Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 8.118
1.
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
2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38731975

Osteoarthritis (OA) is the most prevalent age-related degenerative disorder, which severely reduces the quality of life of those affected. Whilst management strategies exist, no cures are currently available. Virtually all joint resident cells generate extracellular vesicles (EVs), and alterations in chondrocyte EVs during OA have previously been reported. Herein, we investigated factors influencing chondrocyte EV release and the functional role that these EVs exhibit. Both 2D and 3D models of culturing C28I/2 chondrocytes were used for generating chondrocyte EVs. We assessed the effect of these EVs on chondrogenic gene expression as well as their uptake by chondrocytes. Collectively, the data demonstrated that chondrocyte EVs are sequestered within the cartilage ECM and that a bi-directional relationship exists between chondrocyte EV release and changes in chondrogenic differentiation. Finally, we demonstrated that the uptake of chondrocyte EVs is at least partially dependent on ß1-integrin. These results indicate that chondrocyte EVs have an autocrine homeostatic role that maintains chondrocyte phenotype. How this role is perturbed under OA conditions remains the subject of future work.


Chondrocytes , Extracellular Vesicles , Homeostasis , Integrin beta1 , Chondrocytes/metabolism , Extracellular Vesicles/metabolism , Integrin beta1/metabolism , Humans , Cell Differentiation , Osteoarthritis/metabolism , Osteoarthritis/pathology , Chondrogenesis , Animals , Extracellular Matrix/metabolism , Cartilage, Articular/metabolism , Cells, Cultured
3.
PLoS One ; 19(5): e0302906, 2024.
Article En | MEDLINE | ID: mdl-38718039

Osteoarthritis is the most prevalent type of degenerative arthritis. It is characterized by persistent pain, joint dysfunction, and physical disability. Pain relief and inflammation control are prioritised during osteoarthritis treatment Mume Fructus (Omae), a fumigated product of the Prunus mume fruit, is used as a traditional medicine in several Asian countries. However, its therapeutic mechanism of action and effects on osteoarthritis and articular chondrocytes remain unknown. In this study, we analyzed the anti-osteoarthritis and articular regenerative effects of Mume Fructus extract on rat chondrocytes. Mume Fructus treatment reduced the interleukin-1ß-induced expression of matrix metalloproteinase 3, matrix metalloproteinase 13, and a disintegrin and metalloproteinase with thrombospondin type 1 motifs 5. Additionally, it enhanced collagen type II alpha 1 chain and aggrecan accumulation in rat chondrocytes. Furthermore, Mume Fructus treatment regulated the inflammatory cytokine levels, mitogen-activated protein kinase phosphorylation, and nuclear factor-kappa B activation. Overall, our results demonstrated that Mume Fructus inhibits osteoarthritis progression by inhibiting the nuclear factor-kappa B and mitogen-activated protein kinase pathways to reduce the levels of inflammatory cytokines and prevent cartilage degeneration. Therefore, Mume Fructus may be a potential therapeutic option for osteoarthritis.


Cartilage, Articular , Chondrocytes , Interleukin-1beta , NF-kappa B , Osteoarthritis , Plant Extracts , Animals , Chondrocytes/drug effects , Chondrocytes/metabolism , Interleukin-1beta/metabolism , Rats , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , NF-kappa B/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Plant Extracts/pharmacology , Prunus/chemistry , Rats, Sprague-Dawley , Down-Regulation/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Collagen Type II/metabolism , Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Fruit/chemistry , Aggrecans/metabolism , ADAMTS5 Protein/metabolism , ADAMTS5 Protein/genetics , Cells, Cultured , Male , MAP Kinase Signaling System/drug effects
4.
Cell Mol Biol Lett ; 29(1): 64, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698311

Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.


CRISPR-Cas Systems , Gene Editing , Inflammation , Osteoarthritis , Humans , Osteoarthritis/genetics , Osteoarthritis/therapy , CRISPR-Cas Systems/genetics , Inflammation/genetics , Gene Editing/methods , Animals , Genetic Therapy/methods , Cartilage/metabolism , Cartilage/pathology , Cellular Senescence/genetics , Cartilage, Articular/pathology , Cartilage, Articular/metabolism
5.
Clin Epigenetics ; 16(1): 64, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730337

BACKGROUND: Osteoarthritis (OA) is a complex, age-related multifactorial degenerative disease of diarthrodial joints marked by impaired mobility, joint stiffness, pain, and a significant decrease in quality of life. Among other risk factors, such as genetics and age, hyper-physiological mechanical cues are known to play a critical role in the onset and progression of the disease (Guilak in Best Pract Res Clin Rheumatol 25:815-823, 2011). It has been shown that post-mitotic cells, such as articular chondrocytes, heavily rely on methylation at CpG sites to adapt to environmental cues and maintain phenotypic plasticity. However, these long-lasting adaptations may eventually have a negative impact on cellular performance. We hypothesize that hyper-physiologic mechanical loading leads to the accumulation of altered epigenetic markers in articular chondrocytes, resulting in a loss of the tightly regulated balance of gene expression that leads to a dysregulated state characteristic of the OA disease state. RESULTS: We showed that hyper-physiological loading evokes consistent changes in CpGs associated with expression changes (ML-tCpGs) in ITGA5, CAV1, and CD44, among other genes, which together act in pathways such as anatomical structure morphogenesis (GO:0009653) and response to wound healing (GO:0042060). Moreover, by comparing the ML-tCpGs and their associated pathways to tCpGs in OA pathophysiology (OA-tCpGs), we observed a modest but particular interconnected overlap with notable genes such as CD44 and ITGA5. These genes could indeed represent lasting detrimental changes to the phenotypic state of chondrocytes due to mechanical perturbations that occurred earlier in life. The latter is further suggested by the association between methylation levels of ML-tCpGs mapped to CD44 and OA severity. CONCLUSION: Our findings confirm that hyper-physiological mechanical cues evoke changes to the methylome-wide landscape of chondrocytes, concomitant with detrimental changes in positional gene expression levels (ML-tCpGs). Since CAV1, ITGA5, and CD44 are subject to such changes and are central and overlapping with OA-tCpGs of primary chondrocytes, we propose that accumulation of hyper-physiological mechanical cues can evoke long-lasting, detrimental changes in set points of gene expression that influence the phenotypic healthy state of chondrocytes. Future studies are necessary to confirm this hypothesis.


Cartilage, Articular , Chondrocytes , CpG Islands , DNA Methylation , Epigenesis, Genetic , Organoids , Osteoarthritis , DNA Methylation/genetics , Humans , Osteoarthritis/genetics , CpG Islands/genetics , Chondrocytes/metabolism , Organoids/metabolism , Epigenesis, Genetic/genetics , Cartilage, Articular/metabolism
6.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732111

Glycosphingolipids (GSLs), a subtype of glycolipids containing sphingosine, are critical components of vertebrate plasma membranes, playing a pivotal role in cellular signaling and interactions. In human articular cartilage in osteoarthritis (OA), GSL expression is known notably to decrease. This review focuses on the roles of gangliosides, a specific type of GSL, in cartilage degeneration and regeneration, emphasizing their regulatory function in signal transduction. The expression of gangliosides, whether endogenous or augmented exogenously, is regulated at the enzymatic level, targeting specific glycosyltransferases. This regulation has significant implications for the composition of cell-surface gangliosides and their impact on signal transduction in chondrocytes and progenitor cells. Different levels of ganglioside expression can influence signaling pathways in various ways, potentially affecting cell properties, including malignancy. Moreover, gene manipulations against gangliosides have been shown to regulate cartilage metabolisms and chondrocyte differentiation in vivo and in vitro. This review highlights the potential of targeting gangliosides in the development of therapeutic strategies for osteoarthritis and cartilage injury and addresses promising directions for future research and treatment.


Cartilage, Articular , Chondrocytes , Glycosphingolipids , Osteoarthritis , Regeneration , Humans , Osteoarthritis/therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Glycosphingolipids/metabolism , Signal Transduction , Gangliosides/metabolism
7.
ACS Biomater Sci Eng ; 10(5): 3242-3254, 2024 May 13.
Article En | MEDLINE | ID: mdl-38632852

Osteoarthritis is characterized by enzymatic breakdown of the articular cartilage via the disruption of chondrocyte homeostasis, ultimately resulting in the destruction of the articular surface. Decades of research have highlighted the importance of inflammation in osteoarthritis progression, with inflammatory cytokines shifting resident chondrocytes into a pro-catabolic state. Inflammation can result in poor outcomes for cells implanted for cartilage regeneration. Therefore, a method to promote the growth of new cartilage and protect the implanted cells from the pro-inflammatory cytokines found in the joint space is required. In this study, we fabricate two gel types: polymer network hydrogels composed of chondroitin sulfate and hyaluronic acid, glycosaminoglycans (GAGs) known for their anti-inflammatory and prochondrogenic activity, and interpenetrating networks of GAGs and collagen I. Compared to a collagen-only hydrogel, which does not provide an anti-inflammatory stimulus, chondrocytes in GAG hydrogels result in reduced production of pro-inflammatory cytokines and enzymes as well as preservation of collagen II and aggrecan expression. Overall, GAG-based hydrogels have the potential to promote cartilage regeneration under pro-inflammatory conditions. Further, the data have implications for the use of GAGs to generally support tissue engineering in pro-inflammatory environments.


Chondrocytes , Chondroitin Sulfates , Hyaluronic Acid , Hydrogels , Inflammation , Hydrogels/chemistry , Hydrogels/pharmacology , Chondrocytes/drug effects , Chondrocytes/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/chemistry , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cytokines/metabolism , Aggrecans/metabolism , Tissue Engineering/methods , Osteoarthritis/pathology , Osteoarthritis/drug therapy , Osteoarthritis/metabolism
8.
Mol Med ; 30(1): 55, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664616

BACKGROUND: Osteoarthritis (OA), the most common joint disease, is linked with chondrocyte apoptosis and extracellular matrix (ECM) degradation. Charged multivesicular body protein 5 (CHMP5), a member of the multivesicular body, has been reported to serve as an anti-apoptotic protein to participate in leukemia development. However, the effects of CHMP5 on apoptosis and ECM degradation in OA remain unclear. METHODS: In this study, quantitative proteomics was performed to analyze differential proteins between normal and OA patient articular cartilages. The OA mouse model was constructed by the destabilization of the medial meniscus (DMM). In vitro, interleukin-1 beta (IL-1ß) was used to induce OA in human chondrocytes. CHMP5 overexpression and silencing vectors were created using an adenovirus system. The effects of CHMP5 on IL-1ß-induced chondrocyte apoptosis were investigated by CCK-8, flow cytometry, and western blot. The effects on ECM degradation were examined by western blot and immunofluorescence. The potential mechanism was explored by western blot and Co-IP assays. RESULTS: Downregulated CHMP5 was identified by proteomics in OA patient cartilages, which was verified in human and mouse articular cartilages. CHMP5 overexpression repressed cell apoptosis and ECM degradation in OA chondrocytes. However, silencing CHMP5 exacerbated OA chondrocyte apoptosis and ECM degradation. Furthermore, we found that the protective effect of CHMP5 against OA was involved in nuclear factor kappa B (NF-κB) signaling pathway. CONCLUSIONS: This study demonstrated that CHMP5 repressed IL-1ß-induced chondrocyte apoptosis and ECM degradation and blocked NF-κB activation. It was shown that CHMP5 might be a novel potential therapeutic target for OA in the future.


Apoptosis , Chondrocytes , Extracellular Matrix , Hyaluronoglucosaminidase , NF-kappa B , Osteoarthritis , Signal Transduction , Animals , Humans , Male , Mice , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/genetics , Proteomics/methods
9.
Nat Commun ; 15(1): 3225, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38622181

Osteoarthritis (OA) is a painful, incurable disease affecting over 500 million people. Recent clinical trials of the nerve growth factor (NGF) inhibitors in OA patients have suggested adverse effects of NGF inhibition on joint structure. Here we report that nerve growth factor receptor (NGFR) is upregulated in skeletal cells during OA and plays an essential role in the remodeling and repair of osteoarthritic joints. Specifically, NGFR is expressed in osteochondral cells but not in skeletal progenitor cells and induced by TNFα to attenuate NF-κB activation, maintaining proper BMP-SMAD1 signaling and suppressing RANKL expression in mice. NGFR deficiency hyper-activates NF-κB in murine osteoarthritic joints, which impairs bone formation and enhances bone resorption as exemplified by a reduction in subchondral bone and osteophytes. In human OA cartilage, NGFR is also negatively associated with NF-κB activation. Together, this study suggests a role of NGFR in limiting inflammation for repair of diseased skeletal tissues.


Cartilage, Articular , Osteoarthritis , Humans , Mice , Animals , Receptor, Nerve Growth Factor , NF-kappa B , Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor , Inflammation , Cartilage, Articular/metabolism , Joints/metabolism
10.
Exp Gerontol ; 190: 112413, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38570055

BACKGROUND: Osteoporotic osteoarthritis (OP-OA) is a severe pathological form of OA, urgently requiring precise management strategies and more efficient interventions. Emodin (Emo), an effective ingredient found in the traditional Chinese medicine rhubarb, has been dEmonstrated to promote osteogenesis and inhibit extracellular matrix degradation. In this study, we aimed to investigate the interventional effects of Emo on the subchondral bone and cartilage of the knee joints in OP-OA model rats. METHODS: Thirty-two SD rats were randomly and equally divided into sham, OP-OA, Emo low-dose, and Emo high-dose groups. Micro-CT scanning was conducted to examine the bone microstructure of the rat knee joints. H&E and Safranin O and Fast Green staining (SO&FG) were performed for the pathomorphological evaluation of the rat cartilage tissues. ELISA was used to estimate the rat serum expression levels of inflammatory factors, including interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α). Additionally, the CCK-8 assay was utilized for determining the viability of Emo-treated BMSCs. Western blot and real-time PCR analyses were also employed to measure the bone formation indexes and cartilage synthesis and decomposition indexes. Lastly, the osteogenic and chondrogenic differentiation efficiency of the BMSCs was investigated via Alizarin Red and Alcian Blue staining. RESULTS: Emo intervention alleviated the bone microstructural disruption of the subchondral bone and articular cartilage in the OP-OA rats and up-regulated the expression of bone and cartilage anabolic metabolism indicators, decreased the expression of cartilage catabolism indicators, and diminished the expression of inflammatory factors in the rat serum (P<0.05). Furthermore, Emo reversed the decline in the osteogenic and chondrogenic differentiation ability of the BMSCs (P<0.05). CONCLUSION: Emo intervention mitigates bone loss and cartilage damage in OP-OA rats and promotes the osteogenic and chondrogenic differentiation of BMSCs.


Cartilage, Articular , Emodin , Osteoporosis , Rats, Sprague-Dawley , X-Ray Microtomography , Animals , Emodin/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Rats , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Female , Disease Models, Animal , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/pathology
11.
Arthritis Res Ther ; 26(1): 91, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664820

OBJECTIVE: To characterize aspects of triiodothyronine (T3) induced chondrocyte terminal maturation within the molecular osteoarthritis pathophysiology using the previously established T3 human ex vivo osteochondral explant model. DESIGNS: RNA-sequencing was performed on explant cartilage obtained from OA patients (n = 8), that was cultured ex vivo with or without T3 (10 ng/ml), and main findings were validated using RT-qPCR in an independent sample set (n = 22). Enrichment analysis was used for functional clustering and comparisons with available OA patient RNA-sequencing and GWAS datasets were used to establish relevance for OA pathophysiology by linking to OA patient genomic profiles. RESULTS: Besides the upregulation of known hypertrophic genes EPAS1 and ANKH, T3 treatment resulted in differential expression of 247 genes with main pathways linked to extracellular matrix and ossification. CCDC80, CDON, ANKH and ATOH8 were among the genes found to consistently mark early, ongoing and terminal maturational OA processes in patients. Furthermore, among the 37 OA risk genes that were significantly affected in cartilage by T3 were COL12A1, TNC, SPARC and PAPPA. CONCLUSIONS: RNA-sequencing results show that metabolic activation and recuperation of growth plate morphology are induced by T3 in OA chondrocytes, indicating terminal maturation is accelerated. The molecular mechanisms involved in hypertrophy were linked to all stages of OA pathophysiology and will be used to validate disease models for drug testing.


Cartilage, Articular , Chondrocytes , Osteoarthritis , Osteogenesis , Triiodothyronine , Humans , Triiodothyronine/pharmacology , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Chondrocytes/metabolism , Chondrocytes/drug effects , Chondrocytes/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Osteogenesis/drug effects , Osteogenesis/physiology , Osteogenesis/genetics , Female , Biomimetics/methods , Male , Aged , Middle Aged
12.
PLoS One ; 19(4): e0298575, 2024.
Article En | MEDLINE | ID: mdl-38593124

Osteoarthritis (OA) is a widespread chronic, progressive, degenerative joint disease that causes pain and disability. Current treatments for OA have limited effectiveness and new biomarkers need to be identified. Bioinformatics analysis was conducted to explore differentially expressed genes and DNA repair/recombination protein 54 L (RAD54L) was selected. We firstly overexpressed RAD54L in interleukin-1ß (IL-1ß)-induced human articular chondrocytes or in OA rats to investigate its effect on OA. Chondrocyte viability and apoptotic rate were measured by Cell Counting Kit-8 and flow cytometry, respectively. Then we evaluated OA severity in vivo by Hematoxylin-eosin staining and Osteoarthritis Research Society International standards. The expression of inflammatory mediators was tested by enzyme-linked immunosorbent assay. Finally, western blot was performed to determine the relative expression level of hypoxia-inducible factors 1α (HIF-1α) and vascular endothelial growth factor (VEGF). Overexpression of RAD54L promoted cell viability and attenuated apoptosis in IL-1ß-induced human chondrocytes. A lower Osteoarthritis Research Society International score and a remarkable alleviation of chondrocyte disordering and infiltration of inflammatory cells were found in cartilage tissues of OA rats after overexpressing RAD54L. The inflammatory response induced by OA was decreased by RAD54L overexpression in vitro and in vivo. In addition, RAD54L overexpression decreased the relative expression level of HIF-1α and VEGF. Overexpression of RAD54L could attenuate OA by suppressing the HIF-1α/VEGF signaling pathway, indicating that RAD54L may be a potential treatment target for OA.


Cartilage, Articular , Osteoarthritis , Animals , Humans , Rats , Apoptosis , Cartilage, Articular/metabolism , Chondrocytes/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
13.
Immun Inflamm Dis ; 12(4): e1211, 2024 Apr.
Article En | MEDLINE | ID: mdl-38602270

BACKGROUND: Traumatic cartilage injury is an important cause of osteoarthritis (OA) and limb disability, and toll-like receptors (TLRs) mediated innate immune response has been confirmed to play a crucial role in cartilage injury. In the previous study, we found that the activation of TLR8 molecules in injured articular cartilage was more obvious than other TLRs by establishing an animal model of knee impact injury in rabbits, and the changes of TLR8 molecules could significantly affect the process of articular cartilage injury and repair. OBJECTIVE: To verify how mir-99a-5p regulates TLR8 receptor mediated innate immune response to treat traumatic cartilage injury. METHODS: The impact of a heavy object on the medial condyle of the rabbit's knee joint caused damage to the medial condylar cartilage. Through pathological and imaging analysis, it was demonstrated whether the establishment of an animal model of traumatic cartilage injury was successful. Establishing a cell model by virus transfection of chondrocytes to demonstrate the role of TLR8 in the innate immune response to impact cartilage injury. Through transcriptome sequencing, potential targets of TLR8, mir-99a-5p, were predicted, and basic experiments were conducted to demonstrate how they interact with innate immune responses to impact cartilage damage. RESULTS: TLR8 is a receptor protein of the immune system, which is widely expressed in immune cells. In our study, we found that TLR8 expression is localized in lysosomes and endosomes. Mir-99a-5p can negatively regulate TLR8 to activate PI3K-AKT molecular pathway and aggravate cartilage damage. Inhibiting TLR8 expression can effectively reduce the incidence of articular cartilage damage. CONCLUSION: Based on the results from this study, mir-99a-5p may be an effective molecular marker for predicting traumatic cartilage injury and targeting TLR8 is a novel and promising approach for the prevention or early treatment of cartilage damage.


Cartilage, Articular , MicroRNAs , Animals , Rabbits , MicroRNAs/genetics , Toll-Like Receptor 8/metabolism , Phosphatidylinositol 3-Kinases , Knee Joint/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology
14.
BMC Musculoskelet Disord ; 25(1): 253, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38561728

BACKGROUND: The characteristics and therapeutic potential of subtypes of bone marrow mesenchymal stem cells (BMSCs) are largely unknown. Also, the application of subpopulations of BMSCs in cartilage regeneration remains poorly characterized. The aim of this study was to explore the regenerative capacity of CD146-positive subpopulations of BMSCs for repairing cartilage defects. METHODS: CD146-positive BMSCs (CD146 + BMSCs) were sorted by self-developed CD146-specific lipid magnetic spheres (CD146-LMS). Cell surface markers, viability, and proliferation were evaluated in vitro. CD146 + BMSCs were subjected to in vitro chondrogenic induction and evaluated for chondrogenic properties by detecting mRNA and protein expression. The role of the CD146 subpopulation of BMSCs in cartilage damage repair was assessed by injecting CD146 + BMSCs complexed with sodium alginate gel in the joints of a mouse cartilage defect model. RESULTS: The prepared CD146-LMS had an average particle size of 193.7 ± 5.24 nm, an average potential of 41.9 ± 6.21 mv, and a saturation magnetization intensity of 27.2 Am2/kg, which showed good stability and low cytotoxicity. The sorted CD146 + BMSCs highly expressed stem cell and pericyte markers with good cellular activity and cellular value-added capacity. Cartilage markers Sox9, Collagen II, and Aggrecan were expressed at both protein and mRNA levels in CD146 + BMSCs cells after chondrogenic induction in vitro. In a mouse cartilage injury model, CD146 + BMSCs showed better function in promoting the repair of articular cartilage injury. CONCLUSION: The prepared CD146-LMS was able to sort out CD146 + BMSCs efficiently, and the sorted subpopulation of CD146 + BMSCs had good chondrogenic differentiation potential, which could efficiently promote the repair of articular cartilage injury, suggesting that the sorted CD146 + BMSCs subpopulation is a promising seed cell for cartilage tissue engineering.


Cartilage, Articular , Mesenchymal Stem Cells , Animals , Mice , Cartilage, Articular/metabolism , CD146 Antigen/metabolism , Cell Differentiation , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Bone Marrow Cells/metabolism , Chondrogenesis , RNA, Messenger/metabolism , Magnetic Phenomena , Lipids
15.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38673933

The aim of this study was to provide a comprehensive understanding of similarities and differences in mRNAs, lncRNAs, and circRNAs within cartilage for Kashin-Beck disease (KBD) compared to osteoarthritis (OA). We conducted a comparison of the expression profiles of mRNAs, lncRNAs, and circRNAs via whole-transcriptome sequencing in eight KBD and ten OA individuals. To facilitate functional annotation-enriched analysis for differentially expressed (DE) genes, DE lncRNAs, and DE circRNAs, we employed bioinformatic analysis utilizing Gene Ontology (GO) and KEGG. Additionally, using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), we validated the expression levels of four cartilage-related genes in chondrocytes. We identified a total of 43 DE mRNAs, 1451 DE lncRNAs, and 305 DE circRNAs in KBD cartilage tissue compared to OA (q value < 0.05; |log2FC| > 1). We also performed competing endogenous RNA network analysis, which identified a total of 65 lncRNA-mRNA interactions and 4714 miRNA-circRNA interactions. In particular, we observed that circRNA12218 had binding sites for three miRNAs targeting ACAN, while circRNA12487 had binding sites for seven miRNAs targeting COL2A1. Our results add a novel set of genes and non-coding RNAs that could potentially serve as candidate diagnostic biomarkers or therapeutic targets for KBD patients.


Kashin-Beck Disease , Osteoarthritis , RNA, Circular , RNA, Long Noncoding , RNA, Messenger , Transcriptome , Humans , Kashin-Beck Disease/genetics , RNA, Long Noncoding/genetics , Male , Female , Middle Aged , RNA, Circular/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics , Osteoarthritis/genetics , Gene Expression Profiling/methods , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Aged , Knee Joint/pathology , Knee Joint/metabolism , MicroRNAs/genetics , Collagen Type II/genetics , Collagen Type II/metabolism , Computational Biology/methods , Chondrocytes/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism , Gene Expression Regulation , Gene Ontology , Adult
16.
J Cell Mol Med ; 28(8): e18327, 2024 Apr.
Article En | MEDLINE | ID: mdl-38661437

Cartilage defects in the knee are often associated with the progression of degenerative osteoarthritis (OA), and cartilage repair is a useful strategy for managing this disease. However, cartilage repair is challenging because of the unique environment within the tissue. Recently, stem cell-based therapies have shed new light on this issue. In this study, we prepared exosomes (EXOs) from cartilage stem/progenitor cells (CSPCs) and found that treatment with EXOs increased the viability, migration, and proliferation of cultured primary chondrocytes. In a subacute OA rat model, the application of EXOs facilitated cartilage regeneration as evidenced by histological staining. Exosomal protein analysis together with bioinformatics suggested that cyclin-dependent kinase 9 (CDK9) is a key factor for chondrocyte growth and migration. Functional studies confirmed this prediction, that is, inhibiting CDK9 reduced the beneficial effects induced by EXOs in primary chondrocytes; while overexpression of CDK9 recapitulated the EXOs-induced phenotypes. RNA-Seq data showed that a set of genes involved in cell growth and migration were up-regulated by EXOs in chondrocytes. These changes could be partially reproduced by CDK9 overexpression. Overall, our data suggest that EXOs derived from primary CSPCs hold great therapeutic potential for treating cartilage defect-associated disorders such as degenerative OA, and that CDK9 is a key factor in this process.


Cartilage, Articular , Cell Proliferation , Chondrocytes , Disease Models, Animal , Exosomes , Animals , Exosomes/metabolism , Rats , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Stem Cells/metabolism , Stem Cells/cytology , Cell Movement , Rats, Sprague-Dawley , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/genetics , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/therapy , Male , Cells, Cultured , Regeneration , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/therapy
17.
ACS Appl Mater Interfaces ; 16(17): 21383-21399, 2024 May 01.
Article En | MEDLINE | ID: mdl-38626424

Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1ß-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.


Activating Transcription Factors , Chondrocytes , Exosomes , Mitochondria , Osteoarthritis , RNA, Messenger , Unfolded Protein Response , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/therapy , Exosomes/metabolism , Exosomes/chemistry , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Unfolded Protein Response/drug effects , Activating Transcription Factors/metabolism , Activating Transcription Factors/chemistry , Activating Transcription Factors/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Chondrocytes/metabolism , Chondrocytes/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Hydrogels/chemistry , Male , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/drug effects
18.
Sci Adv ; 10(16): eadk8402, 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38640238

Osteoarthritis (OA) treatment is limited by the lack of effective nonsurgical interventions to slow disease progression. Here, we examined the contributions of the subchondral bone properties to OA development. We used parathyroid hormone (PTH) to modulate bone mass before OA initiation and alendronate (ALN) to inhibit bone remodeling during OA progression. We examined the spatiotemporal progression of joint damage by combining histopathological and transcriptomic analyses across joint tissues. The additive effect of PTH pretreatment before OA initiation and ALN treatment during OA progression most effectively attenuated load-induced OA pathology. Individually, PTH directly improved cartilage health and slowed the development of cartilage damage, whereas ALN primarily attenuated subchondral bone changes associated with OA progression. Joint damage reflected early transcriptomic changes. With both treatments, the structural changes were associated with early modulation of immunoregulation and immunoresponse pathways that may contribute to disease mechanisms. Overall, our results demonstrate the potential of subchondral bone-modifying therapies to slow the progression of OA.


Cartilage, Articular , Osteoarthritis , Parathyroid Hormone , Animals , Mice , Alendronate/pharmacology , Alendronate/therapeutic use , Bone and Bones , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Bone Remodeling/drug effects , Weight-Bearing
19.
Med Sci Monit ; 30: e943738, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38664941

BACKGROUND The pathological mechanism of osteoarthritis is still unclear. The regulation of the immune microenvironment has been of growing interest in the progression and treatment of osteoarthritis. Macrophages with different phenotypes, producing different cytokines, have been linked to the mechanism of cartilage injury in osteoarthritis. Copper ions play a role in the immune response and are involved in the pathological mechanisms of osteoarthritis by affecting the metabolism of the cartilage matrix. Bioactive glass (BG) is an osteogenic material with superior biocompatibility. Here, we report on the regulatory behavior of macrophages using a copper-based composite BG material. MATERIAL AND METHODS Cu-BGC powder was prepared by sol-gel method, and scaffolds were fabricated and characterized using 3D printing. Macrophage cultures grown with Cu-BGC were examined for cell culture and proliferation. The effect of Cu-BGC on the degradation metabolism of chondrocytes, cultured in the environment of inflammatory cytokine IL-1ß, was determined. In addition, the morphology of macrophages, secretion of inflammatory cytokines, and expression of surface markers were examined. RESULTS The results show that Cu-BGC promotes macrophage proliferation at a range of concentrations and increases the secretion of anti-inflammatory cytokines while inhibiting proinflammatory cytokines. At the same time, M2-type cell surface markers are definitely expressed and the morphology of macrophages is altered. In addition, Cu-BGC inhibited the degradation metabolism of chondrocytes in the inflammatory environment induced by IL-1ß. CONCLUSIONS These results suggest that Cu-BGC induced macrophage polarization into an M2 type anti-inflammatory phenotype, and inhibition of immune injury response may play a role in delaying cartilage matrix damage in osteoarthritis.


Cell Proliferation , Chondrocytes , Copper , Cytokines , Macrophages , Osteoarthritis , Macrophages/metabolism , Macrophages/drug effects , Osteoarthritis/pathology , Osteoarthritis/metabolism , Animals , Chondrocytes/metabolism , Chondrocytes/drug effects , Chondrocytes/pathology , Copper/metabolism , Copper/pharmacology , Cytokines/metabolism , Mice , Cell Proliferation/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage/metabolism , Cartilage/drug effects , Cartilage/pathology , RAW 264.7 Cells , Glass , Tissue Scaffolds
20.
Int J Biol Macromol ; 266(Pt 2): 131259, 2024 May.
Article En | MEDLINE | ID: mdl-38574937

This study presents an alginate-collagen interpenetrating network (IPN) matrix of incorporating collagen fibrils into an alginate hydrogel by physical mixing and controlled gelation. The resulting matrix closely mimics the physiological and pathological stiffness range of the chondrocyte pericellular matrix (PCM). Chondrocytes were cultured within three-dimensional (3D) alginate-collagen IPN matrices with varying stiffness, namely Firm, Medium, and Soft. Alginate lyase was introduced to study the effects of the changes in stiffness of the Firm on chondrocyte response by in situ softening. The developed alginate-collagen IPN matrix displayed good cell-biocompatibility. Compared with stiffer tissue culture plastic (TCP), chondrocytes grown within Firm displayed a stabilized differentiated phenotype characterized by higher expression levels of aggrecan, collagen II, and SOX-9. Moreover, the developed alginate-collagen IPN matrix exhibited a gradually increased percentage of propidium iodide (PI)-positive dead cells with decreasing stiffness. Softer matrices directed cells towards higher proliferation rates and spherical morphologies while stimulating chondrocyte cluster formation. Furthermore, reducing Firm stiffness by in situ softening decreased aggrecan expression, contributing to matrix degradation similar to that seen in osteoarthritis (OA). Hence, the 3D alginate-collagen IPN constructs hold significant potential for in vitro replicating PCM stiffness changes observed in OA cartilage.


Alginates , Chondrocytes , Collagen , Osteoarthritis , Alginates/chemistry , Chondrocytes/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Collagen/metabolism , Collagen/chemistry , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Hydrogels/chemistry , Animals , Humans , Tissue Scaffolds/chemistry , Cell Proliferation , Cells, Cultured , Aggrecans/metabolism , Aggrecans/genetics , Tissue Engineering/methods
...