Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.958
Filter
1.
Biomaterials ; 312: 122720, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39084098

ABSTRACT

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Nanoparticles , Osteoarthritis , Peptides , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Nanoparticles/chemistry , Humans , Cell Differentiation/drug effects , Peptides/chemistry , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis/drug effects , Mice , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Tissue Engineering/methods , Regeneration/drug effects
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 324: 125000, 2025 Jan 05.
Article in English | MEDLINE | ID: mdl-39180968

ABSTRACT

Fourier transform infrared spectroscopy (FTIRS) can provide rich information on the composition and content of samples, enabling the detection of subtle changes in tissue composition and structure. This study represents the first application of FTIRS to investigate cartilage under microgravity. Simulated microgravity cartilage model was firstly established by tail-suspension (TS) for 7, 14 and 21 days, which would be compared to control samples. A self-developed hollow optical fiber attenuated total reflection (HOF-ATR) probe coupled with a FTIR spectrometer was used for the spectral acquisition of cartilage samples in situ, and one-way analysis of variance (ANOVA) was employed to analyze the changes in the contents of cartilage matrix at different stages. The results indicate that cartilage degenerates in microgravity, the collagen content gradually decreases with the TS time, and the structure of collagen fibers changes. The trends of proteoglycan content and collagen integrity show an initial decrease followed by an increase, ultimately significantly decreasing. The findings provide the basis for the cartilage degeneration in microgravity with TS time, which must be of real significance for space science and health detection.


Subject(s)
Cartilage, Articular , Collagen , Weightlessness Simulation , Spectroscopy, Fourier Transform Infrared/methods , Cartilage, Articular/pathology , Cartilage, Articular/chemistry , Cartilage, Articular/metabolism , Collagen/analysis , Collagen/metabolism , Collagen/chemistry , Animals , Proteoglycans/analysis , Male
3.
Biomolecules ; 14(9)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39334835

ABSTRACT

Introduction: Osteoarthritis is a degenerative condition of the cartilage, often common among the population and occurs frequently with aging. Many factors are decisive for the development of its pathogenesis such as age, obesity, trauma, mechanical load, and modification of synovial biology. The main features of osteoarthritis are chondrocytes and cartilage matrix loss, which lead to pain, loss of function of the whole joint, and disability, representing a relevant health problem. Recently, a new therapeutic approach based on cell therapy has been studying the regenerative ability of mesenchymal stem cells for osteoarthritic chondrocytes. Aim: This in vitro study clarifies the regenerative effects of multipotent adipose-derived stem cells and the pluripotent amniotic epithelial stem cells on arthrosis chondrocytes by performing co-culture experiments. Methods: We studied the regenerative potential of secretome (soluble factors and extracellular vesicles), mesenchymal stem cells, and the adipose stromal vascular fraction. The regenerative effects were evaluated by gene and protein expression analysis of articular cartilage-specific genes and proteins like col2a1, acan, and sox9. Results: Mesenchymal stem cells, secretome, and adipose stromal vascular fractions influenced the cartilage genes and protein expression. Conclusions: The results indicate that the treatment with mesenchymal stem cells could be the best biological approach for cartilage regenerative medicine.


Subject(s)
Cartilage, Articular , Chondrocytes , Mesenchymal Stem Cells , Osteoarthritis , Secretome , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Secretome/metabolism , Chondrocytes/metabolism , Osteoarthritis/therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Coculture Techniques , Collagen Type II/metabolism , Collagen Type II/genetics , Aggrecans/metabolism , Aggrecans/genetics , Cells, Cultured
4.
Nutrients ; 16(18)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39339707

ABSTRACT

BACKGROUND: Hesperetin, a flavonoid derived from citrus fruits, exhibits potent antioxidant and anti-inflammatory activities and has been implicated in cartilage protection. However, its effectiveness against T-2 toxin-induced knee cartilage damage remains unclear. METHODS: In this study, high-throughput sequencing analysis was employed to identify the key signaling pathways involved in T-2 toxin-induced articular cartilage damage in rats. Animal models were divided into the following groups: control, low-dose T-2 toxin, high-dose T-2 toxin, T-2 toxin + hesperetin, hesperetin, and vehicle. Pathological staining and immunohistochemistry were used to assess pathological changes, as well as the expression levels of the cartilage matrix-related proteins MMP13 and collagen II, along with the activation of the p38 MAPK signaling pathway. Additionally, primary rat chondrocytes were cultured to establish an in vitro model for investigating the underlying mechanism. RESULTS: High-throughput sequencing analysis revealed the involvement of the MAPK signaling pathway in T-2 toxin-induced articular cartilage damage in rats. Hesperetin intervention in T-2 toxin-exposed rats attenuated pathological cartilage damage. Immunohistochemistry results demonstrated a significant reduction in collagen II protein expression in the high-dose T-2 toxin group (p < 0.01), accompanied by a significant increase in MMP13 protein expression (p < 0.01). In both the articular cartilage and the epiphyseal plate, the T-2 toxin + hesperetin group exhibited significantly higher collagen II protein expression than the high-dose T-2 toxin group (p < 0.05), along with significantly lower MMP13 protein expression (p < 0.05). Hesperetin inhibited the over-activation of the p38/MEF2C signaling axis induced by T-2 toxin in primary rat chondrocytes. Compared to the T-2 toxin group, the T-2 toxin + hesperetin group showed significantly reduced phosphorylation levels of p38 and protein expression levels of MEF2C (p < 0.001 or p < 0.05). Moreover, the T-2 toxin + hesperetin group exhibited a significant decrease in MMP13 protein expression (p < 0.05) and a significant increase in collagen II protein expression (p < 0.01) compared to the T-2 toxin group. CONCLUSIONS: T-2 toxin activates the p38 MAPK signaling pathway, causing knee cartilage damage in rats. Treatment with hesperetin inhibits the p38/MEF2C signaling axis, regulates collagen II and MMP13 protein expression, and reduces cartilage injury significantly.


Subject(s)
Cartilage, Articular , Chondrocytes , Hesperidin , Rats, Sprague-Dawley , T-2 Toxin , p38 Mitogen-Activated Protein Kinases , Animals , Hesperidin/pharmacology , Chondrocytes/drug effects , Chondrocytes/metabolism , T-2 Toxin/toxicity , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Rats , Male , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Signal Transduction/drug effects , Collagen Type II/metabolism , Disease Models, Animal , Cells, Cultured
5.
Mol Biol Rep ; 51(1): 1018, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331223

ABSTRACT

BACKGROUND: Moderate mechanical stress generated by normal joint loading and movements helps maintain the health of articular cartilage. Despite growing interest in the pathogenesis of cartilage degeneration caused by reduced mechanical stress, its reversibility by mechanical reloading is less understood. This study aimed to investigate the response of articular cartilage exposed to mechanical reloading after unloading in vivo and in vitro. METHODS AND RESULTS: Disuse atrophy was induced in the knee joint cartilage of adult mice through hindlimb unloading by tail suspension. For in vivo experiments, mice were subjected to reloading with or without daily exercise intervention or surgical destabilization of the knee joint. Microcomputed tomography and histomorphometric analyses were performed on the harvested knee joints. Matrix loss and thinning of articular cartilage due to unloading were fully or partially restored by reloading, and exercise intervention enhanced the restoration. Subchondral bone density decreased by unloading and increased to above-normal levels by reloading. The severity of cartilage damage caused by joint instability was not different even with prior non-weight bearing. For in vitro experiments, articular chondrocytes isolated from the healthy or unloaded joints of the mice were embedded in agarose gel. After dynamic compression loading, the expression levels of anabolic (Sox9, Col2a1, and Acan) and catabolic (Mmp13 and Adamts5) factors of cartilage were analyzed. In chondrocytes isolated from the unloaded joints, similar to those from healthy joints, dynamic compression increased the expression of anabolic factors but suppressed the expression of catabolic factors. CONCLUSION: The results of this study indicate that the morphological changes in articular cartilage exposed to mechanical unloading may be restored in response to mechanical reloading by shifting extracellular matrix metabolism in chondrocytes to anabolism.


Subject(s)
ADAMTS5 Protein , Cartilage, Articular , Chondrocytes , Hindlimb Suspension , Stress, Mechanical , Animals , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Mice , Chondrocytes/metabolism , Chondrocytes/pathology , ADAMTS5 Protein/metabolism , ADAMTS5 Protein/genetics , Hindlimb Suspension/adverse effects , Matrix Metalloproteinase 13/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Aggrecans/metabolism , Collagen Type II/metabolism , Male , X-Ray Microtomography , Weight-Bearing/physiology , Atrophy , Knee Joint/pathology , Knee Joint/physiopathology , Knee Joint/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Physical Conditioning, Animal
6.
J Cell Mol Med ; 28(18): e70095, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289794

ABSTRACT

EphrinB2, a member of the Ephrin family, has been linked to several orthopaedic conditions. Nevertheless, the correlation between ephrinB2 and post-traumatic arthritis (PTOA) remains unclear. Human PTOA cartilage from human and mouse knee joints was systematically analysed to investigate the relationship between EphrinB2 and PTOA using SO-FG and toluidine blue staining, micro-CT, histomorphometry, immunohistochemistry, immunofluorescence, lentiviral articular injection and in situ end labeling (TUNEL) assays. EphrinB2 expression was significantly downregulated in PTOA chondrocytes. Blocking EphrinB2 increased the breakdown of cartilage matrix in mice with PTOA via reducing the process of chondrocyte autophagy. The presence of severe cartilage damage was evident, as indicated by a considerable decrease in both cartilage thickness and area, accompanied by an increase in chondrocyte death. Altogether, EphrinB2 is required for the maintenance of cartilage homeostasis in post-traumatic arthritis, and EphrinB2 ablation is associated with accelerated chondrocyte matrix degeneration, finally causing damage to the articular cartilage.


Subject(s)
Autophagy , Cartilage, Articular , Chondrocytes , Ephrin-B2 , Homeostasis , Chondrocytes/metabolism , Chondrocytes/pathology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Ephrin-B2/metabolism , Ephrin-B2/genetics , Humans , Mice , Male , Mice, Inbred C57BL , Female
7.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273553

ABSTRACT

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


Subject(s)
Analgesics , Anti-Inflammatory Agents , Gynostemma , Osteoarthritis , Plant Extracts , Animals , Gynostemma/chemistry , Mice , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , RAW 264.7 Cells , Rats , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Male , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Disease Models, Animal , Rats, Sprague-Dawley , Pain/drug therapy
8.
Physiol Res ; 73(4): 633-642, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39264083

ABSTRACT

The study aims to elucidate the therapeutic mechanism of Baicalin (BAI) in alleviating cartilage injury in osteoarthritic (OA) rat models, concentrating on its regulation of the miR-766-3p/AIFM1 axis. An OA rat model was developed with unilateral anterior cruciate ligament transection (ACLT). Interventions comprised of BAI treatment and intra-articular administration of miR-766-3p inhibitor. For evaluation, histopathological staining was conducted to investigate the pathological severity of knee cartilage injury. The levels of oxidative stress (OS) indicators including MDA, SOD, and GSH-Px, were quantified using colorimetric assays. Inflammatory factors (IFs; TNF-?, IL-1?, and IL-6) in knee joint lavage fluids were assessed using ELISA, while RT-PCR was employed to quantify miR-766-3p expression. TUNEL apoptosis staining was utilized to detect chondrocyte apoptosis, and western blotting examined autophagy-related markers (LC3, Beclin, p62), extracellular matrix (ECM) synthesis-associated indices (COL2A, ACAN, MMP13), and apoptosis-inducing factor mitochondrion-associated 1 (AIFM1). Histological examination revealed a marked amelioration of cartilage injury in the BAI-treated OA rat models compared to controls. BAI treatment significantly reduced inflammation and OS of knee joint fluid, activated autophagy, and decreased chondrocyte apoptosis and ECM degradation. Interestingly, the inhibitory effects of BAI on these pathological markers were significantly decreased by the miR-766-3p inhibitor. Further assessment revealed that BAI efficiently promoted miR-766-3p expression while inhibiting AIFM1 protein expression. BAI potentially mitigates articular cartilage injury in OA rats, likely through modulation of miR-766-3p/AIFM1 axis. Keywords: Baicalin, microRNA, AIFM1, Osteoarthritisv, Rat.


Subject(s)
Flavonoids , MicroRNAs , Rats, Sprague-Dawley , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , MicroRNAs/metabolism , MicroRNAs/genetics , MicroRNAs/biosynthesis , Rats , Male , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Apoptosis Inducing Factor/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects
9.
Int J Rheum Dis ; 27(9): e15323, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39221886

ABSTRACT

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


Subject(s)
ADAM Proteins , Chondrocytes , Disease Progression , Membrane Proteins , RNA, Long Noncoding , STAT3 Transcription Factor , Signal Transduction , Up-Regulation , Animals , Humans , Male , Mice , ADAM Proteins/metabolism , ADAM Proteins/genetics , Adenosine/analogs & derivatives , Apoptosis , Arthritis, Experimental/metabolism , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Interleukin-1beta/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Mice, Inbred C57BL , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
10.
FASEB J ; 38(17): e70013, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39225365

ABSTRACT

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


Subject(s)
Cartilage, Articular , Chondrocytes , Osteoarthritis , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , SOX9 Transcription Factor , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Mice , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Cell Proliferation , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Male
11.
Nat Commun ; 15(1): 7712, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231947

ABSTRACT

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


Subject(s)
Cartilage, Articular , Cellular Senescence , Chondrocytes , Fatty Acids, Monounsaturated , Osteoarthritis , Animals , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Male , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/drug therapy , Osteoarthritis/prevention & control , Mice , Cellular Senescence/drug effects , Humans , Fatty Acids, Monounsaturated/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Mice, Inbred C57BL , Disease Models, Animal , Female
12.
Bone Res ; 12(1): 50, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231936

ABSTRACT

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


Subject(s)
Carrier Proteins , Chondrocytes , Microfilament Proteins , Osteoarthritis , Animals , Humans , Male , Mice , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/genetics , Phenotype , Receptors, Odorant , Signal Transduction
13.
J Orthop Surg Res ; 19(1): 550, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252111

ABSTRACT

Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.


Subject(s)
Cell Death , Chondrocytes , Disease Progression , Osteoarthritis , Chondrocytes/pathology , Humans , Osteoarthritis/pathology , Osteoarthritis/therapy , Cell Death/physiology , Apoptosis/physiology , Cartilage, Articular/pathology , Autophagy/physiology , Animals , Pyroptosis/physiology , Ferroptosis/physiology
14.
Transplant Proc ; 56(8): 1896-1903, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39242314

ABSTRACT

BACKGROUND: Cartilage is a crucial tissue in vascularized composite allotransplantation (VCA) and plays a pivotal role in restoring motor function, especially in joint allotransplantation. Nevertheless, our understanding of immune rejection in cartilage remains limited and contentious. This study seeks to investigate the immune rejection of cartilage in a large animal model of VCA. METHODS: Cartilage, including articular cartilage and meniscus, as well as skin, muscle and lymph node, was retrieved from a swine heterotopic VCA graft when the skin of the graft suffered from grade III-IV rejection. Histologic examination, transmission electron microscopy and immunofluorescent staining were used to investigate immune rejection. RESULTS: Histologic examination revealed the infiltration of inflammatory cells and tissue destruction in cartilage. Transmission electron microscopy confirmed tissue damage and necrosis in cartilage. However, cartilage exhibited milder tissue damage when compared to rejected skin and muscle. Immunofluorescent staining revealed the activation of both the innate and adaptive immune systems, accompanied by an up-regulation of cell death biomarkers, including apoptosis and pyroptosis, in the rejected cartilage. CONCLUSION: Our study demonstrates that cartilage is not immunologically privileged and undergoes immune rejection concurrently with skin and muscle in the VCA graft, though with less severe inflammation and rejection.


Subject(s)
Graft Rejection , Vascularized Composite Allotransplantation , Animals , Graft Rejection/immunology , Swine , Cartilage/transplantation , Cartilage, Articular/pathology , Disease Models, Animal
15.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337321

ABSTRACT

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


Subject(s)
Lab-On-A-Chip Devices , Osteoarthritis , Humans , Osteoarthritis/metabolism , Osteoarthritis/pathology , Glycosaminoglycans/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Oxygen/metabolism , Glucose/metabolism , L-Lactate Dehydrogenase/metabolism , Alkaline Phosphatase/metabolism
16.
Int J Mol Sci ; 25(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39337384

ABSTRACT

Hemophilia, which is a rare disease, results from congenital deficiencies of coagulation factors VIII and IX, respectively, leading to spontaneous bleeding into joints, resulting in hemophilic arthropathy (HA). HA involves complex processes, including synovial proliferation, angiogenesis, and tissue remodeling. Despite ongoing research, factors contributing to HA progression, especially in adults with severe HA experiencing joint pain, remain unclear. Blood markers, particularly collagen-related ones, have been explored to assess joint health in hemophilia. For example, markers like CTX-I and CTX-II reflect bone and cartilage turnover, respectively. Studies indicate elevated levels of certain markers post-bleeding episodes, suggesting joint health changes. However, longitudinal studies on collagen turnover and basement membrane or endothelial cell markers in relation to joint outcomes, particularly during painful episodes, are scarce. Given the role of the CX3CL1/CX3XR1 axis in arthritis, other studies investigate its involvement in HA. The importance of different inflammatory and bone damage biomarkers should be assessed, alongside articular cartilage and synovial membrane morphology, aiming to enhance understanding of hemophilic arthropathy progression.


Subject(s)
Biomarkers , Hemophilia A , Humans , Hemophilia A/complications , Hemophilia A/metabolism , Hemophilia A/pathology , Hemarthrosis/metabolism , Hemarthrosis/etiology , Hemarthrosis/pathology , Joint Diseases/metabolism , Joint Diseases/pathology , Joint Diseases/etiology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology
17.
J Cell Mol Med ; 28(16): e70027, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159149

ABSTRACT

Ageing is the most prominent risk for osteoarthritis (OA) development. This study aimed to investigate the role of phosphoinositide-specific phospholipase Cγ (PLCγ) 1, previously linked to OA progression, in regulating age-related changes in articular cartilage and subchondral bone. d-galactose (d-Gal) was employed to treat chondrocytes from rats and mice or injected intraperitoneally into C57BL/6 mice. RTCA, qPCR, Western blot and immunohistochemistry assays were used to evaluate cell proliferation, matrix synthesis, senescence genes and senescence-associated secretory phenotype, along with PLCγ1 expression. Subchondral bone morphology was assessed through micro-CT. In mice with chondrocyte-specific Plcg1 deficiency (Plcg1flox/flox; Col2a1-CreERT), articular cartilage and subchondral bone were examined over different survival periods. Our results showed that d-Gal induced chondrocyte senescence, expedited articular cartilage ageing and caused subchondral bone abnormalities. In d-Gal-induced chondrocytes, diminished PLCγ1 expression was observed, and its further inhibition by U73122 exacerbated chondrocyte senescence. Plcg1flox/flox; Col2a1-CreERT mice exhibited more pronounced age-related changes in articular cartilage and subchondral bone compared to Plcg1flox/flox mice. Therefore, not only does d-Gal induce senescence in chondrocytes and age-related changes in articular cartilage and subchondral bone, as well as diminished PLCγ1 expression, but PLCγ1 deficiency in chondrocytes may also accelerate age-related changes in articular cartilage and subchondral bone. PLCγ1 may be a promising therapeutic target for mitigating age-related changes in joint tissue.


Subject(s)
Cartilage, Articular , Chondrocytes , Mice, Inbred C57BL , Phospholipase C gamma , Animals , Male , Mice , Rats , Aging/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/diagnostic imaging , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Proliferation , Cellular Senescence , Chondrocytes/metabolism , Estrenes/pharmacology , Galactose/metabolism , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology , Phospholipase C gamma/metabolism , Phospholipase C gamma/genetics , Pyrrolidinones/pharmacology
18.
Am J Sports Med ; 52(10): 2555-2564, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39101608

ABSTRACT

BACKGROUND: Cam morphology develops during skeletal growth, but its influence on cartilage and the labrum in high-impact athletes later in life is unknown. PURPOSE: To (1) explore the association between the presence and duration of cam morphology during adolescence and the cartilage and labral status 7 to 12 years later and (2) report the prevalence of cartilage loss and labral damage in a population of young male athletes (<32 years old) who played professional soccer during skeletal growth. STUDY DESIGN: Cohort study (Prognosis); Level of evidence, 2. METHODS: A total of 89 healthy male academy soccer players from the Dutch soccer club Feyenoord (aged 12-19 years) were included at baseline. At baseline and 2.5- and 5-year follow-ups, standardized supine anteroposterior pelvis and frog-leg lateral radiographs of each hip were obtained. At 12-year follow-up, magnetic resonance imaging of both hips was performed. Cam morphology was defined by a validated alpha angle ≥60° on radiographs at baseline or 2.5- or 5-year follow-up when the growth plates were closed. Hips with the presence of cam morphology at baseline or at 2.5-year follow-up were classified as having a "longer duration" of cam morphology. Hips with cam morphology only present since 5-year follow-up were classified as having a "shorter duration" of cam morphology. At 12-year follow-up, cartilage loss and labral abnormalities were assessed semiquantitatively. Associations were estimated using logistic regression, adjusted for age and body mass index. RESULTS: Overall, 35 patients (70 hips) with a mean age of 28.0 ± 2.0 years and mean body mass index of 24.1 ± 1.8 participated at 12-year follow-up. Cam morphology was present in 56 of 70 hips (80%). The prevalence of cartilage loss was 52% in hips with cam morphology and 21% in hips without cam morphology (adjusted odds ratio, 4.52 [95% CI, 1.16-17.61]; P = .03). A labral abnormality was present in 77% of hips with cam morphology and in 64% of hips without cam morphology (adjusted odds ratio, 1.99 [95% CI, 0.59-6.73]; P = .27). The duration of cam morphology did not influence these associations. CONCLUSION: The development of cam morphology during skeletal growth was associated with future magnetic resonance imaging findings consistent with cartilage loss in young adults but not with labral abnormalities.


Subject(s)
Cartilage, Articular , Soccer , Humans , Male , Adolescent , Prospective Studies , Young Adult , Follow-Up Studies , Soccer/injuries , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/growth & development , Cartilage, Articular/pathology , Child , Magnetic Resonance Imaging , Adult , Bone Development , Radiography , Athletes , Femoracetabular Impingement/diagnostic imaging , Hip Joint/diagnostic imaging , Hip Joint/growth & development
19.
Int Immunopharmacol ; 140: 112858, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39111145

ABSTRACT

OBJECTIVE: The aim of this study was to investigate whether ASA VI controls osteoarthritis (OA) by regulating mitochondrial function. METHODS: Primary chondrocytes were isolated and cultured from rat knee joints. The chondrocytes were treated with ASA VI and interleukin-1ß (IL-1ß) to simulate the inflammatory environment of OA. Cell viability, apoptosis, inflammatory cytokine levels, and extracellular matrix (ECM) component levels were assessed. Mitochondrial function, including ATP levels, mitochondrial membrane potential, reactive oxygen species (ROS) levels, and mitochondrial DNA content, was evaluated. The expression of Sirtuin 3 (Sirt3), a key regulator of mitochondrial homeostasis, was examined. Additionally, a rat OA model was established by destabilizing the medial meniscus, and the effects of ASA VI on cartilage degeneration were assessed. RESULTS: ASA VI treatment improved cell viability, reduced apoptosis, and decreased IL-6 and TNF-α levels in IL-1ß-induced chondrocytes. ASA VI also upregulated Collagen II and Aggrecan expression, while downregulating ADAMTS5 and MMP-13 expression. Furthermore, ASA VI mitigated IL-1ß-induced mitochondrial dysfunction by increasing ATP levels, restoring mitochondrial membrane potential, reducing ROS production, and preserving mitochondrial DNA content. These effects were accompanied by the activation of Sirt3. In the rat OA model, ASA VI treatment increased Sirt3 expression and alleviated cartilage degeneration. CONCLUSION: ASA VI exerts chondroprotective and anti-inflammatory effects on IL-1ß-induced chondrocytes by improving mitochondrial function through Sirt3 activation. ASA VI also attenuates cartilage degeneration in a rat OA model. These findings suggest that ASA VI may be a potential therapeutic agent for the treatment of osteoarthritis by targeting mitochondrial dysfunction.


Subject(s)
Chondrocytes , Homeostasis , Mitochondria , Osteoarthritis , Saponins , Sirtuin 3 , Animals , Male , Mice , Rats , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Saponins/pharmacology
20.
Cell Rep ; 43(8): 114535, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39088322

ABSTRACT

Cartilage maintains the structure and function of joints, with disturbances leading to potential osteoarthritis. N6-methyladenosine (m6A), the most widespread post-transcriptional modification in eukaryotes, plays a crucial role in regulating biological processes. While current research has indicated that m6A affects the progression of osteoarthritis, its function in the development and homeostasis of articular cartilage remains unclear. Here we report that Mettl3 deficiency in chondrocytes leads to mandibular condylar cartilage morphological alterations, early temporomandibular joint osteoarthritis, and diminished adaptive response to abnormal mechanical stimuli. Mechanistically, METTL3 modulates Lats1 mRNA methylation and facilitates its degradation in an m6A-YTHDF2-dependent manner, which subsequently influences the degradation and nuclear translocation of YAP1. Intervention with the Hippo pathway inhibitor XMU-MP-1 alleviates condylar abnormality caused by Mettl3 knockout. Our findings demonstrate the role of METTL3 in cartilage development and homeostasis, offering insights into potential treatment strategies for osteoarthritis.


Subject(s)
Adenosine , Chondrocytes , Homeostasis , Methyltransferases , Protein Serine-Threonine Kinases , RNA Stability , RNA-Binding Proteins , Methyltransferases/metabolism , Methyltransferases/genetics , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Chondrocytes/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , YAP-Signaling Proteins/metabolism , Mice, Knockout , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage/metabolism , Mice, Inbred C57BL , Chondrogenesis/genetics , Methylation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Male , Mandibular Condyle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL