Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.381
Filter
1.
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
2.
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
3.
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
5.
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
6.
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
7.
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
8.
Nat Rev Rheumatol ; 20(5): 258-271, 2024 May.
Article in English | MEDLINE | ID: mdl-38600215

ABSTRACT

In rheumatoid arthritis, juvenile idiopathic arthritis and other forms of inflammatory arthritis, the immune system targets certain joints but not others. The pattern of joints affected varies by disease and by individual, with flares most commonly involving joints that were previously inflamed. This phenomenon, termed joint-specific memory, is difficult to explain by systemic immunity alone. Mechanisms of joint-specific memory include the involvement of synovial resident memory T cells that remain in the joint during remission and initiate localized disease recurrence. In addition, arthritis-induced durable changes in synovial fibroblasts and macrophages can amplify inflammation in a site-specific manner. Together with ongoing systemic processes that promote extension of arthritis to new joints, these local factors set the stage for a stepwise progression in disease severity, a paradigm for arthritis chronicity that we term the joint accumulation model. Although durable drug-free remission through early treatment remains elusive for most forms of arthritis, the joint accumulation paradigm defines new therapeutic targets, emphasizes the importance of sustained treatment to prevent disease extension to new joints, and identifies a rolling window of opportunity for altering the natural history of arthritis that extends well beyond the initiation phase of disease.


Subject(s)
Arthritis, Rheumatoid , Memory T Cells , Humans , Memory T Cells/immunology , Arthritis, Rheumatoid/immunology , Joints/immunology , Joints/pathology , Immunologic Memory/immunology , Disease Progression , Animals , Synovial Membrane/immunology , Synovial Membrane/pathology , Arthritis/immunology
9.
Int Immunopharmacol ; 132: 111913, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38603855

ABSTRACT

Resident synoviocytes and synovial microvasculature, together with immune cells from circulation, contribute to pannus formation, the main pathological feature of rheumatoid arthritis (RA), leading to destruction of adjacent cartilage and bone. Seeds, fibroblast-like synoviocytes (FLSs), macrophages, dendritic cells (DCs), B cells, T cells and endothelial cells (ECs) seeds with high metabolic demands undergo metabolic reprogramming from oxidative phosphorylation to glycolysis in response to poor soil of RA synovium with hypoxia, nutrient deficiency and inflammatory stimuli. Glycolysis provides rapid energy supply and biosynthetic precursors to support pathogenic growth of these seeds. The metabolite lactate accumulated during this process in turn condition the soil microenvironment and affect seeds growth by modulating signalling pathways and directing lactylation modifications. This review explores in depth the survival mechanism of seeds with high metabolic demands in the poor soil of RA synovium, providing useful support for elucidating the etiology of RA. In addition, we discuss the role and major post-translational modifications of proteins and enzymes linked to glycolysis to inspire the discovery of novel anti-rheumatic targets.


Subject(s)
Arthritis, Rheumatoid , Glycolysis , Synovial Membrane , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Humans , Animals , Synovial Membrane/pathology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , Signal Transduction
10.
Int Immunopharmacol ; 132: 112016, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38593506

ABSTRACT

Osteoarthritis (OA) is a low-grade inflammatory joint illness in which monocytes migrate and infiltrate synovial tissue, differentiating into the pro-inflammatory M1 macrophage phenotype. IL-17 is a proinflammatory mediator principally generated by Th17 cells, which is elevated in OA patients; nevertheless, investigators have yet to elucidate the function of IL-17 in M1 polarization during OA development. Our analysis of clinical tissues and results from the open online dataset discovered that the level of M1 macrophage markers is elevated in human OA tissue samples than in normal tissue. High-throughput screening demonstrated that MCP-1 is a potential candidate factor after IL-17 treatment in OA synovial fibroblasts (OASFs). Immunohistochemistry data revealed that the level of MCP-1 is higher in humans and mice with OA than in normal tissues. IL-17 stimulation facilitates MCP-1-dependent macrophage polarization to the M1 phenotype. It also appears that IL-17 enhances MCP-1 synthesis in human OASFs, enhancing monocyte migration via the JAK and STAT3 signaling cascades. Our findings indicate the IL-17/MCP-1 axis as a novel strategy for the remedy of OA.


Subject(s)
Cell Movement , Chemokine CCL2 , Interleukin-17 , Macrophages , Monocytes , Osteoarthritis , Animals , Humans , Male , Mice , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Interleukin-17/metabolism , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/drug effects , Monocytes/metabolism , Osteoarthritis/immunology , Signal Transduction , STAT3 Transcription Factor/metabolism , Synovial Membrane/immunology , Synovial Membrane/pathology
11.
Int J Immunogenet ; 51(3): 130-142, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38462560

ABSTRACT

Osteoarthritis (OA) is one of the most common degenerative diseases characterised by joint pain, swelling and decreased mobility, with its main pathological features being articular synovitis, cartilage degeneration and osteophyte formation. Inflammatory cytokines and chemokines secreted by activated immunocytes can trigger various inflammatory and immune responses in articular cartilage and synovium, contributing to the genesis and development of OA. A series of monocyte/macrophage chemokines, including monocyte chemotaxis protein (MCP)-1/CCL2, MCP2/CCL8, macrophage inflammatory protein (MIP)-1α/CCL3, MIP-1ß/CCL4, MIP-3α/CCL20, regulated upon activation, normal T-cell expressed and secreted /CCL5, CCL17 and macrophage-derived chemokine/CCL22, was proven to transmit cell signals by binding to G protein-coupled receptors on recipient cell surface, mediating and promoting inflammation in OA joints. However, the underlying mechanism of these chemokines in the pathogenesis of OA remains still elusive. Here, published literature was reviewed, and the function and mechanisms of monocyte/macrophage chemokines in OA pathogenesis were summarised. The symptoms and disease progression of OA were found to be effectively alleviated when the expression of these chemokines is inhibited. Elucidating these mechanisms could contribute to further understand how OA develops and provide potential targets for the early diagnosis of arthritis and drug treatment to delay or even halt OA progression.


Subject(s)
Chemokines , Macrophages , Monocytes , Osteoarthritis , Humans , Osteoarthritis/immunology , Osteoarthritis/pathology , Osteoarthritis/metabolism , Chemokines/metabolism , Monocytes/immunology , Monocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Cartilage, Articular/pathology , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Synovial Membrane/immunology , Synovial Membrane/pathology , Synovial Membrane/metabolism
12.
Immunol Med ; 47(2): 58-67, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38168669

ABSTRACT

Synovial tissue-resident macrophages (STRMs) maintain normal joint homeostasis in a steady state. However, it is unclear whether STRMs still play homeostatic roles or change the functions in the joint of rheumatoid arthritis (RA), where infiltrating peripheral blood monocyte-derived macrophages (PBMoMs) play proinflammatory roles. In the present study, we examined changes in the phenotypes and functions of STRMs in response to RA-related stimuli in vitro. STRMs were prepared from non-inflammatory osteoarthritis (OA) joint synovium, which is histologically indistinguishable from normal joint synovium. PBMoMs were prepared and used for comparison. After stimulation with plate-bound IgG, which mimics anti-citrullinated protein antibody immunocomplex formed in RA joints, or with combinations of RA-related inflammatory mediators, namely tumor necrosis factor-α (TNF-α) and prostaglandin E2 or interferon-γ, PBMoMs downregulated surface markers and genes associated with anti-inflammatory macrophages, and upregulated cytokine and marker genes of proinflammatory macrophages in RA. On the other hand, STRMs hardly changed the expression of surface molecules and marker genes but altered the pattern of cytokine gene expression after stimulation like PBMoMs. Furthermore, in vitro stimulated STRMs promote proinflammatory functions of cocultured synovial fibroblasts. Thus, STRMs might play proinflammatory roles in RA joints, while maintaining their phenotypes in the steady state.


Subject(s)
Arthritis, Rheumatoid , Macrophages , Phenotype , Synovial Membrane , Humans , Synovial Membrane/immunology , Macrophages/immunology , Macrophages/physiology , Arthritis, Rheumatoid/immunology , Tumor Necrosis Factor-alpha/metabolism , Cytokines/metabolism , Male , Fibroblasts/immunology , Osteoarthritis/immunology , Osteoarthritis/etiology , Cells, Cultured , Female , Dinoprostone/metabolism , Middle Aged , Aged , Inflammation/immunology , Inflammation Mediators/metabolism
13.
Cell Immunol ; 383: 104655, 2023 01.
Article in English | MEDLINE | ID: mdl-36516652

ABSTRACT

Using a murine collagen-induced arthritis model, we characterized the heterogeneity of synovial CD8+ T cells based on the expression of chemokine receptors, cytokines, and nuclear transcription factors. Four subsets, i.e. CXCR3-CCR4- cells, CXCR3+CCR4- cells, CXCR3+CCR4+ cells, and CXCR3-CCR4+ cells, were present in synovial CD8+CD62L-CCR6+IL-23R+CCR10- T cells. CXCR3-CCR4- cells belonged to exhausted CD8+ T cells. CXCR3+CCR4- cells were Tc17.1 cells expressing both IL-17A and IFN-γ. CXCR3+CCR4+ cells were transitional Tc17.1 cells expressing IL-17A but lower IFN-γ, and CXCR3-CCR4+ cells were Tc17 cells expressing IL-17A but no IFN-γ. Transitional Tc17.1 cells can differentiate into Tc17.1 cells in vitro under the instruction of IL-12. Tc17.1 cells and transitional Tc17.1 cells strongly induced the expression of pro-inflammatory mediators in synovial fibroblasts, whereas Tc17 cells were less potent in doing so. IFN-γ was involved in the higher pathogenicity of Tc17.1 cells and transitional Tc17.1 cells on synovial fibroblasts. This study expands the understanding of Tc17 biology by unveiling the phenotypic and functional heterogeneity of synovial IL-17A-expressing CD8+ T cells. These heterogeneous IL-17A-expressing CD8+ T cells could be novel therapeutic targets in future arthritis treatment.


Subject(s)
Arthritis, Experimental , CD8-Positive T-Lymphocytes , Interleukin-17 , Synovial Membrane , Th17 Cells , Animals , Mice , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD8-Positive T-Lymphocytes/immunology , Interleukin-17/metabolism , Synovial Membrane/immunology , Th17 Cells/immunology
14.
J Cell Physiol ; 237(9): 3541-3553, 2022 09.
Article in English | MEDLINE | ID: mdl-35938550

ABSTRACT

Rheumatoid arthritis (RA) is considered an autoimmune chronic disorder and the most common inflammatory arthropathy. Disease progression in RA begins with asymptomatic autoimmune responses in cases with a genetic or environmental predisposition, that alters to arthralgia phase as autoantibodies reach the joints and subjects begin demonstrating nonspecific musculoskeletal presentations lacking any clinical symptoms of synovial inflammation. After that, patients' symptoms develop to undifferentiated arthritis (UA)/idiopathic arthritis (IA) whenever the subjects progress to clinical synovitis systemic comorbidities affecting the vasculature, metabolism, and bone, and eventually with augmented immune cell infiltration, IA/UA patients progress to clinically classifiable RA. RA is mainly correlated with different immune cells and each of them contributes variously to the pathogenesis of the disease. The pathogenesis of RA is altered by the contribution of both T and B cells in an autoimmune irregularity. Modulation of the immune responses occurs through regulatory and inhibitory molecules that control activation of the adaptive system as well as immune hemostasis. To confine the exorbitant T cell-associated inflammatory reactions, the immune system provides a system of inhibitory feedbacks, collectively named immune checkpoints. In this review, we aimed to discuss about inhibitory members of immune checkpoint molecules, including programmed cell death 1 (PD-1)/PD-L1, cytotoxic-T-lymphocyte-antigen-4, lymphocyte activation gene-3, T cell immunoglobulin-3, V-domain Ig suppressor of T cell activation, B- and T-lymphocyte attenuator, and T cell immunoglobulin and ITIM domain and their role in RA.


Subject(s)
Arthritis, Rheumatoid , Immune Checkpoint Proteins , Synovitis , Autoantibodies , Humans , Prospective Studies , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovitis/immunology , Synovitis/pathology
15.
Sci Immunol ; 7(68): eabf2846, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148199

ABSTRACT

Macrophages regulate protective immune responses to infectious microbes, but aberrant macrophage activation frequently drives pathological inflammation. To identify regulators of vigorous macrophage activation, we analyzed RNA-seq data from synovial macrophages and identified SLAMF7 as a receptor associated with a superactivated macrophage state in rheumatoid arthritis. We implicated IFN-γ as a key regulator of SLAMF7 expression and engaging SLAMF7 drove a strong wave of inflammatory cytokine expression. Induction of TNF-α after SLAMF7 engagement amplified inflammation through an autocrine signaling loop. We observed SLAMF7-induced gene programs not only in macrophages from rheumatoid arthritis patients but also in gut macrophages from patients with active Crohn's disease and in lung macrophages from patients with severe COVID-19. This suggests a central role for SLAMF7 in macrophage superactivation with broad implications in human disease pathology.


Subject(s)
Inflammation/immunology , Macrophage Activation/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , Transcriptome/immunology , Acute Disease , Adult , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , COVID-19/genetics , COVID-19/immunology , COVID-19/metabolism , COVID-19/virology , Cells, Cultured , Chronic Disease , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/metabolism , Female , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophage Activation/genetics , RNA-Seq/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/metabolism , Single-Cell Analysis/methods , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Transcriptome/genetics
16.
Ann Rheum Dis ; 81(2): 193-205, 2022 02.
Article in English | MEDLINE | ID: mdl-34598926

ABSTRACT

OBJECTIVES: This study investigates pathogenic and protective polyfunctional T-cell responses in patient with rheumatoid arthritis (RA), individuals at risk (IAR) and healthy control (HC) synovial-tissue biopsies and identifies the presence of a novel population of pathogenic polyfunctional T-cells that are enriched in the RA joint prior to the development of clinical inflammation. METHODS: Pathway enrichment analysis of previously obtained RNAseq data of synovial biopsies from RA (n=118), IAR (n=20) and HC (n=44) was performed. Single-cell synovial tissue suspensions from RA (n=10), IAR (n=7) and HC (n=7) and paired peripheral blood mononuclear cells (PBMC) were stimulated in vitro and polyfunctional synovial T-cell subsets examined by flow cytometric analysis, simplified presentation of incredibly complex evaluations (SPICE) and FlowSom clustering. Flow-imaging was utilised to confirm specific T-cell cluster identification. Fluorescent lifetime imaging microscopy (FLIM) was used to visualise metabolic status of sorted T-cell populations. RESULTS: Increased plasticity of Tfh cells and CD4 T-cell polyfunctionality with enriched memory Treg cell responses was demonstrated in RA patient synovial tissue. Synovial-tissue RNAseq analysis reveals that enrichment in T-cell activation and differentiation pathways pre-dates the onset of RA. Switch from potentially protective IL-4 and granulocyte macrophage colony stimulating factor (GMCSF) dominated polyfunctional CD4 T-cell responses towards pathogenic polyfunctionality is evident in patient with IAR and RA synovial tissue. Cluster analysis reveals the accumulation of highly polyfunctional CD4+ CD8dim T-cells in IAR and RA but not HC synovial tissue. CD4+ CD8dim T-cells show increased utilisation of oxidative phosphorylation, a characteristic of metabolically primed memory T-cells. Frequency of synovial CD4+ CD8dim T-cells correlates with RA disease activity. CONCLUSION: Switch from potentially protective to pathogenic T-cell polyfunctionality pre-dates the onset of clinical inflammation and constitutes an opportunity for therapeutic intervention in RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Synovial Membrane/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Prodromal Symptoms
17.
Front Immunol ; 12: 778480, 2021.
Article in English | MEDLINE | ID: mdl-34887865

ABSTRACT

Objectives: This study aims to investigate if addition of fibroblast-stromal cell markers to a classification of synovial pathotypes improves their predictive value on clinical outcomes in rheumatoid arthritis (RA). Methods: Active RA patients with a knee needle synovial biopsy at baseline and finished 1-year follow-up were recruited from a real-world prospective cohort. Positive staining for CD20, CD38, CD3, CD68, CD31, and CD90 were scored semiquantitatively (0-4). The primary outcome was radiographic progression defined as a minimum increase of 0.5 units of the modified total Sharp score from baseline to 1 year. Results: Among 150 recruited RA patients, 123 (82%) had qualified synovial tissue. Higher scores of CD20+ B cells, sublining CD68+ macrophages, CD31+ endothelial cells, and CD90+ fibroblasts were associated with less decrease in disease activity and greater increase in radiographic progression. A new fibroblast-based classification of synovial pathotypes giving more priority to myeloid and stromal cells classified samples as myeloid-stromal (57.7%, 71/123), lymphoid (31.7%, 39/123), and paucicellular pathotypes (10.6%, 13/123). RA patients with myeloid-stromal pathotype showed the highest rate of radiographic progression (43.7% vs. 23.1% vs. 7.7%, p = 0.011), together with the lowest rate of Boolean remission at 3, 6, and 12 months. Baseline synovial myeloid-stromal pathotype independently predicted radiographic progression at 1 year (adjusted OR: 3.199, 95% confidence interval (95% CI): 1.278, 8.010). Similar results were obtained in a subgroup analysis of treatment-naive RA. Conclusions: This novel fibroblast-based myeloid-stromal pathotype could predict radiographic progression at 1 year in active RA patients which may contribute to the shift of therapeutic decision in RA.


Subject(s)
Antigens, CD/analysis , Arthritis, Rheumatoid/immunology , Fibroblasts/immunology , Immunohistochemistry , Knee Joint/immunology , Stromal Cells/immunology , Synovial Membrane/immunology , Adult , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Biomarkers/analysis , Biopsy, Needle , Disease Progression , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Knee Joint/diagnostic imaging , Knee Joint/drug effects , Knee Joint/pathology , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Remission Induction , Stromal Cells/drug effects , Stromal Cells/pathology , Synovial Membrane/diagnostic imaging , Synovial Membrane/drug effects , Synovial Membrane/pathology , Time Factors , Treatment Outcome
18.
Front Immunol ; 12: 790122, 2021.
Article in English | MEDLINE | ID: mdl-34899757

ABSTRACT

Rheumatoid arthritis is an autoimmune disease that exhibits significant clinical heterogeneity. There are various treatments for rheumatoid arthritis, including disease-modifying anti-rheumatic drugs (DMARDs), glucocorticoids, non-steroidal anti-inflammatory drugs (NSAIDs), and inflammatory cytokine inhibitors (ICI), typically associated with differentiated clinical effects and characteristics. Personalized responsiveness is observed to the standard treatment due to the pathophysiological heterogeneity in rheumatoid arthritis, resulting in an overall poor prognosis. Understanding the role of individual variation in cellular and molecular mechanisms related to rheumatoid arthritis will considerably improve clinical care and patient outcomes. In this review, we discuss the source of pathophysiological heterogeneity derived from genetic, molecular, and cellular heterogeneity and their possible impact on precision medicine and personalized treatment of rheumatoid arthritis. We provide emphasized description of the heterogeneity derived from mast cells, monocyte cell, macrophage fibroblast-like synoviocytes and, interactions within immune cells and with inflammatory cytokines, as well as the potential as a new therapeutic target to develop a novel treatment approach. Finally, we summarize the latest clinical trials of treatment options for rheumatoid arthritis and provide a suggestive framework for implementing preclinical and clinical experimental results into clinical practice.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Genetic Heterogeneity , Immune System/immunology , Synovial Membrane/immunology , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Cytokines/genetics , Cytokines/metabolism , Genetic Predisposition to Disease , Humans , Immune System/drug effects , Immune System/metabolism , Inflammation Mediators/metabolism , Phenotype , Signal Transduction , Synovial Membrane/drug effects , Synovial Membrane/metabolism
19.
Front Immunol ; 12: 745523, 2021.
Article in English | MEDLINE | ID: mdl-34745117

ABSTRACT

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.


Subject(s)
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
20.
Nat Immunol ; 22(12): 1551-1562, 2021 12.
Article in English | MEDLINE | ID: mdl-34811544

ABSTRACT

Misdirected immunity gives rise to the autoimmune tissue inflammation of rheumatoid arthritis, in which excess production of the cytokine tumor necrosis factor (TNF) is a central pathogenic event. Mechanisms underlying the breakdown of self-tolerance are unclear, but T cells in the arthritic joint have a distinctive metabolic signature of ATPlo acetyl-CoAhi proinflammatory effector cells. Here we show that a deficiency in the production of mitochondrial aspartate is an important abnormality in these autoimmune T cells. Shortage of mitochondrial aspartate disrupted the regeneration of the metabolic cofactor nicotinamide adenine dinucleotide, causing ADP deribosylation of the endoplasmic reticulum (ER) sensor GRP78/BiP. As a result, ribosome-rich ER membranes expanded, promoting co-translational translocation and enhanced biogenesis of transmembrane TNF. ERrich T cells were the predominant TNF producers in the arthritic joint. Transfer of intact mitochondria into T cells, as well as supplementation of exogenous aspartate, rescued the mitochondria-instructed expansion of ER membranes and suppressed TNF release and rheumatoid tissue inflammation.


Subject(s)
Arthritis, Rheumatoid/metabolism , Aspartic Acid/metabolism , CD4-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , ADP-Ribosylation , Adoptive Transfer , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD4-Positive T-Lymphocytes/ultrastructure , Case-Control Studies , Cells, Cultured , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Chaperone BiP/metabolism , Female , Humans , Male , Mice , Mitochondria/immunology , Mitochondria/transplantation , Mitochondria/ultrastructure , Synovial Membrane/immunology , Synovial Membrane/ultrastructure , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...