Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.103
Filtrer
1.
Clin Transl Med ; 14(10): e70029, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39350476

RÉSUMÉ

INTRODUCTION: Temporomandibular joint osteoarthritis (TMJ OA) is a major disease that affects maxillofacial health and is characterised by cartilage degeneration and subchondral bone remodelling. Obesity is associated with the exacerbation of pathological manifestations of TMJ OA. However, the underlying mechanism between adipose tissue and the TMJ axis remains limited. OBJECTIVES: To evaluate the effects of obesity and the adipose tissue on the development of TMJ OA. METHODS: The obesity-related metabolic changes in TMJ OA patients were detected by physical signs and plasma metabolites. The effects of adipose tissue-derived EVs (Ad-EVs) on TMJ OA was investigated through histological and cytological experiments as well as gene editing technology. Alterations of Ad-EVs in obese state were identified by microRNA-seq analysis and the mechanism by which EVs affect TMJ OA was explored in vitro and in vivo. RESULTS: Obesity and the related metabolic changes were important influencing factors for TMJ OA. Ad-EVs from obese mice induced marked chondrocyte apoptosis, cartilage matrix degradation and subchondral bone remodelling, which exacerbated the development of TMJ OA. Depletion of Ad-EVs secretion by knocking out the geranylgeranyl diphosphate synthase (Ggpps) gene in adipose tissue significantly inhibited the obesity-induced aggravation of TMJ OA. MiR-3074-5p played an important role in this process . CONCLUSIONS: Our work unveils an unknown link between obese adipose tissue and TMJ OA. Targeting the Ad-EVs and the miR-3074-5p may represent a promising therapeutic strategy for obesity-related TMJ OA. KEY POINTS: High-fat-diet-induced obesity aggravate the progression of TMJ OA in mice. Obese adipose tissue participates in cartilage damage through the altered miRNA in extracellular vesicles. Inhibition of miR-3074-5p/SMAD4 pathway in chondrocyte alleviated the effect of HFD-EVs on TMJ OA.


Sujet(s)
Tissu adipeux , Vésicules extracellulaires , Obésité , Arthrose , Vésicules extracellulaires/métabolisme , Animaux , Arthrose/métabolisme , Arthrose/étiologie , Obésité/métabolisme , Obésité/complications , Souris , Tissu adipeux/métabolisme , Humains , Mâle , Femelle , Articulation temporomandibulaire/métabolisme , Articulation temporomandibulaire/anatomopathologie , Troubles de l'articulation temporomandibulaire/métabolisme , microARN/génétique , microARN/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine
3.
BMC Musculoskelet Disord ; 25(1): 769, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39354427

RÉSUMÉ

BACKGROUND: Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by chronic inflammation and progressive cartilage degradation, ultimately leading to joint dysfunction and disability. Oleocanthal (OC), a bioactive phenolic compound derived from extra virgin olive oil, has garnered significant attention due to its potent anti-inflammatory properties, which are comparable to those of non-steroidal anti-inflammatory drugs (NSAIDs). This study pioneers the investigation into the effects of OC on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway in OA, aiming to validate its efficacy as a functional food-based therapeutic intervention. METHODS: To simulate cartilage tissue in vitro, human bone marrow-derived mesenchymal stem cells (BMSCs) were differentiated into chondrocytes. An inflammatory OA-like environment was induced in these chondrocytes using lipopolysaccharide (LPS) to mimic the pathological conditions of OA. The therapeutic effects of OC were evaluated by treating these inflamed chondrocytes with various concentrations of OC. The study focused on assessing key inflammatory markers, catabolic enzymes, and mitochondrial function to elucidate the protective mechanisms of OC. Mitochondrial function, specifically mitochondrial membrane potential (ΔΨm), was assessed using Rhodamine 123 staining, a fluorescent dye that selectively accumulates in active mitochondria. The integrity of ΔΨm serves as an indicator of mitochondrial and bioenergetic function. Additionally, Western blotting was employed to analyze protein expression levels, while real-time polymerase chain reaction (RT-PCR) was used to quantify gene expression of inflammatory cytokines and catabolic enzymes. Flow cytometry was utilized to measure cell viability and apoptosis, providing a comprehensive evaluation of OC's therapeutic effects on chondrocytes. RESULTS: The results demonstrated that OC significantly downregulated PAR-2 expression in a dose-dependent manner, leading to a substantial reduction in pro-inflammatory cytokines, including TNF-α, IL-1ß, and MCP-1. Furthermore, OC attenuated the expression of catabolic markers such as SOX4 and ADAMTS5, which are critically involved in cartilage matrix degradation. Importantly, OC was found to preserve mitochondrial membrane potential (ΔΨm) in chondrocytes subjected to inflammatory stress, as evidenced by Rhodamine 123 staining, indicating a protective effect on cellular bioenergetics. Additionally, OC modulated the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)/Receptor Activator of Nuclear Factor Kappa-Β (RANK) pathway, suggesting a broader therapeutic action against the multifactorial pathogenesis of OA. CONCLUSIONS: This study is the first to elucidate the modulatory effects of OC on the PAR-2 mediated inflammatory pathway in OA, revealing its potential as a multifaceted therapeutic agent that not only mitigates inflammation but also protects cartilage integrity. The preservation of mitochondrial function and modulation of the RANKL/RANK pathway further underscores OC's comprehensive therapeutic potential in counteracting the complex pathogenesis of OA. These findings position OC as a promising candidate for integration into nutritional interventions aimed at managing OA. However, further research is warranted to fully explore OC's therapeutic potential across different stages of OA and its long-term effects in musculoskeletal disorders.


Sujet(s)
Anti-inflammatoires , Chondrocytes , Cyclopentane monoterpenes , Cellules souches mésenchymateuses , Arthrose , Récepteur de type PAR-2 , Humains , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Récepteur de type PAR-2/métabolisme , Anti-inflammatoires/pharmacologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cyclopentane monoterpenes/pharmacologie , Cellules cultivées , Aliment fonctionnel , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Lipopolysaccharides/pharmacologie , Aldéhydes , Phénols
4.
J Pineal Res ; 76(6): e12992, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39228264

RÉSUMÉ

Recent evidence indicates that the damaged regions in osteoarthritis are accompanied by the accumulation of iron ions. Ferroptosis, as an iron-dependent form of cell death, holds significant implications in osteoarthritis. Melatonin, a natural product with strong scavenging abilities against reactive oxygen species and lipid peroxidation, plays a crucial role in the treatment of osteoarthritis. This study aims to demonstrate the existence of ferroptosis in osteoarthritis and explore the specific mechanism of melatonin in suppressing ferroptosis and alleviating osteoarthritis. Our findings reveal that melatonin reverses inflammation-induced oxidative stress and lipid peroxidation while promoting the expression of extracellular matrix components in chondrocytes, safeguarding the cells. Our research has revealed that NADPH oxidase 4 (NOX4) serves as a crucial molecule in the ferroptosis process of osteoarthritis. Specifically, NOX4 is located on mitochondria in chondrocytes, which can induce disorders in mitochondrial energy metabolism and dysfunction, thereby intensifying oxidative stress and lipid peroxidation. LC-MS analysis further uncovered that GRP78 is a downstream binding protein of NOX4. NOX4 induces ferroptosis by weakening GRP78's protective effect on GPX4 and reducing its expression. Melatonin can inhibit the upregulation of NOX4 on mitochondria and mitigate mitochondrial dysfunction, effectively suppressing ferroptosis and alleviating osteoarthritis. This suggests that melatonin therapy represents a promising new approach for the treatment of osteoarthritis.


Sujet(s)
Ferroptose , Mélatonine , Mitochondries , NADPH Oxidase 4 , Arthrose , Mélatonine/pharmacologie , Ferroptose/effets des médicaments et des substances chimiques , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , NADPH Oxidase 4/métabolisme , Animaux , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Humains , Souris
5.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39273098

RÉSUMÉ

Osteoarthritis (OA) is a leading cause of pain and disability worldwide in elderly people. There is a critical need to develop novel therapeutic strategies that can effectively manage pain and disability to improve the quality of life for older people. Mesenchymal stem cells (MSCs) have emerged as a promising cell-based therapy for age-related disorders due to their multilineage differentiation and strong paracrine effects. Notably, MSC-derived exosomes (MSC-Exos) have gained significant attention because they can recapitulate MSCs into therapeutic benefits without causing any associated risks compared with direct cell transplantation. These exosomes help in the transport of bioactive molecules such as proteins, lipids, and nucleic acids, which can influence various cellular processes related to tissue repair, regeneration, and immune regulation. In this review, we have provided an overview of MSC-Exos as a considerable treatment option for osteoarthritis. This review will go over the underlying mechanisms by which MSC-Exos may alleviate the pathological hallmarks of OA, such as cartilage degradation, synovial inflammation, and subchondral bone changes. Furthermore, we have summarized the current preclinical evidence and highlighted promising results from in vitro and in vivo studies, as well as progress in clinical trials using MSC-Exos to treat OA.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , Arthrose , Exosomes/métabolisme , Exosomes/transplantation , Humains , Arthrose/thérapie , Arthrose/métabolisme , Arthrose/anatomopathologie , Cellules souches mésenchymateuses/métabolisme , Animaux , Transplantation de cellules souches mésenchymateuses/méthodes
6.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39273304

RÉSUMÉ

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


Sujet(s)
Cannabinoïdes , Membrane synoviale , Humains , Cannabinoïdes/usage thérapeutique , Cannabinoïdes/pharmacologie , Cannabinoïdes/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/effets des médicaments et des substances chimiques , Animaux , Endocannabinoïdes/métabolisme , Rhumatismes/traitement médicamenteux , Rhumatismes/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie
7.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39273346

RÉSUMÉ

Articular cartilage receives nutrients and oxygen from the synovial fluid to maintain homeostasis. However, compared to tissues with abundant blood flow, articular cartilage is exposed to a hypoxic environment (i.e., physioxia) and has an enhanced hypoxic stress response. Hypoxia-inducible factors (HIFs) play a pivotal role in this physioxic environment. In normoxic conditions, HIFs are downregulated, whereas in physioxic conditions, they are upregulated. The HIF-α family comprises three members: HIF-1α, HIF-2α, and HIF-3α. Each member has a distinct function in articular cartilage. In osteoarthritis, which is primarily caused by degeneration of articular cartilage, HIF-1α is upregulated in chondrocytes and is believed to protect articular cartilage by acting anabolically on it. Conversely, in contrast to HIF-1α, HIF-2α exerts a catabolic influence on articular cartilage. It may therefore be possible to develop a new treatment for OA by controlling the expression of HIF-1α and HIF-2α with drugs or by altering the oxygen environment in the joints.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Cartilage articulaire , Chondrocytes , Homéostasie , Sous-unité alpha du facteur-1 induit par l'hypoxie , Arthrose , Humains , Cartilage articulaire/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Arthrose/métabolisme , Chondrocytes/métabolisme , Oxygène/métabolisme , Hypoxie/métabolisme , Hypoxie/physiopathologie
8.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39273553

RÉSUMÉ

Osteoarthritis (OA) is an age-related disease characterized by inflammation, pain, articular cartilage damage, synovitis, and irreversible disability. Gynostemma pentaphyllum (Thunb.) Makino (GP), a herbal medicine traditionally used in East Asia for its anti-inflammatory properties, was investigated for its potential to modulate OA pathology and symptoms. This study evaluated GP's efficacy in inhibiting pain, functional decline, and cartilage destruction in monosodium iodoacetate-induced OA and acetic acid-induced writhing models. Additionally, the effects of GP on OA-related inflammatory targets were assessed via mRNA and protein expression in rat knee cartilage and lipopolysaccharide-induced RAW 264.7 cells. The GP group demonstrated significant pain relief, functional improvement, and cartilage protection. Notably, GP inhibited key inflammatory mediators, including interleukin (IL)-1ß, IL-6, matrix metalloproteinases (MMP)-3 and MMP-13, cyclooxygenase-2, and prostaglandin E receptor 2, surpassing the effects of active controls. These findings suggest that GP is a promising candidate for disease-modifying OA drugs and warrants further comprehensive studies.


Sujet(s)
Analgésiques , Anti-inflammatoires , Gynostemma , Arthrose , Extraits de plantes , Animaux , Gynostemma/composition chimique , Souris , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , Arthrose/induit chimiquement , Arthrose/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Cellules RAW 264.7 , Rats , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique , Analgésiques/pharmacologie , Analgésiques/usage thérapeutique , Mâle , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Modèles animaux de maladie humaine , Rat Sprague-Dawley , Douleur/traitement médicamenteux
9.
FASEB J ; 38(17): e70013, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39225365

RÉSUMÉ

Articular cartilage phenotypic homeostasis is crucial for life-long joint function, but the underlying cellular and molecular mechanisms governing chondrocyte stability remain poorly understood. Here, we show that the protein tyrosine phosphatase SHP2 is differentially expressed in articular cartilage (AC) and growth plate cartilage (GPC) and that it negatively regulates cell proliferation and cartilage phenotypic program. Postnatal SHP2 deletion in Prg4+ AC chondrocytes increased articular cellularity and thickness, whereas SHP2 deletion in Acan+ pan-chondrocytes caused excessive GPC chondrocyte proliferation and led to joint malformation post-puberty. These observations were verified in mice and in cultured chondrocytes following treatment with the SHP2 PROTAC inhibitor SHP2D26. Further mechanistic studies indicated that SHP2 negatively regulates SOX9 stability and transcriptional activity by influencing SOX9 phosphorylation and promoting its proteasome degradation. In contrast to published work, SHP2 ablation in chondrocytes did not impact IL-1-evoked inflammation responses, and SHP2's negative regulation of SOX9 could be curtailed by genetic or chemical SHP2 inhibition, suggesting that manipulating SHP2 signaling has translational potential for diseases of cartilage dyshomeostasis.


Sujet(s)
Cartilage articulaire , Chondrocytes , Arthrose , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Facteur de transcription SOX-9 , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Souris , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Arthrose/métabolisme , Arthrose/anatomopathologie , Prolifération cellulaire , Cellules cultivées , Souris de lignée C57BL , Souris knockout , Mâle
10.
PLoS One ; 19(9): e0309807, 2024.
Article de Anglais | MEDLINE | ID: mdl-39236007

RÉSUMÉ

This study explored the mechanism of curcumin (CUR) suppressing osteoclastogenesis and evaluated its effects on osteoarthritis (OA) mouse. Bone marrow-derived macrophages were isolated as osteoclast precursors. In the presence or absence of CUR, cell proliferation was detected by CCK-8, osteoclastogenesis was detected by tartrate-resistant acid phosphatase (TRAP) staining, F-actin rings formation was detected by immunofluorescence, bone resorption was detected by bone slices, IκBα, nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways were detected using western blot, osteoclastogenesis-related gens were measured using quantitative polymerase chain reaction. A knee OA mouse model was designed by destabilizing the medial meniscus (DMM). Thirty-six male mice were divided into sham+vehicle, OA+vehicle, and OA+CUR groups. Mice were administered with or without CUR at 25 mg/kg/d from the first post-operative day until sacrifice. After 4 and 8 weeks of OA induction, micro-computed tomography was performed to analyze microstructure changes in subchondral bone, hematoxylin and eosin staining was performed to calculate the thickness of the calcified and hyaline cartilage layers, toluidine blue O staining was performed to assess the degenerated cartilage, TRAP-stained osteoclasts were counted, and NF-κB, phosphorylated Jun N-terminal Kinases (p-JNK), and receptor activator of nuclear factor κB ligand (RANKL) were detected using immunohistochemistry. CUR suppressed osteoclastogenesis and bone resorption without cytotoxicity. CUR restrained RANKL-induced activation of NF-κB, p-JNK and up-regulation of osteoclastogenesis-related genes. CUR delayed cartilage degeneration by suppressing osteoclastogenesis and bone resorption in early OA. The mechanism of CUR inhibiting osteoclastogenesis might be associated with NF-κB/JNK signaling pathway, indicating a novel strategy for OA treatment.


Sujet(s)
Curcumine , Système de signalisation des MAP kinases , Facteur de transcription NF-kappa B , Ostéoclastes , Ostéogenèse , Animaux , Souris , Mâle , Facteur de transcription NF-kappa B/métabolisme , Curcumine/pharmacologie , Ostéogenèse/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Résorption osseuse/traitement médicamenteux , Résorption osseuse/métabolisme , Résorption osseuse/anatomopathologie
11.
Chem Biol Drug Des ; 104(3): e14620, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39251394

RÉSUMÉ

Osteoarthritis (OA) is a progressive joint disease characterized by extracellular matrix (ECM) degradation and inflammation, which is involved with pathological microenvironmental alterations induced by damaged chondrocytes. However, current therapies are not effective in alleviating the progression of OA. Isoquercetin is a natural flavonoid glycoside compound that has various pharmacological effects including anticancer, anti-diabetes and blood lipid regulation. Previous evidence suggests that isoquercetin has anti-inflammatory properties in various diseases, but its effect on OA has not been investigated yet. In this study, through western bolt, qRT-PCR and ELISA, it was found that isoquercetin could reduce the increase of ADAMTS5, MMP13, COX-2, iNOS and IL-6 induced by IL-1ß, suggesting that isoquercetin could inhibit the inflammation and ECM degradation of chondrocytes. Through nuclear-plasma separation technique, western blot and immunocytochemistry, it can be found that Nrf2 and NF-κB pathways are activated in this process, and isoquercetin may rely on this process to play its protective role. In vivo, the results of X-ray and SO staining show that intra-articular injection of isoquercetin reduces the degradation of cartilage in the mouse OA model. In conclusion, the present work suggests that isoquercetin may benefit chondrocytes by regulating the Nrf2/NF-κB signaling axis, which supports isoquercetin as a potential drug for the treatment of OA.


Sujet(s)
Chondrocytes , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Arthrose , Quercétine , Transduction du signal , Animaux , Humains , Mâle , Souris , Protéine ADAMTS5/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Anti-inflammatoires/usage thérapeutique , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Cyclooxygenase 2/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-6/métabolisme , Matrix Metalloproteinase 13/métabolisme , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2/effets des médicaments et des substances chimiques , Facteur-2 apparenté à NF-E2/métabolisme , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Nitric oxide synthase type II/métabolisme , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Quercétine/pharmacologie , Quercétine/analogues et dérivés , Quercétine/composition chimique , Quercétine/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques
12.
Int J Rheum Dis ; 27(9): e15323, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39221886

RÉSUMÉ

BACKGROUND: Osteoarthritis (OA) is a prevalent degenerative disease. We explored the role and regulatory mechanisms of lncRNA-FAS-AS1 in OA progression. METHODS: We exposed human immortalized chondrocytes to IL-1ß for 24 h to induce an OA cell model. The target molecule levels were assessed using western blot and quantitative real-time PCR (RT-qPCR). Cell viability and apoptosis were measured using CCK-8 and flow cytometry. The m6A modification of FAS-AS1 was determined using MeRIP. We examined the binding relationships between FAS-AS1, Fragile X mental retardation 1 (FMR1), and A disintegrin and metalloproteinase 8 (ADAM8) using RIP and RNA pull-down. The OA animal model was established by separating the medial collateral ligament and medial meniscus. Safranin-O staining and Mankin's scale were employed to evaluate pathological changes within the cartilage. RESULTS: FAS-AS1, METTL14, and ADAM8 were upregulated, and the JAK/STAT3 signaling pathway was activated in OA mice and IL-1ß-induced chondrocytes. FAS-AS1 knockdown inhibited extracellular matrix degradation in IL-1ß-induced chondrocytes; however, ADAM8 overexpression reversed this effect. FAS-AS1 maintained the stability of ADAM8 mRNA by recruiting FMR1. METTL14 knockdown repressed FAS-AS1 expression in an m6A-dependent manner. FAS-AS1 overexpression reversed the inhibitory effects of METTL14 knockdown on JAK/STAT3 signaling and cartilage damage in the OA model both in vitro and in vivo. CONCLUSION: METTL14-mediated FAS-AS1 promotes OA progression through the FMR1/ADAM8/JAK/STAT3 axis.


Sujet(s)
Protéines ADAM , Chondrocytes , Évolution de la maladie , Protéines membranaires , ARN long non codant , Facteur de transcription STAT-3 , Transduction du signal , Régulation positive , Animaux , Humains , Mâle , Souris , Protéines ADAM/métabolisme , Protéines ADAM/génétique , Adénosine/analogues et dérivés , Apoptose , Arthrite expérimentale/métabolisme , Arthrite expérimentale/génétique , Arthrite expérimentale/anatomopathologie , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Lignée cellulaire , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Modèles animaux de maladie humaine , Interleukine-1 bêta/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Souris de lignée C57BL , Arthrose/métabolisme , Arthrose/génétique , Arthrose/anatomopathologie , Gonarthrose/métabolisme , Gonarthrose/génétique , Gonarthrose/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique
13.
Nat Commun ; 15(1): 7712, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39231947

RÉSUMÉ

Osteoarthritis is a degenerative joint disease with joint pain as the main symptom, caused by fibrosis and loss of articular cartilage. Due to the complexity and heterogeneity of osteoarthritis, there is a lack of effective individualized disease-modifying osteoarthritis drugs in clinical practice. Chondrocyte senescence is reported to participate in occurrence and progression of osteoarthritis. Here we show that small molecule 10-hydroxy-2-decenoic acid suppresses cartilage degeneration and relieves pain in the chondrocytes, cartilage explants from osteoarthritis patients, surgery-induced medial meniscus destabilization or naturally aged male mice. We further confirm that 10-hydroxy-2-decenoic acid exerts a protective effect by targeting the glycosylation site in the Asp_Arg_Hydrox domain of aspartyl ß-hydroxylase. Mechanistically, 10-hydroxy-2-decenoic acid alleviate cellular senescence through the ERK/p53/p21 and GSK3ß/p16 pathways in the chondrocytes. Our study uncovers that 10-hydroxy-2-decenoic acid modulate cartilage metabolism by targeting aspartyl ß-hydroxylase to inhibit chondrocyte senescence in osteoarthritis. 10-hydroxy-2-decenoic acid may be a promising therapeutic drug against osteoarthritis.


Sujet(s)
Cartilage articulaire , Vieillissement de la cellule , Chondrocytes , Acides gras monoinsaturés , Arthrose , Animaux , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Mâle , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/traitement médicamenteux , Arthrose/prévention et contrôle , Souris , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Humains , Acides gras monoinsaturés/pharmacologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Femelle
14.
Bone Res ; 12(1): 50, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39231936

RÉSUMÉ

Osteoarthritis (OA) is the most common form of arthritic disease, and phenotypic modification of chondrocytes is an important mechanism that contributes to the loss of cartilage homeostasis. This study identified that Fascin actin-bundling protein 1 (FSCN1) plays a pivotal role in regulating chondrocytes phenotype and maintaining cartilage homeostasis. Proteome-wide screening revealed markedly upregulated FSCN1 protein expression in human OA cartilage. FSCN1 accumulation was confirmed in the superficial layer of OA cartilage from humans and mice, primarily in dedifferentiated-like chondrocytes, associated with enhanced actin stress fiber formation and upregulated type I and III collagens. FSCN1-inducible knockout mice exhibited delayed cartilage degeneration following experimental OA surgery. Mechanistically, FSCN1 promoted actin polymerization and disrupted the inhibition of Decorin on TGF-ß1, leading to excessive TGF-ß1 production and ALK1/Smad1/5 signaling activation, thus, accelerated chondrocyte dedifferentiation. Intra-articular injection of FSCN1-overexpressing adeno-associated virus exacerbated OA progression in mice, which was mitigated by an ALK1 inhibitor. Moreover, FSCN1 inhibitor NP-G2-044 effectively reduced extracellular matrix degradation in OA mice, cultured human OA chondrocytes, and cartilage explants by suppressing ALK1/Smad1/5 signaling. These findings suggest that targeting FSCN1 represents a promising therapeutic approach for OA.


Sujet(s)
Protéines de transport , Chondrocytes , Protéines des microfilaments , Arthrose , Animaux , Humains , Mâle , Souris , Protéines de transport/métabolisme , Protéines de transport/génétique , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Souris de lignée C57BL , Souris knockout , Protéines des microfilaments/génétique , Protéines des microfilaments/métabolisme , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrose/génétique , Phénotype , Récepteurs olfactifs , Transduction du signal
15.
PeerJ ; 12: e17963, 2024.
Article de Anglais | MEDLINE | ID: mdl-39282111

RÉSUMÉ

The role of mitochondria in the pathogenesis of osteoarthritis (OA) is significant. In this study, we aimed to identify diagnostic signature genes associated with OA from a set of mitochondria-related genes (MRGs). First, the gene expression profiles of OA cartilage GSE114007 and GSE57218 were obtained from the Gene Expression Omnibus. And the limma method was used to detect differentially expressed genes (DEGs). Second, the biological functions of the DEGs in OA were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Wayne plots were employed to visualize the differentially expressed mitochondrial genes (MDEGs) in OA. Subsequently, the LASSO and SVM-RFE algorithms were employed to elucidate potential OA signature genes within the set of MDEGs. As a result, GRPEL and MTFP1 were identified as signature genes. Notably, GRPEL1 exhibited low expression levels in OA samples from both experimental and test group datasets, demonstrating high diagnostic efficacy. Furthermore, RT-qPCR analysis confirmed the reduced expression of Grpel1 in an in vitro OA model. Lastly, ssGSEA analysis revealed alterations in the infiltration abundance of several immune cells in OA cartilage tissue, which exhibited correlation with GRPEL1 expression. Altogether, this study has revealed that GRPEL1 functions as a novel and significant diagnostic indicator for OA by employing two machine learning methodologies. Furthermore, these findings provide fresh perspectives on potential targeted therapeutic interventions in the future.


Sujet(s)
Apprentissage machine , Arthrose , Humains , Arthrose/génétique , Arthrose/diagnostic , Arthrose/métabolisme , Marqueurs biologiques/métabolisme , Analyse de profil d'expression de gènes/méthodes , Mitochondries/génétique , Mitochondries/métabolisme , Algorithmes , Transcriptome/génétique
16.
Sci Adv ; 10(37): eadp4408, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39259800

RÉSUMÉ

The rapid advancement of cell therapies underscores the importance of understanding fundamental cellular attributes. Among these, cell fitness-how transplanted cells adapt to new microenvironments and maintain functional stability in vivo-is crucial. This study identifies a chemical compound, FPH2, that enhances the fitness of human chondrocytes and the repair of articular cartilage, which is typically nonregenerative. Through drug screening, FPH2 was shown to broadly improve cell performance, especially in maintaining chondrocyte phenotype and enhancing migration. Single-cell transcriptomics indicated that FPH2 induced a super-fit cell state. The mechanism primarily involves the inhibition of carnitine palmitoyl transferase I and the optimization of metabolic homeostasis. In animal models, FPH2-treated human chondrocytes substantially improved cartilage regeneration, demonstrating well-integrated tissue interfaces in rats. In addition, an acellular FPH2-loaded hydrogel proved effective in preventing the onset of osteoarthritis. This research provides a viable and safe method to enhance chondrocyte fitness, offering insights into the self-regulatory mechanisms of cell fitness.


Sujet(s)
Cartilage articulaire , Chondrocytes , Régénération , Chondrocytes/métabolisme , Chondrocytes/cytologie , Chondrocytes/effets des médicaments et des substances chimiques , Animaux , Humains , Cartilage articulaire/métabolisme , Rats , Arthrose/métabolisme , Arthrose/thérapie , Hydrogels/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques
17.
J Nanobiotechnology ; 22(1): 555, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261846

RÉSUMÉ

BACKGROUND: The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. METHODS: The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF-α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. RESULTS: The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF-α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF-α, thereby exerting chondroprotective effects. CONCLUSION: TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , Arthrose , Facteur de nécrose tumorale alpha , Exosomes/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Souris , Arthrose/thérapie , Arthrose/métabolisme , Tissu adipeux/cytologie , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Cartilage articulaire/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules cultivées , Humains
18.
Nano Lett ; 24(37): 11697-11705, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39225479

RÉSUMÉ

Osteoarthritis (OA) is a degenerative joint disease characterized by obscure etiology and unsatisfactory therapeutic outcomes, making the development of new efficient therapies urgent. Superfluous reactive oxygen species (ROS) have historically been considered one of the crucial factors inducing the pathological progression of OA. Ultrasmall Prussian blue nanoparticles (USPBNPs), approximately sub-5 nm in size, are developed by regulating the configuration of polyvinylpyrrolidone chains. USPBNPs display an excellent ROS eliminating capacity and catalase-like activity, capable of decomposing hydrogen peroxide (H2O2) into O2. The anti-inflammatory mechanism of USPBNPs can be attributed to repolarizing macrophages from pro-inflammatory M1 to anti-inflammatory M2 phenotype by decreasing the ROS levels accompanied by O2 improvement. Additionally, USPBNPs exhibit an exciting therapeutic efficiency against OA, comparable to that of hydrocortisone in vivo. This study not only develops a new therapeutic agent for OA but also offers an estimable insight into the application of the nanozyme.


Sujet(s)
Hexacyanoferrates II , Macrophages , Arthrose , Espèces réactives de l'oxygène , Hexacyanoferrates II/composition chimique , Hexacyanoferrates II/pharmacologie , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , Arthrose/métabolisme , Espèces réactives de l'oxygène/métabolisme , Animaux , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Nanoparticules/composition chimique , Peroxyde d'hydrogène/composition chimique , Peroxyde d'hydrogène/métabolisme , Humains , Cellules RAW 264.7 , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Phénotype , Taille de particule
19.
Front Biosci (Landmark Ed) ; 29(9): 320, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39344315

RÉSUMÉ

Osteoarthritis (OA) is a prevalent, chronic joint disorder affecting millions of people worldwide, characterized by articular cartilage degradation, subchondral bone remodeling, synovial cytokine secretion, and osteophyte formation. OA primarily affects the hips, knees, hands, and spine. Patients with OA exhibit a higher prevalence of cardiovascular comorbidities and potentially important associations between OA and cardiovascular diseases have prompted investigations into potentially similar pathophysiological associations. This review explores the coexistence of atherosclerotic peripheral vascular disease (ASPVD) in OA patients, including evidence from a contemporary study suggesting associations between OA and arterial wall thickness and blood flow changes which are characteristic of early atherosclerosis, and which stimulate reactive pathology in endothelial cells. Observations from this study demonstrate elevated arterial flow volume and increased intima-media thickness in arteries ipsilateral to OA knees, suggesting a potential link between OA and arterial wall disease. We further explore the intricate relationship between the vascular system and skeletal health, highlighting bidirectional interactions among endothelial cells, inflammatory cells, and various bone cells. Mechanical endothelial cell dysfunction is discussed, emphasizing the impact of vessel wall material changes and endothelial cell responses to alterations in fluid shear stress. Inflammatory changes in OA and ASPVD are also explored, showcasing shared pathophysiological processes involving immune cell infiltration and pro-inflammatory cytokines. Additionally, the role of hypofibrinolysis in OA and ASPVD is discussed, highlighting similarities in elevations of the hypercoagulative and hypofibrinolytic factor, plasminogen activator inhibitor (PAI-1). The review suggests a provocative relationship among low-grade chronic inflammation, endothelial dysfunction, and hypofibrinolytic states in OA and ASPVD, warranting further investigation. In conclusion, this review provides an exploration of the possible associations between OA and ASPVD. While the ongoing study's findings and other reports are observational, they suggest shared pathophysiological processes and emphasize the need for further research to elucidate additional potentially correlative linkages between these conditions. Understanding common molecular pathways may pave a way for targeted interventions that address both OA and ASPVD.


Sujet(s)
Arthrose , Humains , Arthrose/physiopathologie , Arthrose/métabolisme , Athérosclérose/physiopathologie , Cellules endothéliales/métabolisme
20.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39337321

RÉSUMÉ

The existing in vitro and in vivo models for studying osteoarthritis have significant limitations in replicating the complexity of joint tissues. This research aims to validate a Tissue-On-a-Chip system for osteoarthritis research. Osteochondral tissues obtained from knee replacement surgeries of patients with osteoarthritis were cultured in an Organ-On-a-Chip system. This system was designed to supply oxygen and glucose to the cartilage from the bone. The distribution of oxygen and glucose was evaluated by fluorescence using Image-iT Green Hypoxia and 2-NBDG, respectively. Cytotoxicity was measured using lactate dehydrogenase (LDH) levels in chip cultures compared to plate cultures (12 tissues per method). Glycosaminoglycans (GAGs), alkaline phosphatase (ALP), Coll2-1, and procollagen type II N-terminal propeptide (PIINP) were measured in the perfused medium of the Tissue-On-a-Chip over a period of 70 days. Fluorescence of Image-iT Green Hypoxia was observed only in the cartilage area, while 2-NBDG was distributed throughout the tissue. An increase in LDH levels was noted in the plate cultures on day 24 and in the Tissue-On-a-Chip cultures on day 63. Compared to the start of the culture, GAG content increased on day 52, while ALP showed variations. A notable increase in GAG, ALP, and Coll2-1 levels was observed on day 59. PIINP levels remained stable throughout the experiment. The validated osteochondral Tissue-On-a-Chip system can replicate the joint microenvironment, with hypoxic conditions in cartilage and normoxic conditions in bone. Tissue survival and component stability were maintained for approximately two months. This platform is a useful tool for evaluating new drugs and represents a viable alternative to animal models.


Sujet(s)
Laboratoires sur puces , Arthrose , Humains , Arthrose/métabolisme , Arthrose/anatomopathologie , Glycosaminoglycanes/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Oxygène/métabolisme , Glucose/métabolisme , L-Lactate dehydrogenase/métabolisme , Phosphatase alcaline/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE