main protease

主要蛋白酶
  • 文章类型: Journal Article
    主要蛋白酶(Mpros)是冠状病毒中一类保守的半胱氨酸水解酶,在病毒复制中起着至关重要的作用。因此,MPRs是开发泛冠状病毒药物的理想目标。X77,以前针对SARS-CoVMpro开发的,在COVID-19大流行期间被重新用作抗SARS-CoV-2Mpro的非共价紧密结合剂抑制剂。使用X77作为参考,已经发现了许多具有良好功效的新型抑制剂。这表明X77的结构可能是药物设计的有价值的支架。然而,X77的广谱性能和潜在机制仍然知之甚少。这里,我们报道了SARS-CoV-2,SARS-CoV,还有MERS-CoV,和几个来自SARS-CoV-2变体的Mpro突变体与X77结合。对这些结构的详细分析揭示了相互作用所必需的关键结构决定因素,并阐明了X77与不同冠状病毒Mpro的结合模式。通过分子动力学模拟和结合自由能计算进一步评估X77对这些研究的Mpros的效力。这些数据提供了对X77对冠状病毒Mpros的广谱抑制的分子见解,以及X77与各种Mpros结合时的异同。这将促进基于X77的新型抗病毒药物的设计,具有针对不同冠状病毒和SARS-CoV-2变体的广谱功效。
    Main proteases (Mpros) are a class of conserved cysteine hydrolases among coronaviruses and play a crucial role in viral replication. Therefore, Mpros are ideal targets for the development of pan-coronavirus drugs. X77, previously developed against SARS-CoV Mpro, was repurposed as a non-covalent tight binder inhibitor against SARS-CoV-2 Mpro during COVID-19 pandemic. Many novel inhibitors with favorable efficacy have been discovered using X77 as a reference, suggesting that the structure of X77 could be a valuable scaffold for drug design. However, the broad-spectrum performance of X77 and underlying mechanism remain less understood. Here, we reported the crystal structures of Mpros from SARS-CoV-2, SARS-CoV, and MERS-CoV, and several Mpro mutants from SARS-CoV-2 variants bound to X77. A detailed analysis of these structures reveals key structural determinants essential for interaction and elucidates the binding modes of X77 to different coronaviral Mpros. The potencies of X77 against these investigated Mpros were further evaluated through molecular dynamic simulation and binding free energy calculation. These data provide molecular insights into broad-spectrum inhibition against coronaviral Mpros by X77 and the similarities and differences of X77 when bound to various Mpros, which will promote X77-based design of novel antivirals with broad-spectrum efficacy against different coronaviruses and SARS-CoV-2 variants.
    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
    人冠状病毒229E(HCoV-229E)与上呼吸道感染有关,通常会引起轻度呼吸道症状。HCoV-229E感染可导致细胞死亡,但是导致病毒诱导的细胞死亡的分子途径以及病毒蛋白和细胞细胞死亡效应物之间的相互作用对于HCoV-229E仍然缺乏表征。研究HCoV-229E和其他常见的冷冠状病毒如何与细胞死亡途径相互作用并影响细胞死亡途径可能有助于了解其发病机理并将其与高致病性冠状病毒进行比较。这里,我们报告说,HCoV-229E的主要蛋白酶(Mpro)可以在其活性N端结构域内的两个不同位点(Q29和Q193)切割gasderminD(GSDMD),以产生现在无法引起焦亡的片段,通常由这种蛋白质执行的裂解细胞死亡的一种形式。尽管HCoV-229EMpro切割GSDMD,我们显示HCoV-229E感染仍然导致裂解细胞死亡。我们证明,在病毒感染期间,caspase-3切割并激活gasderminE(GSDME),化脓的另一个关键执行者。因此,GSDME敲除细胞在病毒感染时显示裂解细胞死亡的显著减少。最后,我们显示HCoV-229E感染导致表达Mpro不可裂解的GSDMD突变体(GSDMDQ29A+Q193A)的细胞中裂解细胞死亡水平增加.我们得出结论,GSDMD在HCoV-229E感染期间被Mpro灭活,防止GSDMD介导的细胞死亡,并指出caspase-3/GSDME轴在病毒诱导的细胞死亡的执行中具有重要作用。在高致病性冠状病毒的类似报道发现的背景下,我们的结果提示,这些机制并不导致冠状病毒间致病性的差异.尽管如此,了解普通感冒相关冠状病毒及其蛋白质与程序性细胞死亡机制的相互作用,可能为冠状病毒控制策略提供新的线索.
    Human coronavirus 229E (HCoV-229E) is associated with upper respiratory tract infections and generally causes mild respiratory symptoms. HCoV-229E infection can cause cell death, but the molecular pathways that lead to virus-induced cell death as well as the interplay between viral proteins and cellular cell death effectors remain poorly characterized for HCoV-229E. Studying how HCoV-229E and other common cold coronaviruses interact with and affect cell death pathways may help to understand its pathogenesis and compare it to that of highly pathogenic coronaviruses. Here, we report that the main protease (Mpro) of HCoV-229E can cleave gasdermin D (GSDMD) at two different sites (Q29 and Q193) within its active N-terminal domain to generate fragments that are now unable to cause pyroptosis, a form of lytic cell death normally executed by this protein. Despite GSDMD cleavage by HCoV-229E Mpro, we show that HCoV-229E infection still leads to lytic cell death. We demonstrate that during virus infection caspase-3 cleaves and activates gasdermin E (GSDME), another key executioner of pyroptosis. Accordingly, GSDME knockout cells show a significant decrease in lytic cell death upon virus infection. Finally, we show that HCoV-229E infection leads to increased lytic cell death levels in cells expressing a GSDMD mutant uncleavable by Mpro (GSDMD Q29A+Q193A). We conclude that GSDMD is inactivated by Mpro during HCoV-229E infection, preventing GSDMD-mediated cell death, and point to the caspase-3/GSDME axis as an important player in the execution of virus-induced cell death. In the context of similar reported findings for highly pathogenic coronaviruses, our results suggest that these mechanisms do not contribute to differences in pathogenicity among coronaviruses. Nonetheless, understanding the interactions of common cold-associated coronaviruses and their proteins with the programmed cell death machineries may lead to new clues for coronavirus control strategies.
    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
    2019年冠状病毒病(COVID-19)是由一种称为严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的新型冠状病毒引起的,迅速传播到世界各地。主要蛋白酶(Mpro)对病毒的复制和转录具有重要意义,使其成为对抗冠状病毒的有吸引力的药物靶标。这里,我们介绍了一系列通过基于结构的药物设计方法从头设计的新型抑制剂,这些抑制剂具有巨大的体外抑制SARS-CoV-2Mproin的潜力。高分辨率结构表明,这些抑制剂通过新型二溴甲基酮(DBMK)作为反应性弹头与催化半胱氨酸形成共价键。同时,DBMK弹头旁边的设计苯基通过π-π堆叠相互作用插入H41和C145之间的裂缝中,分解催化二重体并破坏质子转移。这种独特的结合模型为SARS-CoV-2以及其他病原体的半胱氨酸蛋白酶抑制剂开发提供了新的线索。
    The coronavirus disease 2019 (COVID-19) is caused by a novel coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which spreads rapidly all over the world. The main protease (Mpro) is significant to the replication and transcription of viruses, making it an attractive drug target against coronaviruses. Here, we introduce a series of novel inhibitors which are designed de novo through structure-based drug design approach that have great potential to inhibit SARS-CoV-2 Mproin vitro. High-resolution structures show that these inhibitors form covalent bonds with the catalytic cysteine through the novel dibromomethyl ketone (DBMK) as a reactive warhead. At the same time, the designed phenyl group beside the DBMK warhead inserts into the cleft between H41 and C145 through π-π stacking interaction, splitting the catalytic dyad and disrupting proton transfer. This unique binding model provides novel clues for the cysteine protease inhibitor development of SARS-CoV-2 as well as other pathogens.
    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
    SARS-CoV-2的爆发,也称为COVID-19大流行,仍然是人类生活和全球经济的关键风险因素。虽然,文献中已经介绍了几种有希望的疗法来抑制SARS-CoV-2,其中大多数是合成药物,可能对人体产生一些不利影响。因此,这项研究的主要目的是对麻黄病的药用特性进行计算机研究(P.alliaceaL.)介导的植物化合物用于治疗SARS-CoV-2感染,因为与合成药物相比,植物化学物质的不良反应较少。目的:探索百里香潜在的植物化合物作为候选药物分子,我们选择SARS-CoV-2的引起感染的主要蛋白酶(Mpro)作为受体蛋白。使用AutoDockVina进行这些受体蛋白与不同植物化合物的分子对接分析。然后,我们选择了三种排名最高的植物化合物(杨梅素,Engeletin,和astilbin)作为候选药物分子,其最高结合亲和力评分为-8.9,-8.7和-8.3(Kcal/mol),分别。然后,使用YASARA软件对它们与Mpro的复合物进行了100ns分子动力学(MD)模拟研究,计算的RMSD,RMSF,PCA,DCCM,MM/PBSA,和自由能源景观(FEL),并发现它们几乎稳定的结合性能。此外,生物活性,ADME/T,DFT,和药物相似度分析显示了所选植物化合物的合适药代动力学特性。因此,在湿实验室和临床试验中进行实验验证后,这项研究的结果可能是制定SARS-CoV-2感染安全治疗计划的有用资源.
    The outbreak of SARS-CoV-2, also known as the COVID-19 pandemic, is still a critical risk factor for both human life and the global economy. Although, several promising therapies have been introduced in the literature to inhibit SARS-CoV-2, most of them are synthetic drugs that may have some adverse effects on the human body. Therefore, the main objective of this study was to carry out an in-silico investigation into the medicinal properties of Petiveria alliacea L. (P. alliacea L.)-mediated phytocompounds for the treatment of SARS-CoV-2 infections since phytochemicals have fewer adverse effects compared to synthetic drugs. To explore potential phytocompounds from P. alliacea L. as candidate drug molecules, we selected the infection-causing main protease (Mpro) of SARS-CoV-2 as the receptor protein. The molecular docking analysis of these receptor proteins with the different phytocompounds of P. alliacea L. was performed using AutoDock Vina. Then, we selected the three top-ranked phytocompounds (myricitrin, engeletin, and astilbin) as the candidate drug molecules based on their highest binding affinity scores of -8.9, -8.7 and -8.3 (Kcal/mol), respectively. Then, a 100 ns molecular dynamics (MD) simulation study was performed for their complexes with Mpro using YASARA software, computed RMSD, RMSF, PCA, DCCM, MM/PBSA, and free energy landscape (FEL), and found their almost stable binding performance. In addition, biological activity, ADME/T, DFT, and drug-likeness analyses exhibited the suitable pharmacokinetics properties of the selected phytocompounds. Therefore, the results of this study might be a useful resource for formulating a safe treatment plan for SARS-CoV-2 infections after experimental validation in wet-lab and clinical trials.
    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主要蛋白酶(Mpro)在病毒扩增中起着至关重要的作用,是抗病毒药物的理想靶标。目前,通过两轮蛋白水解切割制备真正的Mpro。在这种方法中,Mpro在N-末端携带自切割位点和蛋白酶切割位点,随后在C-末端携带亲和标签。本文提出了一种通过单一消化生产正宗Mpro的新方法。通过在N-末端融合含有TEV蛋白酶切割位点的His标签来构建Mpro。表达的重组蛋白用TEV蛋白酶消化,生成的蛋白质分子量降低,活性显著增加,这与之前方法生成的真实Mpro一致。这些发现表明成功获得了真正的Mpro。此外,研究了Mpro的底物特异性。Mpro在P2位置对Phe具有强烈的偏好,这表明S2亚位点是设计抑制剂的突出靶标。本文也为今后制备用于突发冠状病毒感染的Mpro提供了参考。
    The SARS-CoV-2 main protease (Mpro) plays a crucial role in virus amplification and is an ideal target for antiviral drugs. Currently, authentic Mpro is prepared through two rounds of proteolytic cleavage. In this method, Mpro carries a self-cleavage site at the N-terminus and a protease cleavage site followed by an affinity tag at the C-terminus. This article proposes a novel method for producing authentic Mpro through single digestion. Mpro was constructed by fusing a His tag containing TEV protease cleavage sites at the N-terminus. The expressed recombinant protein was digested by TEV protease, and the generated protein had a decreased molecular weight and significantly increased activity, which was consistent with that of authentic Mpro generated by the previous method. These findings indicated that authentic Mpro was successfully obtained. Moreover, the substrate specificity of Mpro was investigated. Mpro had a strong preference for Phe at position the P2, which suggested that the S2 subsite was an outstanding target for designing inhibitors. This article also provides a reference for the preparation of Mpro for sudden coronavirus infection 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
    人冠状病毒是一组主要引起呼吸道和肠道疾病的病原体。感染容易引起呼吸道症状,以及各种严重的并发症。有几种人类冠状病毒,比如SARS-CoV,MERS-CoV,HCoV-229E,HCoV-OC43、HCoV-NL63、HCoV-HKU1和SARS-CoV-2。COVID-19的流行导致人们越来越关注针对人类冠状病毒的药物研究。来自人冠状病毒的主要蛋白酶(Mpro)是相对保守的,其控制病毒复制。通过使用计算机模拟对接,发现X77对SARS-CoV-2Mpro具有极高的抑制活性。在本文中,我们已经解析了与X77络合的HCoV-NL63Mpro的晶体结构,并详细分析了它们的相互作用。该数据为解决其结合模式及其结构决定因素提供了必要的信息。然后,我们详细比较了X77与SARS-CoV-2Mpro和HCoV-NL63Mpro的结合模式。该研究说明了HCoV-NL63Mpro与抑制剂X77结合的结构基础。从这项研究中得出的结构见解将为开发对人类冠状病毒具有广谱抗性的新药提供信息。
    Human coronaviruses are a group of pathogens that primarily cause respiratory and intestinal diseases. Infection can easily cause respiratory symptoms, as well as a variety of serious complications. There are several types of human coronaviruses, such as SARS-CoV, MERS-CoV, HCoV-229E, HCoV-OC43, HCoV-NL63, HCoV-HKU1, and SARS-CoV-2. The prevalence of COVID-19 has led to a growing focus on drug research against human coronaviruses. The main protease (Mpro) from human coronaviruses is a relatively conserved that controls viral replication. X77 was discovered to have extremely high inhibitory activity against SARS-CoV-2 Mpro through the use of computer-simulated docking. In this paper, we have resolved the crystal structure of the HCoV-NL63 Mpro complexed with X77 and analyzed their interaction in detail. This data provides essential information for solving their binding modes and their structural determinants. Then, we compared the binding modes of X77 with SARS-CoV-2 Mpro and HCoV-NL63 Mpro in detail. This study illustrates the structural basis of HCoV-NL63 Mpro binding to the inhibitor X77. The structural insights derived from this study will inform the development of new drugs with broad-spectrum resistance to human coronaviruses.
    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
    在最初症状发作后24天,在COVID-19检测呈阳性的患者的牙周组织中发现SARS-CoV-2RNA,表明口腔可以作为病毒储库。这项研究旨在调查Ovatdiolide的抗病毒能力,介绍了一种用于SARS-CoV-2研究的新型牙周膜类器官模型。我们已经成功地建立了一个可靠的和可扩展的类器官培养人牙周膜,展示上皮干细胞的典型特征。这种类器官模型使我们能够深入研究牙齿上皮干细胞生物学的鲜为人知的方面及其与病毒和口腔组织的相互作用。我们进行了一系列体外和离体研究,以检查Ova对SARS-CoV-2的抑制作用。我们的发现表明Ovatdiolide分子可以有效地结合NRP1活性域。我们的研究确定了NRP1受体b1域内Ovatdiolide(OVA)的潜在相互作用位点。我们在这个位点产生了点突变,产生三个变体:Y25A,T44A,和双突变Y25A/T44A。虽然这些突变不会改变刺突蛋白的结合活性,它们确实影响了抑制所需的OVA浓度.对于Y25A,这些变体的抑制浓度为15μM,T44A为15.2μM,和25μM的双突变体Y25A/T44A。此外,体外抑制实验表明,Ova对SARS-CoV-2病毒主要蛋白酶(Mpro)的EC50为7.316μM。我们的体外研究和牙周膜类器官模型的使用强调了Ovatdiolide作为小分子治疗剂的潜力,阻碍了病毒与宿主细胞上Neuropilin-1受体结合的能力。该研究揭示了Ovatdiolide可以作为有效的抗病毒小分子药物的各种途径和生化策略。
    The discovery of SARS-CoV-2 RNA in the periodontal tissues of patients who tested positive for COVID-19, 24 days post the initial symptom onset, indicates the oral cavity could serve as a viral reservoir. This research aims to investigate the antiviral capabilities of Ovatodiolide, introducing a novel periodontal ligament organoid model for the study of SARS-CoV-2. We have successfully established a reliable and expandable organoid culture from the human periodontal ligament, showcasing characteristics typical of epithelial stem cells. This organoid model enables us to delve into the lesser-known aspects of dental epithelial stem cell biology and their interactions with viruses and oral tissues. We conducted a series of in vitro and ex vivo studies to examine the inhibitory impacts of Ova on SARS-CoV-2. Our findings indicate that Ovatodiolide molecules can bind effectively to the NRP1 active domain. Our study identifies potential interaction sites for Ovatodiolide (OVA) within the b1 domain of the NRP1 receptor. We generated point mutations at this site, resulting in three variants: Y25A, T44A, and a double mutation Y25A/T44A. While these mutations did not alter the binding activity of the spike protein, they did impact the concentration of OVA required for inhibition. The inhibitory concentrations for these variants are 15 μM for Y25A, 15.2 μM for T44A, and 25 μM for the double mutant Y25A/T44A. In addition, in vitro inhibition experiments demonstrate that the EC50 of Ova against the main protease (Mpro) of the SARS-CoV-2 virus is 7.316 μM. Our in vitro studies and the use of the periodontal ligament organoid model highlight Ovatodiolide\'s potential as a small molecule therapeutic agent that impedes the virus\'s ability to bind to the Neuropilin-1 receptor on host cells. The research uncovers various pathways and biochemical strategies through which Ovatodiolide may function as an effective antiviral small molecule drug.
    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
    非结构蛋白5(Nsp5)是SARS-CoV-2的主要蛋白酶,其将病毒多蛋白切割成病毒复制所必需的单个多肽。这里,我们显示Nsp5结合并切割人tRNA甲基转移酶1(TRMT1),tRNA中普遍的转录后修饰所需的宿主酶。感染SARS-CoV-2的人细胞表现出TRMT1蛋白水平降低和TRMT1催化的tRNA修饰,与TRMT1裂解和Nsp5失活一致。Nsp5在与SARS-CoV-2多蛋白切割位点的共有序列相匹配的特定位置切割TRMT1,序列内的单个突变抑制TRMT1的Nsp5依赖性蛋白水解。TRMT1裂解片段表现出改变的RNA结合活性,并且不能挽救TRMT1缺陷型人细胞中的tRNA修饰。与野生型人类细胞相比,用SARS-CoV-2感染的TRMT1缺陷的人细胞表现出降低的细胞内病毒RNA水平。这些发现提供了证据,证明Nsp5依赖性TRMT1的裂解和tRNA修饰模式的扰动有助于SARS-CoV-2感染的细胞发病机理。
    导致COVID-19感染的病毒被称为SARS-CoV-2。像所有的病毒一样,SARS-CoV-2带有制造蛋白质和其他分子的指令,这些分子在使病毒繁殖和传播中起着至关重要的作用。病毒无法自己制造这些分子,所以它们感染细胞,诱使它们制造分子并代表它们组装新的病毒颗粒。当SARS-CoV2感染细胞时,宿主细胞被重新编程,以制造含有几种病毒蛋白的链,这些蛋白需要通过病毒酶彼此切断,称为Nsp5,以使蛋白质正常工作。先前的研究表明,Nsp5也可能与称为TRMT1的人类蛋白质相互作用,这有助于在细胞中产生新的蛋白质。然而,尚不清楚Nsp5如何与TRMT1结合或这种相互作用如何影响宿主细胞。张等人。在人体细胞中使用生化和分子技术来研究Nsp5如何与TRMT1相互作用。实验发现,病毒酶将TRMT1切割成无活性的片段,随后被细胞破坏。此外,Nsp5在对应于病毒蛋白切割位点的完全相同的位置切割TRMT1。TRMT1中序列的突变使得Nsp5在切割蛋白质时无效。SARS-CoV-2感染导致细胞内TRMT1水平降低,反过来,导致TRMT1活性下降。与正常人细胞相比,该病毒在无法产生TRMT1的细胞中繁殖较少,表明该病毒在感染早期受益于TRMT1,在稍后将其停用之前。这些发现表明,SARS-CoV-2导致疾病的一种方式是通过降低调节蛋白质产生的人类蛋白质的水平。在未来,张等人的工作。可以为检测SARS-CoV-2和其他类似病毒的感染提供新的标记,并指导对它们进行更有效的治疗。
    Nonstructural protein 5 (Nsp5) is the main protease of SARS-CoV-2 that cleaves viral polyproteins into individual polypeptides necessary for viral replication. Here, we show that Nsp5 binds and cleaves human tRNA methyltransferase 1 (TRMT1), a host enzyme required for a prevalent post-transcriptional modification in tRNAs. Human cells infected with SARS-CoV-2 exhibit a decrease in TRMT1 protein levels and TRMT1-catalyzed tRNA modifications, consistent with TRMT1 cleavage and inactivation by Nsp5. Nsp5 cleaves TRMT1 at a specific position that matches the consensus sequence of SARS-CoV-2 polyprotein cleavage sites, and a single mutation within the sequence inhibits Nsp5-dependent proteolysis of TRMT1. The TRMT1 cleavage fragments exhibit altered RNA binding activity and are unable to rescue tRNA modification in TRMT1-deficient human cells. Compared to wild-type human cells, TRMT1-deficient human cells infected with SARS-CoV-2 exhibit reduced levels of intracellular viral RNA. These findings provide evidence that Nsp5-dependent cleavage of TRMT1 and perturbation of tRNA modification patterns contribute to the cellular pathogenesis of SARS-CoV-2 infection.
    The virus responsible for COVID-19 infections is known as SARS-CoV-2. Like all viruses, SARS-CoV-2 carries instructions to make proteins and other molecules that play essential roles in enabling the virus to multiply and spread. Viruses are unable to make these molecules themselves, so they infect cells and trick them into making the molecules and assembling new virus particles on their behalf instead. When SARS-CoV2 infects cells, the host cells are reprogrammed to make chains containing several virus proteins that need to be severed from each other by a virus enzyme, known as Nsp5, to enable the proteins to work properly. Previous studies suggested that Nsp5 may also interact with a human protein known as TRMT1, which helps with the production of new proteins in cells. However, it was not clear how Nsp5 may bind to TRMT1 or how this interaction may affect the host cell. Zhang et al. used biochemical and molecular techniques in human cells to study how Nsp5 interacts with TRMT1. The experiments found that the virus enzyme cuts TRMT1 into fragments that are inactive and are subsequently destroyed by the cells. Moreover, Nsp5 cuts TRMT1 at exactly the same position corresponding to the cleavage sites of the viral proteins. Mutation of the sequence in TRMT1 renders Nsp5 ineffective at cutting the protein. SARS-CoV-2 infection caused TRMT1 levels to decrease inside the cells, in turn, leading to a drop in TRMT1 activity. The virus multiplied less in cells that were unable to produce TRMT1 compared to normal human cells, suggesting that the virus benefits from TRMT1 early during infection, before inactivating it at a later point. These findings suggest that one way SARS-CoV-2 causes disease is by decreasing the levels of a human protein that regulates protein production. In the future, the work of Zhang et al. may provide new markers for detecting infections of SARS-CoV-2 and other similar viruses and guide efforts to make more effective therapies against them.
    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
    新型抗病毒药物的开发不仅对于管理当前的COVID-19感染,而且对于解决未来潜在的人畜共患疫情至关重要。SARS-CoV-2主要蛋白酶(Mpro)对于病毒复制和生存力至关重要,因此可作为抗病毒干预的有吸引力的靶标。在这里,我们报道了针对SARS-CoV-2Mpro的mRNA展示选择产生的环肽抑制剂的优化,以增强其细胞通透性和体外抗病毒活性。通过鉴定肽序列中的突变耐受氨基酸残基,我们描述了带有五个环己基丙氨酸残基的第二代Mpro抑制剂的开发。与亲本肽相比,该环肽类似物表现出显著改善的细胞渗透性和抗病毒活性。这种方法突出了优化环状肽命中针对针对细胞内靶标(例如SARS-CoV-2Mpro)的活性的重要性。
    The development of novel antivirals is crucial not only for managing current COVID-19 infections but for addressing potential future zoonotic outbreaks. SARS-CoV-2 main protease (Mpro) is vital for viral replication and viability and therefore serves as an attractive target for antiviral intervention. Herein, we report the optimization of a cyclic peptide inhibitor that emerged from an mRNA display selection against the SARS-CoV-2 Mpro to enhance its cell permeability and in vitro antiviral activity. By identifying mutation-tolerant amino acid residues within the peptide sequence, we describe the development of a second-generation Mpro inhibitor bearing five cyclohexylalanine residues. This cyclic peptide analogue exhibited significantly improved cell permeability and antiviral activity compared to the parent peptide. This approach highlights the importance of optimizing cyclic peptide hits for activity against intracellular targets such as the SARS-CoV-2 Mpro.
    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