Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Eur J Immunol ; : e2350823, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38922875

ABSTRACT

Osteoclast-mediated bone erosion and deformation represent significant pathological features in rheumatoid arthritis (RA). Myeloid-derived suppressor cells (MDSCs) and B cells have emerged as key contributors to the progression of RA. Nevertheless, their involvement, especially the interaction in RA osteoclastogenesis remains elusive. In this study, our results revealed a marked expansion of MDSCs in RA patients, and importantly, their abundance was positively correlated with radiographic damage evaluated by the Sharp/van der Heijde score. Notably, MDSCs derived from both RA patients and arthritic mice exhibited a heightened propensity to differentiate into osteoclasts compared with those from healthy individuals. Intriguingly, we observed that B cells from RA patients could augment the osteoclastogenic potential of MDSCs, which was also observed in arthritic mice. The impact of B cells on MDSC-mediated osteoclastogenesis was found to be most pronounced in switched memory B cells, followed by CD21low B cells and naïve B cells. MDSCs from B-cell-deficient mice exhibited diminished capacity to differentiate into osteoclasts, accompanied by distinct gene expression profiles associated with osteoclastogenesis. Taken together, our findings suggested that MDSCs were important osteoclast precursors primed by B cells in RA, serving as novel therapeutic targets for the persistent disease.

2.
Ann Rheum Dis ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39084885

ABSTRACT

OBJECTIVES: Viruses have been considered as important participants in the development of rheumatoid arthritis (RA). However, the profile of enteric virome and its role in RA remains elusive. This study aimed to investigate the atlas and involvement of virome in RA pathogenesis. METHODS: Faecal samples from 30 pairs of RA and healthy siblings that minimise genetic interferences were collected for metagenomic sequencing. The α and ß diversity of the virome and the virome-bacteriome interaction were analysed. The differential bacteriophages were identified, and their correlations with clinical and immunological features of RA were analysed. The potential involvement of these differential bacteriophages in RA pathogenesis was further investigated by auxiliary metabolic gene annotation and molecular mimicry study. The responses of CD4+ T cells and B cells to the mimotopes derived from the differential bacteriophages were systemically studied. RESULTS: The composition of the enteric bacteriophageome was distorted in RA. The differentially presented bacteriophages correlated with the immunological features of RA, including anti-CCP autoantibody and HLA-DR shared epitope. Intriguingly, the glycerolipid and purine metabolic genes were highly active in the bacteriophages from RA. Moreover, peptides of RA-enriched phages, in particular Prevotella phage and Oscillibacter phage could provoke the autoimmune responses in CD4+ T cells and plasma cells via molecular mimicry of the disease-associated autoantigen epitopes, especially those of Bip. CONCLUSIONS: This study provides new insights into enteric bacteriophageome in RA development. In particular, the aberrant bacteriophages demonstrated autoimmunity-provoking potential that would promote the occurrence of the disease.

3.
Ann Rheum Dis ; 83(5): 576-588, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38302261

ABSTRACT

OBJECTIVES: B10 and B10pro cells suppress immune responses via secreting interleukin (IL)-10. However, their regulators and underlying mechanisms, especially in human autoimmune diseases, are elusive. This study aimed to address these questions in rheumatoid arthritis (RA), one of the most common highly disabling autoimmune diseases. METHODS: The frequencies and functions of B10 and B10pro cells in healthy individuals and patients with RA were first analysed. The effects of proinflammatory cytokines, particularly tumour necrosis factor (TNF)-α on the quantity, stability and pathogenic phenotype of these cells, were then assessed in patients with RA before and after anti-TNF therapy. The underlying mechanisms were further investigated by scRNA-seq database reanalysis, transcriptome sequencing, TNF-α-/- and B cell-specific SHIP-1-/- mouse disease model studies. RESULTS: TNF-α was a key determinant for B10 cells. TNF-α elicited the proinflammatory feature of B10 and B10pro cells by downregulating IL-10, and upregulating interferon-γ and IL-17A. In patients with RA, B10 and B10pro cells were impaired with exacerbated proinflammatory phenotype, while anti-TNF therapy potently restored their frequencies and immunosuppressive functions, consistent with the increased B10 cells in TNF-α-/- mice. Mechanistically, TNF-α diminished B10 and B10pro cells by inhibiting their glycolysis and proliferation. TNF-α also regulated the phosphatidylinositol phosphate signalling of B10 and B10pro cells and dampened the expression of SHIP-1, a dominant phosphatidylinositol phosphatase regulator of these cells. CONCLUSIONS: TNF-α provoked the proinflammatory phenotype of B10 and B10pro cells by disturbing SHIP-1 in RA, contributing to the disease development. Reinstating the immunosuppressive property of B10 and B10pro cells might represent novel therapeutic approaches for RA.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , B-Lymphocytes, Regulatory , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Autoimmune Diseases/metabolism , B-Lymphocytes, Regulatory/metabolism , Phenotype , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
4.
Rheumatology (Oxford) ; 63(3): 846-855, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37462532

ABSTRACT

OBJECTIVE: Leukocyte Ig-like receptor A3 (LILRA3) is a soluble receptor belongs to the immunoglobulin superfamily. Our previous studies demonstrated that LILRA3 is a common genetic risk for multiple autoimmune diseases, including RA. Functional LILRA3 conferred increased risk of joint destruction in patients with early RA. We undertook this study to further investigate the pathological role of LILRA3 in joint inflammation of RA. METHODS: Soluble LILRA3 was measured by ELISA. LILRA3 plasmids were transfected into human fibroblast-like synoviocytes (FLSs) using electroporation. Activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was determined by western blots. Cytokine transcripts were quantified by real-time PCR. Migratory and invasive capacities of FLSs were evaluated using transwell migration and Matrigel invasion assays. FLS apoptosis was analysed using flow cytometry. Colocalization of LILRA3, LILRB1 and HLA-G in RA-FLSs was visualized by immunofluorescence staining. RESULTS: Soluble LILRA3 was specifically expressed in synovial fluid and serum LILRA3 was significantly increased and positively correlated with disease activity/severity in RA patients. LILRA3 induced an increased expression of IL-6, IL-8 and MMP3 in RA-FLSs. In vitro LILRA3 stimulation or overexpression promoted RA-FLS migration and invasion, and enhanced phosphorylation of ERK/JNK. Inhibition of ERK/JNK resulted in suppression of IL-6/IL-8 expression in LILRA3-stimulated RA-FLSs. LILRA3 was co-localized with its homologue LILRB1 and shared ligand HLA-G in RA-FLSs. CONCLUSION: The present study provides the first evidence that soluble LILRA3 is a novel proinflammatory mediator involved in synovial inflammation by promoting RA-FLS activation, migration and invasion, probably through the ERK/JNK signalling pathways.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , HLA-G Antigens , Humans , Leukocyte Immunoglobulin-like Receptor B1 , Interleukin-6 , Interleukin-8 , Inflammation , Receptors, Immunologic
5.
Article in English | MEDLINE | ID: mdl-38781519

ABSTRACT

OBJECTIVES: The routine biomarkers for rheumatoid arthritis (RA), including anticyclic citrullinated peptide antibody (anti-CCP), rheumatoid factor (RF), immunoglobulin M (IgM), erythrocyte sedimentation rate (ESR), and C-reaction protein (CRP) have limited sensitivity and specificity. Scavenger receptor-A (SR-A) is a novel RA biomarker identified by our group recently, especially for seronegative RA. Here, we performed a large-scale multicentre study to further assess the diagnostic value of SR-A in combination with other biomarkers for RA. METHODS: The performance of SR-A in combination with other biomarkers for RA diagnosis was first revealed by a pilot study, and was further elucidated by a large-scale multicentre study. A total of 1129 individuals from 3 cohorts were recruited in the study, including RA patients, healthy controls, and patients with other common rheumatic diseases. Diagnostic properties were evaluated by the covariate-adjusted receiver-operating characteristic (AROC) curve, sensitivity, specificity and clinical association, respectively. RESULTS: Large-scale multicentre analysis showed that SR-A and anti-CCP dual combination was the optimal method for RA diagnosis, increasing the sensitivity of anti-CCP by 13% (87% vs 74%) while maintaining a specificity of 90%. In early RA patients, SR-A and anti-CCP dual combination also showed promising diagnostic value, increasing the sensitivity of anti-CCP by 7% (79% vs 72%) while maintaining a specificity of 94%. Moreover, SR-A and anti-CCP dual combination was correlated with ESR, IgM, and autoantibodies of RA patients, further revealing its clinical significance. CONCLUSION: SR-A and anti-CCP dual combination could potentially improve early diagnosis of RA, thus improving the prognosis and reducing mortality.

6.
Adv Exp Med Biol ; 1445: 91-99, 2024.
Article in English | MEDLINE | ID: mdl-38967752

ABSTRACT

Liver is the largest internal organ of the body with vital functions. In addition to its endocrine and exocrine activities, liver also plays a pivotal role in the immune system, including haematopoietic functions. Liver parenchymal cells, which are epithelial cells, have been found to possess innate immune functions by expressing pattern-recognition receptors (PRRs), producing complement components, and secreting cytokines. Intriguingly, in recent years, it has been discovered that liver epithelial cells also produce immunoglobulins (Igs), which have long been thought to be produced exclusively by B cells. Notably, even liver epithelial cells from B lymphocyte-deficient mice, including SCID mice and µMT mice, could also produce Igs. Compelling evidence has revealed both the physiological and pathological functions of liver-derived Igs. For instance, liver epithelial cells-derived IgM can serve as a source of natural and specific antibodies that contribute to innate immune responses, while liver-produced IgG can act as a growth factor to promote cell proliferation and survival in normal hepatocytes and hepatocarcinoma. Similar to that in B cells, the toll-like receptor 9 (TLR9)-MyD88 signaling pathway is also actively involved in promoting liver epithelial cells to secrete IgM. Liver-derived Igs could potentially serve as biomarkers, prognostic indicators, and therapeutic targets in the clinical setting, particularly for liver cancers and liver injury. Nevertheless, despite significant advances, much remains unknown about the mechanisms governing Ig transcription in liver cells, as well as the detailed functions of liver-derived Igs and their involvement in diseases and adaptive immunity. Further studies are still needed to reveal these underlying, undefined issues related to the role of liver-derived Igs in both immunity and diseases.


Subject(s)
Immunity, Innate , Liver , Animals , Liver/metabolism , Liver/immunology , Humans , Immunoglobulins/metabolism , Immunoglobulins/immunology , Immunoglobulins/genetics , Signal Transduction , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Hepatocytes/metabolism , Hepatocytes/immunology , Clinical Relevance
7.
Clin Exp Rheumatol ; 41(3): 634-641, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36062763

ABSTRACT

OBJECTIVES: It has been proved that B cells play indispensable roles in immunity via producing cytokines and secreting antibodies. Aberrant B cells are considered as the major participants in the pathogenesis of systemic lupus erythematosus (SLE). Recently, perforin (PFP)-producing B cell has been identified, serving as a new type of potential anti-tumour effector cells. However, the roles and characteristics of the PFP-producing B cells in SLE remain unclear. METHODS: The frequencies of PFP-producing B cells in peripheral blood of heathy controls (HC) and SLE patients were detected by flow cytometry, and their correlation with the patient clinical and immunological features were analysed. The capacities of these cells in producing PFP were also compared between HC and SLE by RT-qPCR and ELISpot analyses. RESULTS: In this study, we demonstrated that B cells could produce PFP and was further enhanced upon anti-BCR and IL-21 stimulation. In patients with SLE, the frequencies of these PFP-producing B cells were decreased and negatively correlated with the clinical characteristics. Further analysis revealed that SLE patients with vasculitis and pleurisy showed even lower frequencies of PFP-producing B cells. CONCLUSIONS: These findings revealed that B cells could produce PFP, and a decrease in these cells was associated with SLE pathogenesis.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Perforin , Cytokines , B-Lymphocytes/pathology
8.
Clin Exp Immunol ; 209(3): 270-279, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35951003

ABSTRACT

To investigate the expression and roles of TAM (Tyro3/Axl/Mer) receptor tyrosine kinases (TK) in synovial fluid and synovial tissue of patients with rheumatoid arthritis (RA). The expression of TAM TKs in the synovial fluid and synovial tissues of RA and osteoarthritis (OA) patients was measured by ELISA and immunohistochemistry. The relationships between soluble TAM TKs (sTAM TKs) levels and the clinical features, laboratory parameters and disease activity were analyzed in RA. The concentrations of sTAM TK in the synovial fluids of RA patients were increased in comparison to those of OA patients. Compared with OA patients, the expression of membrane Tyro3 TK (mTyro3 TK) and mMer TK in RA patient synovial tissue were significantly increased, which may partly explain the possible mechanism of elevated levels of sTAM TK in RA patient synovial fluid. sAxl TK levels were decreased in RA patients under sulfasalazine treatment and elevated in patients under Iguratimod treatment. Furthermore, sTyro3 TK levels were positively correlated with erythrocyte sedimentation rate (ESR) and negatively correlated with white blood cells (WBCs), red blood cells (RBCs), and hemoglobin (HB) in RA patients. The levels of sMer TK were positively associated with disease duration and rheumatoid factor (RF) and negatively correlated with HB, complement 3 (C3), and C4. Taken together, TAM TKs might be involved in RA synovial tissue inflammation.


Subject(s)
Arthritis, Rheumatoid , Osteoarthritis , Complement C3/metabolism , Humans , Receptor Protein-Tyrosine Kinases , Rheumatoid Factor , Sulfasalazine/metabolism , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Tyrosine/metabolism
9.
Clin Exp Immunol ; 207(3): 297-306, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35553634

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease characterized by proliferative synovitis with deterioration of cartilage and bone. Osteoclasts (OCs) are the active participants in the bone destruction of RA. Although with great advances, most current therapeutic strategies for RA have limited effects on bone destruction. Macrophage scavenger receptor A (SR-A) is a class of pattern recognition receptors (PRRs) involved in bone metabolism and OC differentiation. More recently, our study revealed the critical role of SR-A in RA diagnosis and pathogenesis. Here, we further demonstrated that serum SR-A levels were positively correlated with bone destruction in patients with RA. Anti-SR-A neutralizing antibodies significantly inhibited OC differentiation and bone absorption in vitro in patients with RA, but not in healthy individuals, dampening the expression of OC-specific genes such as tartrate-resistant acid phosphatase (TRAP), cathepsin K (CTSK), and matrix metalloproteinase-9 (MMP-9). Similar results were also seen in collagen-induced arthritis (CIA) mice in vitro. Moreover, the anti-SR-A neutralizing antibody could further ameliorate osteoclastogenesis in vivo and ex vivo in CIA mice, accompanied by decreased serum levels of C-terminal telopeptide and IL-6, exhibiting potential protective effects. These results suggest that blockade of SR-A using anti-SR-A neutralizing antibodies might provide a promising therapeutic strategy for bone destruction in the RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Antibodies, Neutralizing/metabolism , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/pathology , Humans , Mice , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism
10.
Clin Exp Immunol ; 205(2): 119-127, 2021 08.
Article in English | MEDLINE | ID: mdl-33969476

ABSTRACT

Human B cells could be divided into four classical subsets based on CD27 and immunoglobulin (Ig)D expression. Distinct from the other three well-studied subsets, CD27- IgD- B cells, also termed as double-negative (DN) B cells, have long been neglected. However, in recent years emerging evidence shows that DN B cells are unique memory B cells with important functions. They are expanded in a variety of diseases, especially in autoimmune diseases, contributing to the disease pathogenesis. Here, we briefly review the studies on DN B cells, including their origins, characteristics, subsets and roles in diseases, to try to bring new insights into this under-recognized B cell subset.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , Immunologic Memory/immunology , Animals , Humans , Immunoglobulin D/immunology
11.
Clin Exp Rheumatol ; 39(4): 859-867, 2021.
Article in English | MEDLINE | ID: mdl-32896245

ABSTRACT

OBJECTIVES: Multiple physiological and pathological conditions interfere with the function of the endoplasmic reticulum (ER). However, much remains unknown regarding the impact of ER stress on toll-like receptors (TLRs) -induced inflammatory responses in rheumatoid arthritis (RA). The aim of this study was to reveal the effects of ER stress and its regulator, X-box-binding protein-1 (XBP-1), on the inflammatory response of RA synovial fibroblasts (RASF) to different TLRs ligands. METHODS: ER stress was induced in RASF by incubating with thapsigargin (Tg). TLR2 ligand Pam3CSK4, TLR3 ligand PolyIC, TLR4 ligand LPS were used to stimulate the cells. Effects of ER stress on TLRs-induced inflammatory mediators were determined by using RT-PCR, qPCR and ELISA analysis. Western blots analysis was used to detected the signalling pathways in this process. For gene silencing experiment, control scrambled or XBP-1 specific siRNA were transfected into RASF. T helper (Th)1/Th17 cells expansion was determined by flow cytometry analysis, and IFN-γ/IL-17A production in supernatants were collected for ELISA assay. RESULTS: ER stress potentiated the expression of inflammatory cytokines, MMPs and VEGF in RASF stimulated by different TLRs ligands, which was companied with enhanced the activation of NF-κB and MAPKs signalling pathways. Silencing XBP-1 in RASF could dampen TLRs signalling-simulated inflammatory response under ER stress. Moreover, blockade of XBP-1 reduced the generation of Th1 and Th17 cells mediated by RASF, and suppressed the production of IFN-γ and IL-17A. CONCLUSIONS: Our findings suggest that ER stress and XBP-1 may function in conjunction with TLRs to drive the inflammation of RASF, and this pathway may serve as a therapeutic target for the disease.


Subject(s)
Arthritis, Rheumatoid , Endoplasmic Reticulum Stress , Synovial Membrane , X-Box Binding Protein 1 , Cells, Cultured , Fibroblasts/metabolism , Humans , Inflammation , Signal Transduction , Synovial Membrane/metabolism , Toll-Like Receptors/metabolism
12.
J Autoimmun ; 112: 102504, 2020 08.
Article in English | MEDLINE | ID: mdl-32576412

ABSTRACT

Immune checkpoint molecules play pivotal roles in maintaining the immune homeostasis. Targeting these molecules, such as the classical Cytotoxic T-Lymphocyte Antigen 4 (CTLA4) and Programmed Cell Death Protein 1 (PD1), achieves great success in treating cancers. However, not all the patients respond well. This urges the immunologists to identify novel immune checkpoint molecules. Lymphocyte activation gene-3 (LAG3; CD223) is a newly identified inhibitory receptor. It is expressed on a variety of immune cells, including CD4+ T cells, CD8+ T cells, Tregs, B cells, and NK cells. Its unique intracellular domains, signaling patterns as well as the striking synergy observed in its targeted therapy with anti-PD1 indicate the important role of LAG3 in maintaining immune tolerance. Currently, a variety of agents targeting LAG3 are in clinical trials, revealing great perspectives in the future immunotherapy. In this review, we briefly summarize the studies on LAG3, including its structure, isoforms, ligands, signaling, function, roles in multiple diseases, as well as the latest targeted therapeutic advances, with particular concern on the potential association of LAG3 with autoimmune diseases.


Subject(s)
Antigens, CD/metabolism , Autoimmune Diseases/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/metabolism , Infections/drug therapy , Neoplasms/drug therapy , Animals , Autoimmune Diseases/immunology , Autoimmunity/drug effects , Clinical Trials as Topic , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors/therapeutic use , Infections/immunology , Molecular Targeted Therapy/methods , Neoplasms/immunology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Lymphocyte Activation Gene 3 Protein
13.
Ann Rheum Dis ; 78(6): 773-780, 2019 06.
Article in English | MEDLINE | ID: mdl-30936065

ABSTRACT

OBJECTIVE: The strong genetic contribution of the major histocompatibility complex (MHC) region to rheumatoid arthritis (RA) has been generally attributed to human leukocyte antigen (HLA)-DRB1. However, due to the high polymorphisms and linkage disequilibrium within MHC, it is difficult to define novel and/or independent genetic risks using conventional HLA genotyping or chip-based microarray technology. This study aimed to identify novel RA risk variants by performing deep sequencing for MHC. METHODS: We first conducted target sequencing for the entire MHC region in 357 anticitrullinated protein antibodies (ACPA)-positive patients with RA and 1001 healthy controls, and then performed HLA typing in an independent case-control cohort consisting of 1415 samples for validation. All study subjects were Han Chinese. Genetic associations for RA susceptibility and severity were analysed. Comparative modelling was constructed to predict potential functions for the newly discovered RA association variants. RESULTS: HLA-DQα1:160D conferred the strongest and independent susceptibility to ACPA-positive RA (p=6.16×10-36, OR=2.29). DRß1:37N had an independent protective effect (p=5.81×10-16, OR=0.49). As predicted by comparative modelling, the negatively charged DQα1:160D stabilises the dimer of dimers, thus may lead to an increased T cell activation. The negatively charged DRß1:37N encoding alleles preferentially bind with epitope P9 arginine, thus may result in a decreased RA susceptibility. CONCLUSIONS: We provide the first evidence that HLA-DQα1:160D, instead of HLA-DRB1*0405, is the strongest and independent genetic risk for ACPA-positive RA in Han Chinese. Our study also illustrates the value of deep sequencing for fine-mapping disease risk variants in the MHC region.


Subject(s)
Arthritis, Rheumatoid/genetics , HLA-DQ alpha-Chains/genetics , Adult , Aged , Anti-Citrullinated Protein Antibodies/immunology , Arthritis, Rheumatoid/immunology , Asian People/genetics , Case-Control Studies , Female , Genetic Predisposition to Disease , HLA-DRB1 Chains/genetics , High-Throughput Nucleotide Sequencing/methods , Histocompatibility Testing/methods , Humans , Lymphocyte Activation/genetics , Male , Middle Aged , T-Lymphocytes/immunology
14.
Analyst ; 144(6): 2042-2051, 2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30714583

ABSTRACT

Glycosylation is a post-translational modification essential for maintaining the structure and function of proteins. Abnormal N-glycan patterns have been found in various diseases compared to healthy controls. A decrease in terminal galactosylated N-glycans of serum IgG in rheumatoid arthritis (RA) and osteoarthritis (OA) may be involved in their immunopathogenesis. However, how glycan patterns differ between RA and OA remains unclear. Here, we identified 15 glycan forms of serum IgG from RA and OA using MALDI-TOF MS. We found that IgG galactosylation represented a suitable candidate for differentiating RA from healthy controls (AUC > 0.9). Then, we performed binary logistic regression to screen out three bisecting N-acetylglucosamine (GlcNAc) glycoforms for distinguishing between OA and RA. Combined ROC analysis of the selected glycans yielded an AUC of 0.81 between OA and RA and an AUC of 0.79 between OA and RF/ACPA negative RA. Similar results were found in the validation set. In conclusion, our analysis demonstrates that RA and OA are distinguished on the basis of their different IgG glycan patterns, which thus serve as suitable candidates as biomarkers for reliably identifying clinical conditions such as RA and OA.


Subject(s)
Arthritis, Rheumatoid/diagnosis , Biomarkers/blood , Galactose/chemistry , Immunoglobulin G/blood , Osteoarthritis/diagnosis , Polysaccharides/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Aged , Arthritis, Rheumatoid/blood , Case-Control Studies , Diagnosis, Differential , Female , Glycosylation , Humans , Male , Middle Aged , Osteoarthritis/blood , Protein Processing, Post-Translational
15.
J Immunol ; 199(9): 3063-3073, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972092

ABSTRACT

Unlike conventional B cells, regulatory B cells exhibit immunosuppressive functions to downregulate inflammation via IL-10 production. However, the molecular mechanism regulating the production of IL-10 is not fully understood. In this study, we report the finding that activation-induced cytidine deaminase (AID) is highly upregulated in the IL-10-competent B cell (B10) cell from Innp5dfl/flAicdaCre/+ mice, whereas the 5' inositol phosphatase SHIP-1 is downregulated. Notably, SHIP-1 deficiency in AID+ B cells leads to a reduction in cell count and impaired IL-10 production by B10 cells. Furthermore, the Innp5dfl/flAicdaCre/+ mouse model shows B cell-dependent autoimmune lupus-like phenotypes, such as elevated IgG serum Abs, formation of spontaneous germinal centers, production of anti-dsDNA and anti-nuclear Abs, and the obvious deposition of IgG immune complexes in the kidney with age. We observe that these lupus-like phenotypes can be reversed by the adoptive transfer of B10 cells from control Innp5dfl/fl mice, but not from the Innp5dfl/flAicdaCre/+ mice. This finding highlights the importance of defective B10 cells in Innp5dfl/flAicdaCre/+ mice. Whereas p-Akt is significantly upregulated, MAPK and AP-1 activation is impaired in B10 cells from Innp5dfl/flAicdaCre/+ mice, resulting in the reduced production of IL-10. These results show that SHIP-1 is required for the maintenance of B10 cells and production of IL-10, and collectively suggests that SHIP-1 could be a new potential therapeutic target for the treatment of autoimmune diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , Cytidine Deaminase/immunology , Interleukin-10/immunology , Lupus Erythematosus, Systemic/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/deficiency , Adoptive Transfer , Animals , B-Lymphocyte Subsets/pathology , Cytidine Deaminase/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Germinal Center/immunology , Germinal Center/pathology , Interleukin-10/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Transgenic , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology
16.
Eur J Immunol ; 46(3): 742-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26604208

ABSTRACT

Synovial fibroblast hyperplasia, T-cell hyperactivity, B-cell overactivation, and the self-perpetuating interactions among these cell types are major characteristics of rheumatoid arthritis (RA). The inflamed joints of RA patients are hypoxic, with upregulated expression of hypoxia-inducible factor-1α (HIF-1α) in RA synovial fibroblasts (RASFs). It remains unknown whether HIF-1α regulates interactions between RASFs and T cells and B cells. We report here that HIF-1α promotes the expression of inflammatory cytokines IL-6, IL-8, TNF-α, and IL-1ß, and cell-cell contact mediators IL-15, vascular cell adhesion molecule (VCAM)-1, thrombospondin (TSP)-1, and stromal cell-derived factor (SDF)-1 in RASFs. Furthermore, HIF-1α perpetuates RASF-mediated inflammatory Th1- and Th17-cell expansion while differentially inhibiting regulatory B10 and innate-like B cells, leading to increased IFN-γ, IL-17, and IgG production and decreased protective natural IgM secretion. Our findings suggest that HIF-1α perpetuates the interactions between RASFs and T cells and B cells to induce inflammatory cytokine and autoantibody production, thus exacerbating the severity of RA. Targeting HIF-1α may provide new therapeutic strategies for overcoming this persistent disease.


Subject(s)
Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Fibroblasts/immunology , Fibroblasts/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , T-Lymphocytes/immunology , Autoantibodies/biosynthesis , Cells, Cultured , Chemokine CXCL12/genetics , Cytokines/genetics , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-15/genetics , Interleukin-17/immunology , Interleukin-17/physiology , Interleukin-6/genetics , Interleukin-8/genetics , Synovial Membrane/cytology , Thrombospondin 1/genetics , Vascular Cell Adhesion Molecule-1/genetics
17.
J Autoimmun ; 76: 53-62, 2017 01.
Article in English | MEDLINE | ID: mdl-27640100

ABSTRACT

Regulatory B10 cells were functionally impaired in rheumatoid arthritis (RA), yet the mechanisms were unclear. B cells are recently recognized as important participants in osteoclastogenesis by producing RANKL. In this study, we investigated whether regulatory B10 cells could convert into RANKL-producing cells, thus impairing their immunosuppressive functions in RA and exacerbating the disease progression. Our results showed that human regulatory B10 cells could ectopically express RANKL. Under RA circumstance, RANKL-producing B10 cells expanded dramatically, partially induced by TNF-α. The frequencies of these cells were positively correlated with RA patient disease activities and tender joint counts, but negatively correlated with the frequencies of regulatory B10 cells. Strikingly, RANKL-producing B10 cells from RA patients, but not healthy individuals significantly promoted osteoclast differentiation and bone erosion in a paracrine and cell-cell contact-dependent manner. Moreover, these pathogenic RANKL-producing B10 cells declined while regulatory IL-10-producing B10 cells increased in RA patients with disease remission after therapy. Collectively, these results showed that in RA, regulatory B10 cells demonstrated the potential of converting into RANKL-producing cells, thus exacerbating osteoclast formation, bone destruction and disease progression. Modulating the status of B10 cells might provide novel therapeutic strategies for RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , B-Lymphocytes, Regulatory/cytology , B-Lymphocytes, Regulatory/metabolism , Cell Transdifferentiation , Osteoclasts/cytology , Osteoclasts/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Autoantibodies/immunology , B-Lymphocytes, Regulatory/immunology , Biomarkers , Case-Control Studies , Cell Transdifferentiation/immunology , Ectopic Gene Expression , Humans , Immunophenotyping , Phenotype , RANK Ligand/genetics , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Ann Rheum Dis ; 75(1): 278-85, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25371442

ABSTRACT

OBJECTIVES: Although myeloid-derived suppressor cells (MDSCs) have been linked to T cell tolerance, their role in autoimmune rheumatoid arthritis (RA) remains elusive. Here we investigate the potential association of MDSCs with the disease pathogenesis using a preclinical model of RA and specimen collected from patients with RA. METHODS: The frequency of MDSCs in blood, lymphoid tissues, inflamed paws or synovial fluid and their association with disease severity, tissue inflammation and the levels of pathogenic T helper (Th) 17 cells were examined in arthritic mice or in patients with RA (n=35) and osteoarthritis (n=15). The MDSCs in arthritic mice were also characterised for their phenotype, inflammation status, T cell suppressive activity and their capacity of pro-Th17 cell differentiation. The involvement of MDSCs in the disease pathology and a Th17 response was examined by adoptive transfer or antibody depletion of MDSCs in arthritic mice or by coculturing mouse or human MDSCs with naïve CD4+ T cells under Th17-polarising conditions. RESULTS: MDSCs significantly expanded in arthritic mice and in patients with RA, which correlated positively with disease severity and an inflammatory Th17 response. While displaying T cell suppressive activity, MDSCs from arthritic mice produced high levels of inflammatory cytokines (eg, interleukin (IL)-1ß, TNF-α). Mouse and human MDSCs promoted Th17 cell polarisation ex vivo. Transfer of MDSCs facilitated disease progression, whereas their elimination in arthritic mice ameliorated disease symptoms concomitant with reduction of IL-17A/Th17 cells. CONCLUSIONS: Our studies suggest that proinflammatory MDSCs with their capacity to drive Th17 cell differentiation may be a critical pathogenic factor in autoimmune arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Myeloid Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Coculture Techniques , Disease Progression , Humans , Immune Tolerance/immunology , Male , Mice, Inbred C57BL , Osteoarthritis/immunology , Severity of Illness Index , Synovial Fluid/immunology , Th17 Cells/immunology
19.
J Immunol ; 193(1): 130-8, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24907345

ABSTRACT

The high temperature requirement A1 (HTRA1) is a potent protease involved in many diseases, including rheumatoid arthritis (RA). However, the regulatory mechanisms that control HTRA1 expression need to be determined. In this study, we demonstrated that IFN-γ significantly inhibited the basal and LPS-induced HTRA1 expression in fibroblasts and macrophages, which are two major cells for HTRA1 production in RA. Importantly, the inhibitory effect of IFN-γ on HTRA1 expression was evidenced in collagen-induced arthritis (CIA) mouse models and in human RA synovial cells. In parallel with the enhanced CIA incidence and pathological changes in IFN-γ-deficient mice, HTRA1 expression in the joint tissues was also increased as determined by real-time PCR and Western blots. IFN-γ deficiency increased the incidence of CIA and the pathological severity in mice. Neutralization of HTRA1 by Ab significantly reversed the enhanced CIA frequency and severity in IFN-γ-deficient mice. Mechanistically, IFN-γ negatively controls HTRA1 expression through activation of p38 MAPK/STAT1 pathway. Dual luciferase reporter assay and chromatin immunoprecipitation analysis showed that STAT1 could directly bind to HTRA1 promoter after IFN-γ stimulation. This study offers new insights into the molecular regulation of HTRA1 expression and its role in RA pathogenesis, which may have significant impact on clinical therapy for RA and possibly other HTRA1-related diseases, including osteoarthritis, age-related macular degeneration, and cancer.


Subject(s)
Arthritis, Rheumatoid/immunology , Gene Expression Regulation, Enzymologic/immunology , Interferon-gamma/immunology , Joints/immunology , Serine Endopeptidases/immunology , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Cell Line , Collagen/toxicity , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , High-Temperature Requirement A Serine Peptidase 1 , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Joints/metabolism , Joints/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Promoter Regions, Genetic/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Ann Rheum Dis ; 74(11): 2070-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24906639

ABSTRACT

BACKGROUND: Recently, our research group identified the non-deleted (functional) leucocyte immunoglobulin-like receptor A3 (LILRA3) as a new genetic risk for rheumatoid arthritis. OBJECTIVES: To further investigate whether the functional LILRA3 is a new susceptibility factor for other autoimmune diseases-for example, systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS). METHODS: The LILRA3 deletion polymorphism and its tagging single nucleotide polymorphism rs103294 were genotyped for 1099 patients with SLE, 403 patients with pSS and 2169 healthy controls. Association analyses were performed in whole dataset or clinical/serological subsets. The impact of LILRA3 on SLE activity and LILRA3 expression was evaluated. RESULTS: The functional LILRA3 conferred high susceptibility to both SLE (p=3.51×10(-7), OR=2.03) and pSS (p=1.40×10(-3), OR=2.32). It was associated with almost all the clinical/serological features in SLE, especially with leucopenia (p=4.09×10(-7), OR=2.19) and thrombocytopenia (p=1.68×10(-5), OR=1.70). In pSS, functional LILRA3 was specifically associated with leucopenia (p=4.39×10(-4), OR=3.25), anti-Ro/SSA-positive subphenotypes (p=4.54×10(-3), OR=2.34) and anti-La/SSB-positive subphenotypes (p=0.012, OR=2.49). Functional LILRA3 conferred higher disease activity in patients with SLE (p=0.044) and higher LILRA3 expression in both SLE (p=5.57×10(-8)) and pSS (p=1.49×10(-7)) than in controls. CONCLUSIONS: Functional LILRA3 is a new susceptibility factor for SLE and pSS. It highly predisposes to certain phenotypes such as leucopenia and thrombocytopenia in SLE, and may confer increased disease activity in SLE and a higher risk of leucopenia and autoantibody-positive subphenotypes in pSS.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Receptors, Immunologic/genetics , Sjogren's Syndrome/genetics , Adult , Aged , Antibodies, Antinuclear/immunology , Case-Control Studies , Female , Genetic Predisposition to Disease , Genotype , Humans , Leukopenia/blood , Leukopenia/genetics , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Phenotype , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Severity of Illness Index , Sjogren's Syndrome/blood , Sjogren's Syndrome/immunology , Thrombocytopenia/blood , Thrombocytopenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL