Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.179
Filtrer
1.
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 , Ostéocytes , Facteurs de transcription , Voie de signalisation Wnt , Exosomes/métabolisme , Animaux , Arthrose/métabolisme , Arthrose/anatomopathologie , Souris , Facteurs de transcription/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Ostéocytes/métabolisme , Chondrocytes/métabolisme , Modèles animaux de maladie humaine , Humains , Interleukine-1 bêta/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Apoptose , Cartilage/métabolisme , Cartilage/anatomopathologie , Mâle , Mouvement cellulaire , Survie cellulaire
2.
Commun Biol ; 7(1): 828, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38972919

RÉSUMÉ

Crystallization of monosodium urate monohydrate (MSU) leads to painful gouty arthritis. Despite extensive research it is still unknown how this pathological biomineralization occurs, which hampers its prevention. Here we show how inflammatory MSU crystals form after a non-inflammatory amorphous precursor (AMSU) that nucleates heterogeneously on collagen fibrils from damaged articular cartilage of gout patients. This non-classical crystallization route imprints a nanogranular structure to biogenic acicular MSU crystals, which have smaller unit cell volume, lower microstrain, and higher crystallinity than synthetic MSU. These distinctive biosignatures are consistent with the template-promoted crystallization of biotic MSU crystals after AMSU at low supersaturation, and their slow growth over long periods of time (possibly years) in hyperuricemic gout patients. Our results help to better understand gout pathophysiology, underline the role of cartilage damage in promoting MSU crystallization, and suggest that there is a time-window to treat potential gouty patients before a critical amount of MSU has slowly formed as to trigger a gout flare.


Sujet(s)
Cristallisation , Goutte , Acide urique , Acide urique/métabolisme , Humains , Goutte/métabolisme , Goutte/anatomopathologie , Biominéralisation , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Goutte articulaire/métabolisme , Goutte articulaire/anatomopathologie
3.
Anal Cell Pathol (Amst) ; 2024: 1083143, 2024.
Article de Anglais | MEDLINE | ID: mdl-38946863

RÉSUMÉ

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


Sujet(s)
Chondrocytes , Chondrogenèse , Vésicules extracellulaires , Flavonoïdes , Cellules souches mésenchymateuses , Membrane synoviale , Voie de signalisation Wnt , Animaux , Lapins , Flavonoïdes/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Vésicules extracellulaires/métabolisme , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Membrane synoviale/cytologie , Chondrogenèse/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/effets des médicaments et des substances chimiques
4.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38928059

RÉSUMÉ

Worldwide, osteoarthritis (OA) is the most common cause of joint pain in older people. Many factors contribute to osteoarthritis' development and progression, including secondary osteoarthritis' underlying causes. It is important to note that osteoarthritis affects all four tissues: cartilage, bone, joint capsule, and articular apparatus. An increasingly prominent area of research in osteoarthritis regulation is microRNAs (miRNAs), a small, single-stranded RNA molecule that controls gene expression in eukaryotes. We aimed to assess and summarize current knowledge about the mechanisms of the action of miRNAs and their clinical significance. Osteoarthritis (OA) is affected by the interaction between miRNAs and inflammatory processes, as well as cartilage metabolism. MiRNAs also influence cartilage cell apoptosis, contributing to the degradation of the cartilage in OA. Studies have shown that miRNAs may have both an inhibitory and promoting effect on osteoporosis progression through their influence on molecular mechanisms. By identifying these regulators, targeted treatments for osteoarthritis may be developed. In addition, microRNA may also serve as a biomarker for osteoarthritis. By using these biomarkers, the disease could be detected faster, and early intervention can be instituted to prevent mobility loss and slow deterioration.


Sujet(s)
microARN , Arthrose , microARN/génétique , microARN/métabolisme , Humains , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Animaux , Régulation de l'expression des gènes , Marqueurs biologiques , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie
5.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38928219

RÉSUMÉ

Angiogenesis and vascular endothelial growth factor (VEGF) are involved in osteoarthritis (OA). We previously reported the inhibitory effect of bevacizumab in a rabbit model of OA. In the current study, we investigated the effects of lenvatinib, an angiogenesis inhibitor targeting the VEGF and fibroblast growth factor receptors, on synovitis, osteophyte formation, and cartilage degeneration in a rabbit OA model. Posttraumatic OA was induced by anterior cruciate ligament transection (ACLT) on one knee of each rabbit. Rabbits were placed into four groups according to the following lenvatinib doses: untreated control (n = 12), L0.3: 0.3 mg/kg/day (n = 15), L1.0: 1.0 mg/kg/day (n = 14), and L3.0: 3.0 mg/kg/day (n = 13) groups. We evaluated limb pain using the weight distribution ratio measured with an incapacitance tester, macroscopic osteophyte formation, and femoral condyle synovium and cartilage histology. For cartilage evaluation, the following distal sites of the femur were evaluated separately: femoral-tibial (FT), femoral-patellar (FP), and femoral corner (between FP and FT). The weight distribution ratio at 12 weeks after surgery was higher in the L0.3 and L1.0 groups than in the control group. Osteophyte formation and synovitis scores were significantly lower in the L0.3, L1.0, and L3.0 groups than in the control group. The Osteoarthritis Research Society International scores of the FT, corner, and FP sites in the L0.3 group were lower than in the control group. The cartilage thickness ratio at the FT and corner sites was significantly lower in the L0.3 group than in the control group. Krenn's grading system of cartilage synovitis showed that all lenvatinib-administered groups had significantly lower scores than the control group. MMP3 expression level in cartilage tissue was significantly lower in the L3.0 group compared with the other three groups. ADAMTS5 expression was lower in the L3.0 group compared with the control and L0.3 groups. Oral administration of lenvatinib inhibited synovitis, osteophyte formation, and cartilage degeneration and reduced pain in a rabbit ACLT model. Lenvatinib is an oral VEGF inhibitor that is easier to administer than other VEGF inhibitors and may have potential as a treatment of posttraumatic OA.


Sujet(s)
Gonarthrose , Phénylurées , Quinoléines , Animaux , Lapins , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Phénylurées/pharmacologie , Phénylurées/usage thérapeutique , Gonarthrose/traitement médicamenteux , Gonarthrose/anatomopathologie , Gonarthrose/étiologie , Gonarthrose/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Modèles animaux de maladie humaine , Mâle , Synovite/traitement médicamenteux , Synovite/étiologie , Synovite/anatomopathologie , Synovite/métabolisme , Cartilage articulaire/anatomopathologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Ostéophyte/traitement médicamenteux , Ostéophyte/métabolisme , Ostéophyte/étiologie , Ostéophyte/anatomopathologie
6.
Am J Sports Med ; 52(8): 2119-2128, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38857056

RÉSUMÉ

BACKGROUND: A major limitation of osteochondral allografts (OCA) is the deterioration of cartilage health associated with cell death during prolonged storage. However, little is known about the mechanisms that contribute to chondrocyte death during storage. PURPOSE/HYPOTHESIS: This study aimed to determine whether bioactive lipid metabolites accumulate in the storage media of OCA and whether they are associated with a loss of chondrocyte viability during prolonged storage. It was hypothesized that free fatty acids (FFAs) would accumulate over time in the storage media of OCA and adversely affect cartilage health during storage. STUDY DESIGN: Controlled laboratory study. METHODS: A group of 21 (n = 6-8 OCA/treatment group) fresh human hemicondylar OCA tissues and media were analyzed after 7, 28, and 68 days of prolonged cold (4°C) storage. Targeted mass spectrometry analysis was used to quantify bioactive FFAs, as well as primary (lipid hydroperoxide [ROOH]) and secondary (malondialdehyde) lipid oxidation products. Chondrocyte viability was measured using a fluorescence-based live/dead assay and confocal microscopy. RESULTS: The concentration of all targeted fatty acid metabolites in storage media was significantly increased with increased cold storage time (P < .05). ROOH was significantly higher on day 28 of cold storage. No difference in secondary ROOH products in storage media was observed. Chondrocyte viability significantly declined in both the en face and the vertical cross-sectional analysis with increased cold storage time and inversely correlated with fatty acid metabolites (P < .05). CONCLUSION: It is well established that elevated levels of certain FFAs and lipid oxidation products can alter cell function and cause cell death via lipotoxicity and other mechanisms. This work is the first to identify elevated levels of FFA metabolites and primary oxidation lipid products in the storage media from clinical OCA. The concentrations of FFA metabolites were measured at levels (>100 µM) known to induce cell death and were directly correlated with chondrocyte viability. CLINICAL RELEVANCE: These findings provide important targets for understanding why cartilage health declines during cold storage, which can be used to optimize media formulations and improve graft health.


Sujet(s)
Mort cellulaire , Chondrocytes , Humains , Chondrocytes/métabolisme , Acide gras libre/métabolisme , Survie cellulaire , Allogreffes , Adulte , Adulte d'âge moyen , Mâle , Cartilage articulaire/métabolisme , Femelle , Métabolisme lipidique
7.
Nat Rev Rheumatol ; 20(7): 432-451, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38858605

RÉSUMÉ

Joint kinematic instability, arising from congenital or acquired musculoskeletal pathoanatomy or from imbalances in anabolism and catabolism induced by pathophysiological factors, leads to deterioration of the composition, structure and function of cartilage and, ultimately, progression to osteoarthritis (OA). Alongside articular cartilage degeneration, synovial fluid lubricity decreases in OA owing to a reduction in the concentration and molecular weight of hyaluronic acid and surface-active mucinous glycoproteins that form a lubricating film over the articulating joint surfaces. Minimizing friction between articulating joint surfaces by lubrication is fundamental for decreasing hyaline cartilage wear and for maintaining the function of synovial joints. Augmentation with highly viscous supplements (that is, viscosupplementation) offers one approach to re-establishing the rheological and tribological properties of synovial fluid in OA. However, this approach has varied clinical outcomes owing to limited intra-articular residence time and ineffective mechanisms of chondroprotection. This Review discusses normal hyaline cartilage function and lubrication and examines the advantages and disadvantages of various strategies for restoring normal joint lubrication. These strategies include contemporary viscosupplements that contain antioxidants, anti-inflammatory drugs or platelet-rich plasma and new synthetic synovial fluid additives and cartilage matrix enhancers. Advanced biomimetic tribosupplements offer promise for mitigating cartilage wear, restoring joint function and, ultimately, improving patient care.


Sujet(s)
Arthrose , Viscosupplémentation , Humains , Viscosupplémentation/méthodes , Arthrose/traitement médicamenteux , Acide hyaluronique/usage thérapeutique , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Viscosuppléments/usage thérapeutique , Viscosuppléments/administration et posologie , Synovie/métabolisme , Compléments alimentaires
8.
ACS Biomater Sci Eng ; 10(7): 4437-4451, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38885017

RÉSUMÉ

Osteoarthritis (OA) is a chronic joint disease characterized by cartilage imbalance and disruption of cartilage extracellular matrix secretion. Identifying key genes that regulate cartilage differentiation and developing effective therapeutic strategies to restore their expression is crucial. In a previous study, we observed a significant correlation between the expression of the gene encoding casein kinase-2 interacting protein-1 (CKIP-1) in the cartilage of OA patients and OA severity scores, suggesting its potential involvement in OA development. To test this hypothesis, we synthesized a chondrocyte affinity plasmid, liposomes CKIP-1, to enhance CKIP-1 expression in chondrocytes. Our results demonstrated that injection of CAP-Lipos-CKIP-1 plasmid significantly improved OA joint destruction and restored joint motor function by enhancing cartilage extracellular matrix (ECM) secretion. Histological and cytological analyses confirmed that CKIP-1 maintains altered the phosphorylation of the signal transduction molecule SMAD2/3 of the transforming growth factor-ß (TGF-ß) pathway by promoting the phosphorylation of the 8T, 416S sit. Taken together, this work highlights a novel approach for the precise modulation of chondrocyte phenotype from an inflammatory to a noninflammatory state for the treatment of OA and may be broadly applicable to patients suffering from other arthritic diseases.


Sujet(s)
Chondrocytes , Homéostasie , Liposomes , Arthrose , Chondrocytes/métabolisme , Arthrose/thérapie , Arthrose/anatomopathologie , Arthrose/métabolisme , Liposomes/composition chimique , Humains , Animaux , Protéines de transport/métabolisme , Protéines de transport/génétique , Mâle , Phosphorylation , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Matrice extracellulaire/métabolisme , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Transduction du signal , Plasmides/génétique , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Protéine Smad2/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique
9.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38892389

RÉSUMÉ

Cartilage, a flexible and smooth connective tissue that envelops the surfaces of synovial joints, relies on chondrocytes for extracellular matrix (ECM) production and the maintenance of its structural and functional integrity. Melatonin (MT), renowned for its anti-inflammatory and antioxidant properties, holds the potential to modulate cartilage regeneration and degradation. Therefore, the present study was devoted to elucidating the mechanism of MT on chondrocytes. The in vivo experiment consisted of three groups: Sham (only the skin tissue was incised), Model (using the anterior cruciate ligament transection (ACLT) method), and MT (30 mg/kg), with sample extraction following 12 weeks of administration. Pathological alterations in articular cartilage, synovium, and subchondral bone were evaluated using Safranin O-fast green staining. Immunohistochemistry (ICH) analysis was employed to assess the expression of matrix degradation-related markers. The levels of serum cytokines were quantified via Enzyme-linked immunosorbent assay (ELISA) assays. In in vitro experiments, primary chondrocytes were divided into Control, Model, MT, negative control, and inhibitor groups. Western blotting (WB) and Quantitative RT-PCR (q-PCR) were used to detect Silent information regulator transcript-1 (SIRT1)/Nuclear factor kappa-B (NF-κB)/Nuclear factor erythroid-2-related factor 2 (Nrf2)/Transforming growth factor-beta (TGF-ß)/Bone morphogenetic proteins (BMPs)-related indicators. Immunofluorescence (IF) analysis was employed to examine the status of type II collagen (COL2A1), SIRT1, phosphorylated NF-κB p65 (p-p65), and phosphorylated mothers against decapentaplegic homolog 2 (p-Smad2). In vivo results revealed that the MT group exhibited a relatively smooth cartilage surface, modest chondrocyte loss, mild synovial hyperplasia, and increased subchondral bone thickness. ICH results showed that MT downregulated the expression of components related to matrix degradation. ELISA results showed that MT reduced serum inflammatory cytokine levels. In vitro experiments confirmed that MT upregulated the expression of SIRT1/Nrf2/TGF-ß/BMPs while inhibiting the NF-κB pathway and matrix degradation-related components. The introduction of the SIRT1 inhibitor Selisistat (EX527) reversed the effects of MT. Together, these findings suggest that MT has the potential to ameliorate inflammation, inhibit the release of matrix-degrading enzymes, and improve the cartilage condition. This study provides a new theoretical basis for understanding the role of MT in decelerating cartilage degradation and promoting chondrocyte repair in in vivo and in vitro cultured chondrocytes.


Sujet(s)
Cartilage articulaire , Chondrocytes , Mélatonine , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Transduction du signal , Sirtuine-1 , Facteur de croissance transformant bêta , Animaux , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Mélatonine/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Mâle , Matrice extracellulaire/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie
10.
Front Immunol ; 15: 1407679, 2024.
Article de Anglais | MEDLINE | ID: mdl-38868774

RÉSUMÉ

Background: Cartilage injury is the main pathological manifestation of osteoarthritis (OA). Healthy chondrocyte is a prerequisite for cartilage regeneration and repair. Differences between healthy and OA chondrocyte types and the role these types play in cartilage regeneration and OA progression are unclear. Method: This study conducted single-cell RNA sequencing (scRNA-seq) on the cartilage from normal distal femur of the knee (NC group) and OA femur (OA group) cartilage, the chondrocyte atlas was constructed, and the differences of cell subtypes between the two groups were compared. Pseudo-time and RNA velocity analysis were both performed to verify the possible differentiation sequence of cell subtypes. GO and KEGG pathway enrichment analysis were used to explore the potential functional characteristics of each cell subtype, and to predict the functional changes during cell differentiation. Differences in transcriptional regulation in subtypes were explored by single-cell regulatory network inference and clustering (SCENIC). The distribution of each cell subtype in cartilage tissue was identified by immunohistochemical staining (IHC). Result: A total of 75,104 cells were included, they were divided into 19 clusters and annotated as 11 chondrocyte subtypes, including two new chondrocyte subtypes: METRNL+ and PRG4+ subtype. METRNL+ is in an early stage during chondrocyte differentiation, and RegC-B is in an intermediate state before chondrocyte dedifferentiation. With cell differentiation, cell subtypes shift from genetic expression to extracellular matrix adhesion and collagen remodeling, and signal pathways shift from HIF-1 to Hippo. The 11 subtypes were finally classified as intrinsic chondrocytes, effector chondrocytes, abnormally differentiated chondrocytes and dedifferentiated chondrocytes. IHC was used to verify the presence and distribution of each chondrocyte subtype. Conclusion: This study screened two new chondrocyte subtypes, and a novel classification of each subtype was proposed. METRNL+ subtype is in an early stage during chondrocyte differentiation, and its transcriptomic characteristics and specific pathways provide a foundation for cartilage regeneration. EC-B, PRG4+ RegC-B, and FC are typical subtypes in the OA group, and the HippO-Taz pathway enriched by these cell subtypes may play a role in cartilage repair and OA progression. RegC-B is in the intermediate state before chondrocyte dedifferentiation, and its transcriptomic characteristics may provide a theoretical basis for intervening chondrocyte dedifferentiation.


Sujet(s)
Cartilage articulaire , Chondrocytes , Analyse sur cellule unique , Humains , Chondrocytes/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Analyse de séquence d'ARN , Fémur/métabolisme , Fémur/anatomopathologie , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Différenciation cellulaire , Mâle , Femelle , Transcriptome , Adulte d'âge moyen , Analyse de profil d'expression de gènes , Gonarthrose/métabolisme , Gonarthrose/anatomopathologie , Gonarthrose/génétique
11.
Arthritis Res Ther ; 26(1): 118, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851726

RÉSUMÉ

BACKGROUND: Primary osteoarthritis (OA) occurs without identifiable underlying causes such as previous injuries or specific medical conditions. Age is a major contributing factor to OA, and as one ages, various joint tissues undergo gradual change, including degeneration of the articular cartilage, alterations in subchondral bone (SCB) morphology, and inflammation of the synovium. METHODS: We investigated the prevalence of primary OA in aged, genetically diverse UM-HET3 mice. Articular cartilage (AC) integrity and SCB morphology were assessed in 182 knee joints of 22-25 months old mice using the Osteoarthritis Research Society International (OARSI) scoring system and micro-CT, respectively. Additionally, we explored the effects of methylene blue (MB) and mitoquinone (MitoQ), two agents that affect mitochondrial function, on the prevalence and progression of OA during aging. RESULTS: Aged UM-HET3 mice showed a high prevalence of primary OA in both sexes. Significant positive correlations were found between cumulative AC (cAC) scores and synovitis in both sexes, and osteophyte formation in female mice. Ectopic chondrogenesis did not show significant correlations with cAC scores. Significant direct correlations were found between AC scores and inflammatory markers in chondrocytes, including matrix metalloproteinase-13, inducible nitric oxide synthase, and the NLR family pyrin domain containing-3 inflammasome in both sexes, indicating a link between OA severity and inflammation. Additionally, markers of cell cycle arrest, such as p16 and ß-galactosidase, also correlated with AC scores. In male mice, no significant correlations were found between SCB morphology traits and cAC scores, while in female mice, significant correlations were found between cAC scores and tibial SCB plate bone mineral density. Notably, MB and MitoQ treatments influenced the disease's progression in a sex-specific manner. MB treatment significantly reduced cAC scores at the medial knee joint, while MitoQ treatment reduced cAC scores, but these did not reach significance. CONCLUSIONS: Our study provides comprehensive insights into the prevalence and progression of primary OA in aged UM-HET3 mice, highlighting the sex-specific effects of MB and MitoQ treatments. The correlations between AC scores and various pathological factors underscore the multifaceted nature of OA and its association with inflammation and subchondral bone changes.


Sujet(s)
Vieillissement , Arthrose , Animaux , Mâle , Femelle , Souris , Vieillissement/anatomopathologie , Vieillissement/génétique , Arthrose/génétique , Arthrose/anatomopathologie , Arthrose/métabolisme , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Bleu de méthylène/pharmacologie , Ubiquinones/analogues et dérivés , Ubiquinones/pharmacologie , Modèles animaux de maladie humaine , Évolution de la maladie
12.
J Nanobiotechnology ; 22(1): 325, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858695

RÉSUMÉ

BACKGROUND: Osteoarthritis (OA) is an aging-related degenerative joint disorder marked by joint discomfort and rigidity. Senescent chondrocytes release pro-inflammatory cytokines and extracellular matrix-degrading proteins, creating an inflammatory microenvironment that hinders chondrogenesis and accelerates matrix degradation. Targeting of senescent chondrocytes may be a promising approach for the treatment of OA. Herein, we describe the engineering of an injectable peptide-hydrogel conjugating a stem cell-homing peptide PFSSTKT for carrying plasmid DNA-laden nanoparticles and Tanshinon IIA (pPNP + TIIA@PFS) that was designed to attenuate OA progression by improving the senescent microenvironment and fostering cartilage regeneration. RESULTS: Specifically, pPNP + TIIA@PFS elevates the concentration of the anti-aging protein Klotho and blocks the transmission of senescence signals to adjacent healthy chondrocytes, significantly mitigating chondrocyte senescence and enhancing cartilage integrity. Additionally, pPNP + TIIA@PFS recruit bone mesenchymal stem cells and directs their subsequent differentiation into chondrocytes, achieving satisfactory chondrogenesis. In surgically induced OA model rats, the application of pPNP + TIIA@PFS results in reduced osteophyte formation and attenuation of articular cartilage degeneration. CONCLUSIONS: Overall, this study introduces a novel approach for the alleviation of OA progression, offering a foundation for potential clinical translation in OA therapy.


Sujet(s)
Chondrocytes , Chondrogenèse , Glucuronidase , Hydrogels , Protéines Klotho , Cellules souches mésenchymateuses , Arthrose , Plasmides , Rat Sprague-Dawley , Animaux , Arthrose/thérapie , Arthrose/traitement médicamenteux , Hydrogels/composition chimique , Rats , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Glucuronidase/métabolisme , Glucuronidase/pharmacologie , Chondrogenèse/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Mâle , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Évolution de la maladie , Nanoparticules/composition chimique , Humains , ADN , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques
13.
J Cell Mol Med ; 28(11): e18460, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38864710

RÉSUMÉ

Haemophilic arthropathy (HA), a common comorbidity in haemophilic patients leads to joint pain, deformity and reduced quality of life. We have recently demonstrated that a long non-coding RNA, Neat1 as a primary regulator of matrix metalloproteinase (MMP) 3 and MMP13 activity, and its induction in the target joint has a deteriorating effect on articular cartilage. In the present study, we administered an Adeno-associated virus (AAV) 5 vector carrying an short hairpin (sh)RNA to Neat1 via intra-articular injection alone or in conjunction with systemic administration of a capsid-modified AAV8 (K31Q) vector carrying F8 gene (F8-BDD-V3) to study its impact on HA. AAV8K31Q-F8 vector administration at low dose, led to an increase in FVIII activity (16%-28%) in treated mice. We further observed a significant knockdown of Neat1 (~40 fold vs. untreated injured joint, p = 0.005) in joint tissue of treated mice and a downregulation of chondrodegenerative enzymes, MMP3, MMP13 and the inflammatory mediator- cPLA2, in mice receiving combination therapy. These data demonstrate that AAV mediated Neat1 knockdown in combination with F8 gene augmentation can potentially impact mediators of haemophilic joint disease.


Sujet(s)
Dependovirus , Facteur VIII , Vecteurs génétiques , Hémophilie A , Matrix Metalloproteinase 13 , Matrix metalloproteinase 3 , ARN long non codant , Animaux , Hémophilie A/génétique , Hémophilie A/thérapie , Hémophilie A/complications , Dependovirus/génétique , ARN long non codant/génétique , Matrix Metalloproteinase 13/métabolisme , Matrix Metalloproteinase 13/génétique , Souris , Matrix metalloproteinase 3/génétique , Matrix metalloproteinase 3/métabolisme , Vecteurs génétiques/génétique , Vecteurs génétiques/administration et posologie , Facteur VIII/génétique , Facteur VIII/métabolisme , Maladies articulaires/thérapie , Maladies articulaires/génétique , Maladies articulaires/étiologie , Humains , Thérapie génétique/méthodes , Souris de lignée C57BL , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Modèles animaux de maladie humaine , Mâle
14.
J Nanobiotechnology ; 22(1): 345, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38890638

RÉSUMÉ

Osteoarthritis (OA) is a common degenerative joint disease which currently lacks of effective agents. It is therefore urgent and necessary to seek an effective approach that can inhibit inflammation and promote cartilage matrix homeostasis. Cartilage progenitor cells (CPCs) are identified as a cell population of superficial zone in articular cartilage which possess strong migration ability, proliferative capacity, and chondrogenic potential. Recently, the application of CPCs may represent a novel cell therapy strategy for OA treatment. There is growing evidence that extracellular vesicles (EVs) are primary mediators of the benefits of stem cell-based therapy. In this study, we explored the protective effects of CPCs-derived EVs (CPCs-EVs) on IL-1ß-induced chondrocytes. We found CPCs-EVs exhibited chondro-protective effects in vitro. Furthermore, our study demonstrated that CPCs-EVs promoted matrix anabolism and inhibited inflammatory response at least partially via blocking STAT3 activation. In addition, liquid chromatography-tandem mass spectrometry analysis identified 991 proteins encapsulated in CPCs-EVs. By bioinformatics analysis, we showed that STAT3 regulatory proteins were enriched in CPCs-EVs and could be transported to chondrocytes. To promoting the protective function of CPCs-EVs in vivo, CPCs-EVs were modified with cationic peptide ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) for surface charge reverse. In posttraumatic OA mice, our results showed PPD modified CPCs-EVs (PPD-EVs) effectively inhibited extracellular matrix catabolism and attenuated cartilage degeneration. Moreover, PPD-EVs down-regulated inflammatory factors expressions and reduced OA-related pain in OA mice. In ex-vivo cultured OA cartilage explants, PPD-EVs successfully promoted matrix anabolism and inhibited inflammation. Collectively, CPCs-EVs-based cell-free therapy is a promising strategy for OA treatment.


Sujet(s)
Cartilage articulaire , Chondrocytes , Matrice extracellulaire , Vésicules extracellulaires , Inflammation , Arthrose , Cellules souches , Vésicules extracellulaires/métabolisme , Animaux , Arthrose/thérapie , Arthrose/métabolisme , Matrice extracellulaire/métabolisme , Souris , Chondrocytes/métabolisme , Inflammation/métabolisme , Cartilage articulaire/métabolisme , Cellules souches/métabolisme , Homéostasie , Souris de lignée C57BL , Mâle , Facteur de transcription STAT-3/métabolisme , Cellules cultivées , Interleukine-1 bêta/métabolisme
15.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892089

RÉSUMÉ

Post-traumatic osteoarthritis of the ankle (PTOA) is frequently observed following a debilitating consequence of intra-articular ankle fractures. Numerous risk factors contribute to the pathogenesis of PTOA, including articular incongruity, joint malalignment, and concomitant soft tissue damage. Despite attempts to restore joint anatomy and manage soft tissues to avoid long-term complications after intra-articular ankle fractures, the incidence of PTOA remains markedly elevated. Inflammatory processes triggered by intra-articular ankle fractures have emerged as potential instigators that expedite the progression of PTOA. Injury to the articular cartilage and subchondral bone may lead to the release of inflammatory mediators, which can contribute to cartilage degradation and bone resorption. This study provides a narrative review on the current knowledge concerning the association between inflammation and the development of PTOA following intra-articular ankle fractures. We also discuss novel therapeutic agents that target inflammatory pathways to impede the progression of post-traumatic osteoarthritis after intra-articular ankle fractures. These medication and interventions were summarized within this review article.


Sujet(s)
Inflammation , Arthrose , Humains , Arthrose/étiologie , Arthrose/anatomopathologie , Arthrose/métabolisme , Inflammation/anatomopathologie , Animaux , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Articulation talocrurale/anatomopathologie , Fractures de la cheville/complications , Fractures de la cheville/anatomopathologie , Fractures de la cheville/métabolisme , Traumatismes de la cheville/complications , Traumatismes de la cheville/anatomopathologie
16.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892229

RÉSUMÉ

Synthetic deer antler peptides (TSKYR, TSK, and YR) stimulate the proliferation of human chondrocytes and osteoblasts and increase the chondrocyte content of collagen and glycosamino-glycan in vitro. This study investigated the peptide mixture's pain relief and chondroprotective effect in a rat model of collagenase-induced osteoarthritis. Thirty-six adult male Sprague-Dawley rats were divided into three groups: control (saline), positive control (hyaluronic acid), and ex-perimental (peptides). Intra-articular collagenase injections were administered on days 1 and 4 to induce osteoarthritis in the left knees of the rats. Two injections of saline, hyaluronic acid, or the peptides were injected into the same knees of each corresponding group at the beginning of week one and two, respectively. Joint swelling, arthritic pain, and histopathological changes were evaluated. Injection of the peptides significantly reduced arthritic pain compared to the control group, as evidenced by the closer-to-normal weight-bearing and paw withdrawal threshold test results. Histological analyses showed reduced cartilage matrix loss and improved total cartilage degeneration score in the experimental versus the control group. Our findings suggest that intra-articular injection of synthetic deer antler peptides is a promising treatment for osteoarthritis.


Sujet(s)
Andouillers , Cervidae , Modèles animaux de maladie humaine , Gonarthrose , Peptides , Rat Sprague-Dawley , Animaux , Injections articulaires , Andouillers/composition chimique , Gonarthrose/traitement médicamenteux , Gonarthrose/anatomopathologie , Gonarthrose/induit chimiquement , Mâle , Rats , Peptides/administration et posologie , Peptides/pharmacologie , Peptides/usage thérapeutique , Acide hyaluronique/administration et posologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Collagenases
17.
Front Endocrinol (Lausanne) ; 15: 1393550, 2024.
Article de Anglais | MEDLINE | ID: mdl-38854686

RÉSUMÉ

Osteoarthritis (OA) is an intricate pathological condition that primarily affects the entire synovial joint, especially the hip, hand, and knee joints. This results in inflammation in the synovium and osteochondral injuries, ultimately causing functional limitations and joint dysfunction. The key mechanism responsible for maintaining articular cartilage function is chondrocyte metabolism, which involves energy generation through glycolysis, oxidative phosphorylation, and other metabolic pathways. Some studies have shown that chondrocytes in OA exhibit increased glycolytic activity, leading to elevated lactate production and decreased cartilage matrix synthesis. In OA cartilage, chondrocytes display alterations in mitochondrial activity, such as decreased ATP generation and increased oxidative stress, which can contribute to cartilage deterioration. Chondrocyte metabolism also involves anabolic processes for extracellular matrix substrate production and energy generation. During OA, chondrocytes undergo considerable metabolic changes in different aspects, leading to articular cartilage homeostasis deterioration. Numerous studies have been carried out to provide tangible therapies for OA by using various models in vivo and in vitro targeting chondrocyte metabolism, although there are still certain limitations. With growing evidence indicating the essential role of chondrocyte metabolism in disease etiology, this literature review explores the metabolic characteristics and changes of chondrocytes in the presence of OA, both in vivo and in vitro. To provide insight into the complex metabolic reprogramming crucial in chondrocytes during OA progression, we investigate the dynamic interaction between metabolic pathways, such as glycolysis, lipid metabolism, and mitochondrial function. In addition, this review highlights prospective future research directions for novel approaches to diagnosis and treatment. Adopting a multifaceted strategy, our review aims to offer a comprehensive understanding of the metabolic intricacies within chondrocytes in OA, with the ultimate goal of identifying therapeutic targets capable of modulating chondrocyte metabolism for the treatment of OA.


Sujet(s)
Cartilage articulaire , Chondrocytes , Arthrose , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Humains , Arthrose/métabolisme , Arthrose/anatomopathologie , Animaux , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Glycolyse
18.
Front Immunol ; 15: 1361606, 2024.
Article de Anglais | MEDLINE | ID: mdl-38846937

RÉSUMÉ

Introduction: Pathological changes in the articular cartilage (AC) and synovium are major manifestations of osteoarthritis (OA) and are strongly associated with pain and functional limitations. Exosome-derived microRNAs (miRNAs) are crucial regulatory factors in intercellular communication and can influence the progression of OA by participating in the degradation of chondrocytes and the phenotypic transformation in the polarization of synovial macrophages. However, the specific relationships and pathways of action of exosomal miRNAs in the pathological progression of OA in both cartilage and synovium remain unclear. Methods: This study evaluates the effects of fibroblast-like synoviocyte (FLS)-derived exosomes (FLS-Exos), influenced by miR-146a, on AC degradation and synovial macrophage polarization. We investigated the targeted relationship between miR-146a and TRAF6, both in vivo and in vitro, along with the involvement of the NF-κB signaling pathway. Results: The expression of miR-146a in the synovial exosomes of OA rats was significantly higher than in healthy rats. In vitro, the upregulation of miR-146a reduced chondrocyte apoptosis, whereas its downregulation had the opposite effect. In vivo, exosomes derived from miR-146a-overexpressing FLSs (miR-146a-FLS-Exos) reduced AC injury and chondrocyte apoptosis in OA. Furthermore, synovial proliferation was reduced, and the polarization of synovial macrophages shifted from M1 to M2. Mechanistically, the expression of TRAF6 was inhibited by targeting miR-146a, thereby modulating the Toll-like receptor 4/TRAF6/NF-κB pathway in the innate immune response. Discussion: These findings suggest that miR-146a, mediated through FLS-Exos, may alleviate OA progression by modulating cartilage degradation and macrophage polarization, implicating the NF-κB pathway in the innate immune response. These insights highlight the therapeutic potential of miR-146a as a protective agent in OA, underscoring the importance of exosomal miRNAs in the pathogenesis and potential treatment of the disease.


Sujet(s)
Exosomes , Macrophages , microARN , Arthrose , Cellules synoviales , Facteur-6 associé aux récepteurs de TNF , microARN/génétique , Animaux , Exosomes/métabolisme , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/immunologie , Rats , Macrophages/immunologie , Macrophages/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Mâle , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Chondrocytes/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Rat Sprague-Dawley , Fibroblastes/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/immunologie , Cellules cultivées , Apoptose , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Activation des macrophages
19.
BMC Musculoskelet Disord ; 25(1): 447, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844896

RÉSUMÉ

BACKGROUND: Although various anti-inflammatory medicines are widely recommended for osteoarthritis (OA) treatment, no significantly clinical effect has been observed. This study aims to examine the effects of vitamin B6, a component that has been reported to be capable of alleviating inflammation and cell death in various diseases, on cartilage degeneration in OA. METHODS: Collagen-induced arthritis (CIA) mice model were established and the severity of OA in cartilage was determined using the Osteoarthritis Research Society International (OARSI) scoring system. The mRNA and protein levels of indicators associated with extracellular matrix (ECM) metabolism, apoptosis and inflammation were detected. The effect of vitamin B6 (VB6) on the mice were assessed using HE staining and masson staining. The apoptosis rate of cells was assessed using TdT-mediated dUTP nick end labeling. RESULTS: Our results showed a trend of improved OARSI score in mice treated with VB6, which remarkably inhibited the hyaline cartilage thickness, chondrocyte disordering, and knees hypertrophy. Moreover, the VB6 supplementation reduced the protein expression of pro-apoptosis indicators, including Bax and cleaved caspase-3 and raised the expression level of anti-apoptosis marker Bcl-2. Importantly, VB6 improved ECM metabolism in both in vivo and in vitro experiments. CONCLUSIONS: This study demonstrated that VB6 alleviates OA through regulating ECM metabolism, inflammation and apoptosis in chondrocytes and CIA mice. The findings in this study provide a theoretical basis for targeted therapy of OA, and further lay the theoretical foundation for studies of mechanisms of VB6 in treating OA.


Sujet(s)
Apoptose , Arthrite expérimentale , Chondrocytes , Inflammation , Arthrose , Vitamine B6 , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Vitamine B6/pharmacologie , Vitamine B6/usage thérapeutique , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Mâle , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Souris de lignée DBA , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/anatomopathologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme
20.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891793

RÉSUMÉ

Joint-resident chondrogenic precursor cells have become a significant therapeutic option due to the lack of regenerative capacity in articular cartilage. Progenitor cells are located in the superficial zone of the articular cartilage, producing lubricin/Prg4 to decrease friction of cartilage surfaces during joint movement. Prg4-positive progenitors are crucial in maintaining the joint's structure and functionality. The disappearance of progenitor cells leads to changes in articular hyaline cartilage over time, subchondral bone abnormalities, and the formation of ectopic ossification. Genetic labeling cell technology has been the main tool used to characterize Prg4-expressing progenitor cells of articular cartilage in vivo through drug injection at different time points. This technology allows for the determination of the origin of progenitor cells and the tracking of their progeny during joint development and cartilage damage. We endeavored to highlight the currently known information about the Prg4-producing cell population in the joint to underline the significance of the role of these cells in the development of articular cartilage and its homeostasis. This review focuses on superficial progenitors in the joint, how they contribute to postnatal articular cartilage formation, their capacity for regeneration, and the consequences of Prg4 deficiency in these cells. We have accumulated information about the Prg4+ cell population of articular cartilage obtained through various elegantly designed experiments using transgenic technologies to identify potential opportunities for further research.


Sujet(s)
Cartilage articulaire , Protéoglycanes , Cellules souches , Cartilage articulaire/métabolisme , Cartilage articulaire/cytologie , Animaux , Humains , Cellules souches/métabolisme , Cellules souches/cytologie , Protéoglycanes/métabolisme , Chondrogenèse , Chondrocytes/métabolisme , Chondrocytes/cytologie , Différenciation cellulaire , Régénération
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...