101
|
Hedin CRH, Vavricka SR, Stagg AJ, Schoepfer A, Raine T, Puig L, Pleyer U, Navarini A, van der Meulen-de Jong AE, Maul J, Katsanos K, Kagramanova A, Greuter T, González-Lama Y, van Gaalen F, Ellul P, Burisch J, Bettenworth D, Becker MD, Bamias G, Rieder F. The Pathogenesis of Extraintestinal Manifestations: Implications for IBD Research, Diagnosis, and Therapy. J Crohns Colitis 2019; 13:541-554. [PMID: 30445584 DOI: 10.1093/ecco-jcc/jjy191] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This article reports on the sixth scientific workshop of the European Crohn's and Colitis Organisation [ECCO] on the pathogenesis of extraintestinal manifestations [EIMs] in inflammatory bowel disease [IBD]. This paper has been drafted by 15 ECCO members and 6 external experts [in rheumatology, dermatology, ophthalmology, and immunology] from 10 European countries and the USA. Within the workshop, contributors formed subgroups to address specific areas. Following a comprehensive literature search, the supporting text was finalized under the leadership of the heads of the working groups before being integrated by the group consensus leaders.
Collapse
Affiliation(s)
- C R H Hedin
- Gastroenterology unit, Patient Area Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - S R Vavricka
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - A J Stagg
- Centre for Immunobiology, Bart's and The London Medical School, Queen Mary University of London, London, UK
| | - A Schoepfer
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - T Raine
- Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University Teaching Hospitals NHS Foundation Trust, Cambridge, UK
| | - L Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - U Pleyer
- University Eye Clinic, Uveitis Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - A Navarini
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - J Maul
- Gastroenterologie am Bayerischen Platz, Berlin, Germany.,Department of Medicine (Gastroenterology, Infectious Diseases, Rheumatology), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - K Katsanos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Ioannina School of Medical Sciences, Ioannina, Greece
| | - A Kagramanova
- IBD Department, The Loginov Moscow Clinical Scientific Centre, Moscow, Russia
| | - T Greuter
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Y González-Lama
- IBD Unit, Gastroenterology and Hepatology Department, Puerta de Hierro University Hospital, Majadahonda, Madrid, Spain
| | - F van Gaalen
- Department of Rheumatology, Leiden University Medical Center [LUMC], Leiden, Netherlands
| | - P Ellul
- Department of Medicine, Division of Gastroenterology, Mater Dei Hospital, Msida, Malta
| | - J Burisch
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark.,Abdominal Center K, Medical Section, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - D Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - M D Becker
- Department of Ophthalmology, Triemli Hospital, Zurich, Switzerland & Department of Ophthalmology, University of Heidelberg, Heidelberg, Germany
| | - G Bamias
- National and Kapodistrian University of Athens, GI Unit, 3rd Academic Department of Internal Medicine, Sotiria Hospital, Athens, Greece
| | - F Rieder
- Department of Gastroenterology, Hepatology and Nutrition; Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
102
|
Tang C, Makusheva Y, Sun H, Han W, Iwakura Y. Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. J Leukoc Biol 2019; 106:903-917. [PMID: 30964564 PMCID: PMC6850291 DOI: 10.1002/jlb.2ri0119-031r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Myeloid C‐type lectin receptors (CLRs), which consist of an extracellular carbohydrate recognition domain and intracellular signal transducing motif such as the immunoreceptor tyrosine‐based activation motif (ITAM) or immunoreceptor tyrosine‐based inhibitory motif (ITIM), are innate immune receptors primarily expressed on myeloid lineage cells such as dendritic cells (DCs) and Mϕs. CLRs play important roles in host defense against infection by fungi and bacteria by recognizing specific carbohydrate components of these pathogens. However, these immune receptors also make important contributions to immune homeostasis of mucosa and skin in mammals by recognizing components of microbiota, as well as by recognizing self‐components such as alarmins from dead cells and noncanonical non‐carbohydrate ligands. CLR deficiency not only induces hypersensitivity to infection, but also causes dysregulation of muco‐cutaneous immune homeostasis, resulting in the development of allergy, inflammation, autoimmunity, and tumors. In this review, we introduce recent discoveries regarding the roles of myeloid CLRs in the immune system exposed to the environment, and discuss the roles of these lectin receptors in the development of colitis, asthma, psoriasis, atopic dermatitis, and cancer. Although some CLRs are suggested to be involved in the development of these diseases, the function of CLRs and their ligands still largely remain to be elucidated.
Collapse
Affiliation(s)
- Ce Tang
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Haiyang Sun
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
103
|
Rehaume LM, Matigian N, Mehdi AM, Lachner N, Bowerman KL, Daly J, Lê Cao KA, Hugenholtz P, Thomas R. IL-23 favours outgrowth of spondyloarthritis-associated pathobionts and suppresses host support for homeostatic microbiota. Ann Rheum Dis 2019; 78:494-503. [PMID: 30700427 DOI: 10.1136/annrheumdis-2018-214381] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Certain gut bacterial families, including Bacteroidaceae, Porphyromonadaceae and Prevotellaceae, are increased in people suffering from spondyloarthropathy (SpA), a disease group associated with IL23R signalling variants. To understand the relationship between host interleukin (IL)-23 signalling and gut bacterial dysbiosis in SpA, we inhibited IL-23 in dysbiotic ZAP-70-mutant SKG mice that develop IL-23-dependent SpA-like arthritis, psoriasis-like skin inflammation and Crohn's-like ileitis in response to microbial beta 1,3-glucan (curdlan). METHODS We treated SKG mice weekly with anti-IL-23 or isotype mAb for 3 weeks, rested them for 3 weeks, then administered curdlan or saline. We collected faecal samples longitudinally, assessed arthritis, spondylitis, psoriasis and ileitis histologically, and analysed the microbiota community profiles using next-generation sequencing. We used multivariate sparse partial least squares discriminant analysis to identify operational taxonomic unit (OTU) signatures best classifying treatment groups and linear regression to develop a predictive model of disease severity. RESULTS IL-23p19 inhibition in naïve SKG mice decreased Bacteroidaceae, Porphyromonadaceae and Prevotellaceae. Abundance of Clostridiaceae and Lachnospiraceae families concomitantly increased, and curdlan-mediated SpA development decreased. Abundance of Enterobacteriaceae and Porphyromonadaceae family and reduction in Lachnospiraceae Dorea genus OTUs early in disease course were associated with disease severity in affected tissues. CONCLUSIONS Dysbiosis in SKG mice reflects human SpA and is IL-23p19 dependent. In genetically susceptible hosts, IL-23p19 favours outgrowth of SpA-associated pathobionts and reduces support for homeostatic-inducing microbiota. The relative abundance of specific pathobionts is associated with disease severity.
Collapse
Affiliation(s)
- Linda M Rehaume
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Nicholas Matigian
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
- QFAB Bioinformatics, Institute of Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ahmed M Mehdi
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Nancy Lachner
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland, Australia
| | - Kate L Bowerman
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland, Australia
| | - Joshua Daly
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland, Australia
| | - Kim-Anh Lê Cao
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
104
|
Altered Bacterial-Fungal Interkingdom Networks in the Guts of Ankylosing Spondylitis Patients. mSystems 2019; 4:mSystems00176-18. [PMID: 30944880 PMCID: PMC6435815 DOI: 10.1128/msystems.00176-18] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 03/13/2019] [Indexed: 12/21/2022] Open
Abstract
The human gut is colonized by diverse fungi (mycobiota), and fungi have long been suspected in the pathogenesis of SpA. Our study unraveled a disease-specific interkingdom network alteration in AS, suggesting that fungi, or the interkingdom interactions between bacteria and fungi, may play an essential role in AS development. However, our study is limited by sample size, and in-depth mechanism studies and additional large-scale investigations characterizing the gut mycobiome in AS patients are needed to form a foundation for research into the relationship between mycobiota dysbiosis and AS development. Intestinal bacterial dysbiosis has been increasingly linked to ankylosing spondylitis (AS), which is a prototypic and best studied subtype of spondyloarthritis (SpA). Fungi and bacteria coexist in the human gut and interact with each other. Although they have been shown to contribute actively to health or disease, no studies have investigated whether the fungal microbiota in AS patients is perturbed. In this study, fecal samples from 22 AS patients, with clinical and radiographic assessments, and 16 healthy controls (HCs) were collected to systematically characterize the gut microbiota and mycobiota in AS patients by 16S rRNA gene- and ITS2-based DNA sequencing. Our results showed that the microbiota of AS patients was characterized by increased abundance of Proteobacteria and decreased Bacteroidetes, which was contributed by enrichment of Escherichia-Shigella, Veillonella, Lachnospiraceae NK4A136 group, and reduction of Prevotella strain 9, Megamona, and Fusobacterium. The gut mycobiota of AS patients was characterized by higher levels of Ascomycota, especially the class of Dothideomycetes, and decreased abundance of Basidiomycota, which was mainly contributed by the decease of Agaricales. Compared to HCs, decreased ITS2/16S biodiversity ratios and altered bacterial-fungal interkingdom networks were observed in AS patients. Compared with nonsteroidal anti-inflammatory drugs (NSAIDs), treating AS patients with biological agents induced obvious changes in the gut mycobiota, and this result was highly associated with disease activity indexes, including AS disease activity index (ASDAS) C-reactive protein (asCRP), erythrocyte sedimentation rate (ESR), and Bath AS disease activity index (BASDAI). In addition, altered mycobiota in AS patients was also found associated with the degree of radiographic damage. IMPORTANCE The human gut is colonized by diverse fungi (mycobiota), and fungi have long been suspected in the pathogenesis of SpA. Our study unraveled a disease-specific interkingdom network alteration in AS, suggesting that fungi, or the interkingdom interactions between bacteria and fungi, may play an essential role in AS development. However, our study is limited by sample size, and in-depth mechanism studies and additional large-scale investigations characterizing the gut mycobiome in AS patients are needed to form a foundation for research into the relationship between mycobiota dysbiosis and AS development.
Collapse
|
105
|
Kwon OC, Lee EJ, Lee JY, Youn J, Kim TH, Hong S, Lee CK, Yoo B, Robinson WH, Kim YG. Prefoldin 5 and Anti-prefoldin 5 Antibodies as Biomarkers for Uveitis in Ankylosing Spondylitis. Front Immunol 2019; 10:384. [PMID: 30891043 PMCID: PMC6411661 DOI: 10.3389/fimmu.2019.00384] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/14/2019] [Indexed: 01/31/2023] Open
Abstract
Objective: Uveitis is the most common extra-articular manifestation of ankylosing spondylitis (AS), for which no diagnostic biomarkers have been identified. This study was conducted to identify biomarker for uveitis in AS. Methods: To identify autoantibodies associated with uveitis in AS, we performed human protein microarray analysis using sera derived from various autoimmune diseases and ELISA analysis of sera derived from AS and rheumatoid arthritis patients. In the curdlan-induced SKG mice model, ophthalmic examination was performed at week 8 post-immunization and histologic examination of the ocular lesions performed at week 16 post-immunization. Serum levels of target antibodies were assessed at various time-points. To evaluate the functional role of specific autoantibodies, an in vitro apoptosis assay using ARPE-19 cells was performed. Results: Reactivity against prefoldin subunit 5 (PFDN5) was identified in AS with uveitis. Levels of anti-PFDN5 antibodies and PFDN5 in sera from AS with uveitis patients were significantly higher than those in AS without uveitis. At week 8, half of curdlan-treated SKG mice developed anterior uveitis, while all of them developed histologically confirmed uveitis at week 16. The levels of anti-PFDN5 antibodies increased over time in the sera of curdlan-treated SKG mice along with increased expression of PFDN5 and apoptosis in the ocular lesions. Knockdown of PFDN5 in ARPE19 cells resulted in increased apoptosis, suggesting a protective role of PFDN5 against cell death in uveitis. Conclusion: AS patients with uveitis have increased levels of anti-PFDN5 antibodies, and our findings suggest that anti-PFDN5 antibodies could provide a biomarker for uveitis in AS.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Eun-Ju Lee
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.,Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Joo Yong Lee
- Department of Ophthalmology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Jeehee Youn
- Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Tae-Hwan Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| |
Collapse
|
106
|
Lynde CW, Beecker J, Dutz J, Flanagan C, Guenther LC, Gulliver W, Papp K, Rahman P, Sholter D, Searles GE. Treating to Target(s) With Interleukin-17 Inhibitors. J Cutan Med Surg 2019; 23:3S-34S. [DOI: 10.1177/1203475418824565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: The treat-to-target (T2T) strategy has become established in several medical specialties as a key guidance to optimal therapeutic decision making. T2T may be effective in the assessment of the biologic class of agents called interleukin (IL)-17 inhibitors, which are emerging as a safe and effective treatment option for autoimmune inflammatory conditions such as plaque psoriasis, psoriatic arthritis (PsA), and ankylosing spondylitis (AS). Objective: The objective of this article is to use a T2T approach for the evaluation of the effectiveness and safety of IL-17 inhibitors in the management of patients with plaque psoriasis, PsA, and AS. Methods: Following a comprehensive literature search, a full-day meeting was convened to discuss and identify the T2T targets for psoriasis, PsA, and AS. Clinical trial evidence was presented for the approved IL-17 inhibitors—secukinumab, ixekizumab, and brodalumab—to assess whether these data meet T2T safety and efficacy targets. Results: All 3 approved agents were significantly superior to placebo and active controls in the achievement of T2T targets for psoriasis. Secukinumab and ixekizumab were likewise associated with significantly better outcomes than controls in the PsA targets, and secukinumab resulted in significant AS target improvements vs placebo. The IL-17 inhibitors were also associated with low rates of serious adverse events and exacerbations of common comorbid conditions. Conclusion: Phase III trial results support the T2T benefit and safety of IL-17 inhibitors according to their specific indications for the management of patients with plaque psoriasis, PsA, and AS.
Collapse
Affiliation(s)
- Charles W. Lynde
- University of Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
- Probity Medical Research, Markham, ON, Canada
| | - Jennifer Beecker
- The Ottawa Hospital, ON, Canada
- The University of Ottawa, ON, Canada
- Probity Medical Research, Ottawa, ON, Canada
| | - Jan Dutz
- University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | | | | | - Wayne Gulliver
- Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Kim Papp
- Probity Medical Research, Waterloo, ON, Canada
| | - Proton Rahman
- Memorial University of Newfoundland, St. John’s, NL, Canada
| | | | | |
Collapse
|
107
|
Liu Y, Liao X, Shi G. Autoantibodies in Spondyloarthritis, Focusing on Anti-CD74 Antibodies. Front Immunol 2019; 10:5. [PMID: 30723468 PMCID: PMC6349765 DOI: 10.3389/fimmu.2019.00005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 01/02/2019] [Indexed: 01/03/2023] Open
Abstract
Spondyloarthritis (SpA) is an inflammatory rheumatic disease with diverse clinical presentation. The diagnosis of SpA remains a big challenge in daily clinical practice because of the limitation in specific biomarkers of SpA, more biomarkers are still needed for SpA diagnosis and disease activity monitoring. In the past, SpA was considered predominantly as auto-inflammatory disease vs. autoimmune disease. However, in recent years several researches demonstrated a broad autoantibody response in SpA patients. Study also indicated that mice lack of ZAP70 in T cell develop SpA featured inflammation. These studies indicated the autoimmune features of SpA and gave rise to the potential use of autoantibody in SpA management. In this article, we reviewed recent reports of autoantibodies associated with SpA patients, revealing the autoimmune features of SpA, suggesting the hypothesis that SpA was also an autoimmune disease, studies about the autoimmune features might provide more insights in the pathogenesis of SpA. In addition, as there are two opposite conclusions in the role of anti-CD74 autoantibody in the diagnosis of SpA, we also gave our own data on the diagnostic value of anti-CD74 in Chinese SpA patients. Though our data indicated that anti-CD74 might not be a good biomarker for SpA diagnosis in Asian people, CD74 was still a good molecule target in the research of SpA pathogenesis.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xining Liao
- Medical College, Xiamen University, Xiamen, China
| | - Guixiu Shi
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
108
|
Min HK, Choi J, Lee SY, Seo HB, Jung K, Na HS, Ryu JG, Kwok SK, Cho ML, Park SH. Protein inhibitor of activated STAT3 reduces peripheral arthritis and gut inflammation and regulates the Th17/Treg cell imbalance via STAT3 signaling in a mouse model of spondyloarthritis. J Transl Med 2019; 17:18. [PMID: 30630513 PMCID: PMC6329133 DOI: 10.1186/s12967-019-1774-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/04/2019] [Indexed: 01/03/2023] Open
Abstract
Background Spondyloarthritis (SpA) is chronic inflammatory arthritis, and interleukin (IL)-17 is crucial in SpA pathogenesis. Type 17 helper T (Th17) cells are one of major IL-17-secreting cells. Signal transducer and activator of transcription (STAT)-3 signaling induces Th17 differentiation. This study investigated the effects of protein inhibitor of activated STAT3 (PIAS3) on SpA pathogenesis. Curdlan was injected into SKG ZAP-70W163C mice for SpA induction. Methods The PIAS3 or Mock vector was inserted into mice for 10 weeks. Clinical and histologic scores of the paw, spine, and gut were evaluated. The expression of IL-17, tumor necrosis factor-α (TNF-α), STAT3, and bone morphogenic protein (BMP) was measured. Confocal microscopy and flow cytometry were used to assess Th cell differentiation. Results PIAS3 significantly diminished the histologic scores of the paw and gut. PIAS3-treated mice displayed decreased expression of IL-17, TNF-α, and STAT3 in the paw, spine, and gut. BMP-2/4 expression was lower in the spines of PIAS3-treated mice. Th cell differentiation was polarized toward the upregulation of regulatory T cells (Tregs) and the downregulation of Th17 in PIAS3-treated mice. Conclusion PIAS3 had beneficial effects in mice with SpA by reducing peripheral arthritis and gut inflammation. Pro-inflammatory cytokines and Th17/Treg differentiation were controlled by PIAS3. In addition, BMPs were decreased in the spines of PIAS3-treated mice. These findings suggest that PIAS3 could have therapeutic benefits in patients with SpA.
Collapse
Affiliation(s)
- Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 137-070, South Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - JeongWon Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Seon-Yeong Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Hyeon-Beom Seo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - KyungAh Jung
- Impact Biotech, Korea 505 Banpo-Dong, Seocho-Ku, Seoul, 137-040, South Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea.,Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Geol Ryu
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 137-070, South Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea. .,Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,Rheumatism Research Center, Catholic Institutes of Medical Science, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 137-701, South Korea.
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 137-070, South Korea. .,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea. .,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 137-701, South Korea.
| |
Collapse
|
109
|
Chen Y, Ouyang J, Yan R, Maarouf MH, Wang X, Chen B, Liu S, Hu J, Guo G, Zhang J, Dai SM, Xu H, Chen JL. Silencing SOCS3 Markedly Deteriorates Spondyloarthritis in Mice Induced by Minicircle DNA Expressing IL23. Front Immunol 2018; 9:2641. [PMID: 30487798 PMCID: PMC6246747 DOI: 10.3389/fimmu.2018.02641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/26/2018] [Indexed: 12/23/2022] Open
Abstract
Objective: Despite extensive studies, the precise mechanism underlying spondyloarthritis, especially ankylosing spondylitis, remains elusive. This study aimed to develop an ideal animal model for an insight into mechanism of spondyloarthritis and functional relevance of SOCS3 in spondyloarthritis. Methods: Since SOCS3 is a major regulator of IL23-STAT3 signaling, we generated SOCS3 knockdown transgenic (TG) mice for development of an animal model of spondyloarthritis. A hydrodynamic delivery method was employed to deliver minicircle DNA expressing IL23 (mc-IL23) into wild-type (WT) and the TG mice. Knockdown/overexpression systems mediated by lentivirus and retrovirus were used to determine whether SOCS3 regulated osteoblast differentiation. Results: Forced expression of IL23 induced severe joint destruction and extensive bone loss in SOCS3 knockdown TG mice, while this treatment only caused moderate symptoms in WT mice. Furthermore, severe spondyloarthritis was found in IL23-injected TG mice as compared to mild disease observed in WT controls under same condition. Moreover, our studies showed that IL23 promoted osteoblast differentiation via activation of STAT3 pathway and disruption of SOCS3 expression greatly increased phosphorylation of STAT3. In addition, silencing SOCS3 resulted in enhanced osteoblast differentiation through activation of Smad1/5/9 signaling, as evidenced by elevated phosphorylation level of Smad1/5/9. Experiments further demonstrated that SOCS3 interacted with Smad1 and thus suppressed the BMP2-Smad signaling. Conclusions: The results reveal that SOCS3 is involved in IL23-induced spondyloarthritis and acts as a key regulator of osteoblast differentiation, and suggest that SOCS3 knockdown TG mice may be an ideal animal model for further studies of spondyloarthritis.
Collapse
Affiliation(s)
- Yuhai Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jing Ouyang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Ruoxiang Yan
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mohamed Hassan Maarouf
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, University of Chinese Academy of Sciences, Beijing, China.,International College, University of Chinese Academy of Sciences, Beijing, China
| | - Xuefei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, University of Chinese Academy of Sciences, Beijing, China
| | - Biao Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, University of Chinese Academy of Sciences, Beijing, China
| | - Shasha Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiayue Hu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Guijie Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jing Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Sheng-Ming Dai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University Hospital, Shanghai, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
110
|
Jeong H, Bae EK, Kim H, Lim DH, Chung TY, Lee J, Jeon CH, Koh EM, Cha HS. Spondyloarthritis features in zymosan-induced SKG mice. Joint Bone Spine 2018; 85:583-591. [DOI: 10.1016/j.jbspin.2017.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022]
|
111
|
Napier RJ, Lee EJ, Vance EE, Snow PE, Samson KA, Dawson CE, Moran AE, Stenzel P, Davey MP, Sakaguchi S, Rosenzweig HL. Nod2 Deficiency Augments Th17 Responses and Exacerbates Autoimmune Arthritis. THE JOURNAL OF IMMUNOLOGY 2018; 201:1889-1898. [PMID: 30150283 DOI: 10.4049/jimmunol.1700507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
Arthritis in a genetically susceptible SKG strain of mice models a theoretical paradigm wherein autoimmune arthritis arises because of interplay between preexisting autoreactive T cells and environmental stimuli. SKG mice have a point mutation in ZAP-70 that results in attenuated TCR signaling, altered thymic selection, and spontaneous production of autoreactive T cells that cause arthritis following exposure to microbial β-glucans. In this study, we identify Nod2, an innate immune receptor, as a critical suppressor of arthritis in SKG mice. SKG mice deficient in Nod2 (Nod2-/-SKG) developed a dramatically exacerbated form of arthritis, which was independent of sex and microbiota, but required the skg mutation in T cells. Worsened arthritis in Nod2-/-SKG mice was accompanied by expansion of Th17 cells, which to some measure coproduced TNF, GM-CSF, and IL-22, along with elevated IL-17A levels within joint synovial fluid. Importantly, neutralization of IL-17A mitigated arthritis in Nod2-/-SKG mice, indicating that Nod2-mediated protection occurs through suppression of the Th17 response. Nod2 deficiency did not alter regulatory T cell development or function. Instead, Nod2 deficiency resulted in an enhanced fundamental ability of SKG CD4+ T cells (from naive mice) to produce increased levels of IL-17 and to passively transfer arthritis to lymphopenic recipients on a single-cell level. These data reveal a previously unconsidered role for T cell-intrinsic Nod2 as an endogenous negative regulator of Th17 responses and arthritogenic T cells. Based on our findings, future studies aimed at understanding a negative regulatory function of Nod2 within autoreactive T cells could provide novel therapeutic strategies for treatment of patients with arthritis.
Collapse
Affiliation(s)
- Ruth J Napier
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Ellen J Lee
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Emily E Vance
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Paige E Snow
- Veterans Affairs Portland Health Care System, Portland, OR 97239
| | - Kimberly A Samson
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Clare E Dawson
- Veterans Affairs Portland Health Care System, Portland, OR 97239
| | - Amy E Moran
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239
| | - Peter Stenzel
- Department of Anatomic Pathology, Oregon Health & Science University, Portland, OR 97239
| | - Michael P Davey
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239.,Department of Medicine, Oregon Health & Science University, Portland, OR 97239; and
| | | | - Holly L Rosenzweig
- Veterans Affairs Portland Health Care System, Portland, OR 97239; .,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
112
|
Rosenbaum JT, Dick AD. The Eyes Have it: A Rheumatologist's View of Uveitis. Arthritis Rheumatol 2018; 70:1533-1543. [PMID: 29790291 DOI: 10.1002/art.40568] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/17/2018] [Indexed: 12/18/2022]
Abstract
Uveitis is defined as intraocular inflammation. It is an extraarticular manifestation of many forms of joint disease, which include spondyloarthritis, juvenile idiopathic arthritis, and Behçet's disease. Rheumatologists may be asked to consult on the ophthalmologic care of patients with uveitis in order to identify an associated systemic illness. Diagnoses such as spondyloarthritis, sarcoidosis, and interstitial nephritis with uveitis are frequently overlooked by referring ophthalmologists. Alternatively, rheumatologists may be asked to help manage the patient's immunosuppression, including biologic therapy, which can be required to treat a subset of patients with uveitis. This review is intended to provide rheumatologists with the necessary information to facilitate collaboration in the comanagement of patients with uveitis.
Collapse
Affiliation(s)
- James T Rosenbaum
- Oregon Health & Science University and Legacy Devers Eye Institute, Portland, Oregon
| | - Andrew D Dick
- University College London, National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital, London, UK, and University of Bristol, Bristol Eye Hospital, Bristol, UK
| |
Collapse
|
113
|
Colbert RA, Navid F, Gill T. The role of HLA-B*27 in spondyloarthritis. Best Pract Res Clin Rheumatol 2018; 31:797-815. [PMID: 30509441 DOI: 10.1016/j.berh.2018.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/20/2018] [Indexed: 02/08/2023]
Abstract
The mechanism by which HLA-B*27 predisposes to spondyloarthritis remains unresolved. Arthritogenic peptides have not been defined in humans and are not involved in experimental models of spondyloarthritis. Aberrant properties of HLA-B*27 can activate the IL-23/IL-17 axis in HLA-B*27 transgenic rats and humans. In HLA-B*27-independent rodent models, spondyloarthritis can be driven by IL-23 triggering entheseal-resident CD4-/CD8- T cells or CD4+ Th17 T cells. These findings point toward noncanonical mechanisms linking HLA-B*27 to the disease and provide a potential explanation for HLA-B*27-negative spondyloarthritis. Gut microbial dysbiosis may be important in the development of spondyloarthritis. HLA-B*27-induced changes in gut microbiota are complex and suggest an ecological model of dysbiosis in rodents. The importance of the IL-23/IL-17 axis in ankylosing spondylitis has been demonstrated by studies showing efficacy of IL-17. Although deciphering the precise role(s) of HLA-B*27 in disease requires further investigation, considerable progress has been made in understanding this complex relationship.
Collapse
Affiliation(s)
- Robert A Colbert
- Pediatric Translational Research Branch, NIAMS Intramural Research Program, NIH, USA.
| | - Fatemeh Navid
- Pediatric Translational Research Branch, NIAMS Intramural Research Program, NIH, USA.
| | - Tejpal Gill
- Pediatric Translational Research Branch, NIAMS Intramural Research Program, NIH, USA.
| |
Collapse
|
114
|
van Tok MN, Na S, Lao CR, Alvi M, Pots D, van de Sande MGH, Taurog JD, Sedgwick JD, Baeten DL, van Duivenvoorde LM. The Initiation, but Not the Persistence, of Experimental Spondyloarthritis Is Dependent on Interleukin-23 Signaling. Front Immunol 2018; 9:1550. [PMID: 30038617 PMCID: PMC6046377 DOI: 10.3389/fimmu.2018.01550] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/22/2018] [Indexed: 12/30/2022] Open
Abstract
IL-17A is a central driver of spondyloarthritis (SpA), its production was originally proposed to be IL-23 dependent. Emerging preclinical and clinical evidence suggests, however, that IL-17A and IL-23 have a partially overlapping but distinct biology. We aimed to assess the extent to which IL-17A-driven pathology is IL-23 dependent in experimental SpA. Experimental SpA was induced in HLA-B27/Huβ2m transgenic rats, followed by prophylactic or therapeutic treatment with an anti-IL23R antibody or vehicle control. Spondylitis and arthritis were scored clinically and hind limb swelling was measured. Draining lymph node cytokine expression levels were analyzed directly ex vivo, and IL-17A protein was measured upon restimulation with PMA/ionomycin. Prophylactic treatment with anti-IL23R completely protected against the development of both spondylitis and arthritis, while vehicle-treated controls did develop spondylitis and arthritis. In a therapeutic study, anti-IL23R treatment failed to reduce the incidence or decrease the severity of experimental SpA. Mechanistically, expression of downstream effector cytokines, including IL-17A and IL-22, was significantly suppressed in anti-IL23R versus vehicle-treated rats in the prophylactic experiments. Accordingly, the production of IL-17A upon restimulation was reduced. In contrast, there was no difference in IL-17A and IL-22 expression after therapeutic anti-IL23R treatment. Targeting the IL-23 axis during the initiation phase of experimental SpA-but not in established disease-inhibits IL-17A expression and suppresses disease, suggesting the existence of IL-23-independent IL-17A production. Whether IL-17A can be produced independent of IL-23 in human SpA remains to be established.
Collapse
Affiliation(s)
- Melissa N van Tok
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Songqing Na
- Eli Lilly and Co, San Diego, CA, United States
| | | | - Marina Alvi
- Eli Lilly and Co, San Diego, CA, United States
| | - Desirée Pots
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Marleen G H van de Sande
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Joel D Taurog
- Rheumatic Diseases Division, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
| | - Jonathon D Sedgwick
- Eli Lilly and Co, San Diego, CA, United States.,Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Dominique L Baeten
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Leonie M van Duivenvoorde
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
115
|
Quintin J. Fungal mediated innate immune memory, what have we learned? Semin Cell Dev Biol 2018; 89:71-77. [PMID: 29842944 DOI: 10.1016/j.semcdb.2018.05.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 10/01/2022]
Abstract
The binary classification of mammalian immune memory is now obsolete. Innate immune cells carry memory characteristics. The overall capacity of innate immune cells to remember and alter their responses is referred as innate immune memory and the induction of a non-specific memory resulting in an enhanced immune status is termed "trained immunity". Historically, trained immunity was first described as triggered by the human fungal pathogen Candida albicans. Since, numerous studies have accumulated and deciphered the main characteristics of trained immunity mediated by fungi and fungal components. This review aims at presenting the newly described aspect of memory in innate immunity with an emphasis on the historically fungal mediated one, covering the known molecular mechanisms associated with training. In addition, the review uncovers the numerous non-specific effect that β-glucans trigger in the context of infectious diseases and septicaemia, inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Jessica Quintin
- Immunology of Fungal Infections, Department of Mycology, Institut Pasteur, 25, rue du Docteur Roux, 75015, Paris, France.
| |
Collapse
|
116
|
Affiliation(s)
- Byron B. Au-Yeung
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Neel H. Shah
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | - Lin Shen
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, California 94143, USA;,
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, California 94143, USA;,
- Howard Hughes Medical Institute, University of California, San Francisco, California 94143, USA
| |
Collapse
|
117
|
Laurence M, Asquith M, Rosenbaum JT. Spondyloarthritis, Acute Anterior Uveitis, and Fungi: Updating the Catterall-King Hypothesis. Front Med (Lausanne) 2018; 5:80. [PMID: 29675414 PMCID: PMC5895656 DOI: 10.3389/fmed.2018.00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/09/2018] [Indexed: 12/12/2022] Open
Abstract
Spondyloarthritis is a common type of arthritis which affects mostly adults. It consists of idiopathic chronic inflammation of the spine, joints, eyes, skin, gut, and prostate. Inflammation is often asymptomatic, especially in the gut and prostate. The HLA-B*27 allele group, which presents intracellular peptides to CD8+ T cells, is by far the strongest risk factor for spondyloarthritis. The precise mechanisms and antigens remain unknown. In 1959, Catterall and King advanced a novel hypothesis explaining the etiology of spondyloarthritis: an as-yet-unrecognized sexually acquired microbe would be causing all spondyloarthritis types, including acute anterior uveitis. Recent studies suggest an unrecognized sexually acquired fungal infection may be involved in prostate cancer and perhaps multiple sclerosis. This warrants reanalyzing the Catterall-King hypothesis based on the current literature. In the last decade, many links between spondyloarthritis and fungal infections have been found. Antibodies against the fungal cell wall component mannan are elevated in spondyloarthritis. Functional polymorphisms in genes regulating the innate immune response against fungi have been associated with spondyloarthritis (CARD9 and IL23R). Psoriasis and inflammatory bowel disease, two common comorbidities of spondyloarthritis, are both strongly associated with fungi. Evidence reviewed here lends credence to the Catterall-King hypothesis and implicates a common fungal etiology in prostate cancer, benign prostatic hyperplasia, multiple sclerosis, psoriasis, inflammatory bowel disease, and spondyloarthritis. However, the evidence available at this time is insufficient to definitely confirm this hypothesis. Future studies investigating the microbiome in relation to these conditions should screen specimens for fungi in addition to bacteria. Future clinical studies of spondyloarthritis should consider antifungals which are effective in psoriasis and multiple sclerosis, such as dimethyl fumarate and nystatin.
Collapse
Affiliation(s)
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - James T Rosenbaum
- Department of Ophthalmology, Oregon Health and Science University, Portland, OR, United States.,Department of Medicine, Oregon Health and Science University, Portland, OR, United States.,Department of Cell Biology, Oregon Health and Science University, Portland, OR, United States.,Legacy Devers Eye Institute, Portland, OR, United States
| |
Collapse
|
118
|
Babaie F, Hasankhani M, Mohammadi H, Safarzadeh E, Rezaiemanesh A, Salimi R, Baradaran B, Babaloo Z. The role of gut microbiota and IL-23/IL-17 pathway in ankylosing spondylitis immunopathogenesis: New insights and updates. Immunol Lett 2018; 196:52-62. [DOI: 10.1016/j.imlet.2018.01.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/16/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
|
119
|
Smith JA. Regulation of Cytokine Production by the Unfolded Protein Response; Implications for Infection and Autoimmunity. Front Immunol 2018; 9:422. [PMID: 29556237 PMCID: PMC5844972 DOI: 10.3389/fimmu.2018.00422] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/16/2018] [Indexed: 12/14/2022] Open
Abstract
Protein folding in the endoplasmic reticulum (ER) is an essential cell function. To safeguard this process in the face of environmental threats and internal stressors, cells mount an evolutionarily conserved response known as the unfolded protein response (UPR). Invading pathogens induce cellular stress that impacts protein folding, thus the UPR is well situated to sense danger and contribute to immune responses. Cytokines (inflammatory cytokines and interferons) critically mediate host defense against pathogens, but when aberrantly produced, may also drive pathologic inflammation. The UPR influences cytokine production on multiple levels, from stimulation of pattern recognition receptors, to modulation of inflammatory signaling pathways, and the regulation of cytokine transcription factors. This review will focus on the mechanisms underlying cytokine regulation by the UPR, and the repercussions of this relationship for infection and autoimmune/autoinflammatory diseases. Interrogation of viral and bacterial infections has revealed increasing numbers of examples where pathogens induce or modulate the UPR and implicated UPR-modulated cytokines in host response. The flip side of this coin, the UPR/ER stress responses have been increasingly recognized in a variety of autoimmune and inflammatory diseases. Examples include monogenic disorders of ER function, diseases linked to misfolding protein (HLA-B27 and spondyloarthritis), diseases directly implicating UPR and autophagy genes (inflammatory bowel disease), and autoimmune diseases targeting highly secretory cells (e.g., diabetes). Given the burgeoning interest in pharmacologically targeting the UPR, greater discernment is needed regarding how the UPR regulates cytokine production during specific infections and autoimmune processes, and the relative place of this interaction in pathogenesis.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States.,Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
120
|
Cafaro G, McInnes IB. Psoriatic arthritis: tissue-directed inflammation? Clin Rheumatol 2018; 37:859-868. [DOI: 10.1007/s10067-018-4012-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/28/2022]
|
121
|
Purvis HA, Clarke F, Jordan CK, Blanco CS, Cornish GH, Dai X, Rawlings DJ, Zamoyska R, Cope AP. Protein tyrosine phosphatase PTPN22 regulates IL-1β dependent Th17 responses by modulating dectin-1 signaling in mice. Eur J Immunol 2018; 48:306-315. [PMID: 28948613 PMCID: PMC5859948 DOI: 10.1002/eji.201747092] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/24/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
A single nucleotide polymorphism within the PTPN22 gene is a strong genetic risk factor predisposing to the development of multiple autoimmune diseases. PTPN22 regulates Syk and Src family kinases downstream of immuno-receptors. Fungal β-glucan receptor dectin-1 signals via Syk, and dectin-1 stimulation induces arthritis in mouse models. We investigated whether PTPN22 regulates dectin-1 dependent immune responses. Bone marrow derived dendritic cells (BMDCs) generated from C57BL/6 wild type (WT) and Ptpn22-/- mutant mice, were pulsed with OVA323-339 and the dectin-1 agonist curdlan and co-cultured in vitro with OT-II T-cells or adoptively transferred into OT-II mice, and T-cell responses were determined by immunoassay. Dectin-1 activated Ptpn22-/- BMDCs enhanced T-cell secretion of IL-17 in vitro and in vivo in an IL-1β dependent manner. Immunoblotting revealed that compared to WT, dectin-1 activated Ptpn22-/- BMDCs displayed enhanced Syk and Erk phosphorylation. Dectin-1 activation of BMDCs expressing Ptpn22R619W (the mouse orthologue of human PTPN22R620W ) also resulted in increased IL-1β secretion and T-cell dependent IL-17 responses, indicating that in the context of dectin-1 Ptpn22R619W operates as a loss-of-function variant. These findings highlight PTPN22 as a novel regulator of dectin-1 signals, providing a link between genetically conferred perturbations of innate receptor signaling and the risk of autoimmune disease.
Collapse
Affiliation(s)
- Harriet A Purvis
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Fiona Clarke
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Christine K Jordan
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Cristina Sanchez Blanco
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Georgina H Cornish
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Xuezhi Dai
- Seattle Children's Research Institute and Departments of Pediatrics and ImmunologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - David J Rawlings
- Seattle Children's Research Institute and Departments of Pediatrics and ImmunologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Rose Zamoyska
- Institute of Immunology and Infection ResearchCentre for Immunity, Infection and EvolutionUniversity of EdinburghEdinburghUK
| | - Andrew P Cope
- Academic Department of RheumatologyCentre for Inflammation Biology and Cancer ImmunologyFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
122
|
Kaeley GS, Eder L, Aydin SZ, Gutierrez M, Bakewell C. Enthesitis: A hallmark of psoriatic arthritis. Semin Arthritis Rheum 2018; 48:35-43. [PMID: 29429762 DOI: 10.1016/j.semarthrit.2017.12.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To describe the growing importance of enthesitis in patients with psoriatic arthritis (PsA) and discuss the advantages and disadvantages of clinical and imaging methods currently used to assess enthesitis. METHODS PubMed literature searches were conducted using the terms psoriatic arthritis, entheses, enthesitis, pathology, imaging, ultrasound, magnetic resonance imaging, clinical, and indices. Articles were deemed relevant if they provided insight into the pathology, monitoring, and/or diagnosis of enthesitis in PsA, or if they discussed clinical or imaging indices used to assess enthesitis. RESULTS Enthesitis is an early manifestation of PsA that is associated with increased disease activity and reduced quality of life. A variety of clinical indices exist to assess enthesitis in PsA; however, the Leeds Enthesitis Index and Maastricht Ankylosing Spondylitis Enthesitis Score index have been the most frequently used indices in recent clinical trials. Limitations of these indices include an inability to discern structural involvement, risk of missing subclinical enthesitis, and lack of sensitivity in detecting enthesitis, especially in patients with central sensitization and/or pain amplification. Such limitations have led to the emergent importance of imaging techniques in the assessment of enthesitis. Although there have been recent advances in magnetic resonance imaging, ultrasound (US) appears to be the preferred method for detecting enthesitis because it allows for accurate assessment of the soft-tissue components of entheses and also for new bone formation. Hypoechogenicity, increased thickness of tendon insertion, calcifications, enthesophytes, erosions, and Doppler activity have been identified as important US characteristics of enthesitis. CONCLUSION Enthesitis is thought to be integrally involved in the pathogenesis of PsA and is associated with worse prognostic outcomes in patients with PsA. A validated US index with entheses that are less confounded by mechanical factors and obesity would be the most effective measure of enthesitis in PsA. As imaging techniques continue to advance, our understanding of enthesitis and its involvement in PsA will also improve.
Collapse
Affiliation(s)
- Gurjit S Kaeley
- Division of Rheumatology, University of Florida College of Medicine, Jacksonville, 653-1 West 8th St., LRC 2nd Floor L-14, Jacksonville, FL, 32209.
| | - Lihi Eder
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Sibel Z Aydin
- University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Marwin Gutierrez
- Division of Musculoskeletal and Rheumatic Diseases, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | | |
Collapse
|
123
|
Th17 cell responses in spondyloarthritis. Best Pract Res Clin Rheumatol 2017; 31:777-796. [DOI: 10.1016/j.berh.2018.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/09/2018] [Accepted: 07/17/2018] [Indexed: 01/01/2023]
|
124
|
Rahman MA, Thomas R. The SKG model of spondyloarthritis. Best Pract Res Clin Rheumatol 2017; 31:895-909. [DOI: 10.1016/j.berh.2018.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022]
|
125
|
Jethwa H, Abraham S. The evidence for microbiome manipulation in inflammatory arthritis. Rheumatology (Oxford) 2017; 56:1452-1460. [PMID: 27789760 DOI: 10.1093/rheumatology/kew374] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Indexed: 01/04/2023] Open
Abstract
The human body consists of millions of commensal bacteria (the microbiome), with the intestinal tract being the most prevalent site of colonization. This colonization process begins at birth, and despite numerous factors such as ageing, diet and drug use affecting the microbiome make-up, by adulthood the composition of the gut bacteria is relatively consistent across local populations. The recent advent of new scientific techniques has enabled us to explore how the microbiome affects health and, in particular, has shed light on the involvement of the microbiome in the pathogenesis of inflammatory disease. In this review we highlight the current evidence for microbiome manipulation in inflammatory arthritis in animal and human models and discuss potential therapeutics targeting the microbiome as treatment for these diseases.
Collapse
Affiliation(s)
- Hannah Jethwa
- Rheumatology Department, Wexham Park Hospital, Slough
| | - Sonya Abraham
- NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
126
|
Jeong H, Bae EK, Kim H, Eun YH, Kim IY, Kim H, Lee J, Jeon CH, Koh EM, Cha HS. Estrogen attenuates the spondyloarthritis manifestations of the SKG arthritis model. Arthritis Res Ther 2017; 19:198. [PMID: 28882159 PMCID: PMC5590166 DOI: 10.1186/s13075-017-1407-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/22/2017] [Indexed: 12/16/2022] Open
Abstract
Background Ankylosing spondylitis (AS) is a male-predominant disease, and radiographic evidence of damage is also more severe in males. Estrogen modulates immune-related processes such as T cell differentiation and cytokine production. This study aimed to evaluate the effect of estrogen on the disease activity of spondyloarthritis (SpA). Methods The effects of estrogen on the development of arthritis were evaluated by performing ovariectomy and 17β-estradiol (E2) pellet implantation in zymosan-treated SKG mice. Clinical arthritis scores were measured, and 18F-fluorodeoxyglucose (18F-FDG) small-animal positron emission tomography/computed tomography performed to quantify joint inflammation. The expression of inflammatory cytokines in joint tissue was measured. Results E2-treated mice showed remarkable suppression of arthritis clinically and little infiltration of inflammatory cells in the Achilles tendon and intervertebral disc. 18F-FDG uptake was significantly lower in E2-treated mice than in sham-operated (sham) and ovariectomized mice. Expression of TNF, interferon-γ, and IL-17A was significantly reduced in E2-treated mice, whereas expression of sclerostin and Dickkopf-1 was increased in E2-treated mice compared with sham and ovariectomized mice. Conclusions Estrogen suppressed arthritis development in SKG mice, a model of SpA. Results of this study suggest that estrogen has an anti-inflammatory effect on the spondyloarthritis manifestations of the SKG arthritis model.
Collapse
Affiliation(s)
- Hyemin Jeong
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Hospital, Bucheon, South Korea
| | - Eun-Kyung Bae
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Hunnyun Kim
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Yeong Hee Eun
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - In Young Kim
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hyungjin Kim
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Jaejoon Lee
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Chan Hong Jeon
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Hospital, Bucheon, South Korea
| | - Eun-Mi Koh
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hoon-Suk Cha
- Division of Rheumatology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
127
|
van Tok MN, Satumtira N, Dorris M, Pots D, Slobodin G, van de Sande MG, Taurog JD, Baeten DL, van Duivenvoorde LM. Innate Immune Activation Can Trigger Experimental Spondyloarthritis in HLA-B27/Huβ2m Transgenic Rats. Front Immunol 2017; 8:920. [PMID: 28824645 PMCID: PMC5545590 DOI: 10.3389/fimmu.2017.00920] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/20/2017] [Indexed: 01/06/2023] Open
Abstract
Spondyloarthritis (SpA) does not display the typical features of auto-immune disease. Despite the strong association with MHC class I, CD8+ T cells are not required for disease induction in the HLA-B27/Huβ2m transgenic rats. We used Lewis HLA-B27/Huβ2m transgenic rats [21-3 × 283-2]F1, HLA-B7/Huβ2m transgenic rats [120-4 × 283-2]F1, and wild-type rats to test our hypothesis that SpA may be primarily driven by the innate immune response. In vitro, splenocytes were stimulated with heat-inactivated Mycobacterium tuberculosis and cytokine expression and production was measured. In vivo, male and female rats were immunized with 30, 60, or 90 µg of heat-inactivated M. tuberculosis and clinically monitored for spondylitis and arthritis development. After validation of the model, we tested whether prophylactic and therapeutic TNF targeting affected spondylitis and arthritis. In vitro stimulation with heat-inactivated M. tuberculosis strongly induced gene expression of pro-inflammatory cytokines such as TNF, IL-6, IL-1α, and IL-1β, in the HLA-B27 transgenic rats compared with controls. In vivo immunization induced an increased spondylitis and arthritis incidence and an accelerated and synchronized onset of spondylitis and arthritis in HLA-B27 transgenic males and females. Moreover, immunization overcame the protective effect of orchiectomy. Prophylactic TNF targeting resulted in delayed spondylitis and arthritis development and reduced arthritis severity, whereas therapeutic TNF blockade did not affect spondylitis and arthritis severity. Collectively, these data indicate that innate immune activation plays a role in the initiation of HLA-B27-associated disease and allowed to establish a useful in vivo model to study the cellular and molecular mechanisms of disease initiation and progression.
Collapse
Affiliation(s)
- Melissa N van Tok
- Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Nimman Satumtira
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Martha Dorris
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Desirée Pots
- Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Gleb Slobodin
- Internal Medicine, Bnai Zion Medical Center, Haifa, Israel
| | - Marleen G van de Sande
- Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joel D Taurog
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Dominique L Baeten
- Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Leonie M van Duivenvoorde
- Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
128
|
Zhang JR, Pang DD, Tong Q, Liu X, Su DF, Dai SM. Different Modulatory Effects of IL-17, IL-22, and IL-23 on Osteoblast Differentiation. Mediators Inflamm 2017; 2017:5950395. [PMID: 28831209 PMCID: PMC5555000 DOI: 10.1155/2017/5950395] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/04/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To examine the expressions of IL-17, IL-22, and IL-23 receptors in four osteoblast models and the effects of IL-17, IL-22, and IL-23 on osteoblasts. METHODS Gene expression levels of receptors, alkaline phosphatase (ALP), osteocalcin (OCN), and Runt-related transcription factor 2 (Runx-2), were evaluated by RT-PCR and real-time RT-PCR. Proliferative responses and cell cycle analysis were detected by a CCK-8 assay and flow cytometry, respectively. ALP activity and ALP mass were detected by an ALP activity assay and ALP staining, respectively. RESULTS In primary osteoblasts, only the IL-17 receptor was expressed. In C2C12, MC3T3-E1, and Saos-2 cells, the genes of IL-17, IL-22, and IL-23 receptors were not detectable. None of IL-17, IL-22, and IL-23 had an obvious effect on the proliferation of primary osteoblasts, but IL-17 exhibited an inhibitory effect on the gene expression of ALP, OCN, and Runx-2. The ALP activity and ALP mass of primary osteoblasts were downregulated by IL-17 treatment in a dose-dependent manner, and IL-17 failed to inhibit BMP-2-induced phosphorylation of Smad. CONCLUSION Primary osteoblasts constitutively express IL-17 receptors, but none of C2C12 cells, MC3T3-E1 cells, and Saos-2 cells express any receptors for IL-17, IL-22, and IL-23. IL-17 inhibits BMP-2-induced osteoblast differentiation via the BMP/Smad-independent pathway.
Collapse
Affiliation(s)
- Jing-Ru Zhang
- Department of Rheumatology & Immunology, Changhai Hospital, Second Military Medical University, Shanghai, China
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Dan-Dan Pang
- Department of Rheumatology & Immunology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qiang Tong
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xia Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Sheng-Ming Dai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
129
|
Th17 in Animal Models of Rheumatoid Arthritis. J Clin Med 2017; 6:jcm6070073. [PMID: 28753982 PMCID: PMC5532581 DOI: 10.3390/jcm6070073] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 01/04/2023] Open
Abstract
IL-17-secreting helper CD4 T cells (Th17 cells) constitute a newly identified subset of helper CD4 T cells that play a key role in the development of rheumatoid arthritis (RA) in its animal models. Recently, several models of spontaneous RA, which elucidate the mechanism of RA onset, have been discovered. These animal models shed new light on the role of Th17 in the development of autoimmune arthritis. Th17 cells coordinate inflammation and promote joint destruction, acting on various cells, including neutrophils, macrophages, synovial fibroblasts, and osteoclasts. Regulatory T cells cannot control Th17 cells under conditions of inflammation. In this review, the pathogenic role of Th17 cells in arthritis development, which was revealed by the recent animal models of RA, is discussed.
Collapse
|
130
|
Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, Hirota K, Matsushita M, Furuta Y, Narazaki M, Sakaguchi N, Kayama H, Nakamura S, Iida T, Saeki Y, Kumanogoh A, Sakaguchi S, Takeda K. Dysbiosis Contributes to Arthritis Development via Activation of Autoreactive T Cells in the Intestine. Arthritis Rheumatol 2017; 68:2646-2661. [PMID: 27333153 DOI: 10.1002/art.39783] [Citation(s) in RCA: 466] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/02/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The intestinal microbiota is involved in the pathogenesis of arthritis. Altered microbiota composition has been demonstrated in patients with rheumatoid arthritis (RA). However, it remains unclear how dysbiosis contributes to the development of arthritis. The aim of this study was to investigate whether altered composition of human intestinal microbiota in RA patients contributes to the development of arthritis. METHODS We analyzed the fecal microbiota of patients with early RA and healthy controls, using 16S ribosomal RNA-based deep sequencing. We inoculated fecal samples from RA patients and healthy controls into germ-free arthritis-prone SKG mice and evaluated the immune responses. We also analyzed whether the lymphocytes of SKG mice harboring microbiota from RA patients react with the arthritis-related autoantigen 60S ribosomal protein L23a (RPL23A). RESULTS A subpopulation of patients with early RA harbored intestinal microbiota dominated by Prevotella copri; SKG mice harboring microbiota from RA patients had an increased number of intestinal Th17 cells and developed severe arthritis when treated with zymosan. Lymphocytes in regional lymph nodes and the colon, but not the spleen, of these mice showed enhanced interleukin-17 (IL-17) responses to RPL23A. Naive SKG mouse T cells cocultured with P copri-stimulated dendritic cells produced IL-17 in response to RPL23A and rapidly induced arthritis. CONCLUSION We demonstrated that dysbiosis increases sensitivity to arthritis via activation of autoreactive T cells in the intestine. Autoreactive SKG mouse T cells are activated by dysbiotic microbiota in the intestine, causing joint inflammation. Dysbiosis is an environmental factor that triggers arthritis development in genetically susceptible mice.
Collapse
Affiliation(s)
- Yuichi Maeda
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan
| | | | - Eiji Umemoto
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan
| | | | | | - Kazuyoshi Gotoh
- Osaka University, Osaka, Japan, and Okayama University Graduate School of Medicine, Okayama, Japan
| | - Keiji Hirota
- Kyoto University, Kyoto, Japan, and Osaka University, Osaka, Japan
| | - Masato Matsushita
- National Hospital Organization Osaka Minami Medical Center, Osaka, Japan
| | - Yoki Furuta
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan
| | | | | | - Hisako Kayama
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan
| | | | | | - Yukihiko Saeki
- National Hospital Organization Osaka Minami Medical Center, Osaka, Japan
| | - Atsushi Kumanogoh
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Shimon Sakaguchi
- Japan Agency for Medical Research and Development, Tokyo, Japan, Kyoto University, Kyoto, Japan, and Osaka University, Osaka, Japan
| | - Kiyoshi Takeda
- Osaka University, Osaka, Japan, and Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
131
|
Clavel G, Boissier MC, Sigaux J, Semerano L. Developments with experimental and investigational drugs for axial spondyloarthritis. Expert Opin Investig Drugs 2017; 26:833-842. [DOI: 10.1080/13543784.2017.1337744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Gaëlle Clavel
- UMR 1125, Inserm, Bobigny, France
- Department of Internal Medicine, Fondation Rothschild, Paris, France
| | - Marie-Christophe Boissier
- UMR 1125, Inserm, Bobigny, France
- Sorbonne Paris Cité - Université Paris 13, Bobigny, France
- Service de Rhumatologie, Assistance Publique–Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, Bobigny, France
| | - Johanna Sigaux
- UMR 1125, Inserm, Bobigny, France
- Department of Rheumatology, Nice University Hospital, Nice, France
| | - Luca Semerano
- UMR 1125, Inserm, Bobigny, France
- Sorbonne Paris Cité - Université Paris 13, Bobigny, France
- Service de Rhumatologie, Assistance Publique–Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, Bobigny, France
| |
Collapse
|
132
|
Abstract
Over the past 5 years, advances in high-throughput techniques and studies involving large cohorts of patients have led to considerable advances in the identification of novel genetic associations and immune pathways involved in ankylosing spondylitis (AS). These discoveries include genes encoding cytokine receptors, transcription factors, signalling molecules and transport proteins. Although progress has been made in understanding the functions and potential pathogenic roles of some of these molecules, much work remains to be done to comprehend their complex interactions and therapeutic potential in AS. In this Review, we outline the current knowledge of AS pathogenesis, including genetic risk associations, HLA-B27-mediated pathology, perturbations in antigen-presentation pathways and the contribution of the type 3 immune response.
Collapse
|
133
|
Marchesoni A, De Marco G, Merashli M, McKenna F, Tinazzi I, Marzo-Ortega H, McGonagle DG. The problem in differentiation between psoriatic-related polyenthesitis and fibromyalgia. Rheumatology (Oxford) 2017; 57:32-40. [DOI: 10.1093/rheumatology/kex079] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Indexed: 12/19/2022] Open
|
134
|
Affiliation(s)
- Christopher T Ritchlin
- From the Allergy, Immunology, and Rheumatology Division, University of Rochester Medical School, Rochester, NY (C.T.R.); the Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD (R.A.C.); and the University Health Network (UHN) Krembil Research Institute, the Psoriatic Arthritis Program, and the Centre for Prognosis Studies in the Rheumatic Diseases - all at Toronto Western Hospital, Toronto (D.D.G.)
| | - Robert A Colbert
- From the Allergy, Immunology, and Rheumatology Division, University of Rochester Medical School, Rochester, NY (C.T.R.); the Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD (R.A.C.); and the University Health Network (UHN) Krembil Research Institute, the Psoriatic Arthritis Program, and the Centre for Prognosis Studies in the Rheumatic Diseases - all at Toronto Western Hospital, Toronto (D.D.G.)
| | - Dafna D Gladman
- From the Allergy, Immunology, and Rheumatology Division, University of Rochester Medical School, Rochester, NY (C.T.R.); the Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD (R.A.C.); and the University Health Network (UHN) Krembil Research Institute, the Psoriatic Arthritis Program, and the Centre for Prognosis Studies in the Rheumatic Diseases - all at Toronto Western Hospital, Toronto (D.D.G.)
| |
Collapse
|
135
|
The Bench-to-Bedside Story of IL-17 and the Therapeutic Efficacy of its Targeting in Spondyloarthritis. Curr Rheumatol Rep 2017; 18:33. [PMID: 27105640 DOI: 10.1007/s11926-016-0585-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
TNF-blocking biologics have revolutionized the care of patients with spondyloarthritis, a group of clinically overlapping conditions that includes ankylosing spondylitis and psoriatic arthritis. However, incomplete response rates speak to the need for alternative therapeutic approaches. Over the last decade, animal models, genetics, and translational studies have implicated the excessive production of a pro-inflammatory cytokine interleukin-17 (IL-17) along with another IL-17-promoting cytokine IL-23 in the pathogenesis of spondyloarthritis. Genome-wide studies identified disease associations with multiple genes regulating IL-23/IL-17 immune pathway activity. Direct examination of the patient blood and tissues revealed excessive IL-17 and IL-23 production by diverse cell types. Murine models both underscored the sufficiency of excess IL-23 in driving disease phenotype and predicted utility in IL-23/IL-17 pathway blockade. However, the clinical efficacy of agents such as secukinumab and ustekinumab, which block IL-17 and IL-23/IL-12 respectively, provided exciting proof of concept.
Collapse
|
136
|
Lau MC, Keith P, Costello ME, Bradbury LA, Hollis KA, Thomas R, Thomas GP, Brown MA, Kenna TJ. Genetic association of ankylosing spondylitis with TBX21 influences T-bet and pro-inflammatory cytokine expression in humans and SKG mice as a model of spondyloarthritis. Ann Rheum Dis 2017; 76:261-269. [PMID: 27125523 DOI: 10.1136/annrheumdis-2015-208677] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 04/01/2016] [Accepted: 04/09/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Ankylosing spondylitis (AS) is a highly heritable immune-mediated arthropathy. Inflammation in AS is poorly understood. TBX21 encodes T-bet, a transcription factor, lying within a locus with genome-wide significant association with AS. T-bet is implicated in innate and adaptive immunity. However, the role of T-bet in AS pathogenesis is unclear. METHODS We assessed the importance of T-bet in disease development and progression in peripheral blood mononuclear cells from 172 AS cases and 83 healthy controls carrying either risk or protective alleles of the peak AS-associated TBX21 single nucleotide polymorphism. Kinetics and localisation of T-bet expression in the SKG mouse model of spondyloarthropathy was examined, along with the impact of Tbx21 knockout on arthritis development in SKG mice. RESULTS Patients with AS had higher T-bet expression than healthy individuals, driven predominantly by natural killer and CD8+ T cells, with expression levels in CD8+ T cells completely distinguishing AS cases from healthy controls. T-bet expression was increased in AS cases carrying risk compared with protective alleles of rs11657479. In curdlan-treated SKG mice, T-bet expression increased early after disease initiation and persisted throughout the course of disease. There was marked reduction in gut and peripheral joint inflammation, and less IFNγ-producing and IL-17-producing CD8+ T cells, in Tbx21-/- compared with wild-type SKG mice. CONCLUSIONS AS-associated variants in TBX21 influence T-bet expression. T-bet+ innate and adaptive immune cells have altered IL-17 and IFNγ, and early activation marker CD69 expression than T-bet cells. This indicates that T-bet is a major component of inflammatory pathways of spondyloarthropathy in humans and mice.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- CD8-Positive T-Lymphocytes/immunology
- Case-Control Studies
- Cytokines/biosynthesis
- Female
- Gene Expression Regulation/physiology
- Genetic Predisposition to Disease
- Genotype
- Humans
- Inflammation Mediators/metabolism
- Killer Cells, Natural/immunology
- Lymph Nodes/immunology
- Male
- Mice, Inbred BALB C
- Mice, Knockout
- Middle Aged
- Polymorphism, Single Nucleotide
- Spondylitis, Ankylosing/genetics
- Spondylitis, Ankylosing/immunology
- Spondylitis, Ankylosing/pathology
- T-Box Domain Proteins/biosynthesis
- T-Box Domain Proteins/genetics
- Young Adult
- T-bet Transcription Factor
Collapse
Affiliation(s)
- Max C Lau
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Patricia Keith
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Mary-Ellen Costello
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Linda A Bradbury
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kelly A Hollis
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Gethin P Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Matthew A Brown
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Tony J Kenna
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
137
|
Ridley A, Hatano H, Wong-Baeza I, Shaw J, Matthews KK, Al-Mossawi H, Ladell K, Price DA, Bowness P, Kollnberger S. Activation-Induced Killer Cell Immunoglobulin-like Receptor 3DL2 Binding to HLA-B27 Licenses Pathogenic T Cell Differentiation in Spondyloarthritis. Arthritis Rheumatol 2016; 68:901-14. [PMID: 26841353 PMCID: PMC4855641 DOI: 10.1002/art.39515] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 11/12/2015] [Indexed: 12/30/2022]
Abstract
Objective In thespondyloarthritides (SpA), increased numbers of CD4+ T cells express killer cell immunoglobulin‐like receptor 3DL2 (KIR‐3DL2). The aim of this study was to determine the factors that induce KIR‐3DL2 expression, and to characterize the relationship between HLA–B27 and the phenotype and function of KIR‐3DL2–expressing CD4+ T cells in SpA. Methods In total, 34 B27+patients with SpA, 28 age‐ and sex‐matched healthy controls (20 B27− and 8 B27+), and 9 patients with rheumatoid arthritis were studied. KIR-3DL2 expression and other phenotypic characteristics of peripheral blood and synovial fluid CD4+ T cells were studied by flow cytometry, quantitative polymerase chain reaction, and Western blotting. T cell receptor clonality was determined by template‐switch anchored reverse transcription–polymerase chain reaction and sequencing analysis. Cytokines were measured by enzyme‐linked immunosorbent assay. Results Cellular activation induced KIR‐3DL2 expression on both naive and effector CD4+ T cells. KIR‐3DL2 binding to B27+ cells promoted expression of KIR‐3DL2, the Th17‐specific transcription factor retinoic acid receptor–related orphan nuclear receptor γt, and the antiapoptotic factor B cell lymphoma 2. KIR‐3DL2+CD4+ T cells in patients with ankylosing spondylitis were oligoclonal and enriched for markers of T cell activation and for the gut homing receptor CCR9. In the presence of B27+ antigen‐presenting cells, KIR‐3DL2+CD4+ T cells produced less interleukin‐2 (IL‐2) but more IL‐17. This effect was blocked by HC10, an antibody that inhibits the binding of KIR‐3DL2 to B27 heavy chains. Conclusion KIR‐3DL2 binding to HLA–B27 licenses Th17 cell differentiation in SpA. These findings raise the therapeutic potential of targeting HLA–B27–KIR‐3DL2 interactions for the treatment of B27+ patients with SpA.
Collapse
Affiliation(s)
- Anna Ridley
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Hiroko Hatano
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Isabel Wong-Baeza
- Isabel Wong-Baeza, PhD: Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK, and National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
| | - Jacqueline Shaw
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | | | - Hussein Al-Mossawi
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | | | - David A Price
- Cardiff University School of Medicine, Cardiff, UK, and National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Paul Bowness
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Simon Kollnberger
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford University, Oxford, UK, and Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
138
|
Bae JM, Choo JY, Kim KJ, Park KS. Association of inflammatory bowel disease with ankylosing spondylitis and rheumatoid arthritis: A nationwide population-based study. Mod Rheumatol 2016; 27:435-440. [PMID: 27459267 DOI: 10.1080/14397595.2016.1211229] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Tantalizing connections between autoimmune rheumatic diseases (ARDs) and inflammatory bowel disease (IBD) have become evident with regard to their genetic and immunologic background. However, the association between these two disease entities remains unclear. The aim of this study is to investigate the association between each ARD and IBD. METHODS A nationwide population-based cross-sectional study was performed using the Korean National Health Insurance Claims database. The data of patients with IBD and age- and sex-matched controls between 2009 and 2013 were collected from the database. The prevalence of ARDs, including systemic lupus erythematosus (SLE), inflammatory myositis (polymyositis and dermatomyositis), systemic sclerosis (SSc), Sjögren's syndrome (SjS), ankylosing spondylitis (AS), and rheumatoid arthritis (RA), was determined. The associations between each ARD and IBD were analyzed using multivariate logistic regression models. RESULTS A total of 40,843 IBD patients (28,197 patients with ulcerative colitis and 12,646 with Crohn's disease) and 122,529 controls were enrolled. The nonstratified analysis revealed that patients with IBD had significant risk of being concomitantly affected by AS (odds ratio [OR] 5.140, 95% confidence interval [95% CI] 4.069-6.492) and RA (OR: 3.474, 95% CI: 2.671-4.519) after adjusting for age and sex. No significant association was observed between IBD and other ARDs including SLE, inflammatory myositis, SSc, and SjS. CONCLUSION IBD is significantly associated with AS and RA in the large-scaled population-based study. This result suggests that etiopathogenesis of IBD might be shared with AS and RA.
Collapse
Affiliation(s)
| | | | - Ki-Jo Kim
- b Division of Rheumatology, Department of Internal Medicine , St. Vincent's Hospital, College of Medicine, The Catholic University of Korea , Suwon , Republic of Korea
| | - Kyung-Su Park
- b Division of Rheumatology, Department of Internal Medicine , St. Vincent's Hospital, College of Medicine, The Catholic University of Korea , Suwon , Republic of Korea
| |
Collapse
|
139
|
Forbes JD, Van Domselaar G, Bernstein CN. The Gut Microbiota in Immune-Mediated Inflammatory Diseases. Front Microbiol 2016; 7:1081. [PMID: 27462309 PMCID: PMC4939298 DOI: 10.3389/fmicb.2016.01081] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/28/2016] [Indexed: 12/17/2022] Open
Abstract
The collection of microbes and their genes that exist within and on the human body, collectively known as the microbiome has emerged as a principal factor in human health and disease. Humans and microbes have established a symbiotic association over time, and perturbations in this association have been linked to several immune-mediated inflammatory diseases (IMID) including inflammatory bowel disease, rheumatoid arthritis, and multiple sclerosis. IMID is a term used to describe a group of chronic, highly disabling diseases that affect different organ systems. Though a cornerstone commonality between IMID is the idiopathic nature of disease, a considerable portion of their pathobiology overlaps including epidemiological co-occurrence, genetic susceptibility loci and environmental risk factors. At present, it is clear that persons with an IMID are at an increased risk for developing comorbidities, including additional IMID. Advancements in sequencing technologies and a parallel explosion of 16S rDNA and metagenomics community profiling studies have allowed for the characterization of microbiomes throughout the human body including the gut, in a myriad of human diseases and in health. The main challenge now is to determine if alterations of gut flora are common between IMID or, if particular changes in the gut community are in fact specific to a single disease. Herein, we review and discuss the relationships between the gut microbiota and IMID.
Collapse
Affiliation(s)
- Jessica D. Forbes
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, WinnipegMB, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, WinnipegMB, Canada
| | - Gary Van Domselaar
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, WinnipegMB, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, WinnipegMB, Canada
| | - Charles N. Bernstein
- Department of Internal Medicine and the IBD Clinical and Research Centre, University of Manitoba, WinnipegMB, Canada
| |
Collapse
|
140
|
Min HK, Kim JK, Lee SY, Kim EK, Lee SH, Lee J, Kwok SK, Cho ML, Park SH. Rebamipide prevents peripheral arthritis and intestinal inflammation by reciprocally regulating Th17/Treg cell imbalance in mice with curdlan-induced spondyloarthritis. J Transl Med 2016; 14:190. [PMID: 27350608 PMCID: PMC4924325 DOI: 10.1186/s12967-016-0942-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/11/2016] [Indexed: 12/25/2022] Open
Abstract
Background Spondyloarthritis (SpA) usually manifests as arthritis of the axial and peripheral joints but can also result in extra-articular manifestations such as inflammatory bowel disease. Proinflammatory cytokine interleukin-17 (IL-17) plays a crucial role in the pathogenesis of SpA. Rebamipide inhibits signal transducer and activator of transcription 3 that controls IL-17 production and Th17 cell differentiation. This study examined the effect of rebamipide on SpA development. Methods SKG ZAP-70W163C mice were immunized with curdlan to induce SpA features. The mice were then intraperitoneally injected with rebamipide or vehicle 3 times a week for 14 weeks and their clinical scores were evaluated. Histological scores of the paw and spine and the length of the gut were measured at sacrifice. Immunohistochemical staining of IL-17 and tumor necrosis factor-α (TNF-α) was performed using tissue samples isolated from the axial joints, peripheral joints, and gut. Spleen tissue samples were isolated from both rebamipide- or vehicle-treated mice with SpA at 14 weeks after curdlan injection to determine the effect of rebamipide on Th17 and regulatory T (Treg) cell differentiation. Results Rebamipide decreased the clinical and histological scores of the peripheral joints. The total length of the gut was preserved in rebamipide-treated mice. IL-17 and TNF-α expression in the spine, peripheral joints, and gut was lower in rebamipide-treated mice than in control mice. Th17 cell differentiation was suppressed whereas Treg cell differentiation was upregulated in the spleen of rebamipide-treated mice. Conclusion Rebamipide exerted beneficial effects in mice with SpA by preventing peripheral arthritis and intestinal inflammation and by regulating Th17/Treg cell imbalance, suggesting that it can be used as a potential therapeutic agent for treating arthritis to SpA patients.
Collapse
Affiliation(s)
- Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Jae-Kyung Kim
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Seon-Yeong Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Eun-Kyung Kim
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Seung Hoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea.,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea.
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea. .,Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, 137-040, South Korea.
| |
Collapse
|
141
|
Koenders MI, van den Berg WB. Secukinumab for rheumatology: development and its potential place in therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2069-80. [PMID: 27445458 PMCID: PMC4928657 DOI: 10.2147/dddt.s105263] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rheumatic disease is not a single disorder, but a group of more than 100 diseases that affect joints, connective tissues, and/or internal organs. Although rheumatic diseases like rheumatoid arthritis (RA), psoriatic arthritis, and ankylosing spondylitis (AS) differ in their pathogenesis and clinical presentation, the treatment of these inflammatory disorders overlaps. Non-steroid anti-inflammatory drugs are used to reduce pain and inflammation. Additional disease-modifying anti-rheumatic drugs are prescribed to slowdown disease progression, and is in RA more frequently and effectively applied than in AS. Biologicals are a relatively new class of treatments that specifically target cytokines or cells of the immune system, like tumor necrosis factor alpha inhibitors or B-cell blockers. A new kid on the block is the interleukin-17 (IL-17) inhibitor secukinumab, which has been recently approved by the US Food and Drug Administration for moderate-to-severe plaque psoriasis, psoriatic arthritis, and AS. IL-17 is a proinflammatory cytokine that has an important role in host defense, but its proinflammatory and destructive effects have also been linked to pathogenic processes in autoimmune diseases like RA and psoriasis. Animal models have greatly contributed to further insights in the potential of IL-17 blockade in autoimmune and autoinflammatory diseases, and have resulted in the development of various potential drugs targeting the IL-17 pathway. Secukinumab (AIN457) is a fully human monoclonal antibody that selectively binds to IL-17A and recently entered the market under the brand name Cosentyx(®). By binding to IL-17A, secukinumab prevents it from binding to its receptor and inhibits its ability to trigger inflammatory responses that play a role in the development of various autoimmune diseases. With secukinumab being the first in class to receive Food and Drug Administration approval, this article will further focus on this new biologic agent and review the milestones in its development and marketing.
Collapse
Affiliation(s)
- Marije I Koenders
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wim B van den Berg
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
142
|
Guerard S, Boieri M, Hultqvist M, Holmdahl R, Wing K. The SKG Mutation in ZAP-70 also Confers Arthritis Susceptibility in C57 Black Mouse Strains. Scand J Immunol 2016; 84:3-11. [DOI: 10.1111/sji.12438] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/01/2016] [Indexed: 01/08/2023]
Affiliation(s)
- S. Guerard
- Division of Medical Inflammation Research; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - M. Boieri
- Institute of Basic Medical Sciences; Faculty of Medicine; University of Oslo; Oslo Norway
| | - M. Hultqvist
- Division of Medical Inflammation Research; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
- Redoxis AB; Medicon Village Scheelevägen 2; Lund Sweden
| | - R. Holmdahl
- Division of Medical Inflammation Research; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - K. Wing
- Division of Medical Inflammation Research; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
143
|
Scher JU, Littman DR, Abramson SB. Microbiome in Inflammatory Arthritis and Human Rheumatic Diseases. Arthritis Rheumatol 2016; 68:35-45. [PMID: 26331579 DOI: 10.1002/art.39259] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/23/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Jose U Scher
- New York University School of Medicine and New York University Hospital for Joint Diseases, New York, New York
| | - Dan R Littman
- Kimmel Center for Biology and Medicine of the Skirball Institute and New York University School of Medicine, New York, New York
| | - Steven B Abramson
- New York University School of Medicine and New York University Hospital for Joint Diseases, New York, New York
| |
Collapse
|
144
|
Durham LE, Kirkham BW, Taams LS. Contribution of the IL-17 Pathway to Psoriasis and Psoriatic Arthritis. Curr Rheumatol Rep 2016. [PMID: 26209291 DOI: 10.1007/s11926-015-0529-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Investigators have accrued compelling evidence that the IL-17 pathway is central to the pathogenesis of psoriasis and psoriatic arthritis. The evidence comprises genome-wide association studies (GWAS), data from experimental murine models and findings from in vitro studies on patients' cells or tissue biopsies. More recently, the success of drugs blocking the IL-17 pathway in treating both psoriasis (PsO) and psoriatic arthritis (PsA) confirms that IL-17 is a clinically relevant therapeutic target. However, there remain many unanswered questions: is PsA simply an extension of PsO from the skin to the synovial tissue or are there differences in the underlying pathogenesis of these diseases? Which cell type represents the primary source of IL-17 in PsO and PsA? And how are these cells regulated? This review outlines the IL-17 pathway, summarises the evidence supporting its role in PsO and PsA and discusses recent data that may help to address these yet unresolved questions.
Collapse
Affiliation(s)
- L E Durham
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), Division of Immunology, Infection and Inflammatory Disease, King's College London, London, SE1 1UL, UK,
| | | | | |
Collapse
|
145
|
Vieira-Sousa E, van Duivenvoorde LM, Fonseca JE, Lories RJ, Baeten DL. Review: animal models as a tool to dissect pivotal pathways driving spondyloarthritis. Arthritis Rheumatol 2016. [PMID: 26215401 DOI: 10.1002/art.39282] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Elsa Vieira-Sousa
- University of Lisbon, Hospital de Santa Maria, and Lisbon Academic Medical Center, Lisbon, Portugal
| | - Leonie M van Duivenvoorde
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, and University of Amsterdam, Amsterdam, The Netherlands
| | - João E Fonseca
- University of Lisbon, Hospital de Santa Maria, and Lisbon Academic Medical Center, Lisbon, Portugal
| | - Rik J Lories
- KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Dominique L Baeten
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, and University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
146
|
Kehl AS, Corr M, Weisman MH. Review: Enthesitis: New Insights Into Pathogenesis, Diagnostic Modalities, and Treatment. Arthritis Rheumatol 2016; 68:312-22. [PMID: 26473401 PMCID: PMC5195265 DOI: 10.1002/art.39458] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Amy S. Kehl
- University of California at Los Angeles, Santa Monica, California
| | - Maripat Corr
- University of California at San Diego, La Jolla, California
| | | |
Collapse
|
147
|
Tseng HW, Pitt ME, Glant TT, McRae AF, Kenna TJ, Brown MA, Pettit AR, Thomas GP. Inflammation-driven bone formation in a mouse model of ankylosing spondylitis: sequential not parallel processes. Arthritis Res Ther 2016; 18:35. [PMID: 26831337 PMCID: PMC4734853 DOI: 10.1186/s13075-015-0805-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/28/2015] [Indexed: 01/21/2023] Open
Abstract
Background Ankylosing spondylitis (AS) is an immune-mediated arthritis particularly targeting the spine and pelvis and is characterised by inflammation, osteoproliferation and frequently ankylosis. Current treatments that predominately target inflammatory pathways have disappointing efficacy in slowing disease progression. Thus, a better understanding of the causal association and pathological progression from inflammation to bone formation, particularly whether inflammation directly initiates osteoproliferation, is required. Methods The proteoglycan-induced spondylitis (PGISp) mouse model of AS was used to histopathologically map the progressive axial disease events, assess molecular changes during disease progression and define disease progression using unbiased clustering of semi-quantitative histology. PGISp mice were followed over a 24-week time course. Spinal disease was assessed using a novel semi-quantitative histological scoring system that independently evaluated the breadth of pathological features associated with PGISp axial disease, including inflammation, joint destruction and excessive tissue formation (osteoproliferation). Matrix components were identified using immunohistochemistry. Results Disease initiated with inflammation at the periphery of the intervertebral disc (IVD) adjacent to the longitudinal ligament, reminiscent of enthesitis, and was associated with upregulated tumor necrosis factor and metalloproteinases. After a lag phase, established inflammation was temporospatially associated with destruction of IVDs, cartilage and bone. At later time points, advanced disease was characterised by substantially reduced inflammation, excessive tissue formation and ectopic chondrocyte expansion. These distinct features differentiated affected mice into early, intermediate and advanced disease stages. Excessive tissue formation was observed in vertebral joints only if the IVD was destroyed as a consequence of the early inflammation. Ectopic excessive tissue was predominantly chondroidal with chondrocyte-like cells embedded within collagen type II- and X-rich matrix. This corresponded with upregulation of mRNA for cartilage markers Col2a1, sox9 and Comp. Osteophytes, though infrequent, were more prevalent in later disease. Conclusions The inflammation-driven IVD destruction was shown to be a prerequisite for axial disease progression to osteoproliferation in the PGISp mouse. Osteoproliferation led to vertebral body deformity and fusion but was never seen concurrent with persistent inflammation, suggesting a sequential process. The findings support that early intervention with anti-inflammatory therapies will be needed to limit destructive processes and consequently prevent progression of AS. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0805-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsu-Wen Tseng
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Miranda E Pitt
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Tibor T Glant
- Section of Molecular Medicine, Department of Orthopedic Surgery, Rush University Medical Center, 1735 W. Harrison Street, Cohn Research Building, Chicago, IL, 60612, USA.
| | - Allan F McRae
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia. .,The University of Queensland, Queensland Brain Institute, QBI Building, St Lucia, QLD, 4072, Australia.
| | - Tony J Kenna
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Matthew A Brown
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Allison R Pettit
- The University of Queensland-Mater Research Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Gethin P Thomas
- The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
148
|
Park R, Kim TH, Ji JD. Gene Expression Profile in Patients with Axial Spondyloarthritis: Meta-analysis of Publicly Accessible Microarray Datasets. JOURNAL OF RHEUMATIC DISEASES 2016. [DOI: 10.4078/jrd.2016.23.6.363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Robin Park
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Tae-Hwan Kim
- Division of Rheumatology, Department of Internal Medicine, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Jong Dae Ji
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
149
|
Kabeerdoss J, Sandhya P, Danda D. Gut inflammation and microbiome in spondyloarthritis. Rheumatol Int 2015; 36:457-68. [PMID: 26719306 DOI: 10.1007/s00296-015-3414-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022]
Abstract
Spondyloarthritis (SpA) is chronic inflammatory disease involving joints and the spine. Bowel inflammation is common in SpA, which may be classified as acute or chronic. Chronic gut inflammation is most common in SpA patients with axial involvement as compared to those presenting with peripheral involvement alone. The pathogenesis of gut inflammation in SpA could be explained by two factors-over-activation of immunological cells and altered gut microbiome. This is exemplified by SpA animal models, namely HLA-B27-expressing transgenic animals and SKG mice models. Immunological mechanisms include homing of activated T cells from gut into synovium, excess pro-inflammatory cytokines secretion by immune cells such as IL-23 and genetic variations in immunological genes. The evidence for role of gut microbiome in SpA is gradually emerging. Recently, metagenomic study of gut microbiome by sequencing of microbial nucleic acids has enabled identification of new microbial taxa and their functions in gut of patients with SpA. In SpA, the gut microbiome could emerge as diagnostic and prognostic marker of disease. Modulation of gut microbiome is slated to have therapeutic potential as well.
Collapse
Affiliation(s)
- Jayakanthan Kabeerdoss
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Pulukool Sandhya
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India.
| |
Collapse
|
150
|
|