Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22.044
Filtrer
1.
FASEB J ; 38(13): e23778, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38959010

RÉSUMÉ

The mechanosensitive ion channels Transient Receptor Potential Vanilloid 4 (TRPV4) and PIEZO1 transduce physiologic and supraphysiologic magnitudes of mechanical signals in the chondrocyte, respectively. TRPV4 activation promotes chondrogenesis, while PIEZO1 activation by supraphysiologic deformations drives cell death. The mechanisms by which activation of these channels discretely drives changes in gene expression to alter cell behavior remain to be determined. To date, no studies have contrasted the transcriptomic response to activation of these channels nor has any published data attempted to correlate these transcriptomes to alterations in cellular function. This study used RNA sequencing to comprehensively investigate the transcriptomes associated with activation of TRPV4 or PIEZO1, revealing that TRPV4 and PIEZO drive distinct transcriptomes and also exhibit unique co-regulated clusters of genes. Notably, activation of PIEZO1 through supraphysiologic deformation induced a transient inflammatory profile that overlapped with the interleukin (IL)-1-responsive transcriptome and contained genes associated with cartilage degradation and osteoarthritis progression. However, both TRPV4 and PIEZO1 were also shown to elicit anabolic effects. PIEZO1 expression promoted a pro-chondrogenic transcriptome under unloaded conditions, and daily treatment with PIEZO1 agonist Yoda1 significantly increased sulfated glycosaminoglycan deposition in vitro. These findings emphasize the presence of a broad "mechanome" with distinct effects of TRPV4 and PIEZO1 activation in chondrocytes, suggesting complex roles for PIEZO1 in both the physiologic and pathologic responses of chondrocytes. The identification of transcriptomic profiles unique to or shared by PIEZO1 and TRPV4 (distinct from IL-1-induced inflammation) could inform future therapeutic designs targeting these channels for the management and treatment of osteoarthritis.


Sujet(s)
Chondrocytes , Canaux ioniques , Canaux cationiques TRPV , Transcriptome , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique , Chondrocytes/métabolisme , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Animaux , Mécanotransduction cellulaire , Souris , Chondrogenèse , Humains
2.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(7): 807-811, 2024 Jul 10.
Article de Chinois | MEDLINE | ID: mdl-38946362

RÉSUMÉ

OBJECTIVE: To analyze the genetic variant and molecular pathogenesis in a Chinese pedigree affected with Multiple epiphyseal dysplasia (MED). METHODS: A MED pedigree which had presented at the Beijing Jishuitan Hospital Affiliated to Capital Medical University on September 13, 2020 was selected as the study subject. Clinical data of the pedigree were collected. Peripheral blood samples were drawn from pedigree members for the extraction of genomic DNA. Whole exome sequencing (WES) was carried out for the pedigree. Candidate variant was verified by Sanger sequencing. Wild type and mutant SLC26A2 expression plasmids were constructed and transfected into human primary chondrocytes. The effect of the variants on the protein localization and cell proliferation was determined by immunofluorescence and CCK8 assays. RESULTS: WES and Sanger sequencing revealed that the proband has harbored compound heterozygous variants of the SLC26A2 gene, including a paternally derived c.484G>T (p.Val162Leu) missense variant and a maternally derived c.485_486delTG (p.Val162Glyfs*12) frameshifting variant. The SLC26A2WT and its mutant SLC26A2Val162Leu and SLC26A2Val162Glyfs*12 expression plasmids were distributed in the nuclei and cytoplasm of human primary chondrocytes. Compared with SLC26A2WT, the expressions of SLC26A2Val162Leu and SLC26A2Val162Glyfs*12 were decreased, along with reduced proliferation of human primary chondrocytes. CONCLUSION: The c.484G>T and c.485_486delTG compound heterozygous variants of the SLC26A2 gene may affect the proliferation of human primary chondrocytes and underlay the pathogenesis of MED in this pedigree.


Sujet(s)
Asiatiques , Ostéochondrodysplasies , Pedigree , Transporteurs de sulfate , Humains , Transporteurs de sulfate/génétique , Transporteurs de sulfate/métabolisme , Ostéochondrodysplasies/génétique , Mâle , Femelle , Asiatiques/génétique , Chondrocytes/métabolisme , , Adulte , Chine , Mutation , Variation génétique , Prolifération cellulaire , Peuples d'Asie de l'Est
3.
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
4.
Sci Rep ; 14(1): 15022, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951570

RÉSUMÉ

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


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Chondrogenèse , Hydrogels , Cellules souches mésenchymateuses , Ingénierie tissulaire , Protéine Wnt3A , Protéine Wnt3A/métabolisme , Chondrogenèse/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Humains , Cartilage/métabolisme , Gélatine/composition chimique , Structures d'échafaudage tissulaires/composition chimique , Facteur de croissance transformant bêta-3/métabolisme , Facteur de croissance transformant bêta-3/pharmacologie , Lignée cellulaire , Matrice extracellulaire/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/cytologie , Animaux
5.
Orphanet J Rare Dis ; 19(1): 245, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956600

RÉSUMÉ

BACKGROUND: Multiple epiphyseal dysplasia-4 (MED-4, MIM 226900) is a rare autosomal recessive disease characterized by disproportionate height and early onset osteoarthritis of the lower limbs. MED-4 is caused by homozygous or compound heterozygous pathogenic variants in the SLC26A2 gene. However, the underlying pathogenic mechanisms in chondrocytes remains unknown. This study aimed to identify the pathogenic variants within a MED-4 family and explore the molecular etiology of this condition in human primary chondrocyte cells. METHODS: Clinical data were recorded and peripheral blood samples were collected for analysis. Whole exome sequencing (WES) and bioinformatic analyses were performed to determine causative variants. Wild-type SLC26A2 and corresponding mutant expression plasmids were constructed and transfected into human primary chondrocytes. The expression and subcellular distribution of SLC26A2 protein in chondrocytes were detected by immunoblotting and immunofluorescence. Effects of these variants on chondrocytes viability and apoptosis were measured by Cell Counting Kit-8 (CCK-8) assay. Expression of genes related to cartilage homeostasis was subsequently analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: We identified two compound heterozygous variants c.1020_1022delTGT(p.Val341del) and c.1262 T > C(p.Ile421Thr) in the SLC26A2 gene in the patients. Mutant SLC26A2Val341del and SLC26A2Ile421Thr proteins were distributed in relatively few cells and were observed only within the nucleus. The viability of chondrocytes with the SLC26A2 variant group was similar to the wild-type (WT) group. However, the protein expressions of SLC26A2Val341del and SLC26A2Ile421Thr were decreased compared with SLC26A2WT. Expression levels of matrix metallopeptidase 13 (MMP13), α-1 chain of type X collagen (COL10A1), and Runt-related transcription factor 2 (RUNX2) were significantly decreased in the variant group. However, aggrecan (ACAN) expression was higher in the variant group than the WT group. CONCLUSIONS: Overall, our data demonstrate that the variants p.Val341del and p.Ile421Thr in SLC26A2 cause MED-4 and that these two variants promote chondrocyte proliferation while inhibiting chondrocyte differentiation.


Sujet(s)
Chondrocytes , Ostéochondrodysplasies , Transporteurs de sulfate , Humains , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Transporteurs de sulfate/génétique , Transporteurs de sulfate/métabolisme , Ostéochondrodysplasies/génétique , Ostéochondrodysplasies/métabolisme , Ostéochondrodysplasies/anatomopathologie , Mâle , Femelle , Homéostasie/génétique ,
6.
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
7.
Immun Inflamm Dis ; 12(6): e1334, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38938021

RÉSUMÉ

OBJECTIVE: To explore the efficacy and potential mechanism of Fengshi Gutong capsule (FSGTC) in osteoarthritis (OA) inflammation. METHODS: The impact of FSGTC on laboratory indicators of OA patients was explored using data mining technology and association rule analysis. Then, the OA cell model was constructed by inducing chondrocytes (CHs) with interleukin-1ß (IL-1ß). In the presence of FSGTC intervention, the regulatory mechanism of PACER/COX2/PGE2 in OA-CH viability and inflammatory responses was evaluated. RESULTS: Retrospective data mining showed that FSGTC effectively reduced inflammation indexes (ESR, HCRP) of OA patients. Cell experiments showed that LncRNA PACER (PACER) silencing inhibited the proliferation activity of OA-CHs, increased the level of COX2 protein, elevated the levels of PGE2, TNF-α, and IL-1ß, and decreased the levels of IL-4 and IL-10 (p < .01). On the contrary, FSGTC-containing serum reversed the effect of PACER silencing on OA-CHs (p < .01). After the addition of COX2 pathway inhibitor, the proliferation activity of OA-CHs was enhanced; the levels of PGE2, TNF-α, and IL-1ß were decreased while the levels of IL-4 and IL-10 were increased (p < .01). CONCLUSION: FSGTC inhibits IL-1ß-induced inflammation in CHs and ameliorates OA by upregulating PACER and downregulating COX2/PGE2.


Sujet(s)
Chondrocytes , Cyclooxygenase 2 , Dinoprostone , Inflammation , Interleukine-1 bêta , Arthrose , ARN long non codant , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , ARN long non codant/génétique , Humains , Interleukine-1 bêta/métabolisme , Cyclooxygenase 2/métabolisme , Cyclooxygenase 2/génétique , Dinoprostone/métabolisme , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Médicaments issus de plantes chinoises/pharmacologie , Régulation négative , Mâle , Femelle , Régulation positive , Adulte d'âge moyen
8.
Stem Cell Res Ther ; 15(1): 185, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926793

RÉSUMÉ

Cartilage, an important connective tissue, provides structural support to other body tissues, and serves as a cushion against impacts throughout the body. Found at the end of the bones, cartilage decreases friction and averts bone-on-bone contact during joint movement. Therefore, defects of cartilage can result from natural wear and tear, or from traumatic events, such as injuries or sudden changes in direction during sports activities. Overtime, these cartilage defects which do not always produce immediate symptoms, could lead to severe clinical pathologies. The emergence of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine, providing a promising platform for generating various cell types for therapeutic applications. Thus, chondrocytes differentiated from iPSCs become a promising avenue for non-invasive clinical interventions for cartilage injuries and diseases. In this review, we aim to highlight the current strategies used for in vitro chondrogenic differentiation of iPSCs and to explore their multifaceted applications in disease modeling, drug screening, and personalized regenerative medicine. Achieving abundant functional iPSC-derived chondrocytes requires optimization of culture conditions, incorporating specific growth factors, and precise temporal control. Continual improvements in differentiation methods and integration of emerging genome editing, organoids, and 3D bioprinting technologies will enhance the translational applications of iPSC-derived chondrocytes. Finally, to unlock the benefits for patients suffering from cartilage diseases through iPSCs-derived technologies in chondrogenesis, automatic cell therapy manufacturing systems will not only reduce human intervention and ensure sterile processes within isolator-like platforms to minimize contamination risks, but also provide customized production processes with enhanced scalability and efficiency.


Sujet(s)
Différenciation cellulaire , Chondrogenèse , Cellules souches pluripotentes induites , Médecine de précision , Médecine régénérative , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Médecine régénérative/méthodes , Médecine de précision/méthodes , Chondrocytes/cytologie , Chondrocytes/métabolisme , Animaux
9.
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
10.
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
11.
Cells ; 13(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38920652

RÉSUMÉ

Mesenchymal stem cells (MSCs) of placental origin hold great promise in tissue engineering and regenerative medicine for diseases affecting cartilage and bone. However, their utility has been limited by their tendency to undergo premature senescence and phenotypic drift into adipocytes. This study aimed to explore the potential involvement of a specific subset of aging and antiaging genes by measuring their expression prior to and following in vitro-induced differentiation of placental MSCs into chondrocytes and osteoblasts as opposed to adipocytes. The targeted genes of interest included the various LMNA/C transcript variants (lamin A, lamin C, and lamin A∆10), sirtuin 7 (SIRT7), and SM22α, along with the classic aging markers plasminogen activator inhibitor 1 (PAI-1), p53, and p16INK4a. MSCs were isolated from the decidua basalis of human term placentas, expanded, and then analyzed for phenotypic properties by flow cytometry and evaluated for colony-forming efficiency. The cells were then induced to differentiate in vitro into chondrocytes, osteocytes, and adipocytes following established protocols. The mRNA expression of the targeted genes was measured by RT-qPCR in the undifferentiated cells and those fully differentiated into the three cellular lineages. Compared to undifferentiated cells, the differentiated chondrocytes demonstrated decreased expression of SIRT7, along with decreased PAI-1, lamin A, and SM22α expression, but the expression of p16INK4a and p53 increased, suggesting their tendency to undergo premature senescence. Interestingly, the cells maintained the expression of lamin C, which indicates that it is the primary lamin variant influencing the mechanoelastic properties of the differentiated cells. Notably, the expression of all targeted genes did not differ from the undifferentiated cells following osteogenic differentiation. On the other hand, the differentiation of the cells into adipocytes was associated with decreased expression of lamin A and PAI-1. The distinct patterns of expression of aging and antiaging genes following in vitro-induced differentiation of MSCs into chondrocytes, osteocytes, and adipocytes potentially reflect specific roles for these genes during and following differentiation in the fully functional cells. Understanding these roles and the network of signaling molecules involved can open opportunities to improve the handling and utility of MSCs as cellular precursors for the treatment of cartilage and bone diseases.


Sujet(s)
Différenciation cellulaire , Chondrogenèse , Cellules souches mésenchymateuses , Ostéogenèse , Placenta , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Femelle , Placenta/métabolisme , Placenta/cytologie , Différenciation cellulaire/génétique , Chondrogenèse/génétique , Grossesse , Ostéogenèse/génétique , Marqueurs biologiques/métabolisme , Vieillissement de la cellule/génétique , Chondrocytes/métabolisme , Chondrocytes/cytologie , Vieillissement , Lamine A/métabolisme , Lamine A/génétique
12.
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
13.
Eur J Cell Biol ; 103(2): 151424, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38823166

RÉSUMÉ

Actin is a central mediator of the chondrocyte phenotype. Monolayer expansion of articular chondrocytes on tissue culture polystyrene, for cell-based repair therapies, leads to chondrocyte dedifferentiation. During dedifferentiation, chondrocytes spread and filamentous (F-)actin reorganizes from a cortical to a stress fiber arrangement causing a reduction in cartilage matrix expression and an increase in fibroblastic matrix and contractile molecule expression. While the downstream mechanisms regulating chondrocyte molecular expression by alterations in F-actin organization have become elucidated, the critical upstream regulators of F-actin networks in chondrocytes are not completely known. Tropomyosin (TPM) and the RhoGTPases are known regulators of F-actin networks. The main purpose of this study is to elucidate the regulation of passaged chondrocyte F-actin stress fiber networks and cell phenotype by the specific TPM, TPM3.1, and the RhoGTPase, CDC42. Our results demonstrated that TPM3.1 associates with cortical F-actin and stress fiber F-actin in primary and passaged chondrocytes, respectively. In passaged cells, we found that pharmacological TPM3.1 inhibition or siRNA knockdown causes F-actin reorganization from stress fibers back to cortical F-actin and causes an increase in G/F-actin. CDC42 inhibition also causes formation of cortical F-actin. However, pharmacological CDC42 inhibition, but not TPM3.1 inhibition, leads to the re-association of TPM3.1 with cortical F-actin. Both TPM3.1 and CDC42 inhibition, as well as TPM3.1 knockdown, reduces nuclear localization of myocardin related transcription factor, which suppresses dedifferentiated molecule expression. We confirmed that TPM3.1 or CDC42 inhibition partially redifferentiates passaged cells by reducing fibroblast matrix and contractile expression, and increasing chondrogenic SOX9 expression. A further understanding on the regulation of F-actin in passaged cells may lead into new insights to stimulate cartilage matrix expression in cells for regenerative therapies.


Sujet(s)
Actines , Dédifférenciation cellulaire , Chondrocytes , Fibres de stress , Tropomyosine , Chondrocytes/métabolisme , Chondrocytes/cytologie , Fibres de stress/métabolisme , Animaux , Actines/métabolisme , Tropomyosine/métabolisme , Tropomyosine/génétique , Phénotype , Cellules cultivées , Protéine G cdc42/métabolisme , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique
14.
J Cell Mol Med ; 28(11): e18476, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38842136

RÉSUMÉ

Osteoarthritis (OA) is a complicated disease that involves apoptosis and mitophagy. MST1 is a pro-apoptotic factor. Hence, decreasing its expression plays an anti-apoptotic effect. This study aims to investigate the protective effect of MST1 inhibition on OA and the underlying processes. Immunofluorescence (IF) was used to detect MST1 expression in cartilage tissue. Western Blot, ELISA and IF were used to analyse the expression of inflammation, extracellular matrix (ECM) degradation, apoptosis and mitophagy-associated proteins. MST1 expression in chondrocytes was inhibited using siRNA and shRNA in vitro and in vivo. Haematoxylin-Eosin, Safranin O-Fast Green and alcian blue staining were used to evaluate the therapeutic effect of inhibiting MST1. This study discovered that the expression of MST1 was higher in OA patients. Inhibition of MST1 reduced inflammation, ECM degradation and apoptosis and enhanced mitophagy in vitro. MST1 inhibition slows OA progression in vivo. Inhibiting MST1 suppressed apoptosis, inflammation and ECM degradation via promoting Parkin-mediated mitophagy and the Nrf2-NF-κB axis. The results suggest that MST1 is a possible therapeutic target for the treatment of osteoarthritis as its inhibition delays the progression of OA through the Nrf2-NF-κB axis and mitophagy.


Sujet(s)
Apoptose , Chondrocytes , Évolution de la maladie , Mitophagie , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Arthrose , Transduction du signal , Ubiquitin-protein ligases , Animaux , Humains , Mâle , Souris , Apoptose/génétique , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Matrice extracellulaire/métabolisme , Techniques de knock-down de gènes , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Protéines et peptides de signalisation intracellulaire , Mitophagie/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Facteur de transcription NF-kappa B/métabolisme , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
15.
Aging (Albany NY) ; 16(11): 9558-9568, 2024 05 31.
Article de Anglais | MEDLINE | ID: mdl-38829778

RÉSUMÉ

Osteoarthritis (OA) is one of the most important causes of global disability, and dysfunction of chondrocytes is an important risk factor. The treatment of OA is still a challenge. Orexin-A is a hypothalamic peptide, and its effects in OA are unknown. In this study, we found that exposure to interleukin-1ß (IL-1ß) reduced the expression of orexin-2R, the receptor of orexin-A in TC-28a2 chondrocytes. Importantly, the senescence-associated ß-galactosidase (SA-ß-gal) staining assay demonstrated that orexin-A treatment ameliorates IL-1ß-induced cellular senescence. Importantly, the presence of IL-1ß significantly reduced the telomerase activity of TC-28a2 chondrocytes, which was rescued by orexin-A. We also found that orexin-A prevented IL-1ß-induced increase in the levels of Acetyl-p53 and the expression of p21. It is shown that orexin-A mitigates IL-1ß-induced reduction of sirtuin 3 (SIRT3). Silencing of SIRT3 abolished the protective effects of orexin-A against IL-1ß-induced cellular senescence. These results imply that orexin-A might serve as a promising therapeutic agent for OA.


Sujet(s)
Vieillissement de la cellule , Chondrocytes , Interleukine-1 bêta , Orexines , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Orexines/pharmacologie , Orexines/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Humains , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Animaux , Protéine p53 suppresseur de tumeur/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Récepteurs des orexines/métabolisme , Récepteurs des orexines/génétique , Lignée cellulaire
16.
J Biomech ; 171: 112179, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38852482

RÉSUMÉ

Cell volume and shape changes play a pivotal role in cellular mechanotransduction, governing cellular responses to external loading. Understanding the dynamics of cell behavior under loading conditions is essential to elucidate cell adaptation mechanisms in physiological and pathological contexts. In this study, we investigated the effects of dynamic cyclic compression loading on cell volume and shape changes, comparing them with static conditions. Using a custom-designed platform which allowed for simultaneous loading and imaging of cartilage tissue, tissues were subjected to 100 cycles of mechanical loading while measuring cell volume and shape alterations during the unloading phase at specific time points. The findings revealed a transient decrease in cell volume (13%) during the early cycles, followed by a gradual recovery to baseline levels after approximately 20 cycles, despite the cartilage tissue not being fully recovered at the unloading phase. This observed pattern indicates a temporal cell volume response that may be associated with cellular adaptation to the mechanical stimulus through mechanisms related to active cell volume regulation. Additionally, this study demonstrated that cell volume and shape responses during dynamic loading were significantly distinct from those observed under static conditions. Such findings suggest that cells in their natural tissue environment perceive and respond differently to dynamic compared to static mechanical cues, highlighting the significance of considering dynamic loading environments in studies related to cellular mechanics. Overall, this research contributes to the broader understanding of cellular behavior under mechanical stimuli, providing valuable insights into their ability to adapt to dynamic mechanical loading.


Sujet(s)
Chondrocytes , Mise en charge , Animaux , Chondrocytes/physiologie , Mise en charge/physiologie , Contrainte mécanique , Taille de la cellule , Mécanotransduction cellulaire/physiologie , Résistance à la compression/physiologie , Bovins , Cartilage articulaire/physiologie , Forme de la cellule/physiologie
17.
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
18.
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
19.
Int J Biol Macromol ; 273(Pt 1): 132819, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38830498

RÉSUMÉ

The avascular nature of hyaline cartilage results in limited spontaneous self-repair and regenerative capabilities when damaged. Recent advances in three-dimensional bioprinting have enabled the precise dispensing of cell-laden biomaterials, commonly referred to as 'bioinks', which are emerging as promising solutions for tissue regeneration. An effective bioink for cartilage tissue engineering needs to create a micro-environment that promotes cell differentiation and supports neocartilage tissue formation. In this study, we introduced an innovative bioink composed of photocurable acrylated type I collagen (COLMA), thiol-modified hyaluronic acid (THA), and poly(ethylene glycol) diacrylate (PEGDA) for 3D bioprinting cartilage grafts using human nasal chondrocytes. Both collagen and hyaluronic acid, being key components of the extracellular matrix (ECM) in the human body, provide essential biological cues for tissue regeneration. We evaluated three formulations - COLMA, COLMA+THA, and COLMA+THA+PEGDA - for their printability, cell viability, structural integrity, and capabilities in forming cartilage-like ECM. The addition of THA and PEGDA significantly enhanced these properties, showcasing the potential of this bioink in advancing applications in cartilage repair and reconstructive surgery.


Sujet(s)
Acide hyaluronique , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Ingénierie tissulaire/méthodes , Humains , Structures d'échafaudage tissulaires/composition chimique , Chondrocytes/cytologie , Chondrocytes/effets des médicaments et des substances chimiques , Polyéthylène glycols/composition chimique , Bio-impression/méthodes , Collagène/composition chimique , Impression tridimensionnelle , Cartilage/cytologie , Matrice extracellulaire/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Encre
20.
Int J Biol Macromol ; 273(Pt 2): 133217, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38897519

RÉSUMÉ

Recent research focuses on fabricating scaffolds imitating the extracellular matrix (ECM) in texture, composition, and functionality. Moreover, specific nano-bio-particles can enhance cell differentiation. Decellularized ECM nanoparticles possess all of the mentioned properties. In this research, cartilage ECM, extracted from the cow's femur condyle, was decellularized, and ECM nanoparticles were synthesized. Finally, nanocomposite electrospun fibers containing polyhydroxybutyrate (PHB), chitosan (Cs) nanoparticles, and ECM nanoparticles were fabricated and characterized. TEM and DLS results revealed ECM nanoparticle sizes of 17.51 and 21.6 nm, respectively. Optimal performance was observed in the scaffold with 0.75 wt% ECM nanoparticles (PHB-Cs/0.75E). By adding 0.75 wt% ECM, the ultimate tensile strength and elongation at break increased by about 29 % and 21 %, respectively, while the water contact angle and crystallinity decreased by about 36° and 2 %, respectively. Uneven and rougher surfaces of the PHB-Cs/0.75E were determined by FESEM and AFM images, respectively. TEM images verified the uniform dispersion of nanoparticles within the fibers. After 70 days of degradation in PBS, the PHB-Cs/0.75E and PHB-Cs scaffolds demonstrated insignificant weight loss differences. Eventually, enhanced viability, attachment, and proliferation of the human costal chondrocytes on the PHB-Cs/0.75E scaffold, concluded from MTT, SEM, and DAPI staining, confirmed its potential for cartilage tissue engineering.


Sujet(s)
Cartilage , Chitosane , Matrice extracellulaire , Hydroxy-butyrates , Nanoparticules , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Chitosane/composition chimique , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires/composition chimique , Matrice extracellulaire/composition chimique , Matrice extracellulaire/métabolisme , Nanoparticules/composition chimique , Animaux , Hydroxy-butyrates/composition chimique , Cartilage/cytologie , Cartilage/métabolisme , Polyesters/composition chimique , Humains , Bovins , Chondrocytes/cytologie , Chondrocytes/métabolisme ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...