Osteoclastogenesis

破骨细胞生成
  • 文章类型: Journal Article
    背景:正畸牙齿移动(OTM)是骨骼重塑的动态平衡,涉及新骨的成骨作用和旧骨的破骨细胞作用,这是由机械力介导的。牙周膜(PDL)空间中的牙周膜干细胞(PDLCSs)可以在OTM过程中传递机械信号并调节破骨细胞生成。KAT6A是在干细胞分化中起作用的组蛋白乙酰转移酶。然而,KAT6A是否参与PDLSCs对破骨细胞生成的调节尚不清楚.
    结果:在这项研究中,我们使用力诱导的OTM模型,观察到在OTM期间,KAT6A在PDL的压缩侧增加,并且在体外压缩力下PDLSCs中也增加。KAT6A抑制剂WM1119对KAT6A的抑制作用,OTM的距离明显降低。PDLSCs中KAT6A的敲除降低了RANKL/OPG比率和THP-1的破骨细胞生成。机械上,KAT6A通过结合和乙酰化YAP促进破骨细胞生成,同时调节YAP/TEAD轴并增加PDLSCs中的RANKL/OPG比率。TED-347,一种YAP-TEAD4相互作用抑制剂,部分减弱了机械力引起的RANKL/OPG比率的升高。
    结论:我们的研究表明,在机械力作用下,PDLSCs通过KAT6A/YAP/TEAD4通路调节破骨细胞生成并增加RANKL/OPG比值。KAT6A可能是加速OTM的新目标。
    BACKGROUND: Orthodontic tooth movement (OTM) is a dynamic equilibrium of bone remodeling, involving the osteogenesis of new bone and the osteoclastogenesis of old bone, which is mediated by mechanical force. Periodontal ligament stem cells (PDLCSs) in the periodontal ligament (PDL) space can transmit mechanical signals and regulate osteoclastogenesis during OTM. KAT6A is a histone acetyltransferase that plays a part in the differentiation of stem cells. However, whether KAT6A is involved in the regulation of osteoclastogenesis by PDLSCs remains unclear.
    RESULTS: In this study, we used the force-induced OTM model and observed that KAT6A was increased on the compression side of PDL during OTM, and also increased in PDLSCs under compression force in vitro. Repression of KAT6A by WM1119, a KAT6A inhibitor, markedly decreased the distance of OTM. Knockdown of KAT6A in PDLSCs decreased the RANKL/OPG ratio and osteoclastogenesis of THP-1. Mechanistically, KAT6A promoted osteoclastogenesis by binding and acetylating YAP, simultaneously regulating the YAP/TEAD axis and increasing the RANKL/OPG ratio in PDLSCs. TED-347, a YAP-TEAD4 interaction inhibitor, partly attenuated the elevation of the RANKL/OPG ratio induced by mechanical force.
    CONCLUSIONS: Our study showed that the PDLSCs modulated osteoclastogenesis and increased the RANKL/OPG ratio under mechanical force through the KAT6A/YAP/TEAD4 pathway. KAT6A might be a novel target to accelerate OTM.
    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
    代谢的表观遗传调节深刻影响细胞命运的承诺。在破骨细胞分化过程中,RANK信号的激活伴随着代谢重编程,但是RANK信号诱导这种重编程的表观遗传机制仍然难以捉摸。通过转录序列和ATAC分析,这项研究确定RANK信号的激活通过表观遗传修饰上调PRMT6,触发从脂肪酸氧化向糖酵解的代谢转换。相反,Prmt6缺陷逆转了这种转变,显著减少HIF-1α介导的糖酵解并增强脂肪酸氧化。因此,在卵巢切除(OVX)小鼠中,PRMT6缺乏或抑制剂阻碍破骨细胞分化并减轻骨丢失。在分子水平上,Prmt6缺陷减少了H3R2在包括Ppard在内的基因启动子的不对称二甲基化,Acox3和Cpt1a,增强脂肪酸氧化的基因组可及性。因此,PRMT6成为代谢检查点,介导从脂肪酸氧化到糖酵解的代谢转换,从而支持破骨细胞生成。揭示PRMT6在破骨细胞生成中表观遗传协调代谢变化中的关键作用,为抗再吸收治疗提供了一个有希望的目标。
    Epigenetic regulation of metabolism profoundly influences cell fate commitment. During osteoclast differentiation, the activation of RANK signaling is accompanied by metabolic reprogramming, but the epigenetic mechanisms by which RANK signaling induces this reprogramming remain elusive. By transcriptional sequence and ATAC analysis, this study identifies that activation of RANK signaling upregulates PRMT6 by epigenetic modification, triggering a metabolic switching from fatty acids oxidation toward glycolysis. Conversely, Prmt6 deficiency reverses this shift, markedly reducing HIF-1α-mediated glycolysis and enhancing fatty acid oxidation. Consequently, PRMT6 deficiency or inhibitor impedes osteoclast differentiation and alleviates bone loss in ovariectomized (OVX) mice. At the molecular level, Prmt6 deficiency reduces asymmetric dimethylation of H3R2 at the promoters of genes including Ppard, Acox3, and Cpt1a, enhancing genomic accessibility for fatty acid oxidation. PRMT6 thus emerges as a metabolic checkpoint, mediating metabolic switch from fatty acid oxidation to glycolysis, thereby supporting osteoclastogenesis. Unveiling PRMT6\'s critical role in epigenetically orchestrating metabolic shifts in osteoclastogenesis offers a promising target for anti-resorptive therapy.
    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
    复子汤(FZD)作为经典制剂在治疗类风湿性关节炎(RA)方面具有悠久的应用历史。然而,其潜在机制尚未完全阐明。本研究旨在破译FZD治疗RA的潜在机制,核因子κB受体活化因子/核因子κB受体活化因子配体(RANK/RANKL)信号通路具有特异性。
    在胶原诱导的关节炎大鼠(CIA)中研究了FZD对RA的影响,并在破骨细胞分化细胞模型中研究了其潜在机制。在体内,通过关节炎指数评分评估FZD在各种剂量(2.3,4.6,9.2g/kg/天)的抗关节炎作用,爪子体积,脚趾厚度和关节发炎的组织病理学检查。此外,采用显微CT和番红花O固绿染色测定踝关节组织,以评估滑膜增生和关节软骨损伤。体外,通过TRAP染色在RANKL诱导的骨髓单个核细胞中评估破骨细胞的分化和成熟。通过ELISA试剂盒评估促炎和抗炎细胞因子以及RANKL和OPG的水平。此外,Western印迹用于研究FZD在体内和体外对RANK/RANKL途径激活的影响。
    FZD显著降低关节炎指数评分,爪子体积,CIA大鼠的脚趾厚度和体重减轻,减轻病理性关节改变。与体内结果一致,FZD通过以剂量依赖性方式减少破骨细胞数量而显著抑制RANKL诱导的破骨细胞分化。此外,FZD降低促炎细胞因子IL-6、IL-1β和TNF-α水平,同时增加血清和培养上清液中的抗炎细胞因子IL-10水平。FZD治疗显著降低血清RANKL水平,OPG水平升高,并降低RANKL/OPG比值。在体内和体外环境中,FZD下调RANK的蛋白表达,RANKL,c-Fos,在提高OPG水平的同时,进一步降低RANKL/OPG比值。
    总而言之,FZD通过抑制RANK/RANKL介导的破骨细胞分化在CIA大鼠中发挥治疗作用,这表明FZD是一种有前途的RA治疗方法。
    UNASSIGNED: Fu-zi decoction (FZD) has a long history of application for treating Rheumatoid arthritis (RA) as a classic formulation. However, its underlying mechanisms have not been fully elucidated. This study aimed to decipher the potential mechanism of FZD in treating RA, with a specific focus on receptor activator of nuclear factor κB/receptor activator of nuclear factor κB ligand (RANK/RANKL) signaling pathway.
    UNASSIGNED: The impact of FZD on RA was investigated in collagen-induced arthritis rats (CIA), and the underlying mechanism was investigated in an osteoclast differentiation cell model. In vivo, the antiarthritic effect of FZD at various doses (2.3, 4.6, 9.2 g/kg/day) was evaluated by arthritis index score, paw volume, toe thickness and histopathological examination of inflamed joints. Additionally, the ankle joint tissues were determined with micro-CT and safranin O fast green staining to evaluate synovial hyperplasia and articular cartilage damage. In vitro, osteoclast differentiation and maturation were evaluated by TRAP staining in RANKL-induced bone marrow mononuclear cells. The levels of pro- and anti-inflammatory cytokines as well as RANKL and OPG were evaluated by ELISA kits. In addition, Western blotting was used to investigate the effect of FZD on RANK/RANKL pathway activation both in vivo and in vitro.
    UNASSIGNED: FZD significantly diminished the arthritis index score, paw volume, toe thickness and weigh loss in CIA rats, alleviated the pathological joint alterations. Consistent with in vivo results, FZD markedly inhibited RANKL-induced osteoclast differentiation by decreasing osteoclast numbers in a dose-dependent manner. Moreover, FZD decreased the levels of pro-inflammatory cytokines IL-6, IL-1β and TNF-α, while increasing anti-inflammatory cytokine IL-10 level both in serum and culture supernatants. Treatment with FZD significantly reduced serum RANKL levels, increased OPG levels, and decreased the RANKL/OPG ratio. In both in vivo and in vitro settings, FZD downregulated the protein expressions of RANK, RANKL, and c-Fos, while elevating OPG levels, further decreasing the RANKL/OPG ratio.
    UNASSIGNED: In conclusion, FZD exerts a therapeutic effect in CIA rats by inhibiting RANK/RANKL-mediated osteoclast differentiation, which suggested that FZD is a promising treatment for RA.
    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
    暂无摘要。
    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
    骨吸收破骨细胞(OCL)是通过单核细胞前体细胞的分化和融合形成的,产生大的多核细胞。破骨细胞生成过程中紧密调节的细胞融合导致可吸收的OCL的形成,其大小落在可预测的生理范围内。调节OCL融合的发生及其随后的停滞的分子机制是,然而,很大程度上未知。我们以前已经表明,从小鼠中培养的OCLs在囊泡运输相关蛋白分选nexin10中的R51Q突变纯合,该突变在人类和小鼠中诱导常染色体隐性遗传性骨硬化症,显示失调和连续融合,产生巨大的,非活动OCL。因此,成熟OCL的融合被一个活跃的,基因编码,细胞自主,和SNX10依赖机制。为了直接检查SNX10是否在体内发挥类似的作用,我们产生了SNX10缺陷(SKO)小鼠,并证明它们表现出大量的骨硬化,并且它们的OCLs在培养中不可控制地融合,纯合R51QSNX10(RQ/RQ)小鼠也是如此。缺乏SNX10的OCL在其外周表现出DC-STAMP蛋白的持续存在,这可能有助于它们不受控制的融合。为了在其天然骨环境中可视化内源性SNX10突变OCLs,我们对野生型OCLs进行了遗传标记,使用EGFP的SKO和RQ/RQ小鼠,然后通过双光子可视化常驻OCL和细胞周围骨基质的三维组织,共焦,和二次谐波生成显微镜。我们展示了卷,表面积和,特别是,两种突变株的OCLs中的细胞核数量平均比野生型小鼠的OCLs大2-6倍,表明放松管制,在突变小鼠中发生过度融合。我们得出结论,OCL的融合,因此它们的大小,在体内受到成熟OCL融合的SNX10依赖性阻滞的调节。
    破骨细胞(OCL)是降解骨骼的细胞。这些细胞通过单核细胞前体细胞的融合产生,但是调节这一过程并最终逮捕它的机制是未知的。我们先前已经表明,从蛋白质分选nexin10(SNX10)中携带R51Q突变的小鼠培养的OCL失去了吸收能力,并且由于不受控制的融合而变得巨大。为了检查是否需要SNX10在体内进行OCL融合阻滞,我们灭活了小鼠的Snx10基因,并荧光标记了它们的OCLs和R51QSNX10小鼠的OCLs,隔离他们的股骨,并使用先进的3D显微镜方法来可视化骨基质内的OCL。不出所料,缺乏SNX10的小鼠表现出过多的骨量,表示它们的OCL处于非活动状态。两种突变小鼠品系的骨骼中的OCL平均比对照小鼠大2-6倍,并按比例包含更多的原子核。我们得出结论,OCL融合被控制,但不是SNX10突变体,老鼠,表明成熟OCL的大小在体内受到活性物质的限制,抑制细胞融合的SNX10依赖性机制。
    Bone-resorbing osteoclasts (OCLs) are formed by differentiation and fusion of monocyte precursor cells, generating large multi-nucleated cells. Tightly-regulated cell fusion during osteoclastogenesis leads to formation of resorption-competent OCLs, whose sizes fall within a predictable physiological range. The molecular mechanisms that regulate the onset of OCL fusion and its subsequent arrest are, however, largely unknown. We have previously shown that OCLs cultured from mice homozygous for the R51Q mutation in the vesicle trafficking-associated protein sorting nexin 10, a mutation that induces autosomal recessive osteopetrosis in humans and in mice, display deregulated and continuous fusion that generates gigantic, inactive OCLs. Fusion of mature OCLs is therefore arrested by an active, genetically-encoded, cell-autonomous, and SNX10-dependent mechanism. In order to directly examine whether SNX10 performs a similar role in vivo, we generated SNX10-deficient (SKO) mice and demonstrated that they display massive osteopetrosis and that their OCLs fuse uncontrollably in culture, as do homozygous R51Q SNX10 (RQ/RQ) mice. OCLs that lack SNX10 exhibit persistent presence of DC-STAMP protein at their periphery, which may contribute to their uncontrolled fusion. In order to visualize endogenous SNX10-mutant OCLs in their native bone environment we genetically labelled the OCLs of wild-type, SKO and RQ/RQ mice with EGFP, and then visualized the three-dimensional organization of resident OCLs and the pericellular bone matrix by two-photon, confocal, and second harmonics generation microscopy. We show that the volumes, surface areas and, in particular, the numbers of nuclei in the OCLs of both mutant strains were on average 2-6 fold larger than those of OCLs from wild-type mice, indicating that deregulated, excessive fusion occurs in the mutant mice. We conclude that the fusion of OCLs, and consequently their size, are regulated in vivo by SNX10-dependent arrest of fusion of mature OCLs.
    Osteoclasts (OCLs) are cells that degrade bone. These cells are generated by fusion of monocyte precursor cells, but the mechanisms that regulate this process and eventually arrest it are unknown. We had previously shown that OCLs cultured from mice carrying the R51Q mutation in the protein sorting nexin 10 (SNX10) lose their resorptive capacity and become gigantic due to uncontrolled fusion. To examine whether SNX10 is required for OCL fusion arrest also in vivo, we inactivated the Snx10 gene in mice and fluorescently labelled their OCLs and OCLs of R51Q SNX10 mice, isolated their femurs, and used advanced 3D microscopy methods to visualize OCLs within the bone matrix. As expected, mice lacking SNX10 exhibited excessive bone mass, indicating that their OCLs are inactive. OCLs within bones of both mutant mouse strains were on average 2-6-fold larger than in control mice, and contained proportionally more nuclei. We conclude that OCL fusion is arrested in control, but not SNX10 mutant, mice, indicating that the sizes of mature OCLs are limited in vivo by an active, SNX10-dependent mechanism that suppresses cell fusion.
    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:破骨细胞过度生成是炎性骨丢失的关键驱动因素。抑制破骨细胞生成一直被认为是治疗炎性骨丢失的关键。N-乙酰转移酶10(NAT10)是负责mRNA的N4-乙酰胞苷(ac4C)修饰的唯一酶,并参与细胞发育。然而,其在破骨细胞生成和炎性骨丢失中的作用仍然难以捉摸。
    目的:我们旨在阐明NAT10和ac4C修饰在破骨细胞生成和炎性骨丢失中的调控机制。
    方法:通过定量实时PCR(qPCR)测定破骨细胞形成过程中NAT10的表达和ac4C修饰,西方印迹,斑点印迹和免疫荧光染色,并通过抗酒石酸酸性磷酸酶染色检测NAT10抑制对体外破骨细胞分化的影响,足体带染色测定和骨吸收坑测定。然后,acRIP-qPCR和NAT10RIP-qPCR,AC4C站点预测,进行mRNA衰减测定和荧光素酶报告基因测定以进一步研究其潜在机制。最后,应用炎性骨丢失的小鼠模型来验证NAT10抑制在体内的治疗效果。
    结果:NAT10表达在破骨细胞分化过程中上调,在牙周炎小鼠牙槽骨破骨细胞中高表达。抑制NAT10显著降低体外破骨细胞分化,如酒石酸抗性酸性磷酸阳性多核细胞的大量减少所示,破骨细胞特异性基因表达,F-肌动蛋白环形成和骨吸收能力。机械上,NAT10催化ac4C修饰Fos(编码AP-1组分c-Fos)mRNA并维持其稳定性。此外,NAT10促进MAPK信号通路,从而激活AP-1(c-Fos/c-Jun)转录用于破骨细胞生成。治疗学上,雷德林的管理,NAT10的特异性抑制剂显着阻碍结扎诱导的牙槽骨丢失和脂多糖诱导的炎性颅骨骨溶解。
    结论:我们的研究表明,NAT10介导的ac4C修饰是破骨细胞分化的重要表观遗传调控,并为炎症性骨丢失提出了一个有希望的治疗靶点。
    BACKGROUND: Excessive osteoclastogenesis is a key driver of inflammatory bone loss. Suppressing osteoclastogenesis has always been considered essential for the treatment of inflammatory bone loss. N-acetyltransferase 10 (NAT10) is the sole enzyme responsible for N4-acetylcytidine (ac4C) modification of mRNA, and is involved in cell development. However, its role in osteoclastogenesis and inflammatory bone loss remained elusive.
    OBJECTIVE: We aimed to clarify the regulatory mechanism of NAT10 and ac4C modification in osteoclastogenesis and inflammatory bone loss.
    METHODS: NAT10 expression and ac4C modification during osteoclastogenesis were determined by quantitative real-time PCR (qPCR), western blotting, dot blot and immunofluorescent staining, and the effect of NAT10 inhibition on osteoclast differentiation in vitro was measured by the tartrate-resistant acid phosphatase staining, podosome belts staining assay and bone resorption pit assay. Then, acRIP-qPCR and NAT10RIP-qPCR, ac4C site prediction, mRNA decay assay and luciferase reporter assay were performed to further study the underlying mechanisms. At last, mice models of inflammatory bone loss were applied to verify the therapeutic effect of NAT10 inhibition in vivo.
    RESULTS: NAT10 expression was upregulated during osteoclast differentiation and highly expressed in alveolar bone osteoclasts from periodontitis mice. Inhibition of NAT10 notably reduced osteoclast differentiation in vitro, as indicated by great reduction of tartrated resistant acid phosphatse positive multinuclear cells, osteoclast-specific gene expression, F-actin ring formation and bone resorption capacity. Mechanistically, NAT10 catalyzed ac4C modification of Fos (encoding AP-1 component c-Fos) mRNA and maintained its stabilization. Besides, NAT10 promoted MAPK signaling pathway and thereby activated AP-1 (c-Fos/c-Jun) transcription for osteoclastogenesis. Therapeutically, administration of Remodelin, the specific inhibitor of NAT10, remarkably impeded the ligature-induced alveolar bone loss and lipopolysaccharide-induced inflammatory calvarial osteolysis.
    CONCLUSIONS: Our study demonstrated that NAT10-mediated ac4C modification is an important epigenetic regulation of osteoclast differentiation and proposed a promising therapeutic target for inflammatory bone loss.
    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
    RNA结合蛋白(RBP),通过RNA的转录后修饰来调节基因表达,在多种生物过程中发挥作用,包括骨细胞发育和骨组织形成。RBP失调可能导致异常的骨稳态并导致各种骨疾病。近年来,RBPs在骨生理和病理生理学中的功能以及潜在的分子机制得到了广泛的研究。本文对此类研究进行了综述,强调RBPs作为治疗干预关键目标的潜力。
    RNA-binding proteins (RBPs), which regulate gene expression through post-transcriptional modifications of RNAs, play a role in diverse biological processes that include bone cell development and bone tissue formation. RBP dysregulation may result in aberrant bone homeostasis and contribute to various bone diseases. The function of RBPs in bone physiology and pathophysiology and the underlying molecular mechanisms have been extensively studied in recent years. This article provides a review of such studies, highlighting the potential of RBPs as pivotal targets for therapeutic intervention.
    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
    Keap1通过协助其泛素化和随后的蛋白水解与Nrf2相互作用。通过在RANKL诱导的破骨细胞形成过程中防止ROS积累,Nrf2激活可以阻止破骨细胞的分化。此外,抑制Keap1-Nrf2PPI可能是触发Nrf2调节氧化应激的有效策略。进行基于结构的虚拟筛选以发现潜在的新型Keap1-Nrf2PPI抑制剂,其中鉴定了KCB-F06。通过TRAP染色和骨吸收测定体外研究了KCB-F06对破骨细胞生成的抑制作用。采用卵巢切除诱导的骨质疏松小鼠模型评价KCB-F06的体内治疗效果。最后,使用实时PCR探索了潜在的机制,西方印迹,和co-IP测定。发现KCB-F06是一种新型Keap1-Nrf2PPI抑制剂。因此,抗氧化剂(HO-1和NQO1)的表达受到抑制,从而减少破骨细胞生成过程中的ROS积累。随后,这导致RANKL诱导的IKB/NF-kB信号的失活。这最终导致包括NFATc1在内的破骨细胞特异性蛋白的下调,NFATc1是破骨细胞生成的必需转录因子。这些结果表明,破骨细胞中的Nrf2激活是破骨细胞骨丢失管理的有价值的工具。此外,KCB-F06是治疗破骨细胞相关骨疾病的替代候选物,也是一种新型小分子,可作为进一步开发Keap1-NRF2PPI抑制剂的模型。
    Keap1 interacts with Nrf2 by assisting in its ubiquitination and subsequent proteolysis. By preventing ROS accumulation during RANKL-induced osteoclastogenesis, Nrf2 activation can prevent the differentiation of osteoclasts. Additionally, inhibiting the Keap1-Nrf2 PPI can be an effective strategy for triggering Nrf2 to regulate oxidative stress. Structure-based virtual screening was performed to discover a potentially novel Keap1-Nrf2 PPI inhibitor wherein KCB-F06 was identified. The inhibitory effects of KCB-F06 on osteoclastogenesis were investigated in vitro through TRAP staining and bone resorption assays. An ovariectomy-induced osteoporosis mouse model was applied to evaluate KCB-F06\'s therapeutic effects in vivo. Lastly, the underlying mechanisms were explored using real-time PCR, Western blotting, and co-IP assays. KCB-F06 was discovered as a novel Keap1-Nrf2 PPI inhibitor. As a result, the expression of antioxidants (HO-1 and NQO1) was suppressed, hence reducing ROS accumulation during osteoclastogenesis. Subsequently, this caused the inactivation of RANKL-induced IKB/NF-kB signaling. This eventually led to the downregulation of osteoclast-specific proteins including NFATc1, which is an essential transcription factor for osteoclastogenesis. These results demonstrated that Nrf2 activation in osteoclasts is a valuable tool for osteoclastic bone loss management. In addition, KCB-F06 presents as an alternative candidate for treating osteoclast-related bone diseases and as a novel small molecule that can serve as a model for further Keap1-NRF2 PPI inhibitor development.
    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
    牙周炎是世界范围内广泛流行的口腔疾病,其特征是牙周膜的破坏和随后的牙周袋的发展。以及牙槽骨的损失,并可能最终导致牙齿脱落。这项研究旨在评估欧帕蒂林的抑制作用,一种从艾蒿中获得的黄酮,体外破骨细胞生成和体内牙周炎。我们发现Eupatilin可以有效地阻止RAW264.7和RANKL诱导的骨髓源性巨噬细胞(BMDMs)的分化,导致成熟破骨细胞的形成。始终如一,骨切片吸收试验显示,Eupatilin以剂量依赖性方式显著抑制破骨细胞介导的骨吸收.Eupatilin还下调了Raw264.7和BMDMs中破骨细胞特异性基因和蛋白质的表达。RNA测序显示Eupatilin显著下调了Siglec-15的表达。基因本体论(GO)和京都基因和基因组百科全书(KEGG)途径分析确定了DEGs中显着富集的途径,包括MAPK信号通路。进一步的机制研究证实,Eupatilin抑制MAPKs/NF-κB信号通路。发现Siglec-15过表达逆转了Eupatilin对破骨细胞分化的抑制作用。此外,激活MAPK信号通路可逆转Siglec-15的下调和Eupatilin对破骨细胞生成的抑制作用。总而言之,Eupatilin通过抑制MAPK信号通路降低Siglec-15的表达,最终导致破骨细胞生成的抑制。同时,Eupatilin抑制了体内实验性牙周炎引起的牙槽骨吸收。Eupatilin在牙周炎的治疗中表现出潜在的治疗效果,使其成为有前途的药剂。
    Periodontitis is a widely prevalent oral disease around the world characterized by the disruption of the periodontal ligament and the subsequent development of periodontal pockets, as well as the loss of alveolar bone, and may eventually lead to tooth loss. This research aims to assess the suppressive impact of Eupatilin, a flavone obtained from Artemisia argyi, on osteoclastogenesis in vitro and periodontitis in vivo. We found that Eupatilin can efficiently obstruct the differentiation of Raw264.7 and bone marrow-derived macrophages (BMDMs) induced by RANKL, leading to the formation of mature osteoclasts. Consistently, bone slice resorption assay showed that Eupatilin significantly inhibited osteoclast-mediated bone resorption in a dose-dependent manner. Eupatilin also downregulated the expression of osteoclast-specific genes and proteins in Raw264.7 and BMDMs. RNA sequencing showed that Eupatilin notably downregulated the expression of Siglec-15. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses identified significantly enriched pathways in DEGs, including MAPK signaling pathway. And further mechanistic investigations confirmed that Eupatilin repressed MAPKs/NF-κBsignaling pathways. It was found that Siglec-15 overexpression reversed the inhibitory impact of Eupatilin on the differentiation of osteoclasts. Furthermore, activating MAPK signaling pathway reversed the downregulation of Siglec-15 and the inhibition of osteoclastogenesis by Eupatilin. To sum up, Eupatilin reduced the expression of Siglec-15 by suppressing MAPK signaling pathway, ultimately leading to the inhibition of osteoclastogenesis. Meanwhile, Eupatilin suppressed the alveolar bone resorption caused by experimentalperiodontitis in vivo. Eupatilin exhibits potential therapeutic effects in the treatment of periodontitis, rendering it a promising pharmaceutical agent.
    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
    具有促进骨形成的多功能的人工韧带的发展,抑制骨吸收,预防感染以获得前交叉韧带(ACL)重建的韧带-骨愈合仍然面临巨大挑战。在这里,制造了一种基于PI纤维机织物(PIF)的新型人工韧带,其通过逐层组装方法(PFPG)涂覆有植酸-镓(PA-Ga)网络。与PIF相比,PFPG与PA-Ga涂层显著抑制破骨细胞分化,同时促进了体外成骨细胞的分化。此外,PFPG明显抑制纤维包裹和骨吸收,同时加速体内韧带-骨愈合的新骨再生。PFPG显著杀死细菌和破坏生物膜,表现出优异的体外抗菌性能以及体内抗感染能力,这归因于Ga离子从PA-Ga涂层的释放。表面特性的协同作用(例如,PFPG的亲水性/表面能和蛋白质吸收)和Ga离子的持续释放显着增强成骨,同时抑制破骨细胞生成,从而实现韧带-骨整合以及抗感染。总之,PFPG显著促进成骨细胞分化,虽然它抑制了破骨细胞的分化,从而抑制用于骨吸收的破骨细胞生成,同时加速用于韧带-骨愈合的骨生成。作为一种新型的人工韧带,PFPG代表了ACL重建中移植物选择的吸引人的选择,并在临床上显示出可观的应用前景。
    The development of an artificial ligament with a multifunction of promoting bone formation, inhibiting bone resorption, and preventing infection to obtain ligament-bone healing for anterior cruciate ligament (ACL) reconstruction still faces enormous challenges. Herein, a novel artificial ligament based on a PI fiber woven fabric (PIF) was fabricated, which was coated with a phytic acid-gallium (PA-Ga) network via a layer-by-layer assembly method (PFPG). Compared with PIF, PFPG with PA-Ga coating significantly suppressed osteoclastic differentiation, while it boosted osteoblastic differentiation in vitro. Moreover, PFPG obviously inhibited fibrous encapsulation and bone absorption while accelerating new bone regeneration for ligament-bone healing in vivo. PFPG remarkably killed bacteria and destroyed biofilm, exhibiting excellent antibacterial properties in vitro as well as anti-infection ability in vivo, which were ascribed to the release of Ga ions from the PA-Ga coating. The cooperative effect of the surface characteristics (e.g., hydrophilicity/surface energy and protein absorption) and sustained release of Ga ions for PFPG significantly enhanced osteogenesis while inhibiting osteoclastogenesis, thereby achieving ligament-bone integration as well as resistance to infection. In summary, PFPG remarkably facilitated osteoblastic differentiation, while it suppressed osteoclastic differentiation, thereby inhibiting osteoclastogenesis for bone absorption while accelerating osteogenesis for ligament-bone healing. As a novel artificial ligament, PFPG represented an appealing option for graft selection in ACL reconstruction and displayed considerable promise for application in clinics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号