Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.627
Filtrer
1.
Front Immunol ; 15: 1398990, 2024.
Article de Anglais | MEDLINE | ID: mdl-39086489

RÉSUMÉ

Background: More and more evidence supports the association between myocardial infarction (MI) and osteoarthritis (OA). The purpose of this study is to explore the shared biomarkers and pathogenesis of MI complicated with OA by systems biology. Methods: Gene expression profiles of MI and OA were downloaded from the Gene Expression Omnibus (GEO) database. The Weighted Gene Co-Expression Network Analysis (WGCNA) and differentially expressed genes (DEGs) analysis were used to identify the common DEGs. The shared genes related to diseases were screened by three public databases, and the protein-protein interaction (PPI) network was built. GO and KEGG enrichment analyses were performed on the two parts of the genes respectively. The hub genes were intersected and verified by Least absolute shrinkage and selection operator (LASSO) analysis, receiver operating characteristic (ROC) curves, and single-cell RNA sequencing analysis. Finally, the hub genes differentially expressed in primary cardiomyocytes and chondrocytes were verified by RT-qPCR. The immune cell infiltration analysis, subtypes analysis, and transcription factors (TFs) prediction were carried out. Results: In this study, 23 common DEGs were obtained by WGCNA and DEGs analysis. In addition, 199 common genes were acquired from three public databases by PPI. Inflammation and immunity may be the common pathogenic mechanisms, and the MAPK signaling pathway may play a key role in both disorders. DUSP1, FOS, and THBS1 were identified as shared biomarkers, which is entirely consistent with the results of single-cell RNA sequencing analysis, and furher confirmed by RT-qPCR. Immune infiltration analysis illustrated that many types of immune cells were closely associated with MI and OA. Two potential subtypes were identified in both datasets. Furthermore, FOXC1 may be the crucial TF, and the relationship of TFs-hub genes-immune cells was visualized by the Sankey diagram, which could help discover the pathogenesis between MI and OA. Conclusion: In summary, this study first revealed 3 (DUSP1, FOS, and THBS1) novel shared biomarkers and signaling pathways underlying both MI and OA. Additionally, immune cells and key TFs related to 3 hub genes were examined to further clarify the regulation mechanism. Our study provides new insights into shared molecular mechanisms between MI and OA.


Sujet(s)
Marqueurs biologiques , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Infarctus du myocarde , Arthrose , Cartes d'interactions protéiques , Biologie des systèmes , Infarctus du myocarde/génétique , Infarctus du myocarde/immunologie , Arthrose/génétique , Arthrose/métabolisme , Humains , Bases de données génétiques , Transcriptome , Chondrocytes/métabolisme , Chondrocytes/immunologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Animaux , Biologie informatique/méthodes
2.
Autoimmunity ; 57(1): 2384889, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39086231

RÉSUMÉ

Osteoarthritis (OA) is a worldwide joint disease, leading to the physical pain, stiffness, and even disability. Lactate dehydrogenase A (LDHA) is known as a lactylation mediator that can regulate histone lactylation of its target genes. However, the role of LDHA-mediated histone H3 lysine 18 lactylation (H3K18la) in OA progression is yet to be clarified. Our study aims at revealing the role and mechanism of LDHA-mediated histone lactylation in the glycolysis of chondrocytes. In this study, we determined at first that the H3K18la level was enhanced in OA. Energy metabolism such as glycolysis is often altered in OA progress. Therefore, we further explored the mechanism mediating glycolysis and thus promoting OA progress. Moreover, glycolysis was enhanced in LPS-induced OA cell model, as evidenced by the increased glucose consumption and lactate production. Furthermore, we silenced LDHA for loss-of-function assays. The results showed that knockdown of LDHA suppressed glycolysis of LPS-induced chondrocytes. In vivo animal study demonstrated that knockout of LDHA recovered cartilage injury of OA mice. Mechanistically, we uncovered that LDHA-mediated H3K18la in TPI1 promoter enhanced the transcription activity of TPI1. Mutation of K69 site was found to ameliorate LPS-induced glycolysis in OA cell model. In conclusion, our study reveals the role of LDHA-mediated H3K18la of TPI1 promoter in OA progress.


Sujet(s)
Chondrocytes , Glycolyse , Histone , Arthrose , Arthrose/métabolisme , Arthrose/génétique , Arthrose/anatomopathologie , Animaux , Souris , Histone/métabolisme , Humains , Chondrocytes/métabolisme , Modèles animaux de maladie humaine , Lactate dehydrogenase 5/métabolisme , Mâle , Régulation de l'expression des gènes , Souris knockout , Régions promotrices (génétique) , Transcription génétique
3.
J Nanobiotechnology ; 22(1): 466, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095867

RÉSUMÉ

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive degeneration of articular cartilage, leading to pain, stiffness, and loss of joint function. The pathogenesis of OA involves multiple factors, including increased intracellular reactive oxygen species (ROS), enhanced chondrocyte apoptosis, and disturbances in cartilage matrix metabolism. These processes contribute to the breakdown of the extracellular matrix (ECM) and the loss of cartilage integrity, ultimately resulting in joint damage and dysfunction. RNA interference (RNAi) therapy has emerged as a promising approach for the treatment of various diseases, including hATTR and acute hepatic porphyria. By harnessing the natural cellular machinery for gene silencing, RNAi allows for the specific inhibition of target genes involved in disease pathogenesis. In the context of OA, targeting key molecules such as matrix metalloproteinase-13 (MMP13), which plays a critical role in cartilage degradation, holds great therapeutic potential. RESULTS: In this study, we developed an innovative therapeutic approach for OA using a combination of liposome-encapsulated siMMP13 and NG-Monomethyl-L-arginine Acetate (L-NMMA) to form an injectable hydrogel. The hydrogel served as a delivery vehicle for the siMMP13, allowing for sustained release and targeted delivery to the affected joint. Experiments conducted on destabilization of the medial meniscus (DMM) model mice demonstrated the therapeutic efficacy of this composite hydrogel. Treatment with the hydrogel significantly inhibited the degradation of cartilage matrix, as evidenced by histological analysis showing preserved cartilage structure and reduced loss of proteoglycans. Moreover, the hydrogel effectively suppressed intracellular ROS accumulation in chondrocytes, indicating its anti-oxidative properties. Furthermore, it attenuated chondrocyte apoptosis, as demonstrated by decreased levels of apoptotic markers. CONCLUSION: In summary, the injectable hydrogel containing siMMP13, endowed with anti-ROS and anti-apoptotic properties, may represent an effective therapeutic strategy for osteoarthritis in the future.


Sujet(s)
Apoptose , Chondrocytes , Hydrogels , Matrix Metalloproteinase 13 , Arthrose , Espèces réactives de l'oxygène , Animaux , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Matrix Metalloproteinase 13/métabolisme , Souris , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mâle , Cartilage articulaire/métabolisme , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Liposomes/composition chimique , Humains
4.
J Cell Mol Med ; 28(15): e18574, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39087591

RÉSUMÉ

Osteoarthritis (OA) is a widespread inflammatory joint disease with significant global disability burden. Cuproptosis, a newly identified mode of cell death, has emerged as a crucial factor in various pathological conditions, including OA. In this context, our study aims to investigate the intrinsic relationship between cuproptosis-related genes (CRGs) and OA, and assess their potential as biomarkers for OA diagnosis and treatment. Datasets from the GEO databases were analysed the differential expression of CRGs, leading to the identification of 10 key CRGs (CDKN2A, DLD, FDX1, GLS, LIAS, LIPT1, MTF1, PDHA1, DLAT and PDHB). A logistic regression analysis and calibration curves were used to show excellent diagnostic accuracy. Consensus clustering revealed two CRG patterns, with Cluster 1 indicating a closer association with OA progression. RT-PCR confirmed a significant increase in the expression levels of these nine key genes in IL-1ß-induced C28/i2 cells, and the expression of CDKN2A and FDX1 were also elevated in conditioned monocytes, while the expression of GLS and MTF1 were significantly decreased. In vitro experiments demonstrated that the expression levels of these 7/10 CRGs were significantly increased in chondrocytes induced by IL-1ß, and upon stimulation with cuproptosis inducers, chondrocyte apoptosis was exacerbated, accompanied by an increase in the expression of cuproptosis-related proteins. These further substantiated our research findings and indicated that the nine selected cuproptosis genes have high potential for application in the diagnosis of OA.


Sujet(s)
Chondrocytes , Arthrose , Humains , Arthrose/génétique , Facteurs de risque , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Marqueurs biologiques/métabolisme , Interleukine-1 bêta/génétique , Régulation de l'expression des gènes , Monocytes/métabolisme , Analyse de profil d'expression de gènes
5.
Physiol Res ; 73(3): 415-426, 2024 07 17.
Article de Anglais | MEDLINE | ID: mdl-39027958

RÉSUMÉ

Recent research has identified that miR-539-3p impedes chondrogenic differentiation, yet its specific role and underlying mechanisms in childhood-onset osteoarthritis (OA) remain unclear. This study found that miR-539-3p levels were considerably lower in cartilage samples derived from childhood-onset OA patients compared to the control group. Enhancing miR-539-3p expression or suppressing RUNX2 expression notably reduced apoptosis, inflammation, and extracellular matrix (ECM) degradation in OA chondrocytes. In contrast, reducing miR-539-3p or increasing RUNX2 had the opposite effects. RUNX2 was confirmed as a direct target of miR-539-3p. Further experiments demonstrated that miR-539-3p targeting RUNX2 effectively lessened apoptosis, inflammation, and ECM degradation in OA chondrocytes, accompanied by changes in key molecular markers like reduced caspase-3 and matrix etallopeptidase 13 (MMP-13) levels, and increased B-cell lymphoma 2 (Bcl-2) and collagen type X alpha 1 chain (COL2A1). This study underscores the pivotal role of miR-539-3p in alleviating inflammation and ECM degradation in childhood-onset OA through targeting RUNX2, offering new insights for potential therapeutic strategies against this disease.


Sujet(s)
Apoptose , Chondrocytes , Sous-unité alpha 1 du facteur CBF , Matrice extracellulaire , microARN , Arthrose , Humains , microARN/métabolisme , microARN/génétique , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Enfant , Mâle , Femelle , Cellules cultivées , Adolescent
6.
J Orthop Surg Res ; 19(1): 429, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054551

RÉSUMÉ

BACKGROUND: The relationship between thyroid hormone (TH) levels in vivo and osteoarthritis (OA) remains inconclusive. This study aims to investigate the association between TH levels and OA, analyze the effect of triiodothyronine on hypertrophic chondrocyte differentiation and OA progression, and identify potential target genes of triiodothyronine in OA to evaluate its diagnostic value. METHODS: Two-sample mendelian randomization method was used to probe the causal links between hyperthyroidism and OA. Differentially expressed genes (DEGs) from two RNA-sequencing data in Gene Expression Omnibus (GSE199847 and GSE114007) and enrichment analysis of DEGs (166 commonly upregulated genes and 71 commonly downregulated genes of GSE199847 and GSE114007) was performed to analyze the effect of triiodothyronine (T3) on hypertrophic chondrocyte differentiation and OA. C28/I2 cells treated with T3 and reverse transcription and quantitative real-time polymerase chain reaction were used to validate T3 targeted genes. The diagnostic performance of target genes was assessed by the receiver operating characteristic (ROC) curve and area under the curve (AUC). RESULTS: There was a positive causal association between hyperthyroidism and OA (IVW result, OR = 1.330, 95% CI 1.136-1.557, P = 0.0004). Weighted median and Weighted mode analysis also demonstrated that hyperthyroidism had a positive causal association with OA (p < 0.05, OR > 1). Bioinformatics analysis indicated T3 can partially induce the emergence of late hypertrophic chondrocyte and promote OA through extracellular matrix organization, blood vessel development, skeletal system development and ossification. Post-T3 treatment, MAFB, C1QTNF1, COL3A1 and ANGPTL2 were significantly elevated in C28/I2 cells. ROC curves in GSE114007 showed that AUC of all above genes were ≥ 0.7. CONCLUSIONS: This study identified that hyperthyroidism has a positive causal association with OA by MR analysis. T3 induced hypertrophic chondrocytes promote OA progression by upregulating genes such as MAFB, C1QTNF1, COL3A1 and ANGPTL2, which can also serve as OA diagnosis.


Sujet(s)
Hyperthyroïdie , Analyse de randomisation mendélienne , Arthrose , Analyse de séquence d'ARN , Tri-iodothyronine , Analyse de randomisation mendélienne/méthodes , Arthrose/génétique , Humains , Hyperthyroïdie/génétique , Hyperthyroïdie/complications , Tri-iodothyronine/sang , Analyse de séquence d'ARN/méthodes , Chondrocytes/métabolisme , Différenciation cellulaire/génétique , Évolution de la maladie
7.
J Transl Med ; 22(1): 662, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010104

RÉSUMÉ

BACKGROUND: Temporomandibular joint osteoarthritis (TMJOA) has a high incidence rate, but its pathogenesis remains unclear. Circadian rhythm is an important oscillation in the human body and influences various biological activities. However, it is still unclear whether circadian rhythm affects the onset and development of TMJOA. METHODS: We disrupted the normal rhythm of rats and examined the expression of core clock genes in the mandibular condylar cartilage of the jaw and histological changes in condyles. After isolating rat mandibular condylar chondrocytes, we upregulated or downregulated the clock gene Per1, examined the expression of cartilage matrix-degrading enzymes, tested the activation of the GSK3ß/ß-CATENIN pathway and verified it using agonists and inhibitors. Finally, after downregulating the expression of Per1 in the mandibular condylar cartilage of rats with jet lag, we examined the expression of cartilage matrix-degrading enzymes and histological changes in condyles. RESULTS: Jet lag led to TMJOA-like lesions in the rat mandibular condyles, and the expression of the clock gene Per1 and cartilage matrix-degrading enzymes increased in the condylar cartilage of rats. When Per1 was downregulated or upregulated in mandibular condylar chondrocytes, the GSK3ß/ß-CATENIN pathway was inhibited or activated, and the expression of cartilage matrix-degrading enzymes decreased or increased, which can be rescued by activator and inhibitor of the GSK3ß/ß-CATENIN pathway. Moreover, after down-regulation of Per1 in mandibular condylar cartilage in vivo, significant alleviation of cartilage degradation, cartilage loss, subchondral bone loss induced by jet lag, and inhibition of the GSK3ß/ß-CATENIN signaling pathway were observed. Circadian rhythm disruption can lead to TMJOA. The clock gene Per1 can promote the occurrence of TMJOA by activating the GSK3ß/ß-CATENIN pathway and promoting the expression of cartilage matrix-degrading enzymes. The clock gene Per1 is a target for the prevention and treatment of TMJOA.


Sujet(s)
Chondrocytes , Rythme circadien , Glycogen synthase kinase 3 beta , Condyle mandibulaire , Arthrose , Protéines circadiennes Period , Articulation temporomandibulaire , Régulation positive , bêta-Caténine , Animaux , Glycogen synthase kinase 3 beta/métabolisme , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , bêta-Caténine/métabolisme , Arthrose/anatomopathologie , Arthrose/métabolisme , Protéines circadiennes Period/métabolisme , Protéines circadiennes Period/génétique , Condyle mandibulaire/anatomopathologie , Condyle mandibulaire/métabolisme , Articulation temporomandibulaire/anatomopathologie , Articulation temporomandibulaire/métabolisme , Mâle , Rat Sprague-Dawley , Transduction du signal , Rats
8.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39000370

RÉSUMÉ

Osteoarthritis (OA) is a degenerative joint disorder that is distinguished by inflammation and chronic cartilage damage. Interleukin-1ß (IL-1ß) is a proinflammatory cytokine that plays an important role in the catabolic processes that underlie the pathogenesis of OA. In this study, we investigate the therapeutic efficacy of exosomes derived from untreated bone-marrow-derived mesenchymal stem cells (BMMSC-Exo) and those treated with cinnamaldehyde (BMMSC-CA-Exo) for preventing the in vitro catabolic effects of IL-1ß on chondrocytes. We stimulated chondrocytes with IL-1ß to mimic the inflammatory microenvironment of OA. We then treated these chondrocytes with BMMSC-Exo and BMMSC-CA-Exo isolated via an aqueous two-phase system and evaluated their effects on the key cellular processes using molecular techniques. Our findings revealed that treatment with BMMSC-Exo reduces the catabolic effects of IL-1ß on chondrocytes and alleviates inflammation. However, further studies directly comparing treatments with BMMSC-Exo and BMMSC-CA-Exo are needed to determine if CA preconditioning can provide additional anti-inflammatory benefits to the exosomes beyond those of CA preconditioning or treatment with regular BMMSC-Exo. Through a comprehensive molecular analysis, we elucidated the regulatory mechanisms underlying this protective effect. We found a significant downregulation of proinflammatory signaling pathways in exosome-infected chondrocytes, suggesting the potential modulation of the NF-κB and MAPK signaling cascades. Furthermore, our study identified the molecular cargo of BMMSC-Exo and BMMSC-CA-Exo, determining the key molecules, such as anti-inflammatory cytokines and cartilage-associated factors, that may contribute to their acquisition of chondroprotective properties. In summary, BMMSC-Exo and BMMSC-CA-Exo exhibit the potential as therapeutic agents for OA by antagonizing the in vitro catabolic effects of IL-1ß on chondrocytes. The regulation of the proinflammatory signaling pathways and bioactive molecules delivered by the exosomes suggests a multifaceted mechanism of action. These findings highlight the need for further investigation into exosome-based therapies for OA and joint-related diseases.


Sujet(s)
Acroléine , Chondrocytes , Exosomes , Inflammation , Interleukine-1 bêta , Cellules souches mésenchymateuses , Transduction du signal , Exosomes/métabolisme , Interleukine-1 bêta/métabolisme , Acroléine/analogues et dérivés , Acroléine/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Animaux , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Humains , Cellules cultivées
9.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000568

RÉSUMÉ

Osteoarthritis (OA) is the most common joint disease, causing symptoms such as joint pain, swelling, and deformity, which severely affect patients' quality of life. Despite advances in medical treatment, OA management remains challenging, necessitating the development of safe and effective drugs. Quercetin (QUE), a natural flavonoid widely found in fruits and vegetables, shows promise due to its broad range of pharmacological effects, particularly in various degenerative diseases. However, its role in preventing OA progression and its underlying mechanisms remain unclear. In this study, we demonstrated that QUE has a protective effect against OA development both in vivo and in vitro, and we elucidated the underlying molecular mechanisms. In vitro, QUE inhibited the expression of IL-1ß-induced chondrocyte matrix metalloproteinases (MMP3 and MMP13) and inflammatory mediators such as INOS and COX-2. It also promoted the expression of collagen II, thereby preventing the extracellular matrix (ECM). Mechanistically, QUE exerts its protective effect on chondrocytes by activating the SIRT1/Nrf-2/HO-1 and inhibiting chondrocyte ferroptosis. Similarly, in an OA rat model induced by anterior cruciate ligament transection (ACLT), QUE treatment improved articular cartilage damage, reduced joint pain, and normalized abnormal subchondral bone remodeling. QUE also reduced serum IL-1ß, TNF-α, MMP3, CTX-II, and COMP, thereby slowing the progression of OA. QUE exerts chondroprotective effects by inhibiting chondrocyte oxidative damage and ferroptosis through the SIRT1/Nrf-2/HO-1 pathway, effectively alleviating OA progression in rats.


Sujet(s)
Cartilage articulaire , Chondrocytes , Modèles animaux de maladie humaine , Ferroptose , Facteur-2 apparenté à NF-E2 , Arthrose , Quercétine , Sirtuine-1 , Animaux , Sirtuine-1/métabolisme , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Rats , Quercétine/pharmacologie , Quercétine/usage thérapeutique , Facteur-2 apparenté à NF-E2/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Interleukine-1 bêta/métabolisme , Heme oxygenase (decyclizing)/métabolisme
10.
Tissue Eng Part C Methods ; 30(7): 314-322, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38946581

RÉSUMÉ

Current tissue engineering (TE) methods utilize chondrocytes primarily from costal or articular sources. Despite the robust mechanical properties of neocartilages sourced from these cells, the lack of elasticity and invasiveness of cell collection from these sources negatively impact clinical translation. These limitations invited the exploration of naturally elastic auricular cartilage as an alternative cell source. This study aimed to determine if auricular chondrocytes (AuCs) can be used for TE scaffold-free neocartilage constructs and assess their biomechanical properties. Neocartilages were successfully generated from a small quantity of primary neonatal AuCs of three minipig donors (n = 3). Neocartilage constructs had instantaneous moduli of 200.5 kPa ± 43.34 and 471.9 ± 92.8 kPa at 10% and 20% strain, respectively. TE constructs' relaxation moduli (Er) were 36.99 ± 6.47 kPa Er and 110.3 ± 16.99 kPa at 10% and 20% strain, respectively. The Young's modulus was 2.0 MPa ± 0.63, and the ultimate tensile strength was 0.619 ± 0.177 MPa. AuC-derived neocartilages contained 0.144 ± 0.011 µg collagen, 0.185 µg ± 0.002 glycosaminoglycans per µg dry weight, and 1.7e-3 µg elastin per µg dry weight. In conclusion, this study shows that AuCs can be used as a reliable and easily accessible cell source for TE of biomimetic and mechanically robust elastic neocartilage implants.


Sujet(s)
Chondrocytes , Cartilage de l'oreille , Cartilage élastique , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Animaux , Ingénierie tissulaire/méthodes , Chondrocytes/cytologie , Chondrocytes/métabolisme , Suidae , Cartilage de l'oreille/cytologie , Cartilage de l'oreille/physiologie , Cartilage élastique/cytologie , Structures d'échafaudage tissulaires/composition chimique , Porc miniature , Module d'élasticité , Cellules cultivées , Résistance à la traction
11.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3330-3339, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-39041096

RÉSUMÉ

This study aims to investigate the mechanism of Huangqin Qingre Chubi Capsules(HQC) in delaying chondrocyte senescence of osteoarthritic(OA) rats by regulating the p53/p21 signaling pathway. Rheumatic fever paralysis models of OA rats were induced based on monosodiun iodoacetate(MIA) combined with external rheumatic fever environmental stimuli and divided into normal(Con) group, OA model(MIA) group, OA model+rheumatic fever stimulation model(MIA-M) group, MIA-M+HQC low-dose(MIA-M+HQC-L) group, medium-dose(MIA-M+HQC-M) group, and high-dose(MIA-M+HQC-H) group, and MIA-M+glucosamine(MIA-M+GS) group. The models were successfully prepared and administered by gavage for 30 d. The pathological changes of cartilage were observed by hematoxylin-eosin(HE) and Senna O solid green(SO) staining. The expression of interleukin(IL)-1ß and IL-6 was detected by enzyme-linked immunosorbent assay(ELISA). Flow cytometry(FCM) was used to detect apoptosis and cell cycle. The mRNA expression of MMP13, ADAMTS-5, COLⅡ, and TGF-ß was detected by RT-qPCR. The protein expression of p53/p21, p16, Bax, and Bcl-2 was detected by Western blot. The articular cartilage surface of rats in the Con group was smooth, and the tide line was smooth. The cartilage layer of MIA and MIA-M groups was obviously damaged, and the cartilage matrix was reduced. The above conditions were more severe in the MIA-M group. The cartilage surface of the HQC high-dose group and MIA-M+GS group was basically intact with clear delamination. Compared with the MIA-M+HQC-H group, Mankin's score was higher in the HQC low-dose and medium-dose groups, and the change was not obvious in the MIA-M+GS group. Compared with the Con group, the proportion of chondrocytes G_1 was elevated in the MIA and MIA-M groups, and the proportion of the S phase and G_2 phase was significantly decreased. In addition, the apoptosis rate was increased. Compared with MIA-M, HQC groups inhibited apoptosis and promoted cell proliferation in a concentration-dependent manner. Compared with the MIA-M+HQC-H group, the effect was more significant in the HQC high-dose group than in the HQC medium-low dose, while it was not significant in the MIA-M+GS group. Compared with the Con group, IL-1ß and IL-6 were elevated in the MIA and MIA-M groups, and mRNA levels of MMP13 and ADAMTS-5 were elevated. p53, p21, p16, and Bax protein were elevated, and mRNA levels of COLⅡ and TGF-ß were decreased. Compared with the MIA-M group, IL-1ß and IL-6 decreased after drug interventions of HQC and GS, and mRNA levels of MMP13 and ADAMTS-5, as well as protein levels of p53, p21, Bax, and p16 decreased. In addition, Bcl-2 increased. The improvement of these indexes was significantly better in the MIA-M+HQC-H group than in the HQC low-dose and medium-dose groups, and the difference with the MIA-M+GS group was not significant. HQC delayed MIA-induced chondrocyte senescence in OA rats, inhibited inflammatory response and extracellular matrix(ECM) degradation, and its mechanism may be related to the inhibition of the p53/p21 pathway.


Sujet(s)
Chondrocytes , Médicaments issus de plantes chinoises , Arthrose , Rat Sprague-Dawley , Transduction du signal , Protéine p53 suppresseur de tumeur , Animaux , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Arthrose/génétique , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Mâle , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Capsules , Humains , Apoptose/effets des médicaments et des substances chimiques
12.
Ageing Res Rev ; 99: 102404, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971322

RÉSUMÉ

Osteoarthritis (OA) is a chronic degenerative joint disease with multiple causative factors such as aging, mechanical injury, and obesity. Autophagy is a complex dynamic process that is involved in the degradation and modification of intracellular proteins and organelles under different pathophysiological conditions. Autophagy, as a cell survival mechanism under various stress conditions, plays a key role in regulating chondrocyte life cycle metabolism and cellular homeostasis. Non-coding RNAs (ncRNAs) are heterogeneous transcripts that do not possess protein-coding functions, but they can act as effective post-transcriptional and epigenetic regulators of gene and protein expression, thus participating in numerous fundamental biological processes. Increasing evidence suggests that ncRNAs, autophagy, and their crosstalk play crucial roles in OA pathogenesis. Therefore, we summarized the complex role of autophagy in OA chondrocytes and focused on the regulatory role of ncRNAs in OA-associated autophagy to elucidate the complex pathological mechanisms of the ncRNA-autophagy network in the development of OA, thus providing new research targets for the clinical diagnosis and treatment of OA.


Sujet(s)
Autophagie , Chondrocytes , Arthrose , ARN non traduit , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Humains , Autophagie/physiologie , Autophagie/génétique , ARN non traduit/génétique , Animaux
13.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38981683

RÉSUMÉ

Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Likely owing to the unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific ER proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the easily expandable cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.


Sujet(s)
Collagène de type II , Réticulum endoplasmique , Procollagène , Réponse aux protéines mal repliées , Réticulum endoplasmique/métabolisme , Humains , Procollagène/métabolisme , Collagène de type II/métabolisme , Mutation , Cellules souches pluripotentes induites/métabolisme , Cartilage/métabolisme , Cartilage/anatomopathologie , Pliage des protéines , Arthrite/métabolisme , Arthrite/génétique , Arthrose/métabolisme , Arthrose/génétique , Arthrose/anatomopathologie , Animaux , Chondrocytes/métabolisme
14.
Sci Rep ; 14(1): 15022, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951570

RÉSUMÉ

Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/ß-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-ß3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-ß3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Chondrogenèse , Hydrogels , Cellules souches mésenchymateuses , Ingénierie tissulaire , Protéine Wnt3A , Protéine Wnt3A/métabolisme , Chondrogenèse/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Humains , Cartilage/métabolisme , Gélatine/composition chimique , Structures d'échafaudage tissulaires/composition chimique , Facteur de croissance transformant bêta-3/métabolisme , Facteur de croissance transformant bêta-3/pharmacologie , Lignée cellulaire , Matrice extracellulaire/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/cytologie , Animaux
15.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38982592

RÉSUMÉ

Chondrocyte apoptosis is recognized as one of the pathological features involved in cartilage degeneration driving the onset and progression of knee osteoarthritis (OA). This study aimed to determine the molecular mechanism underlying the effect of clusterin (CLU), anti-apoptotic molecule, in human knee OA chondrocytes. Primary knee OA chondrocytes were isolated from the cartilage of knee OA patients and divided into five groups: (1) the cells treated with interleukin (IL)-1ß, (2) CLU alone, (3) a combination of IL-1ß and CLU, (4) LY294002 (PI3K inhibitor) along with IL-1ß and CLU, and (5) the untreated cells. Production of apoptotic, inflammatory, anabolic, and catabolic mediators in knee OA chondrocytes was determined after treatment for 24 h. Our in vitro study uncovered that CLU significantly suppressed the production of inflammatory mediators [nitric oxide (NO), IL6, and tumor necrosis factor (TNF)-α] and apoptotic molecule (caspase-3, CASP3). CLU significantly upregulated messenger ribonucleic acid (mRNA) expressions of anabolic factors [SRY-box transcription factor-9 (SOX9) and aggrecan (ACAN)], but significantly downregulated mRNA expressions of IL6, nuclear factor kappa-B (NF-κB), CASP3, and matrix metalloproteinase-13 (MMP13). Anti-apoptotic and anti-inflammatory effects of CLU were mediated through activating PI3K/Akt signaling pathway. The findings suggest that CLU might have beneficial effects on knee OA chondrocytes by exerting anti-apoptotic and anti-inflammatory functions via PI3K/Akt pathway, making CLU a promising target for potential therapeutic interventions in knee OA.


Sujet(s)
Apoptose , Chondrocytes , Clusterine , Interleukine-1 bêta , Gonarthrose , Humains , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Gonarthrose/anatomopathologie , Gonarthrose/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Clusterine/métabolisme , Clusterine/génétique , Interleukine-1 bêta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées , Mâle , Adulte d'âge moyen , Sujet âgé , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Femelle , Phosphatidylinositol 3-kinases/métabolisme , Morpholines/pharmacologie , 4H-1-Benzopyran-4-ones/pharmacologie , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Matrix Metalloproteinase 13/métabolisme , Médiateurs de l'inflammation/métabolisme , Monoxyde d'azote/métabolisme
16.
Cell Commun Signal ; 22(1): 366, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026271

RÉSUMÉ

BACKGROUND: Z-DNA binding protein 1 (ZBP1) is a nucleic acid sensor that is involved in multiple inflammatory diseases, but whether and how it contributes to osteoarthritis (OA) are unclear. METHODS: Cartilage tissues were harvested from patients with OA and a murine model of OA to evaluate ZBP1 expression. Subsequently, the functional role and mechanism of ZBP1 were examined in primary chondrocytes, and the role of ZBP1 in OA was explored in mouse models. RESULTS: We showed the upregulation of ZBP1 in articular cartilage originating from OA patients and mice with OA after destabilization of the medial meniscus (DMM) surgery. Specifically, knockdown of ZBP1 alleviated chondrocyte damage and protected mice from DMM-induced OA. Mechanistically, tumor necrosis factor alpha induced ZBP1 overexpression in an interferon regulatory factor 1 (IRF1)-dependent manner and elicited the activation of ZBP1 via mitochondrial DNA (mtDNA) release and ZBP1 binding. The upregulated and activated ZBP1 could interact with receptor-interacting protein kinase 1 and activate the transforming growth factor-beta-activated kinase 1-NF-κB signaling pathway, which led to chondrocyte inflammation and extracellular matrix degradation. Moreover, inhibition of the mtDNA-IRF1-ZBP1 axis with Cyclosporine A, a blocker of mtDNA release, could delay the progression of DMM-induced OA. CONCLUSIONS: Our data revealed the pathological role of the mtDNA-IRF1-ZBP1 axis in OA chondrocytes, suggesting that inhibition of this axis could be a viable therapeutic approach for OA.


Sujet(s)
Chondrocytes , ADN mitochondrial , Facteur-1 de régulation d'interféron , Arthrose , Protéines de liaison à l'ARN , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Animaux , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrose/génétique , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , Souris , Mâle , Souris de lignée C57BL , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Transduction du signal , Modèles animaux de maladie humaine
17.
Autoimmunity ; 57(1): 2361749, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39007896

RÉSUMÉ

BACKGROUND: Dysregulated circular RNAs (circRNAs) are involved in osteoarthritis (OA) progression. OBJECTIVE: We aimed to explore the effect of hsa_circ_0044719 (circTRIM25) on the ferroptosis of chondrocytes. METHODS: Chondrocytes were treated with interleukin (IL)-1ß to generate cell model. Cellular behaviours were measured using cell counting kit-8, enzyme-linked immunosorbent assay, relevant kits, propidium iodide staining, and immunofluorescence assay. Quantitative real-time polymerase chain reaction was performed to examine the expression of circTRIM25, miR-138-5p, and cAMP responsive element binding protein 1 (CREB1), and their interactions were assessed using luciferase reporter analysis and RNA pull-down assay. RESULTS: CircTRIM25 was upregulated in OA tissues and IL-1ß-stimulated chondrocytes. Knockdown of circTRIM25 facilitated the viability and suppressed ferroptosis and inflammation of IL-1ß-induced cells. CircTRIM25 served as a sponge of miR-138-5p, which directly targets CREB1. Downregulation of miR-138-5p abrogated the effect induced by knockdown of circTRIM25. Furthermore, enforced CREB1 reversed the miR-138-5p induced effect. Moreover, knockdown of circTRIM25 attenuated cartilage injury in vivo. CONCLUSION: Silencing of circTRIM25 inhibited ferroptosis of chondrocytes via the miR-138-5p/CREB axis and thus attenuated OA progression.


Sujet(s)
Chondrocytes , Chondrogenèse , Protéine de liaison à l'élément de réponse à l'AMP cyclique , microARN , Arthrose , ARN circulaire , Animaux , Femelle , Humains , Mâle , Souris , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Chondrogenèse/génétique , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/génétique , Régulation de l'expression des gènes , Extinction de l'expression des gènes , Interleukine-1 bêta/métabolisme , microARN/génétique , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , ARN circulaire/génétique , Transduction du signal , Adulte d'âge moyen , Sujet âgé
18.
Bone Res ; 12(1): 41, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39019845

RÉSUMÉ

Mechanical overloading and aging are two essential factors for osteoarthritis (OA) development. Mitochondria have been identified as a mechano-transducer situated between extracellular mechanical signals and chondrocyte biology, but their roles and the associated mechanisms in mechanical stress-associated chondrocyte senescence and OA have not been elucidated. Herein, we found that PDZ domain containing 1 (PDZK1), one of the PDZ proteins, which belongs to the Na+/H+ Exchanger (NHE) regulatory factor family, is a key factor in biomechanically induced mitochondrial dysfunction and chondrocyte senescence during OA progression. PDZK1 is reduced by mechanical overload, and is diminished in the articular cartilage of OA patients, aged mice and OA mice. Pdzk1 knockout in chondrocytes exacerbates mechanical overload-induced cartilage degeneration, whereas intraarticular injection of adeno-associated virus-expressing PDZK1 had a therapeutic effect. Moreover, PDZK1 loss impaired chondrocyte mitochondrial function with accumulated damaged mitochondria, decreased mitochondrion DNA (mtDNA) content and increased reactive oxygen species (ROS) production. PDZK1 supplementation or mitoubiquinone (MitoQ) application alleviated chondrocyte senescence and cartilage degeneration and significantly protected chondrocyte mitochondrial functions. MRNA sequencing in articular cartilage from Pdzk1 knockout mice and controls showed that PDZK1 deficiency in chondrocytes interfered with mitochondrial function through inhibiting Hmgcs2 by increasing its ubiquitination. Our results suggested that PDZK1 deficiency plays a crucial role in mediating excessive mechanical load-induced chondrocyte senescence and is associated with mitochondrial dysfunction. PDZK1 overexpression or preservation of mitochondrial functions by MitoQ might present a new therapeutic approach for mechanical overload-induced OA.


Sujet(s)
Vieillissement de la cellule , Chondrocytes , Souris knockout , Mitochondries , Arthrose , Animaux , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrose/génétique , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Humains , Souris , Mâle , Souris de lignée C57BL , Contrainte mécanique , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Espèces réactives de l'oxygène/métabolisme
19.
BMC Musculoskelet Disord ; 25(1): 565, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033138

RÉSUMÉ

INTRODUCTION: Growth plate damage in long bones often results in progressive skeletal growth imbalance and deformity, leading to significant physical problems. Gangliosides, key glycosphingolipids in cartilage, are notably abundant in articular cartilage and regulate chondrocyte homeostasis. This suggests their significant roles in regulating growth plate cartilage repair. METHODS: Chondrocytes from 3 to 5 day-old C57BL/6 mice underwent glycoblotting and mass spectrometry. Based on the results of the glycoblotting analysis, we employed GD3 synthase knockout mice (GD3-/-), which lack b-series gangliosides. In 3-week-old mice, physeal injuries were induced in the left tibiae, with right tibiae sham operated. Tibiae were analyzed at 5 weeks postoperatively for length and micro-CT for growth plate height and bone volume at injury sites. Tibial shortening ratio and bone mineral density were measured by micro-CT. RESULTS: Glycoblotting analysis indicated that b-series gangliosides were the most prevalent in physeal chondrocytes among ganglioside series. At 3 weeks, GD3-/- exhibited reduced tibial shortening (14.7 ± 0.2 mm) compared to WT (15.0 ± 0.1 mm, P = 0.03). By 5 weeks, the tibial lengths in GD3-/- (16.0 ± 0.4 mm) closely aligned with sham-operated lengths (P = 0.70). Micro-CT showed delayed physeal bridge formation in GD3-/-, with bone volume measuring 168.9 ± 5.8 HU at 3 weeks (WT: 180.2 ± 3.2 HU, P = 0.09), but normalizing by 5 weeks. CONCLUSION: This study highlights that GD3 synthase knockout mice inhibit physeal bridge formation after growth plate injury, proposing a new non-invasive approach for treating skeletal growth disorders.


Sujet(s)
Chondrocytes , Gangliosides , Lame épiphysaire , Souris de lignée C57BL , Souris knockout , Animaux , Lame épiphysaire/anatomopathologie , Lame épiphysaire/métabolisme , Gangliosides/métabolisme , Chondrocytes/métabolisme , Souris , Inégalité de longueur des membres inférieurs , Tibia/imagerie diagnostique , Tibia/anatomopathologie , Tibia/métabolisme , Tibia/croissance et développement , Microtomographie aux rayons X , Sialyltransferases/déficit , Sialyltransferases/génétique , Sialyltransferases/métabolisme , Modèles animaux de maladie humaine
20.
Autoimmunity ; 57(1): 2364686, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38946534

RÉSUMÉ

BACKGROUND: Chondrocyte viability, apoptosis, and migration are closely related to cartilage injury in osteoarthritis (OA) joints. Exosomes are identified as potential therapeutic agents for OA. OBJECTIVE: This study aimed to investigate the role of exosomes derived from osteocytes in OA, particularly focusing on their effects on cartilage repair and molecular mechanisms. METHODS: An injury cell model was established by treating chondrocytes with IL-1ß. Cartilage repair was evaluated using cell counting kit-8, flow cytometry, scratch test, and Western Blot. Molecular mechanisms were analyzed using quantitative real-time PCR, bioinformatic analysis, and Western Blot. An OA mouse model was established to explore the role of exosomal DLX2 in vivo. RESULTS: Osteocyte-released exosomes promoted cell viability and migration, and inhibited apoptosis and extracellular matrix (ECM) deposition. Moreover, exosomes upregulated DLX2 expression, and knockdown of DLX2 activated the Wnt pathway. Additionally, exosomes attenuated OA in mice by transmitting DLX2. CONCLUSION: Osteocyte-derived exosomal DLX2 alleviated IL-1ß-induced cartilage repair and inactivated the Wnt pathway, thereby alleviating OA progression. The findings suggested that osteocyte-derived exosomes may hold promise as a treatment for OA.


Sujet(s)
Chondrocytes , Exosomes , Protéines à homéodomaine , Arthrose , Voie de signalisation Wnt , Animaux , Humains , Mâle , Souris , Apoptose , Cartilage/métabolisme , Cartilage/anatomopathologie , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Mouvement cellulaire , Survie cellulaire , Chondrocytes/métabolisme , Modèles animaux de maladie humaine , Exosomes/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Interleukine-1 bêta/métabolisme , Arthrose/métabolisme , Arthrose/anatomopathologie , Ostéocytes/métabolisme , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE