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
    大脑对感染和炎症引起的损伤高度敏感。单纯疱疹病毒1(HSV-1)是一种嗜神经病毒,是引起单纯疱疹病毒脑炎的原因3。尚不清楚神经元特异性抗病毒因子是否控制病毒复制以防止感染和过度炎症反应,从而保护大脑。在这里,我们使用全基因组CRISPR筛选将TMEFF1鉴定为HSV-1限制因子。TMEFF1在中枢神经系统的神经元中特异性表达,不受I型干扰素的调节,控制病毒感染的最著名的先天抗病毒系统。干细胞来源的人神经元中TMEFF1的耗尽导致HSV-1感染后病毒复制和神经元死亡升高。TMEFF1通过与nectin-1和非肌肉肌球蛋白重链IIA和IIB的相互作用,在病毒进入水平上阻断了HSV-1复制周期,它们是病毒-细胞结合和病毒-细胞融合的核心蛋白,分别4-6.值得注意的是,Tmeff1-/-小鼠在脑中表现出对HSV-1感染的易感性增加,但在外周却没有。在大脑中,在神经元中特别观察到病毒载量升高。我们的研究将TMEFF1确定为预防中枢神经系统中HSV-1复制所必需的神经元特异性限制因子。
    The brain is highly sensitive to damage caused by infection and inflammation1,2. Herpes simplex virus 1 (HSV-1) is a neurotropic virus and the cause of herpes simplex encephalitis3. It is unknown whether neuron-specific antiviral factors control virus replication to prevent infection and excessive inflammatory responses, hence protecting the brain. Here we identify TMEFF1 as an HSV-1 restriction factor using genome-wide CRISPR screening. TMEFF1 is expressed specifically in neurons of the central nervous system and is not regulated by type I interferon, the best-known innate antiviral system controlling virus infections. Depletion of TMEFF1 in stem-cell-derived human neurons led to elevated viral replication and neuronal death following HSV-1 infection. TMEFF1 blocked the HSV-1 replication cycle at the level of viral entry through interactions with nectin-1 and non-muscle myosin heavy chains IIA and IIB, which are core proteins in virus-cell binding and virus-cell fusion, respectively4-6. Notably, Tmeff1-/- mice exhibited increased susceptibility to HSV-1 infection in the brain but not in the periphery. Within the brain, elevated viral load was observed specifically in neurons. Our study identifies TMEFF1 as a neuron-specific restriction factor essential for prevention of HSV-1 replication in the central nervous system.
    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
    TRIM家族与膜相关,以及它在进步中的参与,增长,和各种癌症类型的发展已经被广泛研究。然而,在肝细胞癌(HCC)方面,TRIM5基因在该家族中所起的作用尚未得到很大程度的探索.从癌症基因组图谱(TCGA)数据库中提取与mRNA表达相关的患者数据和诊断为HCC的个体的存活率。UALCAN用于检查TRIM5表达与临床病理特征之间的潜在联系。此外,进行了差异表达基因(DEGs)的富集分析,以破译TRIM5在HCC中的功能和机制。TCGA和TIMER2.0数据库中的数据用于探索TRIM5与HCC中免疫浸润之间的相关性。进行WGCNA作为评估TRIM5相关共表达基因的手段。“OncoPredict”R软件包也用于研究TRIM5与药物敏感性之间的关联。最后,qRT-PCR,蛋白质印迹(WB)和免疫组织化学(IHC)用于探讨TRIM5的差异表达及其在HCC中的临床意义。根据从体外实验中获得的结果,TRIM5的mRNA和蛋白质水平在HCC组织中显示出显着的上调。值得注意的是,发现TRIM5表达水平与多种免疫细胞的浸润具有强相关性,并且显示与几种免疫检查点抑制剂的正相关性。TRIM5表达对HCC患者也显示出有希望的临床预后价值。
    The TRIM family is associated with the membrane, and its involvement in the progression, growth, and development of various cancer types has been researched extensively. However, the role played by the TRIM5 gene within this family has yet to be explored to a great extent in terms of hepatocellular carcinoma (HCC). The data of patients relating to mRNA expression and the survival rate of individuals diagnosed with HCC were extracted from The Cancer Genome Atlas (TCGA) database. UALCAN was employed to examine the potential link between TRIM5 expression and clinicopathological characteristics. In addition, enrichment analysis of differentially expressed genes (DEGs) was conducted as a means of deciphering the function and mechanism of TRIM5 in HCC. The data in the TCGA and TIMER2.0 databases was utilized to explore the correlation between TRIM5 and immune infiltration in HCC. WGCNA was performed as a means of assessing TRIM5-related co-expressed genes. The \"OncoPredict\" R package was also used for investigating the association between TRIM5 and drug sensitivity. Finally, qRT-PCR, Western blotting (WB) and immunohistochemistry (IHC) were employed for exploring the differential expression of TRIM5 and its clinical relevance in HCC. According to the results that were obtained from the vitro experiments, mRNA and protein levels of TRIM5 demonstrated a significant upregulation in HCC tissues. It is notable that TRIM5 expression levels were found to have a strong association with the infiltration of diverse immune cells and displayed a positive correlation with several immune checkpoint inhibitors. The TRIM5 expression also displayed promising clinical prognostic value for HCC patients.
    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
    严重发热伴血小板减少综合征(SFTS)是一种新兴的蜱传出血热疾病,病死率高达10%-20%。仍然缺乏疫苗或特定的治疗措施。人干扰素诱导跨膜蛋白3(hIFITM3)是一种靶向病毒进入的广谱抗病毒因子。然而,hIFITM3对SFTS病毒(SFTSV)的抗病毒活性及其作用机制尚不清楚.在这里,我们证明了内源性IFITM3对SFTSV感染具有保护作用,并参与了Ⅰ型和Ⅲ型干扰素(IFNs)的抗SFTSV作用。IFITM3过表达通过阻断Gn/Gc介导的病毒进入和融合而表现出抗SFTSV功能。进一步的研究表明,IFITM3直接结合SFTSVGc,其膜内结构域(IMD)负责这种相互作用和限制SFTSV进入。IMD上两个相邻半胱氨酸的突变削弱了IFITM3-Gc相互作用并减弱了IFITM3的抗病毒活性,表明IFITM3-Gc相互作用可能部分介导了SFTSV进入的抑制。总的来说,我们的数据首次表明hIFITM3在IFN介导的抗SFTSV反应中起关键作用,并揭示了IFITM3限制SFTSV感染的新机制,强调临床干预SFTS疾病的潜力。
    Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne hemorrhagic fever disease with high fatality rate of 10%-20%. Vaccines or specific therapeutic measures remain lacking. Human interferon inducible transmembrane protein 3 (hIFITM3) is a broad-spectrum antiviral factor targeting viral entry. However, the antiviral activity of hIFITM3 against SFTS virus (SFTSV) and the functional mechanism of IFITM3 remains unclear. Here we demonstrate that endogenous IFITM3 provides protection against SFTSV infection and participates in the anti-SFTSV effect of type Ⅰ and Ⅲ interferons (IFNs). IFITM3 overexpression exhibits anti-SFTSV function by blocking Gn/Gc-mediated viral entry and fusion. Further studies showed that IFITM3 binds SFTSV Gc directly and its intramembrane domain (IMD) is responsible for this interaction and restriction of SFTSV entry. Mutation of two neighboring cysteines on IMD weakens IFITM3-Gc interaction and attenuates the antiviral activity of IFITM3, suggesting that IFITM3-Gc interaction may partly mediate the inhibition of SFTSV entry. Overall, our data demonstrate for the first time that hIFITM3 plays a critical role in the IFNs-mediated anti-SFTSV response, and uncover a novel mechanism of IFITM3 restriction of SFTSV infection, highlighting the potential of clinical intervention on SFTS disease.
    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
    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
    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)

  • 文章类型: Journal Article
    马传染性贫血病毒(EIAV)是逆转录病毒科中慢病毒属的成员,被认为是HIV/AIDS研究的动物模型。一种减毒EIAV疫苗,它是在1970年代通过经典的连续通道技术成功开发的,是迄今为止被广泛使用的第一个也是唯一一个慢病毒疫苗。限制因子是通过干扰病毒复制周期中的各种关键步骤来提供针对病毒复制和传播的早期防线的细胞蛋白质。然而,病毒已经进化出通过适应克服这些宿主障碍的特定机制。病毒和限制因子之间的斗争实际上是病毒复制过程的自然部分,已在人类免疫缺陷病毒1型(HIV-1)中进行了充分的研究。EIAV具有所有慢病毒中最简单的基因组组成,使它成为一个有趣的主题,了解病毒如何利用其有限的病毒蛋白来克服限制因素。在这次审查中,我们总结了当前有关马限制因子与EIAV之间相互作用的文献。马限制因子的特征和EIAV抵消限制的机制表明,慢病毒采用多种策略来抵消先天免疫限制。此外,我们提出了我们对限制因子是否诱导减毒EIAV疫苗表型改变的见解.
    Equine infectious anemia virus (EIAV) is a member of the lentivirus genus in the Retroviridae family and is considered an animal model for HIV/AIDS research. An attenuated EIAV vaccine, which was successfully developed in the 1970s by classical serial passage techniques, is the first and only lentivirus vaccine that has been widely used to date. Restriction factors are cellular proteins that provide an early line of defense against viral replication and spread by interfering with various critical steps in the viral replication cycle. However, viruses have evolved specific mechanisms to overcome these host barriers through adaptation. The battle between the viruses and restriction factors is actually a natural part of the viral replication process, which has been well studied in human immunodeficiency virus type 1 (HIV-1). EIAV has the simplest genome composition of all lentiviruses, making it an intriguing subject for understanding how the virus employs its limited viral proteins to overcome restriction factors. In this review, we summarize the current literature on the interactions between equine restriction factors and EIAV. The features of equine restriction factors and the mechanisms by which the EIAV counteract the restriction suggest that lentiviruses employ diverse strategies to counteract innate immune restrictions. In addition, we present our insights on whether restriction factors induce alterations in the phenotype of the attenuated EIAV vaccine.
    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
    线粒体抗病毒信号蛋白(MAVS)是针对大多数RNA病毒的免疫应答中必不可少的关节蛋白。无论是蝙蝠,大量人畜共患RNA病毒的天然宿主,利用涉及MAVS介导的干扰素(IFN)应答的保守信号通路仍然难以捉摸。在这项研究中,我们进行了蝙蝠MAVS(BatMAVS)的克隆和功能分析。氨基酸序列分析表明,BatMAVS在物种之间的保守性较差,在进化上更接近其他哺乳动物。BatMAVS的过表达通过激活I型IFN途径显著抑制绿色荧光蛋白(GFP)标记的VSV(VSV-GFP)和GFP标记的新城疫病毒(NDV-GFP)的复制,其在转录水平上的表达在VSV-GFP感染晚期上调。我们进一步证明了CARD_2和TM结构域在BatMAVS激活IFN-β的能力中占据很大比例。这些结果表明,BatMAVS在蝙蝠的IFN诱导和抗RNA病毒中起着重要的调节分子的作用。
    Mitochondrial antiviral signaling protein (MAVS) is an essential articulatory protein in immune responses against most RNA viruses. Whether bats, the natural hosts of numerous zoonotic RNA viruses, utilize conserved signaling pathways involving MAVS-mediated interferon (IFN) responses remains elusive. In this study, we performed the cloning and functional analysis of bat MAVS (BatMAVS). Amino acid sequence analysis revealed that BatMAVS was poorly conserved among species and evolutionarily closer to other mammals. Overexpression of BatMAVS significantly inhibited the replication of green fluorescent protein (GFP)-tagged VSV (VSV-GFP) and GFP-tagged Newcastle disease virus (NDV) (NDV-GFP) by activating the type I IFN pathway, and its expression at the transcriptional level was upregulated at the late stage of VSV-GFP infection. We further demonstrated that the CARD_2 and TM domains occupy a large proportion in the ability of BatMAVS to activate IFN-β. These results suggest that BatMAVS acts as an important regulatory molecule in IFN-induction and anti-RNA viruses in bats.
    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
    哺乳动物DDX23参与多种生物过程,如RNA加工和抗病毒反应。然而,硬骨鱼DDX23的功能仍不清楚。在本文中,我们克隆了黑鲤鱼(Mylophingodonpiceus)的DDX23同源物(bcDDX23),并阐明了其在抗病毒先天免疫中的作用。bcDDX23的编码区包含2427个核苷酸并编码809个氨基酸。LPS的刺激促进bcDDX23的转录,聚(I:C),和SVCV;免疫印迹(IB)分析表明,bcDDX23迁移了94.5kDa。免疫荧光(IF)检测显示bcDDX23主要分布在细胞核,SVCV感染后,胞浆bcDDX23的数量显着增加。报告分析显示bcDDX23抑制bcMAVS介导的IFN启动子的转录。并且免疫共沉淀(co-IP)测定鉴定了bcDDX23和bcMAVS之间的相互作用。此外,共表达的bcDDX23在EPC细胞中显著抑制bcMAVS介导的抗SVCV的抗病毒能力,bcDDX23的敲除增强了宿主细胞对SVCV的抗性。总的来说,我们的结果得出结论,bcDDX23靶向bcMAVS并抑制MAVS介导的IFN信号传导,这为硬骨鱼中IFN信号的调节提供了启示。
    Mammalian DDX23 is involved in multiple biological processes, such as RNA processing and antiviral responses. However, the function of teleost DDX23 still remains unclear. In this paper, we have cloned the DDX23 homologue of black carp (Mylopharyngodon piceus) (bcDDX23) and elucidated its role in the antiviral innate immunity. The coding region of bcDDX23 comprises 2427 nucleotides and encodes 809 amino acids. The transcription of bcDDX23 was promoted by the stimulation of LPS, poly(I:C), and SVCV; and immunoblotting (IB) assay showed that bcDDX23 migrated aground 94.5 kDa. Immunofluorescence (IF) assay revealed that bcDDX23 was mainly distributed in the nucleus, and the amount of cytosolic bcDDX23 was significantly increased after SVCV infection. The reporter assay showed that bcDDX23 inhibited bcMAVS-mediated transcription of the IFN promoter. And the co-immunoprecipitation (co-IP) assays identified the interaction between bcDDX23 and bcMAVS. Furthermore, co-expressed bcDDX23 significantly inhibited bcMAVS-mediated antiviral ability against SVCV in EPC cells, and knockdown of bcDDX23 enhanced the resistance of host cells against SVCV. Overall, our results conclude that bcDDX23 targets bcMAVS and suppresses MAVS-mediated IFN signaling, which sheds light on the regulation of IFN signaling in teleost fish.
    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