Middle East respiratory syndrome coronavirus

中东呼吸综合征冠状病毒
  • 文章类型: Journal Article
    中东呼吸综合征冠状病毒(MERS-CoV)是一种影响骆驼和人类的人畜共患疾病。减毒活疫苗代表候选人疫苗,因为它可以在免疫宿主中诱导强免疫应答。高致病性病毒的减毒疫苗株也可用于在BSL2GMP设施中生产基于细胞的疫苗。在这项研究中,我们评估了致病性对致病性野生型病毒的逆转潜力,以确保减毒活疫苗株的安全性.我们在37°C的Vero细胞中在22°C(EMC2012-CA22°C)下将我们先前开发的冷适应减毒活MERS-CoV疫苗株传代15次,以确定hDPP4(人二肽基肽酶4)-转基因小鼠中致病性逆转的潜力,K18-hDPP4。EMC2012-CA22°C在37°C下在Vero细胞中连续传代多达15次没有导致对野生型MERS-CoV的致病性逆转。在感染这种病毒的K18-hDPP4小鼠中,未观察到体重减轻或死亡,在肺等组织中没有检测到病毒,肾,大脑,和鼻甲。此外,用这种病毒免疫的小鼠产生了强大的中和抗体反应,并且完全免受野生型MERS-CoV的致命攻击。冷适应减毒MERS-CoV疫苗株(EMC2012-CA22°C)在37°C下在Vero细胞中传代15次后未恢复为野生型致病病毒。
    Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic disease affecting camels and humans. The live attenuated vaccine represents a candidate human vaccine because it can induce strong immune responses in immunized hosts. The attenuated vaccine strain of the highly pathogenic virus can also be used to produce a cell-based vaccine in the BSL2 GMP facility. In this study, we evaluated the reversion potential of pathogenicity to pathogenic wild-type virus to ensure the safety of the live attenuated vaccine strain. We passaged our previously developed cold-adapted live attenuated MERS-CoV vaccine strain at 22 °C (EMC2012-CA22°C) in Vero cells at 37 °C as often as 15 times to determine the potential of pathogenicity reversion in hDPP4 (human dipeptidyl peptidase 4)-transgenic mice, K18-hDPP4. The serial passage of EMC2012-CA22°C in Vero cells at 37 °C up to 15 times did not result in pathogenicity reversion to wild-type MERS-CoV. In K18-hDPP4 mice infected with this virus, no weight loss or mortality was observed, and no virus was detected in tissues such as the lung, kidney, brain, and nasal turbinate. In addition, mice immunized with this virus produced a robust neutralizing antibody response and were fully protected from lethal challenge with wild-type MERS-CoV. The cold-adapted attenuated MERS-CoV vaccine strain (EMC2012-CA22°C) was not reverted to wild-type pathogenic virus after 15 passages in Vero cells at 37 °C.
    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(CoV)在其正链基因组RNA中具有许多功能性顺式作用元件。尽管这些RNA结构中的大多数参与病毒复制,病毒复制中CoV基因组RNA的RNA结构功能尚不清楚.在这项研究中,我们研究了高阶RNA茎环(SL)结构SL5B的功能,SL5C,和SL5D在中东呼吸综合征冠状病毒(MERS-CoV)的ORF1a编码区的病毒复制。我们的方法,利用细菌人工染色体系统的反向遗传学,揭示SL5B和SL5C在MERS-CoV的不连续转录中起着重要作用。在计算机分析中预测,SL5C与3'非翻译区的隆起茎环(BSL)相互作用,这表明SL5C的RNA结构对于病毒RNA转录很重要。相反,SL5D不影响转录,而是介导正链基因组RNA的合成。此外,SL5D突变体的回复病毒中SL5的RNA二级结构与野生型相似,这表明SL5D的RNA结构可以微调MERS-CoV中的RNA复制。我们的数据表明MERS-CoV基因组RNA中高阶RNA结构的病毒RNA转录和复制的新调控机制。
    Coronavirus (CoV) possesses numerous functional cis-acting elements in its positive-strand genomic RNA. Although most of these RNA structures participate in viral replication, the functions of RNA structures in the genomic RNA of CoV in viral replication remain unclear. In this study, we investigated the functions of the higher-order RNA stem-loop (SL) structures SL5B, SL5C, and SL5D in the ORF1a coding region of Middle East respiratory syndrome coronavirus (MERS-CoV) in viral replication. Our approach, using reverse genetics of a bacterial artificial chromosome system, revealed that SL5B and SL5C play essential roles in the discontinuous transcription of MERS-CoV. In silico analyses predicted that SL5C interacts with a bulged stem-loop (BSL) in the 3\' untranslated region, suggesting that the RNA structure of SL5C is important for viral RNA transcription. Conversely, SL5D did not affect transcription, but mediated the synthesis of positive-strand genomic RNA. Additionally, the RNA secondary structure of SL5 in the revertant virus of the SL5D mutant was similar to that of the wild-type, indicating that the RNA structure of SL5D can finely tune RNA replication in MERS-CoV. Our data indicate novel regulatory mechanisms of viral RNA transcription and replication by higher-order RNA structures in the MERS-CoV genomic RNA.
    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
    中东呼吸综合征(MERS)的传播动力学和控制的新数学模型,由MERS-CoV冠状病毒引起的呼吸道病毒(主要通过单峰骆驼传播给人类)于2012年首次出现在沙特阿拉伯王国(KSA),设计并用于研究该疾病在人类中的传播动态。KSA内的骆驼种群。对模型进行了严格的分析,使用观察到的KSA的MERS-CoV数据进行拟合和交叉验证,表明,每当其繁殖数(用${\\mathbbR}_{0M}$表示)小于1时,其无病平衡都是局部渐近稳定的。利用模型的固定参数和估计参数,KSA的${\\mathbbR}_{0M}$的价值估计为0.84,这表明消除MERS-CoV的前景非常有希望。该模型进行了扩展,以便评估公共卫生干预策略,特别是对人类和骆驼的疫苗的潜在使用,以及人类在公共场合或靠近骆驼时使用口罩。扩展模型的模拟表明,靠近骆驼的人使用面罩,作为唯一的公共卫生干预策略,显着减少了该疾病的人向骆驼和骆驼向人的传播,这种减少取决于社区使用的口罩类型的功效和覆盖范围。例如,如果外科口罩被优先考虑,如果至少45%与骆驼密切接触的个体持续佩戴骆驼,这种疾病可以在人类和骆驼群体中消除。模拟进一步表明,虽然接种疫苗作为唯一的干预策略,对降低人群的疾病负担仅有边际影响。基于给骆驼接种疫苗的干预策略仅导致骆驼疾病负担的显着减少(并且,因此,在人类中也是如此)。因此,这项研究表明,应该把注意力集中在有效防治骆驼种群的疾病上,而不是在人口中。此外,扩展模型用于模拟混合策略,它结合了人类和骆驼的疫苗接种以及人类使用口罩。这个模拟显示人类和骆驼的疾病负担显著减少,随着这种干预措施有效性的提高,与基线方案或上述任何单独的疫苗接种方案相比。总之,这项研究表明,在沙特阿拉伯王国消除MERS-CoV-2的前景有望使用药物(疫苗接种)和非药物(面罩)干预策略,单独实施或(优选地)组合实施,专注于减少骆驼种群的疾病负担。
    A new mathematical model for the transmission dynamics and control of the Middle Eastern respiratory syndrome (MERS), a respiratory virus caused by MERS-CoV coronavirus (and primarily spread to humans by dromedary camels) that first emerged out of the Kingdom of Saudi Arabia (KSA) in 2012, was designed and used to study the transmission dynamics of the disease in a human-camel population within the KSA. Rigorous analysis of the model, which was fitted and cross-validated using the observed MERS-CoV data for the KSA, showed that its disease-free equilibrium was locally asymptotically stable whenever its reproduction number (denoted by $ {\\mathbb R}_{0M} $) was less than unity. Using the fixed and estimated parameters of the model, the value of $ {\\mathbb R}_{0M} $ for the KSA was estimated to be 0.84, suggesting that the prospects for MERS-CoV elimination are highly promising. The model was extended to allow for the assessment of public health intervention strategies, notably the potential use of vaccines for both humans and camels and the use of face masks by humans in public or when in close proximity with camels. Simulations of the extended model showed that the use of the face mask by humans who come in close proximity with camels, as a sole public health intervention strategy, significantly reduced human-to-camel and camel-to-human transmission of the disease, and this reduction depends on the efficacy and coverage of the mask type used in the community. For instance, if surgical masks are prioritized, the disease can be eliminated in both the human and camel population if at least 45% of individuals who have close contact with camels wear them consistently. The simulations further showed that while vaccinating humans as a sole intervention strategy only had marginal impact in reducing the disease burden in the human population, an intervention strategy based on vaccinating camels only resulted in a significant reduction in the disease burden in camels (and, consequently, in humans as well). Thus, this study suggests that attention should be focused on effectively combating the disease in the camel population, rather than in the human population. Furthermore, the extended model was used to simulate a hybrid strategy, which combined vaccination of both humans and camels as well as the use of face masks by humans. This simulation showed a marked reduction of the disease burden in both humans and camels, with an increasing effectiveness level of this intervention, in comparison to the baseline scenario or any of the aforementioned sole vaccination scenarios. In summary, this study showed that the prospect of the elimination of MERS-CoV-2 in the Kingdom of Saudi Arabia is promising using pharmaceutical (vaccination) and nonpharmaceutical (mask) intervention strategies, implemented in isolation or (preferably) in combination, that are focused on reducing the disease burden in the camel population.
    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
    冠状病毒依靠宿主蛋白酶来激活病毒刺突蛋白,这有助于与宿主细胞膜融合并将病毒基因组RNA释放到宿主细胞质中。特定宿主蛋白酶在宿主中的分布决定了宿主,组织,和这些病毒的细胞嗜性。这里,我们确定激肽释放酶(KLK)家族成员KLK5是由人气道细胞分泌的一种主要宿主蛋白酶,并被多种人β冠状病毒利用.KLK5在体外裂解了来自各种人β病毒的刺突蛋白的引发(S1/S2)和激活(S2')位点。相比之下,KLK12和KLK13显示了S2\'或S1/S2站点的首选项,分别。而KLK12和KLK13合作激活SARS-CoV-2和MERS-CoV尖峰蛋白,KLK5本身有效地激活了来自几种人类β病毒的刺突蛋白,包括SARS-CoV-2.人β病毒对分化的人支气管上皮细胞(HBEC)的感染诱导了KLK5的增加,从而促进了病毒的复制。此外,熊果酸和其他抑制KLK5的相关植物来源的三萜类化合物有效抑制SARS-CoV的复制,MERS-CoV,HBEC中的SARS-CoV-2和减轻感染MERS-CoV或SARS-CoV-2的小鼠的肺部炎症。我们认为KLK5是一种泛冠状病毒宿主因子,是当前和未来冠状病毒引起的疾病的有希望的治疗靶点。
    Coronaviruses rely on host proteases to activate the viral spike protein, which facilitates fusion with the host cell membrane and the release of viral genomic RNAs into the host cell cytoplasm. The distribution of specific host proteases in the host determines the host, tissue, and cellular tropism of these viruses. Here, we identified the kallikrein (KLK) family member KLK5 as a major host protease secreted by human airway cells and exploited by multiple human betacoronaviruses. KLK5 cleaved both the priming (S1/S2) and activation (S2\') sites of spike proteins from various human betacoronaviruses in vitro. In contrast, KLK12 and KLK13 displayed preferences for either the S2\' or S1/S2 site, respectively. Whereas KLK12 and KLK13 worked in concert to activate SARS-CoV-2 and MERS-CoV spike proteins, KLK5 by itself efficiently activated spike proteins from several human betacoronaviruses, including SARS-CoV-2. Infection of differentiated human bronchial epithelial cells (HBECs) with human betacoronaviruses induced an increase in KLK5 that promoted virus replication. Furthermore, ursolic acid and other related plant-derived triterpenoids that inhibit KLK5 effectively suppressed the replication of SARS-CoV, MERS-CoV, and SARS-CoV-2 in HBECs and mitigated lung inflammation in mice infected with MERS-CoV or SARS-CoV-2. We propose that KLK5 is a pancoronavirus host factor and a promising therapeutic target for current and future coronavirus-induced diseases.
    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
    中东呼吸综合征冠状病毒(MERS-CoV)在人类中引起严重和致命的急性呼吸道疾病。高死亡率和持续的传染性仍然是全球卫生准备工作的紧迫问题。靶向受体结合域(RBD)的抗体是对抗人类病毒感染的主要对策。这里,我们报告了四种有效的纳米抗体对抗MERS-CoV,从羊驼中分离出来,特别是Nb14的效力在假型病毒检测中最高。结构研究表明,Nb14框架区(FRs)主要参与靶向一个新的表位的相互作用,这与以前报道的所有抗体完全不同,并破坏RBD的残基W535与hDPP4N229连接的碳水化合物部分(hDPP4-N229-聚糖)之间的蛋白质-碳水化合物相互作用。与Nb14不同,Nb9的目标是RBD的神秘面孔,它不同于hDPP4结合位点和Nb14表位,并诱导β5-β6环向RBD的浅槽弯曲,并抑制hDPP4短螺旋的容纳。特别醒目的表位赋予两个Nbs在假型MERS-CoV测定中协同施用。这些结果不仅具有用于抗体识别的前所未有的表位,而且还提供了预防和治疗MERS-CoV感染的有前途的药物。
    The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe and fatal acute respiratory disease in humans. High fatality rates and continued infectiousness remain a pressing concern for global health preparedness. Antibodies targeted at the receptor-binding domain (RBD) are major countermeasures against human viral infection. Here, we report four potent nanobodies against MERS-CoV, which are isolated from alpaca, and especially the potency of Nb14 is highest in the pseudotyped virus assay. Structural studies show that Nb14 framework regions (FRs) are mainly involved in interactions targeting a novel epitope, which is entirely distinct from all previously reported antibodies, and disrupt the protein-carbohydrate interaction between residue W535 of RBD and hDPP4 N229-linked carbohydrate moiety (hDPP4-N229-glycan). Different from Nb14, Nb9 targets the cryptic face of RBD, which is distinctive from the hDPP4 binding site and the Nb14 epitope, and it induces the β5-β6 loop to inflect towards a shallow groove of the RBD and dampens the accommodation of a short helix of hDPP4. The particularly striking epitopes endow the two Nbs administrate synergistically in the pseudotyped MERS-CoV assays. These results not only character unprecedented epitopes for antibody recognition but also provide promising agents for prophylaxis and therapy of MERS-CoV 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
    COVID-19大流行已经在全球蔓延,因此,必须利用所有可用资源来治疗这一祸害。在现实中,新药的开发主要受益于天然产品。广泛研究了菊科物种的广泛药用用途。在这项研究中,从单精子蒿的甲醇提取物中分离的化合物,一种生长在埃及西奈半岛的野生植物。三种化合物,豆甾醇3-O-β-D-吡喃葡萄糖苷1,鼠李糖素3和帕德蛋白6首先从该物种中分离出来。此外,以前报道的五种化合物,分离了阿卡西林2,jaceosidin4,hispidulin5,7-O-甲基乙二醇7和eupatilin8。应用分子模型模拟揭示了两种化合物,arcapillin2和鼠李糖素3在SARS-CoV-2Mpro结合位点内具有最佳的对接相互作用和能量(分别为-6.16和-6.70kcalmol-1)。顶部对接的化合物(2-3)进一步评估抑制浓度(IC50),SARS-CoV-2和MERS-CoV的半最大细胞毒性(CC50)。有趣的是,阿卡西林对SARS-CoV-2和MERS-CoV显示出高抗病毒活性,IC50值分别为190.8μgmL-1和16.58μgmL-1。这些发现可能为进一步的临床前和临床研究带来希望。特别是阿卡西林本身或与其他药物合作用于COVID-19治疗。
    The COVID-19 pandemic has spread throughout the whole globe, so it is imperative that all available resources be used to treat this scourge. In reality, the development of new pharmaceuticals has mostly benefited from natural products. The widespread medicinal usage of species in the Asteraceae family is extensively researched. In this study, compounds isolated from methanolic extract of Artemisia monosperma Delile, a wild plant whose grows in Egypt\'s Sinai Peninsula. Three compounds, stigmasterol 3-O-β-D-glucopyranoside 1, rhamnetin 3, and padmatin 6, were first isolated from this species. In addition, five previously reported compounds, arcapillin 2, jaceosidin 4, hispidulin 5, 7-O-methyleriodictyol 7, and eupatilin 8, were isolated. Applying molecular modelling simulations revealed two compounds, arcapillin 2 and rhamnetin 3 with the best docking interactions and energies within SARS-CoV-2 Mpro-binding site (-6.16, and -6.70 kcal mol-1, respectively). The top-docked compounds (2-3) were further evaluated for inhibitory concentrations (IC50), and half-maximal cytotoxicity (CC50) of both SARS-CoV-2 and MERS-CoV. Interestingly, arcapillin showed high antiviral activity towards SARS-CoV-2 and MERS-CoV, with IC50 values of 190.8 μg mL-1 and 16.58 μg mL-1, respectively. These findings may hold promise for further preclinical and clinical research, particularly on arcapillin itself or in collaboration with other drugs for COVID-19 treatment.
    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
    暂无摘要。
    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
    严重急性呼吸系统综合症冠状病毒2型(SARS-CoV-2)由于其进化和产生新的亚变体的能力,仍然是全球威胁,导致新的感染浪潮.此外,其他冠状病毒,如中东呼吸综合征冠状病毒(MERS-CoV,以前称为hCoV-EMC),这种疾病于2012年首次出现,持续存在并继续对人类构成严重疾病的威胁。新型冠状病毒的持续鉴定,加上不同菌株之间遗传重组的潜力,增加了全球关注的新型冠状病毒进化枝出现的可能性。因此,迫切需要pan-CoV治疗药物和疫苗。在HCV蛋白酶抑制剂筛选的广泛优化之后,发现了一种新型3CLPro抑制剂(MK-7845),随后进行了分析.MK-7845表现出针对一组临床SARS-CoV-2亚变体和MERS-CoV的具有广谱活性的纳摩尔体外效力。此外,口服时,在感染SARS-CoV-2(K18-hACE2小鼠)和MERS-CoV(K18-hDDP4小鼠)的转基因小鼠的肺中,MK-7845显示病毒负荷显著降低>6个对数数量级。
    Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) continues to be a global threat due to its ability to evolve and generate new subvariants, leading to new waves of infection. Additionally, other coronaviruses like Middle East respiratory syndrome coronavirus (MERS-CoV, formerly known as hCoV-EMC), which first emerged in 2012, persist and continue to present a threat of severe illness to humans. The continued identification of novel coronaviruses, coupled with the potential for genetic recombination between different strains, raises the possibility of new coronavirus clades of global concern emerging. As a result, there is a pressing need for pan-CoV therapeutic drugs and vaccines. After the extensive optimization of an HCV protease inhibitor screening hit, a novel 3CLPro inhibitor (MK-7845) was discovered and subsequently profiled. MK-7845 exhibited nanomolar in vitro potency with broad spectrum activity against a panel of clinical SARS-CoV-2 subvariants and MERS-CoV. Furthermore, when administered orally, MK-7845 demonstrated a notable reduction in viral burdens by >6 log orders in the lungs of transgenic mice infected with SARS-CoV-2 (K18-hACE2 mice) and MERS-CoV (K18-hDDP4 mice).
    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
    RNA病毒SARS-CoV,SARS-CoV-2和MERS-CoV编码非结构性Nsp16(2'-O-甲基转移酶),该酶催化甲基从S-腺苷甲硫氨酸(SAM)转移到mRNA中的第一个核糖核苷酸。最近,已经发现,Nsp16和SAM底物之间的键断裂导致mRNA病毒复制的停止。迄今为止,只有有限数量的此类抑制剂被鉴定出来,这可以归因于缺乏有效的“配方”。我们研究的目的是提出并验证专门用于此类目的的快速有效的筛查方案。我们提出了四个描述结构结合强度的新指标(结构结合亲和力,结构-氢键,然后应用结构-空间和结构-蛋白质-配体指数),并显示出非常有助于确定响应于配体结构相对较小的变化而增加或减少结合亲和力的程度。经过初步的预选,基于与SAM的相似性,我们将研究限于967种化合物,所谓的分子变色龙。然后将它们停靠在Nsp16蛋白质口袋中,使用新的结构结合亲和力指数选择10个候选配体。随后,将所选择的10个候选配体和8种已知的抑制剂对接至来自SARS-CoV-2、MERS-CoV和SARS-CoV的Nsp16口袋。根据四个新指数,选择了最好的配体,并通过调整它们设计了一个新的配体。最后,对最佳配体进行了ADMET分析和分子动力学模拟。新的结构结合强度指数不仅可以成功地用于筛选和调整配体,而且还要确定配体对目标病毒实体变化的反应的有效性,这对于在病毒蛋白改变的情况下评估药物有效性特别有用。开发的方法,所谓的变色龙策略,有能力将一种新的通用范式引入药物设计领域,包括RNA抗病毒药物.
    The RNA viruses SARS-CoV, SARS-CoV-2 and MERS-CoV encode the non-structural Nsp16 (2\'-O-methyltransferase) that catalyzes the transfer of a methyl group from S-adenosylmethionine (SAM) to the first ribonucleotide in mRNA. Recently, it has been found that breaking the bond between Nsp16 and SAM substrate results in the cessation of mRNA virus replication. To date, only a limited number of such inhibitors have been identified, which can be attributed to a lack of an effective \"recipe\". The aim of our study was to propose and verify a rapid and effective screening protocol dedicated to such purposes. We proposed four new indices describing structure-binding strength (structure-binding affinity, structure-hydrogen bonding, structure-steric and structure-protein-ligand indices) were then applied and shown to be extremely helpful in determining the degree of increase or decrease in binding affinity in response to a relatively small change in the ligand structure. After initial pre-selection, based on similarity to SAM, we limited the study to 967 compounds, so-called molecular chameleons. They were then docked in the Nsp16 protein pocket, and 10 candidate ligands were selected using the novel structure-binding affinity index. Subsequently the selected 10 candidate ligands and 8 known inhibitors and were docked to Nsp16 pockets from SARS-CoV-2, MERS-CoV and SARS-CoV. Based on the four new indices, the best ligands were selected and a new one was designed by tuning them. Finally, ADMET profiling and molecular dynamics simulations were performed for the best ligands. The new structure-binding strength indices can be successfully applied not only to screen and tune ligands, but also to determine the effectiveness of the ligand in response to changes in the target viral entity, which is particularly useful for assessing drug effectiveness in the case of alterations in viral proteins. The developed approach, the so-called chameleon strategy, has the capacity to introduce a novel universal paradigm to the field of drugs design, including RNA antivirals.
    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
    中东呼吸综合征冠状病毒(MERS-CoV)于2012年首次出现,并在流行地区引起人类感染。针对MERS-CoV开发的疫苗和治疗剂集中于刺突(S)糖蛋白以防止病毒进入靶细胞。这些努力受限于对感染引起的抗体应答的理解不足。这里,我们分析了MERS-CoV感染者血浆中的S-导向抗体反应.我们观察到,结合和中和抗体在症状发作/住院后1-6周达到峰值,坚持至少6个月,并中和人类和骆驼MERS-CoV菌株。我们表明MERS-CoVS1亚基是免疫显性的,并且靶向S1的抗体,特别是受体结合域(RBD),占大多数血浆中和活性。抗原位点定位显示血浆抗体经常靶向RBD表位,而S2亚基表位的靶向是罕见的。我们的数据揭示了MERS-CoV感染引起的体液免疫反应,这将指导疫苗和治疗设计。
    Middle East respiratory syndrome coronavirus (MERS-CoV) first emerged in 2012 and causes human infections in endemic regions. Vaccines and therapeutics in development against MERS-CoV focus on the spike (S) glycoprotein to prevent viral entry into target cells. These efforts are limited by a poor understanding of antibody responses elicited by infection. Here, we analyze S-directed antibody responses in plasma collected from MERS-CoV-infected individuals. We observe that binding and neutralizing antibodies peak 1-6 weeks after symptom onset/hospitalization, persist for at least 6 months, and neutralize human and camel MERS-CoV strains. We show that the MERS-CoV S1 subunit is immunodominant and that antibodies targeting S1, particularly the receptor-binding domain (RBD), account for most plasma neutralizing activity. Antigenic site mapping reveals that plasma antibodies frequently target RBD epitopes, whereas targeting of S2 subunit epitopes is rare. Our data reveal the humoral immune responses elicited by MERS-CoV infection, which will guide vaccine and therapeutic design.
    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