Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.395
Filter
1.
Autoimmunity ; 57(1): 2387076, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39229919

ABSTRACT

OBJECTIVE: This study aims to explore the effect of NONHSAT042241 on the function of rheumatoid arthritis -fibroblast-like synoviocyte (RA-FLS) and the underlying mechanisms. METHODS: RA-FLS was treated with NONHSAT042241 overexpression and NONHSAT042241 knockdown lentiviruses. Cell counting kit-8 (CCK-8) assay, colony formation assay, flow cytometry, Transwell assay, western-blot, ELISA, and qRT-PCR were used to measure the changes of cell proliferation, apoptosis, invasion, secretion of inflammatory cytokines and matrix metalloproteinases (MMPs). Fluorescent in situ hybridization (FISH) assay, RNA pull-down assay, mass spectrometry (MS) and RNA immunoprecipitation (RIP) were used to find the target proteins that bond to NONHSAT042241, and western-blot was used to detect the expression of related proteins of Wnt/ß-catenin signaling pathway. RESULTS: Overexpression of NONHSAT042241 inhibited the proliferation of RA-FLS (p < 0.05), invasion, secretion of pro-inflammatory factors (IL-1and IL-6) and MMPs (MMP-1 and MMP-3) (p < 0.05), and elevated the level of pro-apoptotic factors (Bax and cleaved caspase3), while NONHSAT042241 knockdown had the opposite effect. NONHSAT042241 can directly bind to hnRNP D, and down-regulated the expression of ß-catenin (p < 0.05), p-GSK-3ß (p < 0.05), Cyclin D1 (p < 0.05), PCNA (p < 0.05), and thus reduced the cell proliferation. CONCLUSION: NONHSAT042241 may inhibit FLS-mediated rheumatoid synovial proliferation, inflammation and aggression. The underlying mechanisms may be that NONHSAT042241 inhibits the activity of Wnt/ß-catenin signaling.


Subject(s)
Arthritis, Rheumatoid , Cell Proliferation , Inflammation , RNA, Long Noncoding , Synoviocytes , Wnt Signaling Pathway , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , Synoviocytes/metabolism , Synoviocytes/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Inflammation/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/immunology , Apoptosis , beta Catenin/metabolism , Cells, Cultured
2.
Front Immunol ; 15: 1428773, 2024.
Article in English | MEDLINE | ID: mdl-39161769

ABSTRACT

Rheumatoid arthritis (RA) is a common autoimmune and inflammatory disease characterized by inflammation and hyperplasia of the synovial tissues. RA pathogenesis involves multiple cell types, genes, transcription factors (TFs) and networks. Yet, little is known about the TFs, and key drivers and networks regulating cell function and disease at the synovial tissue level, which is the site of disease. In the present study, we used available RNA-seq databases generated from synovial tissues and developed a novel approach to elucidate cell type-specific regulatory networks on synovial tissue genes in RA. We leverage established computational methodologies to infer sample-specific gene regulatory networks and applied statistical methods to compare network properties across phenotypic groups (RA versus osteoarthritis). We developed computational approaches to rank TFs based on their contribution to the observed phenotypic differences between RA and controls across different cell types. We identified 18 (fibroblast-like synoviocyte), 16 (T cells), 19 (B cells) and 11 (monocyte) key regulators in RA synovial tissues. Interestingly, fibroblast-like synoviocyte (FLS) and B cells were driven by multiple independent co-regulatory TF clusters that included MITF, HLX, BACH1 (FLS) and KLF13, FOSB, FOSL1 (B cells). However, monocytes were collectively governed by a single cluster of TF drivers, responsible for the main phenotypic differences between RA and controls, which included RFX5, IRF9, CREB5. Among several cell subset and pathway changes, we also detected reduced presence of Natural killer T (NKT) cells and eosinophils in RA synovial tissues. Overall, our novel approach identified new and previously unsuspected Key driver genes (KDG), TF and networks and should help better understanding individual cell regulation and co-regulatory networks in RA pathogenesis, as well as potentially generate new targets for treatment.


Subject(s)
Arthritis, Rheumatoid , Gene Regulatory Networks , Synovial Membrane , Humans , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Synovial Membrane/metabolism , Synovial Membrane/immunology , Synovial Membrane/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Profiling , Computational Biology/methods , Synoviocytes/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Gene Expression Regulation , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Transcriptome
3.
RMD Open ; 10(3)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39209371

ABSTRACT

OBJECTIVE: Previous technical limitations prevented the proof of Fcγ-receptor (FcγR)-activation by soluble immune complexes (sICs) in patients. FcγRIIIa (CD16) is a risk factor in rheumatoid arthritis (RA). We aimed at determining the presence of CD16-activating sICs in RA and control diseases. METHODS: Sera from an exploratory cohort (n=50 patients with RA) and a validation cohort (n=106 patients with RA, 20 patients with psoriasis arthritis (PsA), 22 patients with systemic lupus erythematosus (SLE) and 31 healthy controls) were analysed using a new reporter cell assay. Additionally, 26 synovial fluid samples were analysed, including paired serum/synovial samples. RESULTS: For the first time using a reliable and sensitive functional assay, the presence of sICs in RA sera was confirmed. sICs possess an intrinsic capacity to activate CD16 and can be found in both synovial fluid and in blood. In low experimental dilutions, circulating sICs were also detected in a subset of healthy people and in PsA. However, we report a significantly increased frequency of bioactive circulating sICs in RA. While the bioactivity of circulating sICs was low and did not correlate with clinical parameters, synovial sICs were highly bioactive and correlated with serum autoantibody levels. Receiver operator curves indicated that sICs bioactivity in synovial fluid could be used to discriminate immune complex-associated arthritis from non-associated forms. Finally, circulating sICs were more frequently found in SLE than in RA. The degree of CD16 bioactivity showed strong donor-dependent differences, especially in SLE. CONCLUSIONS: RA is characterised by the presence of circulating and synovial sICs that can engage and activate CD16.


Subject(s)
Antigen-Antibody Complex , Arthritis, Rheumatoid , Receptors, IgG , Synovial Fluid , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/blood , Receptors, IgG/metabolism , Antigen-Antibody Complex/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/blood , Male , Female , Middle Aged , Synovial Fluid/immunology , Synovial Fluid/metabolism , Adult , Aged , Biomarkers , Case-Control Studies , Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/metabolism , Synovial Membrane/immunology , Synovial Membrane/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/blood
4.
Int Immunopharmacol ; 141: 112863, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39146779

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease that causes persistent inflammation involving the joints, cartilage, and synovium. In individuals with RA, alterations in the composition of intestinal bacteria suggest the vital role of gut microbiota in immune dysfunction. Multiple therapies commonly used to treat RA can also alter the diversity of gut microbiota, further suggesting the modulation of gut microbiota as a prevention or treatment for RA. Therefore, a better understanding of the changes in the gut microbiota that accompany RA should facilitate the development of novel therapeutic approaches. In this study, B. coagulans BACO-17 not only significantly reduced paw swelling, arthritis scores, and hind paw and forepaw thicknesses but also protected articular cartilage and the synovium against RA degeneration, with a corresponding downregulation of TNF-α expression. The inhibition or even reversing of RA progression highlights B. coagulans BACO-17 as a novel therapeutic for RA worth investigating.


Subject(s)
Arthritis, Rheumatoid , Bacillus coagulans , Disease Progression , Gastrointestinal Microbiome , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Gastrointestinal Microbiome/drug effects , Male , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Tumor Necrosis Factor-alpha/metabolism , Synovial Membrane/immunology , Synovial Membrane/pathology , Synovial Membrane/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Probiotics/therapeutic use , Humans , Mice , Rats , Chondrocytes/drug effects , Chondrocytes/immunology
5.
Front Immunol ; 15: 1381272, 2024.
Article in English | MEDLINE | ID: mdl-39139555

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease with a complex pathological mechanism involving autoimmune response, local inflammation and bone destruction. Metabolic pathways play an important role in immune-related diseases and their immune responses. The pathogenesis of rheumatoid arthritis may be related to its metabolic dysregulation. Moreover, histological techniques, including genomics, transcriptomics, proteomics and metabolomics, provide powerful tools for comprehensive analysis of molecular changes in biological systems. The present study explores the molecular and metabolic mechanisms of RA, emphasizing the central role of metabolic dysregulation in the RA disease process and highlighting the complexity of metabolic pathways, particularly metabolic remodeling in synovial tissues and its association with cytokine-mediated inflammation. This paper reveals the potential of histological techniques in identifying metabolically relevant therapeutic targets in RA; specifically, we summarize the genetic basis of RA and the dysregulated metabolic pathways, and explore their functional significance in the context of immune cell activation and differentiation. This study demonstrates the critical role of histological techniques in decoding the complex metabolic network of RA and discusses the integration of histological data with other types of biological data.


Subject(s)
Arthritis, Rheumatoid , Biomarkers , Metabolomics , Proteomics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Humans , Metabolomics/methods , Proteomics/methods , Genomics/methods , Animals , Metabolic Networks and Pathways , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Multiomics
6.
Nat Commun ; 15(1): 7503, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39209814

ABSTRACT

Rheumatoid arthritis (RA) is a complex immune-mediated inflammatory disorder in which patients suffer from inflammatory-erosive arthritis. Recent advances on histopathology heterogeneity of RA synovial tissue revealed three distinct phenotypes based on cellular composition (pauci-immune, diffuse and lymphoid), suggesting that distinct etiologies warrant specific targeted therapy which motivates a need for cost effective phenotyping tools in preclinical and clinical settings. To this end, we developed an automated multi-scale computational pathotyping (AMSCP) pipeline for both human and mouse synovial tissue with two distinct components that can be leveraged together or independently: (1) segmentation of different tissue types to characterize tissue-level changes, and (2) cell type classification within each tissue compartment that assesses change across disease states. Here, we demonstrate the efficacy, efficiency, and robustness of the AMSCP pipeline as well as the ability to discover novel phenotypes. Taken together, we find AMSCP to be a valuable cost-effective method for both pre-clinical and clinical research.


Subject(s)
Arthritis, Rheumatoid , Synovial Membrane , Humans , Synovial Membrane/pathology , Synovial Membrane/immunology , Animals , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/immunology , Mice , Phenotype , Computational Biology/methods , Inflammation/pathology
7.
Article in English | MEDLINE | ID: mdl-38932672

ABSTRACT

Rheumatoid arthritis (RA) is a complex autoimmune disease that leads to joint destruction. A number of immune cells that affect joint tissues are involved in the pathogenesis of this disease. This leads to the synthesis of many pro-inflammatory mediators. The transport of drugs, as well as many cytokines involved in the development of inflammation in RA patients, is mediated by membrane transporters. Membrane transporters are proteins that mediate the transfer of substrates across biological membranes. But to date there are no studies examining the expression of solute carrier (SLC) transporters in joint tissues. The aim of the study was to evaluate the expression of individual SLC family transporters in the synovial membranes (SMs) and infrapatellar fat pad (Hoffa's pad) of RA patients. The study included 20 patients with rheumatoid arthritis and 20 with osteoarthritis as the control group who were undergoing joint replacement surgery as a normal part of clinical care. In the SM and Hoffa's pad of RA patients the following 17 membrane transporters were defined at relevant expression levels for SLC transporter superfamily: SLC15A2, SLC16A3, SLC19A1, SLC2A9, SLC22A1, SLC22A3, SLC22A4, SLC22A5, SLC22A18, SLC33A1, SLC47A1, SLC51A, SLC7A5, SLC7A6, SLC01C1, SLC02B1, SLC04A1. The confirmed expression of these transporters in the SMs as well as Hoffa's pad of patients with RA and OA, and the differences in their expression between these groups, suggests the involvement of SLC transporters in both the maintenance of homeostasis under physiological conditions in the tissues of the joints, as well as in the inflammatory process in RA.


Subject(s)
Arthritis, Rheumatoid , Solute Carrier Proteins , Synovial Membrane , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/immunology , Female , Synovial Membrane/metabolism , Synovial Membrane/immunology , Middle Aged , Solute Carrier Proteins/metabolism , Male , Aged , Adipose Tissue/metabolism , Adult , Membrane Transport Proteins/metabolism , Biological Transport , Osteoarthritis/metabolism
8.
J Autoimmun ; 147: 103263, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38851089

ABSTRACT

RATIONALE: In inflammatory diseases such as rheumatoid arthritis (RA), steroid metabolism is a central component mediating the actions of immuno-modulatory glucocorticoids and sex steroids. However, the regulation and function of cellular steroid metabolism within key leukocyte populations such as macrophages remain poorly defined. In this study, the inflammatory regulation of global steroid metabolism was assessed in RA macrophages. METHODS: Bulk RNA-seq data from RA synovial macrophages was used to assess transcripts encoding key enzymes in steroid metabolism and signalling. Changes in metabolism were assessed in synovial fluids, correlated to measures of disease activity and functionally validated in primary macrophage cultures. RESULTS: RNA-seq revealed a unique pattern of differentially expressed genes, including changes in genes encoding the enzymes 11ß-HSD1, SRD5A1, AKR1C2 and AKR1C3. These correlated with disease activity, favouring increased glucocorticoid and androgen levels. Synovial fluid 11ß-HSD1 activity correlated with local inflammatory mediators (TNFα, IL-6, IL-17), whilst 11ß-HSD1, SRD5A1 and AKR1C3 activity correlated with systemic measures of disease and patient pain (ESR, DAS28 ESR, global disease activity). Changes in enzyme activity were evident in inflammatory activated macrophages in vitro and revealed a novel androgen activating role for 11ß-HSD1. Together, increased glucocorticoids and androgens were able to suppress inflammation in macrophages and fibroblast-like-synoviocytes. CONCLUSIONS: This study underscores the significant increase in androgen and glucocorticoid activation within inflammatory polarized macrophages of the synovium, contributing to local suppression of inflammation. The diminished profile of inactive steroid precursors in postmenopausal women may contribute to disturbances in this process, leading to increased disease incidence and severity.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Arthritis, Rheumatoid , Inflammation , Macrophages , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Macrophages/metabolism , Macrophages/immunology , Inflammation/metabolism , Inflammation/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Synovial Fluid/metabolism , Synovial Fluid/immunology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Male , Female , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/immunology , Cells, Cultured , Glucocorticoids/metabolism , Steroids/metabolism , Gene Expression Regulation , Hydroxysteroid Dehydrogenases
9.
Nat Commun ; 15(1): 4991, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862501

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease involving antigen-specific T and B cells. Here, we perform single-cell RNA and repertoire sequencing on paired synovial tissue and blood samples from 12 seropositive RA patients. We identify clonally expanded CD4 + T cells, including CCL5+ cells and T peripheral helper (Tph) cells, which show a prominent transcriptomic signature of recent activation and effector function. CD8 + T cells show higher oligoclonality than CD4 + T cells, with the largest synovial clones enriched in GZMK+ cells. CD8 + T cells with possibly virus-reactive TCRs are distributed across transcriptomic clusters. In the B cell compartment, NR4A1+ activated B cells, and plasma cells are enriched in the synovium and demonstrate substantial clonal expansion. We identify synovial plasma cells that share BCRs with synovial ABC, memory, and activated B cells. Receptor-ligand analysis predicted IFNG and TNFRSF members as mediators of synovial Tph-B cell interactions. Together, these results reveal clonal relationships between functionally distinct lymphocyte populations that infiltrate the synovium of patients with RA.


Subject(s)
Arthritis, Rheumatoid , B-Lymphocytes , Synovial Membrane , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Synovial Membrane/immunology , Synovial Membrane/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Female , Male , Middle Aged , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Single-Cell Analysis , Transcriptome , Plasma Cells/immunology , Plasma Cells/metabolism , Aged , Lymphocyte Activation , Adult
10.
Front Immunol ; 15: 1361606, 2024.
Article in English | MEDLINE | ID: mdl-38846937

ABSTRACT

Introduction: Pathological changes in the articular cartilage (AC) and synovium are major manifestations of osteoarthritis (OA) and are strongly associated with pain and functional limitations. Exosome-derived microRNAs (miRNAs) are crucial regulatory factors in intercellular communication and can influence the progression of OA by participating in the degradation of chondrocytes and the phenotypic transformation in the polarization of synovial macrophages. However, the specific relationships and pathways of action of exosomal miRNAs in the pathological progression of OA in both cartilage and synovium remain unclear. Methods: This study evaluates the effects of fibroblast-like synoviocyte (FLS)-derived exosomes (FLS-Exos), influenced by miR-146a, on AC degradation and synovial macrophage polarization. We investigated the targeted relationship between miR-146a and TRAF6, both in vivo and in vitro, along with the involvement of the NF-κB signaling pathway. Results: The expression of miR-146a in the synovial exosomes of OA rats was significantly higher than in healthy rats. In vitro, the upregulation of miR-146a reduced chondrocyte apoptosis, whereas its downregulation had the opposite effect. In vivo, exosomes derived from miR-146a-overexpressing FLSs (miR-146a-FLS-Exos) reduced AC injury and chondrocyte apoptosis in OA. Furthermore, synovial proliferation was reduced, and the polarization of synovial macrophages shifted from M1 to M2. Mechanistically, the expression of TRAF6 was inhibited by targeting miR-146a, thereby modulating the Toll-like receptor 4/TRAF6/NF-κB pathway in the innate immune response. Discussion: These findings suggest that miR-146a, mediated through FLS-Exos, may alleviate OA progression by modulating cartilage degradation and macrophage polarization, implicating the NF-κB pathway in the innate immune response. These insights highlight the therapeutic potential of miR-146a as a protective agent in OA, underscoring the importance of exosomal miRNAs in the pathogenesis and potential treatment of the disease.


Subject(s)
Exosomes , Macrophages , MicroRNAs , Osteoarthritis , Synoviocytes , TNF Receptor-Associated Factor 6 , MicroRNAs/genetics , Animals , Exosomes/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/immunology , Rats , Macrophages/immunology , Macrophages/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , Male , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , NF-kappa B/metabolism , Signal Transduction , Rats, Sprague-Dawley , Fibroblasts/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/immunology , Cells, Cultured , Apoptosis , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Macrophage Activation
11.
Front Immunol ; 15: 1385006, 2024.
Article in English | MEDLINE | ID: mdl-38895122

ABSTRACT

Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.


Subject(s)
Fibroblasts , Fibrosis , Osteoarthritis , Phenotype , Synoviocytes , Humans , Osteoarthritis/pathology , Osteoarthritis/immunology , Osteoarthritis/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/immunology , Animals , Synoviocytes/metabolism , Synoviocytes/pathology , Synoviocytes/immunology , Synovial Membrane/pathology , Synovial Membrane/immunology , Synovial Membrane/metabolism
12.
Arthritis Rheumatol ; 76(9): 1439-1445, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38782587

ABSTRACT

OBJECTIVE: The objective is to characterize transcriptomic profiles and immune cell composition and distribution in juvenile idiopathic arthritis (JIA) synovial biopsies, assess for associations of these features with clinical parameters, and compare JIA and rheumatoid arthritis (RA) synovial features. METHODS: RNA sequencing (RNASeq) was performed on 24 samples, with pathway analysis and inference of relative abundance of immune cell subsets based on gene expression data. Two multiplex fluorescence immunohistochemistry (IHC) panels were performed on 28 samples (including 13 on which RNASeq was performed), staining for CD206- classical and CD206+ nonclassical macrophages, and CD8+ and CD4+ T and B lymphocytes. Data were compared to a published series of early RA synovial biopsies. RESULTS: Pathway analysis of the most variably expressed genes (n = 339) identified a B and plasma cell signature as the main driver of heterogeneity in JIA synovia, with strong overlap between JIA and RA synovitis. Multiplex IHC confirmed heterogeneity of immune cell infiltration. M1-like macrophage-rich synovial lining was associated with greater lining hypertrophy and higher (CD45+) pan-immune cell and CD8+ T cell infiltration. CONCLUSION: Our study indicates significant similarities between JIA and RA synovitis. Similar to RA, JIA synovia may be broadly categorized into two groups: (1) those with an inflammatory/adaptive immune transcriptomic signature, M1-like macrophage and CD8+ T cell infiltration, and thicker, M1-like macrophage-rich synovial lining, and (2) those with an M2-like macrophage transcriptomic signature, greater M2/M1-like macrophage ratios, and thinner, M2-like macrophage-rich synovial lining. Synovial features were not significantly associated with clinical parameters, likely because of group size and heterogeneity.


Subject(s)
Arthritis, Juvenile , Arthritis, Rheumatoid , B-Lymphocytes , Macrophages , Synovial Membrane , Humans , Arthritis, Juvenile/pathology , Arthritis, Juvenile/immunology , Synovial Membrane/pathology , Synovial Membrane/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/genetics , Macrophages/pathology , Macrophages/immunology , Biopsy , Male , Female , Child , B-Lymphocytes/pathology , B-Lymphocytes/immunology , Transcriptome , Adolescent , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , Synovitis/pathology , Synovitis/immunology , Synovitis/genetics , Plasma Cells/pathology , Plasma Cells/immunology , Immunohistochemistry , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology
13.
Immunol Res ; 72(4): 797-810, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38722530

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial hyperplasia, pannus formation, and cartilage and bone destruction. Lysine-specific demethylase 1 (LSD1), an enzyme involved in transcriptional regulation, has an unclear role in synovial inflammation, fibroblast-like synoviocytes migration, and invasion during RA pathogenesis. In this study, we observed increased LSD1 expression in RA synovial tissues and in TNF-α-stimulated MH7A cells. SP2509, an LSD1 antagonist, directly reduced LSD1 expression and reversed the elevated levels of proteins associated with inflammation, apoptosis, proliferation, and autophagy induced by TNF-α. Furthermore, SP2509 inhibited the migratory capacity of MH7A cells, which was enhanced by TNF-α. In CIA models, SP2509 treatment ameliorated RA development, reducing the expression of pro-inflammatory cytokines and alleviating joint pathological symptoms. These findings underscore the significance of LSD1 in RA and propose the therapeutic potential of SP2509.


Subject(s)
Arthritis, Rheumatoid , Histone Demethylases , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/immunology , Histone Demethylases/metabolism , Humans , Animals , Mice , Synoviocytes/metabolism , Synoviocytes/pathology , Synoviocytes/immunology , Disease Progression , Tumor Necrosis Factor-alpha/metabolism , Cell Movement , Cell Line , Male , Synovial Membrane/pathology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Apoptosis , Arthritis, Experimental/immunology , Cytokines/metabolism , Autophagy , Cell Proliferation/drug effects
15.
Int Immunopharmacol ; 136: 112343, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810305

ABSTRACT

Defective clearance of apoptotic cells due to impaired efferocytosis sustains error in self-tolerance that exacerbates rheumatoid arthritis (RA). However, the molecular determinant that directly or specifically impairs efferocytosis in RA is not yet studied. We identified a new perspective that IL-17A significantly impedes efferocytosis via preferential activation of the JAK/STAT-3/ADAM17 signaling axis. In contrast, disruption of the IL-17A/IL-17RA interaction using cyanidin or silencing of IL-17RA obstructed JAK/STAT-3 activation that further abolished ADAM17 expression. Subsequent depletion of ADAM17 inhibited the shedding of Mer tyrosine kinase receptor (MERTK), which significantly increased apoptotic cell intake and restored efferocytosis in adjuvant-induced arthritic (AA) model. Concomitantly, the amplification of the efferocytosis process due to IL-17A/IL-17RA interaction disruption was sensitive to mitochondrial fission mediated via Drp-1 phosphorylation downstream of STAT-3 inhibition. As expected, cyanidin treated AA synovial macrophages that exhibited increased efferocytosis demonstrated a phenotypic shift towards CD163 anti-inflammatory phenotype in a STAT-5 dependent manner. Similar results were obtained in IL-17A-sensitized AA synovial macrophages treated with S3I-201 (a STAT-3 inhibitor) indicating that IL-17A influences efferocytosis via the STAT-3 pathway. In view of our previous work where cyanidin restored Th17/Treg balance, our present investigation fulfils a critical gap by providing scientific validation that cyanidin escalated PD-L1 expression during the efferocytosis process that could have impacted the restoration of Th17/Treg balance in an AA model. Together, these data corroborate the hypothesis that IL-17A signaling can impair efferocytosis via regulating STAT-3/ADAM17/FL-MERTK axis and that its inhibition can amplify a pro-resolution signal against RA progression.


Subject(s)
Arthritis, Rheumatoid , B7-H1 Antigen , Interleukin-17 , Macrophages , Receptors, Interleukin-17 , STAT3 Transcription Factor , Signal Transduction , Interleukin-17/metabolism , Animals , STAT3 Transcription Factor/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/drug therapy , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , B7-H1 Antigen/metabolism , Mice , Receptors, Interleukin-17/metabolism , Receptors, Interleukin-17/genetics , Anthocyanins/pharmacology , Arthritis, Experimental/immunology , Humans , Disease Models, Animal , Phagocytosis/drug effects , Male , Synovial Membrane/immunology , Synovial Membrane/pathology , Mice, Inbred C57BL , Apoptosis/drug effects , Efferocytosis
16.
Front Immunol ; 15: 1355824, 2024.
Article in English | MEDLINE | ID: mdl-38799447

ABSTRACT

Objectives: IL26 levels are elevated in the blood and synovial fluid of patients with inflammatory arthritis. IL26 can be produced by Th17 cells and locally within joints by tissue-resident cells. IL26 induces osteoblast mineralization in vitro. As osteoproliferation and Th17 cells are important factors in the pathogenesis of axial spondyloarthritis (axSpA), we aimed to clarify the cellular sources of IL26 in spondyloarthritis. Methods: Serum, peripheral blood mononuclear cells (n = 15-35) and synovial tissue (n = 3-9) of adult patients with axSpA, psoriatic arthritis (PsA) and rheumatoid arthritis (RA) and healthy controls (HCs, n = 5) were evaluated by ELISA, flow cytometry including PrimeFlow assay, immunohistochemistry and immunofluorescence and quantitative PCR. Results: Synovial tissue of axSpA patients shows significantly more IL26-positive cells than that of HCs (p < 0.01), but numbers are also elevated in PsA and RA patients. Immunofluorescence shows co-localization of IL26 with CD68, but not with CD3, SMA, CD163, cadherin-11, or CD90. IL26 is elevated in the serum of RA and PsA (but not axSpA) patients compared with HCs (p < 0.001 and p < 0.01). However, peripheral blood CD4+ T cells from axSpA and PsA patients show higher positivity for IL26 in the PrimeFlow assay compared with HCs. CD4+ memory T cells from axSpA patients produce more IL26 under Th17-favoring conditions (IL-1ß and IL-23) than cells from PsA and RA patients or HCs. Conclusion: IL26 production is increased in the synovial tissue of SpA and can be localized to CD68+ macrophage-like synoviocytes, whereas circulating IL26+ Th17 cells are only modestly enriched. Considering the osteoproliferative properties of IL26, this offers new therapeutic options independent of Th17 pathways.


Subject(s)
Antigens, CD , Arthritis, Psoriatic , Interleukins , Synoviocytes , Humans , Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/metabolism , Synoviocytes/metabolism , Synoviocytes/immunology , Synoviocytes/pathology , Male , Adult , Female , Antigens, CD/metabolism , Interleukins/metabolism , Interleukins/blood , Middle Aged , Antigens, Differentiation, Myelomonocytic/metabolism , Axial Spondyloarthritis/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Joints/pathology , Joints/immunology , Joints/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology
17.
J Control Release ; 370: 747-762, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740094

ABSTRACT

Rheumatoid arthritis (RA) is a progressive autoimmune disease and drug therapy has been restricted due to poor therapeutic efficacy and adverse effects. In RA synovium, dendritic cells present self-antigens to activate cascade immune pathway. Furthermore, downstream macrophages secrete high levels of pro-inflammatory cytokines; Hyperplasia of activated synovial fibroblasts (FLS) is responsible for hypoxic synovium microenvironment, secretion of cytokines/chemokines and erosion of bone/cartilage tissues. Positive feedback loop of inflammation between macrophages and FLS independent of antigen-presentation is constructed. Herein, an injectable pH-sensitive peptide hydrogel encapsulating siRNA/Methotrexate-polyethyleneimine (siMP, including sip65MP, sip38MP, siCD86MP) and Bismuthene nanosheet/Methotrexate-polyethyleneimine (BiMP) is successfully developed. Among them, siCD86MP reduces protein level of co-stimulatory molecule CD86 while sip65MP and sip38MP separately inhibit NF-κB and MAPK-p38 pathways of macrophages and FLS to suppress secretion of cytokines and MMPs. Meanwhile, reduction in anti-apoptotic property of FLS induced by inhibition of NF-κB pathway has a synergistic effect with photodynamic therapy (PDT) and photothermal therapy (PTT) mediated by BiMP for FLS elimination, effectively ameliorating hypoxic synovium microenvironment. After being injected into synovium, hydrogel responds to acidic microenvironment and serves as a reservoir for sustained drug release and inherent retention capacity of which enables cationic nanoparticles to bypass tissue barrier for precise synovium targeting. This brand-new drug delivery system combines modulating cascade immune pathway from beginning to end by RNAi and eliminating FLS for improving synovium microenvironment by phototherapy together, providing a robust strategy for clinical RA treatment.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Hydrogels , Methotrexate , Synovial Membrane , Fibroblasts/drug effects , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Hydrogels/administration & dosage , Synovial Membrane/immunology , Animals , Methotrexate/administration & dosage , Methotrexate/pharmacology , RNA, Small Interfering/administration & dosage , Photochemotherapy/methods , Mice , Humans , Macrophages/drug effects , Macrophages/immunology , RAW 264.7 Cells , Cytokines/metabolism , Antirheumatic Agents/administration & dosage , Cellular Microenvironment/drug effects , NF-kappa B/metabolism , Phototherapy/methods , Peptides/administration & dosage
18.
Front Immunol ; 15: 1394108, 2024.
Article in English | MEDLINE | ID: mdl-38799455

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent synovial inflammation and progressive joint destruction. Macrophages are key effector cells that play a central role in RA pathogenesis through their ability to polarize into distinct functional phenotypes. An imbalance favoring pro-inflammatory M1 macrophages over anti-inflammatory M2 macrophages disrupts immune homeostasis and exacerbates joint inflammation. Multiple signaling pathways, including Notch, JAK/STAT, NF-κb, and MAPK, regulate macrophage polarization towards the M1 phenotype in RA. Metabolic reprogramming also contributes to this process, with M1 macrophages prioritizing glycolysis while M2 macrophages utilize oxidative phosphorylation. Redressing this imbalance by modulating macrophage polarization and metabolic state represents a promising therapeutic strategy. Furthermore, complex bidirectional interactions exist between synovial macrophages and fibroblast-like synoviocytes (FLS), forming a self-perpetuating inflammatory loop. Macrophage-derived factors promote aggressive phenotypes in FLS, while FLS-secreted mediators contribute to aberrant macrophage activation. Elucidating the signaling networks governing macrophage polarization, metabolic adaptations, and crosstalk with FLS is crucial to developing targeted therapies that can restore immune homeostasis and mitigate joint pathology in RA.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Macrophage Activation , Macrophages , Signal Transduction , Synovial Membrane , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Macrophages/immunology , Macrophages/metabolism , Synovial Membrane/metabolism , Synovial Membrane/immunology , Synovial Membrane/pathology , Fibroblasts/metabolism , Fibroblasts/immunology , Animals , Macrophage Activation/immunology , Cell Communication/immunology , Metabolic Reprogramming
19.
Front Immunol ; 15: 1387311, 2024.
Article in English | MEDLINE | ID: mdl-38711508

ABSTRACT

Background: Rheumatoid arthritis (RA) is a systemic immune-related disease characterized by synovial inflammation and destruction of joint cartilage. The pathogenesis of RA remains unclear, and diagnostic markers with high sensitivity and specificity are needed urgently. This study aims to identify potential biomarkers in the synovium for diagnosing RA and to investigate their association with immune infiltration. Methods: We downloaded four datasets containing 51 RA and 36 healthy synovium samples from the Gene Expression Omnibus database. Differentially expressed genes were identified using R. Then, various enrichment analyses were conducted. Subsequently, weighted gene co-expression network analysis (WGCNA), random forest (RF), support vector machine-recursive feature elimination (SVM-RFE), and least absolute shrinkage and selection operator (LASSO) were used to identify the hub genes for RA diagnosis. Receiver operating characteristic curves and nomogram models were used to validate the specificity and sensitivity of hub genes. Additionally, we analyzed the infiltration levels of 28 immune cells in the expression profile and their relationship with the hub genes using single-sample gene set enrichment analysis. Results: Three hub genes, namely, ribonucleotide reductase regulatory subunit M2 (RRM2), DLG-associated protein 5 (DLGAP5), and kinesin family member 11 (KIF11), were identified through WGCNA, LASSO, SVM-RFE, and RF algorithms. These hub genes correlated strongly with T cells, natural killer cells, and macrophage cells as indicated by immune cell infiltration analysis. Conclusion: RRM2, DLGAP5, and KIF11 could serve as potential diagnostic indicators and treatment targets for RA. The infiltration of immune cells offers additional insights into the underlying mechanisms involved in the progression of RA.


Subject(s)
Arthritis, Rheumatoid , Gene Expression Profiling , Gene Regulatory Networks , Machine Learning , Ribonucleoside Diphosphate Reductase , Humans , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/diagnosis , Transcriptome , Synovial Membrane/metabolism , Synovial Membrane/immunology , Kinesins/genetics , Biomarkers , Databases, Genetic , Computational Biology/methods , Support Vector Machine
20.
Arthritis Rheumatol ; 76(8): 1263-1277, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38570939

ABSTRACT

OBJECTIVE: Subsets of CD21-/low memory B cells (MBCs), including double-negative (DN, CD27-IgD-) and Tbet+CD11c+ cells, are expanded in chronic inflammatory diseases. In rheumatoid arthritis (RA), CD21-/low MBCs correlate with joint destruction. However, whether this is due to the Tbet+CD11c+ subset, its function and pathogenic contribution to RA are unknown. This study aims to investigate the association between CD21-/lowTbet+CD11c+ MBCs and joint destruction as well as other clinical parameters and to elucidate their functional properties in patients with untreated RA (uRA). METHODS: Clinical observations were combined with flow cytometry (n = 36) and single-cell RNA sequencing (scRNA-seq) and V(D)J sequencing (n = 4) of peripheral blood (PB) MBCs from patients with uRA. The transcriptome of circulating Tbet+CD11c+ MBCs was compared with scRNA-seq data of synovial B cells. In vitro coculture of Tbet+CD11c+ B cells with T cells was used to assess costimulatory capacity. RESULTS: CD21-/lowTbet+CD11c+ MBCs in PB correlated with bone destruction but no other clinical parameters analyzed. The Tbet+CD11c+ MBCs have undergone clonal expansion and express somatically mutated V genes. Gene expression analysis of these cells identified a unique signature of more than 150 up-regulated genes associated with antigen presentation functions, including B cell receptor activation and clathrin-mediated antigen internalization; regulation of actin filaments, endosomes, and lysosomes; antigen processing, loading, presentation, and costimulation; a transcriptome mirrored in their synovial tissue counterparts. In vitro, Tbet+CD11c+ B cells induced retinoic acid receptor-related orphan nuclear receptor γT expression in CD4+ T cells, thereby polarizing to Th17 cells, a T cell subset critical for osteoclastogenesis and associated with bone destruction. CONCLUSION: This study suggests that Tbet+CD11c+ MBCs contribute to the pathogenesis of RA by promoting bone destruction through antigen presentation, T cell activation, and Th17 polarization.


Subject(s)
Arthritis, Rheumatoid , CD11c Antigen , Humans , Arthritis, Rheumatoid/immunology , CD11c Antigen/metabolism , CD11c Antigen/immunology , Male , Middle Aged , Female , B-Lymphocytes/immunology , Memory B Cells/immunology , Aged , Antigen Presentation/immunology , Adult , Synovial Membrane/immunology
SELECTION OF CITATIONS
SEARCH DETAIL