Spike Glycoprotein, Coronavirus

刺突糖蛋白 ,冠状病毒
  • 文章类型: Journal Article
    SARS-CoV-2感染是由刺突糖蛋白通过其受体结合域与人血管紧张素转换酶2(ACE2)受体结合而引发的。阻断这种相互作用已被证明是抑制病毒感染的有效方法。在这里,我们报告了一种名为VHH60的中和纳米抗体的发现,它是在很短的时间内从基于商业化纳米抗体的工程纳米抗体库直接生产的。通过结构分析确定,VHH60与人ACE2竞争结合S351,S470-471和S493-494处的Spike蛋白的受体结合域,亲和力为2.56nM。它可以抑制祖先SARS-CoV-2毒株和带有SARS-CoV-2野生型的假型病毒的感染,纳摩尔水平的关键突变或变异。此外,在体内SARS-CoV-2鼻腔感染后,VHH60抑制SARS-CoV-2感染和传播能力提高了50倍,并且保护小鼠免于死亡的时间是对照组的两倍。因此,VHH60不仅是一种强大的纳米抗体,具有疾病控制的前景,而且为产生治疗性纳米抗体的高效和快速方法提供了证据。
    SARS-CoV-2 infection is initiated by Spike glycoprotein binding to the human angiotensin-converting enzyme 2 (ACE2) receptor via its receptor binding domain. Blocking this interaction has been proven to be an effective approach to inhibit virus infection. Here we report the discovery of a neutralizing nanobody named VHH60, which was directly produced from an engineering nanobody library based on a commercialized nanobody within a very short period. VHH60 competes with human ACE2 to bind the receptor binding domain of the Spike protein at S351, S470-471and S493-494 as determined by structural analysis, with an affinity of 2.56 nM. It inhibits infections of both ancestral SARS-CoV-2 strain and pseudotyped viruses harboring SARS-CoV-2 wildtype, key mutations or variants at the nanomolar level. Furthermore, VHH60 suppressed SARS-CoV-2 infection and propagation 50-fold better and protected mice from death for twice as long as the control group after SARS-CoV-2 nasal infections in vivo. Therefore, VHH60 is not only a powerful nanobody with a promising profile for disease control but also provides evidence for a highly effective and rapid approach to generating therapeutic nanobodies.
    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
    严重急性呼吸系统综合症冠状病毒2(SARS-CoV-2)是一种积极意义,含有单链RNA基因组的病毒,已经感染了全世界数百万人。病毒变异得足够快,导致新的变体和子变体的出现,据报道这些变体是从中国武汉市传播的,病毒的中心,到中国和世界各地。病毒基因组中突变的发生,特别是在病毒刺突蛋白区域,已经导致了多种变体和亚变体的进化,这给病毒带来了宿主免疫逃避的好处,从而使现代疫苗和疗法无效。因此,持续需要研究SARS-CoV-2变种的遗传特征和进化动态.因此,在这项研究中,来自中国太原和武汉的SARS-CoV-2变体的832个完整基因组进行了遗传表征,并使用系统发育学研究了它们的系统发育和进化动力学,遗传相似性,和系统发育网络分析。这项研究表明,太原和武汉最普遍的四个谱系如下:Omicron谱系EG.5.1.1,其次是HK.3,FY.3和XBB.1.16(穿山甲分类),和进化枝23F(EG.5.1),其次是23H(HK.3),22F(XBB),和23D(XBB.1.9)(Nextclade分类),谱系B,其次是OmicronFY.3,谱系A,和OmicronFL.3(穿山甲分类),和进化枝19A,其次是22F(XBB),23F(EG.5.1),和23H(HK.3)(下分类),分别。此外,我们的遗传相似性分析表明,来自武汉的SARS-CoV-2进化枝19A-B.4(名称以412981开头)与来自太原的OmicronXBB.2.3.2(名称以18495234开头)的查询序列相比,在基因组的尖峰区域具有约95.5%的遗传相似性。其次是太原的OmicronFR.1.4(名称以18495199开头),相似度为97.2%,OmicronDY.3(名称以17485740开头)相似度为97.9%。其余变体与来自太原的OmicronXBB.2.3.2的查询序列(名称以18495234开头)显示≥98%的相似性。此外,我们的重组分析结果表明,SARS-CoV-2变体具有三个统计学意义的重组事件,这可能导致OmicronXBB.1.16的出现(重组事件3),FY.3(重组事件5),和FL.2.4(重组事件7),暗示了一些关于病毒进化的非常重要的信息。此外,我们的系统发育树和网络分析显示,总共有14个簇和超过10,000个突变,这些突变可能导致了簇I的出现,其次是47个突变,导致II簇的出现等等。这两个城市的病毒变体的聚类揭示了有关其中病毒的系统动力学的重要信息。我们的时间系统发育分析结果表明,太原的变异很可能是独立于武汉变异的独立变异。这项研究,据我们所知,是中国太原和武汉城市之间的首次遗传比较研究。这项研究将帮助我们更好地了解病毒,并应对新变种在本地和国际层面的出现和传播,并随时通知公共卫生当局,以便他们在设计新的病毒疫苗和疗法时做出更好的决定。它还将帮助疫情调查人员更好地检查未来的任何疫情。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense, single-stranded RNA genome-containing virus which has infected millions of people all over the world. The virus has been mutating rapidly enough, resulting in the emergence of new variants and sub-variants which have reportedly been spread from Wuhan city in China, the epicenter of the virus, to the rest of China and all over the world. The occurrence of mutations in the viral genome, especially in the viral spike protein region, has resulted in the evolution of multiple variants and sub-variants which gives the virus the benefit of host immune evasion and thus renders modern-day vaccines and therapeutics ineffective. Therefore, there is a continuous need to study the genetic characteristics and evolutionary dynamics of the SARS-CoV-2 variants. Hence, in this study, a total of 832 complete genomes of SARS-CoV-2 variants from the cities of Taiyuan and Wuhan in China was genetically characterized and their phylogenetic and evolutionary dynamics studied using phylogenetics, genetic similarity, and phylogenetic network analyses. This study shows that the four most prevalent lineages in Taiyuan and Wuhan are as follows: the Omicron lineages EG.5.1.1, followed by HK.3, FY.3, and XBB.1.16 (Pangolin classification), and clades 23F (EG.5.1), followed by 23H (HK.3), 22F (XBB), and 23D (XBB.1.9) (Nextclade classification), and lineage B followed by the Omicron FY.3, lineage A, and Omicron FL.2.3 (Pangolin classification), and the clades 19A, followed by 22F (XBB), 23F (EG.5.1), and 23H (HK.3) (Nextclade classification), respectively. Furthermore, our genetic similarity analysis show that the SARS-CoV-2 clade 19A-B.4 from Wuhan (name starting with 412981) has the least genetic similarity of about 95.5% in the spike region of the genome as compared to the query sequence of Omicron XBB.2.3.2 from Taiyuan (name starting with 18495234), followed by the Omicron FR.1.4 from Taiyuan (name starting with 18495199) with ~97.2% similarity and Omicron DY.3 (name starting with 17485740) with ~97.9% similarity. The rest of the variants showed ≥98% similarity with the query sequence of Omicron XBB.2.3.2 from Taiyuan (name starting with 18495234). In addition, our recombination analysis results show that the SARS-CoV-2 variants have three statistically significant recombinant events which could have possibly resulted in the emergence of Omicron XBB.1.16 (recombination event 3), FY.3 (recombination event 5), and FL.2.4 (recombination event 7), suggesting some very important information regarding viral evolution. Also, our phylogenetic tree and network analyses show that there are a total of 14 clusters and more than 10,000 mutations which may have probably resulted in the emergence of cluster-I, followed by 47 mutations resulting in the emergence of cluster-II and so on. The clustering of the viral variants of both cities reveals significant information regarding the phylodynamics of the virus among them. The results of our temporal phylogenetic analysis suggest that the variants of Taiyuan have likely emerged as independent variants separate from the variants of Wuhan. This study, to the best of our knowledge, is the first ever genetic comparative study between Taiyuan and Wuhan cities in China. This study will help us better understand the virus and cope with the emergence and spread of new variants at a local as well as an international level, and keep the public health authorities informed for them to make better decisions in designing new viral vaccines and therapeutics. It will also help the outbreak investigators to better examine any future outbreak.
    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
    目前,SARS-CoV-2已经演变成各种变体,包括许多高度突变的Omicron子谱系,显著提高免疫逃避能力。这一发展引发了人们对可用疫苗和基于抗体的疗法的有效性可能降低的担忧。这里,我们描述了那些具有代表性的广泛中和抗体(bnAb)类别,它们对包括Omicron亚谱系在内的新出现的变体保留了显著的有效性.分子特征,表位保守,并进一步详述了这些抗体的抗性机制,旨在为治疗性抗体的发展提供建议或方向,并促进具有广谱潜力的疫苗的设计。
    Currently, SARS-CoV-2 has evolved into various variants, including the numerous highly mutated Omicron sub-lineages, significantly increasing immune evasion ability. The development raises concerns about the possibly diminished effectiveness of available vaccines and antibody-based therapeutics. Here, we describe those representative categories of broadly neutralizing antibodies (bnAbs) that retain prominent effectiveness against emerging variants including Omicron sub-lineages. The molecular characteristics, epitope conservation, and resistance mechanisms of these antibodies are further detailed, aiming to offer suggestion or direction for the development of therapeutic antibodies, and facilitate the design of vaccines with broad-spectrum potential.
    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
    突变推动了严重急性呼吸综合征冠状病毒2(SARS-CoV-2)新变种的进化和发展,潜在的传播性增加,疾病严重程度和疫苗逃逸等。基因组测序是一种允许科学家阅读生物体遗传密码的技术,并已成为研究新兴传染病的有力工具。这里,我们在赞比亚东部省的某些地区进行了横断面研究,从2021年11月到2022年2月。我们使用高通量测序分析了SARS-CoV-2样品(n=76)。在69个SARS-CoV-2基因组中鉴定出总共4097个突变,其中47%(1925/4097)的突变发生在刺突蛋白中。我们在七个Omicron亚谱系(BA.1,BA.1.1,BA.1.14,BA.1.18,BA.1.21,BA.2,BA.2.23和XT)的刺突蛋白中鉴定了83个独特的氨基酸突变。其中,43.4%(36/83)存在于受体结合域中,而14.5%(12/83)在受体结合基序中。虽然我们确定了一个潜在的重组XT菌株,高度传播的BA.2亚谱系占主导地位(40.8%)。我们观察到东部省Omicron菌株对其他变体的替代。这项工作表明了大流行防备的重要性以及监测普通人群疾病的必要性。
    Mutations have driven the evolution and development of new variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with potential implications for increased transmissibility, disease severity and vaccine escape among others. Genome sequencing is a technique that allows scientists to read the genetic code of an organism and has become a powerful tool for studying emerging infectious diseases. Here, we conducted a cross-sectional study in selected districts of the Eastern Province of Zambia, from November 2021 to February 2022. We analyzed SARS-CoV-2 samples (n = 76) using high-throughput sequencing. A total of 4097 mutations were identified in 69 SARS-CoV-2 genomes with 47% (1925/4097) of the mutations occurring in the spike protein. We identified 83 unique amino acid mutations in the spike protein of the seven Omicron sublineages (BA.1, BA.1.1, BA.1.14, BA.1.18, BA.1.21, BA.2, BA.2.23 and XT). Of these, 43.4% (36/83) were present in the receptor binding domain, while 14.5% (12/83) were in the receptor binding motif. While we identified a potential recombinant XT strain, the highly transmissible BA.2 sublineage was more predominant (40.8%). We observed the substitution of other variants with the Omicron strain in the Eastern Province. This work shows the importance of pandemic preparedness and the need to monitor disease in the general population.
    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
    背景:尽管COVID-19引起了广泛的神经系统症状,并且在COVID-19患者死后数月的大脑样本中发现了SARS-CoV-2,由于缺乏研究模型,SARS-CoV-2侵入中枢神经系统的确切机制仍不清楚。
    方法:我们收集了近期有COVID-19病史的神经胶质瘤患者的神经胶质瘤组织样本,并检查了SARS-CoV-2刺突蛋白的存在。随后,对正常脑组织进行空间转录组学分析,神经胶质瘤组织,和最近有COVID-19病史的神经胶质瘤患者的神经胶质瘤组织。此外,收集并分析来自神经胶质瘤组织和神经胶质瘤类器官的单细胞测序数据.神经胶质瘤类器官用于评估潜在COVID-19阻断剂的疗效。
    结果:近期有COVID-19病史的神经胶质瘤患者的神经胶质瘤组织显示存在SARS-CoV-2刺突蛋白。近期有COVID-19病史的神经胶质瘤患者的神经胶质瘤组织与主要表现在神经元细胞的健康脑组织之间的差异。值得注意的是,COVID-19病史的神经胶质瘤组织内的神经元细胞显示出对阿尔茨海默病的易感性增加,抑郁症,和突触功能障碍,指示神经元畸变。在神经胶质瘤组织和神经胶质瘤类器官中均证实了SARS-CoV-2进入因子的表达。此外,神经胶质瘤类器官易受假性SARS-CoV-2感染的影响,感染可能被潜在的COVID-19药物部分阻断。
    结论:胶质瘤具有固有的特征,使其对SARS-CoV-2感染易感,导致其具有COVID-19神经系统症状的代表性。这为利用神经胶质瘤类器官作为SARS-CoV-2感染中枢神经系统的研究和阻断药物测试模型的合理性和可行性奠定了生物学基础。
    BACKGROUND: Despite the extensive neurological symptoms induced by COVID-19 and the identification of SARS-CoV-2 in post-mortem brain samples from COVID-19 patients months after death, the precise mechanisms of SARS-CoV-2 invasion into the central nervous system remain unclear due to the lack of research models.
    METHODS: We collected glioma tissue samples from glioma patients who had a recent history of COVID-19 and examined the presence of the SARS-CoV-2 spike protein. Subsequently, spatial transcriptomic analyses were conducted on normal brain tissues, glioma tissues, and glioma tissues from glioma patients with recent COVID-19 history. Additionally, single-cell sequencing data from both glioma tissues and glioma organoids were collected and analyzed. Glioma organoids were utilized to evaluate the efficacy of potential COVID-19 blocking agents.
    RESULTS: Glioma tissues from glioma patients with recent COVID-19 history exhibited the presence of the SARS-CoV-2 spike protein. Differences between glioma tissues from glioma patients who had a recent history of COVID-19 and healthy brain tissues primarily manifested in neuronal cells. Notably, neuronal cells within glioma tissues of COVID-19 history demonstrated heightened susceptibility to Alzheimer\'s disease, depression, and synaptic dysfunction, indicative of neuronal aberrations. Expressions of SARS-CoV-2 entry factors were confirmed in both glioma tissues and glioma organoids. Moreover, glioma organoids were susceptible to pseudo-SARS-CoV-2 infection and the infections could be partly blocked by the potential COVID-19 drugs.
    CONCLUSIONS: Gliomas had inherent traits that render them susceptible to SARS-CoV-2 infection, leading to their representability of COVID-19 neurological symptoms. This established a biological foundation for the rationality and feasibility of utilization of glioma organoids as research and blocking drug testing model in SARS-CoV-2 infection within the central nervous system.
    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
    严重急性呼吸道综合征冠状病毒-2(SARS-CoV-2)是当前冠状病毒疾病大流行的原因。随着变异菌株的快速进化,找到有效的刺突蛋白抑制剂是一个合乎逻辑和关键的优先事项。血管紧张素转换酶2(ACE2)已被确定为SARS-CoV-2病毒进入的功能受体,因此,与刺突蛋白-ACE2相互作用相关的相关治疗方法显示出抑制病毒感染的高度可行性。我们的计算机辅助药物设计(CADD)方法仔细分析了来自美国国家癌症研究所(NCI)数据库的260,000多个化合物记录,确定潜在的尖峰抑制剂。选择刺突蛋白受体结合域(RBD)作为我们的虚拟筛选过程的靶蛋白。在基于单元格的验证中,利用携带报告基因的SARS-CoV-2假病毒来筛选有效的化合物。最终,化合物C2,C8和C10对SARS-CoV-2具有显着的抗病毒活性,估计EC50值为8.8μM,6.7μM,和7.6μM,分别。以上述化合物为模板,产生了十种衍生物,强大的生物测定结果表明,C8.2(EC50=5.9μM)表现出最强的抗病毒功效。化合物C8.2还显示出对Omicron变体的抑制活性,EC50为9.3μM。因此,CADD方法成功地发现了能够抑制病毒感染的与刺突蛋白RBD结合的先导化合物。
    Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the current coronavirus disease pandemic. With the rapid evolution of variant strains, finding effective spike protein inhibitors is a logical and critical priority. Angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor for SARS-CoV-2 viral entry, and thus related therapeutic approaches associated with the spike protein-ACE2 interaction show a high degree of feasibility for inhibiting viral infection. Our computer-aided drug design (CADD) method meticulously analyzed more than 260,000 compound records from the United States National Cancer Institute (NCI) database, to identify potential spike inhibitors. The spike protein receptor-binding domain (RBD) was chosen as the target protein for our virtual screening process. In cell-based validation, SARS-CoV-2 pseudovirus carrying a reporter gene was utilized to screen for effective compounds. Ultimately, compounds C2, C8, and C10 demonstrated significant antiviral activity against SARS-CoV-2, with estimated EC50 values of 8.8 μM, 6.7 μM, and 7.6 μM, respectively. Using the above compounds as templates, ten derivatives were generated and robust bioassay results revealed that C8.2 (EC50 = 5.9 μM) exhibited the strongest antiviral efficacy. Compounds C8.2 also displayed inhibitory activity against the Omicron variant, with an EC50 of 9.3 μM. Thus, the CADD method successfully discovered lead compounds binding to the spike protein RBD that are capable of inhibiting viral 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
    黏膜免疫在对抗和控制高度突变的严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)的传播中起着至关重要的作用。重组亚单位疫苗在临床试验中显示出安全性和有效性,但需要进一步研究以评估其作为粘膜疫苗的可行性。这项研究使用原型菌株的刺突(S)蛋白和omicron变体开发了SARS-CoV-2粘膜疫苗,随着阳离子壳聚糖佐剂,并系统地评估了小鼠初次和加强免疫后的免疫原性。通过腹膜内和鼻内施用S蛋白的初次免疫引发针对原型菌株的交叉反应抗体,以及三角洲和omicron变体,在粘膜疫苗接种后观察到特别强的效果。在用灭活疫苗初次免疫后加强免疫的情况下,与原型疫苗相比,基于omicron的S蛋白粘膜疫苗在血清和呼吸道粘膜中产生了更广泛和更强大的中和抗体反应,增强对不同变体的保护。这些发现表明,在初次免疫和加强免疫期间,用S蛋白进行粘膜接种有可能引发更广泛和更强的抗体反应。使其成为对抗呼吸道病原体的有希望的策略。
    Mucosal immunity plays a crucial role in combating and controlling the spread of highly mutated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Recombinant subunit vaccines have shown safety and efficacy in clinical trials, but further investigation is necessary to evaluate their feasibility as mucosal vaccines. This study developed a SARS-CoV-2 mucosal vaccine using spike (S) proteins from a prototype strain and the omicron variant, along with a cationic chitosan adjuvant, and systematically evaluated its immunogenicity after both primary and booster immunization in mice. Primary immunization through intraperitoneal and intranasal administration of the S protein elicited cross-reactive antibodies against prototype strains, as well as delta and omicron variants, with particularly strong effects observed after mucosal vaccination. In the context of booster immunization following primary immunization with inactivated vaccines, the omicron-based S protein mucosal vaccine resulted in a broader and more robust neutralizing antibody response in both serum and respiratory mucosa compared to the prototype vaccine, enhancing protection against different variants. These findings indicate that mucosal vaccination with the S protein has the potential to trigger a broader and stronger antibody response during primary and booster immunization, making it a promising strategy against respiratory pathogens.
    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
    SARS-CoV-2的Omicron亚变体BQ.1.1,XBB.1.5和XBB.1.16以其擅长逃避免疫反应而闻名。这里,我们分离出一种中和抗体,7F3,具有中和所有测试的SARS-CoV-2变体的能力,包括BQ.1.1、XBB.1.5和XBB.1.16。7F3靶向受体结合基序(RBM)区域,并表现出与一组37个RBD突变蛋白的广泛结合。我们使用7F3和交叉中和抗体GW01开发了IgG样双特异性抗体G7-Fc。G7-Fc对所有28个测试的SARS-CoV-2变体和肉瘤病毒都表现出强大的中和活性,在K18-ACE和BALB/c雌性小鼠中提供针对XBB.1感染的有效预防和治疗功效。与OmicronXBB尖峰(S)三聚体复合物中G7-Fc的冷冻-EM结构分析揭示了三聚体-二聚体构象,G7-Fc协同靶向两个不同的RBD表位并阻断ACE2结合。7F3和LY-CoV1404表位的比较分析突出显示了由7F3结合的RBM区中的独特且高度保守的表位,从而促进免疫逃避性Omicron变体XBB.1.16的中和。G7-Fc有望作为针对SARS-CoV-2的潜在预防对策,特别是针对循环和新出现的变体。
    The Omicron subvariants BQ.1.1, XBB.1.5, and XBB.1.16 of SARS-CoV-2 are known for their adeptness at evading immune responses. Here, we isolate a neutralizing antibody, 7F3, with the capacity to neutralize all tested SARS-CoV-2 variants, including BQ.1.1, XBB.1.5, and XBB.1.16. 7F3 targets the receptor-binding motif (RBM) region and exhibits broad binding to a panel of 37 RBD mutant proteins. We develop the IgG-like bispecific antibody G7-Fc using 7F3 and the cross-neutralizing antibody GW01. G7-Fc demonstrates robust neutralizing activity against all 28 tested SARS-CoV-2 variants and sarbecoviruses, providing potent prophylaxis and therapeutic efficacy against XBB.1 infection in both K18-ACE and BALB/c female mice. Cryo-EM structure analysis of the G7-Fc in complex with the Omicron XBB spike (S) trimer reveals a trimer-dimer conformation, with G7-Fc synergistically targeting two distinct RBD epitopes and blocking ACE2 binding. Comparative analysis of 7F3 and LY-CoV1404 epitopes highlights a distinct and highly conserved epitope in the RBM region bound by 7F3, facilitating neutralization of the immune-evasive Omicron variant XBB.1.16. G7-Fc holds promise as a potential prophylactic countermeasure against SARS-CoV-2, particularly against circulating and emerging variants.
    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
    严重急性呼吸道综合症冠状病毒(SARS-CoV-2)大流行对全球公共卫生安全产生了重大影响,迫切需要开发有效的抗病毒药物。先前的研究发现,与ACE2结合是SARS-CoV-2侵入宿主细胞的关键步骤,因此,阻断ACE2可以抑制病毒的侵袭,但很少有关于这种特异性抑制剂的报道。我们先前的研究发现,三尖杉酯碱(HT)可以抑制SARS-CoV-2刺突假病毒进入ACE2h细胞,但其相对较高的细胞毒性限制了其进一步发展。活性组分的氨基酸修饰可以增加其溶解度并降低其细胞毒性。因此,在这项研究中,通过对其核心结构的氨基酸修饰合成了七种新的衍生物。通过细胞活力测定和SARS-CoV-2刺突假病毒进入测定来评估目标化合物。化合物CET-1显著抑制假病毒进入ACE2h细胞,并且显示出比HT更小的细胞毒性。分子对接结果表明,CET-1与HT一样,可以与ACE2的重要残基TYR83结合。总之,我们的研究提供了一种新的化合物,在抑制SARS-CoV-2尖峰假病毒感染方面具有比HT更高的潜在活性和更低的毒性,这使得将来成为抗病毒药物的先导化合物成为可能。
    The severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic has triggered a significant impact on global public health security, it is urgent to develop effective antiviral drugs. Previous studies have found that binding to ACE2 is a key step in the invasion of SARS-CoV-2 into host cells, so virus invasion can be inhibited by blocking ACE2, but there are few reports on this kind of specific inhibitor. Our previous study found that Harringtonine (HT) can inhibit the entry of SARS-CoV-2 spike pseudovirus into ACE2h cells, but its relatively high cytotoxicity limits its further development. Amino acid modification of the active components can increase their solubility and reduce their cytotoxicity. Therefore, in this study, seven new derivatives were synthesized by amino acid modification of its core structure Cephalotaxine. The target compounds were evaluated by cell viability assay and the SARS-CoV-2 spike pseudovirus entry assay. Compound CET-1 significantly inhibited the entry of pseudovirus into ACE2h cells and showed less cytotoxicity than HT. Molecular docking results showed that CET-1 could bind TYR83, an important residue of ACE2, just like HT. In conclusion, our study provided a novel compound with more potential activity and lower toxicity than HT on inhibiting the SARS-CoV-2 spike pseudovirus infection, which makes it possible to be a lead compound as an antiviral drug in the future.
    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
    治疗性单克隆抗体(mAb)与不断出现的严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)变体之间的博弈有利于该病毒,因为大多数治疗性单克隆抗体都被逃避了。应对这一挑战,在这项研究中,我们系统地探索了可重复的双特异性抗体(bsAb)依赖性协同作用。当其单个mAb中的任何一个被变体逃脱时,它可以有效地恢复bsAb的中和活性。这种协同作用主要归因于受体结合结构域(RBD)-5的结合角度,促进了刺间交联并促进了经典抗体混合物无法实现的隐蔽表位暴露。此外,RBD-5与RBD-2、RBD-6和RBD-7以及RBD-8也表现出显著增强的效果。这项研究不仅改变了理解抗体相互作用的范式,而且为开发针对快速突变的SARS-CoV-2的更有效的治疗性抗体铺平了道路,Dia-19已经显示出针对BA.2.86,EG.1等新兴变体的希望。和JN.1。
    The game between therapeutic monoclonal antibodies (mAbs) and continuously emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has favored the virus, as most therapeutic mAbs have been evaded. Addressing this challenge, we systematically explored a reproducible bispecific antibody (bsAb)-dependent synergistic effect in this study. It could effectively restore the neutralizing activity of the bsAb when any of its single mAbs is escaped by variants. This synergy is primarily attributed to the binding angle of receptor-binding domain (RBD)-5, facilitating inter-spike cross-linking and promoting cryptic epitope exposure that classical antibody cocktails cannot achieve. Furthermore, RBD-5 with RBD-2, RBD-6, and RBD-7, alongside RBD-8, also exhibit significantly enhanced effects. This study not only shifts the paradigm in understanding antibody interactions but paves the way for developing more effective therapeutic antibodies against rapidly mutating SARS-CoV-2, with Dia-19 already showing promise against emerging variants like BA.2.86, EG.5.1, and JN.1.
    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