Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.183
1.
Nature ; 612(7941): 771-777, 2022 12.
Article En | MEDLINE | ID: mdl-36477533

Human leucocyte antigen B*27 (HLA-B*27) is strongly associated with inflammatory diseases of the spine and pelvis (for example, ankylosing spondylitis (AS)) and the eye (that is, acute anterior uveitis (AAU))1. How HLA-B*27 facilitates disease remains unknown, but one possible mechanism could involve presentation of pathogenic peptides to CD8+ T cells. Here we isolated orphan T cell receptors (TCRs) expressing a disease-associated public ß-chain variable region-complementary-determining region 3ß (BV9-CDR3ß) motif2-4 from blood and synovial fluid T cells from individuals with AS and from the eye in individuals with AAU. These TCRs showed consistent α-chain variable region (AV21) chain pairing and were clonally expanded in the joint and eye. We used HLA-B*27:05 yeast display peptide libraries to identify shared self-peptides and microbial peptides that activated the AS- and AAU-derived TCRs. Structural analysis revealed that TCR cross-reactivity for peptide-MHC was rooted in a shared binding motif present in both self-antigens and microbial antigens that engages the BV9-CDR3ß TCRs. These findings support the hypothesis that microbial antigens and self-antigens could play a pathogenic role in HLA-B*27-associated disease.


Autoimmunity , HLA-B Antigens , Peptides , Receptors, Antigen, T-Cell , Humans , Autoantigens/chemistry , Autoantigens/immunology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Synovial Fluid/immunology , Spondylitis, Ankylosing/immunology , Uveitis, Anterior/immunology , Peptide Library , Cross Reactions , Amino Acid Motifs
2.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article En | MEDLINE | ID: mdl-34884486

Lysophosphatidylserine (lysoPS) is known to regulate immune cell functions. Phospholipase A1 member A (PLA1A) can generate this bioactive lipid through hydrolysis of sn-1 fatty acids on phosphatidylserine (PS). PLA1A has been associated with cancer metastasis, asthma, as well as acute coronary syndrome. However, the functions of PLA1A in the development of systemic autoimmune rheumatic diseases remain elusive. To investigate the possible implication of PLA1A during rheumatic diseases, we monitored PLA1A in synovial fluids from patients with rheumatoid arthritis and plasma of early-diagnosed arthritis (EA) patients and clinically stable systemic lupus erythematosus (SLE) patients. We used human primary fibroblast-like synoviocytes (FLSs) to evaluate the PLA1A-induced biological responses. Our results highlighted that the plasma concentrations of PLA1A in EA and SLE patients were elevated compared to healthy donors. High concentrations of PLA1A were also detected in synovial fluids from rheumatoid arthritis patients compared to those from osteoarthritis (OA) and gout patients. The origin of PLA1A in FLSs and the arthritic joints remained unknown, as healthy human primary FLSs does not express the PLA1A transcript. Besides, the addition of recombinant PLA1A stimulated cultured human primary FLSs to secrete IL-8. Preincubation with heparin, autotaxin (ATX) inhibitor HA130 or lysophosphatidic acid (LPA) receptor antagonist Ki16425 reduced PLA1A-induced-secretion of IL-8. Our data suggested that FLS-associated PLA1A cleaves membrane-exposed PS into lysoPS, which is subsequently converted to LPA by ATX. Since primary FLSs do not express any lysoPS receptors, the data suggested PLA1A-mediated pro-inflammatory responses through the ATX-LPA receptor signaling axis.


Arthritis/pathology , Fibroblasts/pathology , Gout/pathology , Lupus Erythematosus, Systemic/pathology , Phospholipases A1/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Synoviocytes/pathology , Arthritis/genetics , Arthritis/immunology , Arthritis/metabolism , Case-Control Studies , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Gout/genetics , Gout/immunology , Gout/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Male , Phospholipases A1/genetics , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Synovial Fluid/immunology , Synovial Fluid/metabolism , Synoviocytes/immunology , Synoviocytes/metabolism
3.
BMC Immunol ; 22(1): 77, 2021 12 17.
Article En | MEDLINE | ID: mdl-34920714

BACKGROUND: Inflammatory arthritis including rheumatoid arthritis (RA) and spondyloarthritis (SpA) is characterized by inflammation and destruction of the joints. Approximately one third of patients do not respond to first-line treatments. Nitro-fatty acids are bioactive lipids with anti-inflammatory properties and tissue-protective functions. The nitro-fatty acid 10-NO2-oleic acid (10-NO2-OA) is being tested in clinical trials for patients with fibrotic and inflammatory conditions. Here, we tested whether 10-NO2-OA could inhibit immune reactions involved in the inflammatory and joint destructive processes in inflammatory arthritis. METHODS: Synovial fluid and blood samples were obtained from 14 patients with active RA or SpA. The in vitro models consisted of synovial fluid mononuclear cells (SFMCs) cultured for 48 h, SFMCs cultured for 21 days, and fibroblast-like synovial cells (FLSs) co-cultured with peripheral blood mononuclear cells (PBMCs) for 48 h. Cells were treated with or without 10-NO2-OA or the tumor necrosis factor alpha (TNFα) inhibitor etanercept. Supernatants were analyzed for type I interferon, monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase 3 (MMP3) and tartrate resistant acid phosphatase (TRAP). RESULTS: In SFMCs cultured for 48 h, 10-NO2-OA dose-dependently decreased the secretion of bioactive type I interferons and MCP-1 but not MMP3 (P = 0.032, P = 0.0001, and P = 0.58, respectively). Both MCP-1 and MMP3 were decreased by etanercept (P = 0.0031 and P = 0.026, respectively). In SFMCs cultured for 21 days, 10-NO2-OA significantly decreased the production of MCP-1 but not TRAP (P = 0.027 and P = 0.1523, respectively). Etanercept decreased the production of TRAP but not MCP-1 (P < 0.001 and P = 0.84, respectively). In co-cultures of FLSs and PBMCs, 10-NO2-OA decreased the production of MCP-1 (P < 0.0001). This decrease in MCP-1 production was not seen with etanercept treatment (P = 0.47). CONCLUSION: 10-NO2-OA decreased the release of MCP-1 in three models of inflammatory arthritis. Of particular interest, 10-NO2-OA inhibited type I interferon, and 10-NO2-OA was more effective in reducing MCP-1 production in cultures dominated by FLSs compared with etanercept. Our results encourage clinical investigations of 10-NO2-OA in patients with inflammatory arthritis.


Anti-Inflammatory Agents/metabolism , Arthritis, Rheumatoid/metabolism , Fibroblasts/physiology , Leukocytes, Mononuclear/immunology , Oleic Acids/metabolism , Spondylitis, Ankylosing/metabolism , Synovial Fluid/immunology , Adult , Cells, Cultured , Chemokine CCL2/metabolism , Coculture Techniques , Etanercept/pharmacology , Female , Humans , Interferon Type I/metabolism , Male , Middle Aged
4.
Front Immunol ; 12: 734322, 2021.
Article En | MEDLINE | ID: mdl-34956173

Osteoarthritis (OA) may result from impaired ability of synovial macrophages to resolve joint inflammation. Increasing macrophage counts in inflamed joints through injection with bone marrow mononuclear cells (BMNC) induces lasting resolution of synovial inflammation. To uncover mechanisms by which BMNC may affect resolution, in this study, differential transcriptional signatures of BMNC in response to normal (SF) and inflamed synovial fluid (ISF) were analyzed. We demonstrate the temporal behavior of co-expressed gene networks associated with traits from related in vivo and in vitro studies. We also identified activated and inhibited signaling pathways and upstream regulators, further determining their protein expression in the synovium of inflamed joints treated with BMNC or DPBS controls. BMNC responded to ISF with an early pro-inflammatory response characterized by a short spike in the expression of a NF-ƙB- and mitogen-related gene network. This response was associated with sustained increased expression of two gene networks comprising known drivers of resolution (IL-10, IGF-1, PPARG, isoprenoid biosynthesis). These networks were common to SF and ISF, but more highly expressed in ISF. Most highly activated pathways in ISF included the mevalonate pathway and PPAR-γ signaling, with pro-resolving functional annotations that improve mitochondrial metabolism and deactivate NF-ƙB signaling. Lower expression of mevalonate kinase and phospho-PPARγ in synovium from inflamed joints treated with BMNC, and equivalent IL-1ß staining between BMNC- and DPBS-treated joints, associates with accomplished resolution in BMNC-treated joints and emphasize the intricate balance of pro- and anti-inflammatory mechanisms required for resolution. Combined, our data suggest that BMNC-mediated resolution is characterized by constitutively expressed homeostatic mechanisms, whose expression are enhanced following inflammatory stimulus. These mechanisms translate into macrophage proliferation optimizing their capacity to counteract inflammatory damage and improving their general and mitochondrial metabolism to endure oxidative stress while driving tissue repair. Such effect is largely achieved through the synthesis of several lipids that mediate recovery of homeostasis. Our study reveals candidate mechanisms by which BMNC provide lasting improvement in patients with OA and suggests further investigation on the effects of PPAR-γ signaling enhancement for the treatment of arthritic conditions.


Bone Marrow Cells/immunology , Leukocytes, Mononuclear/immunology , Osteoarthritis/complications , Osteoarthritis/immunology , Synovitis/complications , Synovitis/immunology , Transcriptome/genetics , Animals , Carpal Joints/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Gene Regulatory Networks , Genomics/methods , Horses , Lipopolysaccharides/adverse effects , Macrophages/immunology , Male , Osteoarthritis/genetics , Synovial Fluid/immunology , Synovitis/chemically induced , Synovitis/genetics
5.
Emerg Microbes Infect ; 10(1): 2151-2168, 2021 Dec.
Article En | MEDLINE | ID: mdl-34723780

Infection by (re-)emerging RNA arboviruses including Chikungunya virus (CHIKV) and Mayaro virus primarily cause acute febrile disease and transient polyarthralgia. However, in a significant subset of infected individuals, debilitating arthralgia persists for weeks over months up to years. The underlying immunopathogenesis of chronification of arthralgia upon primary RNA-viral infection remains unclear. Here, we analysed cell-intrinsic responses to ex vivo arthritogenic alphaviral infection of primary human synovial fibroblasts isolated from knee joints, one the most affected joint types during acute and chronic CHIKV disease. Synovial fibroblasts were susceptible and permissive to alphaviral infection. Base-line and exogenously added type I interferon (IFN) partially and potently restricted infection, respectively. RNA-seq revealed a CHIKV infection-induced transcriptional profile that comprised upregulation of expression of several hundred IFN-stimulated and arthralgia-mediating genes. Single-cell virus-inclusive RNA-seq uncovered a fine-tuned switch from induction to repression of cell-intrinsic immune responses depending on the abundance of viral RNA in an individual cell. Specifically, responses were most pronounced in cells displaying low-to-intermediate amounts of viral RNA and absence of virus-encoded, fluorescent reporter protein expression, arguing for efficient counteraction of innate immunity in cells expressing viral antagonists at sufficient quantities. In summary, cell-intrinsic sensing of viral RNA that potentially persists or replicates at low levels in synovial fibroblasts and other target cell types in vivo may contribute to the chronic arthralgia induced by alphaviral infections. Our findings might advance our understanding of the immunopathophysiology of long-term pathogenesis of RNA-viral infections.


Arbovirus Infections/virology , Arboviruses/physiology , Arthralgia/virology , Immunity, Innate , RNA, Viral/genetics , Synovial Fluid/immunology , Arbovirus Infections/genetics , Arbovirus Infections/immunology , Arboviruses/genetics , Arthralgia/genetics , Arthralgia/immunology , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/virology , Humans , RNA, Viral/metabolism , Single-Cell Analysis , Synovial Fluid/cytology , Virus Replication
6.
Front Immunol ; 12: 745523, 2021.
Article En | MEDLINE | ID: mdl-34745117

Background: Interleukin 40 (IL-40) is a newly identified B cell-associated cytokine implicated in humoral immune responses and B cell homeostasis. As B cells play a pivotal role in autoimmunity, we investigated the function of IL-40 in rheumatoid arthritis (RA). Methods: IL-40 expression was determined in the synovial tissue from RA and osteoarthritis (OA) patients. IL-40 was analysed in the serum/synovial fluid of patients with RA (n=50), systemic lupus erythematosus (SLE, n=69), OA (n=44), and healthy controls (HC, n=50). We assessed the changes of IL-40 levels in RA patients following the B cell depletion by rituximab (n=29) or after the TNF inhibition by adalimumab (n=25). We examined the relationship between IL-40, disease activity, autoantibodies, cytokines, and NETosis markers. Effect of IL-40 on synovial fibroblasts was determined. Results: IL-40 was overexpressed in RA synovial tissue, particularly by synovial lining and infiltrating immune cells. The levels of IL-40 were up-regulated in the synovial fluid of RA versus OA patients (p<0.0001). Similarly, IL-40 was increased in the serum of RA patients compared to HC, OA, or SLE (p<0.0001 for all) and decreased after 16 and 24 weeks (p<0.01 and p<0.01) following rituximab treatment. No significant effect of adalimumab on IL-40 was observed. IL-40 levels in RA patients correlated with rheumatoid factor-IgM and anti-cyclic citrullinated peptides (anti-CCP) in the serum (p<0.0001 and p<0.01), as well as in the synovial fluid (p<0.0001 and p<0.001). Synovial fluid IL-40 was also associated with disease activity score DAS28 (p<0.05), synovial fluid leukocyte count (p<0.01), neutrophil attractants IL-8 (p<0.01), MIP-1α (p<0.01), and markers of neutrophil extracellular traps externalization (NETosis) such as proteinase 3 (p<0.0001) and neutrophil elastase (p<0.0001). Synovial fibroblasts exposed to IL-40 increased the secretion of IL-8 (p<0.01), MCP-1 (p<0.05), and MMP-13 (p<0.01) compared to the unstimulated cells. Conclusions: We show the up-regulation of IL-40 in RA and its decrease following B cell depleting therapy. The association of IL-40 with autoantibodies, chemokines, and markers of NETosis may imply its potential involvement in RA development. Moreover, IL-40 up-regulates the secretion of chemokines and MMP-13 in synovial fibroblasts, indicating its role in the regulation of inflammation and tissue destruction in RA.


Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/therapy , Extracellular Traps/immunology , Interleukins/metabolism , Rituximab/pharmacology , Adalimumab/therapeutic use , Adult , Aged , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/immunology , Autoantibodies/blood , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biomarkers , Cells, Cultured , Cohort Studies , Cytokines/analysis , Female , Fibroblasts , Gene Expression Regulation/drug effects , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Depletion , Male , Matrix Metalloproteinase 13/analysis , Middle Aged , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/metabolism , Rituximab/therapeutic use , Synovial Fluid/chemistry , Synovial Fluid/immunology , Synovial Membrane/chemistry , Synovial Membrane/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
7.
Front Immunol ; 12: 715997, 2021.
Article En | MEDLINE | ID: mdl-34594331

Objective: Antibodies against carbamylated proteins (anti-CarP) are associated with poor prognosis and the development of bone erosions in rheumatoid arthritis (RA). RA neutrophils externalize modified autoantigens through the formation of neutrophil extracellular traps (NETs). Increased levels of the cathelicidin LL37 have been documented in the synovium of RA patients, but the cellular source remains unclear. We sought to determine if post-translational modifications of LL37, specifically carbamylation, occur during NET formation, enhance this protein's autoantigenicity, and contribute to drive bone erosion in the synovial joint. Methods: ELISA and Western blot analyses were used to identify carbamylated LL37 (carLL37) in biological samples. Anti-carLL37 antibodies were measured in the serum of HLA-DRB1*04:01 transgenic mice and in human RA synovial fluid. Results: Elevated levels of carLL37 were found in plasma and synovial fluid from RA patients, compared to healthy controls. RA NETs release carLL37 and fibroblast-like synoviocytes (FLS) internalized NET-bound carLL37 and loaded it into their MHCII compartment. HLA-DRB1*04:01 transgenic mice immunized with FLS containing NETs developed autoantibodies against carLL37. Anti-carLL37 antibodies were present in RA sera and synovial fluid and they correlated with radiologic bone erosion scores of the hands and feet in RA patients. CarLL37-IgG immune complexes enhanced the ability of monocytes to differentiate into osteoclasts and potentiated osteoclast-mediated extracellular matrix resorption. Conclusions: NETs are a source of carLL37 leading to induction of anti-carbamylated autoantibody responses. Furthermore, carLL37-IgG immune complexes may be implicated in the bone damage characteristic of RA. These results support that dysregulated NET formation has pathogenic roles in RA.


Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/pathology , Autoantibodies/immunology , Autoantigens/immunology , Bone Resorption/immunology , Bone Resorption/pathology , Cathelicidins/immunology , Animals , Arthritis, Rheumatoid/metabolism , Bone Resorption/metabolism , Extracellular Traps/immunology , Humans , Mice , Osteoclasts/immunology , Osteoclasts/metabolism , Synovial Fluid/immunology , Synoviocytes/immunology , Synoviocytes/metabolism , Synoviocytes/pathology
8.
Cells ; 10(7)2021 07 02.
Article En | MEDLINE | ID: mdl-34359833

Inflammatory monocyte-derived dendritic cells (Mo-DCs) have been described in several chronic inflammatory disorders, such as rheumatoid arthritis (RA), and are suspected to play a detrimental role by fueling inflammation and skewing adaptive immune responses. However, the characterization of their phenotype is still limited, as well as the comprehension of the factors that govern their differentiation. Here, we show that inflammatory Mo-DCs generated in vitro expressed a large and atypical panel of C-type lectin receptors, including isoforms of CD209 and CD206, CD303 and CD207, as well as intracellular proteins at their surfaces such as the lysosomal protein CD208. Combination of these markers allowed us to identify cells in the synovial fluid of RA patients with a close phenotype of inflammatory Mo-DCs generated in vitro. Finally, we found in coculture experiments that RA synoviocytes critically affected the phenotypic differentiation of monocytes into Mo-DCs, suggesting that the crosstalk between infiltrating monocytes and local mesenchymal cells is decisive for Mo-DCs generation.


Arthritis, Rheumatoid/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Monocytes/immunology , Synoviocytes/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , B7 Antigens/genetics , B7 Antigens/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Differentiation , Coculture Techniques , Dendritic Cells/pathology , Humans , Immunophenotyping , Lectins, C-Type/genetics , Lectins, C-Type/immunology , /immunology , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monocytes/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Phenotype , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction , Synovial Fluid/cytology , Synovial Fluid/immunology , Synoviocytes/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
9.
JCI Insight ; 6(18)2021 09 22.
Article En | MEDLINE | ID: mdl-34403374

Oligoarticular juvenile idiopathic arthritis (oligo JIA) is the most common form of chronic inflammatory arthritis in children, yet the cause of this disease remains unknown. To understand immune responses in oligo JIA, we immunophenotyped synovial fluid T cells with flow cytometry, bulk RNA-Seq, single-cell RNA-Seq (scRNA-Seq), DNA methylation studies, and Treg suppression assays. In synovial fluid, CD4+, CD8+, and γδ T cells expressed Th1-related markers, whereas Th17 cells were not enriched. Th1 skewing was prominent in CD4+ T cells, including Tregs, and was associated with severe disease. Transcriptomic studies confirmed a Th1 signature in CD4+ T cells from synovial fluid. The regulatory gene expression signature was preserved in Tregs, even those exhibiting Th1 polarization. These Th1-like Tregs maintained Treg-specific methylation patterns and suppressive function, supporting the stability of this Treg population in the joint. Although synovial fluid CD4+ T cells displayed an overall Th1 phenotype, scRNA-Seq uncovered heterogeneous effector and regulatory subpopulations, including IFN-induced Tregs, peripheral helper T cells, and cytotoxic CD4+ T cells. In conclusion, oligo JIA is characterized by Th1 polarization that encompasses Tregs but does not compromise their regulatory identity. Targeting Th1-driven inflammation and augmenting Treg function may represent important therapeutic approaches in oligo JIA.


Arthritis, Juvenile/immunology , Cell Polarity , Synovial Fluid/immunology , T-Lymphocytes/physiology , Adolescent , Arthritis, Juvenile/genetics , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Case-Control Studies , Child , Child, Preschool , DNA Methylation , Female , Humans , Immunophenotyping , Infant , Intraepithelial Lymphocytes/physiology , Male , Sequence Analysis, RNA , Single-Cell Analysis , T-Lymphocytes, Regulatory/physiology , Th1 Cells/physiology , Transcriptome
10.
Front Immunol ; 12: 651049, 2021.
Article En | MEDLINE | ID: mdl-34276648

Objective: Autoantibodies are detected in most patients with rheumatoid arthritis (RA) and can be of the IgM, IgG or IgA subclass. Correlations between IgA autoantibodies and more severe disease activity have been previously reported, but the functional role of IgA autoantibodies in the pathogenesis of RA is ill understood. In this study, we explored the effect of IgA immune complexes on osteoclast mediated bone resorption. Methods: Anti-citrullinated peptide antibody (ACPA) and anti-carbamylated protein (anti-CarP) antibody levels of the IgA and IgG isotype and rheumatoid factor (RF) IgA were determined in synovial fluid (SF) of RA patients. Monocytes, neutrophils, and osteoclasts were stimulated with precipitated immune complexes from SF of RA patients or IgA- and IgG-coated beads. Activation was determined by neutrophil extracellular trap (NET) release, cytokine secretion, and bone resorption. Results: NET formation by neutrophils was enhanced by SF immune complexes compared to immune complexes from healthy or RA serum. Monocytes stimulated with isolated SF immune complexes released IL-6 and IL-8, which correlated with the levels of ACPA IgA levels in SF. Osteoclasts cultured in the presence of supernatant of IgA-activated monocytes resorbed significantly more bone compared to osteoclasts that were cultured in supernatant of IgG-activated monocytes (p=0.0233). Osteoclasts expressed the Fc receptor for IgA (FcαRI; CD89) and Fc gamma receptors. IgA-activated osteoclasts however produced significantly increased levels of IL-6 (p<0.0001) and IL-8 (p=0.0007) compared to IgG-activated osteoclasts. Both IL-6 (p=0.03) and IL-8 (p=0.0054) significantly enhanced bone resorption by osteoclasts. Conclusion: IgA autoantibodies induce release of IL-6 and IL-8 by immune cells as well as osteoclasts, which enhances bone resorption by osteoclasts. We anticipate that this will result in more severe disease activity in RA patients. Targeting IgA-FcαRI interactions therefore represents a promising novel therapeutic strategy for RA patients with IgA autoantibodies.


Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Bone Resorption/immunology , Immunoglobulin A/immunology , Osteoclasts/immunology , Animals , Antigen-Antibody Complex/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Bone Resorption/blood , Bone Resorption/pathology , Cattle , Extracellular Traps/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Knee Joint/immunology , Knee Joint/pathology , Osteoclasts/metabolism , Synovial Fluid/immunology
11.
Ann Rheum Dis ; 80(12): 1559-1567, 2021 12.
Article En | MEDLINE | ID: mdl-34226188

OBJECTIVES: A number of immune populations have been implicated in psoriatic arthritis (PsA) pathogenesis. This study used mass cytometry (CyTOF) combined with transcriptomic analysis to generate a high-dimensional dataset of matched PsA synovial fluid (SF) and blood leucocytes, with the aim of identifying cytokine production ex vivo in unstimulated lymphoid and myeloid cells. METHODS: Fresh SF and paired blood were either fixed or incubated with protein transport inhibitors for 6 hours. Samples were stained with two CyTOF panels: a phenotyping panel and an intracellular panel, including antibodies to both T cell and myeloid cell secreted proteins. Transcriptomic analysis by gene array of key expanded cell populations, single-cell RNA-seq, ELISA and LEGENDplex analysis of PsA SF were also performed. RESULTS: We observed marked changes in the myeloid compartment of PsA SF relative to blood, with expansion of intermediate monocytes, macrophages and dendritic cell populations. Classical monocytes, intermediate monocytes and macrophages spontaneously produced significant levels of the proinflammatory mediators osteopontin and CCL2 in the absence of any in vitro stimulation. By contrast minimal spontaneous cytokine production by T cells was detected. Gene expression analysis showed the genes for osteopontin and CCL2 to be among those most highly upregulated by PsA monocytes/macrophages in SF; and both proteins were elevated in PsA SF. CONCLUSIONS: Using multiomic analyses, we have generated a comprehensive cellular map of PsA SF and blood to reveal key expanded myeloid proinflammatory modules in PsA of potential pathogenic and therapeutic importance.


Arthritis, Psoriatic/immunology , Dendritic Cells/cytology , Macrophages/cytology , Monocytes/cytology , Synovial Fluid/cytology , T-Lymphocytes/cytology , Adult , Arthritis, Psoriatic/genetics , Arthritis, Psoriatic/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Profiling , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Osteopontin/genetics , Osteopontin/immunology , Osteopontin/metabolism , RNA-Seq , Single-Cell Analysis , Synovial Fluid/immunology , Synovial Fluid/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Int Immunopharmacol ; 99: 107905, 2021 Oct.
Article En | MEDLINE | ID: mdl-34242997

OBJECTIVE: To determine whether idealized anterior cruciate ligament reconstruction (IACL-R) restores normal gait features, and whether inflammatory factors are involved in the pathogenesisof post-traumatic osteoarthritis (PTOA). METHODS: Fourteen mature female minipigs were allocated to a sham group (n = 7) or an IACL-R group (n = 7). Load asymmetry during gait was recorded using a pressure-sensing walkway measurement system to evaluate the gait features of the right knee joint before and after surgery. Inflammatory factors (including interleukin [IL]-1α, IL-1ß, IL-2, IL-6, IL-8, IL-18, tumor necrosis factor-α, and granulocyte-macrophage colony-stimulating factor) in synovial fluid were measured using Luminex assays before and after surgery. Cartilage integrity and the subchondral bone plate of the right knee were evaluated using histology and imaging at 3 months postoperatively. RESULTS: Swing time and stance time returned to their preoperative values on day 31, while maximum force, contact area, peak force ,and impulse returned to their preoperative values on day 45 after the surgery in the IACL-R group (P = 0.073, 0.053, 0.107, 0.052, 0.152, and 0.059, respectively).Thus, IACL-R restored normal gait. Compared with their preoperative concentrations, all tested inflammatory factors showed significantly increased concentrations in the synovial fluid in the IACL-R group, especially at 3, 7, and 15 days postoperatively. X-ray, computed tomography, magnetic resonance imaging, and histological data showed severe cartilage damage in the IACL-R model. CONCLUSION: IACL-R restored normal gait features but caused significant cartilage damage, indicating that significantly elevated inflammatory factors maybe crucial for the pathogenesis of PTOA.


Anterior Cruciate Ligament Reconstruction , Osteoarthritis, Knee/therapy , Animals , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Cytokines/immunology , Disease Models, Animal , Female , Gait Analysis , Knee Joint/diagnostic imaging , Knee Joint/pathology , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/physiopathology , Swine , Swine, Miniature , Synovial Fluid/immunology
13.
J Immunol ; 207(2): 381-388, 2021 07 15.
Article En | MEDLINE | ID: mdl-34162725

Stem cell-like memory T (Tscm) cells are a subset of memory T cells that have characteristics of stem cells. The characteristics of Tscm cells in patients with rheumatoid arthritis (RA) are not well known. The percentage of CD4+ and CD8+ Tscm cells in PBMCs and synovial fluid mononuclear cells was measured. After confirming the stem cell nature of Tscm cells, we examined their pathogenicity in RA patients and healthy controls (HCs) by assessing T cell activation markers and cytokine secretion after stimulation with anti-CD3/CD28 beads and/or IL-6. Finally, RNA transcriptome patterns in Tscm cells from RA patients were compared with those in HCs. In this study, the percentage of CD4+ and CD8+ Tscm cells in total T cells was significantly higher in RA patients than in HCs. Tscm cells self-proliferated and differentiated into memory and effector T cell subsets when stimulated. Compared with Tscm cells from HCs, Tscm cells from RA patients were more easily activated by anti-CD3/CD28 beads augmented by IL-6. Transcriptome analyses revealed that Tscm cells from RA patients showed a pattern distinct from those in HCs; RA-specific transcriptome patterns were not completely resolved in RA patients in complete clinical remission. In conclusion, Tscm cells from RA patients show a transcriptionally distinct pattern and are easily activated to produce inflammatory cytokines when stimulated by TCRs in the presence of IL-6. Tscm cells can be a continuous source of pathogenicity in RA.


Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Stem Cells/immunology , CD28 Antigens/immunology , Cytokines/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Count/methods , Male , Middle Aged , Synovial Fluid/immunology
14.
Res Vet Sci ; 136: 377-384, 2021 May.
Article En | MEDLINE | ID: mdl-33799167

BACKGROUND: Osteoarthritis is currently one of the most common chronic diseases. As life expectancy increases, its prevalence and incidence are expected to rise. At present, more and more evidences prove the correlation between the complement system and osteoarthritis (OA). This study aims to investigate complement C5's influence on the effect of MK801 on osteoarthritis synovial fibroblasts (OA-SFs). METHODS: We used IL-1b to induce OA-SFs derived from mice to obtain OA-SFs. And we performed RT-PCR and Western Blot assays to evaluate the expression levels of associated mRNA and protein. The alteration of MAC expression on OA-SFs cell membrane was evaluated by immunofluorescence assay. The expression of related inflammatory factors of OA-SFs was evaluated by ELISA experiment. RESULTS: MK801 could significantly inhibit the expression of osteoarthritis (OA) marker factors, such as: membrane attack complex (MAC), tumor necrosis factor-α (TNF-α) and matrix metalloproteinase-13 (MMP13). Meanwhile, MK801 can significantly inhibit the expression of complement C5 (C5) in OA-SFs. Immunofluorescence assay showed that MAC expression on OA-SFs cell membrane was significantly inhibited by MK801. The nucleo-plasmic separation experiment demonstrated that MK801 could significantly inhibit the activation of Nuclear factor-κB (NF-κB) signaling pathway in OA-SFs. Futhermore, koncking down the expression of C5 reversed the inhibition MK801 on the expression of OA-SFs inflammatory factors. CONCLUSIONS: These results illustrated two points: first, MK801 inhibited the generation of MAC and the release of inflammation factors in OA-SFs through C5; second: MK801 inhibited the activation of NF-κB signaling pathway in OA-SFs.


Complement C5/metabolism , Dizocilpine Maleate/therapeutic use , Fibroblasts/drug effects , Osteoarthritis/drug therapy , Animals , Blotting, Western , Cells, Cultured , Complement Membrane Attack Complex/antagonists & inhibitors , Fibroblasts/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , RNA, Messenger , Signal Transduction , Synovial Fluid/drug effects , Synovial Fluid/immunology , Synovial Membrane/cytology , Tumor Necrosis Factor-alpha/metabolism
15.
Nat Rev Rheumatol ; 17(4): 224-237, 2021 04.
Article En | MEDLINE | ID: mdl-33674813

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder that primarily affects the joints. One hypothesis for the pathogenesis of RA is that disease begins at mucosal sites as a consequence of interactions between the mucosal immune system and an aberrant local microbiota, and then transitions to involve the synovial joints. Alterations in the composition of the microbial flora in the lungs, mouth and gut in individuals with preclinical and established RA suggest a role for mucosal dysbiosis in the development and perpetuation of RA, although establishing whether these alterations are the specific consequence of intestinal involvement in the setting of a systemic inflammatory process, or whether they represent a specific localization of disease, is an ongoing challenge. Data from mouse models of RA and investigations into the preclinical stages of disease also support the hypothesis that these alterations to the microbiota predate the onset of disease. In addition, several therapeutic options widely used for the treatment of RA are associated with alterations in intestinal microbiota, suggesting that modulation of intestinal microbiota and/or intestinal barrier function might be useful in preventing or treating RA.


Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/microbiology , Joints/pathology , Microbiota/immunology , Mucous Membrane/immunology , Animals , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Dysbiosis/complications , Dysbiosis/immunology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Humans , Joints/immunology , Male , Mice , Microbiota/drug effects , Mucous Membrane/microbiology , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Synovial Fluid/immunology
16.
BMJ Case Rep ; 14(3)2021 Mar 02.
Article En | MEDLINE | ID: mdl-33653867

A previously healthy 53-year-old man was hospitalised for 12 days due to COVID-19 with shortness of breath. A few days after discharge from hospital, the patient developed fever and severe pain in several joints in the lower extremities. The pain was so severe that the patient was unable to stand on his feet. Synovial fluid from the right-side knee contained a high number of polynuclear cells and a few mononuclear cells. Microscopy, culture and PCR tests for bacterial infection were all negative. Furthermore, the patient tested negative for rheumatoid factor, anti-cyclic citrullinated peptide and human leukocyte antigen (HLA)-B27. Thus, the condition was compatible with reactive arthritis. The condition improved markedly after a few days' treatment with non-steroid anti-inflammatory drugs and prednisolone.


Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Arthritis, Reactive , Arthritis , COVID-19 , Prednisolone/administration & dosage , Synovial Fluid , Anti-Inflammatory Agents/administration & dosage , Arthralgia/diagnosis , Arthralgia/etiology , Arthritis/drug therapy , Arthritis/etiology , Arthritis/physiopathology , Arthritis, Reactive/diagnosis , Arthritis, Reactive/drug therapy , Arthritis, Reactive/physiopathology , Arthritis, Reactive/virology , Arthritis, Rheumatoid/diagnosis , Autoantibodies/analysis , COVID-19/complications , COVID-19/physiopathology , COVID-19/therapy , Diagnosis, Differential , Humans , Knee Joint/diagnostic imaging , Lower Extremity/pathology , Male , Middle Aged , Radiography/methods , Synovial Fluid/cytology , Synovial Fluid/immunology , Treatment Outcome
17.
Front Immunol ; 12: 644725, 2021.
Article En | MEDLINE | ID: mdl-33777041

With ELISAs one detects the ensemble of immunoreactive molecules in biological samples. For biomolecules undergoing proteolysis for activation, potentiation or inhibition, other techniques are necessary to study biology. Here we develop methodology that combines immunosorbent sample preparation and nano-scale liquid chromatography-tandem mass spectrometry (nano-LC-MS/MS) for proteoform analysis (ISTAMPA) and apply this to the aglycosyl chemokine CXCL8. CXCL8, the most powerful human chemokine with neutrophil chemotactic and -activating properties, occurs in different NH2-terminal proteoforms due to its susceptibility to site-specific proteolytic modification. Specific proteoforms display up to 30-fold enhanced activity. The immunosorbent ion trap top-down mass spectrometry-based approach for proteoform analysis allows for simultaneous detection and quantification of full-length CXCL8(1-77), elongated CXCL8(-2-77) and all naturally occurring truncated CXCL8 forms in biological samples. For the first time we demonstrate site-specific proteolytic activation of CXCL8 in synovial fluids from patients with chronic joint inflammation and address the importance of sample collection and processing.


Arthritis/metabolism , Interleukin-8/metabolism , Proteomics , Synovial Fluid/metabolism , Tandem Mass Spectrometry , Arthritis/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-8/immunology , Male , Synovial Fluid/immunology
18.
Clin Exp Immunol ; 205(1): 44-52, 2021 07.
Article En | MEDLINE | ID: mdl-33576004

Arthritis is a common clinical feature of systemic lupus erythematosus (SLE) and is usually non-erosive, as opposed to rheumatoid arthritis (RA). While RA synovial pathology has been extensively studied, little is known about the pathophysiology of lupus arthritis. Here, we aimed to explore the cytokine and cellular compartments in synovial fluids of SLE patients with arthritic manifestations. Acellular synovial fluid and paired serum samples from SLE patients (n = 17) were analyzed with cytokine bead array for T helper-associated cytokines. From two SLE patients, synovial fluid mononuclear cells (SFMC) could also be captured and were analyzed by multiparameter flow cytometry to dissect T cell, B cell, monocyte and dendritic cell phenotypes. SLE-derived SFMC were further stimulated in vitro to measure their capacity for producing interferon (IFN)-γ and interleukin (IL)-17A. All patients fulfilled the ACR 1982 classification criteria for SLE. Clinical records were reviewed to exclude the presence of co-morbidities such as osteoarthritis or overlap with RA. IL-17A and IL-6 levels were high in SLE synovial fluid. A clear subset of the synovial CD4+ T cells expressed CCR6+ , a marker associated with T helper type 17 (Th17) cells. IL-17A-production was validated among CD4+ CCR6+ T cells following in-vitro stimulation. Furthermore, a strong IFN-γ production was observed in both CD4+ and CD8+ cells. Our study shows high IL-17A and IL-6 levels in synovial fluids of patients with lupus arthritis. The Th17 pathway has been implicated in several aspects of SLE disease pathogenesis and our data also point to Th17 involvement for lupus arthritis.


Arthritis/immunology , Interleukin-17/immunology , Interleukin-6/immunology , Lupus Erythematosus, Systemic/immunology , Synovial Fluid/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Female , Humans , Interferon-gamma/immunology , Male , Middle Aged , Th17 Cells/immunology
19.
Eur J Immunol ; 51(4): 903-914, 2021 04.
Article En | MEDLINE | ID: mdl-33347617

This study elucidates the mechanism of CCL25 and CCR9 in rheumatoid arthritis (RA). RA synovial fluid (SF) expresses elevated levels of CCL25 compared to OA SF and plasma from RA and normal. CCL25 was released into RA SF by fibroblasts (FLS) and macrophages (MΦs) stimulated with IL-1ß and IL-6. CCR9 is also presented on IL-1ß and IL-6 activated RA FLS and differentiated MΦs. Conversely, in RA PBMCs neither CCL25 nor CCR9 are impacted by 3-month longitudinal TNF inhibitor therapy. CCL25 amplifies RA FLS and monocyte infiltration via p38 and ERK phosphorylation. CCL25-stimulated RA FLS secrete potentiated levels of IL-8 which is disrupted by p38 and ERK inhibitors. CCL25 polarizes RA monocytes into nontraditional M1 MΦs that produce IL-8 and CCL2. Activation of p38 and ERK cascades are also responsible for the CCL25-induced M1 MΦ development. Unexpectedly, CCL25 was unable to polarize RA PBMCs into effector Th1/Th17 cells. Consistently, lymphokine like RANKL was uninvolved in CCL25-induced osteoclastogenesis; however, this manifestation was regulated by osteoclastic factors such as RANK, cathepsin K (CTSK), and TNF-α. In short, we reveal that CCL25/CCR9 manipulates RA FLS and MΦ migration and inflammatory phenotype in addition to osteoclast formation via p38 and ERK activation.


Arthritis, Rheumatoid/immunology , Cell Differentiation/immunology , Chemokines, CC/immunology , Macrophages/immunology , Osteoclasts/immunology , Receptors, CCR/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemokines, CC/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Interleukin-8/immunology , Interleukin-8/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Phosphorylation , Receptors, CCR/metabolism , Signal Transduction/immunology , Synovial Fluid/cytology , Synovial Fluid/immunology , Synovial Fluid/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Front Immunol ; 12: 722349, 2021.
Article En | MEDLINE | ID: mdl-35095831

Dendritic cells (DC) have a key role in the initiation and progression of inflammatory arthritis (IA). In this study, we identified a DC population that derive from monocytes, characterized as CD209/CD14+ DC, expressing classical DC markers (HLADR, CD11c) and the Mo-DC marker (CD209), while also retaining the monocytic marker CD14. This CD209/CD14+ DC population is present in the circulation of Healthy Control (HC), with increased frequency in Rheumatoid Arthritis (RA) and Psoriatic arthritic (PsA) patients. We demonstrate, for the first time, that circulatory IA CD209/CD14+ DC express more cytokines (IL1ß/IL6/IL12/TNFα) and display a unique chemokine receptor expression and co-expression profiles compared to HC. We demonstrated that CD209/CD14+ DC are enriched in the inflamed joint where they display a unique inflammatory and maturation phenotype, with increased CD40 and CD80 and co-expression of specific chemokine receptors, displaying unique patterns between PsA and RA. We developed a new protocol of magnetic isolation and expansion for CD209+ DC from blood and identified transcriptional differences involved in endocytosis/antigen presentation between RA and PsA CD209+ DC. In addition, we observed that culture of healthy CD209+ DC with IA synovial fluid (SF), but not Osteoarthritis (OA) SF, was sufficient to induce the development of CD209/CD14+ DC, leading to a poly-mature DC phenotype. In addition, differential effects were observed in terms of chemokine receptor and chemokine expression, with healthy CD209+ DC displaying increased expression/co-expression of CCR6, CCR7, CXCR3, CXCR4 and CXCR5 when cultured with RA SF, while an increase in the chemokines CCR3, CXCL10 and CXCL11 was observed when cultured with PsA SF. This effect may be mediated in part by the observed differential increase in chemokines expressed in RA vs PsA SF. Finally, we observed that the JAK/STAT pathway, but not the NF-κB pathway (driven by TNFα), regulated CD209/CD14+ DC function in terms of activation, inflammatory state, and migratory capacity. In conclusion, we identified a novel CD209/CD14+ DC population, which is active in the circulation of RA and PsA, an effect potentiated once they enter the joint. Furthermore, we demonstrated that JAK/STAT inhibition can be used as a therapeutic strategy to decrease the inflammatory state of the pathogenic CD209/CD14+ DC.


Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/immunology , Cell Adhesion Molecules/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Lipopolysaccharide Receptors/immunology , Receptors, Cell Surface/immunology , Synovial Fluid/immunology , Synovial Membrane/immunology , Adult , Aged , Chemokines/immunology , Female , Humans , Male , Middle Aged , Monocytes/immunology
...