3C Viral Proteases

3C 病毒蛋白酶
  • 文章类型: Journal Article
    肠病毒引起广泛的疾病,具有不同的表现和严重程度,一些肠道病毒已经成为严重的公共卫生问题。这些包括柯萨奇病毒B3(CVB3),一种病毒性心肌炎的活跃病原体,和柯萨奇病毒B4(CVB4),这可能会加速1型糖尿病的进展。来自CVB3和CVB4的3C蛋白酶在这些病毒的繁殖中起重要作用。在这项研究中,在大肠杆菌中表达来自CVB3和CVB4的3C蛋白酶,并通过亲和层析和凝胶过滤层析纯化。CVB3和CVB43C蛋白酶的晶体衍射至2.10和2.01µ分辨率,分别。通过分子置换方法解决了晶体结构,并包含典型的胰凝乳蛋白酶样折叠和保守的His40-Glu71-Cys147催化三联体。与来自其他肠道病毒的3C蛋白酶的结构比较显示高度相似性和微小差异,这将指导设计具有广谱特性的3C靶向抑制剂。
    Enteroviruses cause a wide range of disorders with varying presentations and severities, and some enteroviruses have emerged as serious public health concerns. These include Coxsackievirus B3 (CVB3), an active causative agent of viral myocarditis, and Coxsackievirus B4 (CVB4), which may accelerate the progression of type 1 diabetes. The 3C proteases from CVB3 and CVB4 play important roles in the propagation of these viruses. In this study, the 3C proteases from CVB3 and CVB4 were expressed in Escherichia coli and purified by affinity chromatography and gel-filtration chromatography. The crystals of the CVB3 and CVB4 3C proteases diffracted to 2.10 and 2.01 Å resolution, respectively. The crystal structures were solved by the molecular-replacement method and contained a typical chymotrypsin-like fold and a conserved His40-Glu71-Cys147 catalytic triad. Comparison with the structures of 3C proteases from other enteroviruses revealed high similarity with minor differences, which will guide the design of 3C-targeting inhibitors with broad-spectrum properties.
    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
    炎症小体通过加工和促进IL-1β的分泌在炎症中起关键作用。Caspase-1参与IL-1β和IL-18的成熟,而人caspase-4特异性加工IL-18。与Pro-IL-18结合的caspase-4的最新结构研究揭示了caspase-4激活Pro-IL-18的分子基础。然而,caspase-1处理pro-IL-1β和其他IL-1β转化酶的机制仍然难以捉摸。这里,我们观察到猪Pro-IL-1β(sPro-IL-1β)作为寡聚前体存在,与单体人Pro-IL-1β(hPro-IL-1β)不同。有趣的是,塞内卡谷病毒(SVV)3C蛋白酶切割sPro-IL-1β以产生成熟的IL-1β,虽然它切割hPro-IL-1β,但不以特定方式产生成熟的IL-1β。当炎症体被阻塞时,SVV3C通过在猪肺泡巨噬细胞(PAMs)中的直接裂解继续激活IL-1β。通过分子建模和诱变研究,我们发现sPro-IL-1β的pro-结构域充当“exosite”,其疏水残基对接到带正电荷的3C蛋白酶口袋中,从而将底物引导至活性位点。猪IL-1β(sIL-1β)的裂解产生单体和活性形式的sIL-1β,启动下行信令。因此,这些研究提供了IL-1β是一种炎症传感器,其通过与宿主炎性体并行操作的独立途径直接检测病毒蛋白酶。
    Inflammasomes play pivotal roles in inflammation by processing and promoting the secretion of IL-1β. Caspase-1 is involved in the maturation of IL-1β and IL-18, while human caspase-4 specifically processes IL-18. Recent structural studies of caspase-4 bound to Pro-IL-18 reveal the molecular basis of Pro-IL-18 activation by caspase-4. However, the mechanism of caspase-1 processing of pro-IL-1β and other IL-1β-converting enzymes remains elusive. Here, we observed that swine Pro-IL-1β (sPro-IL-1β) exists as an oligomeric precursor unlike monomeric human Pro-IL-1β (hPro-IL-1β). Interestingly, Seneca Valley Virus (SVV) 3C protease cleaves sPro-IL-1β to produce mature IL-1β, while it cleaves hPro-IL-1β but does not produce mature IL-1β in a specific manner. When the inflammasome is blocked, SVV 3C continues to activate IL-1β through direct cleavage in porcine alveolar macrophages (PAMs). Through molecular modeling and mutagenesis studies, we discovered that the pro-domain of sPro-IL-1β serves as an \'exosite\' with its hydrophobic residues docking into a positively charged 3C protease pocket, thereby directing the substrate to the active site. The cleavage of sPro-IL-1β generates a monomeric and active form of IL-1β, initiating the downstream signaling. Thus, these studies provide IL-1β is an inflammatory sensor that directly detects viral protease through an independent pathway operating in parallel with host inflammasomes.
    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
    柯萨奇病毒B3(CVB3)编码对翻译的病毒多蛋白的加工必需的蛋白酶。病毒蛋白酶还靶向宿主蛋白以操纵细胞过程并逃避先天抗病毒反应以促进复制和感染。虽然已经鉴定了CVB33C和2A半胱氨酸蛋白酶的一些宿主蛋白底物,目标的全部曲目尚不清楚。这里,我们利用基于无偏定量蛋白质组学的方法称为底物末端胺同位素标记(TAILS),对感染CVB3的人HeLa和小鼠心肌细胞(HL-1)细胞系中CVB3蛋白酶产生的N末端肽进行了全面分析.我们鉴定了在CVB3感染的HeLa和HL-1细胞中切割的>800种蛋白质,包括病毒多蛋白,已知的病毒3C蛋白酶的底物,如PABP,DDX58和HNRNPsM,K,和D和新的细胞蛋白。网络和GO-term分析显示,包括免疫反应和激活在内的生物过程的富集,RNA加工,和脂质代谢。我们验证了在CVB3感染下裂解的候选底物的子集,其中一些是3C蛋白酶在体外的直接靶标。此外,TAILS鉴定的靶蛋白子集的消耗降低了病毒产量。两种靶蛋白的表征表明,Emerin和氨酰-tRNA合成酶复合物相互作用多功能蛋白2的3Cpro靶向裂解片段的表达调节了自噬和活化B细胞的核因子κ轻链增强子(NF-κB)途径,分别。在病毒感染期间靶向的宿主蛋白的全面鉴定提供了对操纵以促进感染的细胞途径的见解。
    目的:RNA病毒编码负责将病毒蛋白加工成成熟形式的蛋白酶。病毒蛋白酶也靶向和切割宿主细胞蛋白;然而,这些靶蛋白的完整目录是不完整的。我们使用一种称为底物末端胺同位素标记(TAILS)的技术,N-末端组学用于鉴定在病毒感染下裂解的宿主蛋白。我们确定了数百种在感染下裂解的细胞蛋白质,其中一些是病毒蛋白酶直接靶向的。揭示这些靶蛋白提供了对宿主细胞途径和抗病毒信号传导因子的见解,这些因子被调节以促进病毒感染并可能导致病毒诱导的发病机理。
    Coxsackievirus B3 (CVB3) encodes proteinases that are essential for processing of the translated viral polyprotein. Viral proteinases also target host proteins to manipulate cellular processes and evade innate antiviral responses to promote replication and infection. While some host protein substrates of the CVB3 3C and 2A cysteine proteinases have been identified, the full repertoire of targets is not known. Here, we utilize an unbiased quantitative proteomics-based approach termed terminal amine isotopic labeling of substrates (TAILS) to conduct a global analysis of CVB3 protease-generated N-terminal peptides in both human HeLa and mouse cardiomyocyte (HL-1) cell lines infected with CVB3. We identified >800 proteins that are cleaved in CVB3-infected HeLa and HL-1 cells including the viral polyprotein, known substrates of viral 3C proteinase such as PABP, DDX58, and HNRNPs M, K, and D and novel cellular proteins. Network and GO-term analysis showed an enrichment in biological processes including immune response and activation, RNA processing, and lipid metabolism. We validated a subset of candidate substrates that are cleaved under CVB3 infection and some are direct targets of 3C proteinase in vitro. Moreover, depletion of a subset of TAILS-identified target proteins decreased viral yield. Characterization of two target proteins showed that expression of 3Cpro-targeted cleaved fragments of emerin and aminoacyl-tRNA synthetase complex-interacting multifunctional protein 2 modulated autophagy and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, respectively. The comprehensive identification of host proteins targeted during virus infection provides insights into the cellular pathways manipulated to facilitate infection.
    OBJECTIVE: RNA viruses encode proteases that are responsible for processing viral proteins into their mature form. Viral proteases also target and cleave host cellular proteins; however, the full catalog of these target proteins is incomplete. We use a technique called terminal amine isotopic labeling of substrates (TAILS), an N-terminomics to identify host proteins that are cleaved under virus infection. We identify hundreds of cellular proteins that are cleaved under infection, some of which are targeted directly by viral protease. Revealing these target proteins provides insights into the host cellular pathways and antiviral signaling factors that are modulated to promote virus infection and potentially leading to virus-induced pathogenesis.
    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
    肠道病毒D68(EV-D68)是一种与严重呼吸道疾病和麻痹性疾病有关的微小核糖核酸病毒,称为婴儿急性弛缓性脊髓炎。目前,没有保护性疫苗或抗病毒药物来对抗这种病毒。像其他肠道病毒一样,EV-D68使用细胞自噬途径的成分重新连接膜以进行复制。这里,我们显示转录因子EB(TFEB),自噬和溶酶体生物发生的主要转录调节因子,对于EV-D68感染至关重要。TFEB的敲减减弱EV-D68基因组RNA复制,但不影响病毒结合或进入宿主细胞。EV-D68的3C蛋白酶在病毒RNA复制高峰后立即在谷氨酰胺60(Q60)的N末端切割TFEB,破坏TFEB-RagC相互作用并限制TFEB运输到溶酶体表面。尽管如此,TFEB在EV-D68感染期间大部分保持细胞溶质。缺乏RagC结合结构域的TFEB突变体构建体的过表达,但不是野生型结构,在S1HeLa细胞中阻断自噬并增加EV-D68非裂解释放,但在自噬缺陷型ATG7KOS1HeLa细胞中不存在。我们的结果确定TFEB是调节EV-D68生命周期的多个阶段的重要宿主因子,并表明TFEB可能是针对EV-D68的抗病毒开发的有希望的目标。
    目的:肠道病毒是人类疾病的最重要原因之一。一些肠道病毒导致严重的麻痹性疾病,如脊髓灰质炎或急性弛缓性脊髓炎。后一种疾病与多种非脊髓灰质炎肠道病毒有关,包括肠道病毒D68(EV-D68),肠道病毒71型和柯萨奇病毒B3(CVB3)。这里,我们证明EV-D68与宿主转录因子相互作用,转录因子EB(TFEB),促进病毒RNA(vRNA)复制并调节病毒粒子从细胞中的流出。TFEB先前与CVB3的病毒外泄有关,并且病毒蛋白酶3C在感染期间切割TFEB。这里,我们显示EV-D683C蛋白酶也在vRNA复制达到峰值后切割TFEB。这种切割破坏了TFEB与宿主蛋白RagC的相互作用,这改变了TFEB的定位和调控。缺乏RagC结合结构域的TFEB抑制自噬通量并促进病毒外泄。这些机制的见解突出了共同的宿主因素如何影响密切相关的,医学上重要的病毒不同。
    Enterovirus D68 (EV-D68) is a picornavirus associated with severe respiratory illness and a paralytic disease called acute flaccid myelitis in infants. Currently, no protective vaccines or antivirals are available to combat this virus. Like other enteroviruses, EV-D68 uses components of the cellular autophagy pathway to rewire membranes for its replication. Here, we show that transcription factor EB (TFEB), the master transcriptional regulator of autophagy and lysosomal biogenesis, is crucial for EV-D68 infection. Knockdown of TFEB attenuated EV-D68 genomic RNA replication but did not impact viral binding or entry into host cells. The 3C protease of EV-D68 cleaves TFEB at the N-terminus at glutamine 60 (Q60) immediately post-peak viral RNA replication, disrupting TFEB-RagC interaction and restricting TFEB transport to the surface of the lysosome. Despite this, TFEB remained mostly cytosolic during EV-D68 infection. Overexpression of a TFEB mutant construct lacking the RagC-binding domain, but not the wild-type construct, blocks autophagy and increases EV-D68 nonlytic release in H1HeLa cells but not in autophagy-defective ATG7 KO H1HeLa cells. Our results identify TFEB as a vital host factor regulating multiple stages of the EV-D68 lifecycle and suggest that TFEB could be a promising target for antiviral development against EV-D68.
    OBJECTIVE: Enteroviruses are among the most significant causes of human disease. Some enteroviruses are responsible for severe paralytic diseases such as poliomyelitis or acute flaccid myelitis. The latter disease is associated with multiple non-polio enterovirus species, including enterovirus D68 (EV-D68), enterovirus 71, and coxsackievirus B3 (CVB3). Here, we demonstrate that EV-D68 interacts with a host transcription factor, transcription factor EB (TFEB), to promote viral RNA(vRNA) replication and regulate the egress of virions from cells. TFEB was previously implicated in the viral egress of CVB3, and the viral protease 3C cleaves TFEB during infection. Here, we show that EV-D68 3C protease also cleaves TFEB after the peak of vRNA replication. This cleavage disrupts TFEB interaction with the host protein RagC, which changes the localization and regulation of TFEB. TFEB lacking a RagC-binding domain inhibits autophagic flux and promotes virus egress. These mechanistic insights highlight how common host factors affect closely related, medically important viruses differently.
    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
    小核糖核酸病毒基因组编码一个大的,由病毒蛋白酶加工形成活性复制复合物的单一多蛋白。复制复合物与病毒基因组形成,宿主蛋白,和直接从每个病毒基因组产生/翻译的病毒蛋白(以顺式提供的病毒蛋白)。通过反式提供的病毒蛋白在体内复制复合物形成的有效互补,因此,外源或异位表达的病毒蛋白,还有待证明。这里,我们报道了一种有效的反式互补系统,用于通过病毒多蛋白前体在HEK293细胞中复制缺陷型脊髓灰质炎病毒(PV)突变体。病毒3AB中的多蛋白,但不是2BC,完全在顺式中处理。有缺陷的PV复制子突变体的复制,在3Cpro和3Dpol(3C/D[A/G]突变体)之间的病毒3Cpro蛋白酶的裂解位点被破坏,可以通过反式提供的病毒多蛋白来挽救。只有复制子的3Dpol活性缺陷可以反式挽救;2CATPase/hel中的失活突变,3B,和3Cpro的复制子完全废除了反式拯救的复制。以反式提供的3CDpro的3Cpro结构域的完整N末端对于反式活性功能是必需的。通过使用这个反式互补系统,一个高滴度的有缺陷的PV假病毒(PVpv)(>107感染单位/毫升)可以产生与有缺陷的突变体,其复制完全依赖于反式互补。这项工作揭示了外源病毒蛋白在PV复制中的潜在作用,并提供了对微小核糖核酸病毒感染期间蛋白质/蛋白质相互作用的见解。
    目的:病毒多蛋白加工是由多蛋白中编码的病毒蛋白酶精心控制的步骤;完全加工的蛋白质和加工中间体需要正确产生以进行复制,即使多蛋白的小修饰也可能受到不利影响。纯化/分离的病毒蛋白可以保留病毒复制所需的酶活性,如蛋白酶,解旋酶,聚合酶,等。然而,当这些小核糖核酸病毒的蛋白质被外源提供(反式提供)给具有缺陷病毒基因组的病毒复制复合物时,复制通常不被拯救/补充,提示内源性提供(以顺式提供)到复制复合物的病毒蛋白的重要性。在这项研究中,我发现只有脊髓灰质炎病毒(PV)(小核糖核酸病毒家族的典型成员)的病毒聚合酶活性可以通过外源表达的病毒蛋白有效地挽救。目前的研究揭示了外源病毒蛋白在病毒复制中的潜在作用,并提供了对小核糖核酸病毒感染过程中相互作用的见解。
    The picornavirus genome encodes a large, single polyprotein that is processed by viral proteases to form an active replication complex. The replication complex is formed with the viral genome, host proteins, and viral proteins that are produced/translated directly from each of the viral genomes (viral proteins provided in cis). Efficient complementation in vivo of replication complex formation by viral proteins provided in trans, thus exogenous or ectopically expressed viral proteins, remains to be demonstrated. Here, we report an efficient trans complementation system for the replication of defective poliovirus (PV) mutants by a viral polyprotein precursor in HEK293 cells. Viral 3AB in the polyprotein, but not 2BC, was processed exclusively in cis. Replication of a defective PV replicon mutant, with a disrupted cleavage site for viral 3Cpro protease between 3Cpro and 3Dpol (3C/D[A/G] mutant) could be rescued by a viral polyprotein provided in trans. Only a defect of 3Dpol activity of the replicon could be rescued in trans; inactivating mutations in 2CATPase/hel, 3B, and 3Cpro of the replicon completely abrogated the trans-rescued replication. An intact N-terminus of the 3Cpro domain of the 3CDpro provided in trans was essential for the trans-active function. By using this trans complementation system, a high-titer defective PV pseudovirus (PVpv) (>107 infectious units per mL) could be produced with the defective mutants, whose replication was completely dependent on trans complementation. This work reveals potential roles of exogenous viral proteins in PV replication and offers insights into protein/protein interaction during picornavirus infection.
    OBJECTIVE: Viral polyprotein processing is an elaborately controlled step by viral proteases encoded in the polyprotein; fully processed proteins and processing intermediates need to be correctly produced for replication, which can be detrimentally affected even by a small modification of the polyprotein. Purified/isolated viral proteins can retain their enzymatic activities required for viral replication, such as protease, helicase, polymerase, etc. However, when these proteins of picornavirus are exogenously provided (provided in trans) to the viral replication complex with a defective viral genome, replication is generally not rescued/complemented, suggesting the importance of viral proteins endogenously provided (provided in cis) to the replication complex. In this study, I discovered that only the viral polymerase activity of poliovirus (PV) (the typical member of picornavirus family) could be efficiently rescued by exogenously expressed viral proteins. The current study reveals potential roles for exogenous viral proteins in viral replication and offers insights into interactions during picornavirus 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
    SenecaValley病毒(SVV)是最近发现的一种导致水泡病的猪病原体,对全球养猪业构成重大威胁。产生促红细胞生成素的肝癌受体A2(EphA2)参与AKT/mTOR信号通路的激活,参与自噬。然而,SVV和EphA2之间的调控关系尚不清楚.在这项研究中,我们证明EphA2在SVV感染的BHK-21和PK-15细胞中被蛋白水解。EphA2过表达显著抑制SVV复制,正如病毒蛋白表达减少所证明的那样,病毒滴度,和病毒载量,提示EphA2的抗病毒功能。随后,筛选参与EphA2蛋白水解的病毒蛋白,并且发现SVV3C蛋白酶(3Cpro)负责这种切割,取决于它的蛋白酶活性。然而,3Cpro的蛋白酶活性位点不影响3Cpro与EphA2之间的相互作用。我们进一步确定EphA2过表达通过激活mTOR通路和抑制SVV复制来抑制自噬。一起来看,这些结果表明,SVV3Cpro靶向EphA2进行切割,以削弱其EphA2介导的抗病毒活性,并强调在开发针对SVV感染的抗病毒策略中涉及的分子相互作用的潜力.
    The Seneca Valley virus (SVV) is a recently discovered porcine pathogen that causes vesicular diseases and poses a significant threat to the pig industry worldwide. Erythropoietin-producing hepatoma receptor A2 (EphA2) is involved in the activation of the AKT/mTOR signaling pathway, which is involved in autophagy. However, the regulatory relationship between SVV and EphA2 remains unclear. In this study, we demonstrated that EphA2 is proteolysed in SVV-infected BHK-21 and PK-15 cells. Overexpression of EphA2 significantly inhibited SVV replication, as evidenced by decreased viral protein expression, viral titers, and viral load, suggesting an antiviral function of EphA2. Subsequently, viral proteins involved in the proteolysis of EphA2 were screened, and the SVV 3C protease (3Cpro) was found to be responsible for this cleavage, depending on its protease activity. However, the protease activity sites of 3Cpro did not affect the interactions between 3Cpro and EphA2. We further determined that EphA2 overexpression inhibited autophagy by activating the mTOR pathway and suppressing SVV replication. Taken together, these results indicate that SVV 3Cpro targets EphA2 for cleavage to impair its EphA2-mediated antiviral activity and emphasize the potential of the molecular interactions involved in developing antiviral strategies against SVV infection.
    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:人鼻病毒3C蛋白酶(HRV-3Cpro)在病毒增殖中起着至关重要的作用,将其确立为抗病毒治疗的首要目标。然而,关于鉴定HRV-3Cpro抑制剂的研究仍然有限。
    目的:本研究有两个主要目的:为了验证终点比色测定的有效性,以前由我们的团队开发,用于鉴定HRV-3Cpro的潜在抑制剂;其次,在药用植物中发现抑制酶活性的植物化学物质。
    方法:鲁普莱维,一种众所周知的HRV-3Cpro抑制剂,用于验证比色测定。在此之后,我们对2532种植物化学物质进行了两步筛选,这导致了八种活性化合物的鉴定:芹菜素,Carnosol,绿原酸,山奈酚,木犀草素,槲皮素,迷迭香酸,还有Rutin.我们随后在体外评估了这些候选物。为了进一步研究最有希望的候选人的抑制潜力,即,鼠尾草酚和迷迭香酸,进行分子对接研究以分析它们与HRV-3Cpro的结合相互作用。
    结果:我们先前开发的比色测定法可有效鉴定选择性抑制HRV-3Cpro的化合物。鼠尾草酚和迷迭香酸作为强效抑制剂出现,体外抑制HRV-3Cpro活性超过55%。我们的分析表明,鼠尾草酚和迷迭香酸通过竞争机制发挥抑制作用。分子对接证实了它们与酶活性位点的竞争性结合。
    结论:鼠尾草酚和迷迭香酸在开发冷疗方面的潜力值得进一步研究。通过强调这些化合物是有效的HRV-3Cpro抑制剂,我们的研究提供了一个有希望的方法来发现植物化学抑制剂对蛋白酶从类似的病原体。
    BACKGROUND: Human rhinovirus 3C protease (HRV-3Cpro) plays a crucial role in viral proliferation, establishing it as a prime target for antiviral therapy. However, research on identifying HRV-3Cpro inhibitors is still limited.
    OBJECTIVE: This study had two primary objectives: first, to validate the efficacy of an end-point colorimetric assay, previously developed by our team, for identifying potential inhibitors of HRV-3Cpro; and second, to discover phytochemicals in medicinal plants that inhibit the enzyme\'s activity.
    METHODS: Rupintrivir, a well-known inhibitor of HRV-3Cpro, was used to validate the colorimetric assay. Following this, we conducted a two-step in silico screening of 2532 phytochemicals, which led to the identification of eight active compounds: apigenin, carnosol, chlorogenic acid, kaempferol, luteolin, quercetin, rosmarinic acid, and rutin. We subsequently evaluated these candidates in vitro. To further investigate the inhibitory potential of the most promising candidates, namely, carnosol and rosmarinic acid, molecular docking studies were performed to analyze their binding interactions with HRV-3Cpro.
    RESULTS: The colorimetric assay we previously developed is effective in identifying compounds that selectively inhibit HRV-3Cpro. Carnosol and rosmarinic acid emerged as potent inhibitors, inhibiting HRV-3Cpro activity in vitro by over 55%. Our analysis indicated that carnosol and rosmarinic acid exert their inhibitory effects through a competitive mechanism. Molecular docking confirmed their competitive binding to the enzyme\'s active site.
    CONCLUSIONS: Carnosol and rosmarinic acid warrant additional investigation for their potential in the development of common cold treatment. By highlighting these compounds as effective HRV-3Cpro inhibitors, our study presents a promising approach for discovering phytochemical inhibitors against proteases from similar pathogens.
    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
    口蹄疫是一种高度传染性和传染性疾病,影响偶蹄动物。疾病控制因其高度传染性和抗原多样性而变得复杂。宿主microRNA(miRNA)是转录后调节因子,其在病毒感染中促进或抑制病毒复制。在本研究中,我们发现ssc-miR-7139-3p(SusscrofamiR-7139-3p)在口蹄疫病毒(FMDV)感染期间在宿主细胞中显著上调.miR-7139-3p的过表达减弱了FMDV的复制,而抑制促进FMDV复制。此外,通过注射miR-7139-3pagomiR可提高FMDV感染乳鼠的存活率。进一步的研究表明,miR-7139-3p靶向Bcl-2启动凋亡途径和caspase-3裂解3Cpro后174天冬氨酸(D174),这最终促进了3Cpro的降解。总的来说,我们的研究结果表明,miR-7139-3p通过靶向凋亡阴性调控基因Bcl-2促进3Cpro降解,从而抑制FMDV的复制.
    Foot-and-mouth disease is a highly contagious and infectious disease affecting cloven-hoofed animals. Disease control is complicated by its highly contagious nature and antigenic diversity. Host microRNAs (miRNAs) are post-transcriptional regulators that either promote or repress viral replications in virus infection. In the present study, we found that ssc-miR-7139-3p (Sus scrofa miR-7139-3p) was significantly up-regulated in host cells during foot-and-mouth disease virus (FMDV) infection. Overexpression of miR-7139-3p attenuated FMDV replication, whereas inhibition promoted FMDV replication. In addition, the survival rate of FMDV infected suckling mice was increased through injection of miR-7139-3p agomiR. Further studies revealed that miR-7139-3p targets Bcl-2 to initiate the apoptotic pathway and caspase-3 cleaved 3Cpro behind the 174th aspartic acid (D174), which eventually promotes the degradation of 3Cpro. Overall, our findings demonstrate that miR-7139-3p suppresses FMDV replication by promoting degradation of 3Cpro through targeting the apoptosis-negative regulatory gene Bcl-2.
    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:蛋白酶3C(3Cpro)是人类甲型肝炎病毒基因组中唯一编码的蛋白酶,由于其在病毒生命周期中的关键作用,被认为是抗病毒药物的潜在靶标。此外,3Cpro已被确定为铁性凋亡的有效诱导剂,一种新描述的细胞死亡类型。因此,研究3Cpro功能的分子机制可以为病毒与宿主的相互作用和铁凋亡的生物学作用提供新的见解。然而,这样的研究需要一种可靠的技术来生产功能活性的重组酶。
    目标:这里,我们表达了在N端或C端带有六组氨酸标签的3Cpro的不同修饰形式,以研究固定化金属离子亲和层析(IMAC)生产3Cpro的适用性。
    方法:我们在大肠杆菌中表达蛋白质,并使用IMAC纯化它们,然后是凝胶渗透色谱法。使用特定的显色底物测定产生的蛋白质的酶活性。
    结果:我们的发现表明,六组氨酸标签的引入和位置不影响酶的活性。然而,对于7个C末端残基被六组氨酸序列取代的变体,靶蛋白的产量最高.
    结论:我们证明了我们的方法用于生产重组,具有酶活性的3Cpro。
    BACKGROUND: Protease 3C (3Cpro) is the only protease encoded in the human hepatitis A virus genome and is considered as a potential target for antiviral drugs due to its critical role in the viral life cycle. Additionally, 3Cpro has been identified as a potent inducer of ferroptosis, a newly described type of cell death. Therefore, studying the molecular mechanism of 3Cpro functioning can provide new insights into viral-host interaction and the biological role of ferroptosis. However, such studies require a reliable technique for producing the functionally active recombinant enzyme.
    OBJECTIVE: Here, we expressed different modified forms of 3Cpro with a hexahistidine tag on the N- or C-terminus to investigate the applicability of immobilized metal Ion affinity chromatography (IMAC) for producing 3Cpro.
    METHODS: We expressed the proteins in Escherichia coli and purified them using IMAC, followed by gel permeation chromatography. The enzymatic activity of the produced proteins was assayed using a specific chromogenic substrate.
    RESULTS: Our findings showed that the introduction and position of the hexahistidine tag did not affect the activity of the enzyme. However, the yield of the target protein was highest for the variant with seven C-terminal residues replaced by a hexahistidine sequence.
    CONCLUSIONS: We demonstrated the applicability of our approach for producing recombinant, enzymatically active 3Cpro.
    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