Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 10.788
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.
Medicine (Baltimore) ; 103(19): e38065, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728521

Knee varus (KV) deformity leads to abnormal forces in the different compartments of the joint cavity and abnormal mechanical loading thus leading to knee osteoarthritis (KOA). This study used computer-aided design to create 3-dimensional simulation models of KOA with varying varus angles to analyze stress distribution within the knee joint cavity using finite element analysis for different varus KOA models and to compare intra-articular loads among these models. Additionally, we developed a cartilage loading model of static KV deformity to correlate with dynamic clinical cases of cartilage injury. Different KV angle models were accurately simulated with computer-aided design, and the KV angles were divided into (0°, 3°, 6°, 9°, 12°, 15°, and 18°) 7 knee models, and then processed with finite element software, and the Von-Mises stress distribution and peak values of the cartilage of the femoral condyles, medial tibial plateau, and lateral plateau were obtained by simulating the human body weight in axial loading while performing the static extension position. Finally, intraoperative endoscopy visualization of cartilage injuries in clinical cases corresponding to KV deformity subgroups was combined to find cartilage loading and injury correlations. With increasing varus angle, there was a significant increase in lower limb mechanical axial inward excursion and peak Von-Mises stress in the medial interstitial compartment. Analysis of patients' clinical data demonstrated a significant correlation between varus deformity angle and cartilage damage in the knee, medial plateau, and patellofemoral intercompartment. Larger varus deformity angles could be associated with higher medial cartilage stress loads and increased cartilage damage in the corresponding peak stress area. When the varus angle exceeds 6°, there is an increased risk of cartilage damage, emphasizing the importance of early surgical correction to prevent further deformity and restore knee function.


Cartilage, Articular , Finite Element Analysis , Knee Joint , Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/physiopathology , Osteoarthritis, Knee/surgery , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Knee Joint/physiopathology , Male , Weight-Bearing/physiology , Biomechanical Phenomena , Middle Aged , Stress, Mechanical , Female , Computer Simulation , Aged
3.
Jt Dis Relat Surg ; 35(2): 330-339, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38727112

OBJECTIVES: The study aims to investigate the relationship between the vastus medialis obliquus (VMO) muscle distal insertion features and patellar chondral lesion presence. PATIENTS AND METHODS: This cross-sectional study included a total of 100 patients (18 males, 82 females, mean age 67.2±7.1 years; range, 50 to 86 years) who underwent total knee arthroplasty (TKA). Radiological assessments, including merchant view and standing orthoroentgenograms, were conducted. The current osteoarthritis stage, varus angle, quadriceps angle (Q angle), patella-patellar tendon angle (P-PT angle), congruence angle, and sulcus angle were calculated. The VMO tendon length, muscle fiber angle, tendon insertion width measurements, and patellar chondral lesion localization data were obtained intraoperatively. Grouping was done according to the distal insertion width of the VMO tendon to the medial edge of the patella. The medial rim of the patella was divided into three equal-sized sectors. The first group (Group 1, n=31) consisted of patients who had an insertion from the quadriceps tendon into the upper one-third of the patella. The second group (Group 1, n=48) consisted of patients with a distal insertion expanding into the middle one-third of the patella. The third group (Group 3, n=21) consisted of patients who had a distal insertion extending into the distal third region of the medial patella margin. The patella joint surface was divided into sectors, and the presence and location of cartilage lesions were noted in detail. RESULTS: The mean tendon insertion width rate was 45.99±16.886% (range, 16.7 to 83.3%). The mean muscle fiber insertion angle was 51.85±11.67º (range, 20º to 80º). The mean tendon length was 12.45±3.289 (range, 4 to 20) mm. There was no significant difference between the mean age, weight, height, body mass index, BMI, fiber angle, tendon length, varus angle, Q angle, sulcus angle, and congruence angle data among the groups. In terms of the P-PT angle, Groups 1 and 2 had a significant relationship (p=0.008). No relationship was found between the mean fiber insertion angle, mean tendon length, or the presence of chondral lesions. There was a statistically significant difference among the groups regarding the presence of chondral lesions. The highest percentage of chondral lesion frequency was observed in Group 3 (95.24%), followed by Group 1 (90.3%) and Group 2 (89.6%), respectively. Compared to the other two groups, Group 3 had a higher average ratio of lesion areas per patient. CONCLUSION: Our study results demonstrate that the formation and localization of the patellar chondral lesions are affected by the insertion width type of the VMO muscle into the patella. Group 2-type insertion is associated with a lower lesion frequency rate than Groups 1 and 3.


Patella , Quadriceps Muscle , Humans , Female , Male , Middle Aged , Aged , Quadriceps Muscle/pathology , Quadriceps Muscle/diagnostic imaging , Cross-Sectional Studies , Aged, 80 and over , Patella/pathology , Patella/diagnostic imaging , Patella/anatomy & histology , Arthroplasty, Replacement, Knee , Osteoarthritis, Knee/surgery , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/diagnostic imaging , Cartilage, Articular/pathology , Cartilage, Articular/diagnostic imaging , Radiography , Knee Joint/pathology , Knee Joint/surgery , Knee Joint/diagnostic imaging
4.
Jt Dis Relat Surg ; 35(2): 361-367, 2024 Mar 21.
Article En | MEDLINE | ID: mdl-38727116

OBJECTIVES: This study aims to compare the radiological, biomechanical, and histopathological results of microfracture treatment and osteochondral damage repair treatment with a new scaffold product produced by the three-dimensional (3D) bioprinting method containing gelatin-hyaluronic acid-alginate in rabbits with osteochondral damage. MATERIALS AND METHODS: A new 3D bioprinted scaffold consisting of gelatin, hyaluronic acid, and alginate designed by us was implanted into the osteochondral defect created in the femoral trochlea of 10 rabbits. By randomization, it was determined which side of 10 rabbits would be repaired with a 3D bioprinted scaffold, and microfracture treatment was applied to the other knees of the rabbits. After six months of follow-up, the rabbits were sacrificed. The results of both treatment groups were compared radiologically, biomechanically, and histopathologically. RESULTS: None of the rabbits experienced any complications. The magnetic resonance imaging evaluation showed that all osteochondral defect areas were integrated with healthy cartilage in both groups. There was no significant difference between the groups in the biomechanical load test (p=0.579). No statistically significant difference was detected in the histological examination using the modified Wakitani scores (p=0.731). CONCLUSION: Our study results showed that 3D bioprinted scaffolds exhibited comparable radiological, biomechanical, and histological properties to the conventional microfracture technique for osteochondral defect treatment.


Alginates , Bioprinting , Cartilage, Articular , Gelatin , Hyaluronic Acid , Knee Joint , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Rabbits , Alginates/chemistry , Gelatin/chemistry , Hyaluronic Acid/chemistry , Hyaluronic Acid/therapeutic use , Tissue Scaffolds/chemistry , Cartilage, Articular/pathology , Cartilage, Articular/injuries , Cartilage, Articular/surgery , Knee Joint/surgery , Knee Joint/pathology , Bioprinting/methods , Disease Models, Animal , Biomechanical Phenomena , Magnetic Resonance Imaging , Arthroplasty, Subchondral/methods
5.
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
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.
Foot Ankle Clin ; 29(2): 213-224, 2024 Jun.
Article En | MEDLINE | ID: mdl-38679434

Osteochondral lesions of the talus (OLTs) are the lesions that affect the articular cartilage and the subchondral bone of the talus. Symptoms develop between 6 and 12 months after the index trauma and are associated with degradation of quality of life. Two-thirds of the lesions (73%) are located on the medial part of the talus, 28% of the lesions are posteromedial, and 31% of the lesions are centromedial. Currently, OLT of up to 100 mm2 can behave in a more indolent condition, and above that area, the defect tends to transmit more shearing forces to adjacent cartilage and is more symptomatic.


Cartilage, Articular , Quality of Life , Talus , Humans , Talus/injuries , Talus/pathology , Cartilage, Articular/pathology , Cartilage, Articular/injuries , Osteochondritis/surgery
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.
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
13.
Eur Rev Med Pharmacol Sci ; 28(7): 2670-2676, 2024 Apr.
Article En | MEDLINE | ID: mdl-38639506

BACKGROUND: Synovial chondromatosis is a non-malignant synovial disorder characterized by the presence of cartilage formation within the synovial membrane, leading to the emergence of multiple cartilaginous nodules that may be either attached or unattached. The presence of this anatomical feature is frequently observed in articulations such as the knee, hip, elbow, and ankle. CASE REPORT: In this study, we present a case of synovial chondromatosis in the knee joint of a healthy male in his early 60s. Notably, the patient exhibited the simultaneous presence of 87 large loose bodies. The occurrence of a substantial quantity of unattached entities of notable dimensions within the joint is highly uncommon. CONCLUSIONS: The patient had several synovial chondromas, a rare disease. Synovial chondromatosis is a benign disorder; however, growing synovium can cause pyogenic cartilage nodules. Most loose bodies in joints can abrade and degenerate articular cartilage, causing long-term discomfort. Thus, an early-stage procedure to remove loose bodies and carefully excise synovial tissue is necessary to treat this condition.


Cartilage, Articular , Chondromatosis, Synovial , Humans , Male , Chondromatosis, Synovial/diagnostic imaging , Chondromatosis, Synovial/surgery , Chondromatosis, Synovial/pathology , Synovial Membrane/pathology , Knee Joint/diagnostic imaging , Knee Joint/surgery , Knee Joint/pathology , Cartilage, Articular/pathology , Ankle Joint
14.
Sci Rep ; 14(1): 8046, 2024 04 05.
Article En | MEDLINE | ID: mdl-38580748

Osteoarthritis is a common chronic disease and major cause of disability and chronic pain in ageing populations. In this pathology, the entire joint is involved, and the regeneration of articular cartilage still remains one of the main challenges. Here, we investigated the molecular mechanisms underlying cartilage regeneration in young mice using a full-thickness cartilage injury (FTCI) model. FTCI-induced cartilage defects were created in the femoral trochlea of young and adult C57BL/6 mice. To identify key molecules and pathways involved in the early response to cartilage injury, we performed RNA sequencing (RNA-seq) analysis of cartilage RNA at 3 days after injury. Young mice showed superior cartilage regeneration compared to adult mice after cartilage injury. RNA-seq analysis revealed significant upregulation of genes associated with the immune response, particularly in the IFN-γ signaling pathway and qRT-PCR analysis showed macrophage polarization in the early phase of cartilage regeneration (3 days) in young mice after injury, which might promote the removal of damaged or necrotic cells and initiate cartilage regeneration in response to injury. IFN-γR1- and IFN-γ-deficient mice exhibited impaired cartilage regeneration following cartilage injury. DMM-induced and spontaneous OA phenotypes were exacerbated in IFN-γR1-/- mice than in wild-type mice. Our data support the hypothesis that IFN-γ signaling is necessary for cartilage regeneration, as well as for the amelioration of post-traumatic and age-induced OA.


Cartilage, Articular , Osteoarthritis , Animals , Mice , Cartilage, Articular/pathology , Disease Models, Animal , Interferon-gamma/genetics , Mice, Inbred C57BL , Osteoarthritis/metabolism , Regeneration , Signal Transduction
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.
Foot Ankle Clin ; 29(2): 185-192, 2024 Jun.
Article En | MEDLINE | ID: mdl-38679432

The current concepts thoroughly highlight the ankle cartilage cascade focusing on the different stages and the different etiologic factors that can introduce a patient into the cascade. Moreover, the authors will provide the reader with a comprehensive overview of the types of lesions that may present as symptomatic, asymptomatic, and dangerous for progression into osteoarthritis, and the authors supply the reader with considerations and directions for future clinical implications and scientific endeavors.


Ankle Joint , Cartilage, Articular , Humans , Cartilage, Articular/pathology , Osteoarthritis/etiology , Incidence , Ankle Injuries/complications , Terminology as Topic , Cartilage Diseases
17.
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
18.
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
19.
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
20.
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
...