Smad2/3

Smad2 / 3
  • 文章类型: Journal Article
    糖尿病足溃疡(DFU)是糖尿病的主要并发症,预后不良,伴有高截肢率和死亡率。当归四尼汤(DSD),作为中国历史悠久的经典配方,已发现可改善DFU症状。然而,DSD用于DFU治疗的机制尚不清楚,没有系统的阐述。在体内,DFU大鼠模型建立后,DSD干预低,中等剂量和高剂量,以二甲双胍(DM)为阳性对照组。伤口愈合检测,HE染色病理改变,ELISA和qRT-PCR检测炎症因子表达,通过ELISA测定氧化应激水平,通过Westernblot进行AGEs/RAGE/TGF-β/Smad2/3的表达。体外,在人真皮成纤维细胞成体(HDF-a)细胞中进行基于DSD治疗的LY2109761(TGF-β通路抑制剂)干预。CCK8的细胞活力,细胞划痕的迁移能力,通过流式细胞术细胞凋亡,Westernblot检测AGEs/RAGE/TGF-β/Smad2/3的表达。DFU大鼠表现出升高的AGEs/RAGE表达,而TGF-β1和p-Smad3/Smad3蛋白表达降低,伴有较高的IL-1β,IL-6,TNF-α水平,和氧化应激。DSD干预逆转了上述效果。葡萄糖诱导导致较低的细胞活力,迁移,TGF-β1和p-Smad3/Smad3蛋白表达,HDF-a细胞中凋亡和AGEs/RAGE表达增加。这些效果在DSD干预后逆转,和进一步LY2109761干预抑制细胞中的DSD效应。DSD干预可能通过调节AGEs/RAGE/TGF-β/Smad2/3的表达促进DFU创面愈合,为DSD治疗DFU的临床应用提供了科学实验依据。
    Diabetic foot ulcer (DFU) is a predominant complication of diabetes mellitus with poor prognosis accompanied by high amputation and mortality rates. Dang-Gui-Si-Ni decoction (DSD), as a classic formula with a long history in China, has been found to improve DFU symptoms. However, mechanism of DSD for DFU therapy remains unclear with no systematic elaboration. In vivo, following establishment of DFU rat model, DSD intervention with low, medium and high doses was done, with Metformin (DM) as a positive control group. With wound healing detection, pathological changes by HE staining, inflammatory factor expression by ELISA and qRT-PCR, oxidative stress levels by ELISA, and AGEs/RAGE/TGF-β/Smad2/3 expression by Western blot were performed. In vitro, intervention with LY2109761 (TGF-β pathway inhibitor) based on DSD treatment in human dermal fibroblast-adult (HDF-a) cells was made. Cell viability by CCK8, migration ability by cell scratch, apoptosis by flow cytometry, and AGEs/RAGE/TGF-β/Smad2/3 expression by Western blot were measured. DFU rats exhibited elevated AGEs/RAGE expression, whereas decreased TGF-β1 and p-Smad3/Smad3 protein expression, accompanied by higher IL-1β, IL-6, TNF-α levels, and oxidative stress. DSD intervention reversed above effects. Glucose induction caused lower cell viability, migration, TGF-β1 and p-Smad3/Smad3 protein expression, with increased apoptosis and AGEs/RAGE expression in HDF-a cells. These effects were reversed after DSD intervention, and further LY2109761 intervention inhibited DSD effects in cells. DSD intervention may facilitate wound healing in DFU by regulating expression of AGEs/RAGE/TGF-β/Smad2/3, providing scientific experimental evidence for DSD clinical application for DFU 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
    糖尿病肾病(DN)是糖尿病患者常见的微血管并发症之一。骨髓间充质干细胞(MSCs)在DN治疗中受到广泛关注,但其机制尚不清楚。这里,我们证明MSC可以减轻高糖(HG)诱导的人肾小管上皮细胞(HK-2细胞)损伤,并改善DN小鼠的肾损伤。我们确定Smad2/3负责MSC调节的DN进展。Smad2/3的活性在HG诱导的HK-2细胞和DN小鼠中主要上调,并被MSC抑制。通过转化生长因子-β1(TGF-β1)激活Smad2/3可以消除MSCs对HG诱导的HK-2细胞损伤和DN小鼠肾脏损伤的保护作用。据报道,Smad2/3与N6-甲基腺苷(m6A)复合物的甲基转移酶相互作用,我们发现了甲基转移酶,Wilms\'肿瘤1相关蛋白(WTAP),参与MSCs-Smad2/3调控的DN发育。此外,WTAP过表达消除了MSCs对HG诱导的HK-2细胞损伤和DN小鼠肾损伤的改善作用。随后,α-烯醇化酶(ENO1)是WTAP介导的m6A修饰的下游靶标,并有助于MSC介导的调节。总的来说,这些发现揭示了DN进展的分子机制,并表明Smad2/3/WTAP/ENO1可能是MSCs介导的DN治疗的靶点.
    Diabetic nephropathy (DN) is one of the common microvascular complications in diabetic patients. Marrow mesenchymal stem cells (MSCs) have attracted attention in DN therapy but the underlying mechanism remains unclear. Here, we show that MSC administration alleviates high glucose (HG)-induced human kidney tubular epithelial cell (HK-2 cell) injury and ameliorates renal injury in DN mice. We identify that Smad2/3 is responsible for MSCs-regulated DN progression. The activity of Smad2/3 was predominantly upregulated in HG-induced HK-2 cell and DN mice and suppressed with MSC administration. Activation of Smad2/3 via transforming growth factor-β1 (TGF-β1) administration abrogates the protective effect of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Smad2/3 has been reported to interact with methyltransferase of N6-methyladenosine (m6A) complex and we found a methyltransferase, Wilms\' tumor 1-associating protein (WTAP), is involved in MSCs-Smad2/3-regulated DN development. Moreover, WTAP overexpression abrogates the improvement of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Subsequently, α-enolase (ENO1) is the downstream target of WTAP-mediated m6A modification and contributes to the MSCs-mediated regulation. Collectively, these findings reveal a molecular mechanism in DN progression and indicate that Smad2/3/WTAP/ENO1 may present a target for MSCs-mediated DN 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
    导致心脏重构和心力衰竭的主要致病因素是心肌纤维化。最近的研究表明,microRNAs在心脏纤维化的进展中至关重要。心肌纤维化被认为是通过骨形态发生蛋白和激活素膜结合抑制剂(BAMBI)来缓解的,它通过阻断转化生长因子β1(TGF-β1)信号通路来做到这一点。这里,本研究旨在阐明miR-19a-3p对BAMBI的转录后调控及其在TGF-β1诱导的心肌成纤维细胞活化中的作用.横向主动脉缩窄(TAC)引起心肌间质和血管周围胶原沉积。RT-PCR显示miR-19a-3p在心肌纤维化组织中上调,TGF-β1诱导心肌成纤维细胞中miR-19a-3p表达增加。双荧光素酶报告基因试验和qRT-PCR证实miR-19a-3p与BAMBImRNA3'UTR直接结合,从而减少BAMBI表达,这降低了BAMBI抑制TGF-β1的能力。此外,miR-19a-3p模拟物增加TGF-β1/SMAD2/3通路信号的激活,支持心脏成纤维细胞活化,被BAMBI的过表达所阻断。这些发现暗示miR-19a-3p通过抑制BAMBI增强TGF-β1/SMAD2/3的激活,进一步促进心脏成纤维细胞的激活,因此可能提供一种新的策略来解决心肌纤维化。
    The main pathogenic factor leading to cardiac remodeling and heart failure is myocardial fibrosis. Recent research indicates that microRNAs are essential for the progress of cardiac fibrosis. Myocardial fibrosis is considered to be alleviated through the bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI), which does this by blocking the transforming growth factor β1 (TGF-β1) signaling pathway. Here, this study sought to elucidate the post-transcriptional regulation of miR-19a-3p on BAMBI and its role in TGF-β1-induced cardiac fibroblast activation. Transverse aortic constriction (TAC) caused both myocardial interstitial and perivascular collagen deposition. RT-PCR showed that miR-19a-3p was upregulated in the myocardial tissue of cardiac fibrosis, and TGF-β1 induced an increase of miR-19a-3p expression in cardiac fibroblasts. The dual-luciferase reporter test and qRT-PCR confirmed that miR-19a-3p directly combined with BAMBI mRNA 3\'UTR, thus reduced BAMBI expression, which diminished the capability of BAMBI to inhibit TGF-β1. Furthermore, miR-19a-3p mimic increased the activation of TGF-β1/SMAD2/3 pathway signaling, which supported cardiac fibroblast activation, which blocked by overexpression of BAMBI. These findings imply that miR-19a-3p enhances the activation of TGF-β1/SMAD2/3 by inhibiting BAMBI, further boosting the activation of cardiac fibroblasts, and may thus offer a novel strategy to tackling myocardial fibrosis.
    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:多功能促纤维化细胞因子转化生长因子-β2(TGF-β2)与原发性开角型青光眼的病理生理有关。芍药苷(PAE)是一种具有多种药理作用的单萜苷,如抗氧化剂,抗纤维化,和抗炎特性。研究表明,芍药苷保护人角膜上皮细胞,视网膜色素上皮细胞,和视网膜小胶质细胞的损伤。这里,PAE在小梁网(TM)微环境中细胞外基质(ECM)的TGF-β2依赖性重塑中的生物学作用。
    方法:将在无血清培养基中调节的原代或转化的(GTM3)人TM(HTM)细胞与TGF-β2(5ng/mL)孵育。将PAE(300μM)添加到HTM细胞的血清饥饿汇合培养物中2小时,随后与TGF-β2孵育22小时。SB-431542,TGF-β受体抑制剂(10μM),用作阳性对照。通过CellROX绿染料评估细胞内ROS的水平。Western印迹用于测量TGF-β2/Smad2/3信号传导相关分子的水平。通过RT-qPCR评估胶原蛋白1α1,胶原蛋白4α1和结缔组织生长因子(CTGF)的表达。采用免疫荧光法检测HTM细胞中I/IV型胶原的表达。进行Phalloidin染色测定以评估细胞中的F-肌动蛋白应力纤维形成。
    结果:PAE在原代或转化的HTM细胞中减弱TGF-β2诱导的氧化应激并抑制TGF-β2诱导的Smad2/3信号传导。此外,PAE抑制了TGF-β2诱导的胶原蛋白1α1,胶原蛋白4α1和CTGF表达的上调,并降低了TGF-β2介导的胶原蛋白I/IV表达和F-肌动蛋白应激纤维的形成。
    结论:PAE通过Smad2/3信号失活减轻TGF-β2诱导的HTM细胞ECM沉积和氧化应激。
    BACKGROUND: The multifunctional profibrotic cytokine transforming growth factor-beta2 (TGF-β2) is implicated in the pathophysiology of primary open angle glaucoma. Paeoniflorin (PAE) is a monoterpene glycoside with multiple pharmacological efficacies, such as antioxidant, anti-fibrotic, and anti-inflammatory properties. Studies have demonstrated that paeoniflorin protects human corneal epithelial cells, retinal pigment epithelial cells, and retinal microglia from damage. Here, the biological role of PAE in TGF-β2-dependent remodeling of the extracellular matrix (ECM) within the trabecular meshwork (TM) microenvironment.
    METHODS: Primary or transformed (GTM3) human TM (HTM) cells conditioned in serum-free media were incubated with TGF-β2 (5 ng/mL). PAE (300 μM) was added to serum-starved confluent cultures of HTM cells for 2 h, followed by incubation with TGF-β2 for 22 h. SB-431542, a TGF-β receptor inhibitor (10 μM), was used as a positive control. The levels of intracellular ROS were evaluated by CellROX green dye. Western blotting was used to measure the levels of TGF-β2/Smad2/3 signaling-related molecules. Collagen 1α1, collagen 4α1, and connective tissue growth factor (CTGF) expression was evaluated by RT-qPCR. Immunofluorescence assay was conducted to measure collagen I/IV expression in HTM cells. Phalloidin staining assay was conducted for evaluating F-actin stress fiber formation in the cells.
    RESULTS: PAE attenuated TGF-β2-induced oxidative stress and suppressed TGF-β2-induced Smad2/3 signaling in primary or transformed HTM cells. Additionally, PAE repressed TGF-β2-induced upregulation of collagen 1α1, collagen 4α1, and CTGF expression and reduced TGF-β2-mediated collagen I/IV expression and of F-actin stress fiber formation in primary or transformed HTM cells.
    CONCLUSIONS: PAE alleviates TGF-β2-induced ECM deposition and oxidative stress in HTM cells through inactivation of Smad2/3 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
    三阴性乳腺癌(TNBC)是一种侵袭性亚型,预后不良。硫链菌素对包括TNBC在内的几种癌症具有抗肿瘤活性。本文讨论了硫链菌素在TNBC中的新分子机制。硫链菌素抑制MDA-MB-231细胞活力,伴随着c-FLIP和p-SMAD2/3的减少。c-FLIP过表达降低MDA-MB-231细胞对硫链菌素的敏感性,SMAD2/3敲除增加MDA-MB-231细胞对硫链菌素的敏感性。此外,c-FLIP过表达显著增加SMAD2/3蛋白的表达和磷酸化,反之亦然。总之,我们的研究揭示了c-FLIP/SMAD2/3信号通路是硫链菌素抗肿瘤活性的新机制。
    Triple-negative breast cancer (TNBC) is an aggressive subtype with poor prognosis of breast cancer. Thiostrepton exerts anti-tumor activities against several cancers including TNBC. Herein we discussed the new molecular mechanisms of thiostrepton in TNBC. Thiostrepton inhibited MDA-MB-231 cell viability, accompanied by a decrease of c-FLIP and p-SMAD2/3. c-FLIP overexpression reduced the sensitivity of MDA-MB-231 cells to thiostrepton, while SMAD2/3 knockdown increased the sensitivity of MDA-MB-231 cells to thiostrepton. Moreover, c-FLIP overexpression significantly increased the expression and phosphorylation of SMAD2/3 proteins and vice versa. In conclusion, our study reveals c-FLIP/SMAD2/3 signaling pathway as a novel mechanism of antitumor activity of thiostrepton.
    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:阐明超负荷性骨关节炎(OA)的分子机制,寻找新的治疗靶点。
    方法:我们利用人类软骨标本,小鼠软骨细胞,内侧半月板(DMM)小鼠模型的不稳定,和小鼠后肢负重模型,以验证超负荷对软骨细胞衰老和OA发育的作用。然后,我们观察到PIEZO1-miR-155-5p-GDF6-SMAD2/3信号轴在体内过载下对关节代谢稳态的保护作用,体外和体外通过qPCR,蛋白质印迹,酶联免疫吸附测定(ELISA),免疫组织化学,免疫荧光,SA-β-gal染色,CCK8测定,etal.最后,我们验证了关节内注射miR-155-5p抑制剂或重组GDF6对小鼠超负荷诱导的OA模型的治疗效果.
    结果:软骨细胞通过PIEZO1感觉到机械过载,并上调miR-155-5p表达。MiR-155-5p模拟物可以复制过载诱导的软骨细胞衰老和OA的作用。此外,miR-155-5p可以抑制关节内各种组织中Gdf6-Smad2/3的mRNA表达。超负荷可以通过下调软骨细胞和滑膜细胞中合成代谢指标的表达和上调分解代谢指标的表达来破坏关节代谢稳态。而miR-155-5p抑制或GDF6补充可以通过保持关节稳态发挥拮抗作用。最后,在体内过载模型中,关节内注射miR-155-5p抑制剂或重组GDF6可显着减轻即将发生的OA的严重程度,并减轻现有OA的进展.
    结论:GDF6过表达或miR-155-5p抑制可通过PIEZO1-miR-155-5p-GDF6-SMAD2/3信号通路减轻过载诱导的软骨细胞衰老和OA。本研究为超负荷性OA的治疗提供了新的治疗靶点。
    OBJECTIVE: To elucidate the molecular mechanism of overloading-induced osteoarthritis (OA) and to find a novel therapeutic target.
    METHODS: We utilized human cartilage specimens, mouse chondrocytes, a destabilization of the medial meniscus (DMM) mouse model, and a mouse hindlimb weight-bearing model to validate the role of overloading on chondrocyte senescence and OA development. Then, we observed the effect of PIEZO1-miR-155-5p-GDF6-SMAD2/3 signaling axis on the preservation of joint metabolic homeostasis under overloading in vivo, in vitro and ex vivo by qPCR, Western blot, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, immunofluorescence, SA-β-gal staining, CCK8 assay, et al. Finally, we verified the therapeutic effects of intra-articular injection of miR-155-5p inhibitor or recombinant GDF6 on the murine overloading-induced OA models.
    RESULTS: Chondrocytes sensesed the mechanical overloading through PIEZO1 and up-regulated miR-155-5p expression. MiR-155-5p mimics could copy the effects of overloading-induced chondrocyte senescence and OA. Additionally, miR-155-5p could suppress the mRNA expression of Gdf6-Smad2/3 in various tissues within the joint. Overloading could disrupt joint metabolic homeostasis by downregulating the expression of anabolism indicators and upregulating the expression of catabolism indicators in the chondrocytes and synoviocytes, while miR-155-5p inhibition or GDF6 supplementation could exert an antagonistic effect by preserving the joint homeostasis. Finally, in the in vivo overloading models, intra-articular injection of miR-155-5p inhibitor or recombinant GDF6 could significantly mitigate the severity of impending OA and lessened the progression of existing OA.
    CONCLUSIONS: GDF6 overexpression or miR-155-5p inhibition could attenuate overloading-induced chondrocyte senescence and OA through the PIEZO1-miR-155-5p-GDF6-SMAD2/3 signaling pathway. Our study provides a new therapeutic target for the treatment of overloading-induced 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
    目的:驱动蛋白家族成员18A(KIF18A)因其在各种癌症类型中的异常表达而著称,其关键作用是驱动癌症进展。在这项研究中,我们旨在研究KIF18A对HCC进展的影响的复杂分子机制。
    方法:蛋白质印迹试验,我们进行了实时定量PCR和免疫组织化学分析,以定量评估KIF18A在HCC组织中的表达.然后,我们通过使用短发夹RNA(shRNA)沉默内源性KIF18A并引入外源质粒以过表达KIF18A,在HCC细胞内进行遗传操作。我们监测细胞进展,分析细胞周期和细胞凋亡,并在体外和体内评估细胞迁移和侵袭。此外,我们进行了RNA测序,利用Reactome和KEGG富集方法探索KIF18A相关信号通路,并验证了这些通路中的关键介质.
    结果:TCGA-LIHC数据库的分析显示KIF18A在HCC组织中明显过表达,这一发现随后通过分析HCC患者的临床样本得到证实.值得注意的是,沉默KIF18A导致细胞增殖明显抑制,体外迁移和侵袭。此外,在皮下和原位异种移植模型中,抑制KIF18A显著减轻肿瘤重量和肺转移结节的数量。机械上,KIF18A似乎通过上调MAD2和CDK1/CyclinB1表达水平促进细胞增殖,SMAD2/3信号的激活有助于KIF18A驱动的转移。
    结论:我们的研究阐明了KIF18A介导肝癌细胞增殖和转移的分子机制,为潜在的治疗靶点提供新的见解。
    OBJECTIVE: Kinesin family member 18A (KIF18A) is notable for its aberrant expression across various cancer types and its pivotal role is driving cancer progression. In this study, we aim to investigate the intricate molecular mechanisms underlying the impact of KIF18A on the progression of HCC.
    METHODS: Western blotting assays, a quantitative real-time PCR and immunohistochemical analyses were performed to quantitatively assess KIF18A expression in HCC tissues. We then performed genetic manipulations within HCC cells by silencing endogenous KIF18A using short hairpin RNA (shRNA) and introducing exogenous plasmids to overexpress KIF18A. We monitored cell progression, analyzed cell cycle and cell apoptosis and assessed cell migration and invasion both in vitro and in vivo. Moreover, we conducted RNA-sequencing to explore KIF18A-related signaling pathways utilizing Reactome and KEGG enrichment methods and validated these critical mediators in these pathways.
    RESULTS: Analysis of the TCGA-LIHC database revealed pronounced overexpression of KIF18A in HCC tissues, the finding was subsequently confirmed through the analysis of clinical samples obtained from HCC patients. Notably, silencing KIF18A in cells led to an obvious inhibition of cell proliferation, migration and invasion in vitro. Furthermore, in subcutaneous and orthotopic xenograft models, suppression of KIF18A sgnificantly redudce tumor weight and the number of lung metastatic nodules. Mechanistically, KIF18A appears to facilitate cell proliferation by upregulating MAD2 and CDK1/CyclinB1 expression levels, with the activation of SMAD2/3 signaling contributing to KIF18A-driven metastasis.
    CONCLUSIONS: Our study elucidates the molecular mechanism by which KIF18A mediates proliferation and metastasis in HCC cells, offering new insights into potential therapeutic targets.
    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:内皮-间充质转化(EndMT)在动脉粥样硬化的病理进展过程中在内皮功能障碍中起关键作用;然而,其详细机制尚不清楚。在这里,我们探讨了EndMT中上游刺激因子1(USF1)在动脉粥样硬化过程中的生物学功能和机制.
    方法:在高脂饮食喂养的ApoE-/-小鼠和ox-LDL暴露的人脐静脉内皮细胞(HUVECs)中建立体内和体外动脉粥样硬化模型。斑块的形成,胶原蛋白和脂质沉积,并通过苏木精和伊红(HE)评估主动脉组织的形态学变化,Masson,油红O和Verhoeff-VanGieson(EVG)染色,分别。通过EndMT相关蛋白的表达水平测定EndMT。通过RT-qPCR和Western印迹检测靶分子表达。通过ELISA测量促炎细胞因子的释放。通过transwell和划痕测定检测HUVEC的迁移。通过双荧光素酶报告实验研究了分子机制,ChIP,和Co-IP测定。
    结果:USF1在动脉粥样硬化患者中表达上调。USF1敲低通过上调CD31和VE-Cadherin抑制EndMT,同时下调α-SMA和波形蛋白,从而抑制炎症,和在暴露于ox-LDL的HUVECs中的迁移。此外,USF1转录激活泛素特异性蛋白酶14(USP14),这促进了NLR家族CARD结构域含5(NLRC5)的去泛素化和上调,以及随后的Smad2/3途径激活。sh-USF1或sh-USP14对EndMT的抑制作用被USP14或NLRC5过表达部分逆转。最后,USF1敲低通过抑制小鼠EndMT延迟动脉粥样硬化进展。
    结论:我们的发现表明USF1/USP14/NLRC5轴通过促进EndMT对动脉粥样硬化发展的贡献,提供有效的治疗靶点。
    BACKGROUND: Endothelial-to-Mesenchymal Transformation (EndMT) plays key roles in endothelial dysfunction during the pathological progression of atherosclerosis; however, its detailed mechanism remains unclear. Herein, we explored the biological function and mechanisms of upstream stimulating factor 1 (USF1) in EndMT during atherosclerosis.
    METHODS: The in vivo and in vitro atherosclerotic models were established in high fat diet-fed ApoE-/- mice and ox-LDL-exposed human umbilical vein endothelial cells (HUVECs). The plaque formation, collagen and lipid deposition, and morphological changes in the aortic tissues were evaluated by hematoxylin and eosin (HE), Masson, Oil red O and Verhoeff-Van Gieson (EVG) staining, respectively. EndMT was determined by expression levels of EndMT-related proteins. Target molecule expression was detected by RT-qPCR and Western blotting. The release of pro-inflammatory cytokines was measured by ELISA. Migration of HUVECs was detected by transwell and scratch assays. Molecular mechanism was investigated by dual-luciferase reporter assay, ChIP, and Co-IP assays.
    RESULTS: USF1 was up-regulated in atherosclerosis patients. USF1 knockdown inhibited EndMT by up-regulating CD31 and VE-Cadherin, while down-regulating α-SMA and vimentin, thereby repressing inflammation, and migration in ox-LDL-exposed HUVECs. In addition, USF1 transcriptionally activated ubiquitin-specific protease 14 (USP14), which promoted de-ubiquitination and up-regulation of NLR Family CARD Domain Containing 5 (NLRC5) and subsequent Smad2/3 pathway activation. The inhibitory effect of sh-USF1 or sh-USP14 on EndMT was partly reversed by USP14 or NLRC5 overexpression. Finally, USF1 knockdown delayed atherosclerosis progression via inhibiting EndMT in mice.
    CONCLUSIONS: Our findings indicate the contribution of the USF1/USP14/NLRC5 axis to atherosclerosis development via promoting EndMT, which provide effective therapeutic targets.
    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
    右美托咪定(Dex)是一种有效的α2-肾上腺素能受体(α2-AR)激动剂,已被证明可以预防脓毒症引起的肺损伤,然而,这种保护的潜在机制尚未完全理解。自噬和Smad2/3信号通路在脓毒症肺损伤中发挥重要作用,但是Dex和Smad2/3之间的关系并不清楚。本研究旨在探讨自噬和Smad2/3信号通路在Dex介导脓毒症肺损伤治疗中的作用。在C57BL/6J小鼠中使用盲肠结扎和穿孔(CLP)进行脓毒症。
    将小鼠随机分为四组(每组n=6):假手术,CLP,CLP-Dex,和CLP-Dex-YOH,盐酸育亨宾(YOH)是α2-AR阻断剂。小心地分离盲肠以避免血管损伤,并用18号针进行鉴定和穿刺两次。病理变化,炎症因子水平,氧化应激,自噬,肺组织中Smad2/3信号通路相关蛋白水平,测定血清超氧化物歧化酶(SOD)和丙二醛(MDA)活性。
    CLP诱导的肺损伤反映在炎性细胞因子水平的升高,凋亡,和氧化应激,自噬和Smad2/3信号通路相关蛋白的表达增加。Dex可以逆转这些变化,并在脓毒症期间对肺产生保护作用。然而,YOH的给药显著降低了Dex对脓毒症小鼠的积极作用。
    Dex通过调节自噬和Smad2/3信号通路发挥对脓毒症肺损伤的有益作用。
    UNASSIGNED: Dexmedetomidine (Dex) is a potent α2-adrenergic receptor(α2-AR) agonist that has been shown to protect against sepsis-induced lung injury, however, the underlying mechanisms of this protection are not fully understood. Autophagy and the Smad2/3 signaling pathway play important roles in sepsis-induced lung injury, but the relationship between Dex and Smad2/3 is not clear. This study aimed to investigate the role of autophagy and the Smad2/3 signaling pathway in Dex-mediated treatment of sepsis-induced lung injury. Sepsis was performed using cecal ligation and puncture (CLP) in C57BL/6J mice.
    UNASSIGNED: Mice were randomly assigned to four groups (n=6 per group): sham, CLP, CLP-Dex, and CLP-Dex-YOH, Yohimbine hydrochloride (YOH) is an α2-AR blocker. The cecum was carefully separated to avoid blood vessel damage and was identified and punctured twice with an 18-gauge needle. The pathological changes, inflammatory factor levels, oxidative stress, autophagy, Smad2/3 signaling pathway-related protein levels in lung tissues, and the activity of superoxide dismutase (SOD) and malonaldehyde (MDA) in the serum were measured.
    UNASSIGNED: CLP-induced lung injury was reflected by increased levels of inflammatory cytokines, apoptosis, and oxidative stress, along with an increase in the expression of autophagy and Smad2/3 signaling pathway-related proteins. Dex could reverse these changes and confer a protective effect on the lung during sepsis. However, the administration of YOH significantly reduced the positive effects of Dex in mice with sepsis.
    UNASSIGNED: Dex exerts its beneficial effects against sepsis-induced lung injury through the regulation of autophagy and the Smad2/3 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
    目的:食管再狭窄是食管支架置入术后的严重并发症,影响支架植入术的临床预后和患者的生活质量。TGF-β1/Smads信号通路在皮肤创伤后嗜酸性粒细胞性食管炎的发生发展和瘢痕修复中起重要作用。然而,TGF-β1/Smads在食管支架置入后食管再狭窄发展中的作用尚不清楚.我们的研究旨在探讨TGF-β1/Smads是否在食管支架术后再狭窄的发生发展中起重要作用。以及外源性TGF-β1抑制剂补充是否可以改善食管支架术后的食管再狭窄。
    方法:建立大鼠食管支架置入后再狭窄模型,并通过免疫化学(IHC)测定TGF-β1/Smads信号通路的表达水平和成纤维细胞活化的相关标志物,Western印迹和实时qPCR。当暴露于有或没有TGF-β1抑制剂P144的rhTGF-β1时,食管成纤维细胞中的所有指标也被确定。
    结果:支架组血清IL-1β和TNFα水平较空白对照组明显升高,用P144治疗时明显改善。支架组TGF-β1/Smads信号通路及成纤维细胞活化的相关标志物较空白对照组明显增多,用P144治疗时明显改善。当暴露于rhTGF-β1时,所有指标均显着增加,而当P144处理时,所有指标均显着降低。
    结论:TGF-β1抑制剂P144可通过抑制TGF-β1/Smads信号通路下调成纤维细胞活化相关标志物的表达水平,从而保护食管支架术后良性再狭窄。并可作为食管支架置入术后良性再狭窄的新疗法。
    OBJECTIVE: Esophageal restenosis is a serious complication after esophageal stent placement, which influences the clinical prognosis of stent implantation and the patient\'s quality of life. TGF-β1/Smads signaling pathway plays an important role in the development of the eosinophilic esophagitis and scar repair after skin trauma. However, the role of TGF-β1/Smads in the development of esophageal restenosis after esophageal stent placement remains unknown. Our study aimed to investigate whether TGF-β1/Smads plays an important role in the development of esophageal restenosis after esophageal stent, and whether the exogenous TGF-β1 inhibitor supplement could ameliorate the esophageal restenosis after esophageal stent.
    METHODS: We established the model of esophageal restenosis after esophageal stenting in rats, and determined the expression levels of TGF-β1/Smads signaling pathway and the relevant markers of fibroblast activation by immunochemistry (IHC), Western Blot and real time qPCR. Those all the indicators were also determined in esophageal fibroblast when exposed to rhTGF-β1 with or without TGF-β1 inhibitor P144.
    RESULTS: The serum level of IL-1β and TNFα were significantly increased in stent implantation group compared to blank control group, and obviously ameliorated when treated with P144. The TGF-β1/Smads signaling pathway and the relevant markers of fibroblast activation were significantly increased in stent implantation group compared to blank control group, and obviously ameliorated when treated with P144. Those all the indicators were significantly increased when exposed to rhTGF-β1, and obviously decreased when treated with P144.
    CONCLUSIONS: TGF-β1 Inhibitor P144 could protect against benign restenosis after esophageal stenting by down-regulating the expression levels of relevant markers of fibroblast activation through TGF-β1/Smads signaling pathway inhibition, and may be used as a novel therapy for benign restenosis after esophageal stenting.
    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