Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.076
Filter
1.
Int J Rheum Dis ; 27(8): e15282, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39091178

ABSTRACT

OBJECTIVE: To investigate the impact of IGJ on the proliferation, inflammation, and motility of rheumatoid arthritis (RA) fibroblast-like synoviocytes and elucidate the underlying mechanism. METHODS: The expression of IGJ RA fibroblast-like synoviocytes was assessed using immunoblot and qPCR. Cell growth was evaluated using CCK-8 and FCM assays. The effects on inflammatory response were determined by ELISA and immunoblot assays. Cell motility was assessed using transwell and immunoblot assays. The mechanism was further confirmed using immunoblot assays. RESULTS: IGJ expression was found to be elevated in fibroid synovial cells of RA. IGJ ablation inhibited the growth of MH7A cells and suppressed the inflammatory response. Knockdown of IGJ also blocked cell motility. Mechanically, the knockdown of IGJ suppressed the NF-κB axis in MH7A cells. CONCLUSION: IGJ suppresses RA in fibroblast-like synoviocytes via NF-κB pathway.


Subject(s)
Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Fibroblasts , NF-kappa B , Signal Transduction , Synoviocytes , Humans , Synoviocytes/metabolism , Synoviocytes/pathology , Synoviocytes/drug effects , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , NF-kappa B/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Cells, Cultured , Cell Line , Hyaluronoglucosaminidase
2.
Drug Des Devel Ther ; 18: 3523-3545, 2024.
Article in English | MEDLINE | ID: mdl-39135759

ABSTRACT

Purpose: Sinomenine (SIN) is commonly used in Traditional Chinese Medicine (TCM) as a respected remedy for rheumatoid arthritis (RA). Nevertheless, the therapeutic mechanism of SIN in RA remains incompletely understood. This study aimed to delve into the molecular mechanism of SIN in the treatment of RA. Methods: The potential targets of SIN were predicted using the TCMSP server, STITCH database, and SwissTarget Prediction. Differentially expressed genes (DEGs) in RA were obtained from the GEO database. Enrichment analyses and molecular docking were conducted to explore the potential mechanism of SIN in the treatment of RA. In vitro and in vivo studies were conducted to validate the intervention effects of SIN on rheumatoid arthritis, as determined through network pharmacology analyses. Results: A total of 39 potential targets associated with the therapeutic effects of SIN in RA were identified. Enrichment analysis revealed that these potential targets are primarily enriched in PI3K-Akt signaling pathway, and the molecular docking suggests that SIN may act on specific proteins in the pathway. Experimental results have shown that exposure to SIN inhibits cytokine secretion, promotes apoptosis, reduces metastasis and invasion, and blocks the activation of the PI3K-Akt signaling pathway in RA fibroblast-like synoviocytes (RA-FLS). Moreover, SIN treatment alleviated arthritis-related symptoms and regulated the differentiation of CD4+ T cells in the spleen of collagen-induced arthritis (CIA) mice. Conclusion: By utilizing network pharmacology, molecular modeling, and in vitro/in vivo validation, this study demonstrates that SIN can alleviate RA by inhibiting the PI3K-Akt signaling pathway. These findings enhance the understanding of the therapeutic mechanisms of SIN in RA, offering a stronger theoretical foundation for its future clinical application.


Subject(s)
Arthritis, Rheumatoid , Molecular Docking Simulation , Morphinans , Network Pharmacology , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Morphinans/pharmacology , Morphinans/chemistry , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Mice , Animals , Signal Transduction/drug effects , Humans , Phosphatidylinositol 3-Kinases/metabolism , Male , Antirheumatic Agents/pharmacology , Antirheumatic Agents/chemistry , Cells, Cultured , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Mice, Inbred DBA
3.
Pathol Res Pract ; 261: 155508, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39116571

ABSTRACT

Rheumatoid arthritis (RA) is a common autoimmune condition and chronic inflammatory disease, mostly affecting synovial joints. The complex pathogenesis of RA is supportive of high morbidity, disability, and mortality rates. Pathological changes a common characteristic in RA synovial tissue is attributed to the inadequacy of apoptotic pathways. In that regard, apoptotic pathways have been the center of attention in RA therapeutic approaches. As the regulators in the complex network of apoptosis, microRNAs (miRNAs) are found to be vital modulators in both intrinsic and extrinsic pathways through altering their regulatory genes. Indeed, miRNA, a member of the family of non-coding RNAs, are found to be an important player in not even apoptosis, but proliferation, gene expression, signaling pathways, and angiogenesis. Aberrant expression of miRNAs is implicated in attenuation and/or intensification of various apoptosis routes, resulting in culmination of human diseases including RA. Considering the need for more studies focused on the underlying mechanisms of RA in order to elevate the unsatisfactory clinical treatments, this study is aimed to delineate the importance of apoptosis in the pathophysiology of this disease. As well, this review is focused on the critical role of miRNAs in inducing or inhibiting apoptosis of RA-synovial fibroblasts and fibroblast-like synoviocytes and how this mechanism can be exerted for therapeutic purposes for RA.


Subject(s)
Apoptosis , Arthritis, Rheumatoid , MicroRNAs , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Synovial Membrane/pathology , Synovial Membrane/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , Signal Transduction , Fibroblasts/metabolism , Fibroblasts/pathology , Animals
4.
Sci Rep ; 14(1): 18939, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39147768

ABSTRACT

Rheumatoid arthritis (RA) and arthrofibrosis (AF) are both chronic synovial hyperplasia diseases that result in joint stiffness and contractures. They shared similar symptoms and many common features in pathogenesis. Our study aims to perform a comprehensive analysis between RA and AF and identify novel drugs for clinical use. Based on the text mining approaches, we performed a correlation analysis of 12 common joint diseases including arthrofibrosis, gouty arthritis, infectious arthritis, juvenile idiopathic arthritis, osteoarthritis, post infectious arthropathies, post traumatic osteoarthritis, psoriatic arthritis, reactive arthritis, rheumatoid arthritis, septic arthritis, and transient arthritis. 5 bulk sequencing datasets and 4 single-cell sequencing datasets of RA and AF were integrated and analyzed. A novel drug repositioning method was found for drug screening, and text mining approaches were used to verify the identified drugs. RA and AF performed the highest gene similarity (0.77) and functional ontology similarity (0.84) among all 12 joint diseases. We figured out that they share the same key pathogenic cell including CD34 + sublining fibroblasts (CD34-SLF) and DKK3 + sublining fibroblasts (DKK3-SLF). Potential therapeutic target database (PTTD) was established with the differential expressed genes (DEGs) of these key pathogenic cells. Based on the PTTD, 15 potential drugs for AF and 16 potential drugs for RA were identified. This work provides a new perspective on AF and RA study which enhances our understanding of their pathogenesis. It also shed light on their underlying mechanism and open new avenues for drug repositioning studies.


Subject(s)
Arthritis, Rheumatoid , Fibrosis , Synovial Membrane , Humans , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Synovial Membrane/pathology , Synovial Membrane/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Drug Repositioning , Cellular Microenvironment/drug effects , Data Mining
5.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125782

ABSTRACT

Ferroptosis is a form of iron-dependent regulated cell death caused by the accumulation of lipid peroxides. In this review, we summarize research on the impact of ferroptosis on disease models and isolated cells in various types of arthritis. While most studies have focused on rheumatoid arthritis (RA) and osteoarthritis (OA), there is limited research on spondylarthritis and crystal arthropathies. The effects of inducing or inhibiting ferroptosis on the disease strongly depend on the studied cell type. In the search for new therapeutic targets, inhibiting ferroptosis in chondrocytes might have promising effects for any type of arthritis. On the other hand, ferroptosis induction may also lead to a desired decrease of synovial fibroblasts in RA. Thus, ferroptosis research must consider the cell-type-specific effects on arthritis. Further investigation is needed to clarify these complexities.


Subject(s)
Ferroptosis , Osteoarthritis , Humans , Animals , Osteoarthritis/metabolism , Osteoarthritis/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Arthritis/metabolism , Arthritis/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Iron/metabolism
6.
Pharmacol Res ; 207: 107346, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127263

ABSTRACT

Synovitis is characterized by a distinctmetabolic profile featuring the accumulation of lactate, a byproduct of cellular metabolism within inflamed joints. This study reveals that the activation of the CD31 signal by lactate instigates a metabolic shift, specifically initiating endothelial cell autophagy. This adaptive process plays a pivotal role in fulfilling the augmented energy and biomolecule demands associated with the formation of new blood vessels in the synovium of Rheumatoid Arthritis (RA). Additionally, the amino acid substitutions in the CD31 cytoplasmic tail at the Y663F and Y686F sites of the immunoreceptor tyrosine-based inhibitory motifs (ITIM) alleviate RA. Mechanistically, this results in the downregulation of glycolysis and autophagy pathways. These findings significantly advance our understanding of potential therapeutic strategies for modulating these processes in synovitis and, potentially, other autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid , Autophagy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Animals , Humans , Glycolysis , Male , Synovial Membrane/metabolism , Lactic Acid/metabolism , Endothelial Cells/metabolism
7.
Cell Death Dis ; 15(8): 584, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122678

ABSTRACT

TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.


Subject(s)
Arthritis, Rheumatoid , Deubiquitinating Enzyme CYLD , Fibroblasts , I-kappa B Kinase , MAP Kinase Kinase Kinases , Signal Transduction , Synovial Membrane , Fibroblasts/metabolism , Fibroblasts/pathology , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzyme CYLD/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Animals , Synovial Membrane/metabolism , Synovial Membrane/pathology , Mice , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Humans , NF-kappa B/metabolism , Mice, Inbred C57BL , Mice, Knockout
8.
Cell Mol Life Sci ; 81(1): 350, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141086

ABSTRACT

Heparan sulfate (HS) proteoglycans are important regulators of cellular responses to soluble mediators such as chemokines, cytokines and growth factors. We profiled changes in expression of genes encoding HS core proteins, biosynthesis enzymes and modifiers during macrophage polarisation, and found that the most highly regulated gene was Sulf2, an extracellular HS 6-O-sulfatase that was markedly downregulated in response to pro-inflammatory stimuli. We then generated Sulf2+/- bone marrow chimeric mice and examined inflammatory responses in antigen-induced arthritis, as a model of rheumatoid arthritis. Resolution of inflammation was impaired in myeloid Sulf2+/- chimeras, with elevated joint swelling and increased abundance of pro-arthritic Th17 cells in synovial tissue. Transcriptomic and in vitro analyses indicated that Sulf2 deficiency increased type I interferon signaling in bone marrow-derived macrophages, leading to elevated expression of the Th17-inducing cytokine IL6. This establishes that dynamic remodeling of HS by Sulf2 limits type I interferon signaling in macrophages, and so protects against Th17-driven pathology.


Subject(s)
Macrophages , Mice, Inbred C57BL , Signal Transduction , Th17 Cells , Animals , Th17 Cells/immunology , Th17 Cells/metabolism , Mice , Macrophages/metabolism , Macrophages/immunology , Sulfatases/metabolism , Sulfatases/genetics , Sulfotransferases/metabolism , Sulfotransferases/genetics , Myeloid Cells/metabolism , Myeloid Cells/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Inflammation/metabolism , Inflammation/pathology , Mice, Knockout , Interleukin-6/metabolism , Interleukin-6/genetics , Heparitin Sulfate/metabolism
9.
Arthritis Res Ther ; 26(1): 150, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160592

ABSTRACT

OBJECTIVE: Rheumatoid Arthritis (RA) often exhibits suboptimal treatment response despite early diagnosis and treatment. This study aimed to analyze Early Rheumatoid Arthritis (ERA) synovial biopsies through histology and immunohistochemistry (IHC) to identify predictive factors for treatment response to Methotrexate (MTX). METHODS: 140 ERA patients from the UCLouvain Arthritis Cohort underwent synovial biopsy and were monitored after initiating Disease-Modifying Antirheumatic Drug (DMARD) therapy. Histological features [Synovial Hyperplasia, Fibrinoid Necrosis (FN), Hypervascularization and Inflammatory Infiltrate] and IHC (CD3, CD20, CD138, CD68) were each semi-quantitatively assessed on a 0-3 scale with 7 levels. RESULTS: A strong association was observed between synovial CD68 and Fibrinoid Necrosis scores [r = 0.44 (0.27 - 0.56); p < 0.0001]. CD68 correlated with C-Reactive Protein (CRP), DAS28, SDAI and CDAI. Fibrinoid Necrosis score correlated with CRP and DAS28. Patients were then categorized as CD68NecrosisHIGH (CD68 + Necrosis ≥ 3) and CD68NecrosisLOW (CD68 + Necrosis < 3). CD68NecrosisHIGH exhibited higher pre-treatment disease activity [5.48 (1.6) versus 4.8 (1.7); p = 0.03] and a greater fall in DAS28 [1.99 (2.06) versus 1.1 (2.27), p = 0.03], SDAI [21.45 (IQR 23.3) versus 11.65 (IQR 17.5); p = 0.003] and CDAI [16 [14.9] versus 10.5 (20.1), p = 0.04]. CD68NecrosisHIGH patients had a higher EULAR Moderate/Good Response rate. CD68Necrosis score was incorporated into a probability matrix model together with clinical features (SJC44 and DAS28) to predict achieving a Moderate/Good EULAR Response Criteria at 3 months with a good performance (AUC 0.724). CONCLUSION: FN and CD68 + in ERA synovial biopsies identify patients with higher disease activity and predict a better treatment response at three months. A model including synovial CD68 and fibrinoid necrosis with baseline clinical features predicts EULAR response at 3 months.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Methotrexate , Necrosis , Synovial Membrane , Humans , Methotrexate/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Female , Male , Middle Aged , Synovial Membrane/pathology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Antirheumatic Agents/therapeutic use , Aged , Cohort Studies , Adult , Treatment Outcome , Immunohistochemistry
10.
Int J Rheum Dis ; 27(8): e15287, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39175280

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is one autoimmune disease that badly influences the lives of humans. Nuclear factor interleukin 3 (NFIL3) has been elucidated to join into the progression of diversiform diseases. According to a recent report, NFIL3 expression levels are increased in the peripheral blood and synovial tissues of individuals with RA. However, the detailed regulatory impacts of NFIL3 and associated pathways in RA progression need more investigations. METHODS: The mRNA and protein expressions were tested through RT-qPCR and western blot. The cell proliferation was evaluated through CCK-8 and EdU assay. The cell apoptosis was measured through flow cytometry. The levels of TNF-α, IL-6, and IL-8 were assessed through ELISA. The cell migration and invasion were tested through Transwell assay. RESULTS: In this study, NFIL3 exhibited higher expression in RA fibroblast-like synoviocytes (interleukin-1ß [IL-1ß]-triggered MH7A cell model). In addition, knockdown of NFIL3 repressed the growth of IL-1ß-mediated MH7A cells. It was also demonstrated that suppressing NFIL3 resulted in reduced inflammatory reactions in IL-1ß-mediated MH7A cells. Suppression of NFIL3 alleviated cell migration and invasion in the RA cell model. Ultimately, it was demonstrated that NFIL3 retarded the AMPK/mTOR pathway. CONCLUSION: This study demonstrated that the inhibition of NFIL3 effectively controlled the AMPK/mTOR pathway, thereby suppressing the overactive proliferation, inflammation, and migration of fibroblast-like synoviocytes in human RA. This discovery implied that NFIL3 can be a serviceable biomarker for RA therapy.


Subject(s)
AMP-Activated Protein Kinases , Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Signal Transduction , Synoviocytes , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Humans , AMP-Activated Protein Kinases/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , TOR Serine-Threonine Kinases/metabolism , Inflammation Mediators/metabolism , Gene Knockdown Techniques , Cell Line , Apoptosis
11.
Front Immunol ; 15: 1438803, 2024.
Article in English | MEDLINE | ID: mdl-39076977

ABSTRACT

Ferroptosis is an iron-dependent mode of cell death distinct from apoptosis and necrosis. Its mechanisms mainly involve disordered iron metabolism, lipid peroxide deposition, and an imbalance of the antioxidant system. The endoplasmic reticulum is an organelle responsible for protein folding, lipid metabolism, and Ca2+ regulation in cells. It can be induced to undergo endoplasmic reticulum stress in response to inflammation, oxidative stress, and hypoxia, thereby regulating intracellular environmental homeostasis through unfolded protein responses. It has been reported that ferroptosis and endoplasmic reticulum stress (ERS) have an interaction pathway and jointly regulate cell survival and death. Both have also been reported separately in rheumatoid arthritis (RA) mechanism studies. However, studies on the correlation between ferroptosis and ERS in RA have not been reported so far. Therefore, this paper reviews the current status of studies and the potential correlation between ferroptosis and ERS in RA, aiming to provide a research reference for developing treatments for RA.


Subject(s)
Arthritis, Rheumatoid , Endoplasmic Reticulum Stress , Ferroptosis , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Animals , Iron/metabolism , Oxidative Stress , Unfolded Protein Response , Signal Transduction
12.
Open Biol ; 14(7): 240089, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38981514

ABSTRACT

Rheumatoid arthritis is a chronic inflammatory disease that shows characteristic diurnal variation in symptom severity, where joint resident fibroblast-like synoviocytes (FLS) act as important mediators of arthritis pathology. We investigate the role of FLS circadian clock function in directing rhythmic joint inflammation in a murine model of inflammatory arthritis. We demonstrate FLS time-of-day-dependent gene expression is attenuated in arthritic joints, except for a subset of disease-modifying genes. The deletion of essential clock gene Bmal1 in FLS reduced susceptibility to collagen-induced arthritis but did not impact symptomatic severity in affected mice. Notably, FLS Bmal1 deletion resulted in loss of diurnal expression of disease-modulating genes across the joint, and elevated production of MMP3, a prognostic marker of joint damage in inflammatory arthritis. This work identifies the FLS circadian clock as an influential driver of daily oscillations in joint inflammation, and a potential regulator of destructive pathology in chronic inflammatory arthritis.


Subject(s)
ARNTL Transcription Factors , Arthritis, Experimental , Circadian Rhythm , Fibroblasts , Synoviocytes , Animals , Synoviocytes/metabolism , Synoviocytes/pathology , Mice , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Circadian Clocks/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Mice, Knockout , Disease Models, Animal , Gene Expression Regulation , Male
13.
Cell Commun Signal ; 22(1): 373, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049070

ABSTRACT

BACKGROUND: Ascorbic acid can regulate the function of the immune system. This study aimed to investigate the underlying mechanisms of ascorbic acid in plasma cell differentiation and rheumatoid arthritis (RA). METHODS: Mice were intraperitoneally injected with either ascorbic acid or an equivalent volume of phosphate-buffered saline (PBS). To elucidate the effects of ascorbic acid on arthritis, we utilized a collagen induced arthritis mouse model (CIA). To investigate the effects of ascorbic acid on antibody response, mice were immunized with (4-Hydroxy-3-nitrophenylacetyl)-Ficoll (NP-Ficoll) or (4-hydroxy-3-nitrophenyl) acetyl-keyhole limpet hemocyanin (NP-KLH) to elicit a T-cell independent (TI) or T-cell dependent (TD) antibody response. To clarify the ability of ascorbic acid on plasma cell production, we tracked the B cell differentiation fate on the NP-specific B1-8hi BCR transgenic background. RESULTS: Ascorbic acid-injected mice demonstrated significantly delayed disease incidence and decreased disease severity compared to PBS-injected mice. Ascorbic acid can reduce the titers of autoantibodies in both arthritis and lupus mice models. Ascorbic acid can significantly reduce the number of plasma cells and the production of antigen-specific antibodies in TI and TD antibody response. In addition, ascorbic acid can disrupt the antibody affinity maturation. Through B1-8hi adoptive transfer experiments, it has been demonstrated that ascorbic acid restrains B cell differentiation into plasma cells in a cell-intrinsic manner. After in-depth exploration, we found that ascorbic acid can block the cell cycle of B cells and promote cell apoptosis. Mechanistically, ascorbic acid inhibited the production of autoreactive plasma cells by inhibiting the Stat3 signaling pathway. CONCLUSION: Our study demonstrates that ascorbic acid has the ability to suppress the generation of autoreactive plasma cells, diminish the production of autoantibodies, and consequently delay the onset of rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid , Ascorbic Acid , Autoantibodies , Animals , Autoantibodies/immunology , Autoantibodies/blood , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Mice , Arthritis, Experimental/immunology , Arthritis, Experimental/drug therapy , Cell Differentiation/drug effects , Plasma Cells/immunology , Plasma Cells/drug effects
15.
Biomolecules ; 14(7)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39062579

ABSTRACT

The tumor necrosis factor receptor-associated factor 1 (TRAF1) plays a key role in promoting lymphocyte survival, proliferation, and cytokine production. Recent evidence showed that TRAF1 plays opposing roles in monocytes and macrophages where it controls NF-κB activation and limits pro-inflammatory cytokine production as well as inflammasome-dependent IL-1ß secretion. Importantly, TRAF1 polymorphisms have been strongly linked to an increased risk of rheumatoid arthritis (RA). However, whether and how TRAF1 contributes to RA pathogenesis is not fully understood. Moreover, investigating the role of TRAF1 in driving RA pathogenesis is complicated by its multifaceted and opposing roles in various immune cells. In this study, we subjected wildtype (WT) mice to the collagen antibody-induced arthritis (CAIA) model of RA and injected them intra-articularly with WT- or TRAF1-deficient macrophages. We show that mice injected with TRAF1-deficient macrophages exhibited significantly exacerbated joint inflammation, immune cell infiltration, and tissue damage compared to mice injected with WT macrophages. This study may lay the groundwork for novel therapies for RA that target TRAF1 in macrophages.


Subject(s)
Arthritis, Rheumatoid , Macrophages , TNF Receptor-Associated Factor 1 , Animals , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism , TNF Receptor-Associated Factor 1/deficiency , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Macrophages/metabolism , Mice , Arthritis, Experimental/pathology , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Mice, Inbred C57BL , Mice, Knockout
16.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063107

ABSTRACT

It is not entirely clear how the interaction between joint inflammation and the central nervous system (CNS) response in rheumatoid arthritis (RA) works, and what pathophysiology underlies the sex differences in coexisting neuropsychiatric comorbidities. It is known that estrogen hormones reduce inflammation in RA and that this occurs mainly via the stimulation of G protein-coupled receptor-30 (GPR30), also known as G protein-coupled estrogen receptor (GPER) 1. However, changes in GPR30 expression and sex differences induced by local and systemic inflammation in RA are not yet known. Our aim was to reveal sex differences in the expression and association of joint GPR30 with local and systemic inflammation, clinical course and furthermore with hippocampal GPR30 expression during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Furthermore, we demonstrated sex-specific differences in the association between joint and systemic inflammation and hippocampal microglia during PIA. Our results suggest sex-specific differences not only in the clinical course and serum levels of pro-inflammatory cytokines but also in the expression of GPR30. Female rats show greater synovial inflammation and greater damage to the articular cartilage compared to males during PIA attack. Male rats express higher levels of synovial and cartilaginous GPR30 than females during PIA, which correlates with a less severe clinical course. The correlation between synovial and cartilaginous GPR30 and joint inflammation scores (Krenn and Mankin) in male rats suggests that the more severe the joint inflammation, the higher the GPR30 expression. At the same time, there is no particular upregulation of hippocampal GPR30 in males. On the other hand, female rats express higher levels of neuroprotective GPR30 in the hippocampus than male rats at the basic level and during PIA attack. In addition, females have a higher number of Iba-1+ cells in the hippocampus during PIA attack that strongly correlates with the clinical score, serum levels of IL-17A, and Krenn and Mankin scores. These results suggest that male rats are better protected from inflammation in the joints and female rats are better protected from the inflammation in the hippocampus during a PIA attack, independently of microglia proliferation. However, in the remission phase, synovial GPR30 expression suddenly increases in female rats, as does hippocampal GPR30 expression in males. Further experiments with a longer remission period are needed to investigate the molecular background of these sex differences, as well as microglia phenotype profiling.


Subject(s)
Arthritis, Rheumatoid , Disease Models, Animal , Hippocampus , Receptors, G-Protein-Coupled , Animals , Female , Male , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , Hippocampus/metabolism , Rats , Inflammation/metabolism , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Sex Characteristics , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Microglia/metabolism , Sex Factors , Terpenes
17.
PLoS One ; 19(7): e0302119, 2024.
Article in English | MEDLINE | ID: mdl-39083495

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation and swelling. Several studies have demonstrated that RA fibroblast-like synovial cells (RA-FLS) play an important role in RA pathogenesis. Activated RA-FLS contribute to synovial inflammation by secreting inflammatory cytokines including interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α. LMT-28 is derivative of oxazolidone and exerts anti-inflammatory effects on RA via IL-6 signaling pathway regulation. LMT-28 also regulates T cell differentiation in RA condition. However, the effect of LMT-28 on the migration and invasion of RA-FLS remains unknown. Kaempferol has been reported to have pharmacological effects on various diseases, such as inflammatory diseases, autoimmune diseases, and cancer. Additionally, kaempferol has been reported to inhibit RA-FLS migration and invasion, but it is not known about the therapeutic mechanism including molecular mechanism such as receptor. The present study aimed to investigate the synergistic effects of the combined treatment of LMT-28 and kaempferol on RA-FLS activation and RA pathogenesis in mouse model. LMT-28 and kaempferol co-administration inhibited RA disease severity and histological collapse in the joint tissues of CIA mice, as well as downregulated the levels of pro-inflammatory cytokines in mouse serum. Additionally, the combined treatment inhibited excessive differentiation of T helper 17 cells and osteoclasts. Furthermore, compared with single treatments, combined treatment showed enhanced inhibitory effects on the hyperactivation of IL-6-induced signaling pathway in RA-FLS. Combined treatment also inhibited RA-FLS cell proliferation, migration, and invasion and suppressed the expression of matrix metalloproteinase in RA-FLS. Furthermore, we confirmed that the combined treatment inhibited chondrocyte proliferation, migration, and invasion. In conclusion, our results suggest that the combined treatment of LMT-28 and kaempferol exerts a synergistic effect on the RA development via the regulation of IL-6-induced hyperactivation of RA-FLS. Furthermore, this study suggests that combination therapies can be an effective therapeutic option for arthritis.


Subject(s)
Anti-Inflammatory Agents , Arthritis, Experimental , Kaempferols , Animals , Kaempferols/pharmacology , Kaempferols/therapeutic use , Kaempferols/administration & dosage , Mice , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/metabolism , Mice, Inbred DBA , Disease Models, Animal , Male , Cell Movement/drug effects , Interleukin-6/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Cell Differentiation/drug effects , Drug Therapy, Combination , Synoviocytes/drug effects , Synoviocytes/metabolism
18.
Biomaterials ; 311: 122706, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39032219

ABSTRACT

Medicine intervention is the major clinical treatment used to relieve the symptoms and delay the progression of rheumatoid arthritis (RA), but is limited by its poor targeted delivery and short therapeutic duration. Herein, we developed an injectable and bioadhesive gelatin-based (Gel) hydrogel as a local depot of leonurine (Leon)-loaded and folate-functionalized polydopamine (FA-PDA@Leon) nanoparticles for anti-inflammation and chondroprotection in RA. The nanoparticles could protect Leon and facilitate its entry into the M1 phenotype macrophage for intracellular delivery of Leon, while the hydrogel tightly adhered to the tissues in the joint cavity and prolonged the retention of FA-PDA@Leon nanoparticles, thus achieving higher availability and therapeutic efficiency of Leon. In vitro and in vivo experiments demonstrated that the Gel/FA-PDA@Leon hydrogel could strongly suppress the inflammatory response by down-regulating the JAK2/STAT3 signaling pathway in macrophages and protect the chondrocytes from ferritinophagy/ferroptosis. This contributed to maintaining the structural integrity of articular cartilage and accelerating the joint functional recovery. This work provides an effective and convenient strategy to achieve higher bioavailability and long-lasting therapeutic duration of medicine intervention in arthritis diseases.


Subject(s)
Arthritis, Rheumatoid , Ferroptosis , Hydrogels , Inflammation , Nanoparticles , Polymers , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Hydrogels/chemistry , Mice , Nanoparticles/chemistry , Inflammation/drug therapy , Inflammation/pathology , Ferroptosis/drug effects , Polymers/chemistry , RAW 264.7 Cells , Nanomedicine/methods , Indoles/chemistry , Indoles/pharmacology , Indoles/administration & dosage , Male , Macrophages/drug effects , Macrophages/metabolism , Folic Acid/chemistry , Gelatin/chemistry , Humans , Chondrocytes/drug effects , Chondrocytes/metabolism
19.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000375

ABSTRACT

Angiogenesis is critical for rheumatoid arthritis (RA) progression. The effects of tofacitinib, a JAK-STAT inhibitor used for RA treatment, on angiogenesis in RA are unclear. We, therefore, evaluated the levels of angiogenic factors in two systems of a human co-culture of fibroblast (HT1080) and monocytic (U937) cell lines treated with tofacitinib and in serum samples from RA patients before and after six months of tofacitinib treatment. Tofacitinib reduced CD147 levels, matrix metalloproteinase-9 (MMP-9) activity, and angiogenic potential but increased endostatin levels and secreted proteasome 20S activity. In vitro, tofacitinib did not change CD147 mRNA but increased miR-146a-5p expression and reduced STAT3 phosphorylation. We recently showed that CD147 regulates the ability of MMP-9 and secreted proteasome 20S to cleave collagen XVIIIA into endostatin. We show here that tofacitinib-enhanced endostatin levels are mediated by CD147, as CD147-siRNA or an anti-CD147 antibody blocked proteasome 20S activity. The correlation between CD147 and different disease severity scores supported this role. Lastly, tofacitinib reduced endostatin' s degradation by inhibiting cathepsin S activity and recombinant cathepsin S reversed this in both systems. Thus, tofacitinib inhibits angiogenesis by reducing pro-angiogenic factors and enhancing the anti-angiogenic factor endostatin in a dual effect mediated partly through CD147 and partly through cathepsin S.


Subject(s)
Arthritis, Rheumatoid , Basigin , Cathepsins , Endostatins , Piperidines , Pyrimidines , Humans , Basigin/metabolism , Basigin/genetics , Piperidines/pharmacology , Endostatins/metabolism , Endostatins/pharmacology , Pyrimidines/pharmacology , Cathepsins/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , STAT3 Transcription Factor/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Female , Middle Aged , Male , Pyrroles/pharmacology , Cell Line
20.
Arthritis Res Ther ; 26(1): 135, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026358

ABSTRACT

With great interest, we have read the recent article "Expression of HIF1α in intestinal epithelium restricts arthritis inflammation by inhibiting RIPK3-induced cell death machinery" published by Lyu et al. in Annals of the Rheumatic Diseases. The authors pose that the expression of hypoxia-inducible factor 1 alpha in intestinal epithelial cells represents a crucial check point for the development of arthritis by impeding necroptosis of intestinal epithelial cells and safeguarding the intestinal barrier integrity. Previous studies suggest a potential mechanistic link between faulty intestinal barrier function and potentiation of arthritogenic immune cells. From this perspective, bolstering the intestinal barrier integrity arose as an attractive therapeutic strategy for rheumatoid arthritis.


Subject(s)
Intestinal Mucosa , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL