cartilage protection

  • 文章类型: Journal Article
    背景:预防关节水肿对于阻止骨关节炎(OA)进展至关重要。越来越多的临床证据表明,健脾通络方(JTF)可能具有有希望的抗水肿作用。然而,JTF的治疗特性和潜在机制尚不清楚.
    方法:根据动态组织病理学评估和显微CT观察,建立了OA大鼠模型并用于评估JTF的体内药理作用。然后,通过临床转录组数据分析和“疾病基因-药物靶标”网络分析,确定了OA相关基因和JTF的潜在靶标,通过一系列体内实验验证。
    结果:JTF能有效减轻疼痛和关节水肿,抑制基质降解,软骨细胞凋亡,OA大鼠水通道蛋白的表达。值得注意的是,JTF剂量依赖性逆转损伤相关分子模式和炎症因子上调。机械上,我们的“疾病基因-药物靶标”网络分析表明,NCOA4-HMGB1-GSK3B-AQPs轴,与铁凋亡和水通道蛋白失调有关,可能作为JTF对抗OA的目标。因此,JTF减轻了NCOA4、HMGB1和GSK3B的表达,氧化应激,和OA大鼠的铁代谢异常。此外,JTF处理显著减弱OA大鼠软骨组织中观察到的AQP1、AQP3和AQP4蛋白的异常上调。
    结论:我们的数据首次揭示了JTF可能通过抑制NCOA4-HMGB1驱动的铁蛋白凋亡和水通道蛋白失调在骨关节炎治疗中发挥软骨保护和抗水肿作用。
    BACKGROUND: Preventing joint edema is crucial in halting osteoarthritis (OA) progression. Growing clinical evidence indicate that Jianpi-Tongluo Formula (JTF) may have a promising anti-edema effect. However, the therapeutic properties of JTF and the underlying mechanisms remains unclear.
    METHODS: An OA rat model was established and employed to evaluate pharmacological effects of JTF in vivo based on dynamic histopathologic assessments and micro-CT observations. Then, OA-related genes and potential targets of JTF were identified through clinical transcriptomic data analysis and \"disease gene-drug target\" network analysis, which were verified by a series of in vivo experiments.
    RESULTS: JTF administration effectively reduced pain and joint edema, inhibited matrix degradation, chondrocyte apoptosis, and aquaporin expression in OA rats. Notably, JTF dose-dependently reversed damage-associated molecular patterns and inflammatory factor upregulation. Mechanically, our \"disease gene-drug target\" network analysis indicated that the NCOA4-HMGB1-GSK3B-AQPs axis, implicated in ferroptosis and aquaporin dysregulation, may be potentially served as a target of JTF against OA. Accordingly, JTF mitigated NCOA4, HMGB1, and GSK3B expression, oxidative stress, and iron metabolism aberrations in OA rats. Furthermore, JTF treatment significantly attenuated the aberrant upregulation of AQP1, AQP3, and AQP4 proteins observed in cartilage tissues of OA rats.
    CONCLUSIONS: Our data reveal for the first time that JTF may exert cartilage protective and anti-edema effects in osteoarthritis therapy by inhibiting NCOA4-HMGB1-driven ferroptosis and aquaporin dysregulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    包括活性氧(ROS)在内的土壤中的正火,一氧化氮(NO),无细胞DNA,并调节与炎症相关的种子,如巨噬细胞,中性粒细胞,成纤维细胞,代表了维持类风湿关节炎(RA)治疗滑膜组织稳态的有希望的策略。在这里,ROS清除两亲性嵌段共聚物聚乙二醇化胆红素和NO清除聚乙二醇化邻苯二胺被制造成自组装成负载有JAK抑制剂诺波特罗的双重响应纳米颗粒(Not@BR/oPDA-PEG,NBOPNPs)。同时ROS和NO耗竭结合JAK-STAT通路抑制不仅可以促进M2极化,进一步减少ROS和NO的生成,但也减少细胞因子和趋化因子,以防止免疫细胞募集。具体来说,NBOPNPs对高水平ROS和NO的反应,并随着疏水头BR和oPDA转化为亲水头而崩解,在接头中释放出诺普特罗。释放的诺普特罗可以抑制炎症细胞的JAK-STAT通路,减少促炎细胞因子和趋化因子的分泌。该策略代表了通过打破炎症加重的正反馈循环来调节RA土壤和种子的有效方法。在胶原诱导的关节炎大鼠模型中获得优异的抗RA功效。一起来看,我们的工作为调整RA土壤和种子以增强RA治疗提供了参考。
    Normalizing inflamed soils including reactive oxygen species (ROS), nitric oxide (NO), cell-free DNA, and regulating inflammation-related seeds such as macrophages, neutrophils, fibroblasts, represent a promising strategy to maintain synovial tissue homeostasis for rheumatoid arthritis (RA) treatment. Herein, ROS scavenging amphiphilic block copolymer PEGylated bilirubin and NO-scavenging PEGylated o-phenylenediamine were fabricated to self-assemble into a dually responsive nanoparticle loaded with JAK inhibitor notopterol (Not@BR/oPDA-PEG, NBOP NPs). The simultaneous ROS and NO depletion combined with JAK-STAT pathway inhibition could not only promote M2 polarization to reduce further ROS and NO generation, but also decrease cytokines and chemokines to prevent immune cell recruitment. Specifically, NBOP NPs responded to high level ROS and NO, and disintegrated to release notopterol in inflamed joints as the hydrophobic heads BR and oPDA were transformed into hydrophilic ones. The released notopterol could inhibit the JAK-STAT pathway of inflammatory cells to reduce the secretion of pro-inflammatory cytokines and chemokines. This strategy represented an effective way to regulate RA soils and seeds through breaking the positive feedback loop of inflammation aggravation, achieving an excellent anti-RA efficacy in a collagen-induced arthritis rat model. Taken together, our work offered a reference to adjust RA soils and seeds for enhanced RA treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    半月板损伤与软骨退化和骨关节炎(OA)的发展有关。当半月板损伤发生时,保护关节软骨和改善运动是具有挑战性的。在这里,受弯月面的成分和功能的启发,我们开发了一种自润滑和摩擦响应性的水凝胶,其中包含负载有双氯芬酸钠(DS)和KTogenin(KGN)的纳米脂质体,用于抗炎和软骨再生。当水凝胶被注射到半月板损伤部位时,负载药物的纳米脂质体以摩擦响应的方式从水凝胶中释放,并重新组装以形成在运动过程中润滑关节的水合层.同时,DS和KNG不断地从纳米脂质体中释放以减轻炎症并促进软骨再生。此外,这种水凝胶表现出良好的可注射性,机械性能,抗疲劳性,和长时间的退化。体内实验表明,水凝胶的注射有效地改善了运动能力,保护了大鼠的关节软骨,表明它是半月板损伤的潜在治疗方法。
    Meniscus injuries are associated with the degeneration of cartilage and development of osteoarthritis (OA). It is challenging to protect articular cartilage and improve exercise when a meniscus injury occurs. Herein, inspired by the components and functions of the meniscus, we developed a self-lubricating and friction-responsive hydrogel that contains nanoliposomes loaded with diclofenac sodium (DS) and Kartogenin (KGN) for anti-inflammation and cartilage regeneration. When the hydrogel was injected into the meniscus injury site, the drug-loaded nanoliposomes were released from the hydrogel in a friction-responsive manner and reassembled to form hydration layers that lubricate joints during movement. Meanwhile, DS and KNG were constantly released from the nanoliposomes to mitigate inflammation and promote cartilage regeneration. Additionally, this hydrogel exhibited favorable injectability, mechanical properties, fatigue resistance, and prolonged degradation. In vivo experiments demonstrated that injection of the hydrogel effectively improved exercise performance and protected the articular cartilage of rats, suggesting it as a potential therapeutic approach for meniscal injuries.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    类风湿性关节炎(RA)是一种以炎症为特征的慢性自身免疫性疾病,自身免疫功能障碍,软骨和骨骼破坏。在这次审查中,我们总结了关于灵芝多糖(GLP)在抗炎方面对RA的保护作用的现有报道,免疫调节,抗血管生成和骨保护作用。首先,GLP抑制RA滑膜成纤维细胞(RASF)增殖和迁移,调节促炎和抗炎细胞因子并减少滑膜炎症。其次,GLP调节抗原呈递细胞如树突状细胞的增殖和分化,抑制单核巨噬细胞和自然杀伤(NK)细胞的吞噬作用,并调节M1,M2和相关炎性细胞因子的比例。此外,GLP在平衡体液和细胞免疫中产生活性,例如调节免疫球蛋白的产生,调节T和B淋巴细胞增殖反应和细胞因子释放,表现出免疫调节作用。第三,GLP通过直接抑制血管内皮细胞增殖和诱导细胞死亡以及间接抑制细胞中血管内皮生长因子(VEGF)的产生来抑制血管生成。最后,GLP可抑制基质金属蛋白酶的产生,促进成骨细胞的形成,对骨骼和关节软骨发挥保护作用。提示GLP可能是治疗RA的有希望的药物。
    Rheumatoid arthritis (RA) is a chronic and autoimmune disease characterized by inflammation, autoimmune dysfunction, and cartilage and bone destruction. In this review, we summarized the available reports on the protective effects of Ganoderma lucidum polysaccharides (GLP) on RA in terms of anti-inflammatory, immunomodulatory, anti-angiogenic and osteoprotective effects. Firstly, GLP inhibits RA synovial fibroblast (RASF) proliferation and migration, modulates pro- and anti-inflammatory cytokines and reduces synovial inflammation. Secondly, GLP regulates the proliferation and differentiation of antigen-presenting cells such as dendritic cells, inhibits phagocytosis by mononuclear macrophages and nature killer (NK) cells and regulates the ratio of M1, M2 and related inflammatory cytokines. In addition, GLP produced activities in balancing humoral and cellular immunity, such as regulating immunoglobulin production, modulating T and B lymphocyte proliferative responses and cytokine release, exhibiting immunomodulatory effects. Thirdly, GLP inhibits angiogenesis through the direct inhibition of vascular endothelial cell proliferation and induction of cell death and the indirect inhibition of vascular endothelial growth factor (VEGF) production in the cells. Finally, GLP can inhibit the production of matrix metalloproteinases and promote osteoblast formation, exerting protective effects on bone and articular cartilage. It is suggested that GLP may be a promising agent for the treatment of RA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    类风湿性关节炎(RA)是一种自身免疫性疾病,其特征是严重的滑膜炎症和软骨损伤。尽管RA治疗取得了很大进展,仍然缺乏完全治愈RA患者的药物。在这里,我们提出了一种重编程的中性粒细胞细胞药物负载TNFα靶向siRNA(siTNFα)作为RA治疗的替代抗炎方法.负载的siTNFα不仅是抑制炎症滑膜中巨噬细胞产生TNFα的基因治疗剂,而且编辑也将嗜中性粒细胞重新编程为抗炎表型。利用中性粒细胞对炎症的积极倾向,重编程的siTNFα/中性粒细胞药物(siTNFα/TP/NEs)可以快速迁移到发炎的滑膜,将加载的siTNFα转移到巨噬细胞,然后显着降低TNFα表达,规避中性粒细胞的促炎活性,从而导致减轻滑膜炎症和改善软骨保护。我们的工作为RA治疗提供了一种有前途的细胞药物,并提出了基于活中性粒细胞的基因传递平台。
    Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe synovial inflammation and cartilage damage. Despite great progress in RA therapy, there still lacks the drugs to completely cure RA patients. Herein, we propose a reprogrammed neutrophil cytopharmaceuticals loading with TNFα-targeting-siRNA (siTNFα) as an alternative anti-inflammatory approach for RA treatment. The loaded siTNFα act as not only the gene therapeutics to inhibit TNFα production by macrophages in inflamed synovium, but also the editors to reprogram neutrophils to anti-inflammatory phenotypes. Leveraging the active tendency of neutrophils to inflammation, the reprogrammed siTNFα/neutrophil cytopharmaceuticals (siTNFα/TP/NEs) can rapidly migrate to the inflamed synovium, transfer the loaded siTNFα to macrophages followed by the significant reduction of TNFα expression, and circumvent the pro-inflammatory activity of neutrophils, thus leading to the alleviated synovial inflammation and improved cartilage protection. Our work provides a promising cytopharmaceutical for RA treatment, and puts forward a living neutrophil-based gene delivery platform.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨关节炎(OA)是一种以炎症和软骨破坏为特征的进行性关节疾病,其进展与M1/M2滑膜巨噬细胞的失衡密切相关。提出了一种调节细胞内/细胞外一氧化氮(NO)和氢质子重编程M1/M2滑膜巨噬细胞的双管齐下的策略。在“二合一”纳米载体(NAHA-CaP/siRNA纳米颗粒)中,碳酸酐酶IX(CA9)siRNA和NO清除剂的组合是通过清除NO和抑制滑膜巨噬细胞中的CA9表达而开发的,用于进行性OA治疗。体外实验表明,这些NPs可以明显清除与正常组相似的细胞内NO水平,并下调CA9mRNA的表达水平(约90%)。从而将M1巨噬细胞重新极化为M2表型,并增加软骨原TGF-β1mRNA的表达水平(约1.3倍),抑制软骨细胞凋亡。此外,体内实验表明,NPs具有很好的抗炎作用,软骨保护和修复作用,从而有效地减轻单碘乙酸诱导的早期和晚期OA小鼠模型中的OA进展以及内侧半月板诱导的OA大鼠模型的手术失稳。因此,siCA9和NO清除剂“二合一”递送系统是进行性OA治疗的潜在和有效策略。
    Osteoarthritis (OA) is a progressive joint disease characterized by inflammation and cartilage destruction, and its progression is closely related to imbalances in the M1/M2 synovial macrophages. A two-pronged strategy for the regulation of intracellular/extracellular nitric oxide (NO) and hydrogen protons for reprogramming M1/M2 synovial macrophages is proposed. The combination of carbonic anhydrase IX (CA9) siRNA and NO scavenger in \"two-in-one\" nanocarriers (NAHA-CaP/siRNA nanoparticles) is developed for progressive OA therapy by scavenging NO and inhibiting CA9 expression in synovial macrophages. In vitro experiments demonstrate that these NPs can significantly scavenge intracellular NO similar to the levels as those in the normal group and downregulate the expression levels of CA9 mRNA (≈90%), thereby repolarizing the M1 macrophages into the M2 phenotype and increasing the expression levels of pro-chondrogenic TGF-β1 mRNA (≈1.3-fold), and inhibiting chondrocyte apoptosis. Furthermore, in vivo experiments show that the NPs have great anti-inflammation, cartilage protection and repair effects, thereby effectively alleviating OA progression in both monoiodoacetic acid-induced early and late OA mouse models and a surgical destabilization of medial meniscus-induced OA rat model. Therefore, the siCA9 and NO scavenger \"two-in-one\" delivery system is a potential and efficient strategy for progressive OA treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:半月板损伤是运动医学领域的常见疾病。这种疾病的主要治疗方法是缓解疼痛,而不是半月板功能恢复。它可能导致不良预后并加速骨关节炎的进展。在这项研究中,设计半月板支架以达到半月板功能恢复和软骨保护的目的。
    方法:半月板支架是使用三重周期性最小表面(TPMS)方法设计的。使用有限元分析软件将支架模拟为三维(3D)完整的膝盖模型,以获得不同的力学测试结果。机械性能是通过万能机获得的。最后,建立体内模型以评估基于TPMS的半月板支架对软骨保护的影响。进行射线照相和组织学检查以评估软骨和骨结构。使用万能机测试再生半月板的不同区域以评估生物力学功能。
    结果:基于TPMS的半月板支架具有更大的体积分数和更长的功能周期性,表现出更好的机械性能,载荷传递和应力分布更接近天然生物力学环境。TPMS组的射线照相图像和组织学结果在软骨保护方面表现出比网格组更好的性能。TPMS组中的再生弯月面也具有与天然弯月面相似的机械性能。
    结论:TPMS方法可以通过调节体积分数和功能周期性来影响机械性能。基于TPMS的半月板支架显示出半月板再生和软骨保护的适当特征。
    BACKGROUND: The meniscus injury is a common disease in the area of sports medicine. The main treatment for this disease is the pain relief, rather than the meniscal function recovery. It may lead to a poor prognosis and accelerate the progression of osteoarthritis. In this study, we designed a meniscal scaffold to achieve the purposes of meniscal function recovery and cartilage protection.
    METHODS: The meniscal scaffold was designed using the triply periodic minimal surface (TPMS) method. The scaffold was simulated as a three-dimensional (3D) intact knee model using a finite element analysis software to obtain the results of different mechanical tests. The mechanical properties were gained through the universal machine. Finally, an in vivo model was established to evaluate the effects of the TPMS-based meniscal scaffold on the cartilage protection. The radiography and histological examinations were performed to assess the cartilage and bony structures. Different regions of the regenerated meniscus were tested using the universal machine to assess the biomechanical functions.
    RESULTS: The TPMS-based meniscal scaffold with a larger volume fraction and a longer functional periodicity demonstrated a better mechanical performance, and the load transmission and stress distribution were closer to the native biomechanical environment. The radiographic images and histological results of the TPMS group exhibited a better performance in terms of cartilage protection than the grid group. The regenerated meniscus in the TPMS group also had similar mechanical properties to the native meniscus.
    CONCLUSIONS: The TPMS method can affect the mechanical properties by adjusting the volume fraction and functional periodicity. The TPMS-based meniscal scaffold showed appropriate features for meniscal regeneration and cartilage protection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    随着生活方式的改变和人口老龄化,骨关节炎(OA)正在成为全球范围内的主要医疗负担。OA是一种慢性炎症和退行性疾病,最初表现为关节疼痛并最终导致永久性残疾。迄今为止,目前还没有可用于骨关节炎的明确治疗的药物,大多数疗法都是通过缓解症状而不是治愈疾病的。再加上对早期症状和诊断方法的模糊认识,这种疾病仍然是一个全球性问题,需要协调一致的研究努力。一系列事件从促炎细胞因子的产生开始调节骨关节炎的发作和进展,包括白细胞介素(IL)-1β,IL-6,肿瘤坏死因子(TNF)-α;分解代谢酶,如基质金属蛋白酶(MMP)-1,-3和-13,最终导致软骨破坏,失去润滑,疼痛,无法加载关节。虽然小分子和大分子的关节内注射通常是为了缓解症状,在滑膜腔内的低停留时间严重损害其功效。这篇综述将简要描述决定疾病发作和进展的因素,介绍目前临床上批准的治疗和诊断方法,最后阐述了纳米/微米药物在骨关节炎治疗中应用的主要挑战和机遇。因此,未来的治疗方案将受益于同时考虑机械生物学,炎症,和组织退化方面的疾病。本文分为:纳米技术生物学方法>生物学中的纳米级系统可植入材料和外科技术>纳米技术在组织修复和替换。
    With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α; catabolic enzymes, such as matrix metalloproteinases (MMPs)-1, -3, and -13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra-articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    UNASSIGNED: We utilized the destabilization of medial meniscus (DMM)-induced mice to illustrate the osteoarthritis (OA) suppressing and pain-relieving effects of a novel prolonged-release intra-articular (IA)-dexamethasone-loaded thermo-sensitive hydrogel (DLTH).
    UNASSIGNED: The effects of temperature and pH on DLTH formation and in vitro DLTH release profile were assessed. C57BL/6J mice were randomly divided into three groups: Ctrl group, Model group and DLTH group. The DLTH group received joint injections of 10 µL DLTH (1 mg/kg) into the right knee once a week from week 2 to week 11. We performed micro-computed tomography (Micro-CT) and histological analyses of safranin O-fast green, hematoxylin and eosin, and tartrate-resistant acid phosphatase in knee joints. We also carried out immunohistochemical (IHC) staining for matrix metalloproteinase-9 (MMP-9), MMP-13, and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) in cartilage and Ki-67 in synovia. Pain behavioral testing was carried out in all mice. The serum content of prostaglandin E2 (PGE2) and real-time polymerase chain reaction (PCR) of inflammatory cytokines and pain-related factors in dorsal root ganglia (DRGs) were evaluated.
    UNASSIGNED: It took 20 minutes to form DLTH at pH 7.0 and 37 °C. The cumulative release profiles of dexamethasone (Dex) from DLTH at 37 °C revealed a rapid release in the first 24 h and a sustained slow release for 7 days. In vivo study illustrated that DLTH attenuated OA bone destruction and ameliorated synovitis and progression of OA in DMM-induced mice. The chondroprotective effects of DLTH were mediated by decreased expressions of MMP-9, MMP-13, and ADAMTS-5. The results showed that IA-DLTH exerted pain-relieving effects in OA mice. Upregulation of nociceptive response time (NRT) and downregulations of serum PGE2, inflammatory factors, and pain-related mediators in DRGs of mice in the DLTH group were recorded.
    UNASSIGNED: Data presented in this study elucidated that DLTH exhibited a long and lasting Dex release and it is a potential sustainable drug delivery system (DDS) to treat OA locally. IA-DLTH injection exerted chondroprotective and pain-relieving effects in DMM-induced arthritis. The involvement of MMP-9, MMP-13, ADAMTS-5, and inflammatory and pain-related factors, may account for the suppression of OA progression and pain.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Osteoarthritis (OA) is a common disease characterized by cartilage degeneration. In recent years much attention has been paid to Traditional Chinese Medicine (TCM) since its treatments have shown efficacy for ameliorating cartilage degradation with mild side effects. Osteoking is a TCM prescription that has long been used in OA treatment. However, the exact mechanism of Osteoking are not fully elucidated. In the current study, destabilization of the medial meniscus (DMM)-induced OA mice was introduced as a wild type animal model. After 8 weeks of administration of Osteoking, histomorphometry, OARSI scoring, gait analysis, micro-CT, and immunohistochemical staining for Col2, MMP-13, TGFβRII and pSmad-2 were conducted to evaluate the chondroprotective effects of Osteoking in vivo. Further in vitro experiments were then performed to detect the effect of Osteoking on chondrocytes. TGFβRIICol2ER transgenic mice were constructed and introduced in the current study to validate whether Osteoking exerts its anti-OA effects via the TGF-β signaling pathway. Results demonstrated that in wild type DMM mice, Osteoking ameliorated OA-phenotype including cartilage degradation, subchondral bone sclerosis, and gait abnormality. Col2, TGFβRII, and pSmad-2 expressions were also found to be up-regulated after Osteoking treatment, while MMP-13 was down-regulated. In vitro, the mRNA expression of MMP-13 and ADAMTS5 decreased and the mRNA expression of Aggrecan, COL2, and TGFβRII were up-regulated after the treatment of Osteoking in IL-1β treated chondrocytes. The additional treatment of SB505124 counteracted the positive impact of Osteoking on primary chondrocytes. In TGFβRIICol2ER mice, spontaneous OA-liked phenotype was observed and treatment of Osteoking failed to reverse the OA spontaneous progression. In conclusion, Osteoking ameliorates OA progression by decelerating cartilage degradation and alleviating subchondral bone sclerosis partly via the TGF-β signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号