Antiviral Restriction Factors

  • 文章类型: Journal Article
    不同的宿主蛋白靶向不同的HIV蛋白并拮抗它们的功能,取决于HIV生命周期的阶段和感染阶段。同时,HIV蛋白还靶向和拮抗各种不同的宿主蛋白以促进HIV在宿主细胞内的复制。前面相当具体的知识领域在艾滋病毒的发病机理,然而,仍然没有得到足够的理解。因此,我们建议,在这篇评论文章中,研究和讨论对抗那些直接导致HIV感染性增加的宿主限制性蛋白的HIV蛋白。我们详细阐述了拮抗宿主细胞蛋白以促进HIV复制的HIV蛋白,从而感染艾滋病毒。我们研究了Nef的功能和机制,Vif,Vpu,Env,Vpr,和Vpx抵消宿主蛋白,如Ser5,PSGL-1,IFITMS,A3G,Tetherin,GBP5,SAMHD1,STING,HUSH,REAF,和TET2增加艾滋病毒的传染性。Nef拮抗三种宿主蛋白,viz.,Ser5、PSGL1和IFITIM,而Vpx还拮抗三种宿主限制因子,viz.,SAMHD1,STING,和HUSH复合体;因此,这些蛋白可能是HIV感染治疗干预的潜在候选蛋白.Tetherin是Vpu和Env的目标,PSGL1是Nef和Vpu的目标,而Ser5由Nef和Env蛋白靶向。最后,还提出了结论性意见和未来观点。
    Different host proteins target different HIV proteins and antagonize their functions, depending on the stage of the HIV life cycle and the stage of infection. Concurrently, HIV proteins also target and antagonize various different host proteins to facilitate HIV replication within host cells. The preceding quite specific area of knowledge in HIV pathogenesis, however, remains insufficiently understood. We therefore propose, in this review article, to examine and discuss the HIV proteins that counteract those host restriction proteins which results directly in increased infectivity of HIV. We elaborate on HIV proteins that antagonize host cellular proteins to promote HIV replication, and thus HIV infection. We examine the functions and mechanisms via which Nef, Vif, Vpu, Env, Vpr, and Vpx counteract host proteins such as Ser5, PSGL-1, IFITMS, A3G, tetherin, GBP5, SAMHD1, STING, HUSH, REAF, and TET2 to increase HIV infectivity. Nef antagonizes three host proteins, viz., Ser5, PSGL1, and IFITIMs, while Vpx also antagonizes three host restriction factors, viz., SAMHD1, STING, and HUSH complex; therefore, these proteins may be potential candidates for therapeutic intervention in HIV infection. Tetherin is targeted by Vpu and Env, PSGL1 is targeted by Nef and Vpu, while Ser5 is targeted by Nef and Env proteins. Finally, conclusive remarks and future perspectives are also presented.
    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
    病毒感染所施加的进化压力导致了具有有效抗病毒活性的各种细胞蛋白的发展。其中一些被称为抗病毒限制因子。含TRIpartnal基序的蛋白5α(TRIM5α)是逆转录病毒的经过充分研究的限制因子,在保护特定慢病毒的跨灵长类动物传播方面表现出病毒和宿主物种特异性功能。这种特异性是通过主要在其C端B30.2/PRYSPRY结构域内的阳性选择在宿主基因水平上实现的,负责逆转录病毒衣壳的高度特异性识别。然而,最近的工作挑战了这种范式,证明TRIM5α是逆转录元件以及系统发育上不同病毒家族的限制因子,通过B30.2/PRYSPRY识别病毒基因产物起类似作用。该抗病毒活性谱提出了关于该蛋白质的遗传和结构可塑性作为识别潜在的多种病毒分子模式的介体的问题。这篇综述强调了B30.2/PRYSPRY域对逆转录病毒的动态进化足迹,同时探索了TRIM5α的全部其他域所赋予的指导“特异性”,这可能是其最近发现的滥交。
    The evolutionary pressures exerted by viral infections have led to the development of various cellular proteins with potent antiviral activities, some of which are known as antiviral restriction factors. TRIpartite Motif-containing protein 5 alpha (TRIM5α) is a well-studied restriction factor of retroviruses that exhibits virus- and host-species-specific functions in protecting against cross-primate transmission of specific lentiviruses. This specificity is achieved at the level of the host gene through positive selection predominantly within its C-terminal B30.2/PRYSPRY domain, which is responsible for the highly specific recognition of retroviral capsids. However, more recent work has challenged this paradigm, demonstrating TRIM5α as a restriction factor for retroelements as well as phylogenetically distinct viral families, acting similarly through the recognition of viral gene products via B30.2/PRYSPRY. This spectrum of antiviral activity raises questions regarding the genetic and structural plasticity of this protein as a mediator of the recognition of a potentially diverse array of viral molecular patterns. This review highlights the dynamic evolutionary footprint of the B30.2/PRYSPRY domain in response to retroviruses while exploring the guided \'specificity\' conferred by the totality of TRIM5α\'s additional domains that may account for its recently identified promiscuity.
    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
    含有无菌α基序结构域的蛋白9和9样蛋白(SAMD9/9L)与人类危及生命的遗传疾病有关,并且是痘病毒的限制因子。然而,它们的细胞功能和抗病毒作用的程度知之甚少。这里,我们发现干扰素刺激的人SAMD9L在复制的后期限制了HIV-1,在转录后和早熟阶段,影响病毒翻译,可能,内体运输。令人惊讶的是,模拟SAMD9产生了相反的效果,增强HIV-1。更广泛地说,我们发现SAMD9L限制灵长类慢病毒,但不是γ逆转录病毒(MLV),Nor2RNA病毒(沙粒病毒MOPV和弹状病毒VSV)。使用SAMD9L的结构建模和诱变,我们确定了人类和啮齿动物SAMD9L限制HIV-1所必需的保守的Schlafen样活性位点。通过测试SAMD9L相关自身炎症性疾病患者的功能增益组成型活性变异,我们确定SAMD9L的致病功能也取决于Schlafen样活性位点。最后,我们发现组成型活性SAMD9L强烈抑制HIV,MLV,and,在较小程度上,MOPV。这表明SAMD9L的病毒特异性作用可能涉及其差异激活/传感以及病毒逃避SAMD9L限制的能力。总的来说,我们的研究将SAMD9L从细胞自主免疫中鉴定为HIV-1抗病毒因子,并破译了翻译抑制的潜在宿主决定簇.这提供了针对病毒感染和遗传疾病的新联系和治疗途径。
    Sterile alpha motif domain-containing proteins 9 and 9-like (SAMD9/9L) are associated with life-threatening genetic diseases in humans and are restriction factors of poxviruses. Yet, their cellular function and the extent of their antiviral role are poorly known. Here, we found that interferon-stimulated human SAMD9L restricts HIV-1 in the late phases of replication, at the posttranscriptional and prematuration steps, impacting viral translation and, possibly, endosomal trafficking. Surprisingly, the paralog SAMD9 exerted an opposite effect, enhancing HIV-1. More broadly, we showed that SAMD9L restricts primate lentiviruses, but not a gammaretrovirus (MLV), nor 2 RNA viruses (arenavirus MOPV and rhabdovirus VSV). Using structural modeling and mutagenesis of SAMD9L, we identified a conserved Schlafen-like active site necessary for HIV-1 restriction by human and a rodent SAMD9L. By testing a gain-of-function constitutively active variant from patients with SAMD9L-associated autoinflammatory disease, we determined that SAMD9L pathogenic functions also depend on the Schlafen-like active site. Finally, we found that the constitutively active SAMD9L strongly inhibited HIV, MLV, and, to a lesser extent, MOPV. This suggests that the virus-specific effect of SAMD9L may involve its differential activation/sensing and the virus ability to evade from SAMD9L restriction. Overall, our study identifies SAMD9L as an HIV-1 antiviral factor from the cell autonomous immunity and deciphers host determinants underlying the translational repression. This provides novel links and therapeutic avenues against viral infections and genetic 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
    背景:通过宿主模式识别受体检测病毒可诱导I型干扰素(IFN)和IFN刺激基因(ISG)的表达,抑制病毒复制。许多研究已经将HIV-1描述为体外先天免疫的不良激活剂。病毒衣壳在这种免疫逃避中的确切作用尚未完全了解。
    结果:为了更好地理解HIV-1衣壳在感知中的作用,我们测试了通过共表达截短的Gag来制造HIV-1的效果,该Gag编码衣壳的前107个氨基酸与荧光素酶或GFP融合,和野生型Gag-pol一起.我们发现与野生型HIV-1不同,用野生型和与荧光素酶或GFP融合的截短Gag的混合物产生的病毒颗粒在THP-1细胞和巨噬细胞中诱导有效的IFN应答。Gag融合HIV-1的先天性免疫激活依赖于逆转录和DNA传感器cGAS,提示通过病毒DNA激活IFN应答。进一步的研究表明,将Gag-荧光素酶/GFP融合蛋白掺入病毒颗粒中,该病毒颗粒与野生型Gag裂解中的细微缺陷以及饱和限制因子TRIM5α的能力降低有关,可能是由于异常颗粒形成。我们认为Gag融合蛋白的表达会干扰野生型Gag的正确裂解和成熟,产生的病毒颗粒不能有效地屏蔽病毒DNA从先天传感器,包括cGAS检测。
    结论:这些数据强调了衣壳在先天逃避中的关键作用,并支持越来越多的文献,即Gag裂解和衣壳形成的破坏会诱导病毒DNA和cGAS依赖性先天免疫应答。这些数据一起证明了衣壳的保护作用,并表明衣壳靶向抗病毒药物的抗病毒活性可能受益于体内先天和适应性免疫的增强。
    BACKGROUND: Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood.
    RESULTS: To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS.
    CONCLUSIONS: These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity 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
    目的:非洲猪瘟(ASF)是一种高度传染性和急性出血性病毒性疾病,在家猪中死亡率接近100%。ASF病毒(ASFV)它是阿法尔病毒科的唯一成员,是一种非常复杂和大小的dsDNA病毒,编码150多种蛋白质。目前,没有针对ASFV的疫苗。ASFVCP204L代表感染早期表达最丰富的病毒蛋白,在调节ASFV复制中起重要作用。然而,ASFVCP204L与宿主蛋白相互作用影响ASFV复制的机制尚不清楚.在这项研究中,我们证明细胞蛋白SNX32与CP204L相互作用,并通过上调自噬相关蛋白RAB1B降解CP204L.总之,本研究将有助于我们了解CP204L与宿主在感染时的相互作用机制,并为疫苗和抗病毒药物的开发提供新的见解。
    African swine fever virus (ASFV) causes a highly contagious and deadly disease in domestic pigs and European wild boars, posing a severe threat to the global pig industry. ASFV CP204L, a highly immunogenic protein, is produced during the early stages of ASFV infection. However, the impact of CP204L protein-interacting partners on the outcome of ASFV infection is poorly understood. To accomplish this, coimmunoprecipitation and mass spectrometry analysis were conducted in ASFV-infected porcine alveolar macrophages (PAMs). We have demonstrated that sorting nexin 32 (SNX32) is a CP204L-binding protein and that CP204L interacted and colocalized with SNX32 in ASFV-infected PAMs. ASFV growth and replication were promoted by silencing SNX32 and suppressed by overexpressing SNX32. SNX32 degraded CP204L by recruiting the autophagy-related protein Ras-related protein Rab-1b (RAB1B). RAB1B overexpression inhibited ASFV replication, while knockdown of RAB1B had the opposite effect. Additionally, RAB1B, SNX32, and CP204L formed a complex upon ASFV infection. Taken together, this study demonstrates that SNX32 antagonizes ASFV growth and replication by recruiting the autophagy-related protein RAB1B. This finding extends our understanding of the interaction between ASFV CP204L and its host and provides new insights into exploring the relationship between ASFV infection and autophagy.IMPORTANCEAfrican swine fever (ASF) is a highly contagious and acute hemorrhagic viral disease with a high mortality near 100% in domestic pigs. ASF virus (ASFV), which is the only member of the family Asfarviridae, is a dsDNA virus of great complexity and size, encoding more than 150 proteins. Currently, there are no available vaccines against ASFV. ASFV CP204L represents the most abundantly expressed viral protein early in infection and plays an important role in regulating ASFV replication. However, the mechanism by which the interaction between ASFV CP204L and host proteins affects ASFV replication remains unclear. In this study, we demonstrated that the cellular protein SNX32 interacted with CP204L and degraded CP204L by upregulating the autophagy-related protein RAB1B. In summary, this study will help us understand the interaction mechanism between CP204L and its host upon infection and provide new insights for the development of vaccines and antiviral drugs.
    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
    单纯疱疹病毒1(HSV-1)是一种神经亲病毒,在神经元细胞体中仍然潜伏,但在个体的整个生命中重新激活,导致严重的不良反应,如单纯疱疹性脑炎(HSE)。最近,它也与阿尔茨海默病(AD)的病因有关。缺乏有效的疫苗和许多耐药变体的出现要求开发新的抗病毒药物来应对HSV-1感染。宿主靶向抗病毒药物(HTAs)最近已成为有希望的抗病毒化合物,可作用于病毒复制所必需的宿主细胞因子。在这里,我们展示了一类新的针对肽基精氨酸脱亚胺酶(PAD)的HTA,一个催化蛋白质瓜氨酸化的钙依赖性酶家族,对HSV-1表现出显著的抑制活性。此外,我们显示HSV-1感染通过转录激活三种PAD亚型:PAD2,PAD3和PAD4导致蛋白瓜氨酸化增强.有趣的是,通过特定药物或siRNA消除PAD3显著抑制HSV-1复制。最后,对瓜氨酸的分析揭示了细胞和病毒蛋白脱亚胺水平的显著变化,干扰素(IFN)诱导蛋白IFIT1和IFIT2是脱亚胺作用最严重的蛋白之一。由于IFIT1和IFIT2的遗传耗竭强烈增强了HSV-1的生长,我们认为病毒诱导的IFIT1和2瓜氨酸化是一种高效的HSV-1逃避宿主抗病毒抗性的机制。总的来说,我们的发现指出瓜氨酸化在颠覆细胞对病毒感染的反应中的关键作用,并证明PAD抑制剂在体外有效抑制HSV-1感染,这可能为它们重新用作HSV-1抗病毒药物提供了理由。
    Herpes simplex virus 1 (HSV-1) is a neurotropic virus that remains latent in neuronal cell bodies but reactivates throughout an individual\'s life, causing severe adverse reactions, such as herpes simplex encephalitis (HSE). Recently, it has also been implicated in the etiology of Alzheimer\'s disease (AD). The absence of an effective vaccine and the emergence of numerous drug-resistant variants have called for the development of new antiviral agents that can tackle HSV-1 infection. Host-targeting antivirals (HTAs) have recently emerged as promising antiviral compounds that act on host-cell factors essential for viral replication. Here we show that a new class of HTAs targeting peptidylarginine deiminases (PADs), a family of calcium-dependent enzymes catalyzing protein citrullination, exhibits a marked inhibitory activity against HSV-1. Furthermore, we show that HSV-1 infection leads to enhanced protein citrullination through transcriptional activation of three PAD isoforms: PAD2, PAD3, and PAD4. Interestingly, PAD3-depletion by specific drugs or siRNAs dramatically inhibits HSV-1 replication. Finally, an analysis of the citrullinome reveals significant changes in the deimination levels of both cellular and viral proteins, with the interferon (IFN)-inducible proteins IFIT1 and IFIT2 being among the most heavily deiminated ones. As genetic depletion of IFIT1 and IFIT2 strongly enhances HSV-1 growth, we propose that viral-induced citrullination of IFIT1 and 2 is a highly efficient HSV-1 evasion mechanism from host antiviral resistance. Overall, our findings point to a crucial role of citrullination in subverting cellular responses to viral infection and demonstrate that PAD inhibitors efficiently suppress HSV-1 infection in vitro, which may provide the rationale for their repurposing as HSV-1 antiviral drugs.
    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
    静止的人类造血干细胞(HSC)由于其保留的干性和再增殖能力,是基因治疗应用的理想靶标。但是由于它们对基因操纵的抵抗力,它们没有被广泛利用。我们在这里报告了一种慢病毒转导方案的发展,该方案克服了长期重新繁殖的静止HSC中的这种抗性,允许他们有效的遗传操作。机械上,发现静态HSC的慢病毒载体转导被限制在载体进入水平和有限的嘧啶库。通过在慢病毒载体转导期间组合添加环孢菌素H(CsH)和脱氧核苷(dN)克服了这些限制。与标准离体培养的对照相比,临床相关的转导水平与更高的长期再增殖HSC的多克隆移植配对。这些发现确定了限制静止HSC转导的细胞内在屏障,并提供了克服它们的方法。为未受刺激的HSC基因工程铺平了道路。
    Quiescent human hematopoietic stem cells (HSC) are ideal targets for gene therapy applications due to their preserved stemness and repopulation capacities; however, they have not been exploited extensively because of their resistance to genetic manipulation. We report here the development of a lentiviral transduction protocol that overcomes this resistance in long-term repopulating quiescent HSC, allowing their efficient genetic manipulation. Mechanistically, lentiviral vector transduction of quiescent HSC was found to be restricted at the level of vector entry and by limited pyrimidine pools. These restrictions were overcome by the combined addition of cyclosporin H (CsH) and deoxynucleosides (dNs) during lentiviral vector transduction. Clinically relevant transduction levels were paired with higher polyclonal engraftment of long-term repopulating HSC as compared with standard ex vivo cultured controls. These findings identify the cell-intrinsic barriers that restrict the transduction of quiescent HSC and provide a means to overcome them, paving the way for the genetic engineering of unstimulated HSC.
    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
    哺乳动物的先天免疫系统是由相互作用的蛋白质组成的复杂网络,它们共同构成了传染性病原体进入的第一道屏障。E3-泛素连接酶三方基序(TRIM)家族的基因已被证明通过限制不同逆转录病毒物种的活性在先天免疫系统中起重要作用。例如,TRIM5和TRIM22都与HIV限制有关,被认为是哺乳动物抗逆转录病毒机制的关键部分。我们对正选择的分析证实了这些基因对某些哺乳动物群体的重大意义。然而,我们还表明,许多物种完全缺乏TRIM5和TRIM22。通过分析大量的哺乳动物基因组,在这里,我们提供了这些基因在异教徒中进化的第一个全面观点,显示TRIM基因的积累模式在哺乳动物中不同。我们的数据表明,这些差异是由异教徒免疫系统的进化可塑性引起的,他们已经适应了使用不同的策略来对抗逆转录病毒感染。总之,我们的结果提供了对代表性限制因子家族的不同进化的见解,突出了先天免疫系统适应性和特质进化的一个例子。
    The innate immune system of mammals is formed by a complex web of interacting proteins, which together constitute the first barrier of entry for infectious pathogens. Genes from the E3-ubiquitin ligase tripartite motif (TRIM) family have been shown to play an important role in the innate immune system by restricting the activity of different retrovirus species. For example, TRIM5 and TRIM22 have both been associated with HIV restriction and are regarded as crucial parts of the antiretroviral machinery of mammals. Our analyses of positive selection corroborate the great significance of these genes for some groups of mammals. However, we also show that many species lack TRIM5 and TRIM22 altogether. By analyzing a large number of mammalian genomes, here we provide the first comprehensive view of the evolution of these genes in eutherians, showcasing that the pattern of accumulation of TRIM genes has been dissimilar across mammalian orders. Our data suggest that these differences are caused by the evolutionary plasticity of the immune system of eutherians, which have adapted to use different strategies to combat retrovirus infections. Altogether, our results provide insights into the dissimilar evolution of a representative family of restriction factors, highlighting an example of adaptive and idiosyncratic evolution in the innate immune system.
    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
    人腺病毒物种C型5(HAdV-C5)早期1B区55kDa(E1B-55K)蛋白是一种多功能蛋白,可通过各种机制促进病毒复制和腺病毒介导的细胞转化,主要抵消宿主的内在和先天免疫。这些包括利用宿主细胞泛素和小泛素样修饰剂(SUMO)缀合机制来调节抗病毒细胞限制因子的翻译后活性。然而,尽管在这一领域取得了重大进展,迄今为止,管理这些过程的一些潜在机制仍未得到确认。这里,我们在基于细胞培养物(SILAC)的定量SUMO蛋白质组学中通过氨基酸进行稳定同位素标记,以更好地了解E1B-55K介导的宿主细胞调节和腺病毒感染的一般细胞后果.我们评估了野生型HAdV-C5或E1B-55K缺失突变体感染过程中细胞蛋白的丰度变化和SUMO2偶联蛋白质组变化。我们提供的证据表明,SUMO化蛋白质组的变化有可能调节DNA损伤反应,细胞周期控制,染色质组装,和基因转录,并将这些数据作为研究界的资源。引人注目的是,我们确定了一个依赖SUMO的,泛素介导的一些SUMO底物的降解机制,提示E1B-55K可能使用多种机制来改变限制性细胞通路的活性。IMPORTANCEHuman腺病毒(HAdV)通常会引起上呼吸道和胃肠道的轻度和自限性疾病,但对免疫功能低下的患者和儿童构成严重的风险。此外,它们被广泛用作疫苗载体和基于载体的基因治疗方法。因此,彻底表征HAdV基因产物,尤其是HAdV毒力因子至关重要。早期1B区55kDa蛋白(E1B-55K)是一种多功能HAdV编码的癌蛋白,参与促进病毒复制和细胞转化的各种病毒和细胞途径。我们分析了SUMO化的E1B-55K依赖性,翻译后的蛋白质修饰,在感染的细胞中使用定量蛋白质组学。我们发现HAdV增加整体细胞SUMO化,并且这种增加的SUMO化可以靶向影响HAdV复制的抗病毒细胞途径。此外,我们表明E1B-55K协调某些细胞抗病毒因子的SUMO依赖性降解。这些结果再次强调了E1B-55K在生产性HAdV感染中病毒和细胞蛋白调节中的关键作用。
    Human adenoviruses (HAdVs) generally cause mild and self-limiting diseases of the upper respiratory and gastrointestinal tracts but pose a serious risk to immunocompromised patients and children. Moreover, they are widely used as vectors for vaccines and vector-based gene therapy approaches. It is therefore vital to thoroughly characterize HAdV gene products and especially HAdV virulence factors. Early region 1B 55 kDa protein (E1B-55K) is a multifunctional HAdV-encoded oncoprotein involved in various viral and cellular pathways that promote viral replication and cell transformation. We analyzed the E1B-55K dependency of SUMOylation, a post-translational protein modification, in infected cells using quantitative proteomics. We found that HAdV increases overall cellular SUMOylation and that this increased SUMOylation can target antiviral cellular pathways that impact HAdV replication. Moreover, we showed that E1B-55K orchestrates the SUMO-dependent degradation of certain cellular antiviral factors. These results once more emphasize the key role of E1B-55K in the regulation of viral and cellular proteins in productive HAdV infections.
    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
    细胞内先天免疫涉及特异性抑制感染病毒的共同进化的抗病毒限制因子。研究这些限制增加了我们对病毒复制的理解,宿主-病原体相互作用,和发病机制,并代表新型抗病毒治疗的潜在目标。慢病毒限制2(Lv2)被鉴定为HIV-2的未定位的早期限制,后来显示也限制了HIV-1和猿猴免疫缺陷病毒。Lv2易感性的病毒决定簇已被定位到HIV-1和HIV-2中的包膜和衣壳蛋白,以及HIV-1中的病毒蛋白R(Vpr),并且似乎取决于细胞进入机制。全基因组筛选确定了几种可能的宿主因子,包括聚合酶相关因子1(PAF1)和人类沉默中心(HUSH)复合物的成员,和新表征的核前mRNA结构域包含2(RPRD2)的调控。随后,RPRD2(或RNA相关的早期抗病毒因子)已被证明在T细胞激活后上调,在骨髓细胞中高度表达,结合病毒逆转录,并有效限制HIV-1感染。RPRD2也与HIV-1Vpr结合,并在逆转录时被蛋白酶体靶向降解,提示RPRD2阻碍逆转录,Vpr靶向克服了这种阻滞。RPRD2主要定位于细胞核并与RNA结合,DNA,和DNA:RNA杂种。最近,RPRD2已显示出负调节全基因组转录并与抑制HIV转录的HUSH和PAF1复合物相互作用,并与维持HIV潜伏期有关。在这次审查中,我们研究了Lv2限制和RPRD2的抗病毒作用,并考虑了潜在的作用机制.
    Intracellular innate immunity involves co-evolved antiviral restriction factors that specifically inhibit infecting viruses. Studying these restrictions has increased our understanding of viral replication, host-pathogen interactions, and pathogenesis, and represent potential targets for novel antiviral therapies. Lentiviral restriction 2 (Lv2) was identified as an unmapped early-phase restriction of HIV-2 and later shown to also restrict HIV-1 and simian immunodeficiency virus. The viral determinants of Lv2 susceptibility have been mapped to the envelope and capsid proteins in both HIV-1 and HIV-2, and also viral protein R (Vpr) in HIV-1, and appears dependent on cellular entry mechanism. A genome-wide screen identified several likely contributing host factors including members of the polymerase-associated factor 1 (PAF1) and human silencing hub (HUSH) complexes, and the newly characterized regulation of nuclear pre-mRNA domain containing 2 (RPRD2). Subsequently, RPRD2 (or RNA-associated early-stage antiviral factor) has been shown to be upregulated upon T cell activation, is highly expressed in myeloid cells, binds viral reverse transcripts, and potently restricts HIV-1 infection. RPRD2 is also bound by HIV-1 Vpr and targeted for degradation by the proteasome upon reverse transcription, suggesting RPRD2 impedes reverse transcription and Vpr targeting overcomes this block. RPRD2 is mainly localized to the nucleus and binds RNA, DNA, and DNA:RNA hybrids. More recently, RPRD2 has been shown to negatively regulate genome-wide transcription and interact with the HUSH and PAF1 complexes which repress HIV transcription and are implicated in maintenance of HIV latency. In this review, we examine Lv2 restriction and the antiviral role of RPRD2 and consider potential mechanism(s) 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