Vesiculovirus

囊泡病毒
  • 文章类型: Journal Article
    马尔堡病毒(MARV),丝状病毒,1967年在马尔堡首次被发现,德国,贝尔格莱德,前南斯拉夫。从那以后,MARV在非洲部分地区引起了人类疾病的零星爆发,病死率很高,最大的疫情发生在2004/05年安哥拉。从2021年到2023年,几内亚发生了MARV疫情,加纳,几内亚,坦桑尼亚,强调将其流行区扩展到新的地理区域。目前没有批准的针对MARV的疫苗或疗法,但是几种候选疫苗在临床前研究中显示出了希望。我们通过给仓鼠接种单剂量的腺病毒(ChAdOx-1MARV)疫苗,同时比较了三种疫苗平台,一种基于甲病毒复制子的RNA(LION-MARV)疫苗,或重组水泡性口炎病毒(VSV-MARV)疫苗,都表达MARV糖蛋白作为抗原。接种后4周用仓鼠适应的MARV进行致命攻击导致VSV-MARV和LION-MARV组和ChAdOx-1MARV组的83%的保护。对抗原特异性体液反应及其功能的评估揭示了疫苗平台依赖性的差异,特别是在Fc效应子功能中。
    Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.
    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
    有效疫苗递送的一个主要限制是其对稳健的冷链基础设施的依赖性。虽然水泡性口炎病毒(VSV)已被证明是包括埃博拉在内的疾病的有效病毒疫苗载体,其-70°C存储要求是访问弱势位置和人口的重大限制。先前的工作已经显示了具有支链淀粉和海藻糖(PT)干燥膜的组合的病毒疫苗的热稳定性。为了提高VSV的热稳定性,我们优化了PT配方的浓度和成分,以及增强真空干燥的干燥方法。当配制在PT薄膜中时,VSV可以在4°C下储存32周,PFU损失小于2log,在25°C下,PFU损失为2.5log,并且在37°C下具有3.1logPFU损失。这些结果证明了VSV热稳定性的显著进步,降低VSV载体疫苗的冷链需求。
    One major limitation of effective vaccine delivery is its dependency on a robust cold chain infrastructure. While Vesicular stomatitis virus (VSV) has been demonstrated to be an effective viral vaccine vector for diseases including Ebola, its -70 °C storage requirement is a significant limitation for accessing disadvantaged locations and populations. Previous work has shown thermal stabilization of viral vaccines with a combination of pullulan and trehalose (PT) dried films. To improve the thermal stability of VSV, we optimized PT formulation concentrations and components, as well as drying methodology with enhanced vacuum drying. When formulated in PT films, VSV can be stored for 32 weeks at 4 °C with less than 2 log PFU loss, at 25 °C with 2.5 log PFU loss, and at 37 °C with 3.1 log PFU loss. These results demonstrate a significant advancement in VSV thermal stabilization, decreasing the cold chain requirements for VSV vectored vaccines.
    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
    已经研究了组蛋白去乙酰化家族蛋白在通过去乙酰化非组蛋白蛋白调节病毒复制中的功能。RIG-I(维甲酸诱导基因I)是RNA病毒诱导的先天抗病毒信号通路中的关键蛋白。我们之前的研究表明,HDAC8(组蛋白去乙酰化酶8)参与先天抗病毒免疫反应,但病毒感染过程中的潜在机制仍不清楚。在这项研究中,我们表明HDAC8参与了水泡性口炎病毒(VSV)复制的调节。HDAC8的过表达抑制,而敲低促进VSV复制。进一步的探索表明,HDAC8与RIG-I相互作用并脱乙酰,这最终导致增强先天抗病毒免疫反应。总的来说,我们的数据清楚地表明,HDAC8通过促进RIG-I介导的干扰素产生和下游信号通路抑制VSV复制.
    Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream 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
    尽管患者在多种肿瘤中受益于免疫检查点抑制(ICI)治疗,耐药性可能来自免疫抑制肿瘤微环境(TME),这是特别真实的肝细胞癌(HCC)。由于溶瘤病毒(OV)可以产生高度免疫浸润,炎性TME,OV可能会通过招募来恢复ICI的响应能力,启动,和激活抗肿瘤T细胞。在这里,我们发现相反,溶瘤性水疱性口炎病毒,表达干扰素β(VSV-IFNβ),在部分抗PD-L1反应性HCC模型中拮抗抗PD-L1治疗的效果。通过飞行时间的细胞测定法显示,VSV-IFNβ扩增显性抗病毒效应CD8T细胞,伴随着抗肿瘤T细胞群的相对消失,它们是抗PD-L1的靶标。然而,通过在VSV内表达一系列HCC肿瘤抗原,OV和抗PD-L1联合治疗获益可以恢复。我们的数据通过显示显性抗病毒T细胞应答可以抑制亚显性抗肿瘤T细胞应答,为使用高免疫原性病毒作为肿瘤特异性免疫治疗剂提供了警示信息。然而,通过在病毒中编码肿瘤抗原,溶瘤病毒疗法可以产生抗肿瘤T细胞群体,免疫检查点阻断可以有效地发挥作用。
    Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.
    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
    病毒糖蛋白介导进入宿主细胞,从而决定宿主范围和发病机制。此外,它们构成中和抗体反应的主要目标,使它们成为疫苗开发中的重要抗原。编码外源糖蛋白的重组水泡性口炎病毒(VSV)可以提供一种方便安全的替代系统,进化,以及来自难以操作的病毒或需要高生物安全级别遏制的病毒的病毒糖蛋白的抗原性。然而,重组VSV的生产在技术上具有挑战性。在这项工作中,我们提出了一种有效且强大的基于质粒的系统,用于生产编码外源糖蛋白的重组VSV。我们使用来自不同病毒家族的糖蛋白验证该系统,包括沙粒病毒,冠状病毒,和汉坦病毒,以及强调它们在研究突变对病毒适应性的影响方面的效用。总的来说,例如,本文所述的方法可以促进编码外源糖蛋白的天然和重组VSV两者的研究,并且可以用作生产基于VSV的疫苗的基础。
    Viral glycoproteins mediate entry into host cells, thereby dictating host range and pathogenesis. In addition, they constitute the principal target of neutralizing antibody responses, making them important antigens in vaccine development. Recombinant vesicular stomatitis virus (VSV) encoding foreign glycoproteins can provide a convenient and safe surrogate system to interrogate the function, evolution, and antigenicity of viral glycoproteins from viruses that are difficult to manipulate or those requiring high biosafety level containment. However, the production of recombinant VSV can be technically challenging. In this work, we present an efficient and robust plasmid-based system for the production of recombinant VSV encoding foreign glycoproteins. We validate the system using glycoproteins from different viral families, including arenaviruses, coronaviruses, and hantaviruses, as well as highlight their utility for studying the effects of mutations on viral fitness. Overall, the methods described herein can facilitate the study of both native and recombinant VSV encoding foreign glycoproteins and can serve as the basis for the production of VSV-based vaccines.
    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
    溶瘤病毒(OV)治疗的快速发展领域需要开发新的和改进的分析方法,以在生产和开发过程中表征病毒。病毒蛋白的准确监测和绝对定量对于OV产品表征至关重要,可以促进对感染的理解,免疫原性,和病毒复制的发展阶段。像多反应监测(MRM)这样的靶向质谱方法提供了直接检测和定量由替代肽代表的特定靶向蛋白质的可靠方法。我们通过将超高效液相色谱(UPLC)与Sciex6500三级四极杆质谱仪相结合,利用了MRM的功能,开发了一种准确,绝对地定量假型水疱性口炎病毒(VSV)的结构蛋白的测定法。用作新的生物治疗剂(以下称为VSV-GP,以区别于天然VV)。新的UPLC-MRM方法使用重标记的参考标准替代肽提供绝对定量。以已知的精确量添加到标准和样品中时,参考标准对样品制备和/或仪器性能期间的任何小扰动进行标准化和说明,导致准确和精确的定量。由于MRM的多重性质,同时对所有靶向蛋白进行定量。优化的测定已被增强以量化经处理的GP1和GP2蛋白的比率,同时测量包膜蛋白GP复合物(GPC;全长GPC)的任何剩余或未加工形式。
    目的:近年来,溶瘤病毒治疗的发展取得了相当大的势头。水泡性口炎病毒糖蛋白(VSV-GP)是溶瘤病毒治疗平台中出现的一种新的生物治疗剂。可以准确和精确地定量病毒蛋白的新型分析测定法是成功开发作为生物治疗剂的病毒载体的必要条件。我们开发了一种基于超高效液相色谱多反应监测的测定法,以量化VSV-GP不同结构蛋白的绝对浓度。GP复合物(GPC)的完全处理是病毒感染性的先决条件。该测定法扩展了定量全长GPC的潜力,这提供了对GPC处理的理解(以及分别对GP1和GP2的定量)。我们在追踪用VSV-GP感染的HEK-293-F生产细胞系中的GPC处理中使用该测定。
    The rapidly developing field of oncolytic virus (OV) therapy necessitates the development of new and improved analytical approaches for the characterization of the virus during production and development. Accurate monitoring and absolute quantification of viral proteins are crucial for OV product characterization and can facilitate the understanding of infection, immunogenicity, and development stages of viral replication. Targeted mass spectrometry methods like multiple reaction monitoring (MRM) offer a robust way to directly detect and quantify specific targeted proteins represented by surrogate peptides. We have leveraged the power of MRM by combining ultra-high performance liquid chromatography (UPLC) with a Sciex 6500 triple-stage quadrupole mass spectrometer to develop an assay that accurately and absolutely quantifies the structural proteins of a pseudotyped vesicular stomatitis virus (VSV) intended for use as a new biotherapeutic (designated hereafter as VSV-GP to differentiate it from native VSV). The new UPLC-MRM method provides absolute quantification with the use of heavy-labeled reference standard surrogate peptides. When added in known exact amounts to standards and samples, the reference standards normalize and account for any small perturbations during sample preparation and/or instrument performance, resulting in accurate and precise quantification. Because of the multiplexed nature of MRM, all targeted proteins are quantified at the same time. The optimized assay has been enhanced to quantify the ratios of the processed GP1 and GP2 proteins while simultaneously measuring any remaining or unprocessed form of the envelope protein GP complex (GPC; full-length GPC).
    OBJECTIVE: The development of oncolytic viral therapy has gained considerable momentum in recent years. Vesicular stomatitis virus glycoprotein (VSV-GP) is a new biotherapeutic emerging in the oncolytic viral therapy platform. Novel analytical assays that can accurately and precisely quantify the viral proteins are a necessity for the successful development of viral vector as a biotherapeutic. We developed an ultra-high performance liquid chromatography multiple reaction monitoring-based assay to quantify the absolute concentrations of the different structural proteins of VSV-GP. The complete processing of GP complex (GPC) is a prerequisite for the infectivity of the virus. The assay extends the potential for quantifying full-length GPC, which provides an understanding of the processing of GPC (along with the quantification of GP1 and GP2 separately). We used this assay in tracking GPC processing in HEK-293-F production cell lines infected with VSV-GP.
    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
    干扰素(IFN)系统保护哺乳动物免受病毒感染引起的疾病。IFN合成是由病毒感染激活的模式识别受体信号通路诱导的。IFN从感染的细胞分泌,并通过结合细胞表面受体并触发数百个IFN刺激的基因和蛋白质的诱导作用于邻近细胞。其中许多阻止病毒复制的不同步骤。IFN诱导的四三肽重复蛋白(IFIT)是RNA结合蛋白家族。我们和其他人以前曾报道,IFIT2保护小鼠免受许多嗜神经RNA病毒的侵害;事实上,Ifit2-/-小鼠对水泡性口炎病毒(VSV)的鼻内或皮下感染非常敏感。这里,使用新生成的条件敲除鼠标,我们报道,Ifit2表达仅在神经元细胞中的消融足以使小鼠对由鼻内,但不是皮下的,VSV感染。另一种转基因小鼠品系,表达不能结合RNA的突变体IFIT2,与Ifit2-/-小鼠一样容易感染VSV。这些结果表明,IFIT2RNA结合活性对于保护小鼠免受VSV鼻内感染引起的神经系统疾病至关重要。IMPORTANCE干扰素(IFN)抗病毒作用由干扰素刺激基因编码的蛋白质介导。IFN刺激基因(IFIT2)是这样一种蛋白质,抑制许多RNA病毒在小鼠大脑中的复制和由此产生的神经病理学。我们的研究揭示了IFIT2的工作原理。通过仅在神经元细胞中消除Ifit2表达,使用新生成的条件敲除鼠标行,我们表明,感染小鼠的神经元中的Ifit2诱导是干扰素抗病毒功能所必需的。IFIT2没有已知的酶活性;相反,它通过与细胞或病毒蛋白或RNA结合而起作用。我们设计了一种新的小鼠品系,该品系表达了无法结合RNA的突变体IFIT2。这些小鼠对水泡性口炎病毒感染非常敏感,表明IFIT2的RNA结合特性对于其体内抗病毒功能至关重要。
    The interferon (IFN) system protects mammals from diseases caused by virus infections. IFN synthesis is induced by pattern recognition receptor signaling pathways activated by virus infection. IFN is secreted from the infected cells and acts upon neighboring cells by binding cell surface receptors and triggering induction of hundreds of IFN-stimulated genes and proteins, many of which block different steps of virus replication. The IFN-induced tetratricopeptide repeat proteins (IFIT) are a family of RNA-binding proteins. We and others have previously reported that IFIT2 protects mice from many neurotropic RNA viruses; indeed, Ifit2-/- mice are very susceptible to intranasal or subcutaneous infections with vesicular stomatitis virus (VSV). Here, using a newly generated conditional knockout mouse, we report that ablation of Ifit2 expression only in neuronal cells was sufficient to render mice susceptible to neuropathogenesis caused by intranasal, but not subcutaneous, infection of VSV. Another genetically modified mouse line, expressing a mutant IFIT2 that cannot bind RNA, was as susceptible to VSV infection as Ifit2-/- mice. These results demonstrated that IFIT2 RNA-binding activity is essential for protecting mice against neurological diseases caused by intranasal infection of VSV.IMPORTANCEInterferon\'s (IFN\'s) antiviral effects are mediated by the proteins encoded by the interferon-stimulated genes. IFN-stimulated genes (IFIT2) is one such protein, which inhibits replication of many RNA viruses in the mouse brain and the resultant neuropathology. Our study sheds light on how IFIT2 works. By ablating Ifit2 expression only in neuronal cells, using a newly generated conditional knockout mouse line, we showed that Ifit2 induction in the neurons of the infected mouse was necessary for antiviral function of interferon. IFIT2 has no known enzyme activity; instead, it functions by binding to cellular or viral proteins or RNAs. We engineered a new mouse line that expressed a mutant IFIT2 that cannot bind RNA. These mice were very susceptible to infection with vesicular stomatitis virus indicating that the RNA-binding property of IFIT2 was essential for its antiviral function in vivo.
    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
    疱疹病毒B(BV)是一种人畜共患病毒,属于单纯型,与人类单纯疱疹病毒(HSV)相同的属。BV通常在其自然宿主中建立无症状感染,猕猴.然而,在人类中,BV感染导致严重的神经系统疾病和死亡。因此,BV研究只能在高安全级别设施中进行(即,生物安全级别[BSL]4),BV进入的机制尚未完全阐明。在这项研究中,我们产生了假型水泡性口炎病毒(VSV)表达BV糖蛋白使用G-互补VSVΔG系统,我们命名为VSV/BVpv。我们发现四种BV糖蛋白(即,gB,gD,gH,和gL)是生产高滴度VSV/BVpv所需的。此外,VSV/BVpv细胞进入依赖于gD与其细胞受体nectin-1的结合。用内体酸化抑制剂预处理Vero细胞不影响VSV/BVpv感染。结果表明,VSV/BVpv通过与Vero细胞的质膜直接融合而进入,表明其进入途径与天然HSV相似。此外,我们开发了基于VSV/BVpv的化学发光还原中和测试(CRNT),该方法检测了猕猴血浆样品中抗BV的中和抗体,具有较高的灵敏度和特异性。至关重要的是,本研究中产生的VSV/BVpv可在BSL-2条件下通过gD-nectin-1相互作用以及BV与Vero细胞质膜的直接融合来研究初始进入过程。重要疱疹病毒B(BV)是一种针对人类的高致病性人畜共患病毒。BV属于Simplexvius属,与人类单纯疱疹病毒(HSV)相同的属。与HSV相比,BV的细胞进入机制尚不完全清楚。操纵感染性BV的研究程序应在生物安全级别(BSL)-4设施中进行。由于假病毒提供了一个安全的病毒进入模型,因为它们无法产生传染性子代病毒,我们试图通过修饰BV糖蛋白的表达结构来产生带有BV糖蛋白的假型水泡性口炎病毒(VSV/BVpv),并成功获得了高滴度的VSV/BVpv。这项研究为构建VSV/BVpv及其研究BV感染提供了新的信息。
    Herpes B virus (BV) is a zoonotic virus and belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). BV typically establishes asymptomatic infection in its natural hosts, macaque monkeys. However, in humans, BV infection causes serious neurological diseases and death. As such, BV research can only be conducted in a high containment level facility (i.e., biosafety level [BSL] 4), and the mechanisms of BV entry have not been fully elucidated. In this study, we generated a pseudotyped vesicular stomatitis virus (VSV) expressing BV glycoproteins using G-complemented VSV∆G system, which we named VSV/BVpv. We found that four BV glycoproteins (i.e., gB, gD, gH, and gL) were required for the production of a high-titer VSV/BVpv. Moreover, VSV/BVpv cell entry was dependent on the binding of gD to its cellular receptor nectin-1. Pretreatment of Vero cells with endosomal acidification inhibitors did not affect the VSV/BVpv infection. The result indicated that VSV/BVpv entry occurred by direct fusion with the plasma membrane of Vero cells and suggested that the entry pathway was similar to that of native HSV. Furthermore, we developed a VSV/BVpv-based chemiluminescence reduction neutralization test (CRNT), which detected the neutralization antibodies against BV in macaque plasma samples with high sensitivity and specificity. Crucially, the VSV/BVpv generated in this study can be used under BSL-2 condition to study the initial entry process through gD-nectin-1 interaction and the direct fusion of BV with the plasma membrane of Vero cells.IMPORTANCEHerpes B virus (BV) is a highly pathogenic zoonotic virus against humans. BV belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). By contrast to HSV, cell entry mechanisms of BV are not fully understood. The research procedures to manipulate infectious BV should be conducted in biosafety level (BSL)-4 facilities. As pseudotyped viruses provide a safe viral entry model because of their inability to produce infectious progeny virus, we tried to generate a pseudotyped vesicular stomatitis virus bearing BV glycoproteins (VSV/BVpv) by modification of expression constructs of BV glycoproteins, and successfully obtained VSV/BVpv with a high titer. This study has provided novel information for constructing VSV/BVpv and its usefulness to study BV 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
    核蛋白(N)以其在负链RNA病毒基因组RNA的衣壳化中的功能而闻名,这导致核糖核蛋白的形成,作为病毒转录和复制的模板。然而,病毒感染过程中N蛋白在其他方面的功能尚不清楚。在这项研究中,蛇头囊泡病毒(SHVV)的N蛋白,一种鱼弹状病毒,被证明主要通过K63连接的泛素化被泛素化。我们确定了9个与SHVVN相互作用的宿主E3泛素连接酶,其中7种E3泛素连接酶促进N蛋白的泛素化。进一步调查显示,只有两种E3泛素连接酶,SiahE3泛素蛋白连接酶2(Siah2)和富含亮氨酸的重复序列和含有1(LRSAM1)的无菌α基序,介导的N蛋白的K63连接的泛素化。SHVV感染上调Siah2和LRSAM1的表达,维持了SHVVN的稳定性。Siah2或LRSAM1的过表达促进SHVV复制,而Siah2或LRSAM1的敲低抑制SHVV复制。Siah2或LRSAM1的连接酶结构域的缺失不会影响它们与SHVVN的相互作用,但会减少SHVVN和SHVV复制的K63连接的泛素化。总之,Siah2和LRSAM1介导SHVVN的K63连接的泛素化,以促进SHVV复制,这为负链RNA病毒的N蛋白的作用提供了新的见解。
    目的:病毒蛋白的泛素化在病毒复制中起重要作用。然而,很少研究负链RNA病毒核蛋白(N)的泛素化。这项研究旨在研究鱼弹状病毒SHVV(蛇头囊泡病毒)的N蛋白的泛素化,鉴定相关宿主E3泛素连接酶,并确定SHVVN泛素化和宿主E3泛素连接酶在病毒复制中的作用。我们发现SHVVN主要通过K63连接的泛素化被泛素化,这是由宿主E3泛素连接酶Siah2(SiahE3泛素蛋白连接酶2)和LRSAM1(富含亮氨酸的重复序列和含有1的无菌α基序)介导的。数据表明,Siah2和LRSAM1被SHVV劫持以泛素化N蛋白用于病毒复制,它展示了用于药物设计的新型抗SHVV靶标。
    Nucleoprotein (N) is well known for its function in the encapsidation of the genomic RNAs of negative-strand RNA viruses, which leads to the formation of ribonucleoproteins that serve as templates for viral transcription and replication. However, the function of the N protein in other aspects during viral infection is far from clear. In this study, the N protein of snakehead vesiculovirus (SHVV), a kind of fish rhabdovirus, was proved to be ubiquitinated mainly via K63-linked ubiquitination. We identified nine host E3 ubiquitin ligases that interacted with SHVV N, among which seven E3 ubiquitin ligases facilitated ubiquitination of the N protein. Further investigation revealed that only two E3 ubiquitin ligases, Siah E3 ubiquitin protein ligase 2 (Siah2) and leucine-rich repeat and sterile alpha motif containing 1 (LRSAM1), mediated K63-linked ubiquitination of the N protein. SHVV infection upregulated the expression of Siah2 and LRSAM1, which maintained the stability of SHVV N. Besides, overexpression of Siah2 or LRSAM1 promoted SHVV replication, while knockdown of Siah2 or LRSAM1 inhibited SHVV replication. Deletion of the ligase domain of Siah2 or LRSAM1 did not affect their interactions with SHVV N but reduced the K63-linked ubiquitination of SHVV N and SHVV replication. In summary, Siah2 and LRSAM1 mediate K63-linked ubiquitination of SHVV N to facilitate SHVV replication, which provides novel insights into the role of the N proteins of negative-strand RNA viruses.
    OBJECTIVE: Ubiquitination of viral protein plays an important role in viral replication. However, the ubiquitination of the nucleoprotein (N) of negative-strand RNA viruses has rarely been investigated. This study aimed at investigating the ubiquitination of the N protein of a fish rhabdovirus SHVV (snakehead vesiculovirus), identifying the related host E3 ubiquitin ligases, and determining the role of SHVV N ubiquitination and host E3 ubiquitin ligases in viral replication. We found that SHVV N was ubiquitinated mainly via K63-linked ubiquitination, which was mediated by host E3 ubiquitin ligases Siah2 (Siah E3 ubiquitin protein ligase 2) and LRSAM1 (leucine-rich repeat and sterile alpha motif containing 1). The data suggested that Siah2 and LRSAM1 were hijacked by SHVV to ubiquitinate the N protein for viral replication, which exhibited novel anti-SHVV targets for drug design.
    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:为了研究基于紫外线(UV)灭活的重组水泡性口炎(rVSV)的载体的免疫反应和保护能力,该载体表达了由四个拷贝的流感基质2蛋白胞外域(tM2e)和埃博拉糖蛋白(EΔM)的树突状细胞(DC)靶向域组成的融合蛋白,(rVSV-EΔM-tM2e)。
    方法:在我们之前的研究中,我们证明了rVSV-EΔM-tM2e可诱导针对流感M2e的强大免疫反应,并可抵抗H1N1和H3N2毒株的致死性攻击。这里,我们使用紫外线灭活rVSV-EΔM-tM2e,并测试了其在BALB/c小鼠中的免疫原性和对小鼠适应的H1N1流感攻击的保护作用。使用酶联免疫吸附测定(ELISA)和抗体依赖性细胞毒性(ADCC),人类特有的流感抗M2e免疫反应,对诱导的禽流感和猪流感毒株进行了表征。同样,使用荧光激活细胞分选(FACS)分析研究了在识别细胞表面上的M2e抗原时诱导的抗M2e免疫应答的特异性。
    结果:与实时减毒rVSV-EΔM-tM2e一样,紫外线灭活的rVSV-EΔM-tM2e对来自不同宿主株的不同流感M2e具有高度免疫原性,包括人类,猪,和鸟类,并保护小鼠免受H1N1流感攻击。FACS分析表明,诱导的免疫反应可以识别来自人类的流感M2抗原,猪和禽流感毒株。此外,rVSV-EΔM-tM2e还诱导针对来自不同宿主株的流感M2e的ADCC活性。
    结论:这些发现表明UV灭活的rVSV-EΔM-tM2e可用作抗流感病毒的灭活疫苗。
    BACKGROUND: To investigate the immune responses and protection ability of ultraviolet light (UV)-inactivated recombinant vesicular stomatitis (rVSV)-based vectors that expressed a fusion protein consisting of four copies of the influenza matrix 2 protein ectodomain (tM2e) and the Dendritic Cell (DC)-targeting domain of the Ebola Glycoprotein (EΔM), (rVSV-EΔM-tM2e).
    METHODS: In our previous study, we demonstrated the effectiveness of rVSV-EΔM-tM2e to induce robust immune responses against influenza M2e and protect against lethal challenges from H1N1 and H3N2 strains. Here, we used UV to inactivate rVSV-EΔM-tM2e and tested its immunogenicity and protection in BALB/c mice from a mouse-adapted H1N1 influenza challenge. Using Enzyme-Linked Immunosorbent Assay (ELISA) and Antibody-Dependent Cellular Cytotoxicity (ADCC), the influenza anti-M2e immune responses specific to human, avian and swine influenza strains induced were characterized. Likewise, the specificity of the anti-M2e immune responses induced in recognizing M2e antigen on the surface of the cell was investigated using Fluorescence-Activated Cell Sorting (FACS) analysis.
    RESULTS: Like the live attenuated rVSV-EΔM-tM2e, the UV-inactivated rVSV-EΔM-tM2e was highly immunogenic against different influenza M2e from strains of different hosts, including human, swine, and avian, and protected against influenza H1N1 challenge in mice. The FACS analysis demonstrated that the induced immune responses can recognize influenza M2 antigens from human, swine and avian influenza strains. Moreover, the rVSV-EΔM-tM2e also induced ADCC activity against influenza M2e from different host strains.
    CONCLUSIONS: These findings suggest that UV-inactivated rVSV-EΔM-tM2e could be used as an inactivated vaccine against influenza viruses.
    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