Coronavirus OC43, Human

冠状病毒 OC43, 人
  • 文章类型: Journal Article
    在具有流行病或大流行潜力的人类中出现新的呼吸道病毒感染强调了对有效的广谱抗病毒药物(BSA)的迫切需要。源自植物的生物活性化合物可以提供新的BSA候选物的天然来源。这里,我们研究了新型植物复合物制剂SP4™作为针对当前主要人类呼吸道病毒的候选直接作用BSA,包括冠状病毒和流感病毒。SP4™抑制SARS-CoV-2,hCoV-OC43,hCoV-229E,甲型和乙型流感病毒,和低微克范围的呼吸道合胞病毒。使用hCoV-OC43作为代表性呼吸道病毒,观察到SP4™的大部分抗病毒活性主要源于其二聚体A型原花青素(PAC-A)组分。对作用机理模式的进一步研究显示SP4™及其富含PAC-A的部分防止hCoV-OC43附着于靶细胞并发挥杀病毒活性。这是通过它们与hCoV-OC43和SARS-CoV-2的刺突蛋白相互作用而发生的,从而干扰了刺突功能并导致病毒体感染性丧失。总的来说,这些发现支持SP4™作为天然来源的候选BSA用于预防人类呼吸道病毒感染的进一步发展。
    The appearance of new respiratory virus infections in humans with epidemic or pandemic potential has underscored the urgent need for effective broad-spectrum antivirals (BSAs). Bioactive compounds derived from plants may provide a natural source of new BSA candidates. Here, we investigated the novel phytocomplex formulation SP4™ as a candidate direct-acting BSA against major current human respiratory viruses, including coronaviruses and influenza viruses. SP4™ inhibited the in vitro replication of SARS-CoV-2, hCoV-OC43, hCoV-229E, Influenza A and B viruses, and respiratory syncytial virus in the low-microgram range. Using hCoV-OC43 as a representative respiratory virus, most of the antiviral activity of SP4™ was observed to stem primarily from its dimeric A-type proanthocyanidin (PAC-A) component. Further investigations of the mechanistic mode of action showed SP4™ and its PAC-A-rich fraction to prevent hCoV-OC43 from attaching to target cells and exert virucidal activity. This occurred through their interaction with the spike protein of hCoV-OC43 and SARS-CoV-2, thereby interfering with spike functions and leading to the loss of virion infectivity. Overall, these findings support the further development of SP4™ as a candidate BSA of a natural origin for the prevention of human respiratory virus 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
    我们产生了具有复制能力的OC43人类季节性冠状病毒(CoV),其表达了严重的急性呼吸道综合症冠状病毒2(SARS-CoV-2)尖峰,代替了天然尖峰(rOC43-CoV2S)。该病毒在培养的细胞和动物中相对于OC43和SARS-CoV-2是高度减毒的,并且被NIH生物安全委员会分类为生物安全2级(BSL-2)试剂。S特异性单克隆抗体对rOC43-CoV2S和SARS-CoV-2的中和与人血清高度相关,与重组水泡性口炎病毒-CoV2S不同。用rOC43-CoV2S单剂量免疫产生高水平的抗SARS-CoV-2的中和抗体,并完全保护人ACE2转基因小鼠免受SARS-CoV-2致命攻击,尽管在呼吸和非呼吸器官中检测不到复制。rOC43-CoV2S诱导恒河猴的S特异性血清和气道粘膜免疫球蛋白A和IgG反应。rOC43-CoV2S作为BSL-2试剂在真正的CoV的情况下测量S特异性抗体具有巨大价值,并且是候选的减毒SARS-CoV-2粘膜疫苗,优先在上呼吸道复制。
    We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.
    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
    病毒是专性寄生虫,依赖于细胞机制进行繁殖。几种病毒还掺入促进病毒传播的细胞蛋白。定义这些细胞蛋白对于破译病毒生命周期和描绘新的治疗策略至关重要。虽然许多研究探索了宿主蛋白在冠状病毒传播中的重要性,关于它们在成熟病毒体中存在的信息是有限的。在这项研究中,我们开发了一种方案来高度富集成熟的HCoV-OC43病毒体,并通过蛋白质组学对其进行表征。认识到细胞释放细胞外囊泡,其内容受病毒调节,考虑到我们能够从这些囊泡中分离病毒体,我们还分析了它们在未感染和感染细胞中的蛋白质含量。我们发现了与病毒体相关的69种独特的细胞蛋白,包括31种高置信度命中。这些蛋白质主要调节RNA代谢,酶活性,囊泡运输,细胞粘附,代谢物相互转化,和翻译。我们进一步发现该病毒对外泌体成分有深远的影响,与未感染细胞相比,在病毒相关的细胞外囊泡中掺入了47种新型细胞蛋白(11种高置信度),并排除了92种其他蛋白(61种高置信度)。此外,一个dsiRNA屏幕显示,11的18个目标选择显著影响病毒产量,包括病毒体或细胞外囊泡中的蛋白质。总的来说,这项研究为将许多宿主蛋白掺入HCoV-OC43病毒体提供了新的重要见解,它们的生物学意义,以及病毒调节细胞外囊泡的能力。
    目标:近年来,冠状病毒已经引起了全球的关注,这使得开发控制它们和防止未来大流行的方法变得至关重要。除了病毒蛋白,宿主蛋白在病毒繁殖中起着重要作用,并提供了潜在的治疗靶点。靶向宿主蛋白是有利的,因为与病毒蛋白相比,它们不太可能突变和产生抗性,许多抗病毒治疗的常见问题。在这项研究中,我们检查了毒性较低的生物安全级别2HCoV-OC43病毒的蛋白质含量,以代替毒性较大的SARS-CoV-2。我们的发现表明,整合到病毒体中的几种细胞蛋白调节病毒传播。此外,我们报告说,该病毒广泛调节细胞外囊泡的含量,增强病毒传播。这强调了病毒之间的关键相互作用,宿主蛋白,和细胞外囊泡。
    Viruses are obligate parasites that depend on the cellular machinery for their propagation. Several viruses also incorporate cellular proteins that facilitate viral spread. Defining these cellular proteins is critical to decipher viral life cycles and delineate novel therapeutic strategies. While numerous studies have explored the importance of host proteins in coronavirus spread, information about their presence in mature virions is limited. In this study, we developed a protocol to highly enrich mature HCoV-OC43 virions and characterize them by proteomics. Recognizing that cells release extracellular vesicles whose content is modulated by viruses, and given our ability to separate virions from these vesicles, we also analyzed their protein content in both uninfected and infected cells. We uncovered 69 unique cellular proteins associated with virions including 31 high-confidence hits. These proteins primarily regulate RNA metabolism, enzymatic activities, vesicular transport, cell adhesion, metabolite interconversion, and translation. We further discovered that the virus had a profound impact on exosome composition, incorporating 47 novel cellular proteins (11 high confidence) and excluding 92 others (61 high confidence) in virus-associated extracellular vesicles compared to uninfected cells. Moreover, a dsiRNA screen revealed that 11 of 18 select targets significantly impacted viral yields, including proteins found in virions or extracellular vesicles. Overall, this study provides new and important insights into the incorporation of numerous host proteins into HCoV-OC43 virions, their biological significance, and the ability of the virus to modulate extracellular vesicles.
    OBJECTIVE: In recent years, coronaviruses have dominated global attention, making it crucial to develop methods to control them and prevent future pandemics. Besides viral proteins, host proteins play a significant role in viral propagation and offer potential therapeutic targets. Targeting host proteins is advantageous because they are less likely to mutate and develop resistance compared to viral proteins, a common issue with many antiviral treatments. In this study, we examined the protein content of the less virulent biosafety level 2 HCoV-OC43 virus as a stand-in for the more virulent SARS-CoV-2. Our findings reveal that several cellular proteins incorporated into the virion regulate viral spread. In addition, we report that the virus extensively modulates the content of extracellular vesicles, enhancing viral dissemination. This underscores the critical interplay between the virus, host proteins, and extracellular vesicles.
    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
    COVID-19大流行的出现促使人们对季节性人类冠状病毒的兴趣增加。OC43,229E,NL63和HKU1是引起普通感冒的地方性季节性冠状病毒,通常伴有轻度呼吸道症状。在这项研究中,我们鉴定了在被3种冠状病毒感染后表现出细胞病变效应(CPE)的细胞系,并表征了它们的病毒复制动力学和感染对宿主表面受体表达的影响.我们发现NL63在LLC-MK2细胞中产生CPE,而OC43在MRC-5、HCT-8和WI-38细胞系中产生CPE,而229E在感染后第3天在MRC-5和WI-38中产生CPE。我们观察到,从感染后第3天到第5天,所有病毒的核衣壳和刺突病毒RNA(vRNA)急剧增加;然而,在感染细胞的上清液和细胞裂解物中测量的vRNA拷贝的丰度和比例根据病毒-宿主细胞对的不同而有很大差异。重要的是,我们观察到感染时冠状病毒进入和附着受体的调节。229E和OC43的感染分别导致CD13和GD3的下调。相比之下,NL63和OC43感染导致ACE2表达增加。使用可溶性ACE2或抗ACE2单克隆抗体阻断NL63进入的尝试证明了这些策略极大地减少感染的潜力。总的来说,我们的结果使我们能够更好地了解季节性冠状病毒在允许细胞系中的感染动力学,并揭示了可能对促进人类多种冠状病毒共同感染有影响的进入受体调节.IMPORTANCE季节性人类冠状病毒是与一般轻度上呼吸道感染相关的普通感冒的重要原因,可导致某些人的呼吸道并发症。没有针对这些病毒的疫苗,只有有限的抗病毒治疗方案来治疗最严重的病例。更好地了解这些病毒如何与宿主细胞相互作用对于确定预防感染相关并发症的新策略至关重要。通过分析不同允许细胞系中的病毒复制动力学,我们发现细胞依赖性宿主因素影响病毒基因的表达和病毒颗粒的释放。我们还分析了受感染细胞上的进入受体表达,发现这些可以根据感染的冠状病毒进行上调或下调。我们的发现引起了人们对某些冠状病毒共同感染后感染增强的可能性的担忧,这可能有助于基因重组和新变体和菌株的出现。
    The emergence of the COVID-19 pandemic prompted an increased interest in seasonal human coronaviruses. OC43, 229E, NL63, and HKU1 are endemic seasonal coronaviruses that cause the common cold and are associated with generally mild respiratory symptoms. In this study, we identified cell lines that exhibited cytopathic effects (CPE) upon infection by three of these coronaviruses and characterized their viral replication kinetics and the effect of infection on host surface receptor expression. We found that NL63 produced CPE in LLC-MK2 cells, while OC43 produced CPE in MRC-5, HCT-8, and WI-38 cell lines, while 229E produced CPE in MRC-5 and WI-38 by day 3 post-infection. We observed a sharp increase in nucleocapsid and spike viral RNA (vRNA) from day 3 to day 5 post-infection for all viruses; however, the abundance and the proportion of vRNA copies measured in the supernatants and cell lysates of infected cells varied considerably depending on the virus-host cell pair. Importantly, we observed modulation of coronavirus entry and attachment receptors upon infection. Infection with 229E and OC43 led to a downregulation of CD13 and GD3, respectively. In contrast, infection with NL63 and OC43 leads to an increase in ACE2 expression. Attempts to block entry of NL63 using either soluble ACE2 or anti-ACE2 monoclonal antibodies demonstrated the potential of these strategies to greatly reduce infection. Overall, our results enable a better understanding of seasonal coronaviruses infection kinetics in permissive cell lines and reveal entry receptor modulation that may have implications in facilitating co-infections with multiple coronaviruses in humans.IMPORTANCESeasonal human coronavirus is an important cause of the common cold associated with generally mild upper respiratory tract infections that can result in respiratory complications for some individuals. There are no vaccines available for these viruses, with only limited antiviral therapeutic options to treat the most severe cases. A better understanding of how these viruses interact with host cells is essential to identify new strategies to prevent infection-related complications. By analyzing viral replication kinetics in different permissive cell lines, we find that cell-dependent host factors influence how viral genes are expressed and virus particles released. We also analyzed entry receptor expression on infected cells and found that these can be up- or down-modulated depending on the infecting coronavirus. Our findings raise concerns over the possibility of infection enhancement upon co-infection by some coronaviruses, which may facilitate genetic recombination and the emergence of new variants and strains.
    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
    β-冠状病毒引起上呼吸道急性感染,导致各种症状和临床表现。OC43是一种人类β-冠状病毒,可引起轻度临床症状,可以在BSL2实验室进行安全研究。由于风险低,OC43可以成为理解β-冠状病毒发病机制的有价值和可访问的模型。限制病毒感染性的一个潜在目标可能是间隙连接介导的通讯。这项研究旨在揭示人类β-冠状病毒感染中通过间隙连接进行细胞间通讯的状态。用OC43感染导致Cx43在肺上皮癌细胞系A549中的表达降低。这种病毒感染也显示出显着的ER和氧化应激增加。在OC43感染后,在内质网-高尔基中间区室(ERGIC)区域观察到Cx43的内部定位,这削弱了两个相邻细胞之间的间隙连接通信,通过路西法黄染料转移试验证实。它也会影响半通道的形成,如EtBr摄取测定所示。Cx43贩运的损害以及形成半通道和功能性GJIC的能力受到病毒诱导的高尔基体破坏的阻碍。总之,这些结果表明,A549细胞中伴随OC43感染的一些生理变化,可以认为是理解病毒感染后缝隙连接通讯差异的合适模型系统.该模型系统可以为开发针对人类β-冠状病毒感染的疗法提供有价值的见解。重要性最近的SARS-CoV-2大流行的持久影响强调了研究人类β-冠状病毒的重要性,推进我们对未来冠状病毒感染的准备。由于SARS-CoV-2具有高度传染性,另一种人类β-冠状病毒OC43可以被认为是实验模型。可以考虑的关键途径之一是间隙连接通信,因为它对细胞稳态至关重要。我们的研究旨在了解人类β-冠状病毒OC43感染期间Cx43介导的细胞间通讯的变化。体外研究表明,OC43感染后,间隙连接蛋白Cx43的下调和内质网和氧化应激标志物的上调。此外,HCoV-OC43感染导致Cx43贩运减少,通过病毒介导的高尔基体破坏引起功能半通道的损害和GJIC的形成。总的来说,这项研究推断OC43感染重塑了细胞间的通讯,提示该途径可能是通过调节Cx43表达设计高效抗人冠状病毒治疗药物的一个有前景的靶点.
    β-coronaviruses cause acute infection in the upper respiratory tract, resulting in various symptoms and clinical manifestations. OC43 is a human β-coronavirus that induces mild clinical symptoms and can be safely studied in the BSL2 laboratory. Due to its low risk, OC43 can be a valuable and accessible model for understanding β-coronavirus pathogenesis. One potential target for limiting virus infectivity could be gap junction-mediated communication. This study aims to unveil the status of cell-to-cell communications through gap junctions in human β-coronavirus infection. Infection with OC43 leads to reduced expression of Cx43 in A549, a lung epithelial carcinoma cell line. Infection with this virus also shows a significant ER and oxidative stress increase. Internal localization of Cx43 is observed post-OC43 infection in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) region, which impairs the gap junction communication between two adjacent cells, confirmed by Lucifer yellow dye transfer assay. It also affects hemichannel formation, as depicted by the EtBr uptake assay. Impairment of Cx43 trafficking and the ability to form hemichannels and functional GJIC are hampered by virus-induced Golgi apparatus disruption. Altogether, these results suggest that several physiological changes accompany OC43 infection in A549 cells and can be considered an appropriate model system for understanding the differences in gap junction communication post-viral infections. This model system can provide valuable insights for developing therapies against human β-coronavirus infections.IMPORTANCEThe enduring impact of the recent SARS-CoV-2 pandemic underscores the importance of studying human β-coronaviruses, advancing our preparedness for future coronavirus infections. As SARS-CoV-2 is highly infectious, another human β-coronavirus OC43 can be considered an experimental model. One of the crucial pathways that can be considered is gap junction communication, as it is vital for cellular homeostasis. Our study seeks to understand the changes in Cx43-mediated cell-to-cell communication during human β-coronavirus OC43 infection. In vitro studies showed downregulation of the gap junction protein Cx43 and upregulation of the endoplasmic reticulum and oxidative stress markers post-OC43 infection. Furthermore, HCoV-OC43 infection causes reduced Cx43 trafficking, causing impairment of functional hemichannel and GJIC formation by virus-mediated Golgi apparatus disruption. Overall, this study infers that OC43 infection reshapes intercellular communication, suggesting that this pathway may be a promising target for designing highly effective therapeutics against human coronaviruses by regulating Cx43 expression.
    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
    人类冠状病毒(hCoV)OC43是全球人群特有的,通常会导致无症状或轻度上呼吸道疾病。这里,我们证明了用hCoV-OC43刺突糖蛋白的S1B和S1C结构域免疫的羊驼分离的纳米抗体的中和功效。总共40个纳米抗体以1至149nM的亲和力与重组OC43蛋白结合。两个纳米抗体WNb293和WNb294在0.21和1.79nM中和病毒,分别。与Fc结构域融合的WNb293的鼻内和腹膜内递送在hCoV-OC43感染的小鼠模型中显著降低了鼻病毒载量。用X射线晶体学,我们观察到WNb293与OC43S1B结构域上的表位结合,远离与宿主细胞进入有关的唾液酸聚糖结合位点。该结果表明该纳米抗体的中和机制不涉及聚糖结合的破坏。我们的工作提供了针对hCoV-OC43的纳米抗体的表征,可阻止病毒进入并减少体内病毒载量,并可能有助于未来基于纳米抗体的hCoV-OC43感染疗法。
    目的:持续的COVID-19大流行和先前由严重急性呼吸综合征冠状病毒和中东呼吸综合征冠状病毒引起的流行已经证明了冠状病毒带来的大流行潜力。除了这些主要的致病性冠状病毒,有四种地方性冠状病毒感染人类:hCoV-OC43,hCoV-229E,hCoV-HKU1和hCoV-NL63。我们确定了一组针对人类冠状病毒OC43(hCoV-OC43)的纳米抗体,并发现两个高亲和力纳米抗体在低纳摩尔浓度下有效地中和了hCoV-OC43。预防性施用一种中和纳米抗体降低了感染hCoV-OC43的小鼠中的病毒载量,显示了基于纳米抗体的治疗hCoV-OC43感染的潜力。
    Human coronavirus (hCoV) OC43 is endemic to global populations and usually causes asymptomatic or mild upper respiratory tract illness. Here, we demonstrate the neutralization efficacy of isolated nanobodies from alpacas immunized with the S1B and S1C domain of the hCoV-OC43 spike glycoprotein. A total of 40 nanobodies bound to recombinant OC43 protein with affinities ranging from 1 to 149 nM. Two nanobodies WNb 293 and WNb 294 neutralized virus at 0.21 and 1.79 nM, respectively. Intranasal and intraperitoneal delivery of WNb 293 fused to an Fc domain significantly reduced nasal viral load in a mouse model of hCoV-OC43 infection. Using X-ray crystallography, we observed that WNb 293 bound to an epitope on the OC43 S1B domain, distal from the sialoglycan-binding site involved in host cell entry. This result suggests that neutralization mechanism of this nanobody does not involve disruption of glycan binding. Our work provides characterization of nanobodies against hCoV-OC43 that blocks virus entry and reduces viral loads in vivo and may contribute to future nanobody-based therapies for hCoV-OC43 infections.
    OBJECTIVE: The pandemic potential presented by coronaviruses has been demonstrated by the ongoing COVID-19 pandemic and previous epidemics caused by severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus. Outside of these major pathogenic coronaviruses, there are four endemic coronaviruses that infect humans: hCoV-OC43, hCoV-229E, hCoV-HKU1, and hCoV-NL63. We identified a collection of nanobodies against human coronavirus OC43 (hCoV-OC43) and found that two high-affinity nanobodies potently neutralized hCoV-OC43 at low nanomolar concentrations. Prophylactic administration of one neutralizing nanobody reduced viral loads in mice infected with hCoV-OC43, showing the potential for nanobody-based therapies for hCoV-OC43 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
    病毒编码的复制酶通常会产生异常的RNA基因组,称为缺陷病毒基因组(DVG)。当与提供必需蛋白质的辅助病毒共感染时,DVG可以繁殖和传播。虽然DVG依赖于辅助病毒的传播,在某些情况下,它们可以破坏感染性病毒的复制,影响免疫反应,并影响病毒的持久性或进化。在感染期间,了解DVG与标准病毒基因组的动力学仍不清楚。为了解决这个问题,我们对两种β-冠状病毒进行了长期的实验进化,人类冠状病毒OC43(HCoV-OC43)和鼠肝炎病毒(MHV),在高和低感染复数(MOI)的细胞培养物中。然后我们以固定的时间间隔进行RNA-seq,重建的DVG,并分析了它们的积累动态。我们的研究结果表明,DVG进化为表现出更大的多样性和丰度,删除和插入是最常见的类型。值得注意的是,一些高MOI缺失显示出非常有限的暂时存在,而其他人随着时间的推移变得普遍。我们在HCoV-OC43样品中观察到高和低MOI条件之间的DVG丰度的差异。具有缺失的HCoV-OC43基因组的大小分布在高和低MOI传代之间不同。在低MOI谱系中,短而长的DVG是最常见的,在高MOI谱系中有一个额外的集群,随着进化时间的推移变得更加普遍。MHV在不同的MOI条件下也显示出DVG大小分布的变化,尽管与HCoV-OC43相比,它们不太明显,这表明DVG大小分布更为随机。我们确定了在高MOI下进化的缺失的热点区域,主要在编码结构和辅助蛋白的顺反子内。总之,我们的研究说明了DVGs的广泛形成过程中,受MOI和细胞和病毒特异性因素的影响。
    Virus-encoded replicases often generate aberrant RNA genomes, known as defective viral genomes (DVGs). When co-infected with a helper virus providing necessary proteins, DVGs can multiply and spread. While DVGs depend on the helper virus for propagation, they can in some cases disrupt infectious virus replication, impact immune responses, and affect viral persistence or evolution. Understanding the dynamics of DVGs alongside standard viral genomes during infection remains unclear. To address this, we conducted a long-term experimental evolution of two betacoronaviruses, the human coronavirus OC43 (HCoV-OC43) and the murine hepatitis virus (MHV), in cell culture at both high and low multiplicities of infection (MOI). We then performed RNA-seq at regular time intervals, reconstructed DVGs, and analyzed their accumulation dynamics. Our findings indicate that DVGs evolved to exhibit greater diversity and abundance, with deletions and insertions being the most common types. Notably, some high MOI deletions showed very limited temporary existence, while others became prevalent over time. We observed differences in DVG abundance between high and low MOI conditions in HCoV-OC43 samples. The size distribution of HCoV-OC43 genomes with deletions differed between high and low MOI passages. In low MOI lineages, short and long DVGs were the most common, with an additional cluster in high MOI lineages which became more prevalent along evolutionary time. MHV also showed variations in DVG size distribution at different MOI conditions, though they were less pronounced compared to HCoV-OC43, suggesting a more random distribution of DVG sizes. We identified hotspot regions for deletions that evolved at a high MOI, primarily within cistrons encoding structural and accessory proteins. In conclusion, our study illustrates the widespread formation of DVGs during betacoronavirus evolution, influenced by MOI and cell- and virus-specific factors.
    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
    随着COVID-19非药物干预措施的解除,全球有几个国家记录了常见病毒性呼吸道感染的死灰复燃.它促进病毒共感染,进一步负担已经过度紧张的医疗保健系统。需要迅速建立寻找与共感染相关的功效治疗剂的竞赛。然而,它遇到了许多挑战,需要仔细调查。这里,我们介绍了一种潜在的重组微型抗体相关治疗方法,3D8单链可变片段(scFv),它已被开发为一种广谱抗病毒药物,通过其核酸催化和细胞渗透能力发挥作用。在这项研究中,我们证明了3D8scFv在三种已建立的共感染模型中同时对甲型流感病毒(IAV)和冠状病毒具有抗病毒活性,该模型包括两种类型的冠状病毒[β冠状病毒-人冠状病毒OC43(hCoV-OC43)和α冠状病毒-猪流行性腹泻病毒(PEDV)]在VeroE6细胞中,MDCK细胞中的两个IAV[A/波多黎各/8/1934H1N1(H1N1/PR8)和A/X-31(H3N2/X-31)],以及VeroE6细胞中冠状病毒和IAV(hCoV-OC43和适应的H1N1)的组合,通过病毒基因表达的统计学显着降低,蛋白质水平,大约85%左右,65%,和80%的“hCoV-OC43-PEDV”后代,\'H1N1/PR8-H3N2/X-31\',和\'hCoV-OC43-adapted-H1N1\',分别,在3D8scFv的存在下被抽取。一起来看,我们认为3D8scFv是一种有前途的广谱药物,用于治疗共感染的RNA病毒。
    With the lifting of COVID-19 non-pharmaceutical interventions, the resurgence of common viral respiratory infections was recorded in several countries worldwide. It facilitates viral co-infection, further burdens the already over-stretched healthcare systems. Racing to find co-infection-associated efficacy therapeutic agents need to be rapidly established. However, it has encountered numerous challenges that necessitate careful investigation. Here, we introduce a potential recombinant minibody-associated treatment, 3D8 single chain variable fragment (scFv), which has been developed as a broad-spectrum antiviral drug that acts via its nucleic acid catalytic and cell penetration abilities. In this research, we demonstrated that 3D8 scFv exerted antiviral activity simultaneously against both influenza A viruses (IAVs) and coronaviruses in three established co-infection models comprising two types of coronaviruses [beta coronavirus-human coronavirus OC43 (hCoV-OC43) and alpha coronavirus-porcine epidemic diarrhea virus (PEDV)] in Vero E6 cells, two IAVs [A/Puerto Rico/8/1934 H1N1 (H1N1/PR8) and A/X-31 (H3N2/X-31)] in MDCK cells, and a combination of coronavirus and IAV (hCoV-OC43 and adapted-H1N1) in Vero E6 cells by a statistically significant reduction in viral gene expression, proteins level, and approximately around 85%, 65%, and 80% of the progeny of \'hCoV-OC43-PEDV\', \'H1N1/PR8-H3N2/X-31\', and \'hCoV-OC43-adapted-H1N1\', respectively, were decimated in the presence of 3D8 scFv. Taken together, we propose that 3D8 scFv is a promising broad-spectrum drug for treatment against RNA viruses in co-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
    病毒的不断突变,可变的免疫反应,并且可能出现新的病毒威胁需要多种方法来进行新的抗病毒治疗。此外,新的抗病毒药物应具有广谱活性和环境稳定。这里,我们表明,生物相容性锥形CuS纳米颗粒(NPs)有效地凝集冠状病毒,其结合亲和力取决于表面配体和颗粒形状的手性。左旋弯曲顶点的L-青霉胺稳定的NPs对假型SARS-CoV-2病毒和野生型武汉-1SARS-CoV-2病毒显示出半最大抑制浓度(IC50)低至0.66pM(1.4ng/mL)和0.57pM(1.2ng/mL),分别,比抗体(0.73nM)低约1100倍。受益于强大的NPs-蛋白质相互作用,同样的颗粒也对其他冠状病毒株有效,例如HCoV-HKU1,HCoV-OC43,HCoV-NL63和SARS-CoV-2Omicron变体,IC50值低于10pM(21.8ng/mL)。考虑到对疫情的快速反应,暴露于升高的温度不会导致NPs的抗病毒活性变化,而抗体完全失活。在小鼠中的测试表明,手性优化的NP可以用作抗病毒生物制剂的热稳定类似物,以补充当前的治疗谱。
    The incessant mutations of viruses, variable immune responses, and likely emergence of new viral threats necessitate multiple approaches to novel antiviral therapeutics. Furthermore, the new antiviral agents should have broad-spectrum activity and be environmentally stable. Here, we show that biocompatible tapered CuS nanoparticles (NPs) efficiently agglutinate coronaviruses with binding affinity dependent on the chirality of surface ligands and particle shape. L-penicillamine-stabilized NPs with left-handed curved apexes display half-maximal inhibitory concentrations (IC50) as low as 0.66 pM (1.4 ng/mL) and 0.57 pM (1.2 ng/mL) for pseudo-type SARS-CoV-2 viruses and wild-type Wuhan-1 SARS-CoV-2 viruses, respectively, which are about 1,100 times lower than those for antibodies (0.73 nM). Benefiting from strong NPs-protein interactions, the same particles are also effective against other strains of coronaviruses, such as HCoV-HKU1, HCoV-OC43, HCoV-NL63, and SARS-CoV-2 Omicron variants with IC50 values below 10 pM (21.8 ng/mL). Considering rapid response to outbreaks, exposure to elevated temperatures causes no change in the antiviral activity of NPs while antibodies are completely deactivated. Testing in mice indicates that the chirality-optimized NPs can serve as thermally stable analogs of antiviral biologics complementing the current spectrum of treatments.
    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
    多种重组蛋白的液相色谱纯化,并行,如果流程快速,接近传统的鲁棒性,可以催化研究和发现,“一次一种蛋白质”纯化。这里,我们报告一个自动化的,我们设计并验证了用于毫克规模平行蛋白质纯化的四通道色谱平台。该设备可以纯化多达四种蛋白质(每种蛋白质都有自己的单柱),具有多达八种缓冲液或溶剂的输入,可以通过软件驱动的阀门网络引导到四个列中的任何一个,并且包括自动级分收集器,其具有用于1.5或5.0mL收集管的十个位置和用于每个柱输出的用于50mL收集管的四个位置。可以通过Python脚本访问控制软件,让用户完全访问纯化过程的所有步骤,或通过简单导航的触摸屏图形用户界面,不需要命令行或任何编程语言的知识。使用我们的仪器,我们报告了毫克级,并行化,一组哺乳动物细胞表达的冠状病毒的单柱纯化(SARS-CoV-2,HCoV-229E,HCoV-OC43,HCoV-229E)三聚体刺突和单体受体结合域(RBD)抗原,和靶向SARS-CoV-2尖峰(S)和流感血凝素(HA)的单克隆抗体。我们包括详细的硬件构建指南,并使控制软件开源,允许其他人建立和定制自己的蛋白质净化器系统。
    Liquid chromatography purification of multiple recombinant proteins, in parallel, could catalyze research and discovery if the processes are fast and approach the robustness of traditional, \"one-protein-at-a-time\" purification. Here, we report an automated, four channel chromatography platform that we have designed and validated for parallelized protein purification at milligram scales. The device can purify up to four proteins (each with its own single column), has inputs for up to eight buffers or solvents that can be directed to any of the four columns via a network of software-driven valves, and includes an automated fraction collector with ten positions for 1.5 or 5.0 mL collection tubes and four positions for 50 mL collection tubes for each column output. The control software can be accessed either via Python scripting, giving users full access to all steps of the purification process, or via a simple-to-navigate touch screen graphical user interface that does not require knowledge of the command line or any programming language. Using our instrument, we report milligram-scale, parallelized, single-column purification of a panel of mammalian cell expressed coronavirus (SARS-CoV-2, HCoV-229E, HCoV-OC43, HCoV-229E) trimeric Spike and monomeric Receptor Binding Domain (RBD) antigens, and monoclonal antibodies targeting SARS-CoV-2 Spike (S) and Influenza Hemagglutinin (HA). We include a detailed hardware build guide, and have made the controlling software open source, to allow others to build and customize their own protein purifier systems.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号