Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.094
Filtrer
1.
Autoimmunity ; 57(1): 2387076, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39229919

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Prolifération cellulaire , Inflammation , ARN long non codant , Cellules synoviales , Voie de signalisation Wnt , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Inflammation/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/immunologie , Apoptose , bêta-Caténine/métabolisme , Cellules cultivées
2.
Cell Commun Signal ; 22(1): 440, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261837

RÉSUMÉ

BACKGROUND: Bivalent regions of chromatin (BvCR) are characterized by trimethylated lysine 4 (H3K4me3) and lysine 27 on histone H3 (H3K27me3) deposition which aid gene expression control during cell differentiation. The role of BvCR in post-transcriptional DNA damage response remains unidentified. Oncoprotein survivin binds chromatin and mediates IFNγ effects in CD4+ cells. In this study, we explored the role of BvCR in DNA damage response of autoimmune CD4+ cells in rheumatoid arthritis (RA). METHODS: We performed deep sequencing of the chromatin bound to survivin, H3K4me3, H3K27me3, and H3K27ac, in human CD4+ cells and identified BvCR, which possessed all three histone H3 modifications. Protein partners of survivin on chromatin were predicted by integration of motif enrichment analysis, computational machine-learning, and structural modeling, and validated experimentally by mass spectrometry and peptide binding array. Survivin-dependent change in BvCR and transcription of genes controlled by the BvCR was studied in CD4+ cells treated with survivin inhibitor, which revealed survivin-dependent biological processes. Finally, the survivin-dependent processes were mapped to the transcriptome of CD4+ cells in blood and in synovial tissue of RA patients and the effect of modern immunomodulating drugs on these processes was explored. RESULTS: We identified that BvCR dominated by H3K4me3 (H3K4me3-BvCR) accommodated survivin within cis-regulatory elements of the genes controlling DNA damage. Inhibition of survivin or JAK-STAT signaling enhanced H3K4me3-BvCR dominance, which improved DNA damage recognition and arrested cell cycle progression in cultured CD4+ cells. Specifically, BvCR accommodating survivin aided sequence-specific anchoring of the BRG1/SWI chromatin-remodeling complex coordinating DNA damage response. Mapping survivin interactome to BRG1/SWI complex demonstrated interaction of survivin with the subunits anchoring the complex to chromatin. Co-expression of BRG1, survivin and IFNγ in CD4+ cells rendered complete deregulation of DNA damage response in RA. Such cells possessed strong ability of homing to RA joints. Immunomodulating drugs inhibited the anchoring subunits of BRG1/SWI complex, which affected arthritogenic profile of CD4+ cells. CONCLUSIONS: BvCR execute DNA damage control to maintain genome fidelity in IFN-activated CD4+ cells. Survivin anchors the BRG1/SWI complex to BvCR to repress DNA damage response. These results offer a platform for therapeutic interventions targeting survivin and BRG1/SWI complex in autoimmunity.


Sujet(s)
Lymphocytes T CD4+ , Chromatine , Altération de l'ADN , Helicase , Protéines nucléaires , Survivine , Facteurs de transcription , Humains , Survivine/métabolisme , Survivine/génétique , Lymphocytes T CD4+/métabolisme , Chromatine/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Helicase/métabolisme , Helicase/génétique , Histone/métabolisme , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique
3.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39273304

RÉSUMÉ

The management of rheumatic diseases has noticeably changed in recent years with the development of targeted therapeutic agents, namely, biological disease-modifying antirheumatic drugs. Identifying essential signaling pathways and factors crucial for the development and progression of these diseases remains a significant challenge. Therapy could be used to delay the onset or reduce harm. The endocannabinoid system's presence within the synovium can be identified as a suggested target for therapeutic interventions due to its role in modulating pain, inflammation, and joint metabolism. This review brings together the most pertinent information concerning the actions of the endocannabinoid system present in inflamed synovial tissue and its interaction with phytocannabinoids and synthetic cannabinoids, which can be used from a therapeutic perspective to minimize the inflammatory and pain processes typical of osteoarthritis and rheumatoid arthritis.


Sujet(s)
Cannabinoïdes , Membrane synoviale , Humains , Cannabinoïdes/usage thérapeutique , Cannabinoïdes/pharmacologie , Cannabinoïdes/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/effets des médicaments et des substances chimiques , Animaux , Endocannabinoïdes/métabolisme , Rhumatismes/traitement médicamenteux , Rhumatismes/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie
4.
Elife ; 132024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39235454

RÉSUMÉ

miRNAs constitute fine-tuners of gene expression and are implicated in a variety of diseases spanning from inflammation to cancer. miRNA expression is deregulated in rheumatoid arthritis (RA); however, their specific role in key arthritogenic cells such as the synovial fibroblast (SF) remains elusive. Previous studies have shown that Mir221/222 expression is upregulated in RA SFs. Here, we demonstrate that TNF and IL-1ß but not IFN-γ activated Mir221/222 gene expression in murine SFs. SF-specific overexpression of Mir221/222 in huTNFtg mice led to further expansion of SFs and disease exacerbation, while its total ablation led to reduced SF expansion and attenuated disease. Mir221/222 overexpression altered the SF transcriptional profile igniting pathways involved in cell cycle and ECM (extracellular matrix) regulation. Validation of targets of Mir221/222 revealed cell cycle inhibitors Cdkn1b and Cdkn1c, as well as the epigenetic regulator Smarca1. Single-cell ATAC-seq data analysis revealed increased Mir221/222 gene activity in pathogenic SF subclusters and transcriptional regulation by Rela, Relb, Junb, Bach1, and Nfe2l2. Our results establish an SF-specific pathogenic role of Mir221/222 in arthritis and suggest that its therapeutic targeting in specific subpopulations could lead to novel fibroblast-targeted therapies.


Sujet(s)
microARN , Animaux , Souris , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Assemblage et désassemblage de la chromatine , Fibroblastes/métabolisme , Régulation de l'expression des gènes , microARN/métabolisme , microARN/génétique , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie
5.
J Pineal Res ; 76(6): e13009, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39315577

RÉSUMÉ

Melatonin has been reported to regulate circadian rhythms and have anti-inflammatory characteristics in various inflammatory autoimmune diseases, but its effects in diseases-associated muscle atrophy remain controversial. This study is aimed to determine the evidence of melatonin in rheumatoid arthritis (RA)-related pathological muscle atrophy. We used initially bioinformatics results to show that melatonin regulated significantly the correlation between pro-inflammation and myogenesis in RA synovial fibroblasts (RASF) and myoblasts. The conditioned medium (CM) from melatonin-treated RASF was incubated in myoblasts with growth medium and differentiated medium to investigate the markers of pro-inflammation, atrophy, and myogenesis. We found that melatonin regulated RASF CM-induced pathological muscle pro-inflammation and atrophy in myoblasts and differentiated myocytes through NF-κB signaling pathways. We also showed for the first time that miR-30c-1-3p is negatively regulated by three inflammatory cytokines in human RASF, which is associated with murine-differentiated myocytes. Importantly, oral administration with melatonin in a collagen-induced arthritis (CIA) mouse model also significantly improved arthritic swelling, hind limb grip strength as well as pathological muscle atrophy. In conclusion, our study is the first to demonstrate not only the underlying mechanism whereby melatonin decreases pro-inflammation in RA-induced pathological muscle atrophy but also increases myogenesis in myoblasts and differentiated myocytes.


Sujet(s)
Polyarthrite rhumatoïde , Fibroblastes , Mélatonine , Muscles squelettiques , Mélatonine/pharmacologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Humains , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Animaux , Souris , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/anatomopathologie , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/effets des médicaments et des substances chimiques , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/traitement médicamenteux , Mâle , Myoblastes/métabolisme , Myoblastes/effets des médicaments et des substances chimiques , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/traitement médicamenteux , Souris de lignée DBA
6.
Ann Clin Lab Sci ; 54(4): 446-451, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39293832

RÉSUMÉ

OBJECTIVE: Rheumatoid arthritis (RA) is a common chronic autoimmune inflammatory disease. The pathogenesis of RA is complex, and RA lacks effective therapeutic drugs. Heme oxygenase 1 (HO-1) is found to be reduced in RA. However, the role of HO-1 in RA and related mechanisms have not been elucidated. METHODS: RA rat model was established. The expression of HO-1 was upregulated by hemin. The increase weight rate, the degree of toe swelling, and the arthritis index were analyzed to evaluate the therapeutic effect of HO-1 on RA. In vitro RAW264.7 inflammatory cell model was established using 5 ng/mL IL-1. SnPP or hemin were used to inhibit or upregulate HO-1 expression. Tetrazolium salt colorimetric assay (MTT) was selected to test cell proliferation. ELISA was used to determine the concentrations of cellular inflammatory factors IL-1 and IL-6. Reactive oxygen species (ROS) activity was assessed. Western blot was performed to analyze NF-[Formula: see text]B and MMP-3 expressions. RESULTS: The expression of HO-1 was decreased in RA rats, and hemin increased HO-1 level in arthritic rats, which elevated the increase weight rate and decreased toe swelling degree and arthritis index (P<0.05). Hemin significantly upregulated HO-1 expression, inhibited inflammatory cell proliferation, decreased IL-1 and IL-6 expressions, declined ROS level, restrained NF-[Formula: see text]B expression, and enhanced MMP-3 expression in Raw264.7 cells induced by LPS (P<0.05). SnPP obviously inhibited the expression of HO-1, promoted cell proliferation, elevated IL-1 and IL-6 secretions, increased ROS level, promoted NF-[Formula: see text]B expression, and decreased MMP-3 level compared with LPS group (P<0.05). CONCLUSION: Upregulation of HO-1 can improve arthritis symptoms by reducing ROS expression, inhibiting NF-[Formula: see text]B signaling pathway, elevating MMP-3 expression, attenuating inflammatory factor secretion, and suppressing inflammatory cell proliferation.


Sujet(s)
Polyarthrite rhumatoïde , Heme oxygenase-1 , Hémine , Espèces réactives de l'oxygène , Animaux , Souris , Rats , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Heme oxygenase (decyclizing) , Heme oxygenase-1/métabolisme , Hémine/pharmacologie , Inflammation/anatomopathologie , Inflammation/métabolisme , Interleukine-1/métabolisme , Interleukine-6/métabolisme , Matrix metalloproteinase 3/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protoporphyrines/pharmacologie , Cellules RAW 264.7 , Espèces réactives de l'oxygène/métabolisme
7.
Front Immunol ; 15: 1433898, 2024.
Article de Anglais | MEDLINE | ID: mdl-39301019

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by synovitis, degradation of articular cartilage, and bone destruction. Fibroblast-like synoviocytes (FLS) play a central role in RA, producing a significant amount of inflammatory mediators such as tumor necrosis factor(TNF)-α and IL-6, which promote inflammatory responses within the joints. Moreover, FLS exhibit tumor-like behavior, including aggressive proliferation and enhanced anti-apoptotic capabilities, which collectively drive chronic inflammation and joint damage in RA. TNF is a major pro-inflammatory cytokine that mediates a series of signaling pathways through its receptor TNFR1, including NF-κB and MAPK pathways, which are crucial for inflammation and cell survival in RA. The abnormal proliferation and anti-apoptotic characteristics of FLS in RA may result from dysregulation in TNF-mediated cell death pathways such as apoptosis and necroptosis. Ubiquitination is a critical post-translational modification regulating these signaling pathways. E3 ubiquitin ligases, such as cIAP1/2, promote the ubiquitination and degradation of target proteins within the TNF receptor complex, modulating the signaling proteins. The high expression of the BIRC3 gene and its encoded protein, cIAP2, in RA regulates various cellular processes, including apoptosis, inflammatory signaling, immune response, MAPK signaling, and cell proliferation, thereby promoting FLS survival and inflammatory responses. Inhibiting BIRC3 expression can reduce the secretion of inflammatory cytokines by RA-FLS under both basal and inflammatory conditions and inhibit their proliferation. Although BIRC3 inhibitors show potential in RA treatment, their possible side effects must be carefully considered. Further research into the specific mechanisms of BIRC3, including its roles in cell signaling, apoptosis regulation, and immune evasion, is crucial for identifying new therapeutic targets and strategies.


Sujet(s)
Polyarthrite rhumatoïde , Protéine-3 contenant des répétitions IAP baculovirales , Prolifération cellulaire , Fibroblastes , Transduction du signal , Cellules synoviales , Facteur de nécrose tumorale alpha , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/immunologie , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Cellules synoviales/immunologie , Protéine-3 contenant des répétitions IAP baculovirales/métabolisme , Protéine-3 contenant des répétitions IAP baculovirales/génétique , Facteur de nécrose tumorale alpha/métabolisme , Fibroblastes/métabolisme , Apoptose , Ubiquitination , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
8.
Front Immunol ; 15: 1438272, 2024.
Article de Anglais | MEDLINE | ID: mdl-39221253

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. Neutrophil extracellular traps (NETs are NETwork protein structures activated by neutrophils to induce the cleavage and release of DNA-protein complexes). Current studies have shown the critical involvement of NETs in the progression of autoimmune diseases, Neutrophils mostly gather in the inflammatory sites of patients and participate in the pathogenesis of autoimmune diseases in various ways. NETs, as the activated state of neutrophils, have attracted much attention in immune diseases. Many molecules released in NETs are targeted autoantigens in autoimmune diseases, such as histones, citrulline peptides, and myeloperoxidase. All of these suggest that NETs have a direct causal relationship between the production of autoantigens and autoimmune diseases. For RA in particular, as a disorder of the innate and adaptive immune response, the pathogenesis of RA is inseparable from the generation of RA. In this article, we investigate the emerging role of NETs in the pathogenesis of RA and suggest that NETs may be an important target for the treatment of inflammatory autoimmune diseases.


Sujet(s)
Polyarthrite rhumatoïde , Évolution de la maladie , Pièges extracellulaires , Granulocytes neutrophiles , Pièges extracellulaires/immunologie , Pièges extracellulaires/métabolisme , Humains , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/étiologie , Polyarthrite rhumatoïde/anatomopathologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Animaux , Autoantigènes/immunologie
9.
Stem Cell Res Ther ; 15(1): 276, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39227964

RÉSUMÉ

Rheumatoid arthritis (RA) remains a challenging chronic autoimmune disorder characterized by persistent joint inflammation and damage. While modern regenerative strategies, encompassing cell/stem cell-based therapies, gene therapy, and tissue engineering, have advanced tissue repair efforts, a definitive cure for RA remains elusive. Consequently, there is growing interest in developing targeted therapies that directly address the underlying mechanisms driving RA pathogenesis, such as extracellular vesicles (EVs). These small membrane-bound particles can modulate immune responses within the inflammatory microenvironment of damaged cartilage. To launch the clinical potential of EVs, they can be isolated from various cell types through several techniques. EVs can carry various bioactive molecules and anti-inflammatory or pro-regenerative drugs, deliver them directly to the affected joints, and affect the behavior of injured cells, making them a compelling choice for targeted therapy and drug delivery in RA patients. However, there are still several challenges and limitations associated with EV-based therapy, including the absence of standardized protocols for EV isolation, characterization, and delivery. This review provides a comprehensive overview of the cellular sources of EVs in RA and delves into their therapeutic potential and the hurdles they must overcome.


Sujet(s)
Polyarthrite rhumatoïde , Vésicules extracellulaires , Humains , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Vésicules extracellulaires/métabolisme , Animaux
10.
Sci Adv ; 10(39): eadj1252, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39321281

RÉSUMÉ

This study performed an in-depth investigation into the myeloid cellular landscape in the synovium of patients with rheumatoid arthritis (RA), "individuals at risk" of RA, and healthy controls (HC). Flow cytometric analysis demonstrated the presence of a CD40-expressing CD206+CD163+ macrophage population dominating the inflamed RA synovium, associated with disease activity and treatment response. In-depth RNA sequencing and metabolic analysis demonstrated that this macrophage population is transcriptionally distinct, displaying unique inflammatory and tissue-resident gene signatures, has a stable bioenergetic profile, and regulates stromal cell responses. Single-cell RNA sequencing profiling of 67,908 RA and HC synovial tissue cells identified nine transcriptionally distinct macrophage clusters. IL-1B+CCL20+ and SPP1+MT2A+ are the principal macrophage clusters in RA synovium, displaying heightened CD40 gene expression, capable of shaping stromal cell responses, and are importantly enriched before disease onset. Combined, these findings identify the presence of an early pathogenic myeloid signature that shapes the RA joint microenvironment and represents a unique opportunity for early diagnosis and therapeutic intervention.


Sujet(s)
Polyarthrite rhumatoïde , Homéostasie , Macrophages , Membrane synoviale , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Humains , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Macrophages/métabolisme , Macrophages/immunologie , Femelle , Mâle , Adulte d'âge moyen , Analyse sur cellule unique , Analyse de profil d'expression de gènes
11.
Nature ; 633(8029): 442-450, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39143217

RÉSUMÉ

Regulation of neutrophil activation is critical for disease control. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA and neutrophil-derived proteins, are formed following pro-inflammatory signals; however, if this process is uncontrolled, NETs contribute to disease pathogenesis, exacerbating inflammation and host tissue damage1,2. Here we show that myeloid inhibitory C-type lectin-like (MICL), an inhibitory C-type lectin receptor, directly recognizes DNA in NETs; this interaction is vital to regulate neutrophil activation. Loss or inhibition of MICL functionality leads to uncontrolled NET formation through the ROS-PAD4 pathway and the development of an auto-inflammatory feedback loop. We show that in the context of rheumatoid arthritis, such dysregulation leads to exacerbated pathology in both mouse models and in human patients, where autoantibodies to MICL inhibit key functions of this receptor. Of note, we also detect similarly inhibitory anti-MICL autoantibodies in patients with other diseases linked to aberrant NET formation, including lupus and severe COVID-19. By contrast, dysregulation of NET release is protective during systemic infection with the fungal pathogen Aspergillus fumigatus. Together, we show that the recognition of NETs by MICL represents a fundamental autoregulatory pathway that controls neutrophil activity and NET formation.


Sujet(s)
COVID-19 , Pièges extracellulaires , Activation des neutrophiles , Granulocytes neutrophiles , Protein-arginine deiminase Type 4 , Espèces réactives de l'oxygène , Pièges extracellulaires/métabolisme , Pièges extracellulaires/immunologie , Humains , Animaux , Souris , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Protein-arginine deiminase Type 4/métabolisme , COVID-19/immunologie , COVID-19/virologie , Espèces réactives de l'oxygène/métabolisme , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Autoanticorps/immunologie , Femelle , Lectines de type C/métabolisme , Lectines de type C/immunologie , Mâle , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , ADN/métabolisme , ADN/immunologie , Aspergillus fumigatus/immunologie , Aspergillus fumigatus/pathogénicité , Rétrocontrôle physiologique , Modèles animaux de maladie humaine , Inflammation/immunologie , Inflammation/métabolisme
12.
Pathol Res Pract ; 261: 155508, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39116571

RÉSUMÉ

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.


Sujet(s)
Apoptose , Polyarthrite rhumatoïde , microARN , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Humains , microARN/génétique , microARN/métabolisme , Apoptose/génétique , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Transduction du signal , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Animaux
13.
Sci Rep ; 14(1): 18939, 2024 08 15.
Article de Anglais | MEDLINE | ID: mdl-39147768

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Fibrose , Membrane synoviale , Humains , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Repositionnement des médicaments , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Fouille de données
14.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39125782

RÉSUMÉ

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.


Sujet(s)
Ferroptose , Arthrose , Humains , Animaux , Arthrose/métabolisme , Arthrose/anatomopathologie , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Arthrite/métabolisme , Arthrite/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fer/métabolisme
15.
Drug Des Devel Ther ; 18: 3523-3545, 2024.
Article de Anglais | MEDLINE | ID: mdl-39135759

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Simulation de docking moléculaire , Morphinanes , Pharmacologie des réseaux , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Morphinanes/pharmacologie , Morphinanes/composition chimique , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Souris , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Phosphatidylinositol 3-kinases/métabolisme , Mâle , Antirhumatismaux/pharmacologie , Antirhumatismaux/composition chimique , Cellules cultivées , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Souris de lignée DBA
16.
Int Immunopharmacol ; 141: 112987, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39182267

RÉSUMÉ

Rheumatoid arthritis (RA) is an enduring autoimmune inflammatory condition distinguished by continual joint inflammation, hyperplasia of the synovium, erosion of bone, and deterioration of cartilage.Fibroblast-like synoviocytes (FLSs) exhibiting "tumor-like" traits are central to this mechanism.ADP-ribosylation factor-like 4c (ARL4C) functions as a Ras-like small GTP-binding protein, significantly impacting tumor migration, invasion, and proliferation.However, it remains uncertain if ARL4C participates in the stimulation of RA FLSs exhibiting "tumor-like" features, thereby fostering the advancement of RA. In our investigation, we unveiled, for the inaugural instance, via the amalgamated scrutiny of single-cell RNA sequencing (scRNA-seq) and Bulk RNA sequencing (Bulk-seq) datasets, that activated fibroblast-like synoviocytes (FLSs) showcase high expression of ARL4C, and the ARL4C protein expression in FLSs derived from RA patients significantly surpasses that observed in individuals with osteoarthritis (OA) and traumatic injury (trauma).Silencing of the ARL4C gene markedly impeded the proliferation of RA FLSs by hindered the transition of cells from the G0/G1 phase to the S phase, and intensified cell apoptosis and diminished the migratory and invasive capabilities. Co-culture of ARL4C gene-silenced RA FLSs with monocytes/macrophages significantly inhibited the polarization of monocytes/macrophages toward M1 and the repolarization of M2 to M1.Furthermore, intra-articular injection of shARL4C significantly alleviated synovial inflammation and cartilage erosion in collagen-induced arthritis (CIA) rats. In conclusion, our discoveries propose that ARL4C assumes a central role in the synovial inflammation, cartilage degradation, and bone erosion associated with RA by triggering the PI3K/AKT and MAPK signaling pathways within RA FLSs.ARL4C holds promise as a prospective target for the development of pharmaceutical agents targeting FLSs, with the aim of addressing RA.


Sujet(s)
Facteurs d'ADP-ribosylation , Polyarthrite rhumatoïde , Macrophages , Cellules synoviales , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Humains , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Facteurs d'ADP-ribosylation/métabolisme , Facteurs d'ADP-ribosylation/génétique , Animaux , Macrophages/immunologie , Macrophages/métabolisme , Rats , Analyse sur cellule unique , Évolution de la maladie , Prolifération cellulaire , Cellules cultivées
17.
ACS Appl Mater Interfaces ; 16(36): 47206-47215, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39190615

RÉSUMÉ

Rheumatoid arthritis (RA) is a systemic autoimmune disorder that severely compromises joint health. The primary therapeutic strategy for advanced RA aims to inhibit joint inflammation. However, the nonspecific distribution of pharmacological agents has limited therapeutic efficacy and heightens the risks associated with RA treatment. To address this issue, we developed mesenchymal stem cell (MSC)-based biomimetic liposomes, termed MSCsome, which were composed of a fusion between MSC membranes and liposomes. MSC some with relatively simple preparation method effectively enhanced the targeting efficiency of drug to diseased joints. Interaction between lymphocyte function-associated antigen-1 and intercellular adhesion molecule-1 enhanced the affinity of the MSCsome for polarized macrophages, thereby improving its targeting capability to affected joints. The effective targeted delivery facilitated drug accumulation in joints, resulting in the significant inhibition of the inflammation, as well as protection and repair of the cartilage. In conclusion, this study introduced MSCsome as a promising approach for the effective treatment of advanced RA, providing a novel perspective on targeted drug delivery therapy for inflammatory diseases.


Sujet(s)
Polyarthrite rhumatoïde , Dexaméthasone , Systèmes de délivrance de médicaments , Liposomes , Cellules souches mésenchymateuses , Liposomes/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Dexaméthasone/administration et posologie , Humains , Animaux , Souris , Cellules endothéliales de la veine ombilicale humaine , Souris de lignée BALB C , Souris de lignée DBA , Mâle , Biomimétique , Cartilage/effets des médicaments et des substances chimiques , Cartilage/anatomopathologie
18.
Pharmacol Res ; 207: 107346, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39127263

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Autophagie , Antigènes CD31 , Animaux , Humains , Mâle , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Cellules endothéliales/métabolisme , Glycolyse , Acide lactique/métabolisme , Membrane synoviale/métabolisme , Antigènes CD31/génétique , Antigènes CD31/métabolisme
19.
Int J Rheum Dis ; 27(8): e15282, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39091178

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Fibroblastes , Facteur de transcription NF-kappa B , Transduction du signal , Cellules synoviales , Humains , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Cellules synoviales/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/immunologie , Facteur de transcription NF-kappa B/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cellules cultivées , Lignée cellulaire , Hyaluronoglucosaminidase
20.
Nat Commun ; 15(1): 7503, 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39209814

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Membrane synoviale , Humains , Membrane synoviale/anatomopathologie , Membrane synoviale/immunologie , Animaux , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/immunologie , Souris , Phénotype , Biologie informatique/méthodes , Inflammation/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE