Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.839
Filtrer
1.
J Transl Med ; 22(1): 715, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090667

RÉSUMÉ

BACKGROUND: Synovial fibrosis is a common complication of knee osteoarthritis (KOA), a pathological process characterized by myofibroblast activation and excessive extracellular matrix (ECM) deposition. Fibroblast-like synoviocytes (FLSs) are implicated in KOA pathogenesis, contributing to synovial fibrosis through diverse mechanisms. Nuclear protein 1 (NUPR1) is a recently identified transcription factor with crucial roles in various fibrotic diseases. However, its molecular determinants in KOA synovial fibrosis remain unknown. This study aims to investigate the role of NUPR1 in KOA synovial fibrosis through in vivo and in vitro experiments. METHODS: We examined NUPR1 expression in the murine synovium and determined the impact of NUPR1 on synovial fibrosis by knockdown models in the destabilization of the medial meniscus (DMM)-induced KOA mouse model. TGF-ß was employed to induce fibrotic response and myofibroblast activation in mouse FLSs, and the role and molecular mechanisms in synovial fibrosis were evaluated under conditions of NUPR1 downexpression. Additionally, the pharmacological effect of NUPR1 inhibitor in synovial fibrosis was assessed using a surgically induced mouse KOA model. RESULTS: We found that NUPR1 expression increased in the murine synovium after DMM surgical operation. The adeno-associated virus (AAV)-NUPR1 shRNA promoted NUPR1 deficiency, attenuating synovial fibrosis, inhibiting synovial hyperplasia, and significantly reducing the expression of pro-fibrotic molecules. Moreover, the lentivirus-mediated NUPR1 deficiency alleviated synoviocyte proliferation and inhibited fibroblast to myofibroblast transition. It also decreased the expression of fibrosis markers α-SMA, COL1A1, CTGF, Vimentin and promoted the activation of the SMAD family member 3 (SMAD3) pathway. Importantly, trifluoperazine (TFP), a NUPR1 inhibitor, attenuated synovial fibrosis in DMM mice. CONCLUSIONS: These findings indicate that NUPR1 is an antifibrotic modulator in KOA, and its effect on anti-synovial fibrosis is partially mediated by SMAD3 signaling. This study reveals a promising target for developing novel antifibrotic treatment.


Sujet(s)
Fibroblastes , Fibrose , Transduction du signal , Protéine Smad-3 , Cellules synoviales , Animaux , Protéine Smad-3/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Souris de lignée C57BL , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Mâle , Gonarthrose/anatomopathologie , Gonarthrose/métabolisme , Modèles animaux de maladie humaine , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Protéines de liaison à l'ADN , Protéines tumorales
2.
Cells ; 13(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39120270

RÉSUMÉ

Degenerative disorders like osteoarthritis (OA) might impair the ability of tissue-resident mesenchymal stem/stromal cells (MSCs) for tissue regeneration. As primary cells with MSC-like properties are exploited for patient-derived stem cell therapies, a detailed evaluation of their in vitro properties is needed. Here, we aimed to compare synovium-derived and bone-derived MSCs in early hip OA with those of patients without OA (non-OA). Tissues from three synovial sites of the hip (paralabral synovium, cotyloid fossa, inner surface of peripheral capsule) were collected along with peripheral trabecular bone from 16 patients undergoing hip arthroscopy (8 early OA and 8 non-OA patients). Primary cells isolated from tissues were compared using detailed in vitro analyses. Gene expression profiling was performed for the skeletal stem cell markers podoplanin (PDPN), CD73, CD164 and CD146 as well as for immune-related molecules to assess their immunomodulatory potential. Synovium-derived and bone-derived MSCs from early OA patients showed comparable clonogenicity, cumulative population doublings, osteogenic, adipogenic and chondrogenic potential, and immunophenotype to those of non-OA patients. High PDPN/low CD146 profile (reminiscent of skeletal stem cells) was identified mainly for non-OA MSCs, while low PDPN/high CD146 mainly defined early OA MSCs. These data suggest that MSCs from early OA patients are not affected by degenerative changes in the hip. Moreover, the synovium represents an alternative source of MSCs for patient-derived stem cell therapies, which is comparable to bone. The expression profile reminiscent of skeletal stem cells suggests the combination of low PDPN and high CD146 as potential biomarkers in early OA.


Sujet(s)
Cellules souches mésenchymateuses , Membrane synoviale , Humains , Cellules souches mésenchymateuses/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Différenciation cellulaire , Sujet âgé , Arthrose/anatomopathologie , Arthrose/métabolisme , Os et tissu osseux/anatomopathologie , Os et tissu osseux/métabolisme , Adulte , Marqueurs biologiques/métabolisme , Chondrogenèse , Ostéogenèse , Cellules cultivées
3.
Front Immunol ; 15: 1381272, 2024.
Article de Anglais | MEDLINE | ID: mdl-39139555

RÉSUMÉ

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


Sujet(s)
Polyarthrite rhumatoïde , Marqueurs biologiques , Métabolomique , Protéomique , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Humains , Métabolomique/méthodes , Protéomique/méthodes , Génomique/méthodes , Animaux , Voies et réseaux métaboliques , Membrane synoviale/immunologie , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Multi-omique
4.
Front Immunol ; 15: 1428773, 2024.
Article de Anglais | MEDLINE | ID: mdl-39161769

RÉSUMÉ

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


Sujet(s)
Polyarthrite rhumatoïde , Réseaux de régulation génique , Membrane synoviale , Humains , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/immunologie , Membrane synoviale/métabolisme , Membrane synoviale/immunologie , Membrane synoviale/anatomopathologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Analyse de profil d'expression de gènes , Biologie informatique/méthodes , Cellules synoviales/métabolisme , Arthrose/génétique , Arthrose/métabolisme , Régulation de l'expression des gènes , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Transcriptome
5.
Stem Cell Res Ther ; 15(1): 261, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39148121

RÉSUMÉ

BACKGROUND: Human adipose-derived stem cells (ADSCs) exert a strong anti-inflammatory effect, and synovium-derived stem cells (SDSCs) have high chondrogenic potential. Thus, this study aims to investigate whether a combination of human ADSCs and SDSCs will have a synergistic effect that will increase the chondrogenic potential of osteoarthritis (OA) chondrocytes in vitro and attenuate the cartilage degeneration of early and advanced OA in vitro. METHODS: ADSCs, SDSCs, and chondrocytes were isolated from OA patients who underwent total knee arthroplasty. The ADSCs-SDSCs mixed cell ratios were 1:0 (ADSCs only), 8:2, 5:5 (5A5S), 2:8, and 0:1 (SDSCs only). The chondrogenic potential of the OA chondrocytes was evaluated in vitro with a transwell assay or pellet culture with various mixed cell groups. The mixed cell group with the highest chondrogenic potential was then selected and injected into the knee joints of nude rats of early and advanced OA stages in vivo. The animals were then evaluated 12 and 20 weeks after surgery through gait analysis, von frey test, microcomputed tomography, MRI, and immunohistochemical and histological analyses. Finally, the mechanisms underlying these findings were investigated through the RNA sequencing of tissue samples in vivo and Western blot of the OA chondrocyte autophagy pathway. RESULTS: Among the MSCs treatment groups, 5A5S had the greatest synergistic effect that increased the chondrogenic potential of OA chondrocytes in vitro and inhibited early and advanced OA in vivo. The 5A5S group significantly reduced cartilage degeneration, synovial inflammation, pain sensation, and nerve invasion in subchondral nude rat OA, outperforming both single-cell treatments. The underlying mechanism was the activation of chondrocyte autophagy via the FoxO1 signaling pathway. CONCLUSION: A combination of human ADSCs and SDSCs demonstrated higher potential than a single type of stem cell, demonstrating potential as a novel treatment for OA.


Sujet(s)
Autophagie , Chondrocytes , Protéine O1 à motif en tête de fourche , Cellules souches mésenchymateuses , Arthrose , Transduction du signal , Humains , Chondrocytes/métabolisme , Animaux , Rats , Arthrose/thérapie , Arthrose/métabolisme , Arthrose/anatomopathologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Protéine O1 à motif en tête de fourche/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Mâle , Rat nude , Chondrogenèse , Membrane synoviale/métabolisme , Membrane synoviale/cytologie , Adulte d'âge moyen , Femelle
6.
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
7.
J Biomech ; 174: 112272, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39146899

RÉSUMÉ

The synovium plays a crucial role in diarthrodial joint health, and its study has garnered appreciation as synovitis has been linked to osteoarthritis symptoms and progression. Quantitative synovium structure-function data, however, remain sparse. In the present study, we hypothesized that tissue glycosaminoglycan (GAG) content contributes to the low friction properties of the synovium. Bovine and human synovium tribological properties were evaluated using a custom friction testing device in two different cases: (1) proteoglycan depletion to isolate the influence of tissue GAGs in the synovium friction response and (2) interleukin-1 (IL) treatment to observe inflammation-induced structural and functional changes. Following proteoglycan depletion, synovium friction coefficients increased while GAG content decreased. Conversely, synovium explants treated with the proinflammatory cytokine IL exhibited elevated GAG concentrations and decreased friction coefficients. For the first time, a relationship between synovium friction coefficient and GAG concentration is demonstrated. The study of synovium tribology is necessary to fully understand the mechanical environment of the healthy and diseased joint.


Sujet(s)
Friction , Protéoglycanes , Membrane synoviale , Membrane synoviale/métabolisme , Humains , Bovins , Animaux , Protéoglycanes/métabolisme , Glycosaminoglycanes/métabolisme , Interleukine-1/métabolisme
8.
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 , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Animaux , Humains , Glycolyse , Mâle , Membrane synoviale/métabolisme , Acide lactique/métabolisme , Cellules endothéliales/métabolisme
9.
Arthritis Res Ther ; 26(1): 150, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160592

RÉSUMÉ

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.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Méthotrexate , Nécrose , Membrane synoviale , Humains , Méthotrexate/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Membrane synoviale/anatomopathologie , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Antirhumatismaux/usage thérapeutique , Sujet âgé , Études de cohortes , Adulte , Résultat thérapeutique , Immunohistochimie
10.
Cell Death Dis ; 15(8): 584, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122678

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Deubiquitinating enzyme CYLD , Fibroblastes , I-kappa B Kinase , MAP Kinase Kinase Kinases , Transduction du signal , Membrane synoviale , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Deubiquitinating enzyme CYLD/métabolisme , Deubiquitinating enzyme CYLD/génétique , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Animaux , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Souris , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL , Souris knockout
11.
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
12.
Epigenetics ; 19(1): 2375011, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38956836

RÉSUMÉ

Mesenchymal stem cells (MSCs), with the ability to differentiate into osteoblasts, adipocytes, or chondrocytes, show evidence that the donor cell's metabolic type influences the osteogenic process. Limited knowledge exists on DNA methylation changes during osteogenic differentiation and the impact of diverse donor genetic backgrounds on MSC differentiation. In this study, synovial membrane mesenchymal stem cells (SMSCs) from two pig breeds (Angeln Saddleback, AS; German Landrace, DL) with distinct metabolic phenotypes were isolated, and the methylation pattern of SMSCs during osteogenic induction was investigated. Results showed that most differentially methylated regions (DMRs) were hypomethylated in osteogenic-induced SMSC group. These DMRs were enriched with genes of different osteogenic signalling pathways at different time points including Wnt, ECM, TGFB and BMP signalling pathways. AS pigs consistently exhibited a higher number of hypermethylated DMRs than DL pigs, particularly during the peak of osteogenesis (day 21). Predicting transcription factor motifs in regions of DMRs linked to osteogenic processes and donor breeds revealed influential motifs, including KLF1, NFATC3, ZNF148, ASCL1, FOXI1, and KLF5. These findings contribute to understanding the pattern of methylation changes promoting osteogenic differentiation, emphasizing the substantial role of donor the metabolic type and epigenetic memory of different donors on SMSC differentiation.


Sujet(s)
Différenciation cellulaire , Méthylation de l'ADN , Cellules souches mésenchymateuses , Ostéogenèse , Membrane synoviale , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Ostéogenèse/génétique , Suidae , Membrane synoviale/cytologie , Membrane synoviale/métabolisme , Cellules cultivées , Épigenèse génétique
13.
Anal Cell Pathol (Amst) ; 2024: 1083143, 2024.
Article de Anglais | MEDLINE | ID: mdl-38946863

RÉSUMÉ

Objectives: Osteochondral defects (OCDs) are localized areas of damaged cartilage and underlying subchondral bone that can produce pain and seriously impair joint function. Literature reports indicated that icariin (ICA) has the effect of promoting cartilage repair. However, its mechanism remains unclear. Here, we explored the effects of icariin and extracellular vesicles (EVs) from rabbit synovial-derived mesenchymal stem cells (rSMSCs) on repairing of OCDs. Materials and Methods: Rabbit primary genicular chondrocytes (rPGCs), knee skeletal muscle cells (rSMCKs), and rSMSCs, and extracellular vesicles derived from the latter two cells (rSMCK-EVs and rSMSC-EVs) were isolated and identified. The rPGCs were stimulated with ICA, rSMSC-EVs either separately or in combination. The rSMCK-EVs were used as a control. After stimulation, chondrogenic-related markers were analyzed by quantitative RT-PCR and western blotting. Cell proliferation was determined by the CCK-8 assay. The preventative effects of ICA and SMSC-EVs in vivo were determined by H&E and toluidine blue staining. Immunohistochemical analyses were performed to evaluate the levels of COL2A1 and ß-catenin in vivo. Results. In vitro, the proliferation of rPGCs was markedly increased by ICA treatment in a dose-dependent manner. When compared with ICA or rSMSC-EVs treatment alone, combined treatment with ICA and SMSC-EVs produced stronger stimulative effects on cell proliferation. Moreover, combined treatment with ICA and rSMSC-EVs promoted the expression of chondrogenic-related gene, including COL2A1, SOX-9, and RUNX2, which may be via the activation of the Wnt/ß-catenin pathway. In vivo, combined treatment with rSMSC-EVs and ICA promoted cartilage repair in joint bone defects. Results also showed that ICA or rSMSC-EVs both promoted the COL2A1 and ß-catenin protein accumulation in articular cartilage, and that was further enhanced by combined treatment with rSMSC-EVs and ICA. Conclusion: Our findings highlight the promising potential of using combined treatment with ICA and rSMSC-EVs for promoting osteochondral repair.


Sujet(s)
Chondrocytes , Chondrogenèse , Vésicules extracellulaires , Flavonoïdes , Cellules souches mésenchymateuses , Membrane synoviale , Voie de signalisation Wnt , Animaux , Lapins , Flavonoïdes/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Vésicules extracellulaires/métabolisme , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Membrane synoviale/cytologie , Chondrogenèse/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/effets des médicaments et des substances chimiques
14.
Clin Exp Rheumatol ; 42(7): 1387-1397, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38976290

RÉSUMÉ

OBJECTIVES: The imbalance between apoptosis and proliferation in fibroblast-like synoviocytes (FLSs) plays a key role in the pathogenesis of rheumatoid arthritis (RA). This study aims to investigate the potential of all-trans retinoic acid (ATRA) as a supplementary therapeutic agent alongside methotrexate (MTX) for RA, by examining its ability to inhibit synovial cell proliferation and enhance apoptosis through the ROS-JNK signalling pathway. METHODS: The viability, apoptosis, and autophagy levels of human rheumatoid arthritis fibroblast-like synovial cells (HFLS-RA) were evaluated, while ROS generation was measured through the DCFH-DA fluorescence microplate assay. Western blotting was used to analyse the expression levels of JNK signalling pathway-related proteins. To assess therapeutic potential in vivo, a collagen-induced arthritis (CIA) model was established in Wistar rats. RESULTS: Small doses of MTX did not significantly affect the viability of HFLS-RAs or induce apoptosis. However, when ATRA was added to the treatment, the therapy markedly inhibited cell proliferation and induced apoptosis and excessive autophagy. Mechanistically, ATRA activated the ROS/JNK signalling pathway in HFLS-RAs. ROS scavengers and JNK inhibitors significantly attenuated ATRA-induced apoptosis and autophagy. In vivo, the combination therapy demonstrated a remarkable enhancement of the anti-arthritic efficacy in CIA rats. CONCLUSIONS: The ability of ATRA to inhibit proliferation in RA FLSs through autophagy and apoptosis underscores its potential as a supplementary therapeutic agent alongside MTX for RA, particularly when compared to the limited impact of MTX on these processes. This combined strategy holds promise for enhancing therapeutic outcomes and warrants further investigation in the management of RA.


Sujet(s)
Apoptose , Arthrite expérimentale , Polyarthrite rhumatoïde , Autophagie , Prolifération cellulaire , Méthotrexate , Rat Wistar , Espèces réactives de l'oxygène , Cellules synoviales , Trétinoïne , Trétinoïne/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Méthotrexate/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Animaux , Humains , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Espèces réactives de l'oxygène/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie , Cellules synoviales/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Association de médicaments , Antirhumatismaux/pharmacologie , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Mâle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Rats , Lignée cellulaire
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1098-1108, 2024 Jun 20.
Article de Chinois | MEDLINE | ID: mdl-38977339

RÉSUMÉ

OBJECTIVE: To identify the biomarkers for early rheumatoid arthritis (RA) diagnosis and explore the possible immune regulatory mechanisms. METHODS: The differentially expressed genesin RA were screened and functionally annotated using the limma, RRA, batch correction, and clusterProfiler. The protein-protein interaction network was retrieved from the STRING database, and Cytoscape 3.8.0 and GeneMANIA were used to select the key genes and predicting their interaction mechanisms. ROC curves was used to validate the accuracy of diagnostic models based on the key genes. The disease-specific immune cells were selected via machine learning, and their correlation with the key genes were analyzed using Corrplot package. Biological functions of the key genes were explored using GSEA method. The expression of STAT1 was investigated in the synovial tissue of rats with collagen-induced arthritis (CIA). RESULTS: We identified 9 core key genes in RA (CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL), which regulate synovial inflammation primarily through cytokines-related pathways. ROC curve analysis showed a high predictive accuracy of the 9 core genes, among which STAT1 had the highest AUC (0.909). Correlation analysis revealed strong correlations of CD3G, ITGAL, LCK, CD8A, and STAT1 with disease-specific immune cells, and STAT1 showed the strongest correlation with M1-type macrophages (R=0.68, P=2.9e-08). The synovial tissues of the ankle joints of CIA rats showed high expressions of STAT1 and p-STAT1 with significant differential expression of STAT1 between the nucleus and the cytoplasm of the synovial fibroblasts. The protein expressions of p-STAT1 and STAT1 in the cell nuclei were significantly reduced after treatment. CONCLUSION: CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL may serve as biomarkers for early diagnosis of RA. Gene-immune cell pathways such as CD3G/CD8A/LCK-γδ T cells, ITGAL-Tfh cells, and STAT1-M1-type macrophages may be closely related with the development of RA.


Sujet(s)
Polyarthrite rhumatoïde , Marqueurs biologiques , Cartes d'interactions protéiques , Facteur de transcription STAT-1 , Membrane synoviale , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Animaux , Rats , Facteur de transcription STAT-1/métabolisme , Marqueurs biologiques/métabolisme , Membrane synoviale/métabolisme , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/métabolisme , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/génétique , Analyse de profil d'expression de gènes , Bases de données génétiques , Humains , Antigènes CD8/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Syk kinase/métabolisme , Syk kinase/génétique , Courbe ROC
16.
Toxicol Appl Pharmacol ; 490: 117033, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38997070

RÉSUMÉ

Gallic acid (GA) has been found by a large number of studies to have pharmacological effects such as antioxidant and anti-inflammatory properties. However, the underlying therapeutic mechanisms are not fully understood.. Studies have shown that altering the intestinal flora affects host metabolism and effectively mediates the development of synovitis. The aim of this study was to explore the pharmacological effects of GA in the treatment of synovial inflammation and anti-synovial fibrosis in knee osteoarthritis (KOA) and the underlying mechanisms by macrogenomics combined with off-target metabolomics. We established a synovitis model via in vivo and in vitro experiments to observe the effect of GA intervention on synovitis. Moreover, we collected serum and feces from rats and analyzed the changes in intestinal flora by macro-genome sequencing and the changes in metabolites in the serum by untargeted metabolomics. We found that GA reduced the levels of IL-1ß, IL-6, and TNF-α, and decreased the protein expression levels of α-SMA, TGF-ß, and Collagen I in synovial tissues and cells, and the composition and function of the intestinal flora were similarly altered. Combined with macrogenomic pathway enrichment analysis and metabolic pathway enrichment analysis, these findings revealed that GA impacts Bacteroidia and Muribaculaceae abundance, and via the following metabolic pathways: sphingolipid metabolism, glycerophospholipid metabolism, and arginine biology.to ameliorate synovial inflammation and fibrosis in KOA. The therapeutic effect of GA on KOA synovitis and fibrosis is partly attributed to the alleviation of metabolic disorder and the rebalancing of the intestinal flora. These results provides a rationale for the therapeutic application of GA in the treatment of synovitis.


Sujet(s)
Fibrose , Acide gallique , Microbiome gastro-intestinal , Rat Sprague-Dawley , Animaux , Acide gallique/pharmacologie , Acide gallique/usage thérapeutique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Mâle , Rats , Synovite/traitement médicamenteux , Synovite/anatomopathologie , Synovite/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Modèles animaux de maladie humaine , Métabolomique
17.
Biochem Biophys Res Commun ; 729: 150354, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-38981403

RÉSUMÉ

Intra-articular injection of mesenchymal stem cells (MSCs) is envisioned as a solution for knee osteoarthritis (OA). Although synovial MSCs (SyMSCs) are promising for cartilage regeneration, the clinical choice is usually adipose MSCs (AdMSCs). However, the similarities/differences in the mode of action between SyMSCs and AdMSCs remain unclear. Here, we compared factors secreted by human SyMSCs and AdMSCs after injection into OA knees. Human SyMSCs or AdMSCs were injected into the knees of rat partial meniscectomy models. The next day, the knee joints were collected to analyze the distribution of injected MSCs and transcriptome changes in the human MSCs and rat synovium. Non-injected MSCs were mixed with rat synovium as a control. After injection, no difference was apparent in intra-articular distribution of the SyMSCs or AdMSCs. RNA sequencing demonstrated an enrichment of cytokine-cytokine receptor interaction-related genes in both human SyMSCs and AdMSCs after injection. Differentially expressed genes (DEGs) specific to SyMSCs were associated with cartilage matrix synthesis and homeostasis. PCR analysis of the matrisome-related DEGs showed significantly higher expression of PRG4 in SyMSCs than in AdMSCs after injection. Immunostaining also confirmed a significantly greater expression of lubricin by SyMSCs than by AdMSCs. These findings indicate that SyMSCs will be a more promising treatment for OA.


Sujet(s)
Tissu adipeux , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Gonarthrose , Membrane synoviale , Animaux , Cellules souches mésenchymateuses/métabolisme , Humains , Gonarthrose/métabolisme , Gonarthrose/thérapie , Gonarthrose/anatomopathologie , Gonarthrose/génétique , Rats , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Tissu adipeux/métabolisme , Tissu adipeux/cytologie , Injections articulaires , Mâle , Rat Sprague-Dawley , Glycoprotéines/métabolisme , Glycoprotéines/génétique , Cellules cultivées , Protéoglycanes/métabolisme , Protéoglycanes/génétique
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167341, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39025373

RÉSUMÉ

Fibroblast-like synoviocytes (FLS) plays an important role in synovial inflammation and joint damage in rheumatoid arthritis (RA). As the most abundant mRNA modification, N6-methyladenosine (m6A) is involved in the development of various diseases; however, its role in RA remains to be defined. In this study, we reported the elevated expression of the m6A demethylase fat mass and obesity-associated protein (FTO) in FLS and synovium from RA patients. Functionally, FTO knockdown or treatment with FB23-2, an inhibitor of the mRNA m6A demethylase FTO, inhibited the migration, invasion and inflammatory response of RA FLS, however, FTO-overexpressed RA FLS exhibited increased migration, invasion and inflammatory response. We further demonstrated that FTO promoted ADAMTS15 mRNA stability in an m6A-IGF2BP1 dependent manner. Notably, the severity of arthritis was significantly reduced in CIA mice with FB23-2 administration or CIA rats with intra-articular injection of FTO shRNA. Our results illustrate the contribution of FTO-mediated m6A modification to joint damage and inflammation in RA and suggest that FTO might be a potential therapeutic target in RA.


Sujet(s)
Adénosine , Alpha-ketoglutarate-dependent dioxygenase FTO , Polyarthrite rhumatoïde , Inflammation , , Animaux , Humains , Souris , Rats , Adénosine/analogues et dérivés , Adénosine/métabolisme , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Stabilité de l'ARN , ARN messager/génétique , ARN messager/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie
19.
Cell Biochem Funct ; 42(5): e4091, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38973151

RÉSUMÉ

The intron retention (IR) is a phenomenon utilized by cells to allow diverse fates at the same mRNA, leading to a different pattern of synthesis of the same protein. In this study, we analyzed the modulation of phosphoinositide-specific phospholipase C (PI-PLC) enzymes by Harpagophytum procumbens extract (HPE) in synoviocytes from joins of osteoarthritis (OA) patients. In some samples, the PI-PLC γ1 isoform mature mRNA showed the IR and, in these synoviocytes, the HPE treatment increased the phenomenon. Moreover, we highlighted that as a consequence of IR, a lower amount of PI-PLC γ1 was produced. The decrease of PI-PLC γ1 was associated with the decrease of metalloprotease-3 (MMP-3), and MMP-13, and ADAMTS-5 after HPE treatment. The altered expression of MMPs is a hallmark of the onset and progression of OA, thus substances able to decrease their expression are very desirable. The interesting outcomes of this study are that 35% of analyzed synovial tissues showed the IR phenomenon in the PI-PLC γ1 mRNA and that the HPE treatment increased this phenomenon. For the first time, we found that the decrease of PI-PLC γ1 protein in synoviocytes interferes with MMP production, thus affecting the pathways involved in the MMP expression. This finding was validated by the silencing of PI-PLC γ1 in synoviocytes where the IR phenomenon was not present. Our results shed new light on the biochemical mechanisms involved in the degrading enzyme production in the joint of OA patients, suggesting a new therapeutic target and highlighting the importance of personalized medicine.


Sujet(s)
Fibroblastes , Introns , Phospholipase C gamma , ARN messager , Humains , ARN messager/métabolisme , ARN messager/génétique , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Phospholipase C gamma/métabolisme , Phospholipase C gamma/génétique , Cellules cultivées , Arthrose/métabolisme , Arthrose/anatomopathologie , Membrane synoviale/métabolisme , Membrane synoviale/cytologie , Membrane synoviale/effets des médicaments et des substances chimiques , Matrix metalloproteinase 3/métabolisme , Matrix metalloproteinase 3/génétique , Protéine ADAMTS5/métabolisme , Protéine ADAMTS5/génétique , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Matrix Metalloproteinase 13/métabolisme , Matrix Metalloproteinase 13/génétique
20.
Biomolecules ; 14(7)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39062498

RÉSUMÉ

With the rise in longevity within the population, medicine continues to encounter fresh hurdles necessitating prompt actions, among which are those associated with hip joint aging. Age-related arthropathies encompass damage to bones' articulating extremities and their supporting structures, such as articular cartilage, and alterations in the quantity and quality of synovial fluid. This study aims to summarize the biomolecular methods of hip joint evaluation focused on its vascularization, using data correlated with biomolecular research on other joints and tissues, in order to reach an objective opinion of the study prospects in this field. Following a retrospective study on most modern biomolecular research methods on the synovium, the capsule, and the articular cartilage of the hip joint, we have hereby concretized certain future research directions in this field that will improve the qualitative and morphofunctional management of the hip joint at an advanced age, even within population categories at risk of developing various degenerative joint pathologies.


Sujet(s)
Cartilage articulaire , Articulation de la hanche , Humains , Articulation de la hanche/anatomopathologie , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Conflit fémoro-acétabulaire/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE