lytic reactivation

  • 文章类型: Journal Article
    联合抗逆转录病毒疗法(cART)抑制获得性免疫缺陷综合征(AIDS)患者的人类免疫缺陷病毒1型(HIV-1)病毒复制和发病机理。然而,HIV-1通过抑制病毒转录保持在感染的潜伏阶段,这阻碍了HIV-1的治愈。治愈HIV-1的一种方法是“休克和杀死”策略。该策略侧重于重新激活潜伏的HIV-1,诱导病毒细胞病变作用并促进免疫清除以消除潜伏的HIV-1储库。这里,我们报道,H3K4三甲基化(H3K4me3)特异性去甲基酶KDM5A/B在抑制HIV-1潜伏细胞中HIV-1Tat/LTR介导的病毒转录中起作用.此外,我们评估了KDM5特异性抑制剂JQKD82作为HIV-1“休克和杀伤”药物的潜力.我们的结果表明,JQKD82增加了HIV-15'LTR启动子区的H3K4me3水平,HIV-1再激活,以及HIV-1潜伏T细胞模型中的细胞病变效应。此外,我们确定了JQKD82和AZD5582的组合,一种非常规的NF-κB激活剂,对诱导T细胞中的HIV-1裂解剂再激活和细胞死亡产生协同影响。JQKD82和AZD5582对的潜伏期逆转效力也在从HIV-1病毒血症患者分离的外周血单核细胞(PBMC)和HIV-1潜伏单核细胞中得到证实。在大脑潜伏感染的小胶质细胞(HC69)中,KDM5A/B的缺失或抑制导致HIV-1潜伏期的逆转。总的来说,我们得出的结论是,KDM5A/B充当HIV-1裂解剂再激活的宿主抑制因子,从而促进了HIV-1感染水库的潜伏期和存活。
    Combinational antiretroviral therapy (cART) suppresses human immunodeficiency virus type 1 (HIV-1) viral replication and pathogenesis in acquired immunodeficiency syndrome (AIDS) patients. However, HIV-1 remains in the latent stage of infection by suppressing viral transcription, which hinders an HIV-1 cure. One approach for an HIV-1 cure is the \"shock and kill\" strategy. The strategy focuses on reactivating latent HIV-1, inducing the viral cytopathic effect and facilitating the immune clearance for the elimination of latent HIV-1 reservoirs. Here, we reported that the H3K4 trimethylation (H3K4me3)-specific demethylase KDM5A/B play a role in suppressing HIV-1 Tat/LTR-mediated viral transcription in HIV-1 latent cells. Furthermore, we evaluated the potential of KDM5-specific inhibitor JQKD82 as an HIV-1 \"shock and kill\" agent. Our results showed that JQKD82 increases the H3K4me3 level at HIV-1 5\' LTR promoter regions, HIV-1 reactivation, and the cytopathic effects in an HIV-1-latent T cell model. In addition, we identified that the combination of JQKD82 and AZD5582, a non-canonical NF-κB activator, generates a synergistic impact on inducing HIV-1 lytic reactivation and cell death in the T cell. The latency-reversing potency of the JQKD82 and AZD5582 pair was also confirmed in peripheral blood mononuclear cells (PBMCs) isolated from HIV-1 aviremic patients and in an HIV-1 latent monocyte. In latently infected microglia (HC69) of the brain, either deletion or inhibition of KDM5A/B results in a reversal of the HIV-1 latency. Overall, we concluded that KDM5A/B function as a host repressor of the HIV-1 lytic reactivation and thus promote the latency and the survival of HIV-1 infected reservoirs.
    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
    病毒感染导致从难治到流产和完全生产状态的异质性细胞结果。单细胞转录组学能够实现这些不同的感染后状态的高分辨率视图。这里,我们已经询问了EB病毒(EBV)重新激活后的宿主病原体动力学。虽然大多数人是良性的,EBV是导致传染性单核细胞增多症的原因,高达2%的人类癌症,是多发性硬化症发展的诱因。在B细胞中建立潜伏期后,EBV重新激活并在唾液中脱落,从而能够感染新宿主。除了它对传播的重要性,裂解周期也与EBV相关的肿瘤发生有关。相反,在潜伏的EBV阳性肿瘤中诱导裂解再激活提供了新的治疗机会。因此,确定EBV裂解剂再激活的动力学和异质性是更好地理解发病机制和治疗潜力的优先事项.在这项研究中,我们应用单细胞技术分析了两种B细胞模型中裂解液再激活过程中不同的命运轨迹.与以前的工作一致,我们发现细胞周期和MYC表达与细胞难以裂解再激活相关。我们进一步发现,裂解诱导产生从流产到完全再活化的连续过程。流产裂解细胞上调NFκB和IRF3通路靶基因,而经过完整裂解周期的细胞表现出与细胞重编程相关的基因的意外表达。裂解细胞的不同亚群进一步显示已知逃避病毒介导的宿主关闭的转录本的可变概况。这些数据揭示了先前未知和混杂的裂解再激活结果,对病毒复制和EBV相关的肿瘤发生具有广泛的影响。
    病毒感染以增强疾病的方式深刻地改变宿主细胞的生物程序。EB病毒(EBV)是一种与多种癌症和几种自身免疫性疾病相关的特别流行的人类病原体。EBV主要在B细胞中建立潜伏感染,并可以通过充分表征的潜伏癌蛋白的功能促进B细胞恶性肿瘤。病毒裂解周期的各个方面也明显导致EBV相关疾病,虽然裂解剂再激活的病理作用尚不完全清楚。在这里,我们使用单细胞技术来检查多个B细胞模型中对EBV裂解剂再激活的细胞反应。与先前的研究一致,延迟的再激活在某些细胞中是不完整的(失败的),在其他细胞中是成功的。流产和完全裂解轨迹表现出不同的生物学反应,每种反应都可能促进发病机理并增强双峰潜伏裂解控制。有趣的是,细胞上通过裂解周期进行的部分表现出与细胞重编程相关的基因的意外和惊人的表达,多能性,和自我更新。总的来说,这项研究为了解病毒再激活过程中不同的宿主病毒动力学和命运提供了宝贵的资源,并确定了多种模式的EBV裂解发病机制,以便在未来的研究中进行研究。
    Viral infection leads to heterogeneous cellular outcomes ranging from refractory to abortive and fully productive states. Single cell transcriptomics enables a high resolution view of these distinct post-infection states. Here, we have interrogated the host-pathogen dynamics following reactivation of Epstein-Barr virus (EBV). While benign in most people, EBV is responsible for infectious mononucleosis, up to 2% of human cancers, and is a trigger for the development of multiple sclerosis. Following latency establishment in B cells, EBV reactivates and is shed in saliva to enable infection of new hosts. Beyond its importance for transmission, the lytic cycle is also implicated in EBV-associated oncogenesis. Conversely, induction of lytic reactivation in latent EBV-positive tumors presents a novel therapeutic opportunity. Therefore, defining the dynamics and heterogeneity of EBV lytic reactivation is a high priority to better understand pathogenesis and therapeutic potential. In this study, we applied single-cell techniques to analyze diverse fate trajectories during lytic reactivation in two B cell models. Consistent with prior work, we find that cell cycle and MYC expression correlate with cells refractory to lytic reactivation. We further found that lytic induction yields a continuum from abortive to complete reactivation. Abortive lytic cells upregulate NFκB and IRF3 pathway target genes, while cells that proceed through the full lytic cycle exhibit unexpected expression of genes associated with cellular reprogramming. Distinct subpopulations of lytic cells further displayed variable profiles for transcripts known to escape virus-mediated host shutoff. These data reveal previously unknown and promiscuous outcomes of lytic reactivation with broad implications for viral replication and EBV-associated oncogenesis.
    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
    本研究旨在探讨Yes相关蛋白1(YAP1)在EB病毒相关胃癌(EBVaGC)发生发展中的作用及调控机制。在这里,我们发现EBV可以通过其编码的潜伏产物EBV编码的小RNA1(EBER1)和潜伏膜蛋白2A(LMP2A)上调YAP1蛋白的表达和活性,增强EBVaGC细胞的恶性特征。此外,我们还表明,YAP1的过表达诱导EBV编码潜伏和裂解期基因和蛋白质在感染EBV的上皮细胞系AGS-EBV中的表达,增加了EBV基因组的拷贝数,而YAP1表达的缺失降低了上述指标。此外,我们发现YAP1增强了两种已知激活剂诱导的EBV裂解再激活,12-O-十四烷酰基骨醇-13-乙酸酯(TPA)和丁酸钠(NaB)。这些结果表明EBV和YAP1蛋白之间的双向调节机制,为进一步了解胃癌中EBV感染模式和致癌机制的调控提供了新的实验证据。
    This study aims to explore the role and regulatory mechanism of Yes-associated protein 1 (YAP1) in the development of Epstein-Barr virus-associated gastric cancer (EBVaGC). Here we showed that EBV can upregulate the expression and activity of YAP1 protein through its encoded latent products EBV-encoded small RNA 1 (EBER1) and latent membrane protein 2A (LMP2A), enhancing the malignant characteristics of EBVaGC cells. In addition, we also showed that overexpression of YAP1 induced the expression of EBV encoding latent and lytic phase genes and proteins in the epithelial cell line AGS-EBV infected with EBV, and increased the copy number of the EBV genome, while loss of YAP1 expression reduced the aforementioned indicators. Moreover, we found that YAP1 enhanced EBV lytic reactivation induced by two known activators, 12-O-tetradecanoylhorbol-13-acetate (TPA) and sodium butyrate (NaB). These results indicated a bidirectional regulatory mechanism between EBV and YAP1 proteins, providing new experimental evidence for further understanding the regulation of EBV infection patterns and carcinogenic mechanisms in gastric cancer.
    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:缺氧可诱导卡波西肉瘤相关病毒(KSHV)的再激活,这就需要合成关键的结构蛋白。尽管缺氧的不利能量条件,KSHV利用机制来防止成功重新激活所需的基本细胞机器的降解。我们的研究为KSHV感染的细胞通过克服缺氧介导的代谢应激以成功激活来维持稳态转录的策略提供了新的见解。我们发现潜伏期相关的核抗原与HIF1α和NEDD4的相互作用抑制其聚泛素化活性,在缺氧期间阻断RNAPolII的降解,对我们对KSHV生物学的理解做出了重大贡献。这项新发现的知识为开发KSHV相关疾病的新疗法提供了新的线索。
    OBJECTIVE: Hypoxia can induce the reactivation of Kaposi sarcoma-associated virus (KSHV), which necessitates the synthesis of critical structural proteins. Despite the unfavorable energetic conditions of hypoxia, KSHV utilizes mechanisms to prevent the degradation of essential cellular machinery required for successful reactivation. Our study provides new insights on strategies employed by KSHV-infected cells to maintain steady-state transcription by overcoming hypoxia-mediated metabolic stress to enable successful reactivation. Our discovery that the interaction of latency-associated nuclear antigen with HIF1α and NEDD4 inhibits its polyubiquitination activity, which blocks the degradation of RNA Pol II during hypoxia, is a significant contribution to our understanding of KSHV biology. This newfound knowledge provides new leads in the development of novel therapies for KSHV-associated 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
    卡波西肉瘤相关疱疹病毒(KSHV)是一种致癌DNA病毒,其裂解复制周期由病毒转录因子RTA(复制和转录激活因子)诱导和驱动。确定在裂解再激活期间RTA与哪些细胞和病毒蛋白相互作用可以帮助更好地理解RTA如何促进裂解周期。使用蛋白质组学方法,我们确定了细胞E3泛素连接酶复合物RNF20/40是RTA的新型相互作用伴侣。该蛋白质复合物的主要作用之一是组蛋白H2B在赖氨酸120上对细胞染色质的单单双酰化,这是转录活性基因的标志。虽然已经在不同的宿主基因组上研究了RNF20/40的作用,其调节病毒基因的潜力在很大程度上仍未被开发。这里,我们表明,shRNA抑制RNF20表达损害RTA诱导的宿主和病毒裂解基因表达,病毒DNA复制,和病毒生产。此外,我们发现RNF20与KSHV附加体的关联在裂解再激活过程中增加,并且RTA与病毒启动子的结合对于通过RTA和RNF20/40的协同启动子激活是必需的。有趣的是,在KSHV裂解再激活过程中抑制RNF20表达并不影响H2BK120ub水平,但确实降低了RNA聚合酶II在病毒启动子上的占有率.总的来说,我们的数据表明,RTA劫持了细胞E3连接酶复合物RNF20/40,以增加病毒基因启动子上转录活性RNA聚合酶II的水平,从而促进裂解基因表达,从而推进病毒裂解周期.IMPORTANCEKaposi肉瘤相关疱疹病毒(KSHV)是一种致癌的人类疱疹病毒,可在人类中持续感染。裂解病毒周期在终生感染中起着至关重要的作用,因为它涉及病毒传播。KSHV裂解复制周期的主要调节因子是病毒复制和转录激活因子(RTA)蛋白,这是必要的,足以推动病毒从潜伏期进入裂解期。因此,确定RTA用于控制裂解周期的宿主因子有助于找到可用于开发针对KSHV的抗病毒疗法的新靶标。使用蛋白质组学方法,我们已经确定了RTA与细胞E3泛素连接酶复合物RNF20/40之间的新相互作用,我们已经证明这对于促进RTA诱导的KSHV裂解周期是必需的。
    OBJECTIVE: Kaposi\'s sarcoma-associated herpesvirus (KSHV) is a cancer-causing human herpesvirus that establishes a persistent infection in humans. The lytic viral cycle plays a crucial part in lifelong infection as it is involved in the viral dissemination. The master regulator of the KSHV lytic replication cycle is the viral replication and transcription activator (RTA) protein, which is necessary and sufficient to push the virus from latency into the lytic phase. Thus, the identification of host factors utilized by RTA for controlling the lytic cycle can help to find novel targets that could be used for the development of antiviral therapies against KSHV. Using a proteomics approach, we have identified a novel interaction between RTA and the cellular E3 ubiquitin ligase complex RNF20/40, which we have shown to be necessary for promoting RTA-induced KSHV lytic cycle.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    爱泼斯坦巴尔是人类第一个致癌病毒,与许多淋巴增生和恶性疾病密切相关,包括HL,BL,NPC,和GC。EBV建立了描绘双相病毒生命周期的终身持续性感染:潜伏期和裂解复制。B细胞是EBV潜伏基因的关键区域,其中病毒基因表达被抑制,促进病毒基因组维持和免疫识别逃避。裂解液重新激活后,病毒基因表达诱导其复制,后代生产,和传输。TSG表达中表观遗传调控的失调导致癌变。一些研究表明,EBV与异常的病毒DNA和宿主基因组甲基化模式有关。促进免疫监测,识别规避和宿主细胞持久性。在其他表观遗传修饰中,DNA甲基化抑制大多数病毒潜伏基因启动子,保留一些,并作为激活EBV裂解周期的先决条件,产生病毒后代。它通过将细胞重编程为致癌基因来影响宿主的表观基因组,持久的表型,在一些恶性肿瘤中很明显。在其生命周期的每个阶段,EBV利用表观遗传调控的细胞机制,暗示其独特的宿主-病原体关系。本文综述了DNA甲基化在几个EBV相关启动子区的调控作用。以及病理条件下的宿主基因组,突出涉及潜伏的病毒基因,EBV感染的裂解期和潜伏裂解期。此外,它提供了对EBV中基于甲基化的途径的图解见解。
    Epstein Barr is the first-in-human oncogenic virus, closely related to numerous lymphoproliferative and malignant diseases, including HL, BL, NPC, and GC. EBV establishes life-long persistence infection portraying a biphasic viral life cycle: latent period and lytic replication. B-cells serve as critical regions for EBV latent genes, wherein viral gene expression is suppressed, promoting viral genome maintenance and immune recognition evasion. Upon its lytic reactivation, viral gene expression induces its replication, progeny production, and transmission. Dysregulations of epigenetic regulation in expressions of TSGs lead to carcinogenesis. Several studies reveal that EBV is associated with aberrant viral DNA and host genome methylation patterns, promoting immune monitoring, recognition evasiveness and host cell persistence. Among other epigenetic modifications, DNA methylation suppresses the majority of viral latent gene promoters, sparing a few, and acts as a prerequisite for activating EBV\'s lytic cycle, giving rise to viral progeny. It affects the host\'s epigenome via reprogramming cells to oncogenic, long-lasting phenotypes, as evident in several malignancies. At each phase of its life cycle, EBV exploits cellular mechanisms of epigenetic regulation, implying its unique host-pathogen relationship. This review summarized the DNA methylation\'s regulatory roles on several EBV-related promoter regions, along with the host genome in pathological conditions, highlights viral genes involved in a latent, lytic and latent-lytic phase of EBV infection. Moreover, it provides diagrammatic insights into methylation-based pathways in EBV.
    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
    EB病毒(EBV)是一种广泛存在的人类疱疹病毒,与淋巴瘤和上皮细胞癌有关。它建立了两个独立的感染阶段,潜伏和裂解,在主机。感染新的宿主细胞后,病毒激活了几种途径,以诱导裂解性EBV抗原的表达和感染性病毒颗粒的产生。尽管潜伏EBV感染的致癌作用已经确立,最近的研究表明,裂解剂的再激活在致癌作用中也起着重要作用。在这次审查中,我们总结了EBV再激活的机制以及关于病毒溶解抗原在肿瘤形成中作用的最新发现。此外,我们讨论了用溶解激活剂治疗EBV相关肿瘤的方法以及将来可能具有治疗效果的靶点.
    Epstein-Barr virus (EBV) is a widespread human herpes virus associated with lymphomas and epithelial cell cancers. It establishes two separate infection phases, latent and lytic, in the host. Upon infection of a new host cell, the virus activates several pathways, to induce the expression of lytic EBV antigens and the production of infectious virus particles. Although the carcinogenic role of latent EBV infection has been established, recent research suggests that lytic reactivation also plays a significant role in carcinogenesis. In this review, we summarize the mechanism of EBV reactivation and recent findings about the role of viral lytic antigens in tumor formation. In addition, we discuss the treatment of EBV-associated tumors with lytic activators and the targets that may be therapeutically effective in the future.
    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:爱泼斯坦巴尔病毒(EBV)的再激活导致病毒和细胞表位转录组的调节。N6-甲基腺苷(m6A)修饰是一种调节mRNA代谢的RNA修饰。先前的报道表明m6A修饰影响EBV编码的mRNA的稳定性和代谢。然而,再激活对细胞mRNA重编程的影响,这如何有助于成功诱导裂解剂再激活尚不清楚。
    方法:甲基化RNA免疫沉淀测序(MeRIP-seq),转录组RNA测序(RNA-seq)和RNA下拉PCR用于筛选和验证差异甲基化的靶标。西方印迹,采用实时定量PCR(RT-qPCR)和免疫细胞化学方法检测不同蛋白的表达和定位。RNA稳定性和多聚体分析测定用于检测下游基因的半衰期和翻译效率。使用插入点突变以破坏m6A甲基化位点来验证m6A甲基化对其稳定性和表达水平的影响。
    结果:我们报告说,在EBV再激活过程中,m6A橡皮擦ALKBH5显着下调,导致细胞转录本DTX4和TYK2的甲基化增强,从而导致TYK2mRNA的降解和更高的效率DTX4mRNA的翻译。这导致促进病毒裂解复制进展的IFN信号传导的减弱。此外,抑制这些转录物的m6A甲基化导致IFN的产生增加,病毒拷贝数大幅减少,这表明裂解病毒复制的废除。
    结论:我们的发现阐明了m6A修饰在克服EBV再激活过程中的先天免疫应答中的重要性。我们现在报道,在裂解再激活过程中,EBV靶向宿主的RNA甲基化系统,通过抑制促进病毒成功裂解复制的干扰素信号来减弱先天免疫反应。
    BACKGROUND: Reactivation of Epstein Barr virus (EBV) leads to modulation of the viral and cellular epitranscriptome. N6-methyladenosine (m6A) modification is a type of RNA modification that regulates metabolism of mRNAs. Previous reports demonstrated that m6A modification affects the stability and metabolism of EBV encoded mRNAs. However, the effect of reactivation on reprograming of the cellular mRNAs, and how this contributes to successful induction of lytic reactivation is not known.
    METHODS: Methylated RNA immunoprecipitation sequencing (MeRIP-seq), transcriptomic RNA sequencing (RNA-seq) and RNA pull-down PCR were used to screen and validate differentially methylated targets. Western blotting, quantitative real-time PCR (RT-qPCR) and immunocytochemistry were used to investigate the expression and localization of different proteins. RNA stability and polysome analysis assays were used to detect the half-lives and translation efficiencies of downstream genes. Insertion of point mutation to disrupt the m6A methylation sites was used to verify the effect of m6A methylation on its stability and expression levels.
    RESULTS: We report that during EBV reactivation the m6A eraser ALKBH5 is significantly downregulated leading to enhanced methylation of the cellular transcripts DTX4 and TYK2, that results in degradation of TYK2 mRNAs and higher efficiency of translation of DTX4 mRNAs. This resulted in attenuation of IFN signaling that promoted progression of viral lytic replication. Furthermore, inhibition of m6A methylation of these transcripts led to increased production of IFN, and a substantial reduction in viral copy number, which suggests abrogation of lytic viral replication.
    CONCLUSIONS: Our findings illuminate the significance of m6A modification in overcoming the innate immune response during EBV reactivation. We now report that during lytic reactivation EBV targets the RNA methylation system of the host to attenuate the innate immune response by suppressing the interferon signaling which facilitates successful lytic replication of the virus.
    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
    EB病毒(EBV)颠覆宿主表观遗传途径,在病毒潜伏期程序之间切换,定植在B细胞区室,并重新激活。在记忆B细胞内,终生感染的水库,EBV基因组DNA和组蛋白甲基化标记限制基因表达。但是这种表观遗传策略也能使EBV感染的肿瘤,包括伯基特淋巴瘤,逃避免疫检测.关于支持EBV表观基因组景观的宿主细胞代谢途径知之甚少。因此,我们使用了氨基酸限制,代谢组学,和CRISPR方法来确定丰富的甲硫氨酸供应和相互连接的甲硫氨酸和叶酸循环维持BurkittEBV基因沉默。蛋氨酸限制,或蛋氨酸循环扰动,低甲基化EBV基因组和去抑制潜伏膜蛋白和裂解基因表达。甲硫氨酸代谢也形成了B细胞永生化所需的EBV潜伏期基因调节。饮食蛋氨酸限制在体内改变了鼠Burkitt异种移植物代谢组和抑制的EBV免疫原。这些结果突出了支持EBVB细胞生命周期的表观遗传/免疫代谢串扰,并提出了治疗方法。
    Epstein-Barr virus (EBV) subverts host epigenetic pathways to switch between viral latency programs, colonize the B cell compartment, and reactivate. Within memory B cells, the reservoir for lifelong infection, EBV genomic DNA and histone methylation marks restrict gene expression. But this epigenetic strategy also enables EBV-infected tumors, including Burkitt lymphomas, to evade immune detection. Little is known about host cell metabolic pathways that support EBV epigenome landscapes. We therefore used amino acid restriction, metabolomic, and CRISPR approaches to identify that an abundant methionine supply and interconnecting methionine and folate cycles maintain Burkitt EBV gene silencing. Methionine restriction, or methionine cycle perturbation, hypomethylated EBV genomes and de-repressed latent membrane protein and lytic gene expression. Methionine metabolism also shaped EBV latency gene regulation required for B cell immortalization. Dietary methionine restriction altered murine Burkitt xenograft metabolomes and de-repressed EBV immunogens in vivo. These results highlight epigenetic/immunometabolism crosstalk supporting the EBV B cell life cycle and suggest therapeutic approaches.
    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
    EB病毒(EBV)的再激活与EBV相关的恶性肿瘤有关,被认为是治疗的有益靶标。穿心莲内酯据称具有抗病毒和抗肿瘤活性。因此,本研究旨在探讨穿心莲内酯对EBV阳性癌细胞中EBV裂解再激活的抑制作用。首先评估穿心莲内酯在EBV阳性癌细胞中的细胞毒性;P3HR1,AGS-EBV和HONE1-EBV细胞,使用MTT测定。在这里,在穿心莲内酯处理的细胞中,EBV裂解基因BALF5,BRLF1和BZLF1的自发表达受到显着抑制。因此,穿心莲内酯抑制丁酸钠(NaB)诱导的EBV裂解再激活中Zta的表达和病毒的产生。此外,蛋白质组学和生物信息学分析显示,在所有处理的细胞系中,抑制EBV裂解再激活的差异表达蛋白与组蛋白修饰和染色质组织功能相关,如组蛋白H3-K9修饰和组蛋白H3-K27甲基化。一起来看,穿心莲内酯通过抑制EBV裂解基因抑制EBV阳性癌细胞的EBV再激活,可能,通过组蛋白修饰。
    Reactivation of Epstein-Barr virus (EBV) is associated with EBV-associated malignancies and is considered to be a benefit target for treatment. Andrographolide is claimed to have antiviral and anti-tumor activities. Therefore, this study aimed to investigate the effect of andrographolide on the inhibition of EBV lytic reactivation in EBV-positive cancer cells. The cytotoxicity of andrographolide was firstly evaluated in EBV-positive cancer cells; P3HR1, AGS-EBV and HONE1-EBV cells, using an MTT assay. Herein, the spontaneous expression of EBV lytic genes; BALF5, BRLF1 and BZLF1, was significantly inhibited in andrographolide-treated cells. Accordingly, andrographolide inhibited the expression of Zta and viral production in sodium butyrate (NaB)-induced EBV lytic reactivation. Additionally, proteomics and bioinformatics analysis revealed the differentially expressed proteins that inhibit EBV lytic reactivation in all treated cell lines were functionally related with the histone modifications and chromatin organization, such as histone H3-K9 modification and histone H3-K27 methylation. Taken together, andrographolide inhibits EBV reactivation in EBV-positive cancer cells by inhibiting EBV lytic genes, probably, through the histone modifications.
    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