PDCD4

PDCD4
  • 文章类型: Journal Article
    以前的研究结果表明,程序性细胞死亡4(PDCD4)在脑缺血(CI)中过度表达,和母亲对十一项截瘫同系物1(SMAD1)是PDCD4的转录因子,并且在CI中也升高;然而,SMAD1/PDCD4轴在CI中的调控机制尚不清楚。目前的工作旨在探索SMAD1/PDCD4inCI的作用和相关机制。PDCD4和SMAD1的表达已通过实时逆转录聚合酶链反应(RT-qPCR)方法检测,并进行了受试者工作特征(ROC)曲线分析,以确定PDCD4和SMAD1的潜在诊断价值。氧葡萄糖剥夺(OGD)模型已用于研究PDCD4和SMAD1对CI的影响。使用TdT介导的dUTP缺口末端标记(TUNEL)测定评价细胞凋亡。SMAD1和PDCD4轴之间的相互作用已通过使用双荧光素酶报告基因以及染色质免疫沉淀(Ch-IP)测定得到证实。最后,已经检查了SMAD1/PDCD4轴对神经元细胞铁性凋亡的影响。PDCD4在CI患者血液样本中过度表达。ROC分析显示PDCD4的AUC为0.7478,NIHSS和MRS评分与PDCD4表达呈正相关。此外,建立细胞OGD模型,PDCD4敲低抑制神经元凋亡。此外,PDCD4的敲低也在体外抑制OGD处理的神经元细胞的铁凋亡。此外,在CI患者的血液样本中SMAD1上调,NIHSS和MRS评分与SMAD1表达呈正相关,SMAD1是PDCD4的转录因子,SMAD1可以转录调节PDCD4的表达。最后,SMAD1可以通过调节PDCD4的转录来调节神经元细胞的铁凋亡。SMAD1/PDCD4轴调节生长,凋亡,和神经元细胞的铁性凋亡,提示靶向SMAD1/PDCD4轴可能是一种潜在的治疗方法。
    Results of previous studies suggested that programmed cell death 4 (PDCD4) was overexpressed in cerebral ischemia (CI), and mothers against decapentaplegic homolog 1 (SMAD1) is a transcription factor of PDCD4, and it is also elevated in CI; however, the regulatory mechanism of SMAD1/PDCD4 axis in CI remains unclear. The current work has been designed to explore the role and associated mechanisms of SMAD1/PDCD4 in CI. PDCD4 and SMAD1 expressions have been examined by real-time reverse transcription-polymerase chain reaction (RT-qPCR) method, and receiver operating characteristic (ROC) curve analysis has been performed to determine the potential diagnostic value of PDCD4 and SMAD1. An oxygen-glucose deprivation (OGD) model has been used to investigate the effects of PDCD4 and SMAD1 on CI in vitro. Cell apoptosis was evaluated using TdT-mediated dUTP nick end labeling (TUNEL) assays. The interaction between SMAD1 and PDCD4 axis has been confirmed by using dual-luciferase reporter as well as chromatin immunoprecipitation (Ch-IP) assays. Finally, the effects of SMAD1/PDCD4 axis on the ferroptosis of neuron cells have been examined. PDCD4 was overexpressed in blood samples of CI patients. ROC analysis showed the AUC for PDCD4 was 0.7478, and NIHSS and MRS scores were positively correlated with PDCD4 expression. Moreover, the cellular OGD model was established and knockdown of PDCD4 suppressed the apoptosis of neurons. Besides, knockdown of PDCD4 also inhibited ferroptosis of OGD-treated neuron cells in vitro. Additionally, SMAD1 was upregulated in blood samples of CI patients, NIHSS and MRS scores were positively correlated with SMAD1 expression, and SMAD1 is a transcriptional factor of PDCD4, and SMAD1 could transcriptionally regulate the expression of PDCD4. Finally, SMAD1 could regulate the ferroptosis of neuron cells through regulating the transcription of PDCD4. The SMAD1/PDCD4 axis regulates the growth, apoptosis, and ferroptosis of neuron cells, suggesting that targeting the SMAD1/PDCD4 axis may be a potential therapeutic method.
    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
    程序性细胞死亡蛋白4(PDCD4)有助于调节一系列细胞过程,如翻译,凋亡,信号转导,和炎症反应。PDCD4和哺乳动物雷帕霉素靶蛋白(mTOR)通路之间存在显著的负相关,这是细胞生长控制不可或缺的一部分。mTOR的激活与PDCD4的降解有关。尽管PDCD4在肿瘤发生和免疫反应调节中的作用已得到充分确立,其在分枝杆菌感染中的功能及其与mTOR途径的相互作用需要进一步阐明。这项研究调查了在分枝杆菌感染的背景下PDCD4表达的调节,揭示了不同分枝杆菌物种的一致下调模式。这一观察结果强调了PDCD4作为评估此类感染期间mTOR途径激活的生物标志物的潜在效用。在这个发现的基础上,我们采用了一种新的方法,使用基于PDCD4的mTOR(Tor)-信号指示剂(TOSI)报告细胞来高通量筛选FDA批准的药物,专注于mTOR抑制剂。这种方法促进了几种代理人的识别,包括已知的mTOR抑制剂,它上调了PDCD4的表达,并同时表现出阻止巨噬细胞内分枝杆菌增殖的功效。这些结果不仅加强了PDCD4作为理解传染病的关键标志物的重要性,特别是分枝杆菌感染,也阐明了它在鉴定mTOR抑制剂方面的潜力,从而促进治疗策略的发展。
    目的:本研究强调哺乳动物雷帕霉素靶蛋白(mTOR)通路在巨噬细胞对分枝杆菌感染的反应中的关键作用,阐明分枝杆菌如何激活mTOR,导致PDCD4降解。利用(Tor)-信号指示物(TOSI)载体实时监测mTOR活性代表了在分枝杆菌感染期间理解mTOR调节的显著进步。这些发现加深了我们对分枝杆菌先天免疫机制的理解,并将PDCD4作为感染性疾病中mTOR活性的新标志物。重要的是,这项研究为使用FDA批准的药物高通量筛选mTOR抑制剂奠定了基础,提供了重新利用治疗分枝杆菌感染的潜力。抑制mTOR激活的药物的鉴定为宿主定向治疗开辟了新的途径,标志着在抗击结核病和其他分枝杆菌疾病方面迈出了重要的一步。
    Programmed cell death protein 4 (PDCD4) is instrumental in regulating a range of cellular processes such as translation, apoptosis, signal transduction, and inflammatory responses. There is a notable inverse correlation between PDCD4 and the mammalian target of rapamycin (mTOR) pathway, which is integral to cellular growth control. Activation of mTOR is associated with the degradation of PDCD4. Although the role of PDCD4 is well established in oncogenesis and immune response regulation, its function in mycobacterial infections and its interplay with the mTOR pathway necessitate further elucidation. This study investigates the modulation of PDCD4 expression in the context of mycobacterial infections, revealing a consistent pattern of downregulation across diverse mycobacterial species. This observation underscores the potential utility of PDCD4 as a biomarker for assessing mTOR pathway activation during such infections. Building on this finding, we employed a novel approach using PDCD4-based mTOR (Tor)-signal-indicator (TOSI) reporter cells for the high-throughput screening of FDA-approved drugs, focusing on mTOR inhibitors. This methodology facilitated the identification of several agents, inclusive of known mTOR inhibitors, which upregulated PDCD4 expression and concurrently exhibited efficacy in impeding mycobacterial proliferation within macrophages. These results not only reinforce the significance of PDCD4 as a pivotal marker in the understanding of infectious diseases, particularly mycobacterial infections, but also illuminate its potential in the identification of mTOR inhibitors, thereby contributing to the advancement of therapeutic strategies.
    OBJECTIVE: This study emphasizes the critical role of the mammalian target of rapamycin (mTOR) pathway in macrophage responses to mycobacterial infections, elucidating how mycobacteria activate mTOR, resulting in PDCD4 degradation. The utilization of the (Tor)-signal-indicator (TOSI) vector for real-time monitoring of mTOR activity represents a significant advancement in understanding mTOR regulation during mycobacterial infection. These findings deepen our comprehension of mycobacteria\'s innate immune mechanisms and introduce PDCD4 as a novel marker for mTOR activity in infectious diseases. Importantly, this research laid the groundwork for high-throughput screening of mTOR inhibitors using FDA-approved drugs, offering the potential for repurposing treatments against mycobacterial infections. The identification of drugs that inhibit mTOR activation opens new avenues for host-directed therapies, marking a significant step forward in combating tuberculosis and other mycobacterial diseases.
    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
    早期生活压力(ELS)会增加后期患抑郁症的风险。程序性细胞死亡因子4(PDCD4),凋亡相关分子,广泛参与肿瘤发生和炎症性疾病。然而,其参与一个人对ELS相关抑郁症的易感性是未知的。为了研究PDCD4对ELS易损性的影响和潜在机制,我们使用了“两次打击”应激小鼠模型:在出生后第7-9天(P7-P9)对新生小鼠进行腹膜内注射脂多糖(LPS),并在青少年中使用不可避免的足休克(IFS)作为后期挑战.我们的研究表明,与仅暴露于LPS或IFS的小鼠相比,“两次打击”应激小鼠出现了更严重的抑郁/焦虑样行为和社会残疾。我们检测了青春期小鼠海马中PDCD4的水平,发现它们在“两次打击”应激小鼠中显著增加。免疫组织化学染色和Sholl分析结果显示,"两击"应激小鼠海马区小胶质细胞数量明显增加,随着形态的变化,缩短的树枝,数量减少。然而,敲低PDCD4可以防止ELS诱导的小胶质细胞数量和形态变化。此外,我们通过高尔基体染色和免疫组织化学染色结果证实,敲低PDCD4可以改善ELS诱导的突触可塑性损伤。机械上,PDCD4的敲低发挥神经保护作用,可能通过BDNF/AKT/CREB信号的调解。合并,这些结果表明,PDCD4可能在ELS诱导的抑郁症易感性中起重要作用,因此,可能成为抑郁症的治疗目标。
    Early life stress (ELS) increases the risk of depression later in life. Programmed cell death factor 4 (PDCD4), an apoptosis-related molecule, extensively participates in tumorigenesis and inflammatory diseases. However, its involvement in a person\'s susceptibility to ELS-related depression is unknown. To examine the effects and underlying mechanisms of PDCD4 on ELS vulnerability, we used a \"two-hit\" stress mouse model: an intraperitoneal injection of lipopolysaccharide (LPS) into neonatal mice was performed on postnatal days 7-9 (P7-P9) and inescapable foot shock (IFS) administration in adolescent was used as a later-life challenge. Our study shows that compared with mice that were only exposed to the LPS or IFS, the \"two-hit\" stress mice developed more severe depression/anxiety-like behaviors and social disability. We detected the levels of PDCD4 in the hippocampus of adolescent mice and found that they were significantly increased in \"two-hit\" stress mice. The results of immunohistochemical staining and Sholl analysis showed that the number of microglia in the hippocampus of \"two-hit\" stress mice significantly increased, with morphological changes, shortened branches, and decreased numbers. However, knocking down PDCD4 can prevent the number and morphological changes of microglia induced by ELS. In addition, we confirmed through the Golgi staining and immunohistochemical staining results that knocking down PDCD4 can ameliorate ELS-induced synaptic plasticity damage. Mechanically, the knockdown of PDCD4 exerts neuroprotective effects, possibly via the mediation of BDNF/AKT/CREB signaling. Combined, these results suggest that PDCD4 may play an important role in the ELS-induced susceptibility to depression and, thus, may become a therapeutic target for depressive disorders.
    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
    STAT1(信号转导和转录激活因子1),属于STAT蛋白家族,对于细胞因子信号传导至关重要。据报道,它在不同的肿瘤中具有环境依赖性致癌或肿瘤抑制作用。早些时候,我们证明了多形性胶质母细胞瘤(GBMs)过度表达FAT1,一种非典型钙粘蛋白,结果较差。过表达的FAT1促进了肿瘤前炎症,通过下调抑癌基因的迁移/侵袭,PDCD4。这里,我们证明STAT1是FAT1下游的新介质,在GBM中下调PDCD4。GBM数据库的计算机分析以及切除的GBM肿瘤中的q-PCR分析显示STAT1和FAT1mRNA水平之间呈正相关。Kaplan-Meier分析显示具有高FAT1和STAT1表达的GBM患者的存活率较差。SiRNA介导的FAT1敲低降低胶质母细胞瘤细胞(LN229和U87MG)中STAT1和PDCD4表达增加。单独敲除STAT1导致PDCD4表达增加。PDCD4启动子的计算机模拟分析揭示了四个推定的STAT1结合位点(Site1-Site4)。ChIP测定证实了STAT1与位点1的结合。ChIP-PCR显示在FAT1敲低后,STAT1在PDCD4启动子上的结合减少。Site1的定点突变导致PDCD4荧光素酶活性增加,证实STAT1介导的PDCD4抑制。EMSA证实STAT1与位点1序列结合。STAT1敲低导致促炎细胞因子和EMT标志物的表达降低,和减少GBM细胞的迁移/侵袭。因此,这项研究将STAT1确定为FAT1的新型下游介质,促进GBM中的促肿瘤活性,通过抑制PDCD4表达。
    STAT1 (Signal Transducer and Activator of Transcription 1), belongs to the STAT protein family, essential for cytokine signaling. It has been reported to have either context dependent oncogenic or tumor suppressor roles in different tumors. Earlier, we demonstrated that Glioblastoma multiforme (GBMs) overexpressing FAT1, an atypical cadherin, had poorer outcomes. Overexpressed FAT1 promotes pro-tumorigenic inflammation, migration/invasion by downregulating tumor suppressor gene, PDCD4. Here, we demonstrate that STAT1 is a novel mediator downstream to FAT1, in downregulating PDCD4 in GBMs. In-silico analysis of GBM databases as well as q-PCR analysis in resected GBM tumors showed positive correlation between STAT1 and FAT1 mRNA levels. Kaplan-Meier analysis showed poorer survival of GBM patients having high FAT1 and STAT1 expression. SiRNA-mediated knockdown of FAT1 decreased STAT1 and increased PDCD4 expression in glioblastoma cells (LN229 and U87MG). Knockdown of STAT1 alone resulted in increased PDCD4 expression. In silico analysis of the PDCD4 promoter revealed four putative STAT1 binding sites (Site1-Site4). ChIP assay confirmed the binding of STAT1 to site1. ChIP-PCR revealed decrease in the binding of STAT1 on the PDCD4 promoter after FAT1 knockdown. Site directed mutagenesis of Site1 resulted in increased PDCD4 luciferase activity, substantiating STAT1 mediated PDCD4 inhibition. EMSA confirmed STAT1 binding to the Site 1 sequence. STAT1 knockdown led to decreased expression of pro-inflammatory cytokines and EMT markers, and reduced migration/invasion of GBM cells. This study therefore identifies STAT1 as a novel downstream mediator of FAT1, promoting pro-tumorigenic activity in GBM, by suppressing PDCD4 expression.
    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: Review
    The tumor suppressor programmed cell death 4 (PDCD4) is downregulated in various tumor tissues indicating poor prognosis. PDCD4 is the first protein found to resist tumor transformation, invasion, and metastasis by inhibiting translation. The functions of PDCD4 dependent on its structures are affected by extracellular signals. It regulates tumor-related proteins through a variety of mechanisms, especially involved in two major signaling pathways, PI3K-Akt-mTOR and MAPK. By analyzing the relationship between the structures, functions and diseases of PDCD4, this review summarizes the roles of PDCD4 in several physiological processes and diseases such as apoptosis, autophagy, tumor, and inflammation in recent years, thereby providing insights for the study of the signaling pathways of PDCD4 and related proteins and the treatment of diseases targeting them.
    程序性细胞死亡蛋白PDCD4(programmed cell death 4)是一种肿瘤抑制蛋白,在多种肿瘤组织中下调并提示不良预后,是第一种被发现通过抑制翻译抵抗肿瘤转化、侵袭和转移的蛋白质。PDCD4自身结构与功能关系密切,并受细胞外信号影响,其蛋白表达水平和功能与人体两大信号通路PI3K-Akt-mTOR和MAPK密切相关,通过多种机制调节与肿瘤相关的其他蛋白质。本文通过解析PDCD4结构、功能与疾病的关系,总结了近年来PDCD4在凋亡、自噬、肿瘤、炎症等生理过程和疾病中的作用,为PDCD4及相关蛋白的信号传导路径研究和以它们为靶标的疾病治疗提供启发和思路。.
    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
    本研究旨在探讨PDCD4通过调节FGR/NF-κB信号通路对直肠癌放疗后急性肾损伤(AKI)的分子机制。使用基因表达综合(GEO)数据集(用于直肠癌的GSE90627和用于AKI的GSE145085)和R软件鉴定差异表达的基因。人肾小管上皮细胞系,HK-2用于建立放疗诱导的AKI的体外模型。RT-qPCR和Westernblotting检测基因和蛋白表达水平,分别。使用CCK-8测定和流式细胞术评估细胞增殖和凋亡,分别。测定细胞培养上清液中的丙二醛和超氧化物歧化酶水平。此外,使用BALB/c小鼠建立体内AKI模型,和肾脏组织形态学,肾损伤分子KIM-1的表达,肾小管细胞凋亡,并对血清中的TAS和TOS进行评价。生物信息学分析显示PDCD4在AKI中的表达上调。体外实验证明PDCD4通过促进FGR表达诱导肾小管细胞凋亡,激活NF-κB信号通路并引发氧化应激反应。体内动物实验证实PDCD4通过激活FGR/NF-κB信号通路促进氧化应激反应和放疗诱导的AKI。沉默PDCD4减弱放疗诱导的AKI。我们的研究结果表明,PDCD4可能通过促进FGR表达诱导直肠癌放疗诱导的AKI,激活NF-κB信号通路,并引发氧化应激反应。
    This study aimed to investigate the molecular mechanism of the effect of PDCD4 on radiotherapy-induced acute kidney injury (AKI) in rectal cancer through the regulation of FGR/NF-κB signaling. Differentially expressed genes were identified using Gene Expression Omnibus (GEO) datasets (GSE90627 for rectal cancer and GSE145085 for AKI) and R software. The human renal tubular epithelial cell line, HK-2, was used to establish an in vitro model of radiotherapy-induced AKI. RT-qPCR and western blotting were used to detect gene and protein expression levels, respectively. Cell proliferation and apoptosis were assessed using the CCK-8 assay and flow cytometry, respectively. The malondialdehyde and superoxide dismutase levels in the cell culture supernatants were determined. Additionally, an in vivo AKI model was established using BALB/c mice, and kidney tissue morphology, expression of the renal injury molecule KIM-1, apoptosis of renal tubular cells, and TAS and TOS in serum were evaluated. Bioinformatics analysis revealed the upregulated expression of PDCD4 in AKI. In vitro experiments demonstrated that PDCD4 induced apoptosis in renal tubular cells by promoting FGR expression, which activated the NF-κB signaling pathway and triggered an oxidative stress response. In vivo animal experiments confirmed that PDCD4 promoted oxidative stress response and radiotherapy-induced AKI through the activation of the FGR/NF-κB signaling pathway. Silencing PDCD4 attenuated radiotherapy-induced AKI. Our findings suggest that PDCD4 may induce radiotherapy-induced AKI in rectal cancer by promoting FGR expression, activating the NF-κB signaling pathway, and triggering an oxidative stress response.
    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
    作为专性寄生虫,病毒已经进化出多种策略来逃避宿主的免疫防御。操纵宿主蛋白酶体系统以降解特定有害因素是常见的病毒对策。鉴定猪繁殖与呼吸综合征病毒(PRRSV)降解蛋白酶体的宿主蛋白,我们对蛋白酶体抑制剂MG132处理的PRRSV感染的Marc-145细胞进行了定量蛋白质组学筛选.数据显示,程序性细胞死亡4(PDCD4)的表达水平被PRRSV强烈下调,并被MG132显著挽救。进一步的研究证实,PRRSV感染诱导PDCD4从细胞核到细胞质的易位,病毒非结构蛋白9(Nsp9)通过激活Akt-mTOR-S6K1通路促进了PDCD4蛋白酶体在细胞质中的降解。Nsp9的C末端结构域负责PDCD4降解。至于PDCD4在PRRSV感染中的作用,我们证明了PDCD4敲低有利于病毒复制,虽然它的过表达显著减弱了复制,提示PDCD4是PRRSV的限制因子。机械上,我们发现PRRSV需要真核翻译起始因子4A(eIF4A)。PDCD4通过其两个MA3域内的四个位点(E249、D253、D414和D418)与eIF4A相互作用,在PRRSV的5'-非翻译区中破坏eIF4A介导的翻译起始,从而抑制PRRSV感染。一起,我们的研究揭示了PDCD4的抗病毒功能和拮抗PDCD4的病毒策略.这些结果将有助于我们对PRRSV采用的免疫逃避策略的理解,并为开发新的抗病毒靶标提供有价值的见解。猪繁殖与呼吸综合征病毒(PRRSV)感染给全球养猪业造成重大经济损失,难以有效控制。这里,使用定量蛋白质组学筛选,我们鉴定了程序性细胞死亡4(PDCD4)作为PRRSV蛋白酶体降解的宿主蛋白.我们证明了PDCD4通过与真核翻译起始因子4A相互作用来限制PRRSV的复制,这是病毒5'非翻译区中翻译起始所必需的。此外,PDCD4的两个MA3结构域内的四个位点被鉴定为负责其抗病毒功能。相反,PRRSV非结构蛋白9通过激活Akt-mTOR-S6K1通路促进PDCD4蛋白酶体在细胞质中的降解,从而削弱了抗PRRSV的功能。我们的工作揭示了PDCD4是PRRSV以前未被识别的宿主限制因子,并揭示了PRRSV开发了克服PDCD4的对策。这将为病毒-宿主相互作用和新的抗病毒靶标的开发提供新的见解。
    As obligate parasites, viruses have evolved multiple strategies to evade the host immune defense. Manipulation of the host proteasome system to degrade specific detrimental factors is a common viral countermeasure. To identify host proteins targeted for proteasomal degradation by porcine reproductive and respiratory syndrome virus (PRRSV), we conducted a quantitative proteomics screen of PRRSV-infected Marc-145 cells under the treatment with proteasome inhibitor MG132. The data revealed that the expression levels of programmed cell death 4 (PDCD4) were strongly downregulated by PRRSV and significantly rescued by MG132. Further investigation confirmed that PRRSV infection induced the translocation of PDCD4 from the nucleus to the cytoplasm, and the viral nonstructural protein 9 (Nsp9) promoted PDCD4 proteasomal degradation in the cytoplasm by activating the Akt-mTOR-S6K1 pathway. The C-terminal domain of Nsp9 was responsible for PDCD4 degradation. As for the role of PDCD4 during PRRSV infection, we demonstrated that PDCD4 knockdown favored viral replication, while its overexpression significantly attenuated replication, suggesting that PDCD4 acts as a restriction factor for PRRSV. Mechanistically, we discovered eukaryotic translation initiation factor 4A (eIF4A) was required for PRRSV. PDCD4 interacted with eIF4A through four sites (E249, D253, D414, and D418) within its two MA3 domains, disrupting eIF4A-mediated translation initiation in the 5\'-untranslated region of PRRSV, thereby inhibiting PRRSV infection. Together, our study reveals the antiviral function of PDCD4 and the viral strategy to antagonize PDCD4. These results will contribute to our understanding of the immune evasion strategies employed by PRRSV and offer valuable insights for developing new antiviral targets.IMPORTANCEPorcine reproductive and respiratory syndrome virus (PRRSV) infection results in major economic losses in the global swine industry and is difficult to control effectively. Here, using a quantitative proteomics screen, we identified programmed cell death 4 (PDCD4) as a host protein targeted for proteasomal degradation by PRRSV. We demonstrated that PDCD4 restricts PRRSV replication by interacting with eukaryotic translation initiation factor 4A, which is required for translation initiation in the viral 5\'-untranslated region. Additionally, four sites within two MA3 domains of PDCD4 are identified to be responsible for its antiviral function. Conversely, PRRSV nonstructural protein 9 promotes PDCD4 proteasomal degradation in the cytoplasm by activating the Akt-mTOR-S6K1 pathway, thus weakening the anti-PRRSV function. Our work unveils PDCD4 as a previously unrecognized host restriction factor for PRRSV and reveals that PRRSV develops countermeasures to overcome PDCD4. This will provide new insights into virus-host interactions and the development of new antiviral 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
    目的:冬凌草乙酸乙酯提取物(COE)是中草药冬凌草茎的主要提取物,具有抗肿瘤和抗炎的生物学作用。我们之前的研究表明,COE对大鼠胃癌前病变(PLGC)有一定的逆转作用,但是确切的作用机制仍然难以捉摸。我们旨在探讨COE对PLGC的治疗作用及其潜在机制。
    方法:通过N-甲基-N'-硝基-N-亚硝基胍(MNNG)多因素诱导法成功构建了PLGC大鼠模型。然后,制备COE以治疗PLGC大鼠模型。苏木精伊红染色观察大鼠胃粘膜病变,采用AB-PAS和HID-AB染色观察肠上皮化生。通过免疫组织化学(IHC)和逆转录聚合酶链反应(RT-PCR)检测体内PDCD4-ATG5信号通路,通过IHC检测自噬水平,透射电子显微镜,和体内RT-PCR。此外,用MNNG处理GES-1细胞成功构建PLGC(MC)细胞模型。然后,形态学,扩散,和MC细胞的凋亡,通过COE和过表达PDCD4治疗后,评估了PDCD4-ATG5信号通路和自噬在MC细胞中的作用。
    结果:COE明显改善大鼠胃粘膜损伤和细胞异形,延缓PLGC的进展。进一步的研究表明,COE不仅抑制了自噬水平,而且干扰了体内PDCD4-ATG5信号通路。另一方面,COE治疗能有效逆转MC细胞损伤,抑制MC细胞增殖,促进MC细胞凋亡。此外,COE还在体外促进PDCD4和抑制ATG5表达,PDCD4过表达后,COE对ATG5介导的自噬的抑制作用进一步增强。
    结论:研究表明,COE可以通过调节PDCD4-ATG5信号通路抑制胃上皮细胞的自噬,这有助于逆转PLGC的进展。
    OBJECTIVE: Celastrus orbiculatus ethyl acetate extract (COE) is the main extract of the stem of the Chinese herbal C. orbiculatus, which has anti-tumor and anti-inflammatory biological effects. Our previous study showed that COE had a certain reversal effect on the precancerous lesions of gastric cancer (PLGC) in rats, but the exact mechanism of action remains elusive. We aimed to explore the therapeutic effects of COE on PLGC and the potential mechanisms.
    METHODS: The PLGC rat model was successfully constructed by N-methyl-N´-nitro-N-nitrosoguanidine (MNNG) multifactorial induction method. Then, COE was prepared to treat the PLGC rat model. Hematoxylin & eosin staining was used to observe gastric mucosal lesions in rats, AB-PAS and HID-AB staining were used to observe intestinal metaplasia. PDCD4-ATG5 signaling pathway was detected by immunohistochemistry (IHC) and reverse transcription polymerase chain reaction (RT-PCR) in vivo, and autophagy level was detected by IHC, transmission electron microscopy, and RT-PCR in vivo. Besides, the PLGC (MC) cell model was successfully constructed by treating GES-1 cells with MNNG. Then, the morphology, proliferation, and apoptosis of MC cells, and the role of the PDCD4-ATG5 signaling pathway and autophagy in MC cells were evaluated by COE and after the overexpression of PDCD4 treatment.
    RESULTS: COE significantly improved gastric mucosal injury and cellular heteromorphism and retarded the progression of PLGC in rats. Further studies indicated COE not only inhibited the level of autophagy but also interfered with the PDCD4-ATG5 signaling pathway in vivo. On the other hand, COE treatment could effectively reverse MC cell damage, inhibit MC cell proliferation, and promote MC cell apoptosis. Furthermore, COE also promoted PDCD4 and inhibited ATG5 expression in vitro, and the inhibitory effect of COE on ATG5-mediated autophagy was further enhanced after the overexpression of PDCD4.
    CONCLUSIONS: The study revealed that COE could regulate the PDCD4-ATG5 signaling pathway to inhibit autophagy in gastric epithelial cells, which contributes to reversing the progression of PLGC.
    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: English Abstract
    目的:阐明程序性细胞死亡因子4(PDCD4)在脓毒症相关血管内皮损伤所致线粒体功能障碍中的作用。
    方法:用靶向PDCD4的小干扰RNA转染培养的人脐静脉内皮细胞(HUVEC)和小鼠血管内皮细胞(C166细胞),然后单独用脂多糖(LPS)或与羰基氰化物3-氯苯腙(FCCP)组合处理。使用LC-MS/MS技术分析PDCD4敲低后细胞中的蛋白质组变化。RT-PCR检测PDCD4mRNA及与细胞炎症和凋亡相关基因的表达,使用Western印迹法确定线粒体分裂和融合关键的FIS1,DRP1和OPA1蛋白的表达。采用JC-1和MitoSOX荧光探针在激光共聚焦显微镜下观察线粒体膜电位和线粒体活性氧水平的变化。
    结果:LPS刺激细胞显著增加白细胞介素-6(IL-6)的mRNA表达,肿瘤坏死因子-α(TNF-α)和单核细胞趋化蛋白1(MCP1)均能增强PDCD4的表达(P<0.05)。蛋白质组学分析表明PDCD4敲低与细胞中线粒体动力学变化之间存在相关性。LPS处理显著增加细胞线粒体分裂蛋白FIS1和DRP1的表达,降低融合蛋白OPA1的表达(P<0.05),还引起线粒体氧化应激和线粒体膜电位降低(P<0.05)。在HUVEC中,用FCCP治疗可显着减弱PDCD4敲低的保护作用,抑制LPS诱导的炎症反应和氧化应激,恢复线粒体分裂和融合的平衡。
    结论:PDCD4敲低通过抑制线粒体裂变和氧化应激保护血管内皮细胞免受LPS诱导的损伤,促进线粒体融合,维持正常的线粒体功能.
    OBJECTIVE: To elucidate the role of programmed cell death factor 4 (PDCD4) in mitochondrial dysfunction caused by sepsis-related vascular endothelial damage.
    METHODS: Cultured human umbilical vein endothelial cells (HUVECs) and mouse vascular endothelial cells (C166 cells) were transfected with a small interfering RNA targeting PDCD4 followed by treatment with lipopolysaccharide (LPS) alone or in combination with carbonyl cyanide 3-chlorophenylhydrazone (FCCP). The proteomic changes in the cells after PDCD4 knockdown were analyzed using LC-MS/MS technique. The mRNA expressions of PDCD4 and the genes associated with cell inflammation and apoptosis were detected with RT-PCR, and the expressions of FIS1, DRP1 and OPA1 proteins key to mitochondrial fission and fusion were determined using Western blotting. JC-1 and MitoSOX fluorescent probes were used to observe the changes in mitochondrial membrane potential and mitochondrial reactive oxygen species levels under by a laser confocal microscope.
    RESULTS: LPS stimulation of the cells significantly increased the mRNA expressions of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein 1 (MCP1) and enhanced the cellular expression of PDCD4 (P < 0.05). Proteomic analysis suggested a correlation between PDCD4 knockdown and changes in mitochondrial dynamics in the cells. LPS treatment significantly increased the expressions of mitochondrial fission proteins FIS1 and DRP1 and lowered the expression of the fusion protein OPA1 in the cells (P < 0.05), causing also mitochondrial oxidative stress and reduction of the mitochondrial membrane potential (P < 0.05). In HUVECs, treatment with FCCP significantly attenuated the protective effect of PDCD4 knockdown, which inhibited LPS-induced inflammation and oxidative stress and restored the balance between mitochondrial fission and fusion.
    CONCLUSIONS: PDCD4 knockdown protects vascular endothelial cells against LPS-induced damages by repressing mitochondrial fission and oxidative stress, promoting mitochondrial fusion, and maintaining normal mitochondrial function.
    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
    探讨微小RNA(miR)-181b-5p通过靶向程序性细胞死亡因子4(PDCD4)促进甲状腺癌(TC)进展的潜在机制。对miR-181b-5p和PDCD4在TC中的表达进行分析。miR-181b-5p和PDCD4对细胞增殖的影响,迁移,入侵,并检查了TC细胞的凋亡。预测并验证miR-181b-5p与PDCD4的结合关系。miR-181b-5p在TC中上调,而PDCD4下调。下调miR-181b-5p或上调PDCD4抑制增殖,迁移,和入侵TC细胞,促进细胞凋亡。PDCD4是miR-181b-5p的下游靶标,PDCD4的下调抵消了miR-181b-5p下调对细胞增殖的抑制作用,迁移,以及对TC细胞的侵袭和对凋亡的促进作用。miR-181b-5p抑制增殖,迁移,并通过靶向PDCD4促进TC细胞的侵袭和细胞凋亡。
    To explore the potential mechanism of microRNA (miR)-181b-5p promoting the progression of thyroid cancer (TC) by targeting programmed cell death 4 (PDCD4). Analysis of miR-181b-5p and PDCD4 expression in TC was performed. The impact of miR-181b-5p and PDCD4 on proliferation, migration, invasion, and apoptosis of TC cells was examined. The binding relationship between miR-181b-5p and PDCD4 was predicted and verified. miR-181b-5p was up-regulated in TC, while PDCD4 was down-regulated. Down-regulating miR-181b-5p or up-regulating PDCD4 inhibited the proliferation, migration, and invasion of TC cells, and promoted cell apoptosis. PDCD4 was the downstream target of miR-181b-5p, and down-regulation of PDCD4 counteracted the inhibitory effect of down-regulation of miR-181b-5p on the proliferation, migration, and invasion of TC cells and the promoting effect on apoptosis. miR-181b-5p inhibits the proliferation, migration, and invasion of TC cells and promotes cell apoptosis by targeting PDCD4.
    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