Foot-and-mouth disease virus (FMDV)

口蹄疫病毒 ( FMDV )
  • 文章类型: Journal Article
    背景:口蹄疫(FMD)是一种影响偶蹄动物的破坏性疾病,这导致受影响国家和地区的重大经济损失。目前,有明显倾向于利用纳米粒子作为创新疫苗开发的强大平台。因此,这项研究开发了一种基于铁蛋白的纳米颗粒(FNP)疫苗,该疫苗在FNP表面上显示口蹄疫病毒(FMDV)VP1(aa140-158)的中和表位,并评估了这些FNP在小鼠和豚鼠模型中的免疫原性和保护功效,为开发潜在的FMD疫苗提供了策略。
    结果:本研究表达了重组蛋白Hpf,HPF-NE和HPF-T34E通过大肠杆菌表达系统。结果表明,重组蛋白Hpf,Hpf-NE和Hpf-T34E可以有效地组装成纳米颗粒。随后,我们评估了Hpf的免疫原性,小鼠Hpf-NE和Hpf-T34E蛋白,以及Hpf-T34E蛋白在豚鼠中的免疫原性和保护性。小鼠试验成果显示Hpf-T34E组的免疫效力年夜于Hpf-NE组。用Hpf-T34E免疫的豚鼠的结果表明,免疫效力与FMD灭活疫苗(IV)的免疫原性基本一致,并且可以在豚鼠中提供针对FMDV攻击的部分保护。
    结论:Hpf-T34E纳米颗粒作为抗口蹄疫亚单位疫苗候选物较好的选择,在FMDV感染的模型动物中提供有效的保护。基于FNP的疫苗表现出优异的安全性和免疫原性,因此代表了继续开发高效和安全的FMD疫苗的有希望的策略。
    BACKGROUND: Foot-and-mouth disease (FMD) is a devastating disease affecting cloven-hoofed animals, that leads to significant economic losses in affected countries and regions. Currently, there is an evident inclination towards the utilization of nanoparticles as powerful platforms for innovative vaccine development. Therefore, this study developed a ferritin-based nanoparticle (FNP) vaccine that displays a neutralizing epitope of foot-and-mouth disease virus (FMDV) VP1 (aa 140-158) on the surface of FNP, and evaluated the immunogenicity and protective efficacy of these FNPs in mouse and guinea pig models to provide a strategy for developing potential FMD vaccines.
    RESULTS: This study expressed the recombinant proteins Hpf, HPF-NE and HPF-T34E via an E. coli expression system. The results showed that the recombinant proteins Hpf, Hpf-NE and Hpf-T34E could be effectively assembled into nanoparticles. Subsequently, we evaluated the immunogenicity of the Hpf, Hpf-NE and Hpf-T34E proteins in mice, as well as the immunogenicity and protectiveness of the Hpf-T34E protein in guinea pigs. The results of the mouse experiment showed that the immune efficacy in the Hpf-T34E group was greater than the Hpf-NE group. The results from guinea pigs immunized with Hpf-T34E showed that the immune efficacy was largely consistent with the immunogenicity of the FMD inactivated vaccine (IV) and could confer partial protection against FMDV challenge in guinea pigs.
    CONCLUSIONS: The Hpf-T34E nanoparticles stand out as a superior choice for a subunit vaccine candidate against FMD, offering effective protection in FMDV-infected model animals. FNP-based vaccines exhibit excellent safety and immunogenicity, thus representing a promising strategy for the continued development of highly efficient and safe FMD 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
    为了研制出安全有效的口蹄疫广谱疫苗,在这里,我们基于三种不同的FMDV拓扑类型开发了重组FMD多表位三价疫苗.通过在猪中的免疫效力评价疫苗的效力。结果表明,抗原不低于25μg的疫苗通过一级加强方案引发FMDV血清型O特异性抗体和中和抗体,并为猪提供免疫保护。更重要的是,该疫苗不仅引发了针对FMDV三种不同拓扑类型的相同水平的中和抗体,但也提供了完全保护猪免受三种相应的病毒攻击。在整个实验过程中,完全受保护的猪都无法产生抗3ABC抗体,这意味着疫苗可以提供无菌免疫。该疫苗可引发持久的高水平抗体,并在免疫后六个月内有效保护猪免受毒力攻击。因此,我们认为这种疫苗将来可能用于FMD的预防和控制。
    In order to develop a safe and effective broad-spectrum vaccine for foot-and-mouth disease (FMDV), here, we developed a recombinant FMD multiple-epitope trivalent vaccine based on three distinct topotypes of FMDV. Potency of the vaccine was evaluated by immune efficacy in pigs. The results showed that the vaccine with no less than 25 μg of antigen elicited FMDV serotype O specific antibodies and neutralization antibodies by primary-booster regime, and offered immune protection to pigs. More importantly, the vaccine elicited not only the same level of neutralization antibodies against the three distinct topotypes of FMDV, but also provided complete protection in pigs from the three corresponding virus challenge. None of the fully protected pigs were able to generate anti-3ABC antibodies throughout the experiment, which implied the vaccine can offer sterilizing immunity. The vaccine elicited lasting-long high-level antibodies and effectively protected pigs from virulent challenge within six months of immunization. Therefore, we consider that this vaccine may be used in the future for the prevention and control of FMD.
    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
    内部核糖体进入位点(IRES)元件构成顺式作用RNA调节序列,以不依赖帽的方式募集核糖体起始复合物,由各种RNA结合蛋白和IRES反式作用因子辅助。口蹄疫病毒(FMDV)含有功能性IRES元件,并利用该元件颠覆宿主翻译机制。我们的研究发现了一种新机制,其中RALY,属于RNA结合蛋白的异质核核糖核蛋白(hnRNP)家族的成员,通过其RNA识别基序残基与FMDVIRES的结构域3结合。这种相互作用通过抑制IRES驱动的翻译导致FMDV复制的下调。此外,我们的发现表明,RALY对FMDV复制的抑制作用不是归因于FMDVIRES介导的翻译起始复合物的组装,而是归因于与40S核糖体结合后80S核糖体复合物形成的障碍。相反,FMDV的3Cpro通过泛素-蛋白酶体途径抵消RALY介导的抑制。因此,这些结果表明,RALY,作为一种新型的关键IRES结合蛋白,通过阻断80S核糖体的形成来抑制FMDV的复制,提供对病毒如何招募和操纵宿主因素的更深入的了解。重要性由IRES元件驱动的FMDV基因组RNA的翻译是病毒感染的关键步骤。许多宿主蛋白被劫持以调节FMDVIRES依赖性翻译,但调控机制尚不清楚。这里,我们首次报道细胞RALY特异性地与FMDV的IRES相互作用,并通过阻断FMDVIRES上的80S核糖体组装来负调控病毒复制。相反,RALY介导的抑制通过泛素-蛋白酶体途径被病毒3C蛋白酶拮抗。这些结果将有助于进一步了解病毒感染过程中病毒与宿主的相互作用和翻译控制。
    The internal ribosome entry site (IRES) element constitutes a cis-acting RNA regulatory sequence that recruits the ribosomal initiation complex in a cap-independent manner, assisted by various RNA-binding proteins and IRES trans-acting factors. Foot-and-mouth disease virus (FMDV) contains a functional IRES element and takes advantage of this element to subvert host translation machinery. Our study identified a novel mechanism wherein RALY, a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family belonging to RNA-binding proteins, binds to the domain 3 of FMDV IRES via its RNA recognition motif residue. This interaction results in the downregulation of FMDV replication by inhibiting IRES-driven translation. Furthermore, our findings reveal that the inhibitory effect exerted by RALY on FMDV replication is not attributed to the FMDV IRES-mediated assembly of translation initiation complexes but rather to the impediment of 80S ribosome complex formation after binding with 40S ribosomes. Conversely, 3Cpro of FMDV counteracts RALY-mediated inhibition by the ubiquitin-proteasome pathway. Therefore, these results indicate that RALY, as a novel critical IRES-binding protein, inhibits FMDV replication by blocking the formation of 80S ribosome, providing a deeper understanding of how viruses recruit and manipulate host factors.
    OBJECTIVE: The translation of FMDV genomic RNA driven by IRES element is a crucial step for virus infections. Many host proteins are hijacked to regulate FMDV IRES-dependent translation, but the regulatory mechanism remains unknown. Here, we report for the first time that cellular RALY specifically interacts with the IRES of FMDV and negatively regulates viral replication by blocking 80S ribosome assembly on FMDV IRES. Conversely, RALY-mediated inhibition is antagonized by the viral 3C protease by the ubiquitin-proteasome pathway. These results would facilitate further understanding of virus-host interactions and translational control during viral 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
    口蹄疫(FMD)是一种引起重大经济损失的急性人畜共患病。迫切需要能够刺激有效保护性免疫应答的疫苗。在这项研究中,使用强大的插件和展示SpyTag/SpyCatcher系统,将血清型A和OFMD病毒(FMDV)的大肠杆菌衍生的重组VP1与热稳定的支架lumazine合酶(LS)或耐热拟杆菌封装蛋白(QtEnc)缀合,以产生多聚纳米疫苗。这些纳米疫苗在接种的小鼠中诱导高度有效的抗体应答。第一次免疫后第14天,抗体滴度比单体抗原高约100倍。两种疫苗均诱导高和长期IgG抗体产生。此外,QtEnc-VP1纳米疫苗诱导的抗体滴度高于LS-VP1纳米疫苗。纳米疫苗还诱导Th1偏向的免疫应答和更高水平的中和抗体。这些数据表明,通过将VP1与热稳定的支架缀合而产生的FMDV纳米疫苗是低资源地区FMDV控制的高度免疫原性和理想候选物。
    Foot-and-mouth disease (FMD) is an acute zoonosis causes significant economic losses. Vaccines able to stimulate efficient protective immune responses are urgently needed. In this study, Escherichia coli-derived recombinant VP1 of serotype A and O FMD virus (FMDV) was conjugated to thermostable scaffold lumazine synthase (LS) or Quasibacillus thermotolerans encapsulin (QtEnc) using a robust plug-and-display SpyTag/SpyCatcher system to generate multimeric nanovaccines. These nanovaccines induced highly potent antibody responses in vaccinated mice. On day 14 after the first immunisation, antibody titres were approximately 100 times higher than those of monomer antigens. Both vaccines induced high and long-term IgG antibody production. Moreover, the QtEnc-VP1 nanovaccine induced higher antibody titres than the LS-VP1 nanovaccine. The nanovaccines also induced Th1-biased immune responses and higher levels of neutralising antibodies. These data indicated that FMDV nanovaccines generated by conjugating VP1 with a thermostable scaffold are highly immunogenic and ideal candidates for FMDV control in low-resource areas.
    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: English Abstract
    瞬时表达是在哺乳动物细胞中表达口蹄疫病毒(FMDV)衣壳蛋白的主要方法。为了实现FMDV衣壳蛋白的稳定表达和病毒样颗粒(VLPs)在细胞中的高效组装,构建piggyBac(PB)转座子组成型表达质粒和PB转座子-四环素(Tet)诱导型表达载体。通过荧光蛋白测试质粒的功能。通过添加抗生素,组成型细胞池(C-WT,C-L127P)表达P12A3C(WT/L127P)基因和可诱导细胞池(I-WT,产生了表达P12A3C(WT/L127P)的I-L127P)基因。绿色荧光蛋白的基因,3C蛋白酶和反向四环素反式激活剂(rtTA)整合到染色体上,荧光观察和PCR检测证实了这一点。细胞池I-L127P具有更强的衣壳蛋白和VLP生产能力,通过蛋白质印迹和酶联免疫吸附测定(ELISA)证实,分别。总之,首次报道了诱导FMDV衣壳蛋白的染色体表达,这可能有助于哺乳动物生产FMDVVLP疫苗的技术过程以及其他蛋白质的哺乳动物诱导型表达系统的构建。
    Transient expression is the major method to express foot-and-mouth disease virus (FMDV) capsid proteins in mammalian cells. To achieve stable expression of FMDV capsid proteins and efficient assembly of virus like particles (VLPs) in cells, the plasmids of piggyBac (PB) transposon-constitutive expression and PB transposon-tetracycline (Tet) inducible expression vectors were constructed. The function of the plasmids was tested by fluorescent proteins. By adding antibiotics, the constitutive cell pools (C-WT, C-L127P) expressing P12A3C (WT/L127P) genes and the inducible cell pools (I-WT, I-L127P) expressing P12A3C (WT/L127P) genes were generated. The genes of green fluorescent protein, 3C protease and reverse tetracycline transactivator (rtTA) were integrated into chromosome, which was confirmed by fluorescence observation and PCR testing. The cell pool I-L127P has a stronger production capacity of capsid proteins and VLPs, which was confirmed by Western blotting and enzyme linked immunosorbent assay (ELISA), respectively. In conclusion, inducing the chromosomal expression of FMDV capsid proteins was firstly reported, which may facilitate the technical process of mammalian production of FMDV VLPs vaccine and the construction of mammalian inducible expression systems for other proteins.
    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
    口蹄疫(FMD)是偶蹄动物的高度传染性病毒性疾病,具有重大的社会经济影响。与这种疾病相关的一个问题是其病因的能力,口蹄疫病毒(FMDV),通过仍有待阐明的潜在机制在其宿主中持续存在。虽然在牛和小反刍动物中已经描述了持久性,它不太可能发生在猪身上。限制FMDV持久性理解进展的因素之一,特别是,差异持久性是缺乏合适的体外模型。源自背侧软腭的原始牛细胞模型,这是FMDV在牛中复制和持续的主要位点,已经开发出来了,这似乎与建立类似的猪模型有关。来自猪FMDV复制的两个位点的细胞,即,背侧软腭和口咽扁桃体,被分离和培养。然后通过免疫荧光评估来自背侧软腭的细胞的上皮特性。用FMDVO/FRA/1/2001克隆2.2单层感染后评估这些细胞的FMDV敏感性。这些细胞也在气-液界面的多层中生长,以模拟对FMDV感染敏感的复层上皮。与在猪体内显示的情况一致,我们的研究表明,没有证据表明FMDV在单层或多层模型中持续存在,感染后28天未检测到感染性病毒。这种模型的发展为猪细胞中FMDV的研究和诊断开辟了新的可能性。
    Foot-and-mouth disease (FMD) is a highly contagious viral disease of cloven-hoofed animals that has a significant socio-economic impact. One concern associated with this disease is the ability of its etiological agent, the FMD virus (FMDV), to persist in its hosts through underlying mechanisms that remain to be elucidated. While persistence has been described in cattle and small ruminants, it is unlikely to occur in pigs. One of the factors limiting the progress in understanding FMDV persistence and, in particular, differential persistence is the lack of suitable in vitro models. A primary bovine cell model derived from the dorsal soft palate, which is the primary site of replication and persistence of FMDV in cattle, has been developed, and it seemed relevant to develop a similar porcine model. Cells from two sites of FMDV replication in pigs, namely, the dorsal soft palate and the oropharyngeal tonsils, were isolated and cultured. The epithelial character of the cells from the dorsal soft palate was then assessed by immunofluorescence. The FMDV-sensitivity of these cells was assessed after monolayer infection with FMDV O/FRA/1/2001 Clone 2.2. These cells were also grown in multilayers at the air-liquid interface to mimic a stratified epithelium susceptible to FMDV infection. Consistent with what has been shown in vivo in pigs, our study showed no evidence of persistence of FMDV in either the monolayer or multilayer model, with no infectious virus detected 28 days after infection. The development of such a model opens up new possibilities for the study and diagnosis of FMDV in porcine 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
    口蹄疫(FMD)是偶蹄动物的高度传染性疾病,由口蹄疫病毒(FMDV)引起。它在亚洲和非洲流行,但在世界各地零星传播,造成畜牧业的重大损失。有效的抗FMDV疗法可能是支持性控制策略。在这里,我们利用计算机辅助,基于结构的虚拟筛选,以FMDV3C蛋白酶(3Cpro)为靶标,过滤来自美国国家癌症研究所(NCI)多样性和机械库的先导化合物。通过基于细胞的抗病毒和细胞内蛋白酶测定进一步检查了七个命中化合物,其中两个化合物(NSC116640和NSC332670)强烈抑制FMDV,EC50值在微摩尔水平为2.88µM(SI=73.15)和5.92µM(SI=11.11),分别。通过在0-60分钟内将病毒滴度的1.00降低至2.27logTCID50,这些化合物可以在杀病毒测定中直接灭活细胞外病毒。此外,添加时间测定显示NSC116640在感染早期(0-8小时)抑制FMDV,而NSC332670在感染时(0小时)同时添加时降低了病毒滴度。两种化合物均显示出良好的FMDV3Cpro抑制作用,IC50值为10.85µM(NSC116640)和4.21µM(NSC332670)。化合物在FMDV3Cpro上的分子对接显示了它们与FMDV3Cpro催化三联体中氨基酸的特异性相互作用。在物理化学和ADME分析研究中,两者都优先与酶和蛋白酶反应。结果揭示了两种新的小分子对FMDV和可能相关的小核糖核酸病毒具有抗病毒活性。
    Foot-and-mouth disease (FMD) is a highly contagious disease in cloven-hoofed animals, caused by the foot-and-mouth disease virus (FMDV). It is endemic in Asia and Africa but spreads sporadically throughout the world, resulting in significant losses in the livestock industry. Effective anti-FMDV therapeutics could be a supportive control strategy. Herein, we utilized computer-aided, structure-based virtual screening to filter lead compounds from the National Cancer Institute (NCI) diversity and mechanical libraries using FMDV 3C protease (3Cpro) as the target. Seven hit compounds were further examined via cell-based antiviral and intracellular protease assays, in which two compounds (NSC116640 and NSC332670) strongly inhibited FMDV, with EC50 values at the micromolar level of 2.88 µM (SI = 73.15) and 5.92 µM (SI = 11.11), respectively. These compounds could inactivate extracellular virus directly in a virucidal assay by reducing 1.00 to 2.27 log TCID50 of the viral titers in 0-60 min. In addition, the time-of-addition assay revealed that NSC116640 inhibited FMDV at the early stage of infection (0-8 h), while NSC332670 diminished virus titers when added simultaneously at infection (0 h). Both compounds showed good FMDV 3Cpro inhibition with IC50 values of 10.85 µM (NSC116640) and 4.21 µM (NSC332670). The molecular docking of the compounds on FMDV 3Cpro showed their specific interactions with amino acids in the catalytic triad of FMDV 3Cpro. Both preferentially reacted with enzymes and proteases in physicochemical and ADME analysis studies. The results revealed two novel small molecules with antiviral activities against FMDV and probably related picornaviruses.
    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
    严重急性呼吸系统综合症冠状病毒2(SARS-CoV-2)是潜在严重呼吸系统疾病的病原体,2019年冠状病毒病(COVID-19),持续的大流行,治疗选择有限。这里,我们评估了模拟口蹄疫病毒(FMDV)基因组(ncRNA)非编码区特定结构域的合成RNA的抗冠状病毒活性.已知这些分子在细胞培养中发挥广谱抗病毒活性,小鼠和猪有效触发宿主的先天免疫应答。在人肠道Caco-2和肺上皮Calu-32B4细胞中转染后,ncRNA显示出对SARS-CoV-2的有效抗病毒活性。当在K18-hACE2小鼠中评估FMDVncRNAs的体内功效时,与未经治疗的感染小鼠相比,鼻内感染SARS-CoV-2之前施用裸ncRNA显着降低了肺部的病毒载量和促炎细胞因子水平。当在肝细胞来源的Huh-7和猪睾丸ST细胞中检测针对普通人类HCoV-229E和猪传染性胃肠炎病毒(TGEV)时,ncRNA也是高度有效的。分别。这些结果证明了FMDVncRNAs的泛冠状病毒抗病毒活性的概念,包括人类和动物不同的冠状病毒,并可能增强我们对抗未来新兴变体的能力。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a potentially severe respiratory disease, the coronavirus disease 2019 (COVID-19), an ongoing pandemic with limited therapeutic options. Here, we assessed the anti-coronavirus activity of synthetic RNAs mimicking specific domains in the non-coding regions of the foot-and-mouth disease virus (FMDV) genome (ncRNAs). These molecules are known to exert broad-spectrum antiviral activity in cell culture, mice and pigs effectively triggering the host innate immune response. The ncRNAs showed potent antiviral activity against SARS-CoV-2 after transfection in human intestinal Caco-2 and lung epithelium Calu-3 2B4 cells. When the in vivo efficacy of the FMDV ncRNAs was assessed in K18-hACE2 mice, administration of naked ncRNA before intranasal SARS-CoV-2 infection significantly decreased the viral load and the levels of pro-inflammatory cytokines in the lungs compared with untreated infected mice. The ncRNAs were also highly efficacious when assayed against common human HCoV-229E and porcine transmissible gastroenteritis virus (TGEV) in hepatocyte-derived Huh-7 and swine testis ST cells, respectively. These results are a proof of concept of the pan-coronavirus antiviral activity of the FMDV ncRNAs including human and animal divergent coronaviruses and potentially enhance our ability to fight future emerging variants.
    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
    口蹄疫(FMD)是一种高度传染性的病毒性疾病,影响偶蹄动物。与这种疾病有关的问题之一是其致病因子的持续存在,口蹄疫病毒(FMDV)。虽然FMDV持续存在的机制尚不清楚,有线索表明,它可能与病毒蛋白和参与干扰素(IFN)反应的细胞蛋白之间的蛋白-蛋白相互作用(PPI)有关。由于已经在牛中描述了FMDV的持久性,绵羊和山羊,但不是猪,我们筛选了PPI涉及FMDV蛋白和16个主要的I型IFN途径蛋白从这四个物种通过纳米荧光素酶-2-杂交互补试验,以鉴定新的PPI并确定其宿主特异性。由于关于3Dpol在免疫逃逸中的作用的数据有限,因此有关3Dpol的结果是最有趣的,我们决定特别关注这种蛋白质。确定的PPI通过GST拉低得到确认。我们鉴定了3Dpol和7种IFN途径蛋白之间的PPI,即,IKKα,IKKε,IRF3,IRF7,NEMO,MDA5和MAVS。这些PPI在四个研究物种中是保守的,除了3Dpol和MAVS之间的一个,只在猪蛋白中发现。我们还展示了,使用荧光素酶报告基因测定,3Dpol可以抑制IFN途径的诱导阶段。这些结果表明,第一次,3Dpol在FMDV先天免疫逃逸中的推定作用。
    Foot-and-mouth disease (FMD) is a highly contagious viral disease affecting cloven-hoofed animals. One of the issues related to this disease is the persistence of its causative agent, foot-and-mouth disease virus (FMDV). While the mechanisms of FMDV persistence remain unclear, there are clues that it may be related to protein-protein interactions (PPI) between viral proteins and cellular proteins involved in the interferon (IFN) response. Since FMDV persistence has been described in cattle, sheep and goats but not in swine, we screened PPI involving FMDV proteins and sixteen major type-I IFN pathway proteins from these four species by nanoluciferase-2-hybrid complementation assay, in order to identify new PPI and determine their host specificity. As the results concerning the 3Dpol were the most interesting in view of the limited data concerning its role in immune escape, we decided to focus particularly on this protein. The identified PPI were confirmed by GST pull-down. We identified PPI between 3Dpol and seven IFN pathway proteins, namely, IKKα, IKKε, IRF3, IRF7, NEMO, MDA5 and MAVS. These PPI are conserved among the four studied species, with the exception of the one between 3Dpol and MAVS, which was only found with the swine protein. We also showed, using luciferase reporter assays, that 3Dpol could inhibit the induction phase of the IFN pathway. These results demonstrate, for the first time, a putative role for 3Dpol in FMDV innate immune escape.
    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
    口蹄疫病毒(FMDV)已经开发了各种策略来拮抗宿主的先天免疫。FMDVLpro和3Cpro通过不同的机制干扰I型IFN。FMDV的结构蛋白VP3降解Janus激酶1以抑制IFN-γ信号转导。FMDV的非结构蛋白是否参与抑制II型IFN信号通路尚不清楚。在这项研究中,研究表明,感染建立后,FMDV复制对IFN-γ治疗具有抗性,并且FMDV抑制II型IFN诱导的IFN-γ刺激基因(ISG)的表达。我们还首次显示FMDV非结构蛋白3C通过阻断STAT1核易位来拮抗IFN-γ刺激的JAK-STAT信号通路。3Cpro表达显着降低了ISGs转录水平和回文γ激活序列(GAS)启动子活性,在不影响蛋白质水平的情况下,酪氨酸磷酸化,和STAT1的同源二聚化。最后,我们提供的证据表明,3C蛋白酶活性在降解KPNA1中起着至关重要的作用,因此抑制了ISGsmRNA和GAS启动子的活性。我们的结果揭示了FMDV非结构蛋白拮抗宿主II型IFN信号的新机制。
    Foot-and-mouth disease virus (FMDV) has developed various strategies to antagonize the host innate immunity. FMDV Lpro and 3Cpro interfere with type I IFNs through different mechanisms. The structural protein VP3 of FMDV degrades Janus kinase 1 to suppress IFN-γ signaling transduction. Whether non-structural proteins of FMDV are involved in restraining type II IFN signaling pathways is unknown. In this study, it was shown that FMDV replication was resistant to IFN-γ treatment after the infection was established and FMDV inhibited type II IFN induced expression of IFN-γ-stimulated genes (ISGs). We also showed for the first time that FMDV non-structural protein 3C antagonized IFN-γ-stimulated JAK-STAT signaling pathway by blocking STAT1 nuclear translocation. 3Cpro expression significantly reduced the ISGs transcript levels and palindromic gamma-activated sequences (GAS) promoter activity, without affecting the protein level, tyrosine phosphorylation, and homodimerization of STAT1. Finally, we provided evidence that 3C protease activity played an essential role in degrading KPNA1 and thus inhibited ISGs mRNA and GAS promoter activities. Our results reveal a novel mechanism by which an FMDV non-structural protein antagonizes host type II IFN signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号