in human and mouse RA macrophages. Deletion of RGS12 in myeloid lineage or globally inhibits the development of collagen-induced arthritis including joint swelling and bone destruction. Mechanistically, RGS12 associates with NF-κB(p65) to activate its phosphorylation and nuclear translocation through PTB domain, and NF-κB(p65) regulates RGS12 expression in a transcriptional manner. The nuclear translocation ability of NF-κB(p65) and RGS12 can both be enhanced by cyclooxygenase-2 (COX2). Furthermore, ablation of RGS12 via RNA interference significantly blocks the inflammatory process in vivo and in vitro. These results demonstrate that RGS12 plays a critical role in the pathogenesis of inflammatory arthritis.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuting Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Ng
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Chuanyun Fu
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Merry Jo Oursler
- Department of Medicine, Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Lewis BJ, Branch DR. Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Fc Receptor-Targeting Biologics. Pharmacology 2020; 105:618-629. [DOI: 10.1159/000508239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). <b><i>Summary:</i></b> Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. <b><i>Key Message:</i></b> The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.
Collapse
|
36
|
Fujimoto K, Uematsu S. Vaccine therapy for dysbiosis-related diseases. World J Gastroenterol 2020; 26:2758-2767. [PMID: 32550752 PMCID: PMC7284185 DOI: 10.3748/wjg.v26.i21.2758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/27/2020] [Accepted: 05/23/2020] [Indexed: 02/06/2023] Open
Abstract
Progress in genomic analysis has resulted in the proposal that the intestinal microbiota is a crucial environmental factor in the development of multifactorial diseases, such as obesity, diabetes, rheumatoid arthritis, and inflammatory bowel diseases represented by Crohn’s disease and ulcerative colitis. Dysregulated gut microbiome contributes to the pathogenesis of such disorders; however, there are few effective treatments for controlling only disease-mediating bacteria. Here, we review current knowledge about the intestinal microbiome in health and disease, and discuss a regulatory strategy using a parenteral vaccine with emulsified curdlan and CpG oligodeoxynucleotides, which we have recently developed. Unlike other conventional injectable immunizations, our vaccine contributes to the induction of antigen-specific systemic and mucosal immunity. This vaccine strategy can prevent infectious diseases such as Streptococcus pneumoniae infection, and control metabolic symptoms mediated by intestinal bacteria (e.g. Clostridium ramosum) by induction of high titers of antigen-specific IgA at target mucosal sites. In the future, our vaccination approach could be an effective therapy for common infectious diseases and dysbiosis-related disorders that have been difficult to control so far.
Collapse
MESH Headings
- Administration, Mucosal
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/microbiology
- Arthritis, Rheumatoid/therapy
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/microbiology
- Diabetes Mellitus, Type 2/therapy
- Dysbiosis/complications
- Dysbiosis/immunology
- Dysbiosis/microbiology
- Dysbiosis/therapy
- Gastrointestinal Microbiome/immunology
- Humans
- Immunity, Mucosal
- Immunization Schedule
- Immunization, Secondary
- Immunoglobulin A/immunology
- Immunoglobulin A/metabolism
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/microbiology
- Inflammatory Bowel Diseases/therapy
- Injections, Intramuscular
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Obesity/immunology
- Obesity/microbiology
- Obesity/therapy
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Polysaccharides, Bacterial/administration & dosage
- Polysaccharides, Bacterial/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- beta-Glucans/administration & dosage
- beta-Glucans/immunology
Collapse
Affiliation(s)
- Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 113-8654, Japan
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 113-8654, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 113-8654, Japan
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 113-8654, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
37
|
Ricciuto A, Sherman PM, Laxer RM. Gut microbiota in chronic inflammatory disorders: A focus on pediatric inflammatory bowel diseases and juvenile idiopathic arthritis. Clin Immunol 2020; 215:108415. [DOI: 10.1016/j.clim.2020.108415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
|
38
|
Williamson MG, Finelli MJ, Sleigh JN, Reddington A, Gordon D, Talbot K, Davies KE, Oliver PL. Neuronal over-expression of Oxr1 is protective against ALS-associated mutant TDP-43 mislocalisation in motor neurons and neuromuscular defects in vivo. Hum Mol Genet 2020; 28:3584-3599. [PMID: 31642482 PMCID: PMC6927465 DOI: 10.1093/hmg/ddz190] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/04/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
A common pathological hallmark of amyotrophic lateral sclerosis (ALS) and the related neurodegenerative disorder frontotemporal dementia, is the cellular mislocalization of transactive response DNA-binding protein 43 kDa (TDP-43). Additionally, multiple mutations in the TARDBP gene (encoding TDP-43) are associated with familial forms of ALS. While the exact role for TDP-43 in the onset and progression of ALS remains unclear, the identification of factors that can prevent aberrant TDP-43 localization and function could be clinically beneficial. Previously, we discovered that the oxidation resistance 1 (Oxr1) protein could alleviate cellular mislocalization phenotypes associated with TDP-43 mutations, and that over-expression of Oxr1 was able to delay neuromuscular abnormalities in the hSOD1G93A ALS mouse model. Here, to determine whether Oxr1 can protect against TDP-43-associated phenotypes in vitro and in vivo, we used the same genetic approach in a newly described transgenic mouse expressing the human TDP-43 locus harbouring an ALS disease mutation (TDP-43M337V). We show in primary motor neurons from TDP-43M337V mice that genetically-driven Oxr1 over-expression significantly alleviates cytoplasmic mislocalization of mutant TDP-43. We also further quantified newly-identified, late-onset neuromuscular phenotypes of this mutant line, and demonstrate that neuronal Oxr1 over-expression causes a significant reduction in muscle denervation and neuromuscular junction degeneration in homozygous mutants in parallel with improved motor function and a reduction in neuroinflammation. Together these data support the application of Oxr1 as a viable and safe modifier of TDP-43-associated ALS phenotypes.
Collapse
Affiliation(s)
- Matthew G Williamson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Mattéa J Finelli
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - James N Sleigh
- Department of Neuromuscular Diseases, Institute of Neurology, University College London, London WC1N 3BG, UK.,UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Amy Reddington
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Kay E Davies
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK.,MRC Harwell Institute, Harwell Campus, Didcot, Oxfordshire, OX11 0RD, UK
| |
Collapse
|
39
|
Relationship between T cells and microbiota in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 171:95-129. [PMID: 32475529 DOI: 10.1016/bs.pmbts.2020.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past decades, the fields of microbiology and immunology have largely advanced by using germ-free animals and next-generation sequencing. Many studies revealed the relationship among gut microbiota, activation of immune system, and various diseases. Especially, some gut commensals can generate their antigen-specific T cells. It is becoming clear that commensal bacteria have important roles in various autoimmune and inflammatory diseases, such as autism, rheumatoid arthritis (RA), and inflammatory bowel diseases (IBD). Recently, it was reported that commensals contribute to the cancer immune therapy. However, how commensal-specific T cells contribute to the disease development and cancer treatment are not fully understood yet. In this chapter, we will summarize the decade history of the studies associated with commensal-induced T cells and commensal-causing diseases.
Collapse
|
40
|
Tuncel J, Holmberg J, Haag S, Hopkins MH, Wester-Rosenlöf L, Carlsen S, Olofsson P, Holmdahl R. Self-reactive T cells induce and perpetuate chronic relapsing arthritis. Arthritis Res Ther 2020; 22:95. [PMID: 32345366 PMCID: PMC7187533 DOI: 10.1186/s13075-020-2104-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/13/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND CD4+ T cells play a central role during the early stages of rheumatoid arthritis (RA), but to which extent they are required for the perpetuation of the disease is still not fully understood. The aim of the current study was to obtain conclusive evidence that T cells drive chronic relapsing arthritis. METHODS We used the rat pristane-induced arthritis model, which accurately portrays the chronic relapsing-remitting disease course of RA, to examine the contribution of T cells to chronic arthritis. RESULTS Rats subjected to whole-body irradiation and injected with CD4+ T cells from lymph nodes of pristane-injected donors developed chronic arthritis that lasted for more than 4 months, whereas T cells from the spleen only induced acute disease. Thymectomy in combination with irradiation enhanced the severity of arthritis, suggesting that sustained lymphopenia promotes T cell-driven chronic inflammation in this model. The ability of T cells to induce chronic arthritis correlated with their expression of Th17-associated transcripts, and while depletion of T cells in rats with chronic PIA led to transient, albeit significant, reduction in disease, neutralization of IL-17 resulted in almost complete and sustained remission. CONCLUSION These findings show that, once activated, self-reactive T cells can sustain inflammatory responses for extended periods of time and suggest that such responses are promoted in the presence of IL-17.
Collapse
Affiliation(s)
- Jonatan Tuncel
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Holmberg
- Section for Medical Inflammation Research, BMCI11, Lund University, Lund, Sweden
| | - Sabrina Haag
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Lena Wester-Rosenlöf
- Section for Medical Inflammation Research, BMCI11, Lund University, Lund, Sweden
| | - Stefan Carlsen
- Section for Medical Inflammation Research, BMCI11, Lund University, Lund, Sweden
| | - Peter Olofsson
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden. .,Section for Medical Inflammation Research, BMCI11, Lund University, Lund, Sweden.
| |
Collapse
|
41
|
Tessandier N, Melki I, Cloutier N, Allaeys I, Miszta A, Tan S, Milasan A, Michel S, Benmoussa A, Lévesque T, Côté F, McKenzie SE, Gilbert C, Provost P, Brisson AR, Wolberg AS, Fortin PR, Martel C, Boilard É. Platelets Disseminate Extracellular Vesicles in Lymph in Rheumatoid Arthritis. Arterioscler Thromb Vasc Biol 2020; 40:929-942. [PMID: 32102567 PMCID: PMC8073225 DOI: 10.1161/atvbaha.119.313698] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The lymphatic system is a circulatory system that unidirectionally drains the interstitial tissue fluid back to blood circulation. Although lymph is utilized by leukocytes for immune surveillance, it remains inaccessible to platelets and erythrocytes. Activated cells release submicron extracellular vesicles (EV) that transport molecules from the donor cell. In rheumatoid arthritis, EV accumulate in the joint where they can interact with numerous cellular lineages. However, whether EV can exit the inflamed tissue to recirculate is unknown. Here, we investigated whether vascular leakage that occurs during inflammation could favor EV access to the lymphatic system. Approach and Results: Using an in vivo model of autoimmune inflammatory arthritis, we show that there is an influx of platelet EV, but not EV from erythrocytes or leukocytes, in joint-draining lymph. In contrast to blood platelet EV, lymph platelet EV lacked mitochondrial organelles and failed to promote coagulation. Platelet EV influx in lymph was consistent with joint vascular leakage and implicated the fibrinogen receptor α2bβ3 and platelet-derived serotonin. CONCLUSIONS These findings show that platelets can disseminate their EV in fluid that is inaccessible to platelets and beyond the joint in this disease.
Collapse
Affiliation(s)
- Nicolas Tessandier
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Imene Melki
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Nathalie Cloutier
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Isabelle Allaeys
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Adam Miszta
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill (A.M., A.S.W.)
- Montreal Heart Institute, Quebec, Canada (A.M., C.M.)
| | - Sisareuth Tan
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France (S.T., A.R.B.)
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine (A.M., C.M.), Université de Montréal, Quebec, Canada
| | - Sara Michel
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Abderrahim Benmoussa
- Department of Nutrition, CHU Sainte-Justine (A.B.), Université de Montréal, Quebec, Canada
| | - Tania Lévesque
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Francine Côté
- Institut Imagine, Inserm U1163, Laboratoire Olivier Hermine, Paris, France (F.C.)
| | - Steven E McKenzie
- Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (S.E.M.)
| | - Caroline Gilbert
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Patrick Provost
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Alain R Brisson
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France (S.T., A.R.B.)
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill (A.M., A.S.W.)
| | - Paul R Fortin
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec-Université Laval, Québec, Canada (P.R.F., E.B.)
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine (A.M., C.M.), Université de Montréal, Quebec, Canada
- Montreal Heart Institute, Quebec, Canada (A.M., C.M.)
| | - Éric Boilard
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec-Université Laval, Québec, Canada (P.R.F., E.B.)
| |
Collapse
|
42
|
Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunol Rev 2020; 294:164-176. [PMID: 31944330 DOI: 10.1111/imr.12841] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022]
Abstract
Mutations of the genes encoding T-cell receptor (TCR)-proximal signaling molecules, such as ZAP-70, can be causative of immunological diseases ranging from T-cell immunodeficiency to T-cell-mediated autoimmune disease. For example, SKG mice, which carry a hypomorphic point mutation of the Zap-70 gene, spontaneously develop T-cell-mediated autoimmune arthritis immunopathologically similar to human rheumatoid arthritis (RA). The Zap-70 mutation alters the sensitivity of developing T cells to thymic positive/negative selection by self-peptides/MHC complexes, shifting self-reactive TCR repertoire to include a dominant arthritogenic specificity and also affecting thymic development and function of autoimmune suppressive regulatory T (Treg) cells. Polyclonal self-reactive T cells, including potentially arthritogenic T cells, thus produced by the thymus recognize self-peptide/MHC complexes on antigen-presenting cells (APCs) in the periphery and stimulate them to produce cytokines including IL-6 to drive the arthritogenic T cells to differentiate into arthritogenic T-helper 17 (Th17) cells. Insufficient Treg suppression or activation of APCs via microbial and other environmental stimuli evokes arthritis by activating granulocyte-macrophage colony-stimulating factor-secreting effector Th17 cells, mediating chronic bone-destructive joint inflammation by activating myeloid cells, innate lymphoid cells, and synoviocytes in the joint. These findings obtained from the study of SKG mouse arthritis are instrumental in understanding how arthritogenic T cells are produced, become activated, and differentiate into effector T cells mediating arthritis, and may help devising therapeutic measures targeting autoimmune pathogenic Th17 cells or autoimmune-suppressing Treg cells to treat and prevent RA.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
43
|
Host-microbiota interactions in rheumatoid arthritis. Exp Mol Med 2019; 51:1-6. [PMID: 31827063 PMCID: PMC6906371 DOI: 10.1038/s12276-019-0283-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/08/2019] [Accepted: 05/22/2019] [Indexed: 12/30/2022] Open
Abstract
The gut microbiota has been proposed to be an important environmental factor in the development of rheumatoid arthritis (RA). Here, we review a growing body of evidence from human and animal studies that supports the hypothesis that intestinal microbiota play a role in RA. Previous studies from we and others showed an altered composition of the microbiota in early RA patients. A recent study demonstrated that Prevotella species are dominant in the intestine of patients in the preclinical stages of RA. In addition, Prevotella-dominated microbiota isolated from RA patients contributes to the development of Th17 cell-dependent arthritis in SKG mice. Moreover, it was reported that periodontal bacteria correlates with the pathogenesis of RA. In this review, we discuss the link between oral bacteria and the development of arthritis. However, many questions remain to be elucidated in terms of molecular mechanisms for the involvement of intestinal and oral microbiota in RA pathogenesis. Microbes living in the gut and mouth have been implicated in the development of rheumatoid arthritis (RA) and treatments that promote the growth of healthier bacterial communities may help weaken this autoimmune disease. Yuichi Maeda and Kiyoshi Takeda from Osaka University, Japan, review data from mice and humans linking RA to altered microbial compositions in the gut. They focus on a particular bacterium called Prevotella copri, which is found at much higher numbers in the gastrointestinal tracts of people with newly diagnosed RA than in those without the disease. Certain mouth-dwelling bacteria may also help exacerbate RA through the induction of antibodies directed against the host. The exact molecular mechanism by which gut and oral microbes contribute to RA remains unclear.
Collapse
|
44
|
Lewis BJB, Ville J, Blacquiere M, Cen S, Spirig R, Zuercher AW, Käsermann F, Branch DR. Using the K/BxN mouse model of endogenous, chronic, rheumatoid arthritis for the evaluation of potential immunoglobulin-based therapeutic agents, including IVIg and Fc-μTP-L309C, a recombinant IgG1 Fc hexamer. BMC Immunol 2019; 20:44. [PMID: 31801459 PMCID: PMC6894239 DOI: 10.1186/s12865-019-0328-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/21/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND High-dose intravenous immunoglobulin (IVIg), and more recently, subcutaneously-delivered Ig (SCIg), are used to treat a variety of autoimmune diseases; however, there are challenges associated with product production, availability, access and efficacy. These challenges have provided incentives to develop a human recombinant Fc as a more potent alternative to IVIg and SCIg for the treatment of autoimmune diseases. Recently, a recombinant human IgG1 Fc hexamer (Fc-μTP-L309C) was shown to be more efficacious than IVIg in a variety of autoimmune mouse models. We have now examined its efficacy compared to IVIg and SCIg in the K/BxN mouse model of endogenous, chronic rheumatoid arthritis (RA). RESULT Using the serum-transfer K/BxN model and the endogenous autoimmune model, amelioration of the arthritis was achieved. Effective treatment required high and frequent doses of IVIg, SCIg and Fc-μTP-L309C. However, Fc-μTP-L309C was efficacious at 10-fold lower doses that IVIg/SCIg. Also, arthritis could be prevented when Fc-μTP-L309C was given prior to onset of the arthritis in both the endogenous model and in the serum transfer model. CONCLUSIONS Our results show that Fc-μTP-L309C is a powerful treatment for the prevention and amelioration of severe, chronic arthritis in a true autoimmune mouse model of RA. Thus, the K/BxN endogenous arthritis model should be useful for testing potential therapeutics for RA. Our findings provide rationale for further examination of the treatment efficacy of immunoglobulin-based therapeutics in rheumatoid arthritis.
Collapse
Affiliation(s)
- Bonnie J B Lewis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 67 College St., Toronto, Ontario, M5G 2M1, Canada.,Centre for Innovation, Canadian Blood Services, 67 College St., Toronto, Ontario, M5G 2M1, Canada
| | - Jade Ville
- Centre for Innovation, Canadian Blood Services, 67 College St., Toronto, Ontario, M5G 2M1, Canada.,School for Biology-Biochemistry-Biotechnology, Catholic University of Lyon, 10 place des Archives, 69288, Lyon Cedex 02, France
| | - Megan Blacquiere
- Centre for Innovation, Canadian Blood Services, 67 College St., Toronto, Ontario, M5G 2M1, Canada
| | - Selena Cen
- Centre for Innovation, Canadian Blood Services, 67 College St., Toronto, Ontario, M5G 2M1, Canada
| | - Rolf Spirig
- CSL Behring, Research, CSL Biologics Research Center, Wankdorfstrasse 10, 3010, Bern, Switzerland
| | - Adrian W Zuercher
- CSL Behring, Research, CSL Biologics Research Center, Wankdorfstrasse 10, 3010, Bern, Switzerland
| | - Fabian Käsermann
- CSL Behring, Research, CSL Biologics Research Center, Wankdorfstrasse 10, 3010, Bern, Switzerland
| | - Donald R Branch
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 67 College St., Toronto, Ontario, M5G 2M1, Canada. .,Centre for Innovation, Canadian Blood Services, 67 College St., Toronto, Ontario, M5G 2M1, Canada. .,Department of Medicine, University of Toronto, 67 College St., Toronto, Ontario, M5G 2M1, Canada.
| |
Collapse
|
45
|
Chemokines in rheumatic diseases: pathogenic role and therapeutic implications. Nat Rev Rheumatol 2019; 15:731-746. [PMID: 31705045 DOI: 10.1038/s41584-019-0323-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/20/2022]
Abstract
Chemokines, a family of small secreted chemotactic cytokines, and their G protein-coupled seven transmembrane spanning receptors control the migratory patterns, positioning and cellular interactions of immune cells. The levels of chemokines and their receptors are increased in the blood and within inflamed tissue of patients with rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, vasculitis or idiopathic inflammatory myopathies. Chemokine ligand-receptor interactions control the recruitment of leukocytes into tissue, which are central to the pathogenesis of these rheumatic diseases. Although the blockade of various chemokines and chemokine receptors has yielded promising results in preclinical animal models of rheumatic diseases, human clinical trials have, in general, been disappointing. However, there have been glimmers of hope from several early-phase clinical trials that suggest that sufficiently blocking the relevant chemokine pathway might in fact have clinical benefits in rheumatic diseases. Hence, the chemokine system remains a promising therapeutic target for rheumatic diseases and requires further study.
Collapse
|
46
|
Kurata I, Matsumoto I, Ohyama A, Osada A, Ebe H, Kawaguchi H, Kaneko S, Kondo Y, Tsuboi H, Tomioka A, Kaji H, Sumida T. Potential involvement of OX40 in the regulation of autoantibody sialylation in arthritis. Ann Rheum Dis 2019; 78:1488-1496. [PMID: 31300460 DOI: 10.1136/annrheumdis-2019-215195] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/18/2019] [Accepted: 07/03/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVE An increased proportion of circulating follicular helper T (Tfh) cells was reported in rheumatoid arthritis (RA), but it remains uncertain how Tfh cells affect antibody hyposialylation. We investigated the regulation of autoantibody hyposialylation by Tfh cells in RA using murine model. METHODS Behaviours of Tfh cells and their function on B cell promotion were analysed. Change of arthritogenicity and sialylation of autoantibodies during the course of arthritis was examined by mass spectrometry. Tfh-mediated regulation of hyposialylation was investigated, and the responsible cell surface molecule was specified both in vitro and in vivo. The relation between circulating Tfh cells and hyposialylation was analysed in patients with RA. RESULTS An increase in Tfh, particularly interleukin-17 producing Tfh (Tfh17) cells, at the onset of arthritis and their enhancement of autoantibody production were found. Autoantibodies at the onset phase demonstrated stronger inflammatory properties than those at the resolution phase, and mass spectrometric analysis revealed their difference in sialylation. In vitro coculture showed enhanced hyposialylation by the Tfh cells via OX40, which was highly expressed in the Tfh and Tfh17 cells. Blockade of OX40 prevented the development of arthritis with reduction in Tfh17 cells and recovery of autoantibody sialylation. Analysis of patients with RA showed abundance of OX40-overexpressing Tfh17 cells, and their proportion correlated negatively with the expression of α2,6-sialyltransferase 1, an enzyme responsible for sialylation. CONCLUSIONS OX40 expressed on Tfh cells can regulate autoantibody sialylation and play a crucial role in the development of autoimmune arthritis.
Collapse
Affiliation(s)
- Izumi Kurata
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Isao Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ayako Ohyama
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsumu Osada
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ebe
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hoshimi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shunta Kaneko
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuya Kondo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroto Tsuboi
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Azusa Tomioka
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Hiroyuki Kaji
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takayuki Sumida
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
47
|
Mast cells drive IgE-mediated disease but might be bystanders in many other inflammatory and neoplastic conditions. J Allergy Clin Immunol 2019; 144:S19-S30. [DOI: 10.1016/j.jaci.2019.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 01/05/2023]
|
48
|
Matsumoto I, Kurata I, Ohyama A, Kawaguchi H, Ebe H, Osada A, Kondo Y, Tsuboi H, Sumida T. Revisit of autoimmunity to glucose-6-phosphate isomerase in experimental and rheumatoid arthritis. Mod Rheumatol 2019; 30:232-238. [PMID: 31441345 DOI: 10.1080/14397595.2019.1659539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is an inflammatory disorder characterized by synovial inflammation in multiple joints. Autoantibodies (Abs) are the hallmark of RA, and as disease-specific and diagnostic markers, rheumatoid factor and anti-citrullinated protein antibody (ACPA) are produced pre-clinically, but their pathogenic roles in RA remain elusive. In this review, we focus on one of the candidate autoantigens in RA; glucose-6-phosphate isomerase (GPI). The arthritogenic role of GPI has been confirmed in two different mouse models: the K/BxN- and GPI-induced arthritis models. Both anti-GPI Abs and citrullinated-GPI peptide Abs have been detected in human RA. Studies conducted in these rodent models have confirmed that the pathogenesis of arthritis involves the localization of autoantigens not only in the joints but also in the circulation. In this review, we revisit and summarize the arthritogenic relevance of GPI in animal RA models and in human RA, and extend the discussion to joint-specific inflammation.
Collapse
Affiliation(s)
- Isao Matsumoto
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Izumi Kurata
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ayako Ohyama
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hoshimi Kawaguchi
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ebe
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsumu Osada
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuya Kondo
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroto Tsuboi
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
49
|
Takeuchi Y, Hirota K, Sakaguchi S. Synovial Tissue Inflammation Mediated by Autoimmune T Cells. Front Immunol 2019; 10:1989. [PMID: 31497022 PMCID: PMC6712680 DOI: 10.3389/fimmu.2019.01989] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/06/2019] [Indexed: 01/08/2023] Open
Abstract
In rheumatoid arthritis (RA), various hematopoietic and non-hematopoietic cells present in the synovial tissue secrete numerous inflammatory mediators including pro-inflammatory cytokines critical for the induction of chronic joint inflammation and bone destruction. Fibroblast-like synoviocytes (FLSs) in the non-hematopoietic cell compartment are key inflammatory cells activated in inflamed joints and driving the disease; yet how synovial tissue inflammation is modulated by autoimmune T cells is not fully understood. In this review, mainly based on recent findings with a mouse model of spontaneous autoimmune arthritis, we discuss the mechanism of Th17-mediated synovial tissue inflammation; that is, what environmental stimuli and arthritogenic self-antigens trigger arthritis, how arthritogenic T cells initiate joint inflammation by stimulating FLSs, and how the cellular sources of GM-CSF from lymphoid and tissue stromal cells in the synovium contribute to the development of arthritis. We also highlight possible plasticity of Th17 cells toward pathogenic GM-CSF producers, and the functional instability of regulatory T cells under inflammatory conditions in RA joints.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
50
|
Flak MB, Colas RA, Muñoz-Atienza E, Curtis MA, Dalli J, Pitzalis C. Inflammatory arthritis disrupts gut resolution mechanisms, promoting barrier breakdown by Porphyromonas gingivalis. JCI Insight 2019; 4:125191. [PMID: 31292292 PMCID: PMC6629160 DOI: 10.1172/jci.insight.125191] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis is linked with altered host immune responses and severe joint destruction. Recent evidence suggests that loss of gut homeostasis and barrier breach by pathobionts, including Porphyromonas gingivalis, may influence disease severity. The mechanism(s) leading to altered gut homeostasis and barrier breakdown in inflammatory arthritis are poorly understood. In the present study, we found a significant reduction in intestinal concentrations of several proresolving mediators during inflammatory arthritis, including downregulation of the gut-protective mediator resolvin D5n-3 DPA (RvD5n-3 DPA). This was linked with increased metabolism of RvD5n-3 DPA to its inactive 17-oxo metabolite. We also found downregulation of IL-10 expression in the gut of arthritic mice that was coupled with a reduction in IL-10 and IL-10 receptor (IL-10R) in lamina propria macrophages. These changes were linked with a decrease in the number of mucus-producing goblet cells and tight junction molecule expression in the intestinal epithelium of arthritic mice when compared with naive mice. P. gingivalis inoculation further downregulated intestinal RvD5n-3 DPA and Il-10 levels and the expression of gut tight junction proteins. RvD5n-3 DPA, but not its metabolite 17-oxo-RvD5n-3 DPA, increased the expression of both IL-10 and IL-10R in macrophages via the upregulation of the aryl hydrocarbon receptor agonist l-kynurenine. Administration of RvD5n-3 DPA to arthritic P. gingivalis-inoculated mice increased intestinal Il-10 expression, restored gut barrier function, and reduced joint inflammation. Together, these findings uncover mechanisms in the pathogenesis of rheumatoid arthritis, where disruption of the gut RvD5n-3 DPA-IL-10 axis weakens the gut barrier, which becomes permissive to the pathogenic actions of the pathobiont P. gingivalis.
Collapse
Affiliation(s)
- Magdalena B. Flak
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
| | - Romain A. Colas
- Lipid Mediator Unit, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Estefanía Muñoz-Atienza
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
| | | | - Jesmond Dalli
- Lipid Mediator Unit, William Harvey Research Institute, QMUL, London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, QMUL, London, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, QMUL, London, United Kingdom
| |
Collapse
|