receptor binding domain

受体结合域
  • 文章类型: Journal Article
    通过使用人单克隆抗体组(PD4,PD5,sc23和sc29)成像,在Vero细胞中检查了重组SARS-CoV-2S蛋白的表达。PD4和sc29抗体识别内质网和高尔基复合体的S2蛋白亚基中的构象特异性表位。虽然PD5和sc23在高尔基体的S1蛋白亚基中检测到构象特异性表位,只有PD5识别受体结合域(RBD)。PD5与识别S1亚基和RBD的非构象特异性抗体的染色模式的比较表明,PD5识别S1蛋白亚基内的构象结构。我们的数据表明,在S蛋白转运过程中,在分泌途径的不同位置形成的人单克隆抗体识别的抗体结合表位,但是高尔基复合体S1蛋白亚基的构象变化形成了抗体结合表位,这些表位被病毒中和抗体识别。
    Recombinant SARS-CoV-2 S protein expression was examined in Vero cells by imaging using the human monoclonal antibody panel (PD4, PD5, sc23, and sc29). The PD4 and sc29 antibodies recognised conformational specific epitopes in the S2 protein subunit at the Endoplasmic reticulum and Golgi complex. While PD5 and sc23 detected conformationally specific epitopes in the S1 protein subunit at the Golgi complex, only PD5 recognised the receptor binding domain (RBD). A comparison of the staining patterns of PD5 with non-conformationally specific antibodies that recognises the S1 subunit and RBD suggested the PD5 recognised a conformational structure within the S1 protein subunit. Our data suggests the antibody binding epitopes recognised by the human monoclonal antibodies formed at different locations in the secretory pathway during S protein transport, but a conformational change in the S1 protein subunit at the Golgi complex formed antibody binding epitopes that are recognised by virus neutralising antibodies.
    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
    严重急性呼吸综合征冠状病毒2(SARS-CoV-2)Omicron变体的重组谱系,名为XBB,出现于2022年底,并进化出后代,先后席卷了当地和全球人口。XBB谱系成员因其改善的免疫逃避和传播性而被注意到。这里,我们确定了XBB.1.5,XBB.1.16,EG.5和EG.5.1尖峰(S)胞外域的低温电子显微镜(cryo-EM)结构,以揭示先前在BA.1和BA.2中观察到的增强的3受体结合域(RBD)-down受体-不可接近的封闭状态改进的XBB.1.5和XBB.1.16RBD稳定性补偿了早期Omicron突变引起的稳定性损失,而F456L取代降低EG.5RBD稳定性。S1亚基突变对S2亚基的构象和表位呈递有长期影响。我们的结果表明,通过同时优化多个参数,持续的S蛋白进化,包括稳定性,受体结合,和免疫逃避,以及相对较少的残基取代在改变S蛋白构象格局中的戏剧性影响。
    A recombinant lineage of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant, named XBB, appeared in late 2022 and evolved descendants that successively swept local and global populations. XBB lineage members were noted for their improved immune evasion and transmissibility. Here, we determine cryoelectron microscopy (cryo-EM) structures of XBB.1.5, XBB.1.16, EG.5, and EG.5.1 spike (S) ectodomains to reveal reinforced 3-receptor binding domain (RBD)-down receptor-inaccessible closed states mediated by interprotomer RBD interactions previously observed in BA.1 and BA.2. Improved XBB.1.5 and XBB.1.16 RBD stability compensated for stability loss caused by early Omicron mutations, while the F456L substitution reduced EG.5 RBD stability. S1 subunit mutations had long-range impacts on conformation and epitope presentation in the S2 subunit. Our results reveal continued S protein evolution via simultaneous optimization of multiple parameters, including stability, receptor binding, and immune evasion, and the dramatic effects of relatively few residue substitutions in altering the S protein conformational landscape.
    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: English Abstract
    本研究旨在开发一种有效的二价亚单位疫苗,该疫苗有望同时预防猪三角洲冠状病毒(PDCoV)和猪流行性腹泻病毒(PEDV)。将PDCoV和PEDV的受体结合域(RBD)融合并克隆到真核表达载体pCDNA3.1()中。通过ExpiCHOTM表达系统表达并纯化融合蛋白PDCoV-RBD-PEDV-RBD(pdRBD-peRBD)。用三种不同剂量(10、20和30μg)的融合蛋白免疫小鼠。通过ELISA和流式细胞术评价融合蛋白诱导的体液免疫应答和细胞免疫应答。通过微中性化试验确定免疫小鼠针对PDCoV和PEDV的血清的中和滴度。结果表明,加强免疫后3个不同剂量组均能诱导高水平的IgG抗体,不同剂量组之间的抗体水平没有显着差异,表明10μg的免疫剂量可以达到良好的免疫效果。流式细胞仪检测结果显示,免疫组CD3+CD4+T细胞比例升高,CD3+CD8+T细胞比例降低,这与亚单位疫苗诱导的体液免疫应答的预期一致。同时,测定血清中白细胞介素(IL)-2,IL-4和干扰素(IFN)-γ的水平。结果表明,融合蛋白可诱导体液免疫效应和细胞免疫应答。中和实验结果表明,10μg融合蛋白诱导的抗体在体外能中和PDCoV和PEDV,滴度分别为1:179.25和1:141.21。以上结果表明,pdRBD-peRBD在10μg剂量下可诱导高水平的体液免疫应答,诱导的抗体可以中和PDCoV和PEDV。因此,pdRBD-peRBD融合蛋白有望成为能同时预防PDCoV和PEDV的有效亚单位疫苗。
    This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 μg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 μg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 μg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 μg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.
    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(SARS-CoV-2)的刺突蛋白负责感染宿主细胞。它有两个部分,S1和S2。S1区段具有与宿主受体血管紧张素转换酶2(ACE2)连接的受体结合域(RBD)。S2区段通过两个七肽重复结构域产生六螺旋束帮助病毒细胞膜的融合。为了开发针对COVID-19的有效疫苗和疗法,表达和纯化SARS-CoV-2Spike蛋白至关重要。已经对完整重组刺突蛋白或其片段的表达进行了广泛的研究。这篇综述提供了用于刺突蛋白表达的不同表达系统的深入分析,以及它们的优点和缺点。
    The spike protein of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is responsible for infecting host cells. It has two segments, S1 and S2. The S1 segment has a receptor-binding domain (RBD) that attaches to the host receptor angiotensin-converting enzyme 2 (ACE2). The S2 segment helps in the fusion of the viral cell membrane by creating a six-helical bundle through the two-heptad repeat domain. To develop effective vaccines and therapeutics against COVID-19, it is critical to express and purify the SARS-CoV-2 Spike protein. Extensive studies have been conducted on expression of a complete recombinant spike protein or its fragments. This review provides an in-depth analysis of the different expression systems employed for spike protein expression, along with their advantages and disadvantages.
    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
    2019年冠状病毒病(COVID-19)大流行,由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)引起,造成了毁灭性的人员和经济损失。疫苗接种仍然是控制COVID-19大流行的最有效方法。尽管如此,SARS-CoV-2变种的持续进化引起了科学界对下一代COVID-19疫苗开发的担忧。其中,考虑到他们的安全,免疫原性,以及展示各种天然表位的灵活性,基于病毒样颗粒(VLP)的疫苗是最有前途的下一代疫苗之一。在这次审查中,我们总结了VLP平台的优势和特点,抗原展示策略,和目前基于VLP平台的SARS-CoV-2疫苗的临床试验进展。重要的是,SARS-CoV-2VLP疫苗开发的经验和教训为制定基于VLP疫苗的预防未来冠状病毒大流行和其他流行病的策略提供了见解。
    The Coronavirus Disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has incurred devastating human and economic losses. Vaccination remains the most effective approach for controlling the COVID-19 pandemic. Nonetheless, the sustained evolution of SARS-CoV-2 variants has provoked concerns among the scientific community regarding the development of next-generation COVID-19 vaccines. Among these, given their safety, immunogenicity, and flexibility to display varied and native epitopes, virus-like particle (VLP)-based vaccines represent one of the most promising next-generation vaccines. In this review, we summarize the advantages and characteristics of VLP platforms, strategies for antigen display, and current clinical trial progress of SARS-CoV-2 vaccines based on VLP platforms. Importantly, the experience and lessons learned from the development of SARS-CoV-2 VLP vaccines provide insights into the development of strategies based on VLP vaccines to prevent future coronavirus pandemics and other epidemics.
    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:猪繁殖与呼吸综合征病毒(PRRSV)是一种流行的猪病原体,近30年来,这对全球养猪业造成了不利影响。然而,由于病毒引起的免疫抑制和PRRSV的遗传多样性,目前还没有成功开发针对病毒的广泛中和策略。抗病毒肽和纳米抗体以其易于生产和在实践中的功效引起了广泛的关注。在这项研究中,通过将PRRSV特异性非中和纳米抗体与靶向PRRSV蛋白的受体结合域(RBD)的CD163衍生肽组合,开发了四种新的融合蛋白,称为纳米抗体肽缀合物(NPC)。
    结果:使用两个纳米抗体分别针对PRRSVN和nsp9成功构建了四个NPC,分别与来自猪CD163的两种抗病毒肽4H7或8H2重组。所有四种NPC均表现出与PRRSV结合的特异性能力以及以剂量依赖性方式对PRRSV的各种谱系的广泛抑制作用。NPC在Nb组分的辅助下通过CD163表位肽干扰PRRSV蛋白的RBD与PRRSV预附着阶段中的CD163的结合。NPC还在附着后阶段抑制病毒复制,抑制作用取决于NPCs中Nb部分的抗病毒功能,包括在长病毒RNA合成中的干扰,NF-κB和IFN-β激活。此外,预测了NPC-N/nsp9-4H7的中和结构域4H7的aaK31和T32位点与PRRSVGP2a的基序171NLRLTG176之间的相互作用。NPC-N/nsp9-8H2的中和结构域8H2的基序28SSS30也可以形成氢与PRRSVGP3的基序152NAFLP156结合。该研究为PRRSV蛋白的RBD的结构特征和潜在功能意义提供了有价值的见解。最后,如小鼠模型所示,体内鼻内接种12-24小时的NPC维持针对PRRSV的显著中和活性。这些发现激发了NPC作为预防措施的潜力,可以降低宿主人群针对PRRSV等呼吸道感染因子的传播风险。
    结论:本研究的目的是开发基于肽的生物活性化合物以中和各种PRRSV毒株。新的抗病毒NPC(纳米抗体肽缀合物)由靶向病毒蛋白的特异性纳米抗体和用于病毒阻断的中和CD163表位肽组成,并提供显著的抗病毒活性。该研究将极大地促进针对PRRSV的抗病毒药物的研发,并启发针对其他病毒性疾病的新策略。
    BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent swine pathogen, which has caused adverse impact on the global swine industry for almost 30 years. However, due to the immune suppression caused by the virus and the genetic diversity in PRRSV, no virus-targeting broad neutralizing strategy has been successfully developed yet. Antiviral peptide and nanobody have attracted extensive attention with the ease in production and the efficacy in practice. In this study, four new fusion proteins named nanobody peptide conjugates (NPCs) were developed by combining PRRSV specific non-neutralizing nanobodies with CD163-derived peptides targeting the receptor binding domain (RBD) of PRRSV proteins.
    RESULTS: Four NPCs were successfully constructed using two nanobodies against PRRSV N and nsp9 individually, recombining with two antiviral peptides 4H7 or 8H2 from porcine CD163 respectively. All four NPCs demonstrated specific capability of binding to PRRSV and broad inhibitory effect against various lineages of PRRSV in a dose-dependent manner. NPCs interfere with the binding of the RBD of PRRSV proteins to CD163 in the PRRSV pre-attachment stage by CD163 epitope peptides in the assistance of Nb components. NPCs also suppress viral replication during the stage of post-attachment, and the inhibitory effects depend on the antiviral functions of Nb parts in NPCs, including the interference in long viral RNA synthesis, NF-κB and IFN-β activation. Moreover, an interaction was predicted between aa K31 and T32 sites of neutralizing domain 4H7 of NPC-N/nsp9-4H7 and the motif 171NLRLTG176 of PRRSV GP2a. The motif 28SSS30 of neutralizing domain 8H2 of NPC-N/nsp9-8H2 could also form hydrogens to bind with the motif 152NAFLP156 of PRRSV GP3. The study provides valuable insights into the structural characteristics and potential functional implications of the RBD of PRRSV proteins. Finally, as indicated in a mouse model, NPC intranasally inoculated in vivo for 12-24 h sustains the significant neutralizing activity against PRRSV. These findings inspire the potential of NPC as a preventive measure to reduce the transmission risk in the host population against respiratory infectious agents like PRRSV.
    CONCLUSIONS: The aim of the current study was to develop a peptide based bioactive compound to neutralize various PRRSV strains. The new antiviral NPC (nanobody peptide conjugate) consists of a specific nanobody targeting the viral protein and a neutralizing CD163 epitope peptide for virus blocking and provides significant antiviral activity. The study will greatly promote the antiviral drug R&D against PRRSV and enlighten a new strategy against other viral diseases.
    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
    由于严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)导致的2019年冠状病毒大流行严重影响了全球公共卫生安全。疫苗研究,中和抗体(NAb)和小分子抗病毒药物目前正在进行。特别是,NAb由于其明确的机制而成为有前途的治疗剂,高特异性,卓越的安全性,易于大规模生产和同时应用于预防和治疗病毒感染。许多NAb疗法已经进入临床研究阶段,显示有希望的治疗和预防效果。这些代理商已根据紧急授权程序用于预防和控制疫情。本综述总结了SARS-CoV-2相关NAb的分子靶标以及NAb开发的筛选和鉴定技术。此外,讨论了目前使用NAb仍然存在的缺点和挑战。本综述的目的是为未来任何突发传染病的NAb的发展提供参考,包括SARS-CoV-2。
    The coronavirus disease 2019 pandemic due to severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) seriously affected global public health security. Studies on vaccines, neutralizing antibodies (NAbs) and small molecule antiviral drugs are currently ongoing. In particular, NAbs have emerged as promising therapeutic agents due to their well‑defined mechanism, high specificity, superior safety profile, ease of large‑scale production and simultaneous application for both prevention and treatment of viral infection. Numerous NAb therapeutics have entered the clinical research stages, demonstrating promising therapeutic and preventive effects. These agents have been used for outbreak prevention and control under urgent authorization processes. The present review summarizes the molecular targets of SARS‑CoV‑2‑associated NAbs and screening and identification techniques for NAb development. Moreover, the current shortcomings and challenges that persist with the use of NAbs are discussed. The aim of the present review is to offer a reference for the development of NAbs for any future emergent infectious diseases, including SARS‑CoV‑2.
    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
    Omicron变体及其亚谱系是目前世界上唯一流行的SARS-CoV-2病毒。在这项研究中,详细检查了Omicron刺突蛋白的分离受体结合域(RBD)的构象稳定性。亲本Omicron谱系在RBD的ACE2结合区中具有超过10个突变,其与其β发夹环结构域特异性相关。通过生物物理分子计算证明,β发夹环结构域中的突变显着增加了环内和环-RBD相互作用的蛋白质内相互作用能。相互作用能增加包括在β发夹环结构域中形成新的氢键,其有助于稳定该关键ACE2结合区。我们的结果也与最近关于Omicron核心β桶域稳定性的实验一致,在其循环域之外,并有助于证明OmicronRBD的整体构象稳定性。通过动态模拟进一步表明,OmicronRBD的未结合状态与结合状态配置保持紧密对齐,这对于野生型RBD没有观察到。总的来说,这些研究证明Omicron的构象稳定性显著高于其野生型构型,并提出了一些问题,即构象稳定性是否可能是SARS-CoV-2病毒突变变化的正选择特征.
    The Omicron variant and its sub-lineages are the only current circulating SARS-CoV-2 viruses worldwide. In this study, the conformational stability of the isolated Receptor Binding Domain (RBD) of Omicron\'s spike protein is examined in detail. The parent Omicron lineage has over ten mutations in the ACE2 binding region of the RBD that are specifically associated with its β hairpin loop domain. It is demonstrated through biophysical molecular computations that the mutations in the β hairpin loop domain significantly increase the intra-protein interaction energies of intra-loop and loop-RBD interactions. The interaction energy increases include the formation of new hydrogen bonds in the β hairpin loop domain that help stabilize this critical ACE2 binding region. Our results also agree with recent experiments on the stability of Omicron\'s core β barrel domain, outside of its loop domain, and help demonstrate the overall conformational stability of the Omicron RBD. It is further shown here through dynamic simulations that the unbound state of the Omicron RBD remains closely aligned with the bound state configuration, which was not observed for the wild-type RBD. Overall, these studies demonstrate the significantly increased conformational stability of Omicron over its wild-type configuration and raise a number of questions on whether conformational stability could be a positive selection feature of SARS-CoV-2 viral mutational changes.
    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
    细小病毒B19(B19V)是一种人类病原体,是婴儿和成人几种疾病的病原体。由于缺乏针对这种病毒的抗病毒药物,治疗方案有限。B19V的次要衣壳蛋白具有独特的N末端,命名为VP1u,这对感染至关重要。VP1u编码受体结合域(RBD),宿主细胞进入所必需的,和磷脂酶A2(PLA2)结构域,对于细胞运输过程中的内体逃逸至关重要。这两个领域都是感染不可或缺的,使RBD成为B19V抑制剂的合理药物靶标,因为它位于病毒的外表面。迄今为止,没有任何细小病毒的VP1u组件的实验性结构信息。在这里,我们报告了B19V的RBD的主链NMR共振分配,并证明了它形成了稳定的结构。主链化学位移与AlphaFold预测的结构非常吻合,验证RBD包含通过紧密转弯连接的三个螺旋。这种RBD构建体现在可以用于进一步的NMR研究,包括全长VP1u的赋值,蛋白质-蛋白质相互作用界面的测定,以及RBD域特异性B19抗病毒药物的开发。
    Parvovirus B19 (B19V) is a human pathogen that is the causative agent of several diseases in infants and adults. Due to a lack of antivirals against this virus, treatment options are limited. The minor capsid protein of B19V has a unique N terminus, named VP1u, which is essential for infection. The VP1u encodes a receptor binding domain (RBD), necessary for host cell entry, and a phospholipase A2 (PLA2) domain, crucial for endosomal escape during cellular trafficking. Both domains are indispensable for infection, making the RBD a plausible drug target for inhibitors against B19V, as it is located on the exterior surface of the virus. To date, no experimental structural information has been available for the VP1u component for any Parvovirus. Here we report the backbone NMR resonance assignments for the RBD of B19V and demonstrate it forms a stable structure. The backbone chemical shifts are in good agreement with a structure predicted by AlphaFold, validating that the RBD contains three helices connected by tight turns. This RBD construct can now be used for further NMR studies, including assignment of full-length VP1u, determination of protein-protein interaction interfaces, and development of B19 antivirals specific to the RBD domain.
    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
    细小病毒B19(B19V)是一种人类病原体,是婴儿和成人几种疾病的病原体。由于缺乏针对这种病毒的抗病毒药物,治疗方案有限。B19V的次要衣壳蛋白具有独特的N末端,命名为VP1u,这对感染至关重要。VP1u编码受体结合域(RBD),宿主细胞进入所必需的,和磷脂酶A2(PLA2)结构域,对于细胞运输过程中的内体逃逸至关重要。这两个领域都是感染不可或缺的,使RBD成为B19V抑制剂的合理药物靶标,因为它位于病毒的外表面。迄今为止,没有任何细小病毒的VP1u组件的实验性结构信息。在这里,我们报告了B19V的RBD的主链NMR共振分配,并证明了它形成了稳定的结构。主链化学位移与AlphaFold预测的结构非常吻合,验证RBD包含通过紧密转弯连接的三个螺旋。这种RBD构建体现在可以用于进一步的NMR研究,包括全长VP1u的赋值,蛋白质-蛋白质相互作用界面的测定,以及RBD域特异性B19抗病毒药物的开发。数据库:BMRB提交代码:52440。
    Parvovirus B19 (B19V) is a human pathogen that is the causative agent of several diseases in infants and adults. Due to a lack of antivirals against this virus, treatment options are limited. The minor capsid protein of B19V has a unique N terminus, named VP1u, which is essential for infection. The VP1u encodes a receptor binding domain (RBD), necessary for host cell entry, and a phospholipase A2 (PLA2) domain, crucial for endosomal escape during cellular trafficking. Both domains are indispensable for infection, making the RBD a plausible drug target for inhibitors against B19V, as it is located on the exterior surface of the virus. To date, no experimental structural information has been available for the VP1u component for any Parvovirus. Here we report the backbone NMR resonance assignments for the RBD of B19V and demonstrate it forms a stable structure. The backbone chemical shifts are in good agreement with a structure predicted by AlphaFold, validating that the RBD contains three helices connected by tight turns. This RBD construct can now be used for further NMR studies, including assignment of full-length VP1u, determination of protein-protein interaction interfaces, and development of B19 antivirals specific to the RBD domain. Database: BMRB submission code: 52440.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号