51
|
Guggino G, Mauro D, Rizzo A, Alessandro R, Raimondo S, Bergot AS, Rahman MA, Ellis JJ, Milling S, Lories R, Elewaut D, Brown MA, Thomas R, Ciccia F. Inflammasome Activation in Ankylosing Spondylitis Is Associated With Gut Dysbiosis. Arthritis Rheumatol 2021; 73:1189-1199. [PMID: 33452867 DOI: 10.1002/art.41644] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE We undertook this study to evaluate the activation and functional relevance of inflammasome pathways in ankylosing spondylitis (AS) patients and rodent models and their relationship to dysbiosis. METHODS An inflammasome pathway was evaluated in the gut and peripheral blood from 40 AS patients using quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), flow cytometry, and confocal microscopy, and was compared to that of 20 healthy controls and 10 patients with Crohn's disease. Bacteria was visualized using silver stain in human samples, and antibiotics were administered to HLA-B27-transgenic rats. The NLRP3 inhibitor MCC950 was administered to SKG mice, and ileal and joint tissues were assessed by IHC analysis and real-time qRT-PCR. The role of inflammasome in modulating the interleukin-23 (IL-23)/IL-17 axis was studied ex vivo. RESULTS Expression levels of Nlrp3, Nlrc4, and Aim2 were increased in the gut of HLA-B27-transgenic rats and reduced by antibiotic treatment (P < 0.05). In curdlan-treated SKG mice, NLRP3 blockade prevented ileitis and delayed arthritis onset (P < 0.05). Compared to healthy controls, AS patients demonstrated overexpression of NLRP3 (fold induction 2.33 versus 22.2; P < 0.001), NLRC4 (fold induction 1.90 versus 6.47; P < 0.001), AIM2 (fold induction 2.40 versus 20.8; P < 0.001), CASP1 (fold induction 2.53 versus 24.8; P < 0.001), IL1B (fold induction 1.07 versus 10.93; P < 0.001), and IL18 (fold induction 2.56 versus 15.67; P < 0.001) in the ileum, and caspase 1 activity was increased (P < 0.01). The score of adherent and invasive mucosa-associated bacteria was higher in AS (P < 0.01) and correlated with the expression of inflammasome components in peripheral blood mononuclear cells (P < 0.001). NLRP3 expression was associated with disease activity (the Ankylosing Spondylitis Disease Activity Score using the C-reactive protein level) (r2 = 0.28, P < 0.01) and with IL23A expression (r2 = 0.34, P < 0.001). In vitro, inflammasome activation in AS monocytes was paralleled by increased serum levels of IL-1β and IL-18. Induction of IL23A, IL17A, and IL22 was IL-1β-dependent. CONCLUSION Inflammasome activation occurs in rodent models of AS and in AS patients, is associated with dysbiosis, and is involved in triggering ileitis in SKG mice. Inflammasomes drive type III cytokine production with an IL-1β-dependent mechanism in AS patients.
Collapse
Affiliation(s)
| | - Daniele Mauro
- Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Aroldo Rizzo
- Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | | | | | - Anne-Sophie Bergot
- University of Queensland Diamantina Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - M Arifur Rahman
- University of Queensland Diamantina Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Jonathan J Ellis
- NIHR Guy's and St, Thomas' Biomedical Research Centre, London, UK
| | | | - Rik Lories
- Katholieke Universiteit Leuven, Leuven, Belgium
| | - Dirk Elewaut
- Ghent Universityand VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Matthew A Brown
- NIHR Guy's and St, Thomas' Biomedical Research Centre, London, UK
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Francesco Ciccia
- Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
52
|
Mandour M, Chen S, van de Sande MGH. The Role of the IL-23/IL-17 Axis in Disease Initiation in Spondyloarthritis: Lessons Learned From Animal Models. Front Immunol 2021; 12:618581. [PMID: 34267743 PMCID: PMC8276000 DOI: 10.3389/fimmu.2021.618581] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Spondyloarthritis (SpA) is a spectrum of chronic inflammatory joint diseases that frequently presents with inflammation of the axial skeleton, peripheral joints, entheses, skin, and gut. Understanding SpA pathogenesis has been proven challenging due to the limited availability of human target tissues. In recent years, the interleukin (IL)-23/IL-17 pathway has been implicated in the pathogenesis of SpA, in addition to the Tumor Necrosis Factor Alpha (TNF-α) cytokine. The underlying molecular mechanisms by which the IL-23/IL-17 pathway triggers disease initiation, both in the joints as well as at extra-musculoskeletal sites, are not precisely known. Animal models that resemble pathological features of human SpA have provided possibilities for in-depth molecular analyses of target tissues during various phases of the disease, including the pre-clinical initiation phase of the disease before arthritis and spondylitis are clinically present. Herein, we summarize recent insights gained in SpA animal models on the role of the IL-23/IL-17 pathway in immune activation across affected sites in SpA, which include the joint, entheses, gut and skin. We discuss how local activation of the IL-23/IL-17 axis may contribute to the development of tissue inflammation and the onset of clinically manifest SpA. The overall aim is to provide the reader with an overview of how the IL-23/IL-17 axis could contribute to the onset of SpA pathogenesis. We discuss how insights from animal studies into the initiation phase of disease could instruct validation studies in at-risk individuals and thereby provide a perspective for potential future preventive treatment.
Collapse
Affiliation(s)
- Mohamed Mandour
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sijia Chen
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Marleen G. H. van de Sande
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
53
|
Berthelot JM, Darrieutort-Laffite C, Trang C, Maugars Y, Le Goff B. Contribution of mycobiota to the pathogenesis of spondyloarthritis. Joint Bone Spine 2021; 88:105245. [PMID: 34166798 DOI: 10.1016/j.jbspin.2021.105245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
This review lists current evidences for a contribution of gut mycobiota to the pathogenesis of SpA and related conditions. Gut mycobiota has a small size as compared to bacterial microbiota, but an even greater inter- and intra-individual variability. Although most fungi (brought by food or air) are only transitory present, a core mycobiota of gut resident fungi exists, and interplays with bacteria in a complex manner. A dysbiosis of this gut mycobiota has been observed in Crohn's disease and sclerosing cholangitis, with decreased proportion of Saccharomyces cerevisiae and outgrowth of more pathogenic gut fungi. Fungal-induced lower number of commensal gut bacteria can promote translocation of some bacterial/fungal antigens through mucosae, and live fungi can also cross the epithelial border in Crohn's disease. This dysbiosis also lower the ability of bacteria to metabolize tryptophan into regulatory metabolites, consequently enhancing tryptophan metabolism within human cells, which might contribute to fatigue. Translocation of mycobiotal antigens like curdlan (beta-glucan), which plays a major role in the pathogenesis of SpA in the SGK mice, has been observed in humans. This translocation of fungal antigens in human SpA might account for the anti-Saccharomyces antibodies found in this setting. Contribution of fungal antigens to psoriasis and hidradenitis suppurativa would fit with the preferential homing of fungi in the skin area most involved in those conditions. Fungal antigens also possess autoimmune uveitis-promoting function. As genes associated with SpA (CARD9 and IL23R) strongly regulate the innate immune response against fungi, further studies on fungi contribution to SpA are needed.
Collapse
Affiliation(s)
- Jean-Marie Berthelot
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France.
| | | | - Caroline Trang
- Service de gastro-entérologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| | - Yves Maugars
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| | - Benoît Le Goff
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| |
Collapse
|
54
|
Ankylosing spondylitis: an autoimmune or autoinflammatory disease? Nat Rev Rheumatol 2021; 17:387-404. [PMID: 34113018 DOI: 10.1038/s41584-021-00625-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disorder of unknown aetiology. Unlike other systemic autoimmune diseases, in AS, the innate immune system has a dominant role characterized by aberrant activity of innate and innate-like immune cells, including γδ T cells, group 3 innate lymphoid cells, neutrophils, mucosal-associated invariant T cells and mast cells, at sites predisposed to the disease. The intestine is involved in disease manifestations, as it is at the forefront of the interaction between the mucosal-associated immune cells and the intestinal microbiota. Similarly, biomechanical factors, such as entheseal micro-trauma, might also be involved in the pathogenesis of the articular manifestation of AS, and sentinel immune cells located in the entheses could provide links between local damage, genetic predisposition and the development of chronic inflammation. Although these elements might support the autoinflammatory nature of AS, studies demonstrating the presence of autoantibodies (such as anti-CD74, anti-sclerostin and anti-noggin antibodies) and evidence of activation and clonal expansion of T cell populations support an autoimmune component to the disease. This Review presents the evidence for autoinflammation and the evidence for autoimmunity in AS and, by discussing the pathophysiological factors associated with each, aims to reconcile the two hypotheses.
Collapse
|
55
|
Selective estrogen receptor modulator lasofoxifene suppresses spondyloarthritis manifestation and affects characteristics of gut microbiota in zymosan-induced SKG mice. Sci Rep 2021; 11:11923. [PMID: 34099783 PMCID: PMC8184804 DOI: 10.1038/s41598-021-91320-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022] Open
Abstract
Ankylosing spondylitis is a male-predominant disease and previous study revealed that estrogens have an anti-inflammatory effect on the spondyloarthritis (SpA) manifestations in zymosan-induced SKG mice. This study aimed to evaluate the effect of selective estrogen receptor modulator (SERM) lasofoxifene (Laso) on disease activity of SpA. Mice were randomized into zymosan-treated, zymosan + 17β-estradiol (E2)-treated, and zymosan + Laso-treated groups. Arthritis was assessed by 18F-fluorodeoxyglucose (18F-FDG) small-animal positron emission tomography/computed tomography and bone mineral density (BMD) was measured. Fecal samples were collected and 16S ribosomal RNA gene sequencing was used to determine gut microbiota differences. Both zymosan + E2-treated mice and zymosan + Laso-treated mice showed lower arthritis clinical scores and lower 18F-FDG uptake than zymosan-treated mice. BMD was significantly higher in zymosan + E2-treated mice and zymosan + Laso-treated mice than zymosan-treated mice, respectively. Fecal calprotectin levels were significantly elevated at 8 weeks after zymosan injection in zymosan-treated mice, but it was not significantly changed in zymosan + E2-treated mice and zymosan + Laso-treated mice. Gut microbiota diversity of zymosan-treated mice was significantly different from zymosan + E2-treated mice and zymosan + Laso-treated mice, respectively. There was no significant difference in gut microbiota diversity between zymosan + E2-treated mice and zymosan + Laso -treated mice. Laso inhibited joint inflammation and enhanced BMD in SKG mice, a model of SpA. Laso also affected the composition and biodiversity of gut microbiota. This study provides new knowledge regarding that selected SpA patients could benefit from SERM treatment.
Collapse
|
56
|
Furesi G, Fert I, Beaufrère M, Araujo LM, Glatigny S, Baschant U, von Bonin M, Hofbauer LC, Horwood NJ, Breban M, Rauner M. Rodent Models of Spondyloarthritis Have Decreased White and Bone Marrow Adipose Tissue Depots. Front Immunol 2021; 12:665208. [PMID: 34149700 PMCID: PMC8207134 DOI: 10.3389/fimmu.2021.665208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
Bone marrow adipose tissue (BMAT) has recently been recognized as a distinct fat depot with endocrine functions. However, if and how it is regulated by chronic inflammation remains unknown. Here, we investigate the amount of white fat and BMAT in HLA-B27 transgenic rats and curdlan-challenged SKG mice, two well-established models of chronic inflammatory spondyloarthritis (SpA). Subcutaneous and gonadal white adipose tissue and BMAT was reduced by 65-70% and by up to 90% in both experimental models. Consistently, B27 rats had a 2-3-fold decrease in the serum concentrations of the adipocyte-derived cytokines adiponectin and leptin as well as a 2-fold lower concentration of triglycerides. The bone marrow of B27 rats was further characterized by higher numbers of neutrophils, lower numbers of erythroblast precursors, and higher numbers of IL-17 producing CD4+ T cells. IL-17 concentration was also increased in the serum of B27 rats. Using a cell culture model, we show that high levels of IL-17 in the serum of B27 rats negatively impacted adipogenesis (-76%), an effect that was reversed in the presence of neutralizing anti-IL-17 antibody. In summary, these findings show BMAT is severely reduced in two experimental models of chronic inflammatory SpA and suggest that IL-17 is involved in this process.
Collapse
Affiliation(s)
- Giulia Furesi
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ingrid Fert
- Laboratoire Infection et inflammation, UMR U1173 INSERM/Université de Versailles-Saint-Quentin-Paris-Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Marie Beaufrère
- Laboratoire Infection et inflammation, UMR U1173 INSERM/Université de Versailles-Saint-Quentin-Paris-Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Luiza M Araujo
- Laboratoire Infection et inflammation, UMR U1173 INSERM/Université de Versailles-Saint-Quentin-Paris-Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Simon Glatigny
- Laboratoire Infection et inflammation, UMR U1173 INSERM/Université de Versailles-Saint-Quentin-Paris-Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Ulrike Baschant
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Malte von Bonin
- Department of Medicine I, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Nicole J Horwood
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Maxime Breban
- Laboratoire Infection et inflammation, UMR U1173 INSERM/Université de Versailles-Saint-Quentin-Paris-Saclay, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
57
|
Atzeni F, Carriero A, Boccassini L, D’Angelo S. Anti-IL-17 Agents in the Treatment of Axial Spondyloarthritis. Immunotargets Ther 2021; 10:141-153. [PMID: 33977094 PMCID: PMC8104974 DOI: 10.2147/itt.s259126] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
Axial spondyloarthritis (axSpA) describes a group of chronic inflammatory rheumatic diseases primarily involving the axial skeleton. IL-17 is involved in the pathogenesis of numerous inflammatory diseases, including inflammatory arthritis. Until a few years ago, the only biological agents licensed for the treatment of axSpA and nr-axSpA were TNF inhibitors. However, as some patients did not respond to TNF inhibition or experienced secondary failure, the introduction of the first two IL-17 inhibitors (secukinumab [SEC] and ixekizumab [IXE]) has extended the treatment options, and there are now three others (bimekizumab, brodalumab and netakimab) in various stages of clinical development. The last ten years have seen the development of a number of therapeutic recommendations that aimed at improving the management of axSpA patients. The aim of this narrative review of the published literature concerning the role of IL-17 in the pathogenesis of SpA, and the role of IL-17 inhibitors in the treatment of axSpA, is to provide a comprehensive picture of the clinical efficacy and safety of the drugs themselves, and the treatment strategies recommended in the international guidelines.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Antonio Carriero
- Rheumatology Institute of Lucania (IReL), Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
- Translational and Clinical Medicine, Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Laura Boccassini
- Rheumatology Unit, Internal Medicine Department, ASST Fatebenefratelli-Sacco, University School of Medicine, Milan, Italy
| | - Salvatore D’Angelo
- Rheumatology Institute of Lucania (IReL), Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| |
Collapse
|
58
|
Moentadj R, Wang Y, Bowerman K, Rehaume L, Nel H, O Cuiv P, Stephens J, Baharom A, Maradana M, Lakis V, Morrison M, Wells T, Hugenholtz P, Benham H, Le Cao KA, Thomas R. Streptococcus species enriched in the oral cavity of patients with RA are a source of peptidoglycan-polysaccharide polymers that can induce arthritis in mice. Ann Rheum Dis 2021; 80:573-581. [PMID: 33397732 DOI: 10.1136/annrheumdis-2020-219009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Analysis of oral dysbiosis in individuals sharing genetic and environmental risk factors with rheumatoid arthritis (RA) patients may illuminate how microbiota contribute to disease susceptibility. We studied the oral microbiota in a prospective cohort of patients with RA, first-degree relatives (FDR) and healthy controls (HC), then genomically and functionally characterised streptococcal species from each group to understand their potential contribution to RA development. METHODS After DNA extraction from tongue swabs, targeted 16S rRNA gene sequencing and statistical analysis, we defined a microbial dysbiosis score based on an operational taxonomic unit signature of disease. After selective culture from swabs, we identified streptococci by sequencing. We examined the ability of streptococcal cell walls (SCW) from isolates to induce cytokines from splenocytes and arthritis in ZAP-70-mutant SKG mice. RESULTS RA and FDR were more likely to have periodontitis symptoms. An oral microbial dysbiosis score discriminated RA and HC subjects and predicted similarity of FDR to RA. Streptococcaceae were major contributors to the score. We identified 10 out of 15 streptococcal isolates as S. parasalivarius sp. nov., a distinct sister species to S. salivarius. Tumour necrosis factor and interleukin 6 production in vitro differed in response to individual S. parasalivarius isolates, suggesting strain specific effects on innate immunity. Cytokine secretion was associated with the presence of proteins potentially involved in S. parasalivarius SCW synthesis. Systemic administration of SCW from RA and HC-associated S. parasalivarius strains induced similar chronic arthritis. CONCLUSIONS Dysbiosis-associated periodontal inflammation and barrier dysfunction may permit arthritogenic insoluble pro-inflammatory pathogen-associated molecules, like SCW, to reach synovial tissue.
Collapse
Affiliation(s)
- Rabia Moentadj
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Yiwen Wang
- School of Mathematics and Statistics, Melbourne Integrative Genomics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kate Bowerman
- Australian Centre for Ecogenomics, The University of Queensland - Saint Lucia Campus, Saint Lucia, Queensland, Australia
| | - Linda Rehaume
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Hendrik Nel
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Paraic O Cuiv
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Current address: Microba Life Sciences, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Juliette Stephens
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Amalina Baharom
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Muralidhara Maradana
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Vanessa Lakis
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Mark Morrison
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Timothy Wells
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, The University of Queensland - Saint Lucia Campus, Saint Lucia, Queensland, Australia
| | - Helen Benham
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Department of Rheumatology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Kim-Anh Le Cao
- School of Mathematics and Statistics, Melbourne Integrative Genomics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| |
Collapse
|
59
|
Olejniczak-Staruch I, Ciążyńska M, Sobolewska-Sztychny D, Narbutt J, Skibińska M, Lesiak A. Alterations of the Skin and Gut Microbiome in Psoriasis and Psoriatic Arthritis. Int J Mol Sci 2021; 22:ijms22083998. [PMID: 33924414 PMCID: PMC8069836 DOI: 10.3390/ijms22083998] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous scientific studies in recent years have shown significant skin and gut dysbiosis among patients with psoriasis. A significant decrease in microbiome alpha-diversity (abundance of different bacterial taxa measured in one sample) as well as beta-diversity (microbial diversity in different samples) was noted in psoriasis skin. It has been proven that the representation of Cutibacterium, Burkholderia spp., and Lactobacilli is decreased and Corynebacterium kroppenstedii, Corynebacterium simulans, Neisseria spp., and Finegoldia spp. increased in the psoriasis skin in comparison to healthy skin. Alterations in the gut microbiome in psoriasis are similar to those observed in patients with inflammatory bowel disease. In those two diseases, the F. prausnitzii, Bifidobacterium spp., Lactobacillus spp., Parabacteroides and Coprobacillus were underrepresented, while the abundance of Salmonella sp., Campylobacter sp., Helicobacter sp., Escherichia coli, Alcaligenes sp., and Mycobacterium sp. was increased. Several research studies provided evidence for the significant influence of psoriasis treatments on the skin and gut microbiome and a positive influence of orally administered probiotics on the course of this dermatosis. Further research is needed to determine the influence of the microbiome on the development of inflammatory skin diseases. The changes in microbiome under psoriasis treatment can serve as a potential biomarker of positive response to the administered therapy.
Collapse
Affiliation(s)
- Irmina Olejniczak-Staruch
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
- Dermoklinika Centrum Medyczne, 90-436 Lodz, Poland
- Correspondence: ; Tel.: +48-42-230-9657
| | - Magdalena Ciążyńska
- Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, 93-513 Lodz, Poland;
| | - Dorota Sobolewska-Sztychny
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Joanna Narbutt
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Małgorzata Skibińska
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Aleksandra Lesiak
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| |
Collapse
|
60
|
Swann JW, Koneva LA, Regan-Komito D, Sansom SN, Powrie F, Griseri T. IL-33 promotes anemia during chronic inflammation by inhibiting differentiation of erythroid progenitors. J Exp Med 2021; 217:151849. [PMID: 32520308 PMCID: PMC7478740 DOI: 10.1084/jem.20200164] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
An important comorbidity of chronic inflammation is anemia, which may be related to dysregulated activity of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Among HSPCs, we found that the receptor for IL-33, ST2, is expressed preferentially and highly on erythroid progenitors. Induction of inflammatory spondyloarthritis in mice increased IL-33 in BM plasma, and IL-33 was required for inflammation-dependent suppression of erythropoiesis in BM. Conversely, administration of IL-33 in healthy mice suppressed erythropoiesis, decreased hemoglobin expression, and caused anemia. Using purified erythroid progenitors in vitro, we show that IL-33 directly inhibited terminal maturation. This effect was dependent on NF-κB activation and associated with altered signaling events downstream of the erythropoietin receptor. Accordingly, IL-33 also suppressed erythropoietin-accelerated erythropoiesis in vivo. These results reveal a role for IL-33 in pathogenesis of anemia during inflammatory disease and define a new target for its treatment.
Collapse
Affiliation(s)
- James W Swann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lada A Koneva
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Stephen N Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Thibault Griseri
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
61
|
Gill T, Rosenbaum JT. Putative Pathobionts in HLA-B27-Associated Spondyloarthropathy. Front Immunol 2021; 11:586494. [PMID: 33537028 PMCID: PMC7848169 DOI: 10.3389/fimmu.2020.586494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Spondyloarthritis (SpA) is a group of immune mediated inflammatory diseases with a strong association to the major histocompatibility (MHC) class I molecule, HLA-B27. Although the association between HLA-B27 and AS has been known for almost 50 years, the mechanisms underlying disease pathogenesis are elusive. Over the years, three hypotheses have been proposed to explain HLA-B27 and disease association: 1) HLA B27 presents arthritogenic peptides and thus creates a pathological immune response; 2) HLA-B27 misfolding causes endoplasmic reticulum (ER) stress which activates the unfolded protein response (UPR); 3) HLA-B27 dimerizes on the cell surface and acts as a target for natural killer (NK) cells. None of these hypotheses explains SpA pathogenesis completely. Evidence supports the hypothesis that HLA-B27-related diseases have a microbial pathogenesis. In animal models of various SpAs, a germ-free environment abrogates disease development and colonizing these animals with gut commensal microbes can restore disease manifestations. The depth of microbial influence on SpA development has been realized due to our ability to characterize microbial communities in the gut using next-generation sequencing approaches. In this review, we will discuss various putative pathobionts in the pathogenesis of HLA-B27-associated diseases. We pursue whether a single pathobiont or a disruption of microbial community and function is associated with HLA-B27-related diseases. Furthermore, rather than a specific pathobiont, metabolic functions of various disease-associated microbes might be key. While the use of germ-free models of SpA have facilitated understanding the role of microbes in disease development, future studies with animal models that mimic diverse microbial communities instead of mono-colonization are indispensable. We discuss the causal mechanisms underlying disease pathogenesis including the role of these pathobionts on mucin degradation, mucosal adherence, and gut epithelial barrier disruption and inflammation. Finally, we review the various uses of microbes as therapeutic modalities including pre/probiotics, diet, microbial metabolites and fecal microbiota transplant. Unravelling these complex host-microbe interactions will lead to the development of new targets/therapies for alleviation of SpA and other HLA-B27 associated diseases.
Collapse
Affiliation(s)
- Tejpal Gill
- Division of Arthritis and Rheumatic Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - James T Rosenbaum
- Departments of Ophthalmology, Medicine, and Cell Biology, Oregon Health & Science University, Portland, OR, United States.,Legacy Devers Eye Institute, Portland, OR, United States
| |
Collapse
|
62
|
Rosine N, Miceli-Richard C. Innate Cells: The Alternative Source of IL-17 in Axial and Peripheral Spondyloarthritis? Front Immunol 2021; 11:553742. [PMID: 33488572 PMCID: PMC7821711 DOI: 10.3389/fimmu.2020.553742] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Spondyloarthritis (SpA) is a chronic inflammatory rheumatism characterized by inflammation of sacroiliac joints, peripheral joints, and spine. The Assessment of SpondyloArthritis Society describes three disease forms: axial (axSpA), peripheral, and enthesitic SpA. Each may be associated with extra-articular manifestations: psoriasis, inflammatory bowel disease, and acute anterior uveitis. Genome-wide association studies performed in axSpA and psoriatic arthritis (PsA) have shown a shared genetic background, especially the interleukin 23 (IL-23)/IL-17 pathway, which suggests pathophysiological similarities. The convincing positive results of clinical trials assessing the effect of secukinumab and ixekizumab (anti-IL-17A monoclonal antibodies) in axSpA and PsA have reinforced the speculated crucial role of IL-17 in SpA. Nevertheless, and obviously unexpectedly, the differential efficacy of anti-IL-23–targeted treatments between axSpA (failure) and PsA (success) has profoundly disrupted our presumed knowledge of disease pathogeny. The cells able to secrete IL-17, their dependence on IL-23, and their respective role according to the clinical form of the disease is at the heart of the current debate to potentially explain these observed differences in efficacy of IL-23/IL-17–targeted therapy. In fact, IL-17 secretion is usually mainly related to T helper 17 lymphocytes. Nevertheless, several innate immune cells express IL-23 receptor and can produce IL-17. To what extent these alternative cell populations can produce IL-17 independent of IL-23 and their respective involvement in axSpA and PsA are the crucial scientific questions in SpA. From this viewpoint, this is a nice example of a reverse path from bedside to bench, in which the results of therapeutic trials allow for reflecting more in depth on the pathophysiology of a disease. Here we provide an overview of each innate immunity-producing IL-17 cell subset and their respective role in disease pathogeny at the current level of our knowledge.
Collapse
Affiliation(s)
- Nicolas Rosine
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Immunoregulation Unit, Paris, France
| | - Corinne Miceli-Richard
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Immunoregulation Unit, Paris, France.,Paris University, Department of Rheumatology-Hôpital Cochin. Assistance Publique-Hôpitaux de Paris, EULAR Center of Excellence, Paris, France
| |
Collapse
|
63
|
Abstract
Axial spondyloarthritis (axSpA) is an inflammatory disease of the axial skeleton associated with significant pain and disability. Previously, the diagnosis of ankylosing spondylitis required advanced changes on plain radiographs of the sacroiliac joints. Classification criteria released in 2009, however, identified a subset of patients, under the age of 45, with back pain for more than three months in the absence of radiographic sacroiliitis who were classified as axSpA based on a positive magnetic resonance imaging or HLAB27 positivity and specific clinical features. This subgroup was labeled non-radiographic (nr)-axSpA. These patients, compared with those identified by the older New York criteria, contained a larger percentage of women and demonstrated less structural damage. However, their clinical manifestations and response to biologics were similar to radiographic axSpA. The discovery of the interleukin (IL) IL-23/IL-17 pathway revealed key molecules involved in the pathophysiology of axSpA. This discovery propelled the generation of antibodies directed toward IL-17A, which are highly effective and demonstrate treatment responses in axSpA that are similar to those observed with anti-TNF agents. The finding that agents that block IL-23 were not effective in axSpA came as a surprise and the potential underlying mechanisms underlying this lack of response are discussed. New agents with dual inhibition of the IL-17A and F isoforms and some oral small molecule agents that target the Jak-STAT pathway, have also shown efficacy in axSpA.
Collapse
Affiliation(s)
- Christopher Ritchlin
- Allergy, Immunology & Rheumatology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Iannis E Adamopoulos
- Rheumatology, Allergy & Clinical Immunology Division, University of California, Davis, Shriners Hospital, Sacramento, California, USA
| |
Collapse
|
64
|
Gracey E, Hromadová D, Lim M, Qaiyum Z, Zeng M, Yao Y, Srinath A, Baglaenko Y, Yeremenko N, Westlin W, Masse C, Müller M, Strobl B, Miao W, Inman RD. TYK2 inhibition reduces type 3 immunity and modifies disease progression in murine spondyloarthritis. J Clin Invest 2020; 130:1863-1878. [PMID: 32149730 PMCID: PMC7108927 DOI: 10.1172/jci126567] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/14/2020] [Indexed: 12/17/2022] Open
Abstract
Spondyloarthritis (SpA) represents a family of inflammatory diseases of the spine and peripheral joints. Ankylosing spondylitis (AS) is the prototypic form of SpA in which progressive disease can lead to fusion of the spine. Therapeutically, knowledge of type 3 immunity has translated into the development of IL-23– and IL-17A–blocking antibodies for the treatment of SpA. Despite being able to provide symptomatic control, the current biologics do not prevent the fusion of joints in AS patients. Thus, there is an unmet need for disease-modifying drugs. Genetic studies have linked the Janus kinase TYK2 to AS. TYK2 is a mediator of type 3 immunity through intracellular signaling of IL-23. Here, we describe and characterize a potentially novel small-molecule inhibitor of TYK2 that blocked IL-23 signaling in vitro and inhibited disease progression in animal models of SpA. The effect of the inhibitor appears to be TYK2 specific, using TYK2-inactive mice, which further revealed a duality in the induction of IL-17A and IL-22 by IL-23. Specifically, IL-22 production was TYK2/JAK2/STAT3 dependent, while IL-17A was mostly JAK2 dependent. Finally, we examined the effects of AS-associated TYK2 SNPs on TYK2 expression and function and correlated them with AS disease progression. This work provides evidence that TYK2 inhibitors have great potential as an orally delivered therapeutic for SpA.
Collapse
Affiliation(s)
- Eric Gracey
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Melissa Lim
- Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Zoya Qaiyum
- Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Michael Zeng
- Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Yuchen Yao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Archita Srinath
- Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Yuriy Baglaenko
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Natalia Yeremenko
- Division of Clinical Immunology and Rheumatology, Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | | | - Craig Masse
- Nimbus Therapeutics, Cambridge, Massachusetts, USA
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Wenyan Miao
- Nimbus Therapeutics, Cambridge, Massachusetts, USA
| | - Robert D Inman
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Motta F, Gershwin ME, Selmi C. Mushrooms and immunity. J Autoimmun 2020; 117:102576. [PMID: 33276307 DOI: 10.1016/j.jaut.2020.102576] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022]
Abstract
In the wide field of nutraceuticals, the effects of mushrooms on immunity, cancer and including autoimmunity have been proposed for centuries but in recent years a growing interest has led scientists to elucidate which specific compounds have bioactive properties and through which mechanisms. Glucans and specific proteins are responsible for most of the biological effects of mushrooms, particularly in terms of immunomodulatory and anti-tumor results. Proteins with bioactive effects include lectins, fungal immunomodulatory proteins (FIPs), ribosome inactivating proteins (RIPs), ribonucleases, laccases, among others. At the present status of knowledge, numerous studies have been performed on cell lines and murine models while only a few clinical trials have been conducted. As in most cases of dietary components, the multitude of variables implicated in the final effect and an inadequate standardization are expected to affect the observed differences, thus making the available evidence insufficient to justify the treatment of human diseases with mushrooms extracts. We will herein provide a comprehensive review and critically discussion the biochemical changes induced by different mushroom compounds as observed in in vitro studies, particularly on macrophages, dendritic cells, T cells, and NK cells, compared to in vivo and human studies. Additional effects are represented by lipids which constitute a minor part of mushrooms but may have a role in reducing serum cholesterol levels or phenols acting as antioxidant and reducing agents. Human studies provide a minority of available data, as well illustrated by a placebo-controlled study of athletes treated with β-glucan from Pleurotus ostreatus. Variables influencing study outcomes include different mushrooms strains, growing conditions, developmental stage, part of mushroom used, extraction method, and storage conditions. We foresee that future rigorous research will be needed to determine the potential of mushroom compounds for human health to reproduce the effects of some compounds such as lentinan which a metaanalysis demonstrated to increase the efficacy of chemotherapy in the treatment of lung cancer and in the improvement of the patients quality of life.
Collapse
Affiliation(s)
- Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
66
|
Polivka L, Frenzel L, Jouzeau JY, Hermine O, Moulin D. Mast cells in spondyloarthritis, more than simple inflammatory bystanders? Ther Adv Musculoskelet Dis 2020; 12:1759720X20971907. [PMID: 33240405 PMCID: PMC7675857 DOI: 10.1177/1759720x20971907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Laura Polivka
- Department of Dermatology, Reference Center for
Genodermatoses (MAGEC), Necker-Enfants Malades Hospital (AP-HP5), Paris-Centre
University; Imagine Institute, INSERM U1163, Laboratory of Molecular Mechanisms of
Hematologic Disorders and Therapeutic Implications, Paris, France; French National
Reference Center for Mastocytosis (CEREMAST)
| | - Laurent Frenzel
- Department of Hematology, Necker-Enfants Malades Hospital
(AP-HP5), Paris-Centre University; Imagine Institute, INSERM U1163, Laboratory of
Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, Paris,
France; French National Reference Center for Mastocytosis (CEREMAST)
| | - Jean-Yves Jouzeau
- IMoPA, UMR7365 CNRS-Université de Lorraine, Vandœuvre-lès
Nancy, France
| | | | | |
Collapse
|
67
|
Vural M, Gilbert B, Üstün I, Caglar S, Finckh A. Mini-Review: Human Microbiome and Rheumatic Diseases. Front Cell Infect Microbiol 2020; 10:491160. [PMID: 33304855 PMCID: PMC7693548 DOI: 10.3389/fcimb.2020.491160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/14/2020] [Indexed: 12/16/2022] Open
Abstract
Rheumatoid arthritis and spondyloarthropathy are the most common inflammatory rheumatic diseases. As the human microbiome is involved in the immune homeostasis, it has the potential to be a key factor in the development of autoimmune diseases and rheumatic diseases. In this article, we review the role of various human microbiota on the pathogenesis of rheumatic diseases, focusing on spondylarthritis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Meltem Vural
- Physical Medicine and Rehabilitation Clinic, University of Health Sciences, Bakırkoy Dr. Sadi Konuk Training Hospital, Istanbul, Turkey
| | - Benoit Gilbert
- Rheumatology Division, Department of Medicine, Geneva University Hospital (HUG), Geneva, Switzerland
| | - Işıl Üstün
- Physical Medicine and Rehabilitation Clinic, University of Health Sciences, Bakırkoy Dr. Sadi Konuk Training Hospital, Istanbul, Turkey
| | - Sibel Caglar
- Physical Medicine and Rehabilitation Clinic, University of Health Sciences, Bakırkoy Dr. Sadi Konuk Training Hospital, Istanbul, Turkey
| | - Axel Finckh
- Rheumatology Division, Department of Medicine, Geneva University Hospital (HUG), Geneva, Switzerland
| |
Collapse
|
68
|
So J, Tam LS. Gut Microbiome and Its Interaction with Immune System in Spondyloarthritis. Microorganisms 2020; 8:microorganisms8111727. [PMID: 33158167 PMCID: PMC7694200 DOI: 10.3390/microorganisms8111727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence suggests there is a gut-joint axis in spondyloarthritis (SpA). In a study, subclinical gut inflammation occurred in nearly 50% of SpA. Chronic gut inflammation also correlated with disease activity in SpA. Trillions of microorganisms reside in the human gut and interact with the human immune system. Dysbiosis affects gut immune homeostasis and triggers different autoimmune diseases including SpA. The absence of arthritis in HLA-B27 germ-free mice and the development of arthritis after the introduction of commensal bacteria to HLA-B27 germ-free mice proved to be the important role of gut bacteria in shaping SpA, other than the genetic factor. The recent advance in gene sequencing technology promotes the identification of microorganisms. In this review, we highlighted current evidence supporting the link between gut and axial SpA (axSpA). We also summarized available findings of gut microbiota and its interaction with the immune system in axSpA. Future research may explore the way to modulate gut microorganisms in axSpA and bring gut microbiome discoveries towards application.
Collapse
Affiliation(s)
| | - Lai-Shan Tam
- Correspondence: ; Tel.: +(852)-3505-3128; Fax: +(852)-2637-3852
| |
Collapse
|
69
|
Hsieh WC, Svensson MN, Zoccheddu M, Tremblay ML, Sakaguchi S, Stanford SM, Bottini N. PTPN2 links colonic and joint inflammation in experimental autoimmune arthritis. JCI Insight 2020; 5:141868. [PMID: 33055428 PMCID: PMC7605542 DOI: 10.1172/jci.insight.141868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/09/2020] [Indexed: 12/28/2022] Open
Abstract
Loss-of-function variants of protein tyrosine phosphatase non-receptor type 2 (PTPN2) enhance risk of inflammatory bowel disease and rheumatoid arthritis; however, whether the association between PTPN2 and autoimmune arthritis depends on gut inflammation is unknown. Here we demonstrate that induction of subclinical intestinal inflammation exacerbates development of autoimmune arthritis in SKG mice. Ptpn2-haploinsufficient SKG mice — modeling human carriers of disease-associated variants of PTPN2 — displayed enhanced colitis-induced arthritis and joint accumulation of Tregs expressing RAR-related orphan receptor γT (RORγt) — a gut-enriched Treg subset that can undergo conversion into FoxP3–IL-17+ arthritogenic exTregs. SKG colonic Tregs underwent higher conversion into arthritogenic exTregs when compared with peripheral Tregs, which was exacerbated by haploinsufficiency of Ptpn2. Ptpn2 haploinsufficiency led to selective joint accumulation of RORγt-expressing Tregs expressing the colonic marker G protein–coupled receptor 15 (GPR15) in arthritic mice and selectively enhanced conversion of GPR15+ Tregs into exTregs in vitro and in vivo. Inducible Treg-specific haploinsufficiency of Ptpn2 enhanced colitis-induced SKG arthritis and led to specific joint accumulation of GPR15+ exTregs. Our data validate the SKG model for studies at the interface between intestinal and joint inflammation and suggest that arthritogenic variants of PTPN2 amplify the link between gut inflammation and arthritis through conversion of colonic Tregs into exTregs. Loss of protein tyrosine phosphatase non-receptor type 2 amplifies the link between gut and joint inflammation through conversion of colonic Tregs into arthritogenic exTregs.
Collapse
Affiliation(s)
- Wan-Chen Hsieh
- Department of Medicine, UCSD School of Medicine, La Jolla, California, USA
| | - Mattias Nd Svensson
- Department of Medicine, UCSD School of Medicine, La Jolla, California, USA.,Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martina Zoccheddu
- Department of Medicine, UCSD School of Medicine, La Jolla, California, USA
| | - Michael L Tremblay
- Rosalind and Morris Goodman Cancer Research Centre.,Department of Biochemistry, and.,Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Nunzio Bottini
- Department of Medicine, UCSD School of Medicine, La Jolla, California, USA
| |
Collapse
|
70
|
Liang J, Zhang JJ, Huang HI, Kanayama M, Youssef N, Jin YJ, Reyes EY, Abram CL, Yang S, Lowell CA, Wang D, Shao L, Shinohara ML, Zhang JY, Hammer GE. The Ubiquitin-Modifying Enzyme A20 Terminates C-Type Lectin Receptor Signals and Is a Suppressor of Host Defense against Systemic Fungal Infection. Infect Immun 2020; 88:e00048-20. [PMID: 32540868 PMCID: PMC7440764 DOI: 10.1128/iai.00048-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/01/2020] [Indexed: 01/02/2023] Open
Abstract
C-type lectin receptors (CLRs) play key roles in antifungal defense. CLR-induced NF-κB is central to CLR functions in immunity, and thus, molecules that control the amplitude of CLR-induced NF-κB could profoundly influence host defense against fungal pathogens. However, little is known about the mechanisms that negatively regulate CLR-induced NF-κB, and molecules which act on the CLR family broadly and which directly regulate acute CLR-signaling cascades remain unidentified. Here, we identify the ubiquitin-editing enzyme A20 as a negative regulator of acute NF-κB activation downstream of multiple CLR pathways. Absence of A20 suppression results in exaggerated CLR responses in cells which are A20 deficient and also cells which are A20 haplosufficient, including multiple primary immune cells. Loss of a single allele of A20 results in enhanced defense against systemic Candida albicans infection and prolonged host survival. Thus, A20 restricts CLR-induced innate immune responses in vivo and is a suppressor of host defense against systemic fungal infection.
Collapse
Affiliation(s)
- Jie Liang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Junyi J Zhang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Hsin-I Huang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Masashi Kanayama
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nourhan Youssef
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yingai J Jin
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Estefany Y Reyes
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Clare L Abram
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Shigao Yang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Clifford A Lowell
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Donghai Wang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Ling Shao
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer Y Zhang
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Gianna Elena Hammer
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
71
|
Hanson AL, Nel HJ, Bradbury L, Phipps J, Thomas R, Lê Cao KA, Kenna TJ, Brown MA. Altered Repertoire Diversity and Disease-Associated Clonal Expansions Revealed by T Cell Receptor Immunosequencing in Ankylosing Spondylitis Patients. Arthritis Rheumatol 2020; 72:1289-1302. [PMID: 32162785 DOI: 10.1002/art.41252] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/05/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) is a common spondyloarthropathy primarily affecting the axial skeleton and strongly associated with HLA-B*27 carriage. Genetic evidence implicates both autoinflammatory processes and autoimmunity against an HLA-B*27-restricted autoantigen in immunopathology. In addition to articular symptoms, up to 70% of AS patients present with concurrent bowel inflammation, suggesting that adverse interactions between a genetically primed host immune system and the gut microbiome contribute to the disease. Accordingly, this study aimed to characterize adaptive immune responses to antigenic stimuli in AS. METHODS The peripheral CD4 and CD8 T cell receptor (TCR) repertoire was profiled in AS patients (n = 47) and HLA-B*27-matched healthy controls (n = 38). Repertoire diversity was estimated using the Normalized Shannon Diversity Entropy (NSDE) index, and univariate and multivariate statistical analyses were performed to characterize AS-associated clonal signatures. Furthermore, T cell proliferation and cytokine production in response to immunogenic antigen exposure were investigated in vitro in peripheral blood mononuclear cells from AS patients (n = 19) and HLA-B*27-matched healthy controls (n = 14). RESULTS Based on the NSDE measure of sample diversity across CD4 and CD8 T cell repertoires, AS patients showed increased TCR diversity compared to healthy controls (for CD4 T cells, P = 7.8 × 10-6 ; for CD8 T cells, P = 9.3 × 10-4 ), which was attributed to a significant reduction in the magnitude of peripheral T cell expansions globally. Upon in vitro stimulation, fewer T cells from AS patients than from healthy controls expressed interferon-γ (for CD8 T cells, P = 0.03) and tumor necrosis factor (for CD4 T cells, P = 0.01; for CD8 T cells, P = 0.002). In addition, the CD8 TCR signature was altered in HLA-B*27+ AS patients compared to healthy controls, with significantly expanded Epstein-Barr virus-specific clonotypes (P = 0.03) and cytomegalovirus-specific clonotypes (P = 0.02). HLA-B*27+ AS patients also showed an increased incidence of "public" CD8 TCRs, representing identical clonotypes emerging in response to common antigen encounters, including homologous clonotypes matching those previously isolated from individuals with bacterial-induced reactive arthritis. CONCLUSION The dynamics of peripheral T cell responses in AS patients are altered, suggesting that differential antigen exposure and disrupted adaptive immunity are underlying features of the disease.
Collapse
Affiliation(s)
- Aimee L Hanson
- University of Queensland, Brisbane, Queensland, Australia
| | - Hendrik J Nel
- University of Queensland, Brisbane, Queensland, Australia
| | - Linda Bradbury
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Julie Phipps
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- University of Queensland, Brisbane, Queensland, Australia
| | | | - Tony J Kenna
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Matthew A Brown
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia, and Guy's and St Thomas' NHS Foundation Trust and King's College London NIHR Biomedical Research Centre, King's College London, UK
| |
Collapse
|
72
|
Abstract
Host-microbiota interactions are fundamental for the development of the immune system. Drastic changes in modern environments and lifestyles have led to an imbalance of this evolutionarily ancient process, coinciding with a steep rise in immune-mediated diseases such as autoimmune, allergic and chronic inflammatory disorders. There is an urgent need to better understand these diseases in the context of mucosal and skin microbiota. This Review discusses the mechanisms of how the microbiota contributes to the predisposition, initiation and perpetuation of immune-mediated diseases in the context of a genetically prone host. It is timely owing to the wealth of new studies that recently contributed to this field, ranging from metagenomic studies in humans and mechanistic studies of host-microorganism interactions in gnotobiotic models and in vitro systems, to molecular mechanisms with broader implications across immune-mediated diseases. We focus on the general principles, such as breaches in immune tolerance and barriers, leading to the promotion of immune-mediated diseases by gut, oral and skin microbiota. Lastly, the therapeutic avenues that either target the microbiota, the barrier surfaces or the host immune system to restore tolerance and homeostasis will be explored.
Collapse
|
73
|
β-Glucan augments IL-1β production by activating the JAK2/STAT3 pathway in cultured rabbit keratinocytes. Microb Pathog 2020; 144:104175. [PMID: 32224209 DOI: 10.1016/j.micpath.2020.104175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 11/23/2022]
Abstract
Trichophyton mentagrophytes (T. mentagrophytes) is the main cause of rabbit dermatophytosis. As the main pathogen-associated molecular pattern of T. mentagrophytes, the role of β-glucan in the pathogenesis of rabbit dermatophytosis remains elusive. Keratinocytes (KC) are the main cellular component and the first defensive line against fungal pathogens in the skin. Therefore, the present study investigated the effects of β-glucan on rabbit KC from dorsal skin. β-glucan was found to inhibit KC proliferation by 10% at 20 ug/ml and this concentration was thus considered as optimal. Next, 20 ug/ml β-glucan stimulation for 24 h significantly increased CXCL8, CXCL11, and IL-1β secretions in KC. Furthermore, β-glucan exposure induced the expressions of JAK2 mRNA, STAT3 mRNA, and p-STAT3 protein. Silencing JAK2 expression inhibited p-STAT3 protein expression and β-glucan-induced IL-1β secretion. And overexpression of JAK2 further promoted β-glucan-mediated p-STAT3 protein and IL-1β productions. These results suggested that β-glucan-induced CXCL8, CXCL11, and IL-1β secretions in rabbit KC might be involved in the inflammatory response of T. mentagrophytes infected rabbit dorsal skin. However, only IL-1β secretion was promoted by the JAK2/STAT3 signaling pathway. In conclusion, this study is a necessary step toward elucidating the mechanisms that underlie skin immune system injury stimulated by β-glucan.
Collapse
|
74
|
Abstract
Rheumatoid Arthritis (RA) is a severe, chronic autoimmune disease that affects 1% of the world's population. Familial risk contributes 50% of the risk of seropositive RA, with strongest risks seen in first-degree relatives. Smoking increases the risk of developing anti-citrullinated peptide antibody (ACPA)+ RA, particularly in individuals with high-risk RA-susceptibility alleles. Other contributory environmental risks including particulate exposure, periodontal disease, bronchiectasis, diet, obesity and the oral contraceptive impact respiratory, oral, intestinal and genital tract mucosal sites. Furthermore, the first signs of autoimmunity may appear at mucosal sites e.g. sputum ACPA-IgA and IgG. While oral and faecal dysbiosis are well described, there is no consistent single bacterial species that appears to drive RA. Animal and human data suggest a model in which multiple environmental influences impact mucosal immune function through the host genetics through enhanced mucosal permeability and the traffic of pro-inflammatory PAMPs and the amplification of autoimmune responses. In some cases, autoimmunity may be driven by cross-reactivity, or mimicry, to pathogen-specific antigens, particularly where the host immune system fails to support their rapid control and elimination.
Collapse
Affiliation(s)
- Anne-Sophie Bergot
- The University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Brisbane, 4102, QLD, Australia
| | - Rabina Giri
- Mater Research Institute-UQ, Faculty of Medicine, University of Queensland, Brisbane, 4102, QLD, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Brisbane, 4102, QLD, Australia.
| |
Collapse
|
75
|
Abstract
Spondyloarthritis (SpA) is a group of chronic, inflammatory rheumatic diseases mainly affecting the axial skeleton. Although the pathogenesis of the disease remains elusive, alterations of intestinal microbial composition have been demonstrated in patients with SpA and associated with intestinal and systemic immune alterations. Substantial data have been published in recent years in ethnically different patient populations, demonstrating in a consolidated way the presence of alterations in the composition of the microbial flora in patients with SpA. It is not currently possible to establish whether these alterations are intrinsically inherent in the disease, for example, the effect of particular genes that confer susceptibility to the disease itself, or are a consequence of a more systemic inflammatory process that also involves the intestine. However, data deriving from animal models and studies on relatives of patients with SpA strongly suggest that these alterations might precede the onset of the disease. In this review, we will try to critically analyze studies on dysbiosis in SpA and animal models of SpA, analyzing their functional consequences and the impact of biotechnological therapies on intestinal bacterial composition.
Collapse
Affiliation(s)
- Daniele Mauro
- Department of Precision Medicine, Università Degli Studi Della Campania L. Vanvitelli, Naples, Italy
| | - Francesco Ciccia
- Department of Precision Medicine, Università Degli Studi Della Campania L. Vanvitelli, Naples, Italy.
| |
Collapse
|
76
|
Żelechowska P, Różalska S, Wiktorska M, Brzezińska-Błaszczyk E, Agier J. Curdlan stimulates tissue mast cells to synthesize pro-inflammatory mediators, generate ROS, and migrate via Dectin-1 receptor. Cell Immunol 2020; 351:104079. [PMID: 32115182 DOI: 10.1016/j.cellimm.2020.104079] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 01/10/2023]
Abstract
Mast cells (MCs) are engaged in host defense against various pathogens as they are equipped with pattern recognition receptors (PRRs). Among PRRs expressed on MCs, there are also molecules recognizing components of the fungal cell wall, which are able to induce cellular activation and response. However, little information is available concerning the MC activation by various fungal-derived components. The aim of the study was to determine whether curdlan, a model fungal particle of β-(1,3)-glucan, can directly stimulate tissue MCs. We demonstrated that curdlan triggers MCs to initiate pro-inflammatory response as it activates these cells to synthesize essential pro-inflammatory and/or immunoregulatory factors. We also showed that curdlan serves as a potent chemoattractant for MCs and stimulates those cells to generate reactive oxygen species (ROS). Finally, we documented that curdlan induces MC response via Dectin-1. Our observations support the idea that MCs serve as important sentinels modulating immune response during fungal infection.
Collapse
Affiliation(s)
- Paulina Żelechowska
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, 92-213 Lodz, Poland
| | - Sylwia Różalska
- Department of Industrial Microbiology and Biotechnology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
| | - Magdalena Wiktorska
- Department of Molecular Cell Mechanisms, Faculty of Health Sciences, Medical University of Lodz, 92-215 Lodz, Poland
| | - Ewa Brzezińska-Błaszczyk
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Justyna Agier
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, 92-213 Lodz, Poland
| |
Collapse
|
77
|
Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020; 16:193-207. [PMID: 32080619 DOI: 10.1038/s41584-019-0364-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.
Collapse
|
78
|
Busch R, Kollnberger S, Mellins ED. HLA associations in inflammatory arthritis: emerging mechanisms and clinical implications. Nat Rev Rheumatol 2020; 15:364-381. [PMID: 31092910 DOI: 10.1038/s41584-019-0219-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our understanding of the mechanisms underlying HLA associations with inflammatory arthritis continues to evolve. Disease associations have been refined, and interactions of HLA genotype with other genes and environmental risk factors in determining disease risk have been identified. This Review provides basic information on the genetics and molecular function of HLA molecules, as well as general features of HLA associations with disease. Evidence is discussed regarding the various peptide-dependent and peptide-independent mechanisms by which HLA alleles might contribute to the pathogenesis of three types of inflammatory arthritis: rheumatoid arthritis, spondyloarthritis and systemic juvenile idiopathic arthritis. Also discussed are HLA allelic associations that shed light on the genetic heterogeneity of inflammatory arthritides and on the relationships between adult and paediatric forms of arthritis. Clinical implications range from improved diagnosis and outcome prediction to the possibility of using HLA associations in developing personalized strategies for the treatment and prevention of these diseases.
Collapse
Affiliation(s)
- Robert Busch
- Department of Life Sciences, University of Roehampton, Whitelands College, London, UK.
| | - Simon Kollnberger
- School of Medicine, Cardiff University, UHW Main Building, Heath Park, Cardiff, UK
| | - Elizabeth D Mellins
- Department of Pediatrics, Program in Immunology, Stanford University Medical Center, Stanford, CA, USA.
| |
Collapse
|
79
|
McGonagle D, Tan AL, Watad A, Helliwell P. Pathophysiology, assessment and treatment of psoriatic dactylitis. Nat Rev Rheumatol 2020; 15:113-122. [PMID: 30610219 DOI: 10.1038/s41584-018-0147-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dactylitis is diffuse swelling of the digits that is usually related to an underlying inflammatory or infiltrative disorder. Psoriatic arthritis (PsA) is the most common severe disease thought to cause dactylitis. Our understanding of the pathogenesis of PsA-related dactylitis comes from experimental animal models of PsA-like disease, as well as advances in imaging and other clinical studies. Clinical trials in PsA have increasingly included dactylitis as an important secondary outcome measure. These studies indicate that cytokines drive multi-locus microanatomical pan-digital pathology. Given the importance of pro-inflammatory cytokines, the pathogenesis of dactylitis is best understood as an initial aberrant innate immune response to biomechanical stress or injury, with subsequent adaptive immune mechanisms amplifying the dactylitis inflammatory response. Regarding the treatment of dactylitis, no studies have been conducted using dactylitis as the primary outcome measure, and the current knowledge comes from analysis of dactylitis as a secondary outcome measure.
Collapse
Affiliation(s)
- Dennis McGonagle
- NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK. .,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
| | - Ai Lyn Tan
- NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Department of Medicine 'B', Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Philip Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Rheumatology department, Bradford Hospitals NHS Foundation Trust, Bradford, UK
| |
Collapse
|
80
|
Pang DD, Cai L, Zhang JR, Dai SM. IL-23 induces the expression of pro-osteogenic factors in
osteoclasts. AKTUEL RHEUMATOL 2020. [DOI: 10.1055/a-1099-9028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Abstract
Background The mechanism for the new bone formation in ankylosing
spondylitis (AS) is still unclear. Although it has been demonstrated that
IL-23 plays a pivotal role in the pathophysiology of AS, IL-23 has no direct
effects on osteoblasts but modulates the function of osteoclasts.
Aims To explore whether IL-23 indirectly facilitates new bone
formation through osteoclasts in AS, here we analyzed whether IL-23 enhances
the expression levels of pro-osteogenic factors by osteoclasts.
Methods Mononuclear cells were harvested from mouse bone marrow and
cultured in the presence of M-CSF (50 ng/ml) and RANKL
(30 ng/ml) to trigger the production of osteoclasts. Protein
and mRNA expression levels of Semaphorin 4D, Ephrin B2, BMP2, BMP6, SPHK1,
HtrA1 and Wnt10b were measured using Western blot and qRT-PCR.
Results Primary mononuclear cells were transformed into osteoclasts
with RANKL and M-CSF. The increased expression of NFATc1 and TRAP together
with TRAP staining of>3 nuclei were used to identify mature
osteoclasts. The mRNA expression levels of BMP2, Ephrin B2 and SPHK1 were
enhanced by 1.46, 2.1 and 2.46 folds after exposure to IL-23. Confirmation
of increased levels of Ephrin B2 and SPHK1 in IL-23-stimulated osteoclasts
was provided by Western blot analysis. IL-23 had no effects on the
expression of BMP6 or Wnt10b, or on the anti-osteogenic factors Semaphorin
4D or HtrA1.
Conclusions IL-23 induces osteoclasts to express pro-osteogenic
factors rather than anti-osteogenic factors, suggesting IL-23 might
indirectly promote the differentiation of osteoblasts through activated
osteoclasts in ankylosing spondylitis.
Collapse
Affiliation(s)
- Dan-Dan Pang
- Department of Rheumatology & Immunology, Changhai Hospital,
Second Military Medical University, Shanghai, China
- Department of Rheumatology & Immunology, Shanghai Jiao Tong
University Affiliated Sixth People’s Hospital, Shanghai,
China
| | - Li Cai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong
University Affiliated Sixth People’s Hospital, Shanghai,
China
| | - Jing-Ru Zhang
- Department of Rheumatology & Immunology, Changhai Hospital,
Second Military Medical University, Shanghai, China
- Department of Rheumatology & Immunology, First Affiliated
Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Sheng-Ming Dai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong
University Affiliated Sixth People’s Hospital, Shanghai,
China
| |
Collapse
|
81
|
Bridgewood C, Sharif K, Sherlock J, Watad A, McGonagle D. Interleukin-23 pathway at the enthesis: The emerging story of enthesitis in spondyloarthropathy. Immunol Rev 2020; 294:27-47. [PMID: 31957051 DOI: 10.1111/imr.12840] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
The inflammatory disorders collectively termed the seronegative spondyloarthropathies (SpA) include ankylosing spondylitis (AS), psoriatic arthritis (PsA), reactive arthritis, the arthritis associated with inflammatory bowel disease including Crohn's disease and ulcerative colitis, the arthritis related to anterior uveitis, and finally, somewhat controversially Behcet's disease. All of these diseases are associated with SNPs in the IL-23R or the interleukin-23 (IL-23) cytokine itself and related downstream signaling JAK pathway genes and the interleukin-17 (IL-17) pathway. In rheumatoid arthritis, the target of the immune response is the synovium but the SpA disorders target the tendon, ligament, and joint capsule skeletal anchorage points that are termed entheses. The discovery that IL-23R-expressing cells were ensconced in healthy murine enthesis, and other extraskeletal anchorage points including the aortic root and the ciliary body of the eye and that systemic overexpression of IL-23 resulted in a severe experimental SpA, confirmed a fundamentally different immunobiology to rheumatoid arthritis. Recently, IL-23R-expressing myeloid cells and various innate and adaptive T cells that produce IL-17 family cytokines have also been described in the human enthesis. Blockade of IL-23 pathway with either anti-p40 or anti-p19 subunits has resulted in some spectacular therapeutic successes in psoriasis and PsA including improvement in enthesitis in the peripheral skeleton but has failed to demonstrate efficacy in AS that is largely a spinal polyenthesitis. Herein, we discuss the known biology of IL-23 at the human enthesis and highlight the remarkable emerging story of this unique skeletal tissue.
Collapse
Affiliation(s)
- Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Kassem Sharif
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Department of Medicine "B", Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Jonathan Sherlock
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Department of Medicine "B", Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton, Leeds Teaching Hospital Trust, Leeds, UK
| |
Collapse
|
82
|
Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunol Rev 2020; 294:164-176. [PMID: 31944330 DOI: 10.1111/imr.12841] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022]
Abstract
Mutations of the genes encoding T-cell receptor (TCR)-proximal signaling molecules, such as ZAP-70, can be causative of immunological diseases ranging from T-cell immunodeficiency to T-cell-mediated autoimmune disease. For example, SKG mice, which carry a hypomorphic point mutation of the Zap-70 gene, spontaneously develop T-cell-mediated autoimmune arthritis immunopathologically similar to human rheumatoid arthritis (RA). The Zap-70 mutation alters the sensitivity of developing T cells to thymic positive/negative selection by self-peptides/MHC complexes, shifting self-reactive TCR repertoire to include a dominant arthritogenic specificity and also affecting thymic development and function of autoimmune suppressive regulatory T (Treg) cells. Polyclonal self-reactive T cells, including potentially arthritogenic T cells, thus produced by the thymus recognize self-peptide/MHC complexes on antigen-presenting cells (APCs) in the periphery and stimulate them to produce cytokines including IL-6 to drive the arthritogenic T cells to differentiate into arthritogenic T-helper 17 (Th17) cells. Insufficient Treg suppression or activation of APCs via microbial and other environmental stimuli evokes arthritis by activating granulocyte-macrophage colony-stimulating factor-secreting effector Th17 cells, mediating chronic bone-destructive joint inflammation by activating myeloid cells, innate lymphoid cells, and synoviocytes in the joint. These findings obtained from the study of SKG mouse arthritis are instrumental in understanding how arthritogenic T cells are produced, become activated, and differentiate into effector T cells mediating arthritis, and may help devising therapeutic measures targeting autoimmune pathogenic Th17 cells or autoimmune-suppressing Treg cells to treat and prevent RA.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
83
|
Abstract
PURPOSE OF REVIEW This article aims to review recent literature linking epithelial barrier inflammation and arthritis in spondyloarthritis (SpA), with a critical view on how they are bound by genetic, immunological and environmental ties. RECENT FINDINGS The epithelia-joint axis has become an intense area of both basic and clinical SpA research. The penultimate goal is to understand the immunopathologic links between epithelial inflammation and arthritis in SpA. Inflammatory bowel disease (IBD) and psoriasis (PsO) have strong links to SpA at several levels. Clinically, there is a strong association of IBD, PsO and SpA. Genetically, there are many shared risk factors; however, there are also distinct differences in the genetics of the respective diseases. Immunologically, type 3 immunity, especially interleukin (IL)-17 and IL-23 dysregulation, has been shown to play a central role in IBD, PsO and SpA. Environmentally, a microbial dysbiosis has been noted in each of these diseases, but whether the microbial signature is similar between diseases is not clear, nor is the effect of dysbiosis on the immune response known. SUMMARY It will be crucial to determine whether the relationship between epithelia inflammation and SpA is truly causal for both the understanding of pathogenesis and for future treatment strategies.
Collapse
|
84
|
Regan-Komito D, Swann JW, Demetriou P, Cohen ES, Horwood NJ, Sansom SN, Griseri T. GM-CSF drives dysregulated hematopoietic stem cell activity and pathogenic extramedullary myelopoiesis in experimental spondyloarthritis. Nat Commun 2020; 11:155. [PMID: 31919358 PMCID: PMC6952438 DOI: 10.1038/s41467-019-13853-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Dysregulated hematopoiesis occurs in several chronic inflammatory diseases, but it remains unclear how hematopoietic stem cells (HSCs) in the bone marrow (BM) sense peripheral inflammation and contribute to tissue damage in arthritis. Here, we show the HSC gene expression program is biased toward myelopoiesis and differentiation skewed toward granulocyte-monocyte progenitors (GMP) during joint and intestinal inflammation in experimental spondyloarthritis (SpA). GM-CSF-receptor is increased on HSCs and multipotent progenitors, favoring a striking increase in myelopoiesis at the earliest hematopoietic stages. GMP accumulate in the BM in SpA and, unexpectedly, at extramedullary sites: in the inflamed joints and spleen. Furthermore, we show that GM-CSF promotes extramedullary myelopoiesis, tissue-toxic neutrophil accumulation in target organs, and GM-CSF prophylactic or therapeutic blockade substantially decreases SpA severity. Surprisingly, besides CD4+ T cells and innate lymphoid cells, mast cells are a source of GM-CSF in this model, and its pathogenic production is promoted by the alarmin IL-33.
Collapse
Affiliation(s)
- Daniel Regan-Komito
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK
| | - James W Swann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Philippos Demetriou
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK
| | - E Suzanne Cohen
- Biopharmaceutical Research Division, AstraZeneca, Cambridge, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK
| | - Stephen N Sansom
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Thibault Griseri
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, UK.
| |
Collapse
|
85
|
Choi SS, Jang E, Jang K, Jung SJ, Hwang KG, Youn J. Autoantibody-Mediated Dysfunction of Salivary Glands Leads to Xerostomia in SKG Mice. Immune Netw 2019; 19:e44. [PMID: 31921474 PMCID: PMC6943169 DOI: 10.4110/in.2019.19.e44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/29/2019] [Accepted: 12/13/2019] [Indexed: 01/10/2023] Open
Abstract
Sjögren's syndrome (SS) is a chronic heterogeneous disease that mainly affects exocrine glands, leading to sicca syndromes such as xerostomia. Despite the second highest prevalence rate among systemic autoimmune diseases, its pathophysiology remains largely unknown. Here we report that SKG mice, a cardinal model of Th17 cell-mediated arthritis, also develop a secondary form of SS-like disorder upon systemic exposure to purified curdlan, a type of β-glucan. The reduced production of saliva was not caused by focal immune cell infiltrates but was associated with IgG deposits in salivary glands. Sera from curdlan-injected SKG mice contained elevated titers of IgG (predominantly IgG1), autoantibody to the muscarinic type 3 receptor (M3R) and inhibited carbachol-induced Ca2+ signaling in salivary acinar cells. These results suggest that the Th17 cells that are elicited in SKG mice promote the production of salivary gland-specific autoantibodies including anti-M3R IgG; the antibodies are then deposited on acinar cells and inhibit M3R-mediated signaling required for salivation, finally leading to hypofunction of the salivary glands. This type II hypersensitivity reaction may explain the origin of secondary SS occurring without focal leukocyte infiltrates.
Collapse
Affiliation(s)
- Suk San Choi
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Eunkyeong Jang
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Kiseok Jang
- Department of Pathology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Sung Jun Jung
- Department of Physiology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Kyung-Gyun Hwang
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Jeehee Youn
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, Hanyang University College of Medicine, Seoul 04763, Korea
| |
Collapse
|
86
|
Rosenbaum JT. The eye in spondyloarthritis ✰. Semin Arthritis Rheum 2019; 49:S29-S31. [PMID: 31779847 DOI: 10.1016/j.semarthrit.2019.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 10/25/2022]
Abstract
Acute anterior uveitis is the most common extra-articular clinical manifestation of spondyloarthropathy. Rheumatologists should be aware of uveitis, know how it presents, understand the differential diagnosis of uveitis and arthritis, and be familiar with the role of systemic medications in the treatment or prevention of uveitis.
Collapse
Affiliation(s)
- James T Rosenbaum
- Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States; Legacy Devers Eye Institute, Suite 200, Portland, OR 97210, United States.
| |
Collapse
|
87
|
Lim DH, Lee EJ, Kwon OC, Hong S, Lee CK, Yoo B, Youn J, Kim TH, Kim YG. Effect of tumor necrosis factor inhibition on spinal inflammation and spinal ankylosis in SKG mice. Sci Rep 2019; 9:18000. [PMID: 31784598 PMCID: PMC6884438 DOI: 10.1038/s41598-019-54549-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
To prevent spinal progression in ankylosing spondylitis, initiating TNF-inhibitor treatment as early as possible is suggested. However, the outcomes are inconsistent in previous clinical studies. Here, we investigated the effect of TNF inhibition alone on spinal progression when used during arthritis development in a murine model. We injected 8-week-old SKG mice with curdlan (curdlan group). We injected adalimumab at 3 and 9 weeks after the first curdlan injection (ADA group). The clinical scores of peripheral arthritis decreased in the ADA group at 3 weeks after first adalimumab injection. Using positron emission tomography–magnetic resonance imaging and histologic examination, spinal inflammation was observed in the curdlan group, and was significantly deceased in the ADA group. However, spinal osteoblast activities by imaging using OsteoSense 680 EX and bone metabolism-related cytokines such as receptor activator of nuclear factor-kappa B ligand, osteoprotegerin, Dickkopf-1, and sclerostin levels except IL-17A level were not different between the two groups. We conclude that treating TNF inhibitor alone reduced peripheral arthritis score and spinal inflammation in curdlan-injected SKG mice but did not decrease the spinal osteoblast activity, suggesting little effect on spinal ankylosis.
Collapse
Affiliation(s)
- Doo-Ho Lim
- Division of Rheumatology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Eun-Ju Lee
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeehee Youn
- Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Tae-Hwan Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
88
|
Veale DJ, McGonagle D, McInnes IB, Krueger JG, Ritchlin CT, Elewaut D, Kanik KS, Hendrikx T, Berstein G, Hodge J, Telliez JB. The rationale for Janus kinase inhibitors for the treatment of spondyloarthritis. Rheumatology (Oxford) 2019; 58:197-205. [PMID: 29618084 PMCID: PMC6343466 DOI: 10.1093/rheumatology/key070] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Indexed: 12/25/2022] Open
Abstract
The pathogenesis of SpA is multifactorial and involves a range of immune cell types and cytokines, many of which utilize Janus kinase (JAK) pathways for signaling. In this review, we summarize the animal and pre-clinical data that have demonstrated the effects of JAK blockade on the underlying molecular mechanisms of SpA and provide a rationale for JAK inhibition for the treatment of SpA. We also review the available clinical trial data evaluating JAK inhibitors tofacitinib, baricitinib, peficitinib, filgotinib and upadacitinib in PsA, AS and related inflammatory diseases, which have demonstrated the efficacy of these agents across a range of SpA-associated disease manifestations. The available clinical trial data, supported by pre-clinical animal model studies demonstrate that JAK inhibition is a promising therapeutic strategy for the treatment of SpA and may offer the potential for improvements in multiple articular and extra-articular disease manifestations of PsA and AS.
Collapse
Affiliation(s)
- Douglas J Veale
- The Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland.,University College, Dublin, Ireland
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - James G Krueger
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, USA
| | - Christopher T Ritchlin
- Allergy, Immunology & Rheumatology Division, University of Rochester Medical Center, Rochester, NY, USA
| | - Dirk Elewaut
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Center, Ghent University and Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | |
Collapse
|
89
|
Korman NJ. Management of psoriasis as a systemic disease: what is the evidence? Br J Dermatol 2019; 182:840-848. [PMID: 31225638 PMCID: PMC7187293 DOI: 10.1111/bjd.18245] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2019] [Indexed: 12/16/2022]
Abstract
Background Psoriasis is a chronic, systemic immune‐mediated disease characterized by development of erythematous, indurated, scaly, pruritic and often painful skin plaques. Psoriasis pathogenesis is driven by proinflammatory cytokines and psoriasis is associated with increased risk for comorbidities, including, but not limited to, psoriatic arthritis, cardiovascular disease, diabetes mellitus, obesity, inflammatory bowel disease and nonalcoholic fatty liver disease compared with the general population. Objectives To explore the pathophysiological relationship between psoriasis and its common comorbidities and discuss the need for new treatment paradigms that include strategies to reduce systemic inflammation in patients with moderate‐to‐severe psoriasis. Methods This narrative review summarizes the published evidence related to the ability of biological therapies to ameliorate the consequences of systemic inflammation in patients with psoriasis. Results Current evidence suggests that preventing damage associated with inflammation, and preventing development of future inflammatory damage and comorbidities, may be a potentially achievable treatment goal for many patients with moderate‐to‐severe plaque psoriasis when biological therapies are utilized early in the disease. Encouraging data from recent studies suggest that the loftier goal of reversing existing inflammatory damage and improving signs and symptoms of inflammatory comorbidities could also possibly be attainable. Conclusions Results from ongoing prospective studies regarding the effects of biologics on markers of systemic inflammation in patients with psoriasis will strengthen the clinical evidence base that can be used to inform treatment decisions for patients with moderate‐to‐severe psoriasis. What's already known about this topic? Psoriasis is a systemic inflammatory disease and treatments are needed to optimize patient outcomes.
What does this study add? This review discusses new psoriasis treatment paradigms that may potentially reduce effects of systemic inflammation. Evidence demonstrating that biological treatment may prevent or reverse inflammatory damage associated with psoriasis comorbidities is reviewed.
Linked Comment:https://doi.org/10.1111/bjd.18456
Collapse
Affiliation(s)
- N J Korman
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, U.S.A.,University Hospitals Cleveland Medical Center, Cleveland, OH, U.S.A
| |
Collapse
|
90
|
Galea R, Nel HJ, Talekar M, Liu X, Ooi JD, Huynh M, Hadjigol S, Robson KJ, Ting YT, Cole S, Cochlin K, Hitchcock S, Zeng B, Yekollu S, Boks M, Goh N, Roberts H, Rossjohn J, Reid HH, Boyd BJ, Malaviya R, Shealy DJ, Baker DG, Madakamutil L, Kitching AR, O’Sullivan BJ, Thomas R. PD-L1- and calcitriol-dependent liposomal antigen-specific regulation of systemic inflammatory autoimmune disease. JCI Insight 2019; 4:126025. [PMID: 31487265 PMCID: PMC6795297 DOI: 10.1172/jci.insight.126025] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 08/21/2019] [Indexed: 12/29/2022] Open
Abstract
Autoimmune diseases resulting from MHC class II-restricted autoantigen-specific T cell immunity include the systemic inflammatory autoimmune conditions rheumatoid arthritis and vasculitis. While currently treated with broad-acting immunosuppressive drugs, a preferable strategy is to regulate antigen-specific effector T cells (Teffs) to restore tolerance by exploiting DC antigen presentation. We targeted draining lymph node (dLN) phagocytic DCs using liposomes encapsulating 1α,25-dihydroxyvitamin D3 (calcitriol) and antigenic peptide to elucidate mechanisms of tolerance used by DCs and responding T cells under resting and immunized conditions. PD-L1 expression was upregulated in dLNs of immunized relative to naive mice. Subcutaneous administration of liposomes encapsulating OVA323-339 and calcitriol targeted dLN PD-L1hi DCs of immunized mice and reduced their MHC class II expression. OVA323-339/calcitriol liposomes suppressed expansion, differentiation, and function of Teffs and induced Foxp3+ and IL-10+ peripheral Tregs in an antigen-specific manner, which was dependent on PD-L1. Peptide/calcitriol liposomes modulated CD40 expression by human DCs and promoted Treg induction in vitro. Liposomes encapsulating calcitriol and disease-associated peptides suppressed the severity of rheumatoid arthritis and Goodpasture's vasculitis models with suppression of antigen-specific memory T cell differentiation and function. Accordingly, peptide/calcitriol liposomes leverage DC PD-L1 for antigen-specific T cell regulation and induce antigen-specific tolerance in inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Ryan Galea
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Hendrik J. Nel
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Meghna Talekar
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Xiao Liu
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Joshua D. Ooi
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Megan Huynh
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Sara Hadjigol
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Kate J. Robson
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Yi Tian Ting
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Suzanne Cole
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Karyn Cochlin
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Shannon Hitchcock
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Bijun Zeng
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Suman Yekollu
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Martine Boks
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Natalie Goh
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | | | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Hugh H. Reid
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ben J. Boyd
- Drug Delivery, Disposition and Dynamics and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Ravi Malaviya
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - David J. Shealy
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Daniel G. Baker
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Loui Madakamutil
- Discovery Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - A. Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
- Departments of Nephrology and Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Brendan J. O’Sullivan
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
91
|
McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL-17A in axial spondyloarthritis and psoriatic arthritis: recent advances and controversies. Ann Rheum Dis 2019; 78:1167-1178. [PMID: 31278139 PMCID: PMC6788885 DOI: 10.1136/annrheumdis-2019-215356] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022]
Abstract
Although the pathogenic mechanisms underlying axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) are not fully elucidated, several lines of evidence suggest that immune responses mediated by interleukin 17A (IL-17A) play a pivotal role in both diseases. This is best highlighted by the significant clinical efficacy shown with inhibitors of IL-17A in treating axSpA and PsA. Nevertheless, a number of knowledge gaps exist regarding the role of IL-17A in the pathophysiology of spondyloarthritis in man, including its cellular origin, its precise role in discrete disease processes such enthesitis, bone erosion, and bone formation, and the reasons for the discrepant responses to IL-17A inhibition observed in certain other spondyloarthritis manifestations. In this review, we focus on the latest data from studies investigating the role of IL-17A in ankylosing spondylitis (AS) and PsA that build on existing and emerging scientific knowledge in the field. Key remaining research questions are also highlighted to guide future research.
Collapse
Affiliation(s)
- Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton, Leeds Teaching Hospital Trust, Leeds, UK
| | - Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sherlock
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Robert Moots
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Department of Academic Rheumatology, Aintree University Hospital, Liverpool, UK
| |
Collapse
|
92
|
Takeuchi Y, Hirota K, Sakaguchi S. Synovial Tissue Inflammation Mediated by Autoimmune T Cells. Front Immunol 2019; 10:1989. [PMID: 31497022 PMCID: PMC6712680 DOI: 10.3389/fimmu.2019.01989] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/06/2019] [Indexed: 01/08/2023] Open
Abstract
In rheumatoid arthritis (RA), various hematopoietic and non-hematopoietic cells present in the synovial tissue secrete numerous inflammatory mediators including pro-inflammatory cytokines critical for the induction of chronic joint inflammation and bone destruction. Fibroblast-like synoviocytes (FLSs) in the non-hematopoietic cell compartment are key inflammatory cells activated in inflamed joints and driving the disease; yet how synovial tissue inflammation is modulated by autoimmune T cells is not fully understood. In this review, mainly based on recent findings with a mouse model of spontaneous autoimmune arthritis, we discuss the mechanism of Th17-mediated synovial tissue inflammation; that is, what environmental stimuli and arthritogenic self-antigens trigger arthritis, how arthritogenic T cells initiate joint inflammation by stimulating FLSs, and how the cellular sources of GM-CSF from lymphoid and tissue stromal cells in the synovium contribute to the development of arthritis. We also highlight possible plasticity of Th17 cells toward pathogenic GM-CSF producers, and the functional instability of regulatory T cells under inflammatory conditions in RA joints.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
93
|
Abstract
Spondyloarthritis (SpA) is a term that refers to a group of inflammatory diseases that includes psoriatic arthritis, axial SpA and nonradiographic axial SpA, reactive arthritis, enteropathic arthritis and undifferentiated SpA. The disease subtypes share clinical and immunological features, including joint inflammation (peripheral and axial skeleton); skin, gut and eye manifestations; and the absence of diagnostic autoantibodies (seronegative). The diseases also share genetic factors. The aetiology of SpA is still the subject of research by many groups worldwide. Evidence from genetic, experimental and clinical studies has accumulated to indicate a clear role for the IL-17 pathway in the pathogenesis of SpA. The IL-17 family consists of IL-17A, IL-17B, IL-17C, IL-17D, IL-17E and IL-17F, of which IL-17A is the best studied. IL-17A is a pro-inflammatory cytokine that also has the capacity to promote angiogenesis and osteoclastogenesis. Of the six family members, IL-17A has the strongest homology with IL-17F. In this Review, we discuss how IL-17A and IL-17F and their cellular sources might contribute to the immunopathology of SpA.
Collapse
|
94
|
Ciccia F, Guggino G, Zeng M, Thomas R, Ranganathan V, Rahman A, Alessandro R, Rizzo A, Saieva L, Macaluso F, Peralta S, Di Liberto D, Dieli F, Cipriani P, Giacomelli R, Baeten D, Haroon N. Proinflammatory CX3CR1+CD59+Tumor Necrosis Factor-Like Molecule 1A+Interleukin-23+ Monocytes Are Expanded in Patients With Ankylosing Spondylitis and Modulate Innate Lymphoid Cell 3 Immune Functions. Arthritis Rheumatol 2019; 70:2003-2013. [PMID: 29869839 DOI: 10.1002/art.40582] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 05/31/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Gut-derived innate lymphoid cell 3 (ILC3) has been shown to participate in the pathogenesis of ankylosing spondylitis (AS). CX3 CR1+ mononuclear phagocytes (MNPs) have been demonstrated to modulate ILC3 function in the gut. This study was undertaken to investigate the role of proinflammatory CX3 CR1+CD59+ MNPs in modulating ILC3 function in AS patients. METHODS MNP subsets in the blood of AS patients and controls were analyzed by flow cytometry. The presence of CX3 CR1+CD59+ cells in tissue was confirmed by confocal microscopy. Expression of the proinflammatory chemokines CX3 CL1 and CCL2 and decoy receptor 6 (DcR-6) was analyzed. Peripheral CX3 CR1+CD59+ cells were cocultured with ILC3, and changes in their frequency were evaluated by flow cytometry. Transcriptome analysis of circulating CX3 CR1+ monocytes was also performed. RESULTS DcR-6 deficiency and CCL2 overexpression were observed in inflamed tissues from AS patients. In the gut, the proinflammatory CX3 CR1+CD59+ MNP population was expanded, correlated with the presence of bacteria, and produced high levels of tumor necrosis factor-like molecule 1A (TL1A) and interleukin-23 (IL-23). MNPs positive for CD11b, CD11c, and major histocompatibility complex class II, predominantly expressing CX3 CR1, were also expanded in the small intestines of treatment-naive SKG relative to BALB/c mice. The frequency of gut-derived CX3 CR1+CD59+CCR9+TL1A+IL-23+ MNPs was significantly higher in the peripheral blood and synovial fluid of AS patients than controls. CCR9+CX3 CR1+CD59+ monocytes were also expanded in AS synovial and bone marrow samples. Transcriptome analysis of isolated CX3 CR1+CD59+ monocytes demonstrated a specific proinflammatory profile in AS. Isolated proinflammatory CX3 CR1+CD59+ MNPs from AS patients induced the expansion and activation of ILC3. CONCLUSION Proinflammatory CX3 CR1+CD59+TL1A+IL-23+ MNPs are expanded in AS patients and display a specific proinflammatory transcriptome profile. Given the ability of these cells to support ILC3 expansion, they may promote a sustained proinflammatory status in AS.
Collapse
Affiliation(s)
| | | | - Michael Zeng
- Krembil Research Institute, Toronto, Ontario, Canada
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | | | - Arifur Rahman
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Riccardo Alessandro
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Palermo, Italy
| | | | - Laura Saieva
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Palermo, Italy
| | | | | | - Diana Di Liberto
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Palermo, Italy
| | - Francesco Dieli
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Palermo, Italy
| | | | | | | | - Nigil Haroon
- Krembil Research Institute, University of Toronto, and University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
95
|
Bridgewood C, Watad A, Russell T, Palmer TM, Marzo-Ortega H, Khan A, Millner PA, Dunsmuir R, Rao A, Loughenbury P, Wittmann M, Cuthbert RJ, McGonagle DG. Identification of myeloid cells in the human enthesis as the main source of local IL-23 production. Ann Rheum Dis 2019; 78:929-933. [PMID: 31018959 PMCID: PMC6585277 DOI: 10.1136/annrheumdis-2018-214944] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/19/2019] [Accepted: 03/23/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We investigated whether the normal human spinal enthesis contained resident myeloid cell populations, capable of producing pivotal proinflammatory cytokines including tumour necrosis factor (TNF) and interleukin (IL)-23 and determined whether these could be modified by PDE4 inhibition. METHODS Normal human enthesis soft tissue (ST) and adjacent perientheseal bone (PEB) (n=15) were evaluated using immunohistochemistry (IHC), digested for myeloid cell phenotyping, sorted and stimulated with different adjuvants (lipopolysaccharide and mannan). Stimulated enthesis fractions were analysed for inducible production of spondyloarthropathy disease-relevant mediators (IL-23 full protein, TNF, IL-1β and CCL20). Myeloid populations were also compared with matched blood populations for further mRNA analysis and the effect of PDE4 inhibition was assessed. RESULTS A myeloid cell population (CD45+ HLADR+ CD14+ CD11c+) phenotype was isolated from both the ST and adjacent PEB and termed 'CD14+ myeloid cells' with tissue localisation confirmed by CD14+ IHC. The CD14- fraction contained a CD123+ HLADR+ CD11c- cell population (plasmacytoid dendritic cells). The CD14+ population was the dominant entheseal producer of IL-23, IL-1β, TNF and CCL20. IL-23 and TNF from the CD14+ population could be downregulated by a PDE4I and other agents (histamine and 8-Bromo-cAMP) which elevate cAMP. Entheseal CD14+ cells had a broadly similar gene expression profile to the corresponding CD14+ population from matched blood but showed significantly lower CCR2 gene expression. CONCLUSIONS The human enthesis contains a CD14+ myeloid population that produces most of the inducible IL-23, IL-1β, TNF and CCL20. This population has similar gene expression profile to the matched blood CD14+ population.
Collapse
Affiliation(s)
- Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- Department of Medicine 'B' and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tobias Russell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Timothy M Palmer
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- National Institute for HealthResearch (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| | - Almas Khan
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | | - Abhay Rao
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- National Institute for HealthResearch (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| | - Richard J Cuthbert
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- National Institute for HealthResearch (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| |
Collapse
|
96
|
Izumiyama T, Mori Y, Mori S, Mori N, Kodama T, Itoi E. The effect of anti-IL-6 receptor antibody for the treatment of McH-lpr/lpr-RA1 mice that spontaneously developed destructive arthritis and enthesitis. BMC Musculoskelet Disord 2019; 20:286. [PMID: 31200688 PMCID: PMC6570918 DOI: 10.1186/s12891-019-2664-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 06/04/2019] [Indexed: 11/28/2022] Open
Abstract
Background McH-lpr/lpr-RA1 mice are a new strain of mice which spontaneously develop destructive arthritis and enthesitis in the ankle. There is no published data that drug treatment has been trialed on these mice. This study examined the effect of the mouse anti-IL-6 receptor antibody, MR16–1, for the treatment of arthritis and enthesitis in McH-lpr/lpr-RA1 mice. Methods Male McH-lpr/lpr-RA1 mice were randomly divided into control and treatment groups. MR16–1 was administered from 10 weeks of age for the treatment group. Saline was applied for the control group. The drug was administered once a week, at an initial dose of 2 mg, then maintained at 0.5 mg once per week thereafter. The effects were evaluated by the histopathological synovitis score, in vivo imaging using indocyanine green liposomes, and analysis of the gene expression of inflammatory cytokines. Results Tissue analyses were carried out at 14, 17 and 20 weeks of age. The synovitis scores of treated groups were significantly lower compared with those of the control group at 14 and 17 weeks of age. The kappa coefficient was 0.77. However, progression of entheseal ossification persisted in the MR16–1 treated group. In vivo imaging using indocyanine green liposomes showed significant decreases in signal intensities of treated groups at week 14, but no significant differences were observed at week 18. Blood serum amyloid A levels in treated groups were significantly lower at 17 weeks of age. The gene expression levels of Tnf and Il17 were also significantly lower in MR16–1 treated groups. Conclusions Administration of the anti-IL-6 receptor antibody is effective for the treatment of synovitis and bone destruction of McH-lpr/lpr-RA1 mice. McH-lpr/lpr-RA1 mice may be a suitable experimental model for the development of new treatments for destructive arthritis and enthesitis. IL-6 signal blockade could contribute to the treatment of destructive arthritis, and further studies should be carried out to confirm its potential in the prevention of enthesopathy developed to ossification.
Collapse
Affiliation(s)
- Takuya Izumiyama
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Yu Mori
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan.
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Naoko Mori
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| |
Collapse
|
97
|
Abstract
PURPOSE OF REVIEW A growing body of evidence supports the relevance of the interleukin-23/interleukin-17 (IL-23/IL-17) pathway for the pathogenesis of axial spondyloarthritis (axSpA) and its treatment. Recently, innate lymphoid cells (ILC), a heterogeneous family of immune effector cells, have been identified as a relevant contributor in tissue homeostasis, partially via IL-23/IL-17 axis. This review describes the biology and the origins of the group 3 ILCs (ILC3s) in humans, focusing on their role in the pathogenesis of axSpA. RECENT FINDINGS Clinical trials showed the effectiveness of IL23/IL-17 axis inhibition in both spondyloarthritis (SpA) and Inflammatory Bowel Disease (IBD). Recent findings confirm the high prevalence of subclinical gut inflammation in patients with SpA. Translational data in humans have demonstrated an increase in the number of ILC3s responsive to IL-23 and producing either IL-22 or IL-17 in the gut of SpA patients. The observation of gut-derived ILC3s in circulation and at inflamed tissues in patients with SpA suggest a recirculation of ILCs from mucosal site to lymphoid tissues and possibly enthesis and joints. Multiple observations demonstrate the expansion of IL-17- and IL-22-producing ILC3 in the subclinically inflamed gut of SpA patients. These innate immune cells, also observed in normal entheses, seem to be able to re-circulate from the gut to inflamed tissues of SpA patients, thus contributing to the disease perpetuation. The development of tools that can provide access to diseased tissue from sacroiliac joint and spinal entheses will provide valuable knowledge on the role of ILC3 in axSpA pathogenesis.
Collapse
|
98
|
Kalinkovich A, Livshits G. A cross talk between dysbiosis and gut-associated immune system governs the development of inflammatory arthropathies. Semin Arthritis Rheum 2019; 49:474-484. [PMID: 31208713 DOI: 10.1016/j.semarthrit.2019.05.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/09/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Emerging evidence suggests that dysbiosis, imbalanced gut microbial community, might be a key player in the development of various diseases, including inflammatory arthropathies, such as rheumatoid arthritis, spondyloarthritis (mainly, ankylosing spondylitis and psoriatic arthritis), and osteoarthritis. Yet, the underlying mechanisms and corresponding interactions remain poorly understood. METHODS We conducted a critical and extensive literature review to explore the association between dysbiosis and the development of inflammatory arthropathies. We also reviewed the literature to assess the perspectives that ameliorate inflammatory arthropathies by manipulating the microbiota with probiotics, prebiotics or fecal microbiota transplantation. RESULTS Some bacterial species (e.g. Prevotella, Citrobacter rodentium, Collinsella aerofaciens, Segmented filamentous bacteria) participate in the creation of the pro-inflammatory immune status, presumably via epitope mimicry, modification of self-antigens, enhanced cell apoptosis mechanisms, and destruction of tight junction proteins and intestinal barrier integrity, all leading to the development and maintainance of inflammatory arthropathies. Whether dysbiosis is an epiphenomenon or is an active driver of these disorders remains unclear, yet, recent observations clearly suggest that dysbiosis precedes and triggers their development implying a causative relationship between dysbiosis and inflammatory arthropathies. The underlying mechanisms include dysbiosis-mediated changes in the functional activity of the intestinal immune cell subsets, such as innate lymphoid cells, mucosa-associated invariant T cells, invariant natural killer T cells, T-follicular helper and T-regulatory cells. In turn, disturbed functionality of the gut-associated immune system is shown to promote the overgrowth of many bacteria, thus establishing a detrimental vicious circle of actively maintaining arthritis. CONCLUSIONS Analysis of the data described in the review supports the notion that a close, dynamic and tightly regulated cross talk between dysbiosis and the gut-associated immune system governs the development of inflammatory arthropathies.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gregory Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
99
|
Choi RY, Asquith M, Rosenbaum JT. Fecal transplants in spondyloarthritis and uveitis: ready for a clinical trial? Curr Opin Rheumatol 2019. [PMID: 29538010 DOI: 10.1097/bor.0000000000000506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The intestinal microbiome is thought to play a role in the pathogenesis of inflammatory bowel disease (IBD). There are many shared clinical manifestations between IBD and spondyloarthritis (SpA), of which the most common are peripheral arthritis and uveitis. Clinical overlap along with similar genetics between these diseases suggests a possible shared pathogenetic mechanism, which might center on the intestinal microbiota. In this review, we discuss the available evidence that SpA is a microbiome-driven disease and indicate how SpA-associated uveitis could be tied to gut dysbiosis. We conclude by discussing different treatment paradigms targeting the intestinal microbiome for SpA. RECENT FINDINGS Recent studies support the growing evidence of the intestinal microbiome as a crucial player in SpA disease pathogenesis. There is emerging evidence that the gut microbiome may play a causative role in uveitis. SUMMARY The field is beginning to discover a new level of understanding how the intestinal microbiome is involved in SpA. Treatment methods to alter intestinal microbiota to treat SpA-related diseases are still in its infancy.
Collapse
Affiliation(s)
- Rene Y Choi
- Department of Ophthalmology, Casey Eye Institute
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Department of Ophthalmology, Oregon Health and Science University
| | - James T Rosenbaum
- Department of Ophthalmology, Casey Eye Institute.,Division of Arthritis and Rheumatic Diseases, Department of Ophthalmology, Oregon Health and Science University.,Legacy Devers Eye Institute, Portland, Oregon, USA
| |
Collapse
|
100
|
Hanson AL, Morton CJ, Parker MW, Bessette D, Kenna TJ. The genetics, structure and function of the M1 aminopeptidase oxytocinase subfamily and their therapeutic potential in immune-mediated disease. Hum Immunol 2019; 80:281-289. [DOI: 10.1016/j.humimm.2018.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/16/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022]
|