restriction factor

制约因素
  • 文章类型: Journal Article
    线粒体是抗病毒反应的关键协调器,可作为先天免疫信号复合物组装和激活的平台。为了应对病毒感染,线粒体可以被触发释放免疫刺激分子,这些分子可以促进干扰素的产生。这些相同的分子可以通过受损的线粒体释放来诱导致病,在没有感染的情况下的抗病毒样免疫反应。这篇综述探讨了含三方基序(TRIM)蛋白家族的成员,它们在抗病毒防御中的作用被认可,调节基于线粒体的先天免疫激活。在抗病毒防御中,TRIM是免疫信号转导途径的重要组成部分,并作为直接作用的病毒限制因子发挥作用。当控制与线粒体相关的免疫激活时,TRIM在概念上进行相似的活动。首先,它们调节可被线粒体分子激活的免疫信号通路。第二,它们协调通过线粒体自噬直接去除线粒体和相关的免疫激活因子。这些见解拓宽了TRIM在先天免疫中的作用范围,并可能暗示TRIM在与线粒体衍生的炎症相关的疾病中。
    Mitochondria are key orchestrators of antiviral responses that serve as platforms for the assembly and activation of innate immune-signaling complexes. In response to viral infection, mitochondria can be triggered to release immune-stimulatory molecules that can boost interferon production. These same molecules can be released by damaged mitochondria to induce pathogenic, antiviral-like immune responses in the absence of infection. This review explores how members of the tripartite motif-containing (TRIM) protein family, which are recognized for their roles in antiviral defense, regulate mitochondria-based innate immune activation. In antiviral defense, TRIMs are essential components of immune signal transduction pathways and function as directly acting viral restriction factors. TRIMs carry out conceptually similar activities when controlling immune activation related to mitochondria. First, they modulate immune-signaling pathways that can be activated by mitochondrial molecules. Second, they co-ordinate the direct removal of mitochondria and associated immune-activating factors through mitophagy. These insights broaden the scope of TRIM actions in innate immunity and may implicate TRIMs in diseases associated with mitochondria-derived inflammation.
    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
    人APOBEC3酶是单链(ss)DNA和RNA胞苷脱氨酶家族,其作为针对病毒和逆转录元件的固有免疫的一部分。这些酶使胞嘧啶脱氨基以形成尿嘧啶,尿嘧啶可以在功能上失活或引起病毒或逆转录元件基因组的降解。此外,APOBEC3通过蛋白质和核酸相互作用具有非脱氨作用的抗病毒活性。如果表达水平失调,一些APOBEC3酶可以进入人类基因组,导致脱氨和诱变,有助于癌症的发生和进化。虽然已知APOBEC3酶与大核糖核蛋白复合物相互作用,功能和RNA依赖性尚未完全了解。为了进一步了解它们的细胞作用,我们通过亲和纯化质谱(AP-MS)确定了人类APOBEC3酶的蛋白质相互作用网络,并绘制了一组不同的蛋白质-蛋白质和蛋白质-RNA介导的相互作用。我们的分析确定了APOBEC3C之间的新型RNA介导的相互作用,APOBEC3H单倍型I和II,和带有剪接体蛋白的APOBEC3G,以及APOBEC3G和APOBEC3H单倍型I,其蛋白质参与tRNA甲基化和ncRNA从细胞核输出。此外,我们鉴定了不依赖RNA的蛋白质-蛋白质与APOBEC3B的相互作用,APOBEC3D,和APOBEC3F和蛋白质折叠伴侣的prefoldin家族。prefoldin5(PFD5)和APOBEC3B之间的相互作用破坏了PFD5诱导癌基因cMyc降解的能力,在癌症中涉及APOBEC3B蛋白相互作用网络。总之,结果揭示了APOBEC3家族的新功能和相互作用,并表明它们可能在细胞RNA生物学中具有基本作用,它们的蛋白质-蛋白质相互作用不是多余的,与肿瘤抑制因子有蛋白质-蛋白质相互作用,表明在癌症生物学中的作用。数据可通过具有标识符PXD044275的ProteomeXchange获得。
    Human APOBEC3 enzymes are a family of single-stranded (ss)DNA and RNA cytidine deaminases that act as part of the intrinsic immunity against viruses and retroelements. These enzymes deaminate cytosine to form uracil which can functionally inactivate or cause degradation of viral or retroelement genomes. In addition, APOBEC3s have deamination-independent antiviral activity through protein and nucleic acid interactions. If expression levels are misregulated, some APOBEC3 enzymes can access the human genome leading to deamination and mutagenesis, contributing to cancer initiation and evolution. While APOBEC3 enzymes are known to interact with large ribonucleoprotein complexes, the function and RNA dependence are not entirely understood. To further understand their cellular roles, we determined by affinity purification mass spectrometry (AP-MS) the protein interaction network for the human APOBEC3 enzymes and mapped a diverse set of protein-protein and protein-RNA mediated interactions. Our analysis identified novel RNA-mediated interactions between APOBEC3C, APOBEC3H Haplotype I and II, and APOBEC3G with spliceosome proteins, and APOBEC3G and APOBEC3H Haplotype I with proteins involved in tRNA methylation and ncRNA export from the nucleus. In addition, we identified RNA-independent protein-protein interactions with APOBEC3B, APOBEC3D, and APOBEC3F and the prefoldin family of protein-folding chaperones. Interaction between prefoldin 5 (PFD5) and APOBEC3B disrupted the ability of PFD5 to induce degradation of the oncogene cMyc, implicating the APOBEC3B protein interaction network in cancer. Altogether, the results uncover novel functions and interactions of the APOBEC3 family and suggest they may have fundamental roles in cellular RNA biology, their protein-protein interactions are not redundant, and there are protein-protein interactions with tumor suppressors, suggesting a role in cancer biology. Data are available via ProteomeXchange with the identifier PXD044275.
    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
    染色质重塑因子SPOC1(PHF13)是转录共调节因子,已被鉴定为针对各种病毒的限制因子,包括人巨细胞病毒(HCMV)。对于HCMV,SPOC1显示在低感染复数(MOI)下阻断立即早期(IE)基因表达的发作。这里,我们证明SPOC1介导的IE表达限制被增加的病毒滴度中和。有趣的是,我们的研究表明,SPOC1发挥额外的抗病毒功能超出IE阶段的HCMV复制。SPOC1在高MOI条件下的表达导致严重受损的病毒DNA复制和病毒颗粒释放,这可能归因于低效的病毒转录。随着点击化学的使用,在感染后的晚期时间点研究病毒DNA的定位.有趣的是,我们检测到SPOC1,RNA聚合酶IIS5P和polycomb阻遏复合物2(PRC2)组分的共定位,在假设携带病毒转录位点的区域与病毒DNA非常接近.我们进一步鉴定了SPOC1的N末端结构域负责与PRC2复合物的亚基EZH2相互作用。通过这项研究,我们报道了SPOC1对HCMV的新的和有效的抗病毒功能,即使不受限制的IE基因表达也是有效的。
    The chromatin-remodeler SPOC1 (PHF13) is a transcriptional co-regulator and has been identified as a restriction factor against various viruses, including human cytomegalovirus (HCMV). For HCMV, SPOC1 was shown to block the onset of immediate-early (IE) gene expression under low multiplicities of infection (MOI). Here, we demonstrate that SPOC1-mediated restriction of IE expression is neutralized by increasing viral titers. Interestingly, our study reveals that SPOC1 exerts an additional antiviral function beyond the IE phase of HCMV replication. Expression of SPOC1 under conditions of high MOI resulted in severely impaired viral DNA replication and viral particle release, which may be attributed to inefficient viral transcription. With the use of click chemistry, the localization of viral DNA was investigated at late time points after infection. Intriguingly, we detected a co-localization of SPOC1, RNA polymerase II S5P and polycomb repressor complex 2 (PRC2) components in close proximity to viral DNA in areas that are hypothesized to harbor viral transcription sites. We further identified the N-terminal domain of SPOC1 to be responsible for interaction with EZH2, a subunit of the PRC2 complex. With this study, we report a novel and potent antiviral function of SPOC1 against HCMV that is efficient even with unrestricted IE gene expression.
    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:哺乳动物雷帕霉素靶标(mTOR)信号通路调节蛋白质磷酸化并控制主要细胞过程。mTOR被大脑中富含的小G蛋白Ras同源物(Rheb)激活,其由Rheb1和Rheb样-1(RhebL1)基因编码。目前缺乏关于RhebL1的作用的信息,特别是其在病毒感染中的参与。在本研究中,我们研究了RhebL1在人类流感A/NWS/33(NWS/33)(H1N1)病毒感染恒河猴肾(LLC-MK2)细胞和人类II型肺泡上皮(A549)细胞中的作用。
    方法:为了评估NWS/33病毒复制的效率,通过间接免疫荧光(IIF)检查病毒核蛋白的表达,并通过50%的组织培养感染剂量测定法检查病毒产量。使用RNA介导的RNA干扰方法来研究RhebL1在NWS/33感染期间的作用。通过IIF评估RhebL1表达,西方印迹,和酶联免疫吸附测定。应用双尾学生t检验来评估组间的差异。
    结果:RhebL1在本研究中使用的细胞模型中差异表达。沉默RhebL1基因导致A549细胞中NWS/33病毒感染增加,但不在LLC-MK2细胞中。此外,高磷酸化细胞角蛋白8的表达,NWS/33病毒感染效率的标志,在耗竭RhebL1的A549细胞中增加,但在LLC-MK2细胞中几乎保持不变。
    结论:这些是首次结果显示内源性RhebL1蛋白在病毒感染过程中的参与。我们的数据表明,RhebL1在流感病毒感染期间发挥宿主细胞依赖性调节作用。RhebL1似乎是A549细胞中NWS/33病毒复制的限制性因素,但不是在LLC-MK2中。
    BACKGROUND: The Mammalian Target of Rapamycin (mTOR) signaling pathway regulates protein phosphorylation and exerts control over major cellular processes. mTOR is activated by the small G-protein Ras Homolog Enriched in Brain (Rheb), which is encoded by the Rheb1 and Rheb-like-1 (RhebL1) genes. There is currently a paucity of information on the role of RhebL1, and specifically its involvement in viral infection. In the present study we investigated the role of RhebL1 during human influenza A/NWS/33 (NWS/33) (H1N1) virus infection of rhesus monkey-kidney (LLC-MK2) cells and human type II alveolar epithelial (A549) cells.
    METHODS: To assess the efficiency of NWS/33 virus replication, the expression of viral nucleoprotein was examined by indirect immunofluorescence (IIF) and the viral yield by fifty percent tissue culture infectious dose assay. An RNA-mediated RNA interference approach was used to investigate the role of RhebL1 during NWS/33 infection. RhebL1 expression was evaluated by IIF, Western blotting, and enzyme-linked immunosorbent assays. A two-tailed Student\'s t-test was applied to evaluate differences between groups.
    RESULTS: RhebL1 was differentially expressed in the cell models used in this study. Silencing of the RhebL1 gene led to increased NWS/33 virus infection in A549 cells, but not in LLC-MK2 cells. Moreover, the expression of hyperphosphorylated cytokeratin 8, a marker of NWS/33 virus infection efficiency, increased in A549 cells depleted of RhebL1 but remained almost unchanged in LLC-MK2 cells.
    CONCLUSIONS: These are the first results showing involvement of the endogenous RhebL1 protein during viral infection. Our data suggests that RhebL1 exerts a host cell-dependent modulatory role during influenza virus infection. RhebL1 appears to be a restrictive factor against NWS/33 virus replication in A549 cells, but not in LLC-MK2.
    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
    宿主抗病毒蛋白通过靶向病毒生命周期的许多特征来抑制灵长类慢病毒和其他逆转录病毒。慢病毒衣壳蛋白和组装的病毒核心已知通过多种抑制,直接作用的抗病毒蛋白。几种表型,包括被称为Lv1至Lv5的那些,已经被描述为针对一些但不是所有灵长类动物慢病毒感染的细胞类型特异性阻断。在这里,我们回顾了已知的衣壳靶向阻断感染的重要特征,以及几种阻断感染的重要特征,其中负责抑制的基因仍有待鉴定。我们概述了这些区块的特征,以及当前的方法现在如何非常适合找到这些抗病毒基因,并解决了HIV和逆转录病毒学领域中这些长期存在的谜团。
    Host antiviral proteins inhibit primate lentiviruses and other retroviruses by targeting many features of the viral life cycle. The lentiviral capsid protein and the assembled viral core are known to be inhibited through multiple, directly acting antiviral proteins. Several phenotypes, including those known as Lv1 through Lv5, have been described as cell type-specific blocks to infection against some but not all primate lentiviruses. Here we review important features of known capsid-targeting blocks to infection together with several blocks to infection for which the genes responsible for the inhibition still remain to be identified. We outline the features of these blocks as well as how current methodologies are now well suited to find these antiviral genes and solve these long-standing mysteries in the HIV and retrovirology fields.
    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
    内源性逆转录病毒(ERV)是祖先病毒感染的残余物。猫白血病病毒(FeLV)是家猫的外源性和内源性逆转录病毒。它分为几个子组(A,B,C,D,E,和T)基于病毒受体干扰特性或受体使用。ERV衍生的分子有益于动物,赋予对传染病的抵抗力。然而,内源性FeLV(enFeLV)的包膜缺陷(env)基因编码的可溶性蛋白在FeLV亚群T感染中起辅助因子的作用。因此,该基因是否有助于病毒感染尚不清楚.基于ERV衍生分子的性质,我们假设有缺陷的env基因具有对宿主有利的抗病毒活性,因为FeLV亚群B(FeLV-B),来自enFeLVenv的重组病毒,仅限于家猫之间的病毒传播。当测试来自enFeLV的可溶性截短Env蛋白对enFeLV和FeLV-B的抑制作用时,他们抑制病毒感染。值得注意的是,这种抗病毒机制被扩展到长臂猿白血病病毒感染,考拉逆转录病毒A,和赫维翼龙γ病毒。尽管这些病毒使用猫磷酸盐转运蛋白1(fePit1)和磷酸盐转运蛋白2作为受体,抑制机制涉及fepit1依赖性方式的竞争性受体结合。受体使用的转变可能是为了避免抑制作用而发生的。总的来说,这些发现强调了来自enFeLV的可溶性截短Env蛋白作为抗逆转录病毒感染的限制因子的可能出现,并将有助于通过控制逆转录病毒传播来发展宿主免疫和抗病毒防御.IMPORTANCERAQ病毒在使用逆转录酶将RNA基因组转化为DNA方面是独特的,感染生殖细胞,并传给后代。许多古老的逆转录病毒序列被称为内源性逆转录病毒(ERV)。来源于ERV的可溶性Env蛋白作为辅助FeLV-T感染的辅因子发挥作用。然而,在这里,我们表明,可溶性Env蛋白表现出抗病毒活性,并通过竞争性受体结合提供对哺乳动物逆转录病毒感染的抗性。特别是,这一发现可以解释为什么在家猫中没有观察到FeLV-B传播。ERV衍生的分子可以在进化军备竞赛中有益于动物,强调ERV的双刃剑性质。
    Endogenous retroviruses (ERVs) are remnants of ancestral viral infections. Feline leukemia virus (FeLV) is an exogenous and endogenous retrovirus in domestic cats. It is classified into several subgroups (A, B, C, D, E, and T) based on viral receptor interference properties or receptor usage. ERV-derived molecules benefit animals, conferring resistance to infectious diseases. However, the soluble protein encoded by the defective envelope (env) gene of endogenous FeLV (enFeLV) functions as a co-factor in FeLV subgroup T infections. Therefore, whether the gene emerged to facilitate viral infection is unclear. Based on the properties of ERV-derived molecules, we hypothesized that the defective env genes possess antiviral activity that would be advantageous to the host because FeLV subgroup B (FeLV-B), a recombinant virus derived from enFeLV env, is restricted to viral transmission among domestic cats. When soluble truncated Env proteins from enFeLV were tested for their inhibitory effects against enFeLV and FeLV-B, they inhibited viral infection. Notably, this antiviral machinery was extended to infection with the Gibbon ape leukemia virus, Koala retrovirus A, and Hervey pteropid gammaretrovirus. Although these viruses used feline phosphate transporter 1 (fePit1) and phosphate transporter 2 as receptors, the inhibitory mechanism involved competitive receptor binding in a fePit1-dependent manner. The shift in receptor usage might have occurred to avoid the inhibitory effect. Overall, these findings highlight the possible emergence of soluble truncated Env proteins from enFeLV as a restriction factor against retroviral infection and will help in developing host immunity and antiviral defense by controlling retroviral spread.IMPORTANCERetroviruses are unique in using reverse transcriptase to convert RNA genomes into DNA, infecting germ cells, and transmitting to offspring. Numerous ancient retroviral sequences are known as endogenous retroviruses (ERVs). The soluble Env protein derived from ERVs functions as a co-factor that assists in FeLV-T infection. However, herein, we show that the soluble Env protein exhibits antiviral activity and provides resistance to mammalian retrovirus infection through competitive receptor binding. In particular, this finding may explain why FeLV-B transmission is not observed among domestic cats. ERV-derived molecules can benefit animals in an evolutionary arms race, highlighting the double-edged-sword nature of ERVs.
    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
    丝氨酸掺入剂(SERINC)蛋白家族的成员发挥广泛的抗病毒活性,许多病毒编码SERINC拮抗剂来规避这些限制。最近对介导限制和拮抗作用的机制获得了重要的新见解。在这次审查中,我们总结了目前对SERINC蛋白在HIV-1感染中的作用方式和相关性的理解.将特别关注最近的发现,这些发现为限制HIV-1病毒的传染性提供了重要的新机制见解。包括SERINC的脂质加扰酶活性的发现及其被HIV-1致病因子Nef的拮抗作用。我们还讨论了SERINC蛋白增强促炎信号并减少骨髓细胞中病毒基因表达的几种其他抗病毒活性的鉴定和含义。SERINC蛋白作为抗HIV-1感染的细胞固有免疫的通用和多功能调节剂出现。
    Members of the serine incorporator (SERINC) protein family exert broad antiviral activity, and many viruses encode SERINC antagonists to circumvent these restrictions. Significant new insight was recently gained into the mechanisms that mediate restriction and antagonism. In this review, we summarize our current understanding of the mode of action and relevance of SERINC proteins in HIV-1 infection. Particular focus will be placed on recent findings that provided important new mechanistic insights into the restriction of HIV-1 virion infectivity, including the discovery of SERINC\'s lipid scramblase activity and its antagonism by the HIV-1 pathogenesis factor Nef. We also discuss the identification and implications of several additional antiviral activities by which SERINC proteins enhance pro-inflammatory signaling and reduce viral gene expression in myeloid cells. SERINC proteins emerge as versatile and multifunctional regulators of cell-intrinsic immunity against HIV-1 infection.
    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
    人类鼻病毒是慢性呼吸道疾病严重加重期间最常见的病毒,比如慢性阻塞性肺病.在这种疾病中,肺泡巨噬细胞显示显著降低的吞噬功能,这可能与细菌超感染有关。然而,人类鼻病毒如何影响巨噬细胞的功能在很大程度上是未知的。用HRV16处理的巨噬细胞表现出缺乏的细菌杀伤活性,吞噬溶酶体生物发生受损,并改变了细胞内区室。使用RNA测序,我们确定了在原代人巨噬细胞中由病毒上调的小GTP酶ARL5b.重要的是,ARL5b的消耗挽救了HRV16暴露后巨噬细胞中细菌清除和内体标志物的定位。在许可细胞中,ARL5b的消耗增加了HRV16病毒粒子的分泌。因此,我们确定ARL5b是巨噬细胞细胞内运输动力学和吞噬溶酶体生物发生的新型调节因子,也是允许细胞中HRV16的限制因子.
    Human rhinovirus is the most frequently isolated virus during severe exacerbations of chronic respiratory diseases, like chronic obstructive pulmonary disease. In this disease, alveolar macrophages display significantly diminished phagocytic functions that could be associated with bacterial superinfections. However, how human rhinovirus affects the functions of macrophages is largely unknown. Macrophages treated with HRV16 demonstrate deficient bacteria-killing activity, impaired phagolysosome biogenesis, and altered intracellular compartments. Using RNA sequencing, we identify the small GTPase ARL5b to be upregulated by the virus in primary human macrophages. Importantly, depletion of ARL5b rescues bacterial clearance and localization of endosomal markers in macrophages upon HRV16 exposure. In permissive cells, depletion of ARL5b increases the secretion of HRV16 virions. Thus, we identify ARL5b as a novel regulator of intracellular trafficking dynamics and phagolysosomal biogenesis in macrophages and as a restriction factor of HRV16 in permissive cells.
    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:m6ARNA修饰通路在多种细胞过程和病毒生命周期中发挥重要作用。这里,我们研究了PIAS1与m6A阅读蛋白YTHDF2之间的关系,该蛋白通过与m6A修饰的RNA结合而参与调节RNA稳定性。我们发现YTHDF2的N端和C端区域都与PIAS1相互作用。我们显示PIAS1促进YTHDF2在三个特定赖氨酸残基处的SUMOylation。我们还证明PIAS1增强YTHDF2的抗EBV活性。我们进一步揭示了PIAS1介导其他YTHDF家族成员的SUMOylation,即,YTHDF1和YTHDF3,以限制EBV的复制。这些发现共同阐明了YTHDF蛋白在通过PIAS1介导的SUMO化控制病毒RNA衰变和EBV复制中的重要调节机制。
    YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) is a member of the YTH protein family that binds to N6-methyladenosine (m6A)-modified RNA, regulating RNA stability and restricting viral replication, including Epstein-Barr virus (EBV). PIAS1 is an E3 small ubiquitin-like modifier (SUMO) ligase known as an EBV restriction factor, but its role in YTHDF2 SUMOylation remains unclear. In this study, we investigated the functional regulation of YTHDF2 by PIAS1. We found that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues (K281, K571, and K572). Importantly, PIAS1 synergizes with wild-type YTHDF2, but not a SUMOylation-deficient mutant, to limit EBV lytic replication. Mechanistically, YTHDF2 lacking SUMOylation exhibits reduced binding to EBV transcripts, leading to increased viral mRNA stability. Furthermore, PIAS1 mediates SUMOylation of YTHDF2\'s paralogs, YTHDF1 and YTHDF3, to restrict EBV replication. These results collectively uncover a unique mechanism whereby YTHDF family proteins control EBV replication through PIAS1-mediated SUMOylation, highlighting the significance of SUMOylation in regulating viral mRNA stability and EBV replication.IMPORTANCEm6A RNA modification pathway plays important roles in diverse cellular processes and viral life cycle. Here, we investigated the relationship between PIAS1 and the m6A reader protein YTHDF2, which is involved in regulating RNA stability by binding to m6A-modified RNA. We found that both the N-terminal and C-terminal regions of YTHDF2 interact with PIAS1. We showed that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues. We also demonstrated that PIAS1 enhances the anti-EBV activity of YTHDF2. We further revealed that PIAS1 mediates the SUMOylation of other YTHDF family members, namely, YTHDF1 and YTHDF3, to limit EBV replication. These findings together illuminate an important regulatory mechanism of YTHDF proteins in controlling viral RNA decay and EBV replication through PIAS1-mediated SUMOylation.
    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
    冠状病毒感染诱导干扰素刺激的基因,其中一个编码Tetherin,一种跨膜蛋白,抑制各种包膜病毒从感染细胞中释放。先前的研究表明,SARS-CoV编码两种Tetherin拮抗剂:Spike蛋白(S),诱导Tetherin的溶酶体降解,ORF7a,改变其糖基化。同样,SARS-CoV-2还显示在Tetherin存在下使用ORF7a和Spike增强病毒体释放。这里,我们直接比较这两种病毒蛋白对抗Tetherin的能力和机制。因此,使用流式细胞术和蛋白质印迹分析研究了ORF7a或S表达时的细胞表面和总Tetherin水平。SARS-CoV和SARS-CoV-2S仅以细胞类型依赖性方式略微降低了Tetherin细胞表面水平。在HEK293T细胞中,在高外源Tetherin表达的条件下,SARS-CoV-2S和ORF7a比来自SARS-CoV的相应对应物更有效地降低了总细胞Tetherin水平。然而,来自两个物种的ORF7a能够改变Tetherin糖基化。在来自不同SARS-CoV-2变体的S蛋白中,降低Tetherin总蛋白水平的能力是保守的(α,γ,δ,ο)。虽然SARS-CoV-2S和ORF7a都与Tetherin共定位,在双杂交试验中,只有ORF7a与限制因子直接相互作用.尽管存在多种Tetherin拮抗剂,Tetherin敲除后,Caco-2细胞中的SARS-CoV-2复制进一步增强。总之,我们的数据显示,内源性Tetherin限制了SARS-CoV-2的复制,Tetherin的抗病毒活性仅被具有不同和互补作用模式的病毒拮抗剂部分抵消.
    Coronavirus infection induces interferon-stimulated genes, one of which encodes Tetherin, a transmembrane protein inhibiting the release of various enveloped viruses from infected cells. Previous studies revealed that SARS-CoV encodes two Tetherin antagonists: the Spike protein (S), inducing lysosomal degradation of Tetherin, and ORF7a, altering its glycosylation. Similarly, SARS-CoV-2 has also been shown to use ORF7a and Spike to enhance virion release in the presence of Tetherin. Here, we directly compare the abilities and mechanisms of these two viral proteins to counteract Tetherin. Therefore, cell surface and total Tetherin levels upon ORF7a or S expression were investigated using flow cytometry and Western blot analysis. SARS-CoV and SARS-CoV-2 S only marginally reduced Tetherin cell surface levels in a cell type-dependent manner. In HEK293T cells, under conditions of high exogenous Tetherin expression, SARS-CoV-2 S and ORF7a reduced total cellular Tetherin levels much more efficiently than the respective counterparts derived from SARS-CoV. Nevertheless, ORF7a from both species was able to alter Tetherin glycosylation. The ability to decrease total protein levels of Tetherin was conserved among S proteins from different SARS-CoV-2 variants (α, γ, δ, ο). While SARS-CoV-2 S and ORF7a both colocalized with Tetherin, only ORF7a directly interacted with the restriction factor in a two-hybrid assay. Despite the presence of multiple Tetherin antagonists, SARS-CoV-2 replication in Caco-2 cells was further enhanced upon Tetherin knockout. Altogether, our data show that endogenous Tetherin restricts SARS-CoV-2 replication and that the antiviral activity of Tetherin is only partially counteracted by viral antagonists with differential and complementary modes of action.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号