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
    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
    LC/MS支持的基础目标化合物的筛选,MS/MS和全球天然产物Social(GNPS)用于鉴定Buteamonsperma的化合物1-10。在感染人类冠状病毒OC43(HCoV-OC43)的人类恶性胚胎横纹肌瘤细胞(RD细胞)中对它们进行了评估,并显示出显着的抑制活性。目标抑制试验表明,化合物6和8抑制蛋白水解酶3CLpro,它广泛存在于冠状病毒中,在复制过程中起着重要作用,具有有效的IC50值。该研究证实,化合物8的二甲醛可能是抑制冠状病毒的关键活性片段(EC507.2μM,SI>139.1)。结果导致鉴定出可能抑制HCoV-OC43的天然生物活性化合物并开发中药(TCM)药物。
    The screening of based target compounds supported by LC/MS, MS/MS and Global Natural Products Social (GNPS) used to identify the compounds 1-10 of Butea monsperma. They were evaluated in human malignant embryonic rhabdomyoma cells (RD cells) infected with Human coronavirus OC43 (HCoV-OC43) and showed significant inhibitory activity. Target inhibition tests showed that compounds 6 and 8 inhibited the proteolytic enzyme 3CLpro, which is widely present in coronavirus and plays an important role in the replication process, with an effective IC50 value. The study confirmed that dioxymethylene of compound 8 may be a key active fragment in inhibiting coronavirus (EC50 7.2 μM, SI > 139.1). The results have led to identifying natural bioactive compounds for possible inhibiting HCoV-OC43 and developing drug for Traditional Chinese Medicine (TCM).
    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
    在硬质和软质无孔材料上评估了三种抗菌剂对两种严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)替代品-牛冠状病毒(BCoV)和人冠状病毒(HCoV)OC43的疗效。具有三种不同活性成分(氯,过氧化氢,和季铵化合物+醇)进行了研究。最初,每种抗菌剂的中和方法进行了优化。然后,我们确定了它们在悬浮液中以及在用聚对苯二甲酸乙二醇酯(PET)塑料和乙烯基装饰织物制成的表面上对BCoV和HCoVOC43的功效。所有测试均在周围环境条件下进行,土壤负荷为5%胎牛血清。暴露2分钟后,所有三种抗微生物剂在悬浮液中的病毒滴度降低>3.0log10.所有这三者还将两种表面材料上的病毒感染性降低到检测极限(0.6log10TCID50/载体)以下。其中病毒滴度降低<3.0log10的处理归因于在消毒前干燥期间载体上的动态范围降低。载体数据显示,两种替代物在乙烯基上或在高相对湿度的条件下更快速地失活(p<0.05)。三类抗菌剂对两种SARS-CoV-2替代病毒均有效,与HCoVOC43相比,BCoV的灵敏度略低。这些发现还说明了(1)优化中和法和(2)考虑相对湿度作为功效测试的关键因素的重要性。
    The efficacy of three antimicrobials was evaluated against two severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) surrogates - bovine coronavirus (BCoV) and human coronavirus (HCoV) OC43 - on hard and soft nonporous materials. Three antimicrobials with three different active ingredients (chlorine, hydrogen peroxide, and quaternary ammonium compound + alcohol) were studied. Initially, a neutralization method was optimized for each antimicrobial. Then, we determined their efficacy against BCoV and HCoV OC43 in both suspension and on surfaces made with polyethylene terephthalate (PET) plastic and vinyl upholstery fabric. All tests were conducted under ambient environmental conditions with a soil load of 5% fetal bovine serum. After a 2-min exposure, all three antimicrobials achieved a >3.0 log10 reduction in viral titers in suspension. All three also reduced virus infectivity on both surface materials below the detection limit (0.6 log10 TCID50/carrier). Treatments in which the reduction in virus titer was <3.0 log10 were attributed to a decreased dynamic range on the carrier during drying prior to disinfection. The carrier data revealed that both surrogates were inactivated more rapidly (p <0.05) on vinyl or under conditions of high relative humidity. Three classes of antimicrobials were efficacious against both SARS-CoV-2 surrogate viruses, with BCoV demonstrating slightly less sensitivity compared to HCoV OC43. These findings also illustrate the importance of (1) optimizing the neutralization method and (2) considering relative humidity as a key factor for efficacy testing.
    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
    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
    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

公众号