Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.542
1.
J Cell Mol Med ; 28(11): e18472, 2024 Jun.
Article En | MEDLINE | ID: mdl-38842129

Excessive load on the temporomandibular joint (TMJ) is a significant factor in the development of TMJ osteoarthritis, contributing to cartilage degeneration. The specific mechanism through which excessive load induces TMJ osteoarthritis is not fully understood; however, mechanically-activated (MA) ion channels play a crucial role. Among these channels, Piezo1 has been identified as a mediator of chondrocyte catabolic responses and is markedly increased in osteoarthritis. Our observations indicate that, under excessive load conditions, endoplasmic reticulum stress in chondrocytes results in apoptosis of the TMJ chondrocytes. Importantly, using the Piezo1 inhibitor GsMTx4 demonstrates its potential to alleviate this condition. Furthermore, Piezo1 mediates endoplasmic reticulum stress in chondrocytes by inducing calcium ion influx. Our research substantiates the role of Piezo1 as a pivotal ion channel in mediating chondrocyte overload. It elucidates the link between excessive load, cell apoptosis, and calcium ion influx through Piezo1. The findings underscore Piezo1 as a key player in the pathogenesis of TMJ osteoarthritis, shedding light on potential therapeutic interventions for this condition.


Apoptosis , Calcium , Chondrocytes , Endoplasmic Reticulum Stress , Ion Channels , Osteoarthritis , Temporomandibular Joint , Chondrocytes/metabolism , Chondrocytes/pathology , Ion Channels/metabolism , Ion Channels/genetics , Animals , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Calcium/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Humans , Mice , Signal Transduction , Spider Venoms , Intercellular Signaling Peptides and Proteins
2.
J Cell Mol Med ; 28(11): e18476, 2024 Jun.
Article En | MEDLINE | ID: mdl-38842136

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.


Apoptosis , Chondrocytes , Disease Progression , Mitophagy , NF-E2-Related Factor 2 , NF-kappa B , Osteoarthritis , Signal Transduction , Ubiquitin-Protein Ligases , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/genetics , Mitophagy/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Apoptosis/genetics , Male , Mice , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins
3.
Front Immunol ; 15: 1361606, 2024.
Article En | MEDLINE | ID: mdl-38846937

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


Exosomes , Macrophages , MicroRNAs , Osteoarthritis , Synoviocytes , TNF Receptor-Associated Factor 6 , MicroRNAs/genetics , Animals , Exosomes/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/immunology , Rats , Macrophages/immunology , Macrophages/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , Male , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , NF-kappa B/metabolism , Signal Transduction , Rats, Sprague-Dawley , Fibroblasts/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/immunology , Cells, Cultured , Apoptosis , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Macrophage Activation
4.
BMC Musculoskelet Disord ; 25(1): 447, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844896

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.


Apoptosis , Arthritis, Experimental , Chondrocytes , Inflammation , Osteoarthritis , Vitamin B 6 , Animals , Apoptosis/drug effects , Mice , Vitamin B 6/pharmacology , Vitamin B 6/therapeutic use , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Osteoarthritis/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Mice, Inbred DBA , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism
5.
BMC Musculoskelet Disord ; 25(1): 451, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844905

OBJECTIVE: Temporomandibular joint osteoarthritis (TMJOA) is a chronic degenerative joint disorder characterized by extracellular matrix degeneration and inflammatory response of condylar cartilage. ß-arrestin2 is an important regulator of inflammation response, while its role in TMJOA remains unknown. The objective of this study was to investigate the role of ß-arrestin2 in the development of TMJOA at the early stage and the underlying mechanism. METHODS: A unilateral anterior crossbite (UAC) model was established on eight-week-old wild-type (WT) and ß-arrestin2 deficiency mice to simulate the progression of TMJOA. Hematoxylin-eosin (HE) staining and microcomputed tomography (micro-CT) analysis were used for histological and radiographic assessment. Immunohistochemistry was performed to detect the expression of inflammatory and degradative cytokines, as well as autophagy related factors. Terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) assay was carried out to assess chondrocyte apoptosis. RESULTS: The loss of ß-arrestin2 aggravated cartilage degeneration and subchondral bone destruction in the model of TMJOA at the early stage. Furthermore, in UAC groups, the expressions of degradative (Col-X) and inflammatory (TNF-α and IL-1ß) factors in condylar cartilage were increased in ß-arrestin2 null mice compared with WT mice. Moreover, the loss of ß-arrestin2 promoted apoptosis and autophagic process of chondrocytes at the early stage of TMJOA. CONCLUSION: In conclusion, we demonstrated for the first time that ß-arrestin2 plays a protective role in the development of TMJOA at the early stage, probably by inhibiting apoptosis and autophagic process of chondrocytes. Therefore, ß-arrestin2 might be a potential therapeutic target for TMJOA, providing a new insight for the treatment of TMJOA at the early stage.


Cartilage, Articular , Disease Models, Animal , Mandibular Condyle , Mice, Knockout , Osteoarthritis , Temporomandibular Joint Disorders , beta-Arrestin 2 , Animals , Osteoarthritis/metabolism , Osteoarthritis/pathology , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Mandibular Condyle/pathology , Mandibular Condyle/metabolism , Mandibular Condyle/diagnostic imaging , Mice , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint Disorders/etiology , Chondrocytes/metabolism , Chondrocytes/pathology , Mice, Inbred C57BL , Apoptosis , Temporomandibular Joint/pathology , Temporomandibular Joint/metabolism , Temporomandibular Joint/diagnostic imaging , Male , X-Ray Microtomography , Autophagy/physiology
6.
PeerJ ; 12: e17417, 2024.
Article En | MEDLINE | ID: mdl-38827307

Background: Osteoarthritis (OA) is a degenerative disease requiring additional research. This study compared gene expression and immune infiltration between lesioned and preserved subchondral bone. The results were validated using multiple tissue datasets and experiments. Methods: Differentially expressed genes (DEGs) between the lesioned and preserved tibial plateaus of OA patients were identified in the GSE51588 dataset. Moreover, functional annotation and protein-protein interaction (PPI) network analyses were performed on the lesioned and preserved sides to explore potential therapeutic targets in OA subchondral bones. In addition, multiple tissues were used to screen coexpressed genes, and the expression levels of identified candidate DEGs in OA were measured by quantitative real-time polymerase chain reaction. Finally, an immune infiltration analysis was conducted. Results: A total of 1,010 DEGs were identified, 423 upregulated and 587 downregulated. The biological process (BP) terms enriched in the upregulated genes included "skeletal system development", "sister chromatid cohesion", and "ossification". Pathways were enriched in "Wnt signaling pathway" and "proteoglycans in cancer". The BP terms enriched in the downregulated genes included "inflammatory response", "xenobiotic metabolic process", and "positive regulation of inflammatory response". The enriched pathways included "neuroactive ligand-receptor interaction" and "AMP-activated protein kinase signaling". JUN, tumor necrosis factor α, and interleukin-1ß were the hub genes in the PPI network. Collagen XI A1 and leucine-rich repeat-containing 15 were screened from multiple datasets and experimentally validated. Immune infiltration analyses showed fewer infiltrating adipocytes and endothelial cells in the lesioned versus preserved samples. Conclusion: Our findings provide valuable information for future studies on the pathogenic mechanism of OA and potential therapeutic and diagnostic targets.


Protein Interaction Maps , Humans , Gene Expression Profiling , Osteoarthritis/genetics , Osteoarthritis/immunology , Osteoarthritis/pathology , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/metabolism , Male , Tibia/pathology , Tibia/immunology , Tibia/metabolism , Down-Regulation , Female
7.
Mol Med ; 30(1): 74, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38831316

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage destruction and inflammation. CC chemokine receptor 1 (CCR1), a member of the chemokine family and its receptor family, plays a role in the autoimmune response. The impact of BX471, a specific small molecule inhibitor of CCR1, on CCR1 expression in cartilage and its effects on OA remain underexplored. METHODS: This study used immunohistochemistry (IHC) to assess CCR1 expression in IL-1ß-induced mouse chondrocytes and a medial meniscus mouse model of destabilization of the medial meniscus (DMM). Chondrocytes treated with varying concentrations of BX471 for 24 h were subjected to IL-1ß (10 ng/ml) treatment. The levels of the aging-related genes P16INK4a and P21CIP1 were analyzed via western blotting, and senescence-associated ß-galactosidase (SA-ß-gal) activity was measured. The expression levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), aggrecan (AGG), and the transcription factor SOX9 were determined through western blotting and RT‒qPCR. Collagen II, matrix metalloproteinase 13 (MMP13), and peroxisome proliferator-activated receptor (PPAR)-γ expression was analyzed via western blot, RT‒qPCR, and immunofluorescence. The impact of BX471 on inflammatory metabolism-related proteins under PPAR-γ inhibition conditions (using GW-9662) was examined through western blotting. The expression of MAPK signaling pathway-related molecules was assessed through western blotting. In vivo, various concentrations of BX471 or an equivalent medium were injected into DMM model joints. Cartilage destruction was evaluated through Safranin O/Fast green and hematoxylin-eosin (H&E) staining. RESULTS: This study revealed that inhibiting CCR1 mitigates IL-1ß-induced aging, downregulates the expression of iNOS, COX-2, and MMP13, and alleviates the IL-1ß-induced decrease in anabolic indices. Mechanistically, the MAPK signaling pathway and PPAR-γ may be involved in inhibiting the protective effect of CCR1 on chondrocytes. In vivo, BX471 protected cartilage in a DMM model. CONCLUSION: This study demonstrated the expression of CCR1 in chondrocytes. Inhibiting CCR1 reduced the inflammatory response, alleviated cartilage aging, and retarded degeneration through the MAPK signaling pathway and PPAR-γ, suggesting its potential therapeutic value for OA.


Chondrocytes , Disease Models, Animal , Osteoarthritis , PPAR gamma , Receptors, CCR1 , Animals , Mice , Osteoarthritis/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/pathology , PPAR gamma/metabolism , Chondrocytes/metabolism , Chondrocytes/drug effects , Receptors, CCR1/metabolism , Receptors, CCR1/antagonists & inhibitors , Male , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Cyclooxygenase 2/metabolism , Nitric Oxide Synthase Type II/metabolism
8.
Sci Rep ; 14(1): 10568, 2024 05 08.
Article En | MEDLINE | ID: mdl-38719877

Early diagnosis and treatment of pre- and early-stage osteoarthritis (OA) is important. However, the cellular and cartilaginous changes occurring during these stages remain unclear. We investigated the histological and immunohistochemical changes over time between pre- and early-stage OA in a rat model of traumatic injury. Thirty-six male rats were divided into two groups, control and OA groups, based on destabilization of the medial meniscus. Histological and immunohistochemical analyses of articular cartilage were performed on days 1, 3, 7, 10, and 14 postoperatively. Cell density of proteins associated with cartilage degradation increased from postoperative day one. On postoperative day three, histological changes, including chondrocyte death, reduced matrix staining, and superficial fibrillation, were observed. Simultaneously, a compensatory increase in matrix staining was observed. The Osteoarthritis Research Society International score increased from postoperative day seven, indicating thinner cartilage. On postoperative day 10, the positive cell density decreased, whereas histological changes progressed with fissuring and matrix loss. The proteoglycan 4-positive cell density increased on postoperative day seven. These findings will help establish an experimental model and clarify the mechanism of the onset and progression of pre- and early-stage traumatic OA.


Cartilage, Articular , Disease Models, Animal , Disease Progression , Immunohistochemistry , Osteoarthritis , Animals , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Male , Rats , Osteoarthritis/pathology , Osteoarthritis/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Rats, Sprague-Dawley , Proteoglycans/metabolism
9.
Sci Rep ; 14(1): 10232, 2024 05 03.
Article En | MEDLINE | ID: mdl-38702404

This study aimed to quantitatively assess three-dimensional changes in the mandibular condyle with osteoarthritis using cone-beam computed tomography (CBCT). Pre- and post-treatment CBCT images of temporomandibular joints (TMJs) from 66 patients were used to assess longitudinal changes in condylar volume within individual patients using 3D slicer software. Total volume difference (dV), net increase (dV + , bone deposition), and net decrease (dV- , bone resorption) after treatment were analyzed based on clinical and radiological factors. Condyles with surface erosion at their first visit showed significantly decreased volume after treatment compared to condyles without erosion (p < 0.05). Amounts of bone resorption and deposition were higher in condyles with surface erosion (both p < 0.01). In patients with condylar erosion, the presence of joint pain was associated with a decrease in condylar volume and an increase in net resorption (both p < 0.01). When both joint pain and condylar erosion were present, patients with parafunctional habits showed reduced condylar volume after treatment (p < 0.05). Condylar volume change after treatment was negatively correlated with the duration of pain relief (R = - 0.501, p < 0.05). These results indicate that condylar erosion and TMJ pain could be significant variables affecting TMJ volume changes after treatment. Establishing appropriate treatment strategies is crucial for managing condylar erosion and TMJ pain.


Cone-Beam Computed Tomography , Mandibular Condyle , Osteoarthritis , Humans , Cone-Beam Computed Tomography/methods , Female , Male , Mandibular Condyle/diagnostic imaging , Mandibular Condyle/pathology , Osteoarthritis/diagnostic imaging , Osteoarthritis/pathology , Middle Aged , Adult , Temporomandibular Joint/diagnostic imaging , Temporomandibular Joint/pathology , Aged , Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint Disorders/pathology , Imaging, Three-Dimensional/methods
10.
Front Cell Infect Microbiol ; 14: 1401963, 2024.
Article En | MEDLINE | ID: mdl-38803575

The understanding of the link between the gut-bone axis is growing yearly, but the mechanisms involved are not yet clear. Our study analyzed the role of Sestrin2 (SESN2)pathway in the gut-bone axis. We established an osteoarthritis (OA) model in Sprague-Dawley (SD) rats using the anterior cruciate ligament transection (ACLT) procedure, followed by a dietary intervention with varying levels of dietary fiber content for 8 weeks. By 16S rRNA sequencing of the gut microbiota, we found that high dietary fiber (HDF) intake could significantly increase the Bacillota-dominant gut microbiota. Meanwhile, enzyme linked immunosorbent assay (ELISA) and histological analysis showed that intervention with HDF could reduce the degree of bone and joint lesions and inflammation. We hypothesize that HDF increased the dominant flora of Bacillota, up-regulated the expression of SESN2 in knee joint, and reduced gut permeability, thereby reducing systemic inflammatory response and the degree of bone and joint lesions. Therefore, the present study confirms that changes in gut microbiota induced by increased dietary fiber intake delayed the onset of OA by promoting up-regulation of SESN2 expression at the knee joint to maintain chondrocyte activity and reduce synovial inflammation.


Chondrocytes , Dietary Fiber , Disease Models, Animal , Gastrointestinal Microbiome , Osteoarthritis , Rats, Sprague-Dawley , Animals , Chondrocytes/metabolism , Osteoarthritis/microbiology , Osteoarthritis/pathology , Rats , Male , RNA, Ribosomal, 16S/genetics , Knee Joint/microbiology , Knee Joint/pathology
11.
BMC Oral Health ; 24(1): 569, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745274

BACKGROUND: Extracellular matrix (ECM) protein malfunction or defect may lead to temporomandibular joint osteoarthritis (TMJ OA). Dentin sialophophoprotein (DSPP) is a mandibular condylar cartilage ECM protein, and its deletion impacted cell proliferation and other extracellular matrix alterations of postnatal condylar cartilage. However, it remains unclear if long-term loss of function of DSPP leads to TMJ OA. The study aimed to test the hypothesis that long-term haploinsufficiency of DSPP causes TMJ OA. MATERIALS AND METHODS: To determine whether Dspp+/- mice exhibit TMJ OA but no severe tooth defects, mandibles of wild-type (WT), Dspp+/-, and Dspp homozygous (Dspp-/-) mice were analyzed by Micro-computed tomography (micro-CT). To characterize the progression and possible mechanisms of osteoarthritic degeneration over time in Dspp+/- mice over time, condyles of Dspp+/- and WT mice were analyzed radiologically, histologically, and immunohistochemically. RESULTS: Micro-CT and histomorphometric analyses revealed that Dspp+/- and Dspp-/- mice had significantly lower subchondral bone mass, bone volume fraction, bone mineral density, and trabecular thickness compared to WT mice at 12 months. Interestingly, in contrast to Dspp-/- mice which exhibited tooth loss, Dspp+/- mice had minor tooth defects. RNA sequencing data showed that haplodeficency of DSPP affects the biological process of ossification and osteoclast differentiation. Additionally, histological analysis showed that Dspp+/- mice had condylar cartilage fissures, reduced cartilage thickness, decreased articular cell numbers and severe subchondral bone cavities, and with signs that were exaggerated with age. Radiographic data showed an increase in subchondral osteoporosis up to 18 months and osteophyte formation at 21 months. Moreover, Dspp+/- mice showed increased distribution of osteoclasts in the subchondral bone and increased expression of MMP2, IL-6, FN-1, and TLR4 in the mandibular condylar cartilage. CONCLUSIONS: Dspp+/- mice exhibit TMJ OA in a time-dependent manner, with lesions in the mandibular condyle attributed to hypomineralization of subchondral bone and breakdown of the mandibular condylar cartilage, accompanied by upregulation of inflammatory markers.


Extracellular Matrix Proteins , Osteoarthritis , Phosphoproteins , Sialoglycoproteins , Temporomandibular Joint Disorders , X-Ray Microtomography , Animals , Osteoarthritis/pathology , Osteoarthritis/diagnostic imaging , Osteoarthritis/genetics , Mice , Extracellular Matrix Proteins/metabolism , Temporomandibular Joint Disorders/pathology , Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint Disorders/etiology , Temporomandibular Joint Disorders/genetics , Phosphoproteins/genetics , Mandibular Condyle/pathology , Mandibular Condyle/diagnostic imaging , Temporomandibular Joint/pathology , Temporomandibular Joint/diagnostic imaging
12.
J Orthop Surg (Hong Kong) ; 32(2): 10225536241254588, 2024.
Article En | MEDLINE | ID: mdl-38758016

PURPOSE: The abnormal function and survival of chondrocytes result in articular cartilage failure, which may accelerate the onset and development of osteoarthritis (OA). This study is aimed to investigate the role of LINC01094 in chondrocyte apoptosis. METHODS: The viability and apoptosis of lipopolysaccharide (LPS)-induced chondrocytes were evaluated through CCK-8 assay and flow cytometry analysis, respectively. The expression levels of LINC01094, miR-577 and MTF1 were detected by qRT-PCR. Dual luciferase reporter tests were implemented for the verification of targeted relationships among them. Western blotting was employed to measure the levels of pro-apoptotic proteins (Caspase3 and Caspase9). RESULTS: The viability of LPS-induced chondrocytes was overtly promoted by loss of LINC01094 or miR-577 upregulation, but could be repressed via MTF1 overexpression. The opposite results were observed in apoptosis rate and the levels of Caspase3 and Caspase9. LINC01094 directly bound to miR-577, while MTF1 was verified to be modulated by miR-577. Both LINC01094 and MTF1 were at high levels, whereas miR-577 was at low level in OA synovial fluid and LPS-induced chondrocytes. Furthermore, the highly expressed miR-577 abolished the influences of MTF1 overexpression on LPS-induced chondrocytes. CONCLUSIONS: Silencing of LINC01094 represses the apoptosis of chondrocytes through upregulating miR-577 expression and downregulating MTF1 levels, providing a preliminary insight for the treatment of OA in the future.


Apoptosis , Chondrocytes , MicroRNAs , Osteoarthritis , RNA, Long Noncoding , Transcription Factors , Chondrocytes/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factor MTF-1 , Cells, Cultured , Gene Knockdown Techniques , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Lipopolysaccharides
13.
Sci Rep ; 14(1): 11237, 2024 05 16.
Article En | MEDLINE | ID: mdl-38755283

Osteoarthritis (OA) is the most prevalent form of arthritis, characterized by a complex pathogenesis. One of the key factors contributing to its development is the apoptosis of chondrocytes triggered by oxidative stress. Involvement of peroxisome proliferator-activated receptor gamma (PPARγ) has been reported in the regulation of oxidative stress. However, there remains unclear mechanisms that through which PPARγ influences the pathogenesis of OA. The present study aims to delve into the role of PPARγ in chondrocytes apoptosis induced by oxidative stress in the context of OA. Primary human chondrocytes, both relatively normal and OA, were isolated and cultured for the following study. Various assessments were performed, including measurements of cell proliferation, viability and cytotoxicity. Additionally, we examined cell apoptosis, levels of reactive oxygen species (ROS), nitric oxide (NO), mitochondrial membrane potential (MMP) and cytochrome C release. We also evaluated the expression of related genes and proteins, such as collagen type II (Col2a1), aggrecan, inducible nitric oxide synthase (iNOS), caspase-9, caspase-3 and PPARγ. Compared with relatively normal cartilage, the expression of PPARγ in OA cartilage was down-regulated. The proliferation of OA chondrocytes decreased, accompanied by an increase in the apoptosis rate. Down-regulation of PPARγ expression in OA chondrocytes coincided with an up-regulation of iNOS expression, leading to increased secretion of NO, endogenous ROS production, and decrease of MMP levels. Furthermore, we observed the release of cytochrome C, elevated caspase-9 and caspase-3 activities, and reduction of the components of extracellular matrix (ECM) Col2a1 and aggrecan. Accordingly, utilization of GW1929 (PPARγ Agonists) or Z-DEVD-FMK (caspase-3 inhibitor) can protect chondrocytes from mitochondrial-related apoptosis and alleviate the progression of OA. During the progression of OA, excessive oxidative stress in chondrocytes leads to apoptosis and ECM degradation. Activation of PPARγ can postpone OA by down-regulating caspase-3-dependent mitochondrial apoptosis pathway.


Apoptosis , Caspase 3 , Chondrocytes , Mitochondria , Osteoarthritis , PPAR gamma , Reactive Oxygen Species , Humans , Chondrocytes/metabolism , Chondrocytes/pathology , PPAR gamma/metabolism , Caspase 3/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress , Membrane Potential, Mitochondrial , Cell Proliferation , Nitric Oxide/metabolism , Cells, Cultured , Middle Aged , Aged , Female , Male
14.
J Nanobiotechnology ; 22(1): 255, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755672

Age is the most important risk factor in degenerative diseases such as osteoarthritis (OA), which is associated with the accumulation of senescent cells in the joints. Here, we aimed to assess the impact of senescence on the therapeutic properties of extracellular vesicles (EVs) from human fat mesenchymal stromal cells (ASCs) in OA. We generated a model of DNA damage-induced senescence in ASCs using etoposide and characterized EVs isolated from their conditioned medium (CM). Senescent ASCs (S-ASCs) produced 3-fold more EVs (S-EVs) with a slightly bigger size and that contain 2-fold less total RNA. Coculture experiments showed that S-ASCs were as efficient as healthy ASCs (H-ASCs) in improving the phenotype of OA chondrocytes cultured in resting conditions but were defective when chondrocytes were proliferating. S-EVs were also impaired in their capacity to polarize synovial macrophages towards an anti-inflammatory phenotype. A differential protein cargo mainly related to inflammation and senescence was detected in S-EVs and H-EVs. Using the collagenase-induced OA model, we found that contrary to H-EVs, S-EVs could not protect mice from cartilage damage and joint calcifications, and were less efficient in protecting subchondral bone degradation. In addition, S-EVs induced a pro-catabolic and pro-inflammatory gene signature in the joints of mice shortly after injection, while H-EVs decreased hypertrophic, catabolic and inflammatory pathways. In conclusion, S-EVs are functionally impaired and cannot protect mice from developing OA.


Cellular Senescence , Chondrocytes , Extracellular Vesicles , Mesenchymal Stem Cells , Osteoarthritis , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Animals , Humans , Mice , Chondrocytes/metabolism , Cells, Cultured , Male , Mice, Inbred C57BL , DNA Damage
15.
PeerJ ; 12: e17032, 2024.
Article En | MEDLINE | ID: mdl-38770093

Purpose: This study seeks to identify potential clinical biomarkers for osteoarthritis (OA) using bioinformatics and investigate OA mechanisms through cellular assays. Methods: Differentially Expressed Genes (DEGs) from GSE52042 (four OA samples, four control samples) were screened and analyzed with protein-protein interaction (PPI) analysis. Overlapping genes in GSE52042 and GSE206848 (seven OA samples, and seven control samples) were identified and evaluated using Gene Set Enrichment Analysis (GSEA) and clinical diagnostic value analysis to determine the hub gene. Finally, whether and how the hub gene impacts LPS-induced OA progression was explored by in vitro experiments, including Western blotting (WB), co-immunoprecipitation (Co-IP), flow cytometry, etc. Result: Bioinformatics analysis of DEGs (142 up-regulated and 171 down-regulated) in GSE52042 identified two overlapping genes (U2AF2, TPX2) that exhibit significant clinical diagnostic value. These genes are up-regulated in OA samples from both GSE52042 and GSE206848 datasets. Notably, TPX2, which AUC = 0.873 was identified as the hub gene. In vitro experiments have demonstrated that silencing TPX2 can alleviate damage to chondrocytes induced by lipopolysaccharide (LPS). Furthermore, there is a protein interaction between TPX2 and MMP13 in OA. Excessive MMP13 can attenuate the effects of TPX2 knockdown on LPS-induced changes in OA protein expression, cell growth, and apoptosis. Conclusion: In conclusion, our findings shed light on the molecular mechanisms of OA and suggested TPX2 as a potential therapeutic target. TPX2 could promote the progression of LPS-induced OA by up-regulating the expression of MMP13, which provides some implications for clinical research.


Cell Cycle Proteins , Chondrocytes , Disease Progression , Lipopolysaccharides , Matrix Metalloproteinase 13 , Microtubule-Associated Proteins , Osteoarthritis , Up-Regulation , Lipopolysaccharides/pharmacology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/chemically induced , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/drug effects , Computational Biology , Protein Interaction Maps
16.
Cells ; 13(10)2024 May 16.
Article En | MEDLINE | ID: mdl-38786073

In this study of the alterations of Glypicans 1 to 6 (GPCs) and Notum in plasma, bone marrow mesenchymal stromal cells (BM-MSCs) and osteoblasts in Osteoarthritis (OA), the levels of GPCs and Notum in the plasma of 25 patients and 24 healthy subjects were measured. In addition, BM-MSCs from eight OA patients and eight healthy donors were cultured over a period of 21 days using both a culture medium and an osteogenic medium. Protein and gene expression levels of GPCs and Notum were determined using ELISA and qPCR at 0, 7, 14 and 21 days. GPC5 and Notum levels decreased in the plasma of OA patients, while the BM-MSCs of OA patients showed downexpression of GPC6 and upregulation of Notum. A decrease in GPC5 and Notum proteins and an increase in GPC3 were found. During osteogenic differentiation, elevated GPCs 2, 4, 5, 6 and Notum mRNA levels and decreased GPC3 were observed in patients with OA. Furthermore, the protein levels of GPC2, GPC5 and Notum decreased, while the levels of GPC3 increased. Glypicans and Notum were altered in BM-MSCs and during osteogenic differentiation from patients with OA. The alterations found point to GPC5 and Notum as new candidate biomarkers of OA pathology.


Glypicans , Mesenchymal Stem Cells , Osteoarthritis , Osteoblasts , Humans , Mesenchymal Stem Cells/metabolism , Osteoarthritis/blood , Osteoarthritis/pathology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Male , Female , Glypicans/metabolism , Glypicans/blood , Glypicans/genetics , Middle Aged , Cell Differentiation , Osteogenesis/genetics , Aged , Case-Control Studies , Cells, Cultured , Bone Marrow Cells/metabolism
17.
Nat Aging ; 4(5): 664-680, 2024 May.
Article En | MEDLINE | ID: mdl-38760576

Hyaline cartilage fibrosis is typically considered an end-stage pathology of osteoarthritis (OA), which results in changes to the extracellular matrix. However, the mechanism behind this is largely unclear. Here, we found that the RNA helicase DDX5 was dramatically downregulated during the progression of OA. DDX5 deficiency increased fibrosis phenotype by upregulating COL1 expression and downregulating COL2 expression. In addition, loss of DDX5 aggravated cartilage degradation by inducing the production of cartilage-degrading enzymes. Chondrocyte-specific deletion of Ddx5 led to more severe cartilage lesions in the mouse OA model. Mechanistically, weakened DDX5 resulted in abundance of the Fn1-AS-WT and Plod2-AS-WT transcripts, which promoted expression of fibrosis-related genes (Col1, Acta2) and extracellular matrix degradation genes (Mmp13, Nos2 and so on), respectively. Additionally, loss of DDX5 prevented the unfolding Col2 promoter G-quadruplex, thereby reducing COL2 production. Together, our data suggest that strategies aimed at the upregulation of DDX5 hold significant potential for the treatment of cartilage fibrosis and degradation in OA.


Alternative Splicing , DEAD-box RNA Helicases , Fibrosis , G-Quadruplexes , Osteoarthritis , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Mice , Osteoarthritis/pathology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Fibrosis/metabolism , Fibrosis/genetics , Fibrosis/pathology , Humans , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Male
18.
Acta Biomater ; 181: 297-307, 2024 Jun.
Article En | MEDLINE | ID: mdl-38710401

The pericellular matrix (PCM) serves a critical role in signal transduction and mechanoprotection in chondrocytes. Osteoarthritis (OA) leads to a gradual deterioration of the cartilage, marked by a shift in the spatial arrangement of chondrocytes from initially isolated strands to large cell clusters in end-stage degeneration. These changes coincide with progressive enzymatic breakdown of the PCM. This study aims to assess the role and involvement of specific matrix metalloproteinases (MMPs) in PCM degradation during OA. We selected cartilage samples from 148 OA patients based on the predominant spatial chondrocyte patterns. The presence of various MMPs (-1,-2,-3,-7,-8,-9,-10,-12,-13) was identified by multiplexed immunoassays. For each pattern and identified MMP, the levels and activation states (pro-form vs. active form) were measured by zymograms and western blots. The localization of these MMPs was determined using immunohistochemical labeling. To verify these results, healthy cartilage was exposed to purified MMPs, and the consecutive structural integrity of the PCM was analyzed through immunolabeling and proximity ligation assay. Screening showed elevated levels of MMP-1,-2,-3,-7, and -13, with their expression profile showing a clear dependency of the degeneration stage. MMP-2 and -7 were localized in the PCM, whereas MMP-1,-7, and -13 were predominantly intracellular. We found that MMP-2 and -3 directly disrupt collagen type VI, and MMP-3 and -7 destroy perlecan. MMP-2, -3, and -7 emerge as central players in early PCM degradation in OA. With the disease's initial stages already displaying elevated peaks in MMP expression, this insight may guide early targeted therapies to halt abnormal PCM remodeling. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) causes a gradual deterioration of the articular cartilage, accompanied by a progressive breakdown of the pericellular matrix (PCM). The PCM's crucial function in protecting and transmitting signals within chondrocytes is impaired in OA. By studying 148 OA-patient cartilage samples, the involvement of matrix metalloproteinases (MMPs) in PCM breakdown was explored. Findings highlighted elevated levels of certain MMPs linked to different stages of degeneration. Notably, MMP-2, -3, and -7 were identified as potent contributors to early PCM degradation, disrupting key components like collagen type VI and perlecan. Understanding these MMPs' roles in initiating OA progression, especially in its early stages, provides insights into potential targets for interventions to preserve PCM integrity and potentially impeding OA advancement.


Extracellular Matrix , Matrix Metalloproteinases , Osteoarthritis , Proteolysis , Humans , Matrix Metalloproteinases/metabolism , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/enzymology , Extracellular Matrix/metabolism , Male , Female , Middle Aged , Aged , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/enzymology , Cartilage, Articular/pathology , Cartilage, Articular/metabolism
19.
Acta Biomater ; 181: 425-439, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729544

Synovial macrophages play an important role in the progression of osteoarthritis (OA). In this study, we noted that synovial macrophages can activate pyroptosis in a gasdermin d-dependent manner and produce reactive oxygen species (ROS), aberrantly activating the mammalian target of rapamycin complex 1 (mTORC1) pathway and matrix metalloproteinase-9 (MMP9) expression in synovial tissue samples collected from both patients with OA and collagen-induced osteoarthritis (CIOA) mouse model. To overcome this, we constructed rapamycin- (RAPA, a mTORC1 inhibitor) loaded mesoporous Prussian blue nanoparticles (MPB NPs, for catalyzing ROS) and modified the NPs with MMP9-targeted peptides (favor macrophage targeting) to develop RAPA@MPB-MMP9 NPs. The inherent enzyme-like activity and RAPA released from RAPA@MPB-MMP9 NPs synergistically impeded the pyroptosis of macrophages and the activation of the mTORC1 pathway. In particular, the NPs decreased pyroptosis-mediated ROS generation, thereby inhibiting cGAS-STING signaling pathway activation caused by the release of mitochondrial DNA. Moreover, the NPs promoted macrophage mitophagy to restore mitochondrial stability, alleviate pyroptosis-related inflammatory responses, and decrease senescent synoviocytes. After the as-prepared NPs were intra-articularly injected into the CIOA mouse model, they efficiently attenuated synovial macrophage pyroptosis and cartilage degradation. In conclusion, our study findings provide a novel therapeutic strategy for OA that modulates the pyroptosis and mitophagy of synovial macrophage by utilizing functionalized NPs. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) presents a significant global challenge owing to its complex pathogenesis and finite treatment options. Synovial macrophages have emerged as key players in the progression of OA, managing inflammation and tissue destruction. In this study, we discovered a novel therapeutic strategy in which the pyroptosis and mitophagy of synovial macrophages are targeted to mitigate OA pathology. For this, we designed and prepared rapamycin-loaded mesoporous Prussian blue nanoparticles (RAPA@MPB-MMP9 NPs) to specifically target synovial macrophages and modulate their inflammatory responses. These NPs could efficiently suppress macrophage pyroptosis, diminish reactive oxygen species production, and promote mitophagy, thereby alleviating inflammation and protecting cartilage integrity. Our study findings not only clarify the intricate mechanisms underlying OA pathogenesis but also present a promising therapeutic approach for effectively managing OA by targeting dysregulation in synovial macrophages.


Macrophages , Mitophagy , Nanoparticles , Osteoarthritis , Pyroptosis , Reactive Oxygen Species , Osteoarthritis/pathology , Osteoarthritis/drug therapy , Animals , Pyroptosis/drug effects , Nanoparticles/chemistry , Macrophages/metabolism , Macrophages/drug effects , Macrophages/pathology , Mitophagy/drug effects , Mice , Humans , Reactive Oxygen Species/metabolism , Male , Sirolimus/pharmacology , Matrix Metalloproteinase 9/metabolism , Disease Progression , Mechanistic Target of Rapamycin Complex 1/metabolism , Synovial Membrane/pathology , Synovial Membrane/drug effects , Mice, Inbred C57BL , Ferrocyanides
20.
Int J Mol Sci ; 25(10)2024 May 20.
Article En | MEDLINE | ID: mdl-38791601

Osteoarthritis (OA) is a common joint disorder characterized by cartilage degeneration, often leading to pain and functional impairment. Minced cartilage implantation (MCI) has emerged as a promising one-step alternative for large cartilage defects. However, the source of chondrocytes for MCI remains a challenge, particularly in advanced OA, as normal cartilage is scarce. We performed in vitro studies to evaluate the feasibility of MCI using osteophyte cartilage, which is present in patients with advanced OA. Osteophyte and articular cartilage samples were obtained from 22 patients who underwent total knee arthroplasty. Chondrocyte migration and proliferation were assessed using cartilage fragment/atelocollagen composites to compare the characteristics and regenerative potential of osteophytes and articular cartilage. Histological analysis revealed differences in cartilage composition between osteophytes and articular cartilage, with higher expression of type X collagen and increased chondrocyte proliferation in the osteophyte cartilage. Gene expression analysis identified distinct gene expression profiles between osteophytes and articular cartilage; the expression levels of COL2A1, ACAN, and SOX9 were not significantly different. Chondrocytes derived from osteophyte cartilage exhibit enhanced proliferation, and glycosaminoglycan production is increased in both osteophytes and articular cartilage. Osteophyte cartilage may serve as a viable alternative source of MCI for treating large cartilage defects in OA.


Cartilage, Articular , Cell Proliferation , Chondrocytes , Osteoarthritis , Osteophyte , Humans , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/surgery , Chondrocytes/metabolism , Chondrocytes/pathology , Osteophyte/metabolism , Osteophyte/pathology , Male , Female , Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/surgery , Middle Aged , Collagen Type II/metabolism , Collagen Type II/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Cells, Cultured , Cell Movement
...