Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.744
Filtrer
1.
J Nanobiotechnology ; 22(1): 466, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095867

RÉSUMÉ

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


Sujet(s)
Apoptose , Chondrocytes , Hydrogels , Matrix Metalloproteinase 13 , Arthrose , Espèces réactives de l'oxygène , Animaux , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Matrix Metalloproteinase 13/métabolisme , Souris , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mâle , Cartilage articulaire/métabolisme , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Liposomes/composition chimique , Humains
2.
Biointerphases ; 19(4)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39051723

RÉSUMÉ

Once damaged, cartilage has poor intrinsic capacity to repair itself. Current cartilage repair strategies cannot restore the damaged tissue sufficiently. It is hypothesized that biomimetic scaffolds, which can recapitulate important properties of the cartilage extracellular matrix, play a beneficial role in supporting cell behaviors such as growth, cartilage differentiation, and integration with native cartilage, ultimately facilitating tissue recovery. Adipose-derived stem cells regenerated cartilage upon the sequential release of transforming growth factor ß1(TGFß1) and fibroblast growth factor 2(FGF2) using a nanofibrous scaffold, in order to get the recovery of functional cartilage. Experiments in vitro have demonstrated that the release sequence of growth factors FGF2 to TGFß1 is the most essential to promote adipose-derived stem cells into chondrocytes that then synthesize collagen II. Mouse subcutaneous implantation indicated that the treatment sequence of FGF2 to TGFß1 was able to significantly induce multiple increase in cartilage regeneration in vivo. This result demonstrates that the group treated with FGF2 to TGFß1 released from a nanofibrous scaffold provides a good strategy for cartilage regeneration by making a favorable microenvironment for cell growth and cartilage regeneration.


Sujet(s)
Différenciation cellulaire , Facteur de croissance fibroblastique de type 2 , Nanofibres , Cellules souches , Structures d'échafaudage tissulaires , Facteur de croissance transformant bêta-1 , Animaux , Facteur de croissance fibroblastique de type 2/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/pharmacologie , Souris , Nanofibres/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Structures d'échafaudage tissulaires/composition chimique , Cellules souches/cytologie , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/physiologie , Chondrogenèse/effets des médicaments et des substances chimiques , Cartilage/effets des médicaments et des substances chimiques , Cartilage/cytologie , Cartilage/physiologie , Tissu adipeux/cytologie , Chondrocytes/cytologie , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/physiologie , Cellules cultivées , Ingénierie tissulaire/méthodes
3.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39000370

RÉSUMÉ

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


Sujet(s)
Acroléine , Chondrocytes , Exosomes , Inflammation , Interleukine-1 bêta , Cellules souches mésenchymateuses , Transduction du signal , Exosomes/métabolisme , Interleukine-1 bêta/métabolisme , Acroléine/analogues et dérivés , Acroléine/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Animaux , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Humains , Cellules cultivées
4.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000568

RÉSUMÉ

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


Sujet(s)
Cartilage articulaire , Chondrocytes , Modèles animaux de maladie humaine , Ferroptose , Facteur-2 apparenté à NF-E2 , Arthrose , Quercétine , Sirtuine-1 , Animaux , Sirtuine-1/métabolisme , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/anatomopathologie , Rats , Quercétine/pharmacologie , Quercétine/usage thérapeutique , Facteur-2 apparenté à NF-E2/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Interleukine-1 bêta/métabolisme , Heme oxygenase (decyclizing)/métabolisme
5.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3330-3339, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-39041096

RÉSUMÉ

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


Sujet(s)
Chondrocytes , Médicaments issus de plantes chinoises , Arthrose , Rat Sprague-Dawley , Transduction du signal , Protéine p53 suppresseur de tumeur , Animaux , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Arthrose/génétique , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Mâle , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Capsules , Humains , Apoptose/effets des médicaments et des substances chimiques
6.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38982592

RÉSUMÉ

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


Sujet(s)
Apoptose , Chondrocytes , Clusterine , Interleukine-1 bêta , Gonarthrose , Humains , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Gonarthrose/anatomopathologie , Gonarthrose/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Clusterine/métabolisme , Clusterine/génétique , Interleukine-1 bêta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées , Mâle , Adulte d'âge moyen , Sujet âgé , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Femelle , Phosphatidylinositol 3-kinases/métabolisme , Morpholines/pharmacologie , 4H-1-Benzopyran-4-ones/pharmacologie , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Matrix Metalloproteinase 13/métabolisme , Médiateurs de l'inflammation/métabolisme , Monoxyde d'azote/métabolisme
7.
Nutrients ; 16(14)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39064775

RÉSUMÉ

The most widely used synthetic glucocorticoid, dexamethasone (DEX), causes stunted growth in children when used excessively or for long periods of time; however, there are still plenty of pediatric patients require long-term treatment with DEX. As an alternative, growth hormone is used in combination, but it has side effects, a high cost, and psychological factors, and it is not satisfactory in terms of effectiveness. It is necessary to develop a safe and affordable treatment that can replace it. The Korean Food and Drug Administration approved HT042, a standardized functional food ingredient, with the claim that it can help height growth of children. In this study, it was found that HT042 activated the Indian hedgehog/parathyroid hormone-related protein signaling pathway and enhanced the number of growth hormone receptors and insulin-like growth factor-1 receptors on the growth plate surface, which were reduced by DEX treatment, and restored growth retardation. In metatarsal bone and primary chondrocyte models, it was found that HT042 can promote the length of growth plate and recover DEX-induced growth retardation. It was also found that HT042 promotes cell proliferation using bromodeoxyuridine and terminal deoxynucleotidyl transferase dUTP nick end labeling assays; moreover, we verified increased expression of GHR/IGF-1R and Ihh/PTHrP pathway activity using qRT-PCR, western blotting, and siRNA analyses to verify its direct action on the growth plate. The anti-apoptotic effect of HT042 was identified by regulating the expression of apoptotic factors such as caspase-3, Bcl2, Bclx, and Bax. These results were identified using both ex vivo and in vitro models. Our study verified that co-administration of HT042 could recover the DEX induced growth retardation.


Sujet(s)
Prolifération cellulaire , Dexaméthasone , Lame épiphysaire , Os du métatarse , Extraits de plantes , Transduction du signal , Animaux , Dexaméthasone/pharmacologie , Os du métatarse/effets des médicaments et des substances chimiques , Lame épiphysaire/effets des médicaments et des substances chimiques , Rats , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Mâle , Protéines Hedgehog/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Protéine apparentée à l'hormone parathyroïdienne/métabolisme , Rat Sprague-Dawley , Apoptose/effets des médicaments et des substances chimiques , Récepteur STH/métabolisme , Récepteur STH/génétique , Récepteur IGF de type 1/métabolisme , Récepteur IGF de type 1/génétique , Développement osseux/effets des médicaments et des substances chimiques , Troubles de la croissance/induit chimiquement
8.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-39062860

RÉSUMÉ

The actions of the retinoic acid nuclear receptor gamma (RARγ) agonist, palovarotene, on pre-existing osteochondromas were investigated using a mouse multiple osteochondroma model. This approach was based on the knowledge that patients often present to the clinic after realizing the existence of osteochondroma masses, and the findings from preclinical investigations are the effects of drugs on the initial formation of osteochondromas. Systemic administration of palovarotene, with increased doses (from 1.76 to 4.0 mg/kg) over time, fully inhibited tumor growth, keeping the tumor size (0.31 ± 0.049 mm3) similar to the initial size (0.27 ± 0.031 mm3, p = 0.66) while the control group tumor grew (1.03 ± 0.23 mm3, p = 0.023 to the drug-treated group). Nanoparticle (NP)-based local delivery of the RARγ agonist also inhibited the growth of osteochondromas at an early stage (Control: 0.52 ± 0.11 mm3; NP: 0.26 ± 0.10, p = 0.008). Transcriptome analysis revealed that the osteoarthritis pathway was activated in cultured chondrocytes treated with palovarotene (Z-score = 2.29), with the upregulation of matrix catabolic genes and the downregulation of matrix anabolic genes, consistent with the histology of palovarotene-treated osteochondromas. A reporter assay performed in cultured chondrocytes demonstrated that the Stat3 pathway, but not the Stat1/2 pathway, was stimulated by RARγ agonists. The activation of Stat3 by palovarotene was confirmed using immunoblotting and immunohistochemistry. These findings suggest that palovarotene treatment is effective against pre-existing osteochondromas and that the Stat3 pathway is involved in the antitumor actions of palovarotene.


Sujet(s)
Chondrocytes , Modèles animaux de maladie humaine , Ostéochondrome , Récepteurs à l'acide rétinoïque , , Animaux , Souris , Récepteurs à l'acide rétinoïque/agonistes , Récepteurs à l'acide rétinoïque/métabolisme , Ostéochondrome/traitement médicamenteux , Ostéochondrome/anatomopathologie , Ostéochondrome/métabolisme , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Mâle
9.
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
10.
ACS Appl Mater Interfaces ; 16(30): 39153-39164, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39018481

RÉSUMÉ

Temporomandibular joint osteoarthritis (TMJ OA) is characterized by the degeneration of cartilage and subchondral bone. In this study, we observed a significant increase in cell-free DNA (cfDNA) levels during the progression of TMJ OA. Bioinformatics analysis identified TLR9 as a pivotal molecule in TMJ OA pathogenesis. The polyamidoamine (PAMAM) dendrimer characterized by a well-structured, highly branched, and reactive nature, exhibits robust binding and clearance capabilities for cfDNA. However, the abundant amino groups on the surface of PAMAM lead to its inherent toxicity. To mitigate this, PEG-5000 was conjugated to the surface of PAMAM dendrimers, enhancing safety. Our results indicate that PEG-PAMAM effectively inhibits the upregulation of the TLR9 protein in TMJ OA, significantly suppressing the activation of the p-IκBα/p-NF-κB signaling pathway and subsequently decreasing chondrocyte inflammation and apoptosis, as evidenced by both in vivo and in vitro experiments. We conclude that PEG-PAMAM is a safe and effective material for in vivo applications, offering a promising therapeutic strategy for TMJ OA by targeting cfDNA clearance.


Sujet(s)
Acides nucléiques acellulaires , Dendrimères , Arthrose , Polyéthylène glycols , Articulation temporomandibulaire , Dendrimères/composition chimique , Dendrimères/pharmacologie , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , Arthrose/métabolisme , Animaux , Polyéthylène glycols/composition chimique , Articulation temporomandibulaire/anatomopathologie , Articulation temporomandibulaire/effets des médicaments et des substances chimiques , Articulation temporomandibulaire/métabolisme , Adsorption , Humains , Récepteur-9 de type Toll-like/métabolisme , Mâle , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Nylons/composition chimique , Nylons/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Souris
11.
Int Immunopharmacol ; 138: 112601, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38971106

RÉSUMÉ

Osteoarthritis (OA) is a joint disease caused by inflammation of cartilage and synovial tissue. Suppressing the process of inflammatory reaction and the generation of oxidative stress is an effective strategy to alleviate the progression of OA. Liensinine is one of the main components of lotus seeds, which has anti-hypertensive and anti-arrhythmia activities. In this study, we aimed to determine the anti-inflammatory effect of liensinine in an OA. Here, we found that liensinine significantly inhibited the inflammatory response of SW1353 cells and primary chondrocytes by inhibiting the release of inflammatory cytokines and oxidative stress. Moreover, we showed that liensinine was able to inhibit the activation of the NF-κB signaling pathway in IL-1ß-induced SW1353 cells. Lastly, we found that liensinine significantly ameliorated cartilage damage and inflammatory response in papain-induced rats. Our study demonstrated a significant protective effect of liensinine against OA, which might be by inhibiting the activation of the NF-κB signaling pathway, and provide a new insight for the treatment of OA using liensinine.


Sujet(s)
Anti-inflammatoires , Chondrocytes , Interleukine-1 bêta , Facteur de transcription NF-kappa B , Arthrose , Animaux , Humains , Mâle , Rats , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Cellules cultivées , Chondrocytes/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Interleukine-1 bêta/métabolisme , Isoquinoléines , Facteur de transcription NF-kappa B/métabolisme , Arthrose/traitement médicamenteux , Arthrose/induit chimiquement , Stress oxydatif/effets des médicaments et des substances chimiques , Papaïne , Phénols , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques
12.
ACS Appl Mater Interfaces ; 16(29): 37683-37697, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-38980692

RÉSUMÉ

Tissue engineering is theoretically considered a promising approach for repairing osteochondral defects. Nevertheless, the insufficient osseous support and integration of the cartilage layer and the subchondral bone frequently lead to the failure of osteochondral repair. Drawing from this, it was proposed that incorporating glycine-modified attapulgite (GATP) into poly(1,8-octanediol-co-citrate) (POC) scaffolds via the one-step chemical cross-linking is proposed to enhance cartilage and subchondral bone defect repair simultaneously. The effects of the GATP incorporation ratio on the physicochemical properties, chondrocyte and MC3T3-E1 behavior, and osteochondral defect repair of the POC scaffold were also evaluated. In vitro studies indicated that the POC/10% GATP scaffold improved cell proliferation and adhesion, maintained cell phenotype, and upregulated chondrogenesis and osteogenesis gene expression. Animal studies suggested that the POC/10% GATP scaffold has significant repair effects on both cartilage and subchondral bone defects. Therefore, the GATP-incorporated scaffold system with dual-lineage bioactivity showed potential application in osteochondral regeneration.


Sujet(s)
Ingénierie tissulaire , Structures d'échafaudage tissulaires , Animaux , Structures d'échafaudage tissulaires/composition chimique , Souris , Chondrocytes/cytologie , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Régénération osseuse/effets des médicaments et des substances chimiques , Chondrogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lapins , Os et tissu osseux/effets des médicaments et des substances chimiques , Régénération/effets des médicaments et des substances chimiques
13.
J Nanobiotechnology ; 22(1): 445, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39069607

RÉSUMÉ

BACKGROUND: The incidence of osteochondral defects caused by trauma, arthritis or tumours is increasing annually, but progress has not been made in terms of treatment methods. Due to the heterogeneous structure and biological characteristics of cartilage and subchondral bone, the integration of osteochondral repair is still a challenge. RESULTS: In the present study, a novel bilayer hydrogel scaffold was designed based on anatomical characteristics to imitate superficial cartilage and subchondral bone. The scaffold showed favourable biocompatibility, and the addition of an antioxidant nanozyme (LiMn2O4) promoted reactive oxygen species (ROS) scavenging by upregulating antioxidant proteins. The cartilage layer effectively protects against chondrocyte degradation in the inflammatory microenvironment. Subchondral bionic hydrogel scaffolds promote osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) by regulating the AMPK pathway in vitro. Finally, an in vivo rat preclinical osteochondral defect model confirmed that the bilayer hydrogel scaffold efficiently promoted cartilage and subchondral bone regeneration. CONCLUSIONS: In general, our biomimetic hydrogel scaffold with the ability to regulate the inflammatory microenvironment can effectively repair osteochondral defects. This strategy provides a promising method for regenerating tissues with heterogeneous structures and biological characteristics.


Sujet(s)
Régénération osseuse , Hydrogels , Cellules souches mésenchymateuses , Ostéogenèse , Rat Sprague-Dawley , Structures d'échafaudage tissulaires , Animaux , Hydrogels/composition chimique , Hydrogels/pharmacologie , Structures d'échafaudage tissulaires/composition chimique , Rats , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Régénération osseuse/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Chondrocytes/effets des médicaments et des substances chimiques , Mâle , Différenciation cellulaire/effets des médicaments et des substances chimiques , Inflammation , Ingénierie tissulaire/méthodes , Espèces réactives de l'oxygène/métabolisme , Chondrogenèse/effets des médicaments et des substances chimiques , Cartilage/effets des médicaments et des substances chimiques , Cartilage articulaire/effets des médicaments et des substances chimiques , Cellules cultivées
14.
BMC Musculoskelet Disord ; 25(1): 601, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080620

RÉSUMÉ

BACKGROUND: This study aimed to investigate functions of GLP-1R agonist by liraglutide (LIRA) and revealing the mechanism related to AGEs/RAGE in chondrocytes. METHODS: To illustrate potential effect of GLP-1R agonist on AGEs induced chondrocytes, chondrocytes were administrated by AGEs with LIRA and GLP-1R inhibitor exendin. Inflammatory factors were assessed using ELISA. Real-time PCR was used to evaluate the catabolic activity MMPs and ADAMTS mRNA level, as well as anabolic activity (aggrecan and collagen II). RAGE expression was investigated by Western blotting. TUNEL, caspase3 activity and immunofluorescence were performed to test the apoptotic activity. RESULTS: Our results showed that treatment with LIRA at > 100 nM attenuated the AGE-induced chondrocyte viability. Western bolt demonstrated that GLP-1R activation by LIRA treatment reduced RAGE protein expression compared with the AGEs groups. ELISA showed that LIRA hindered the AGEs-induced production of inflammatory cytokines (IL-6, IL-12 and TNF-α) in primary chondrocytes. AGEs induced catabolism levels (MMP-1, -3, -13 and ADAMTS-4, 5) are also attenuated by LIRA, causing the retention of more extracellular matrix (Aggrecan and Collagen II). TUNEL, caspase3 activity and immunofluorescence results indicated that LIRA inhibited the AGEs-induced production of inflammatory cytokines in primary chondrocytes and attenuated the caspase 3 level, leading to the reduced apoptotic activity. All the protective effects are reversed by exendin (GLP-1R blockers). CONCLUSIONS: The present study demonstrates for the first time that LIRA, an agonist for GLP-1R which is commonly used in type 2 diabetes reverses AGEs induced chondrocyte inflammation and apoptosis through suppressing RAGE signaling, contributing to reduced catabolism and retention of more extracellular matrix. The above results indicate the possible effect of GLP-1R agonist on treating OA.


Sujet(s)
Apoptose , Chondrocytes , Récepteur du peptide-1 similaire au glucagon , Produits terminaux de glycation avancée , Inflammation , Liraglutide , Récepteur spécifique des produits finaux de glycosylation avancée , Transduction du signal , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Liraglutide/pharmacologie , Produits terminaux de glycation avancée/métabolisme , Récepteur du peptide-1 similaire au glucagon/agonistes , Récepteur du peptide-1 similaire au glucagon/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Cellules cultivées
15.
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
16.
Carbohydr Polym ; 339: 122251, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38823918

RÉSUMÉ

In this study, the disulfide-linked hyaluronic acid (HA) hydrogels were optimised for potential application as a scaffold in tissue engineering through the Quality by Design (QbD) approach. For this purpose, HA was first modified by incorporating the cysteine moiety into the HA backbone, which promoted the formation of disulfide cross-linked HA hydrogel at physiological pH. Utilising a Design of Experiments (DoE) methodology, the critical factors to achieve stable biomaterials, i.e. the degree of HA substitution, HA molecular weight, and coupling agent ratio, were explored. To establish a design space, the DoE was performed with 65 kDa, 138 kDa and 200 kDa HA and variable concentrations of coupling agent to optimise conditions to obtain HA hydrogel with improved rheological properties. Thus, HA hydrogel with a 12 % degree of modification, storage modulus of ≈2321 Pa and loss modulus of ≈15 Pa, was achieved with the optimum ratio of coupling agent. Furthermore, biocompatibility assessments in C28/I2 chondrocyte cells demonstrated the non-toxic nature of the hydrogel, underscoring its potential for tissue regeneration. Our findings highlight the efficacy of the QbD approach in designing HA hydrogels with tailored properties for biomedical applications.


Sujet(s)
Matériaux biocompatibles , Chondrocytes , Disulfures , Acide hyaluronique , Hydrogels , Rhéologie , Ingénierie tissulaire , Acide hyaluronique/composition chimique , Hydrogels/composition chimique , Hydrogels/synthèse chimique , Disulfures/composition chimique , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/cytologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/synthèse chimique , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires/composition chimique , Animaux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Concentration en ions d'hydrogène
17.
Int J Biol Sci ; 20(8): 2994-3007, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904008

RÉSUMÉ

Osteoarthritis (OA) is a challenging degenerative joint disease to manage. Previous research has indicated that cell-free fat extract (CEFFE) may hold potential for OA treatment. This study investigated the role of Annexin A5 (AnxA5) within CEFFE in regulating macrophage polarization and protecting chondrocytes. In vitro experiments demonstrated that AnxA5 effectively inhibited M1 macrophage polarization by facilitating toll-like receptor (TLR) 4 internalization and lysosomal degradation through calcium-dependent endocytosis. This process decreased TLR4 expression, suppressed pro-inflammatory mediator release, and reduced the production of reactive oxygen species. Furthermore, AnxA5 displayed protective effects against chondrocyte necrosis and apoptosis. In vivo, studies revealed that intra-articular administration of AnxA5 ameliorated pain symptoms in a monosodium iodoacetate-induced osteoarthritis rat model. Histological analyses indicated a decrease in synovial inflammation and mitigation of cartilage damage following AnxA5 treatment. These results underscored the potential of AnxA5 as a therapeutic option for OA due to its capacity to regulate macrophage polarization and maintain chondrocyte viability. Further investigation into the specific mechanisms and clinical applications of AnxA5 may help improve the management of OA.


Sujet(s)
Annexine A5 , Chondrocytes , Macrophages , Arthrose , Rat Sprague-Dawley , Animaux , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/induit chimiquement , Rats , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Annexine A5/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Mâle , Récepteur de type Toll-4/métabolisme , Souris , Cellules RAW 264.7 , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques
18.
Cell Commun Signal ; 22(1): 335, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890746

RÉSUMÉ

OBJECTIVE: Kappa opioid receptor (KOR) signaling is involved in joint development and inflammation in Osteoarthritis (OA), while the biochemical mechanism remains unclarified. This study aims to investigate downstream molecular events of KOR activation, to provide novel perspectives in OA pathology. METHODS: U50,488H, a selective KOR agonist, was intra-articularly injected in mice upon destabilization of the medial meniscus (DMM) as OA models, with PBS injection as control. The behavioral and histological evaluation was assessed by hot plate test and red solid green staining, respectively. Alterations in mRNA and protein expression were assessed by RNA-seq, RT-qPCR, immunohistochemistry and western blotting (WB) in chondrocytes treated with TNF-α or TNF-α + U50,488H. Proteins interacted with KOR were explored using proximity labeling followed by mass spectrometry and then testified by co-immunoprecipitation (Co-IP) assay and immunofluorescence (IF). RESULTS: OA-induced pain was reduced and cartilage degeneration was alleviated upon KOR activation in DMM mice. In chondrocytes, activation of KOR reversed the upregulation of MMPs, IL-6, IL-1ß and phosphorylated(p-) STAT3, stimulated by TNF-α, while the expression of NF-κB, MAPKs and AKT signaling weren't reversed. RNA-seq and IF results presented that KOR activation evidently reduced STAT3 nuclear translocation in chondrocytes upon TNF-α stimuli. The reduction may be resulted from the binding of KOR and STAT3 in the plasma membrane, revealed by proximity labeling and Co-IP results. CONCLUSIONS: KOR activation protects cartilage from OA, and this protective effect is mainly exerted via sequestering STAT3 on the plasma membrane, resulting in inactivation of STAT3-dependent immune responses which otherwise contributes to OA.


Sujet(s)
Membrane cellulaire , Chondrocytes , Arthrose , Récepteur kappa , Facteur de transcription STAT-3 , Animaux , Mâle , Souris , 2-(3,4-Dichlorophényl)-N-méthyl-N-((1S,2S)-2-(pyrrolidin-1-yl)cyclohexyl)acétamide/pharmacologie , Membrane cellulaire/métabolisme , Membrane cellulaire/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Chondrocytes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Arthrose/anatomopathologie , Arthrose/métabolisme , Récepteur kappa/métabolisme , Récepteur kappa/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme
19.
J Mater Chem B ; 12(25): 6242-6256, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38842217

RÉSUMÉ

Designing artificial nano-enzymes for scavenging reactive oxygen species (ROS) in chondrocytes (CHOs) is considered the most feasible pathway for the treatment of osteoarthritis (OA). However, the accumulation of ROS due to the amount of nano-enzymatic catalytic site exposure and insufficient oxygen supply seriously threatens the clinical application of this therapy. Although metal-organic framework (MOF) immobilization of artificial nano-enzymes to enhance active site exposure has been extensively studied, artificial nano-enzymes/MOFs for ROS scavenging in OA treatment are still lacking. In this study, a biocompatible lubricating hydrogel-loaded iron-doped zeolitic imidazolate framework-8 (Fe/ZIF-8/Gel) centrase was engineered to scavenge endogenous overexpressed ROS synergistically generating dissolved oxygen and enhancing sustained lubrication for CHOs as a ternary artificial nano-enzyme. This property enabled the nano-enzymatic hydrogels to mitigate OA hypoxia and inhibit oxidative stress damage successfully. Ternary strategy-based therapies show excellent cartilage repair in vivo. The experimental results suggest that nano-enzyme-enhanced lubricating hydrogels are a potentially effective OA treatment and a novel strategy.


Sujet(s)
Chondrocytes , Hydrogels , Espèces réactives de l'oxygène , Hydrogels/composition chimique , Hydrogels/pharmacologie , Animaux , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/cytologie , Espèces réactives de l'oxygène/métabolisme , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Arthrose/traitement médicamenteux , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Cartilage/effets des médicaments et des substances chimiques , Cartilage/métabolisme , Taille de particule , Humains , Zéolites/composition chimique
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE