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

    求助全文

  • 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

    求助全文

  • 文章类型: Journal Article
    人类感染冠状病毒(HCoV)家族对全球健康构成严重威胁,包括几种导致严重呼吸道疾病的高致病性菌株。我们必须开发有效的广谱抗HCoV药物,为未来的疫情做好准备。在这项研究中,我们使用ProteesolutionTrogeting嵌合体(PROTAC)技术专注于HCoV主要蛋白酶(Mpro)的降解,一种对病毒复制和致病性至关重要的保守酶。通过调整Mpro抑制剂GC376,我们生产了两种新型PROTACs,P2和P3对感染人类的CoVHCoV-229E显示出相对广谱的活性,HCoV-OC43和SARS-CoV-2。这些使病毒复制减少50%的PROTACs的浓度范围为0.71至4.6μM,并且在100μM时均未显示细胞毒性。此外,机制结合研究表明,P2和P3有效地靶向HCoV-229E,HCoV-OC43和SARS-CoV-2通过体外降解细胞内的Mpro。这项研究强调了PROTAC技术在开发广谱抗HCoV药物方面的潜力,提出了一种应对未来病毒爆发的新方法,特别是那些源于CoV的。
    The family of human-infecting coronaviruses (HCoVs) poses a serious threat to global health and includes several highly pathogenic strains that cause severe respiratory illnesses. It is essential that we develop effective broad-spectrum anti-HCoV agents to prepare for future outbreaks. In this study, we used PROteolysis TArgeting Chimera (PROTAC) technology focused on degradation of the HCoV main protease (Mpro), a conserved enzyme essential for viral replication and pathogenicity. By adapting the Mpro inhibitor GC376, we produced two novel PROTACs, P2 and P3, which showed relatively broad-spectrum activity against the human-infecting CoVs HCoV-229E, HCoV-OC43, and SARS-CoV-2. The concentrations of these PROTACs that reduced virus replication by 50 % ranged from 0.71 to 4.6 μM, and neither showed cytotoxicity at 100 μM. Furthermore, mechanistic binding studies demonstrated that P2 and P3 effectively targeted HCoV-229E, HCoV-OC43, and SARS-CoV-2 by degrading Mpro within cells in vitro. This study highlights the potential of PROTAC technology in the development of broad-spectrum anti-HCoVs agents, presenting a novel approach for dealing with future viral outbreaks, particularly those stemming from CoVs.
    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:鳞翅目的根茎(鳞翅目科,RDC),一种传统的东亚草药,具有广泛的药用特性,包括消炎药,抗癌,抗菌,和抗病毒活性。
    目的:本研究调查了RDC的30%乙醇提取物对人冠状病毒OC43(HCoV-OC43)的抗病毒潜力,严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),及其变种感染。
    方法:RDC或其成分的30%乙醇提取物,用人冠状病毒感染(HCoV-OC43,SARS-CoV-2及其变体)治疗了丝状酸ABA(PubChemCID:15081408)和干crassinABBA(PubChemCID:3082025)。使用UPLC-Q/TOFMass评估RDC的基峰色谱图,以鉴定RDC,使用LC-MS/MS进行RDC化合物的定量分析。细胞病变效应(CPE)减少试验,westernblot和免疫荧光染色病毒蛋白表达,进行定量病毒RNA拷贝数的qRT-PCR以确定抗病毒活性。添加时间测定,病毒附件,穿透力,和杀病毒试验,SARS-CoV-2Mpro和PLpro活性测定用于阐明作用方式。
    结果:RDC表现出剂量依赖性抑制HCoV-OC43诱导的细胞病变效应,降低病毒RNA拷贝数和病毒蛋白水平。添加时间测定表明,RDC靶向HCoV-OC43生命周期的早期阶段,抑制病毒体附着和渗透具有杀病毒活性。值得注意的是,丝状酸ABA和干蛋白酶ABBA,RDC的组成部分,降低HCoV-OC43病毒RNA载量。此外,RDC在假型化慢病毒检测中有效阻断病毒进入,涉及SARS-CoV-2Deltaplus和南非变体的刺突蛋白,以及表达水泡性口炎病毒糖蛋白G的对照慢病毒颗粒。RDC证明通过靶向病毒蛋白酶抑制SARS-CoV-2感染及其变体,即主要蛋白酶(Mpro)和木瓜蛋白酶(PLpro)。
    结论:这些发现强调了RDC通过阻止病毒进入和抑制病毒蛋白酶活性来靶向病毒感染的多阶段方法。因此,RDC作为一种强有力的承诺,广谱抗冠状病毒治疗剂。
    BACKGROUND: The rhizome of Dryopteris crassirhizoma Nakai (Dryopteridaceae, RDC), a traditional East Asian herbal medicine, possesses a broad spectrum of medicinal properties, including anti-inflammatory, anticancer, antibacterial, and antiviral activities.
    OBJECTIVE: This study investigates the 30% ethanolic extract of RDC\'s antiviral potential against human coronavirus OC43 (HCoV-OC43), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and its variants infections.
    METHODS: A 30% ethanolic extract of RDC or its components, filixic acid ABA (PubChem CID: 15081408) and dryocrassin ABBA (PubChem CID: 3082025) were treated with Human Coronavirus infection (HCoV-OC43, SARS-CoV-2 and its variants). The base peak chromatogram of RDC was evaluated using UPLC-Q/TOF Mass to identify the RDC, and the quantitative analysis of RDC compounds was performed using LC-MS/MS. A cytopathic effect (CPE) reduction assay, Western blot, immunofluorescence staining of viral protein expression, and qRT-PCR were performed to quantify the viral RNA copy numbers and determine the antiviral activity. The time-of-addition assay, the virus attachment, penetration, and virucidal assays, and SARS-CoV-2 Mpro and PLpro activity assay were used to elucidate the mode of action.
    RESULTS: RDC exhibited dose-dependent inhibition of HCoV-OC43-induced cytopathic effects, reducing viral RNA copy numbers and viral protein levels. Time-of-addition assays indicated that RDC targets the early stages of the HCoV-OC43 life cycle, inhibiting virion attachment and penetration with virucidal activity. Notably, filixic acid ABA and dryocrassin ABBA, constituents of RDC, reduced HCoV-OC43 viral RNA loads. Furthermore, RDC effectively blocked viral entry in pseudotyped lentivirus assays, involving spike proteins of SARS-CoV-2 Delta plus and South Africa variants, as well as control lentiviral particles expressing vesicular stomatitis virus glycoprotein G. Additionally, RDC demonstrated inhibition of SARS-CoV-2 infection and its variants by targeting viral proteases, namely main protease (Mpro) and papain-like protease (PLpro).
    CONCLUSIONS: These findings underscore RDC\'s multistage approach to targeting viral infections by impeding virus entry and inhibiting viral protease activity. Therefore, RDC holds promise as a potent, broad-spectrum anticoronaviral therapeutic agent.
    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
    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
    病毒感染的进展涉及许多转录调控事件。鉴定新合成的转录本有助于我们了解病毒的复制机制和发病机理。这里,我们利用一种称为硫醇(SH)连接烷基化的代谢RNA标记方法的时间分辨技术对RNA进行代谢测序(SLAM-seq),以差异阐明BHK21细胞系响应人冠状病毒OC43(HCoV-OC43)感染时稳态和新合成的RNA水平.我们的结果表明,响应于HCoV-OC43感染,Wnt/β-catenin信号通路显着富集了BHK21细胞系的新合成转录本。此外,Wnt通路的抑制促进了病毒在感染早期的复制,但是在感染的后期抑制了它。此外,remesivir抑制HCoV-OC43早期感染诱导的Wnt/β-catenin信号通路的上调。总的来说,我们的研究表明Wnt/β-catenin通路在HCoV-OC43感染的不同阶段的不同作用,提示抗病毒治疗的潜在目标。此外,尽管HCoV-OC43感染在BHK21细胞中诱导细胞病变效应,抑制细胞凋亡不影响病毒的细胞内复制。基于这种时间分辨方法监测新合成的RNA是研究病毒感染机制的非常有前途的方法。
    The progress of viral infection involves numerous transcriptional regulatory events. The identification of the newly synthesized transcripts helps us to understand the replication mechanisms and pathogenesis of the virus. Here, we utilized a time-resolved technique called metabolic RNA labeling approach called thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) to differentially elucidate the levels of steady-state and newly synthesized RNAs of BHK21 cell line in response to human coronavirus OC43 (HCoV-OC43) infection. Our results showed that the Wnt/β-catenin signaling pathway was significantly enriched with the newly synthesized transcripts of BHK21 cell line in response to HCoV-OC43 infection. Moreover, inhibition of the Wnt pathway promoted viral replication in the early stage of infection, but inhibited it in the later stage of infection. Furthermore, remdesivir inhibits the upregulation of the Wnt/β-catenin signaling pathway induced by early infection with HCoV-OC43. Collectively, our study showed the diverse roles of Wnt/β-catenin pathway at different stages of HCoV-OC43 infection, suggesting a potential target for the antiviral treatment. In addition, although infection with HCoV-OC43 induces cytopathic effects in BHK21 cells, inhibiting apoptosis does not affect the intracellular replication of the virus. Monitoring newly synthesized RNA based on such time-resolved approach is a highly promising method for studying the mechanism of viral infections.
    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
    冠状病毒的跨物种传播一直对公共卫生构成重大挑战。猪,作为许多人畜共患病毒的主要动物库,经常介导病毒传播给人类。本研究通过深入的生物信息学分析,全面绘制了人和猪冠状病毒之间的关系。我们发现人类冠状病毒OC43和猪冠状病毒PHEV具有密切的系统发育关系,高度的基因组同源性证明,相似的密码子使用模式和类似的三级结构的刺突蛋白。在源自猪小肠和大肠的类器官中接种感染性OC43病毒表明,猪肠类器官(pIOs)对人冠状病毒OC43感染高度敏感,并支持感染性病毒的产生。使用透射电子显微镜,我们在细胞内和细胞外区室观察到OC43病毒颗粒,并观察到感染的类器官细胞中多个细胞器的异常。pIOs中强健的OC43感染导致类器官活力的显著降低和广泛的细胞死亡。这项研究对于更好地理解人类冠状病毒OC43的进化起源具有重要意义,并为使用pIOs作为模型来研究人类冠状病毒的跨物种传播提供了概念证明。
    Cross-species transmission of coronaviruses has been continuously posing a major challenge to public health. Pigs, as the major animal reservoirs for many zoonotic viruses, frequently mediate viral transmission to humans. This study comprehensively mapped the relationship between human and porcine coronaviruses through in-depth bioinformatics analysis. We found that human coronavirus OC43 and porcine coronavirus PHEV share a close phylogenetic relationship, evidenced by high genomic homology, similar codon usage patterns and comparable tertiary structure in spike proteins. Inoculation of infectious OC43 viruses in organoids derived from porcine small and large intestine demonstrated that porcine intestinal organoids (pIOs) are highly susceptible to human coronavirus OC43 infection and support infectious virus production. Using transmission electron microscopy, we visualized OC43 viral particles in both intracellular and extracellular compartments, and observed abnormalities of multiple organelles in infected organoid cells. Robust OC43 infections in pIOs result in a significant reduction of organoids viability and widespread cell death. This study bears essential implications for better understanding the evolutionary origin of human coronavirus OC43, and provides a proof-of-concept for using pIOs as a model to investigate cross-species transmission of human coronavirus.
    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
    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
    人类呼吸道病毒对国家卫生系统产生巨大影响,社会,由于这种病原体的快速空气传播和流行传播,而有效的特异性抗病毒药物来对抗感染仍然缺乏。这里,我们确定了两种Keggin型多金属氧酸盐(POM),[TiW11CoO40]8-(TiW11Co)和[Ti2PW10O40]7-(Ti2PW10),具有针对有包膜和无包膜人类呼吸道病毒的广谱活性,即,冠状病毒(HCoV-OC43),鼻病毒(HRV-A1),呼吸道合胞病毒(RSV-A2),和腺病毒(AdV-5)。Ti2PW10对所有测试的病毒显示出非常有利的选择性指数(SI>700),并进一步研究了其针对人类冠状病毒和鼻病毒的抗病毒潜力。发现该POM抑制多种HCoV和HRV菌株的复制,在不同的细胞系统中。Ti2PW10不影响病毒结合或细胞内病毒复制,但选择性地抑制了病毒的进入。在POM存在下病毒的连续传代揭示了HRV-A1或HCoV-OC43的Ti2PW10抗性变体的发展的高度障碍。此外,Ti2PW10能够在人类鼻上皮的3D模型中抑制HRV-A1的产生,重要的是,抗病毒治疗未确定细胞毒性或组织损伤.还为Ti2PW10开发了用于鼻递送的粘膜粘附热敏原位水凝胶制剂。总的来说,对细胞系和人鼻上皮具有良好的生物相容性,广谱活动,和缺乏抗病毒抗性的发展揭示了Ti2PW10作为治疗急性呼吸道病毒性疾病发展的抗病毒候选物的潜力,保证进一步的研究,以确定聚阴离子的具体目标,并评估其临床潜力。
    Human respiratory viruses have an enormous impact on national health systems, societies, and economy due to the rapid airborne transmission and epidemic spread of such pathogens, while effective specific antiviral drugs to counteract infections are still lacking. Here, we identified two Keggin-type polyoxometalates (POMs), [TiW11CoO40]8- (TiW11Co) and [Ti2PW10O40]7- (Ti2PW10), endowed with broad-spectrum activity against enveloped and non-enveloped human respiratory viruses, i.e., coronavirus (HCoV-OC43), rhinovirus (HRV-A1), respiratory syncytial virus (RSV-A2), and adenovirus (AdV-5). Ti2PW10 showed highly favorable selectivity indexes against all tested viruses (SIs >700), and its antiviral potential was further investigated against human coronaviruses and rhinoviruses. This POM was found to inhibit replication of multiple HCoV and HRV strains, in different cell systems. Ti2PW10 did not affect virus binding or intracellular viral replication, but selectively inhibited the viral entry. Serial passaging of virus in presence of the POM revealed a high barrier to development of Ti2PW10-resistant variants of HRV-A1 or HCoV-OC43. Moreover, Ti2PW10 was able to inhibit HRV-A1 production in a 3D model of the human nasal epithelium and, importantly, the antiviral treatment did not determine cytotoxicity or tissue damage. A mucoadhesive thermosensitive in situ hydrogel formulation for nasal delivery was also developed for Ti2PW10. Overall, good biocompatibility on cell lines and human nasal epithelia, broad-spectrum activity, and absence of antiviral resistance development reveal the potential of Ti2PW10 as an antiviral candidate for the development of a treatment of acute respiratory viral diseases, warranting further studies to identify the specific target/s of the polyanion and assess its clinical potential.
    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)

公众号