Extracellular matrix degradation

细胞外基质降解
  • 文章类型: Journal Article
    骨性关节炎(OA)是一种因软骨结构和功能恶化而引起的慢性疾病。涉及软骨细胞外基质的进行性降解。组织蛋白酶,溶酶体半胱氨酸蛋白酶,在各种生物和病理过程中发挥关键作用,特别是在蛋白质降解中。据报道,过量的组织蛋白酶水平有助于OA的发展。然而,组织蛋白酶家族与膝髋OA之间的因果关系尚不确定.因此,本研究利用双向孟德尔随机化(MR)分析来探索这种因果关系.我们的结果表明,血清组织蛋白酶O水平升高会增加膝关节OA的总体风险,而血清组织蛋白酶H水平升高会增加髋关节OA的风险。相反,反向MR分析未显示两者之间存在反向因果关系.总之,不同解剖位置的OA可能是由不同血清组织蛋白酶同工型的病理升高引起的,可在临床实践中用作诊断和治疗靶标。
    Osteoarthritis (OA) is a chronic disease due to the deterioration of cartilage structure and function, involving the progressive degradation of the cartilage extracellular matrix. Cathepsins, lysosomal cysteine proteases, play pivotal roles in various biological and pathological processes, particularly in protein degradation. Excess cathepsins levels are reported to contribute to the development of OA. However, the causal relationship between the cathepsin family and knee and hip OA remains uncertain. Therefore, this study utilized bidirectional Mendelian Randomization (MR) analyses to explore this causal association. Our results indicated that elevated serum levels of cathepsin O increase the overall risk of knee OA, while increased serum levels of cathepsin H enhance the risk of hip OA. Conversely, the reverse MR analyses did not reveal a reverse causal relationship between them. In summary, OA in different anatomical locations may genetically result from pathological elevations in different serum cathepsin isoforms, which could be utilized as diagnostic and therapeutic targets in clinical practice.
    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
    胸主动脉夹层(TAD)是一种危及生命的血管疾病,表现为胸主动脉中层的壁内出血。TAD的关键组织病理学特征是内侧变性,以血管平滑肌细胞(VSMC)耗竭和细胞外基质(ECM)降解为特征。MicroRNA,作为重要的表观遗传调节因子,可以在不改变序列的情况下抑制靶基因的蛋白质表达。本研究旨在阐明miR-20a的作用和潜在机制。miR-17-92簇的成员,在TAD发病过程中调节ECM降解。与从正常胸主动脉分离的收缩性VSMC相比,miR-17-92簇的表达在源自TAD病变的合成VSMC中显著增加。值得注意的是,VSMC中miR-20a的表达在血清暴露和各种刺激下增加.在TAD病变中,miR-20a的表达与弹性蛋白的表达呈显著负相关。在富马酸β-氨基丙腈和血管紧张素II诱导的TAD小鼠的胸主动脉中也观察到miR-20a的表达升高。通过模拟转染过表达miR-20a增强了VSMC的生长和侵袭能力,对它们的迁移活性或表型标记(α-SMA,SM22和OPN)。用抑制剂转染沉默miR-20a减轻了PDGF-bb刺激的VSMC中MMP2的过度激活,如活性MMP2水平降低和pro-MMP2水平升高所证明。随后,TIMP2被鉴定为miR-20a的新靶基因。miR-20a在促进MMP2激活中的作用是通过抑制VSMC中的TIMP2表达来介导的。此外,在VSMC中发现miR-20a的表达升高直接由Nanog驱动.总的来说,这些发现表明miR-20a在TAD发病过程中维持胸主动脉壁的稳态中起着至关重要的作用,并且可能是TAD的潜在治疗靶点.
    Thoracic aortic dissection (TAD) is a life-threatening vascular disease manifested as intramural bleeding in the medial layers of the thoracic aorta. The key histopathologic feature of TAD is medial degeneration, characterized by depletion of vascular smooth muscle cells (VSMCs) and degradation of extracellular matrix (ECM). MicroRNA, as essential epigenetic regulators, can inhibit the protein expression of target genes without modifying the sequences. This study aimed to elucidate the role and underlying mechanism of miR-20a, a member of the miR-17-92 cluster, in regulating ECM degradation during the pathogenesis of TAD. The expression of the miR-17-92 cluster was significantly increased in synthetic VSMCs derived from TAD lesions compared to contractile VSMCs isolated from normal thoracic aortas. Notably, the expression of miR-20a was increased in VSMCs in response to serum exposure and various stimuli. In TAD lesions, the expression of miR-20a was significantly negatively correlated with that of elastin. Elevated expression of miR-20a was also observed in thoracic aortas of TAD mice induced by β-aminopropionitrile fumarate and angiotensin II. Overexpression of miR-20a via mimic transfection enhanced the growth and invasive capabilities of VSMCs, with no significant impact on their migratory activity or the expression of phenotypic markers (α-SMA, SM22, and OPN). Silencing of miR-20a with inhibitor transfection mitigated the hyperactivation of MMP2 in VSMCs stimulated by PDGF-bb, as evidenced by reduced levels of active-MMP2 and increased levels of pro-MMP2. Subsequently, TIMP2 was identified as a novel target gene of miR-20a. The role of miR-20a in promoting the activation of MMP2 was mediated by the suppression of TIMP2 expression in VSMCs. In addition, the elevated expression of miR-20a was found to be directly driven by Nanog in VSMCs. Collectively, these findings indicate that miR-20a plays a crucial role in maintaining the homeostasis of the thoracic aortic wall during TAD pathogenesis and may represent a potential therapeutic target for TAD.
    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)是一种常见的慢性疾病,发病率和患病率均与年龄相关。细胞周期蛋白依赖性激酶5(CDK5),是CDK家族的一员,与许多慢性疾病有关。本研究旨在探讨CDK5在OA中的作用及详细的分子机制。逆转录-定量PCR(RT-qPCR)和Westernblot检测转染前后CDK5和ELF3的表达。5-乙炔基-2'-脱氧尿苷(Edu)和末端脱氧核苷酸转移酶介导的缺口末端标记(TUNEL)测定法用于检测C28/I2细胞的增殖和凋亡。使用酶联免疫吸附测定(ELISA)评估炎性细胞因子的水平,而使用westernblot检测与细胞外基质(ECM)降解和凋亡有关的蛋白质的表达。此外,使用荧光素酶活性测定和染色质免疫沉淀(CHIP)测定检测CDK5启动子的活性及其与ELF3的结合。在本研究中,发现在IL-1β诱导的C28/I2细胞中CDK5的mRNA和蛋白表达明显增加。在消耗CDK5表达后,细胞凋亡,IL-1β诱导的C28/I2细胞中的炎症和ECM被抑制。还发现ELF3表达在IL-1β诱导的C28/I2细胞中增加,并充当与CDK5启动子结合的转录因子以调节其转录表达。进一步的实验证明,ELF3过表达部分逆转了CDK5缺乏对IL-1β诱导的细胞凋亡的抑制作用,C28/I2细胞中的炎症和ECM。总的来说,通过ELF3转录上调的CDK5可以促进OA的发展。
    Osteoarthritis (OA) is a common chronic disease with age-associated increase in both incidence and prevalence. The cyclin-dependent kinase 5 (CDK5), which is a member of the CDK family, is involved in many chronic diseases. This study was performed to explore the functional role of CDK5 in OA and to discuss the detailed molecular mechanisms. The expressions of CDK5 and ELF3 before or after transfection were detected with reverse transcription-quantitative PCR (RT-qPCR) and western blot. 5-ethynyl-2\'-deoxyuridine (Edu) and terminal deoxynucleoitidyl transferase-mediated nick-end labeling (TUNEL) assays were used to detect the proliferation and apoptosis of C28/I2 cells. The levels of inflammatory cytokines were estimated using enzyme-linked immunosorbent assay (ELISA) while the expressions of proteins implicated in extracellular matrix (ECM) degradation- and apoptosis were detected using western blot. Additionally, the activity of CDK5 promoters and its binding with ELF3 were detected using luciferase activity assay and chromatin immunoprecipitation (CHIP) assay. In the present study, it was discovered that the mRNA and protein expressions of CDK5 were significantly increased in IL-1β-induced C28/I2 cells. After depleting CDK5 expression, the apoptosis, inflammation and ECM in C28/I2 cells with IL-1β induction were suppressed. It was also found that ELF3 expression was increased in IL-1β-induced C28/I2 cells and acted as a transcription factor binding to the CDK5 promoter to regulate its transcriptional expression. The further experiments evidenced that ELF3 overexpression partially reversed the inhibitory effects of CDK5 deficiency on IL-1β-induced apoptosis, inflammation and ECM in C28/I2 cells. Collectively, CDK5 that upregulated by ELF3 transcription could promote the development of OA.
    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
    阿尔茨海默病是进行性神经退行性疾病,其特征是存在β淀粉样蛋白斑块的细胞外积累和Tau神经原纤维缠结的细胞内沉积。除轴突沉积外,已知病理性聚集的Tau蛋白分泌到细胞外空间并通过播种机制传播。小胶质细胞,大脑的免疫细胞表现出适度的内化细胞外Tau并通过内溶酶体途径降解它的能力。然而,病理蛋白的过度负担削弱了小胶质细胞的吞噬能力。ω-3脂肪酸(n-3)的细胞外补充可以调节小胶质细胞的吞噬作用,因为它们通过膜脂质组成变化介导小胶质细胞的抗炎极化。小胶质细胞中细胞外Tau的内化受皮质膜相关肌动蛋白重塑的调节,该重塑由肌动蛋白结合蛋白的相互作用驱动。另一方面,由于结构中富含脯氨酸的结构域的存在,Tau展示能力与各种肌动蛋白结合蛋白结合并相互作用,并调节其激活。在这项研究中,我们假设在omega-3脂肪酸存在下Tau的内化将传播Tau介导的肌动蛋白结合蛋白和细胞外基质的激活,进而调节皮质肌动蛋白重塑以进行吞噬作用.
    Alzheimer\'s disease is progressive neurodegenerative disease characterize by the presence of extracellular accumulation of amyloid-β plaques and intracellular deposits of neurofibrillary tangles of Tau. Apart from axonal depositions pathological aggregated Tau protein is known to secrete into extracellular spaces and propagate through seeding mechanism. Microglia, the immune cells of the brain display modest ability to internalize the extracellular Tau and degrade it through endolysosomal pathway. However, the excessive burden of pathoproteins weakens the phagocytic ability of microglia. Extracellular supplementation of omega-3 fatty acids (n-3) may regulate the phagocytosis of microglia as they mediate the anti-inflammatory polarization of microglia through membrane lipid compositions changes. The internalization of extracellular Tau in the microglia is regulated by cortical membrane-associated actin remodeling driven by interplay of actin-binding proteins. On the other hand, Tau display capability bind and interact with various actin-binding protein owing to the presence of proline-rich domain in the structure and regulate their activation. In this study, we hypothesize that internalization of Tau in the presence of omega-3 fatty acids would propagate the Tau-mediated activation of actin-binding proteins as well as extracellular matrix and in turn modulate cortical actin remodeling for phagocytosis.
    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
    目的:椎间盘退变(IVDD)是下背痛(LBP)的主要原因。β-抑制蛋白1(ARRB1)是一种多功能蛋白,可调节多种病理过程。本研究旨在探讨ARRB1在IVDD中的作用。
    方法:测定IVDD大鼠髓核中ARRB1的表达。接下来,用含有shArrb1(LV-shArrb1)和过表达Arrb1(LV-oeArrb1)的慢病毒感染大鼠髓核细胞(NPC)。通过测量细胞凋亡来研究Arrb1在血清剥夺型NPCs中的作用,细胞外基质降解,和自噬通量。对于体内实验,将LV-oeArrb1慢病毒注射到IVDD大鼠的NP组织中,以评估Arrb1过表达对NP的影响。
    结果:在IVDD大鼠的NP组织中,ARRB1和caspase-3表达增加,LC3II/LC3I蛋白表达比例上调。Arrb1敲低加重细胞外基质降解,细胞凋亡,以及血清剥夺条件下大鼠NPCs自噬通量的损害,而Arrb1过表达显著逆转了这些效应。ARRB1与Beclin1相互作用,而Arrb1敲低抑制了Beclin1-PIK3C3核心复合物的形成。自噬抑制剂3-甲基腺嘌呤(3-MA)抵消了血清剥夺型NPC中Arrb1过表达的保护作用。此外,Arrb1过表达抑制细胞凋亡和细胞外基质降解,促进NP中的自噬,并延缓了大鼠IVDD的发展。
    结论:ARRB1通过上调自噬和改善IVDD进展,防止NPCs细胞外基质降解和凋亡,提出了治疗IVDD的创新策略。
    OBJECTIVE: Intervertebral disc degeneration (IVDD) is the leading cause of lower back pain (LBP). β-arrestin 1 (ARRB1) is a multifunctional protein that regulates numerous pathological processes. The aim of this study was to investigate the role of ARRB1 in IVDD.
    METHODS: The expression of ARRB1 in nucleus pulposus (NP) of rats with IVDD was assayed. Next, rat nucleus pulposus cells (NPCs) were infected with lentiviruses containing shArrb1 (LV-shArrb1) and overexpressing Arrb1 (LV-oeArrb1). The roles of Arrb1 in serum-deprived NPCs were investigated by measuring apoptosis, extracellular matrix degradation, and autophagic flux. For experiments in vivo, LV-oeArrb1 lentivirus was injected into the NP tissues of IVDD rats to evaluate the effects of Arrb1 overexpression on NP.
    RESULTS: In the NP tissues of IVDD rats, ARRB1 and cleaved caspase-3 expression increased, and the ratio of LC3II/LC3I protein expression was upregulated. Arrb1 knockdown aggravated extracellular matrix degradation, cellular apoptosis, and impairment of autophagic flux in rat NPCs under serum-deprived conditions, whereas Arrb1 overexpression significantly reversed these effects. ARRB1 interacted with Beclin 1, and Arrb1 knockdown suppressed the formation of the Beclin1-PIK3C3 core complex. The autophagy inhibitor 3-methyladenine (3-MA) offset the protective effects of Arrb1 overexpression in serum-deprived NPCs. Furthermore, Arrb1 overexpression inhibited apoptosis and extracellular matrix degradation, promoted autophagy in NP, and delayed the development of IVDD in rats.
    CONCLUSIONS: ARRB1 prevents extracellular matrix degradation and apoptosis of NPCs by upregulating autophagy and ameliorating IVDD progression, presenting an innovative strategy for the treatment of IVDD.
    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
    维生素K2(VK2)是一种有效的抗铁细胞凋亡和抗骨质疏松的化合物,Semensojaepraeparatum(中文为Dandouchi)是VK2的主要来源。软骨细胞铁性凋亡和细胞外基质(ECM)降解在骨关节炎(OA)的发病机理中起作用。谷胱甘肽过氧化物酶4(GPX4)是调节OA进展的两种机制的交叉点。但尚无研究阐明VK2对OA的治疗作用和机制。本研究利用通过前交叉韧带横切(ACLT)创建的体内大鼠OA模型和TBHP诱导的体外软骨细胞氧化损伤模型来研究VK2在OA中的保护作用和作用机制。使用VonFrey测试评价小鼠的膝关节疼痛。采用Micro-CT和SafraninO-FastGreen染色观察胫骨软骨和软骨下骨的损伤程度,而免疫组织化学和PCR用于检测关节软骨中的GPX4水平。使用CCK-8和流式细胞术测定评估VK2对大鼠软骨细胞活力的影响。甲苯胺蓝和阿辛蓝染色观察软骨细胞形态。使用荧光染色和流式细胞术观察VK2对细胞内铁凋亡相关标志物的影响。通过免疫荧光和Westernblot分析检测蛋白表达变化。此外,特异性蛋白质抑制剂用于证实VK2对GPX4的双重调节作用。VK2可以增加胫骨软骨下骨的骨量和软骨厚度,减轻OA引起的疼痛和OARSI评分。免疫组织化学结果表明,VK2通过调节GPX4延迟ECM降解来发挥其抗OA作用。VK2可以抑制细胞内GPX4表达降低引起的MAPK/NFκB信号通路的激活,从而减少ECM降解。此外,VK2可以逆转RSL3对GPX4的抑制作用,增加细胞内GSH含量和GSH/GSSG比值,降低MDA含量,和拯救软骨细胞的铁性凋亡。VK2的保护机制可能涉及其对GPX4的双靶点调控,减少软骨细胞的铁凋亡,抑制MAPK/NFκB信号通路,减缓软骨细胞细胞外基质的降解。
    Vitamin K2 (VK2) is an effective compound for anti-ferroptosis and anti-osteoporosis, and Semen sojae praeparatum (Dandouchi in Chinese) is the main source of VK2. Chondrocyte ferroptosis and extracellular matrix (ECM) degradation playing a role in the pathogenesis of osteoarthritis (OA). Glutathione peroxidase 4 (GPX4) is the intersection of two mechanisms in regulating OA progression. But no studies have elucidated the therapeutic effects and mechanisms of VK2 on OA. This study utilized an in vivo rat OA model created via anterior cruciate ligament transection (ACLT) and an in vitro chondrocyte oxidative damage model induced by TBHP to investigate the protective effects and mechanisms of action of VK2 in OA. Knee joint pain in mice was evaluated using the Von Frey test. Micro-CT and Safranin O-Fast Green staining were employed to observe the extent of damage to the tibial cartilage and subchondral bone, while immunohistochemistry and PCR were used to examine GPX4 levels in joint cartilage. The effects of VK2 on rat chondrocyte viability were assessed using CCK-8 and flow cytometry assays, and chondrocyte morphology was observed with toluidine blue and alcian blue staining. The impact of VK2 on intracellular ferroptosis-related markers was observed using fluorescent staining and flow cytometry. Protein expression changes were detected by immunofluorescence and Western blot analysis. Furthermore, specific protein inhibitors were applied to confirm the dual-regulatory effects of VK2 on GPX4. VK2 can increase bone mass and cartilage thickness in the subchondral bone of the tibia, and reduce pain and the OARSI score induced by OA. Immunohistochemistry results indicate that VK2 exerts its anti-OA effects by regulating GPX4 to delay ECM degradation. VK2 can inhibit the activation of the MAPK/NFκB signaling pathway caused by reduced expression of intracellular GPX4, thereby decreasing ECM degradation. Additionally, VK2 can reverse the inhibitory effect of RSL3 on GPX4, increase intracellular GSH content and the GSH/GSSG ratio, reduce MDA content, and rescue chondrocyte ferroptosis. The protective mechanism of VK2 may involve its dual-target regulation of GPX4, reducing chondrocyte ferroptosis and inhibiting the MAPK/NFκB signaling pathway to decelerate the degradation of the chondrocyte extracellular matrix.
    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
    Notch信号调节软骨形成和体内平衡。大骨节病(KBD),地方性骨软骨病,以严重的软骨退化为特征。KBD的病因与HT-2毒素的暴露有关,一种霉菌毒素和T-2毒素的主要代谢产物。本研究旨在探讨HT-2毒素在hiPSCs-软骨细胞Notch信号调节和细胞外基质(ECM)代谢中的作用。采用免疫组织化学和qRT-PCR检测Notch通路分子在KBD关节软骨和原代软骨细胞中的表达。hiPSCs-软骨细胞,来自hiPSCs,用100ng/mLHT-2毒素和γ-分泌酶抑制剂(DAPT)处理48小时,分别。使用qRT-PCR和Western印迹评估与Notch信号通路和ECM相关的标志物。Notch通路失调在KBD软骨中突出。HT-2毒素暴露导致hiPSCs-软骨细胞的细胞毒性,并通过增加NOTCH1和HES1的mRNA和蛋白质水平来激活Notch信号。HT-2毒素还上调ECM分解代谢酶和下调ECM成分(COL2A1和ACAN),指示ECM降解。DAPT介导的Notch信号传导抑制抑制ADAMTS5表达的mRNA和蛋白质水平,同时增强hiPSCs-软骨细胞中的ECM组分表达。这项研究表明,HT-2毒素可能通过激活Notch信号在hiPSC-软骨细胞中诱导ECM降解。
    Notch signaling regulates cartilage formation and homeostasis. Kashin-Beck Disease (KBD), an endemic osteochondropathy, is characterized by severe cartilage degradation. The etiology of KBD is related to the exposure of HT-2 toxin, a mycotoxin and primary metabolite of T-2 toxin. This study aims to explore the role of HT-2 toxin in the Notch signaling regulation and extracellular matrix (ECM) metabolism of hiPSCs-Chondrocytes. Immunohistochemistry and qRT-PCR were employed to investigate the expression of Notch pathway molecules in KBD articular cartilage and primary chondrocytes. hiPSCs-Chondrocytes, derived from hiPSCs, were treated with 100 ng/mL HT-2 toxin and the γ-secretase inhibitor (DAPT) for 48h, respectively. The markers related to the Notch signaling pathway and ECM were assessed using qRT-PCR and Western blot. Notch pathway dysregulation was prominent in KBD cartilage. HT-2 toxin exposure caused cytotoxicity in hiPSCs-Chondrocytes, and activated Notch signaling by increasing the mRNA and protein levels of NOTCH1 and HES1. HT-2 toxin also upregulated ECM catabolic enzymes and downregulated ECM components (COL2A1 and ACAN), indicating ECM degradation. DAPT-mediated Notch signaling inhibition suppressed the mRNA and protein level of ADAMTS5 expression while enhancing ECM component expression in hiPSCs-Chondrocytes. This study suggests that HT-2 toxin may induce ECM degradation in hiPSCs-Chondrocytes through activating Notch signaling.
    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
    骨关节炎(OA)是一种慢性退行性疾病,患病率很高,会导致软骨损伤并导致残疾。导致OA发生和发展的主要因素包括炎症和细胞外基质的变性。Cathelicidin-BF(BF-30),一种来自Bungarusfasciatus毒液的天然肽,已显示出多种重要的药理作用。然而,BF-30在OA治疗中的作用机制尚待阐明。在这项研究中,X射线和SafraninO染色用于评估体内小鼠膝关节的影像学和组织形态学差异。蛋白质印迹分析等技术,RT-qPCR,ELISA,和免疫荧光染色用于检测体外实验中基因和蛋白质水平的变化。结果发现,BF-30显著降低炎症和增强细胞外基质代谢。第一次,结果表明,BF-30的积极作用是通过激活AMPK/SIRT1/NF-κB途径介导的。此外,当BF-30与化合物C共同给药时,AMPK抑制剂,BF-30的治疗益处在体内和体外均被逆转.总之,研究结果表明,BF-30可能是改善OA的新型治疗剂.
    Osteoarthritis (OA) is a chronic degenerative disease with a significant prevalence that causes cartilage damage and can lead to disability. The main factors contributing to the onset and progression of OA include inflammation and degeneration of the extracellular matrix. Cathelicidin-BF (BF-30), a natural peptide derived from Bungarus fasciatus venom, has shown multiple important pharmacological effects. However, the action mechanism of BF-30 in OA treatment remains to be elucidated. In this research, X-ray and Safranin O staining were employed to evaluate the imageology and histomorphology differences in the knee joints of mice in vivo. Techniques such as Western blot analysis, RT-qPCR, ELISA, and immunofluorescence staining were applied to examine gene and protein level changes in in vitro experiments. It was found that BF-30 significantly decreased inflammation and enhanced extracellular matrix metabolism. For the first time, it was demonstrated that the positive effects of BF-30 are mediated through the activation of the AMPK/SIRT1/NF-κB pathway. Moreover, when BF-30 was co-administered with Compound C, an AMPK inhibitor, the therapeutic benefits of BF-30 were reversed in both in vivo and in vitro settings. In conclusion, the findings suggest that BF-30 could be a novel therapeutic agent for OA improvement.
    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
    骨关节炎(OA)是一种与年龄相关的关节疾病,其特征是关节软骨和软骨下骨的进行性异质性变化。破骨细胞刺激因子1(OSTF1)是一种参与骨形成和骨吸收的小细胞内蛋白。然而,据我们所知,其在OA中的作用尚不清楚。在这项研究中,采用前交叉韧带横断术(ALCT)建立OA大鼠模型。OSTF1在OA患者和OA年夜鼠的软骨组织中增高。接下来,OSTF1在白细胞介素-1β(IL-1β)诱导的软骨细胞凋亡中的作用,炎症和细胞外基质降解通过功能丧失测定进行探索。引人注目的是,OSTF1敲低减轻IL-1β诱导的软骨细胞凋亡,切割的caspase-3和切割的PARP水平降低。此外,OSTF1敲低抑制IL-1β诱导的炎症反应和软骨细胞细胞外基质的降解。随后,探讨了OSTF1的分子机制。转录组分析揭示了OSTF1敲低调控的潜在基因网络图。一些差异表达基因(DEGs)参与调控NF-κB信号通路。此外,我们的结果表明,OSTF1敲低抑制IL-1β激活NF-κB信号通路。最终,我们分析了OSTF1及其下游NF-κB调控的潜在基因网络图。生物信息学分析表明,OSTF1沉默的软骨细胞中的18个DEGs与NF-κB下游靶标重叠。总的来说,我们的发现表明OSTF1敲低通过抑制NF-κB信号通路减轻IL-1β诱导的软骨细胞损伤。
    Osteoarthritis (OA) is an age-related joint disease characterized by progressive heterogeneous changes in articular cartilage and subchondral bone. Osteoclast stimulating factor 1 (OSTF1) is a small intracellular protein involved in bone formation and bone resorption. However, to our best knowledge, its role in OA is still unclear. In this study, an OA rat model was established by anterior cruciate ligament transection (ALCT). OSTF1 was increased in the cartilage tissues of OA patients and OA rats. Next, the role of OSTF1 in interleukin-1β (IL-1β)-induced chondrocyte apoptosis, inflammation and extracellular matrix degradation was explored through loss of function assays. Strikingly, OSTF1 knockdown relieved IL-1β-induced chondrocyte apoptosis, with decreased cleaved caspase-3 and cleaved PARP levels. Besides, OSTF1 knockdown restrained IL-1β-induced inflammation and degradation of extracellular matrix of chondrocytes. Subsequently, the molecular mechanism of OSTF1 was explored. Transcriptomic analysis revealed the potential gene network map regulated by OSTF1 knockdown. Some differentially expressed genes (DEGs) were involved in regulating the NF-κB signaling pathway. Furthermore, our results demonstrated that OSTF1 knockdown inhibited IL-1β-activated the NF-κB signaling pathway. Ultimately, we analyzed the potential gene network map regulated by OSTF1 and its downstream NF-κB. Bioinformatics analysis showed that 18 DEGs in OSTF1-silenced chondrocytes overlapped with the NF-κB downstream targets. Collectively, our findings indicate that OSTF1 knockdown mitigates IL-1β-induced chondrocyte injury via inhibiting the NF-κB signaling pathway.
    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
    背景:骨关节炎(OA),最常见的关节疾病,与软骨细胞凋亡和细胞外基质(ECM)降解有关。带电多囊体蛋白5(CHMP5),多囊身体的成员,据报道,作为抗凋亡蛋白参与白血病的发展。然而,CHMP5对OA细胞凋亡和ECM降解的影响尚不清楚。
    方法:在本研究中,进行了定量蛋白质组学分析正常和OA患者关节软骨之间的差异蛋白。通过内侧半月板(DMM)的去稳定来构建OA小鼠模型。体外,白细胞介素-1β(IL-1β)用于在人软骨细胞中诱导OA。使用腺病毒系统产生CHMP5过表达和沉默载体。CHMP5对IL-1β诱导的软骨细胞凋亡的影响通过CCK-8、流式细胞术、和westernblot.通过蛋白质印迹和免疫荧光检查对ECM降解的影响。通过蛋白质印迹和Co-IP测定探索了潜在的机制。
    结果:通过蛋白质组学在OA患者软骨中鉴定出下调的CHMP5,这在人类和小鼠的关节软骨中得到了证实。CHMP5过表达抑制OA软骨细胞中的细胞凋亡和ECM降解。然而,沉默CHMP5会加剧OA软骨细胞凋亡和ECM降解。此外,我们发现CHMP5对OA的保护作用涉及核因子κB(NF-κB)信号通路。
    结论:本研究表明,CHMP5抑制IL-1β诱导的软骨细胞凋亡和ECM降解,阻断NF-κB活化。研究表明,CHMP5可能是未来OA新的潜在治疗靶点。
    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-1β) 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-1β-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-1β-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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号