Senecavirus A

Senecavirus A
  • 文章类型: Journal Article
    背景:SenecavirusA(SVA)是小核糖核酸科中Senecavirus属病毒的唯一成员,是猪水疱病的病原体之一。SVA在美国有报道,加拿大,中国,泰国,和哥伦比亚。
    方法:在本研究中,通过RT-PCR在从安徽省不同规模的猪场收集的病态材料中检测到SVA阳性感染。
    结果:在这项研究中,通过在BHK21细胞上进行病毒分离,成功获得了SVA的强毒株,并命名为SVA-CH-AHAU-1。同时,一个简单的,本研究建立了快速、准确的检测SVA感染的纳米PCR方法,使用重组质粒pClone-SVA-3D作为模板。
    结论:SVA-CH-AHAU-1的完整基因组为7286bp,包括5'非编码区(UTR),6546个核苷酸的开放阅读框(ORF),编码2182个氨基酸(aa),和具有Poly(A)特征的3'UTR,系统发育分析表明,该分离株与美国分离株US-15-41901SD的核苷酸同源性最高(97.9%)。在这项研究中,发现毒株SVA-CH-AHAU-1在ORF区域与分离株SVA-CH-SDGT-2017和SVA/Canada/ON/FMA-2015-0024T2/2015重组.完整的基因组已提交给Genebank,登录号为OM654411。此外,我们的结果表明,建立的纳米PCR检测方法可以作为一种经济的,SVA方法的现场诊断的可靠和灵敏的方法,特别是在资源有限的地区。
    BACKGROUND: Senecavirus A (SVA) is the only member of the genus Senecavirus in the family Picornaviridae, and is one of the pathogens of porcine blistering disease. SVA has been reported in the United States, Canada, China, Thailand, and Colombia.
    METHODS: In this study, positive SVA infection was detected by RT-PCR in sick materials collected from pig farms of different sizes in Anhui Province.
    RESULTS: In this study, a virulent strain of SVA was successfully obtained by viral isolation on BHK21 cells and named SVA-CH-AHAU-1. Meanwhile, a simple, rapid and accurate nano-PCR method for the detection of SVA infection was established in this study, using the recombinant plasmid pClone-SVA-3D as a template.
    CONCLUSIONS: The complete genome of SVA-CH-AHAU-1 is 7286 bp, including a 5\' non-coding region (UTR), an open reading frame (ORF) of 6546 nucleotides, encoding 2182 amino acids (aa), and a 3\' UTR with Poly(A) features, and phylogenetic analysis showed that this isolate had the highest nucleotide homology (97.9 %) with the US isolate US-15-41901SD. In this study, the virulent strain SVA-CH-AHAU-1 was found to recombine in the ORF region with isolates SVA-CH-SDGT-2017 and SVA/Canada/ON/FMA-2015-0024 T2/2015. The complete genome has been submitted to GeneBank with the accession number OM654411. In addition, our results suggest that the established nano-PCR assay can be used as an economical, reliable and sensitive method for the field diagnosis of SVA method, especially in resource-limited 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: Journal Article
    病毒部署复杂的策略来劫持宿主的翻译机制,以促进病毒蛋白质合成并抵消先天的细胞防御。然而,关于SenecavirusA(SVA)控制宿主翻译的机制知之甚少。使用一系列复杂的分子细胞操作技术,异质核核糖核蛋白A2B1(hnRNPA2B1)被鉴定为参与SVA感染细胞翻译控制的必需宿主因子。还确定了SVA结构蛋白,VP3与hnRNPA2B1结合并重新定位,hnRNPA2B1干扰宿主的蛋白质合成机制,通过双管齐下的策略建立促进病毒繁殖的细胞环境:首先,hnRNPA2B1作为一个有效的内部核糖体进入位点(IRES)反式作用因子,通过支持翻译起始复合物的组装,选择性地选择促进病毒IRES驱动的翻译。第二,在VP3-hnRNPA2B1相互作用的背景下发生宿主细胞翻译的强烈抑制,导致干扰素响应的衰减。这是第一项证明SVAVP3和hnRNPA2B1之间相互作用并表征它们在操纵翻译中的关键作用的研究。这种新颖的双重机制,调节病毒感染细胞的选择性mRNA翻译和免疫逃避,强调VP3-hnRNPA2B1复合物是开发修饰的抗病毒或溶瘤试剂的潜在靶标。
    目的:病毒的繁殖取决于病毒蛋白的合成,这完全依赖于主机的翻译机器。因此,病毒通常需要控制细胞翻译装置,以有利于病毒蛋白的生产和避免宿主先天防御。SenecavirusA(SVA)是一种重要的病毒,既是猪肉行业的新兴病原体,也是神经内分泌癌症的潜在溶瘤病毒。这里,异质核核糖核蛋白A2B1(hnRNPA2B1)被确定为SVA感染过程中翻译景观的关键调节因子。这项研究支持了一个模型,即SVA的VP3蛋白通过其结合和重新定位hnRNPA2B1的能力有效地颠覆宿主的蛋白质合成机制,不仅选择性地促进病毒内部核糖体进入位点驱动的翻译,而且导致整体翻译关闭和免疫逃避。一起,这些数据为翻译机制之间的复杂相互作用提供了新的见解,SVA,和先天免疫有助于SVA的致病性。
    Viruses deploy sophisticated strategies to hijack the host\'s translation machinery to favor viral protein synthesis and counteract innate cellular defenses. However, little is known about the mechanisms by which Senecavirus A (SVA) controls the host\'s translation. Using a series of sophisticated molecular cell manipulation techniques, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) was identified as an essential host factor involved in translation control in SVA-infected cells. It was also determined that the SVA structural protein, VP3, binds to and relocalizes hnRNPA2B1, which interferes with the host\'s protein synthesis machinery to establish a cellular environment that facilitates viral propagation via a two-pronged strategy: first, hnRNPA2B1 serves as a potent internal ribosome entry site (IRES) trans-acting factor, which is selectively co-opted to promote viral IRES-driven translation by supporting the assembly of translation initiation complexes. Second, a strong repression of host cell translation occurs in the context of the VP3-hnRNPA2B1 interaction, resulting in attenuation of the interferons response. This is the first study to demonstrate the interaction between SVA VP3 and hnRNPA2B1, and to characterize their key roles in manipulating translation. This novel dual mechanism, which regulates selective mRNA translation and immune evasion of virus-infected cells, highlights the VP3-hnRNPA2B1 complex as a potential target for the development of modified antiviral or oncolytic reagents.
    OBJECTIVE: Viral reproduction is contingent on viral protein synthesis, which relies entirely on the host\'s translation machinery. As such, viruses often need to control the cellular translational apparatus to favor viral protein production and avoid host innate defenses. Senecavirus A (SVA) is an important virus, both as an emerging pathogen in the pork industry and as a potential oncolytic virus for neuroendocrine cancers. Here, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) was identified as a critical regulator of the translational landscape during SVA infection. This study supports a model whereby the VP3 protein of SVA efficiently subverts the host\'s protein synthesis machinery through its ability to bind to and relocalize hnRNPA2B1, not only selectively promoting viral internal ribosome entry site-driven translation but also resulting in global translation shutdown and immune evasion. Together, these data provide new insights into how the complex interactions between translation machinery, SVA, and innate immunity contribute to the pathogenicity of the SVA.
    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
    SenecavirusA(SVA)是一种在猪中流行的小核糖核酸病毒,引起的水泡疾病在临床上与其他水泡疾病没有区别,比如口蹄疫.广泛的病毒循环,不断进化,和给养猪业造成的经济损失强调需要采取措施控制代理商。在这项研究中,我们使用当代巴西SVA代表性毒株在Balb/ByJ小鼠中评估了全病毒灭活疫苗的免疫原性.通过肌内途径用两个剂量接种动物。通过用于IgG检测的内部ELISA测定来评估由疫苗接种诱导的体液应答。在来自接种和未接种组的脾细胞培养物中体外SVA刺激后,通过流式细胞术评估细胞应答。在用同源病毒口服攻击后,在实验组中评估针对SVA的保护。接种疫苗诱导高水平的IgG抗体和CD45R/B220+sIgM+的增殖,CD3e+CD69+,和CD3e+CD4+CD44+CD62L-细胞。这些结果表明疫苗制剂在鼠模型中的免疫原性和安全性以及针对SVA的体液和细胞应答的诱导。
    Senecavirus A (SVA) is a picornavirus that is endemic in swine, causing a vesicular disease clinically indistinguishable from other vesicular diseases, like foot-and-mouth disease. The widespread viral circulation, constant evolution, and economic losses caused to the swine industry emphasize the need for measures to control the agent. In this study, we evaluated the immunogenicity of a whole-virus-inactivated vaccine using a representative contemporary Brazilian SVA strain in Balb/ByJ mice. The animals were vaccinated with two doses by an intramuscular route. The humoral response induced by the vaccination was evaluated by an in-house ELISA assay for IgG detection. The cellular response was assessed by flow cytometry after in vitro SVA stimulation in splenocyte cultures from vaccinated and non-vaccinated groups. Protection against SVA was assessed in the experimental groups following an oral challenge with the homologous virus. The vaccination induced high levels of IgG antibodies and the proliferation of CD45R/B220+sIgM+, CD3e+CD69+, and CD3e+CD4+CD44+CD62L- cells. These results indicate the immunogenicity and safety of the vaccine formulation in a murine model and the induction of humoral and cellular response against SVA.
    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
    SenecavirusA(SVA)是一种新兴的病毒,对全世界的猪群构成威胁。迄今为止,尚未评估三方基序5(TRIM5)在病毒复制中的作用。这里,据报道,TRIM5通过促进由视黄酸诱导型基因I(RIG-I)介导的I型干扰素(IFN)抗病毒应答来抑制SVA复制。TRIM5表达在SVA感染的细胞中显著上调,TRIM5过表达抑制病毒复制并促进IFN-α,IFN-β,白细胞介素-1β(IL-1β),IL-6和IL-18表达。相反,干扰TRIM5的表达具有相反的效果。病毒吸附和进入实验表明,TRIM5不影响SVA的吸附,但抑制其进入。此外,TRIM5促进RIG-I和RIG-I介导的IFN和促炎细胞因子的表达,通过抑制TRIM5的表达也证明了这种作用。这些发现扩大了有关抑制SVA复制的宿主因子的知识范围,并表明靶向TRIM5可能有助于开发针对SVA的新药物。
    Senecavirus A (SVA) is an emerging virus that poses a threat to swine herds worldwide. To date, the role of tripartite motif 5 (TRIM5) in the replication of viruses has not been evaluated. Here, TRIM5 was reported to inhibit SVA replication by promoting the type I interferon (IFN) antiviral response mediated by retinoic acid-inducible gene I (RIG-I). TRIM5 expression was significantly upregulated in SVA-infected cells, and TRIM5 overexpression inhibited viral replication and promoted IFN-α, IFN-β, interleukin-1beta (IL-1β), IL-6, and IL-18 expression. Conversely, interfering with the expression of TRIM5 had the opposite effect. Viral adsorption and entry assays showed that TRIM5 did not affect the adsorption of SVA but inhibited its entry. In addition, TRIM5 promoted the expression of RIG-I and RIG-I-mediated IFNs and proinflammatory cytokines, and this effect was also proven by inhibiting the expression of TRIM5. These findings expand the scope of knowledge on host factors inhibiting the replication of SVA and indicate that targeting TRIM5 may aid in the development of new agents against SVA.
    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
    肝细胞癌(HCC)是一种高度侵袭性的肿瘤,治疗选择有限,死亡率高。SenecavirusA(SVA)已显示出选择性靶向肿瘤同时保留健康组织的潜力。本研究旨在研究SVA在体内外对肝癌细胞的影响,并阐明其作用机制。
    进行细胞计数试剂盒-8测定和集落形成测定以检查细胞增殖。流式细胞术和细胞核染色分析细胞周期分布和凋亡发生。使用HepG2细胞在体内创建皮下肿瘤异种移植HCC小鼠模型,评估肿瘤组织中Ki67的表达。采用末端脱氧核苷酸转移酶dUTP缺口末端标记法以及苏木精和伊红染色来评估HCC细胞凋亡和SVA对小鼠器官的毒性。
    体外,SVA通过诱导细胞凋亡和细胞周期阻滞有效抑制肿瘤细胞的生长。然而,它对正常肝细胞(MIHA细胞)没有显着影响。在体内环境中,SVA有效抑制小鼠模型中HCC的生长。SVA处理导致Ki67表达的显著降低和肿瘤细胞凋亡的增加。在SVA施用期间,在小鼠的器官中未观察到显著的组织病理学改变。
    SVA通过诱导细胞周期阻滞和凋亡抑制HCC细胞的生长。它不会对重要器官造成任何明显的毒性。
    UNASSIGNED: Hepatocellular carcinoma (HCC) is a highly aggressive tumor with limited treatment options and high mortality. Senecavirus A (SVA) has shown potential in selectively targeting tumors while sparing healthy tissues. This study aimed to investigate the effects of SVA on HCC cells in vitro and in vivo and to elucidate its mechanisms of action.
    UNASSIGNED: The cell counting kit-8 assay and colony formation assay were conducted to examine cell proliferation. Flow cytometry and nuclear staining were employed to analyze cell cycle distribution and apoptosis occurrence. A subcutaneous tumor xenograft HCC mouse model was created in vivo using HepG2 cells, and Ki67 expression in the tumor tissues was assessed. The terminal deoxynucleotidyl transferase dUTP nick end labeling assay and hematoxylin and eosin staining were employed to evaluate HCC apoptosis and the toxicity of SVA on mouse organs.
    UNASSIGNED: In vitro, SVA effectively suppressed the growth of tumor cells by inducing apoptosis and cell cycle arrest. However, it did not have a notable effect on normal hepatocytes (MIHA cells). In an in vivo setting, SVA effectively suppressed the growth of HCC in a mouse model. SVA treatment resulted in a significant decrease in Ki67 expression and an increase in apoptosis of tumor cells. No notable histopathological alterations were observed in the organs of mice during SVA administration.
    UNASSIGNED: SVA inhibits the growth of HCC cells by inducing cell cycle arrest and apoptosis. It does not cause any noticeable toxicity to vital organs.
    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
    SenecavirusA(SVA)是一种引起猪水泡病的新兴病毒,临床上与其他高后果的水疱性疾病难以区分。该病毒属于小牛科Senecavirus属。它的基因组是一个积极意义,单链RNA,长度约为7,300nt,具有3\'聚(A)尾,但没有5\'端封端结构。SVA可以在不同的细胞中有效繁殖,包括一些非猪来源的细胞系。先前在我们的实验室中使用反向遗传学从其cDNA克隆中拯救了野生型SVA。在本研究中,用传代-5SVA接种BSR-T7/5细胞系。接种后12小时,独立收集SVA感染和未感染的细胞用于比较转录组学分析。结果显示共628个差异表达基因,包括565个上调和63个下调,表明SVA感染显著刺激细胞中的转录起始。GO和KEGG富集分析表明SVA对细胞中免疫相关途径发挥多种作用。此外,RNA测序数据进行了其他深入分析,比如单核苷酸多态性,转录因子,和蛋白质-蛋白质相互作用。本研究,随着我们以前的蛋白质组学和代谢组学研究,提供了对SVA与其宿主之间相互作用的多组学洞察。
    Senecavirus A (SVA) is an emerging virus that causes the vesicular disease in pigs, clinically indistinguishable from other high consequence vesicular diseases. This virus belongs to the genus Senecavirus in the family Picornaviridae. Its genome is a positive-sense, single-stranded RNA, approximately 7,300 nt in length, with a 3\' poly(A) tail but without 5\'-end capped structure. SVA can efficiently propagate in different cells, including some non-pig-derived cell lines. A wild-type SVA was previously rescued from its cDNA clone using reverse genetics in our laboratory. In the present study, the BSR-T7/5 cell line was inoculated with the passage-5 SVA. At 12 h post-inoculation, SVA-infected and non-infected cells were independently collected for the analysis on comparative transcriptomics. The results totally showed 628 differentially expressed genes, including 565 upregulated and 63 downregulated ones, suggesting that SVA infection significantly stimulated the transcription initiation in cells. GO and KEGG enrichment analyses demonstrated that SVA exerted multiple effects on immunity-related pathways in cells. Furthermore, the RNA sequencing data were subjected to other in-depth analyses, such as the single-nucleotide polymorphism, transcription factors, and protein-protein interactions. The present study, along with our previous proteomics and metabolomics researches, provides a multi-omics insight into the interaction between SVA and its hosts.
    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:SenecavirusA(SV-A)是一种RNA病毒,属于小RNA病毒科中的Senecavirus属。本研究旨在分析影响SenecavirusA分子诊断的因素,如寡核苷酸,RNA提取方法,和RT-qPCR试剂盒。
    方法:对巴西猪的水疱病疑似病例进行了口蹄疫分析,猪水疱病,和水泡性口炎.所有这些疾病的测试均为阴性,但SV-A为阳性。使用RT-qPCR测试,比较不同的试剂盒和RNA提取方法。灵敏度和重复性进行了评估,证明在临床样品中检测SV-A的功效。
    结果:在RNA提取中,在初始稀释时观察到Cq值显着降低,特别是较大的上清液体积。Trizol和Maxwell在自动化设备协议中表现出更高的灵敏度,尽管组织测试的结果各不相同。RT-qPCR试剂盒比较揭示了使用病毒RNA的扩增差异,但与质粒DNA的差异最小。方法之间的灵敏度是可比的,在非扩增样品中略有变化。重复性测试显示RT-qPCRs之间的结果一致,尽管Cq值存在微小差异,但仍证明方法之间的相似性。
    结论:Trizol,硅胶柱,和半自动化提取进行了比较,以及不同的RT-qPCR试剂盒。该研究发现了可能影响最终诊断的重大变化。
    BACKGROUND: Senecavirus A (SV-A) is an RNA virus that belongs to the genus Senecavirus within the family Picornaviridae. This study aimed to analyze factors that can influence the molecular diagnosis of Senecavirus A, such as oligonucleotides, RNA extraction methods, and RT-qPCR kits.
    METHODS: Samples from suspected cases of vesicular disease in Brazilian pigs were analyzed for foot-and-mouth disease, swine vesicular disease, and vesicular stomatitis. All tested negative for these diseases but positive for SV-A. RT-qPCR tests were used, comparing different reagent kits and RNA extraction methods. Sensitivity and repeatability were evaluated, demonstrating efficacy in detecting SV-A in clinical samples.
    RESULTS: In RNA extraction, significant reduction in Cq values was observed with initial dilutions, particularly with larger supernatant volumes. Trizol and Maxwell showed greater sensitivity in automated equipment protocols, though results varied in tissue tests. RT-qPCR kit comparison revealed differences in amplification using viral RNA but minimal differences with plasmid DNA. Sensitivity among methods was comparable, with slight variations in non-amplified samples. Repeatability tests showed consistent results among RT-qPCRs, demonstrating similarity between methods despite minor discrepancies in Cq values.
    CONCLUSIONS: Trizol, silica columns, and semi-automated extraction were compared, as well as different RT-qPCR kits. The study found significant variations that could impact the final diagnosis.
    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
    背景:SenecavirusA(SVA),2002年发现,已知可导致猪特发性水疱病(PIVD),表现出类似于其他水泡疾病的症状。这种相似性使现场诊断复杂化。传统的分子诊断技术受到成本的限制,灵敏度,和复杂仪表的要求。因此,开发有效和准确的诊断方法对于及时识别和隔离受影响的猪至关重要,从而防止疾病进一步传播。
    方法:在本研究中,我们开发了一种由CRISPR/Cas12a提供支持的高度特异性和超灵敏的SVA检测方法。为了提高不同设备条件的实验室的可用性,介绍了酶标仪和紫外光透射仪。此外,PCR扩增也已被并入该方法中以提高灵敏度。在重组Cas12a蛋白的制备和基于CRISPR/Cas12a的反式切割系统的优化之后,确定该方法的特异性和灵敏度。
    结果:该方法与10种猪病毒无交叉反应。激活实质性反式切割活性所需的最小模板浓度被确定为106拷贝/μLSVA模板。然而,当PCR扩增被纳入,该方法实现了每个反应一个SVA模板拷贝的检测限.它还在模拟样品测试中表现出100%的准确度。完整的测试过程不超过三个小时。
    结论:重要的是,这种方法利用标准的实验室设备,使其可在资源有限的环境中使用,并在流行病期间促进广泛和超敏感的筛查。总的来说,这种方法的发展不仅扩大了可用于检测SVA的工具的范围,而且对控制PIVD的传播具有重要的前景。
    BACKGROUND: Senecavirus A (SVA), identified in 2002, is known to cause porcine idiopathic vesicular disease (PIVD), which presents with symptoms resembling other vesicular diseases. This similarity complicates field diagnosis. Conventional molecular diagnostic techniques are limited by their cost, sensitivity, and requirement for complicated instrumentation. Therefore, developing an effective and accurate diagnostic method is crucial for timely identification and isolation of affected pigs, thereby preventing further disease spread.
    METHODS: In this study, we developed a highly-specific and ultra-sensitive SVA detection method powered by CRISPR/Cas12a. To enhance the availability in laboratories with varied equipment conditions, microplate reader and ultraviolet light transilluminator were introduced. Moreover, PCR amplification has also been incorporated into this method to improve sensitivity. The specificity and sensitivity of this method were determined following the preparation of the recombinant Cas12a protein and optimization of the CRISPR/Cas12a-based trans-cleavage system.
    RESULTS: The method demonstrated no cross-reactivity with ten kinds of viruses of swine. The minimum template concentration required to activate substantial trans-cleavage activity was determined to be 106 copies/µL of SVA templates. However, when PCR amplification was incorporated, the method achieved a detection limit of one copy of SVA templates per reaction. It also exhibited 100% accuracy in simulated sample testing. The complete testing process does not exceed three hours.
    CONCLUSIONS: Importantly, this method utilizes standard laboratory equipment, making it accessible for use in resource-limited settings and facilitating widespread and ultra-sensitive screening during epidemics. Overall, the development of this method not only broadens the array of tools available for detecting SVA but also holds significant promise for controlling the spread of PIVD.
    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
    SenecavirusA(SVA),一种小核糖核酸病毒,导致猪的水泡病和流行性短暂性新生儿损失,对养猪业产生了多方面的经济影响。SVA通过促进病毒感染和传播的多种策略来抵消宿主的抗病毒反应。然而,SVA如何调节干扰素(IFN)反应的机制仍然难以捉摸。这里,我们证明SVA3C蛋白酶(3Cpro)阻断Janus激酶-信号转导子和转录激活因子(JAK-STAT)信号通路的转导,从而拮抗I型IFN应答。机械上,3Cpro通过其蛋白酶活性选择性地切割和降解STAT1和STAT2,而不靶向JAK1、JAK2和IRF9。值得注意的是,SVA3Cpro在亮氨酸(L)-天冬氨酸(D)基序上切割人和猪的STAT1,特别是L693/D694。在STAT2的情况下,确定了两个切割位点:在人和猪中都确定了谷氨酰胺(Q)707,而第二种分裂模式不同,在人STAT2中具有残基754-757(缬氨酸-亮氨酸-谷氨酰胺-丝氨酸基序),在猪STAT2中具有Q758。SVA3Cpro的这些切割模式与以前报道的其他小核糖核酸病毒3Cpro识别的经典基序部分不同,突出了SVA3Cpro的特点。一起,这些结果揭示了SVA3Cpro拮抗IFN诱导的抗病毒反应的机制,但也扩大了我们对微小核糖核酸病毒3Cpro的底物识别模式的认识。IMPORTANCESenecavirusA(SVA),小牛科Senecavirus属中唯一的成员,导致猪的水泡病在临床上与口蹄疫(FMD)没有区别,世界动物卫生组织(WOAH)列出的一种高度传染性病毒性疾病。干扰素(IFN)介导的抗病毒反应在限制和控制病毒感染中起着关键作用。小核糖核酸病毒进化出许多拮抗宿主抗病毒反应的策略。然而,SVA如何调节JAK-STAT信号通路,影响I型IFN应答,仍然难以捉摸。这里,我们确定3Cpro,SVA的一种蛋白酶,作为IFN应答的拮抗剂。3Cpro利用其蛋白酶活性来切割STAT1和STAT2,从而减少宿主IFN应答以促进SVA感染。我们的发现强调了3Cpro作为SVA感染过程中I型信号通路拮抗作用的关键毒力因子的重要性。
    Senecavirus A (SVA), a picornavirus, causes vesicular diseases and epidemic transient neonatal losses in swine, resulting in a multifaceted economic impact on the swine industry. SVA counteracts host antiviral response through multiple strategies facilitatng viral infection and transmission. However, the mechanism of how SVA modulates interferon (IFN) response remains elusive. Here, we demonstrate that SVA 3C protease (3Cpro) blocks the transduction of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway to antagonize type I IFN response. Mechanistically, 3Cpro selectively cleaves and degrades STAT1 and STAT2 while does not target JAK1, JAK2, and IRF9, through its protease activity. Notably, SVA 3Cpro cleaves human and porcine STAT1 on a Leucine (L)-Aspartic acid (D) motif, specifically L693/D694. In the case of STAT2, two cleavage sites were identified: glutamine (Q) 707 was identified in both human and porcine, while the second cleavage pattern differed, with residues 754-757 (Valine-Leucine-Glutamine-Serine motifs) in human STAT2 and Q758 in porcine STAT2. These cleavage patterns by SVA 3Cpro partially differ from previously reported classical motifs recognized by other picornaviral 3Cpro, highlighting the distinct characteristics of SVA 3Cpro. Together, these results reveal a mechanism by which SVA 3Cpro antagonizes IFN-induced antiviral response but also expands our knowledge about the substrate recognition patterns for picornaviral 3Cpro.IMPORTANCESenecavirus A (SVA), the only member in the Senecavirus genus within the Picornaviridae family, causes vesicular diseases in pigs that are clinically indistinguishable from foot-and-mouth disease (FMD), a highly contagious viral disease listed by the World Organization for Animal Health (WOAH). Interferon (IFN)-mediated antiviral response plays a pivotal role in restricting and controlling viral infection. Picornaviruses evolved numerous strategies to antagonize host antiviral response. However, how SVA modulates the JAK-STAT signaling pathway, influencing the type I IFN response, remains elusive. Here, we identify that 3Cpro, a protease of SVA, functions as an antagonist for the IFN response. 3Cpro utilizes its protease activity to cleave STAT1 and STAT2, thereby diminishing the host IFN response to promote SVA infection. Our findings underscore the significance of 3Cpro as a key virulence factor in the antagonism of the type I signaling pathway during SVA 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
    SenecavirusA(SVA)属于小RNA科的Senecavirus属。这种病毒具有积极意义,单链RNA基因组,长度约为7,200nt,由一个单独的5个未翻译区域组成,编码区域和3'个未翻译区域。在这项研究中,一个用增强的绿色FIN荧光蛋白(eGFP)序列标记的重组SVA,rSVA-eGFP,使用反向遗传学从其cDNA克隆中拯救出来。第5代(P5)rSVA-eGFP总共进行了55轮连续的荧光斑块到荧光斑块(FP-FP)转移,和一个额外的体外普通传代。通过下一代测序分析P61病毒原种。结果显示rSVA-eGFP基因组中存在10个单核苷酸突变(SNMs),包括9个过渡和只有一个变性。P61后代仍然显示完整的eGFP序列,表明在连续FP-FP转移期间在eGFP区域内没有发生拷贝选择重组。换句话说,该后代在eGFP序列(RES)的重组中具有遗传缺陷,即,缺乏RES的菌株。在十个SNM中,三个是错义突变,导致单氨基酸突变(SAAMs):L蛋白中的F15V,A74T在VP2中,而E53R在3D蛋白中。预测E53R在空间上与3D蛋白的RNA通道相邻,可能与RES缺乏菌株的出现有关。总之,这项研究揭示了连续FP-FP转移后rSVA-eGFP基因组的全球格局,此外,还揭示了可能与RES缺乏机制有关的推定SAAM。
    Senecavirus A (SVA) belongs to the genus Senecavirus in the family Picornaviridae. This virus possesses a positive-sense, single-stranded RNA genome, approximately 7200 nt in length, composed of a single 5\' untranslated region, encoding region and 3\' untranslated region. In this study, a recombinant SVA tagged with enhanced green fluorescent protein (eGFP) sequence, rSVA-eGFP, was rescued from its cDNA clone using reverse genetics. The passage-5 (P5) rSVA-eGFP was totally subjected to 55 rounds of consecutive fluorescent plaque-to-fluorescent plaque (FP-FP) transfers, and one extra common passaging in vitro. The P61 viral stock was analyzed by next-generation sequencing. The result showed ten single-nucleotide mutations (SNMs) in the rSVA-eGFP genome, including nine transitions and only one transversion. The P61 progeny still showed a complete eGFP sequence, indicating no occurrence of copy-choice recombination within the eGFP region during serial FP-FP transfers. In other words, this progeny was genetically deficient in the recombination of eGFP sequence (RES), namely, an RES-deficient strain. Out of ten SNMs, three were missense mutations, leading to single-amino acid mutations (SAAMs): F15V in L protein, A74T in VP2, and E53R in 3D protein. The E53R was predicted to be spatially adjacent to the RNA channel of 3D protein, perhaps involved in the emergence of RES-deficient strain. In conclusion, this study uncovered a global landscape of rSVA-eGFP genome after serial FP-FP transfers, and moreover shed light on a putative SAAM possibly related to the RES-deficient mechanism.
    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