main protease

主要蛋白酶
  • 文章类型: Journal Article
    COVID-19目前被认为是第九大致命的流行病,通过与感染者直接或间接接触传播。它给许多国家的财政和医疗保健资源带来了持续的压力。为了应对这一挑战,迫切需要开发新的潜在治疗药物来治疗这种疾病。为了确定潜在的抗病毒剂作为SARS-CoV-2的新型双重抑制剂,我们从12种选定的药用抗病毒植物中检索了404生物碱,并对其进行了虚拟筛选,以对抗SARS-CoV-2的两种必需蛋白的著名催化位点和有利的相互作用残基,即,主要蛋白酶和刺突糖蛋白。根据对接分数,对12种具有双重抑制潜力的代谢物进行药物相似度,生物活性评分,和类似药物的能力分析。这些分析包括配体-受体稳定性和靶蛋白潜在活性位点的相互作用,通过对三种铅代谢物的分子动力学模拟进行分析和证实。我们还使用分子力学技术对关键的SARS-CoV-2蛋白抑制剂进行了详细的结合自由能分析,以揭示它们的相互作用动力学和稳定性。总的来说,我们的结果表明,12种生物碱,即,阿杜西汀Y,evodiamideC,麦角醇,Hayatinine,(+)-高芳啉,乙苯硫醚C,N,α-L-鼠李糖基长春胺,pelosine,利血平,toddalidimerine,Toddayanis,和zanthocadinanine,根据它们与靶蛋白的相互作用作为代谢物入围。与标准品相比,所有12种铅代谢物均表现出较高的未结合分数,因此分布更大。特别是,阿杜西汀Y显示出较高的对接分数,但表现出非自发结合谱。相比之下,麦角苷和evodiamideC在分子动力学模拟中显示出良好的结合相互作用和优越的稳定性。Ergosine在几个关键的制药指标中表现出卓越的性能。药代动力学评估显示,麦角苷表现出明显的生物活性,吸收好,和最佳生物利用度。此外,预计不会引起皮肤敏感,并且发现非肝毒性。重要的是,由于其强大的结合亲和力和类似药物的能力,麦角苷和evodiamideC成为SARS-CoV-2双重抑制的优异药物候选物,与已知的抑制剂如N3和莫那普拉韦相当。这项研究受到其计算机性质的限制,需要未来的体外和体内研究来证实这些发现。
    COVID-19 is currently considered the ninth-deadliest pandemic, spreading through direct or indirect contact with infected individuals. It has imposed a consistent strain on both the financial and healthcare resources of many countries. To address this challenge, there is a pressing need for the development of new potential therapeutic agents for the treatment of this disease. To identify potential antiviral agents as novel dual inhibitors of SARS-CoV-2, we retrieved 404 alkaloids from 12 selected medicinal antiviral plants and virtually screened them against the renowned catalytic sites and favorable interacting residues of two essential proteins of SARS-CoV-2, namely, the main protease and spike glycoprotein. Based on docking scores, 12 metabolites with dual inhibitory potential were subjected to drug-likeness, bioactivity scores, and drug-like ability analyses. These analyses included the ligand-receptor stability and interactions at the potential active sites of target proteins, which were analyzed and confirmed through molecular dynamic simulations of the three lead metabolites. We also conducted a detailed binding free energy analysis of pivotal SARS-CoV-2 protein inhibitors using molecular mechanics techniques to reveal their interaction dynamics and stability. Overall, our results demonstrated that 12 alkaloids, namely, adouetine Y, evodiamide C, ergosine, hayatinine, (+)-homoaromoline, isatithioetherin C, N,alpha-L-rhamnopyranosyl vincosamide, pelosine, reserpine, toddalidimerine, toddayanis, and zanthocadinanine, are shortlisted as metabolites based on their interactions with target proteins. All 12 lead metabolites exhibited a higher unbound fraction and therefore greater distribution compared with the standards. Particularly, adouetine Y demonstrated high docking scores but exhibited a nonspontaneous binding profile. In contrast, ergosine and evodiamide C showed favorable binding interactions and superior stability in molecular dynamics simulations. Ergosine demonstrated exceptional performance in several key pharmaceutical metrics. Pharmacokinetic evaluations revealed that ergosine exhibited pronounced bioactivity, good absorption, and optimal bioavailability. Additionally, it was predicted not to cause skin sensitivity and was found to be non-hepatotoxic. Importantly, ergosine and evodiamide C emerged as superior drug candidates for dual inhibition of SARS-CoV-2 due to their strong binding affinity and drug-like ability, comparable to known inhibitors like N3 and molnupiravir. This study is limited by its in silico nature and demands the need for future in vitro and in vivo studies to confirm these findings.
    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
    冠状病毒对人类人口构成重大威胁。严重急性呼吸道综合症冠状病毒-2,SARS-CoV-2,是一种高致病性人类冠状病毒,已引起COVID-19大流行。它导致了全球病毒爆发,传播异常,死亡人数众多,强调需要有效的抗病毒策略。3-胰凝乳蛋白酶样蛋白酶(3CLpro),SARS-CoV-2中的主要蛋白酶通过切割病毒多蛋白以产生病毒复制所必需的11个单独的非结构蛋白而在SARS-CoV-2病毒生命周期中起着不可或缺的作用。3CLpro是具有产生新病毒颗粒功能的两种蛋白酶之一。它是一种高度保守的半胱氨酸蛋白酶,在所有已知的人冠状病毒中具有相同的结构折叠。与3CLpro高亲和力结合的抑制剂将防止病毒多蛋白的裂解,从而阻碍病毒复制。已经实施了多种策略来筛选针对3CLpro的抑制剂,包括共价和非共价结合活性位点的肽样和小分子抑制剂,分别。此外,已经确定了3CLpro的变构位点来筛选可以制造3CLpro的非竞争性抑制剂的小分子。实质上,这篇综述是理解3CLpro的结构复杂性和功能动力学的全面指南,强调阐明其作为SARS-CoV-2主要蛋白酶的作用的关键发现。值得注意的是,该综述是认识到在确定和开发3CLpro抑制剂作为针对COVID-19的有效抗病毒策略方面取得进展的重要资源,其中一些已经被批准用于COVID-19患者的临床应用。
    Coronaviruses constitute a significant threat to the human population. Severe acute respiratory syndrome coronavirus-2, SARS-CoV-2, is a highly pathogenic human coronavirus that has caused the coronavirus disease 2019 (COVID-19) pandemic. It has led to a global viral outbreak with an exceptional spread and a high death toll, highlighting the need for effective antiviral strategies. 3-Chymotrypsin-like protease (3CLpro), the main protease in SARS-CoV-2, plays an indispensable role in the SARS-CoV-2 viral life cycle by cleaving the viral polyprotein to produce 11 individual non-structural proteins necessary for viral replication. 3CLpro is one of two proteases that function to produce new viral particles. It is a highly conserved cysteine protease with identical structural folds in all known human coronaviruses. Inhibitors binding with high affinity to 3CLpro will prevent the cleavage of viral polyproteins, thus impeding viral replication. Multiple strategies have been implemented to screen for inhibitors against 3CLpro, including peptide-like and small molecule inhibitors that covalently and non-covalently bind the active site, respectively. In addition, allosteric sites of 3CLpro have been identified to screen for small molecules that could make non-competitive inhibitors of 3CLpro. In essence, this review serves as a comprehensive guide to understanding the structural intricacies and functional dynamics of 3CLpro, emphasizing key findings that elucidate its role as the main protease of SARS-CoV-2. Notably, the review is a critical resource in recognizing the advancements in identifying and developing 3CLpro inhibitors as effective antiviral strategies against COVID-19, some of which are already approved for clinical use in COVID-19 patients.
    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
    冠状病毒病(COVID-19)是二十世纪最大的挑战之一。最近,计算机工具有助于预测SARS-CoV-2的新抑制剂。在这项研究中,设计了基于remdesivir结构的新化合物(12个化合物)。
    在3CLpro活性位点研究了remdesivir与设计化合物的主要相互作用。通过MM-GBSA方法计算了化合物的结合自由能,并将最佳化合物(值为-88.173kcal/mol的化合物12)引入分子动力学模拟研究。
    将模拟结果与不存在抑制剂和存在remdesivir的蛋白质模拟结果进行比较。此外,蛋白质骨架的RMSD结果表明,化合物12在第二个50纳秒内的波动小于单独的蛋白质和存在remdesivir的情况下,这表明化合物在Mpro蛋白活性位点的稳定性。此外,在不存在化合物和存在化合物12和remdesivir的情况下研究蛋白质紧密度。Rg图显示了大约0.05A的波动,这表明在存在和不存在化合物的情况下蛋白质的可压缩性。RMSF图的结果还显示了蛋白质结合过程中必需氨基酸的稳定性。
    在理论结果的支持下,化合物12可能具有抑制3CLpro酶的潜力,这需要进一步的体外研究,酶抑制也必须在蛋白质水平上得到证实。
    UNASSIGNED: Coronavirus disease (COVID-19) is one of the greatest challenges of the twentieth century. Recently, in silico tools help to predict new inhibitors of SARS-CoV-2. In this study, the new compounds based on the remdesivir structure (12 compounds) were designed.
    UNASSIGNED: The main interactions of remdesivir and designed compounds were investigated in the 3CLpro active site. The binding free energy of compounds by the MM-GBSA method was calculated and the best compound (compound 12 with the value of -88.173 kcal/mol) was introduced to the molecular dynamics simulation study.
    UNASSIGNED: The simulation results were compared with the results of protein simulation without the presence of an inhibitor and in the presence of remdesivir. Additionally, the RMSD results for the protein backbone showed that compound 12 in the second 50 nanoseconds has less fluctuation than the protein alone and in the presence of remdesivir, which indicates the stability of the compound in the active site of the Mpro protein. Furthermore, protein compactness was investigated in the absence of compounds and the presence of compound 12 and remdesivir. The Rg diagram shows a fluctuation of approximately 0.05 A, which indicates the compressibility of the protein in the presence and absence of compounds. The results of the RMSF plot also show the stability of essential amino acids during protein binding.
    UNASSIGNED: Supported by the theoretical results, compound 12 could have the potential to inhibit the 3CLpro enzyme, which requires further in vitro studies and enzyme inhibition must also be confirmed at protein levels.
    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
    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
    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
    开发了SARS-CoV-2主要蛋白酶(Mpro)的滴定剂,第一次,通过活性位点滴定法精确测定酶活性Mpro的浓度。通过确定酶-滴定剂复合物的晶体结构,阐明了四肽滴定剂的共价结合模式。Mpro的四种荧光底物,包括一个原型,内部猝灭的Dabcyl-EDANS肽,在典型测定条件下的溶解度方面进行了比较。通过利用新的滴定剂,确定了Mpro催化裂解这些底物的关键动力学参数。
    A titrant for the SARS-CoV-2 main protease (Mpro) was developed that enables, for the first time, the exact determination of the concentration of the enzymatically active Mpro by active-site titration. The covalent binding mode of the tetrapeptidic titrant was elucidated by the determination of the crystal structure of the enzyme-titrant complex. Four fluorogenic substrates of Mpro, including a prototypical, internally quenched Dabcyl-EDANS peptide, were compared in terms of solubility under typical assay conditions. By exploiting the new titrant, key kinetic parameters for the Mpro-catalyzed cleavage of these substrates were determined.
    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
    自SARS-CoV-2首次报告以来已经过去了四年多,人类经历了一场具有前所未有影响的大流行。此外,新的变种使情况变得更糟。在病毒酶中,SARS-CoV-2主要蛋白酶(Mpro)被认为是有希望的药物靶标。COVID-19。的确,Mpro是病毒复制的关键酶,它在冠状病毒中高度保守。它显示了对酶活性至关重要的蛋白酶残基的高度保守性,强调其作为药物靶标的潜力,以开发不仅有效的广谱抗病毒药物SARS-CoV-2变体,但也针对其他冠状病毒。即使FDA批准的药物Nirmatrelvir,Mpro抑制剂,该药物已经促进了治疗COVID-19的抗病毒治疗,但该药物显示出一些缺点,阻碍了其临床应用。在这里,我们报道了对SARS-CoV-2Mpro具有微摩尔抑制效力的新型噻唑烷-4-酮衍生物的合成。计算机模拟研究揭示了与高度保守的酶残基结合的关键结构要求,表明噻唑啉酮核充当天然底物的Gln氨基酸的模拟物,并且硝基取代的芳族部分在与催化性His-41残基建立π-π堆叠相互作用中的核心作用。
    It has been more than four years since the first report of SARS-CoV-2, and humankind has experienced a pandemic with an unprecedented impact. Moreover, the new variants have made the situation even worse. Among viral enzymes, the SARS-CoV-2 main protease (Mpro) has been deemed a promising drug target vs. COVID-19. Indeed, Mpro is a pivotal enzyme for viral replication, and it is highly conserved within coronaviruses. It showed a high extent of conservation of the protease residues essential to the enzymatic activity, emphasizing its potential as a drug target to develop wide-spectrum antiviral agents effective not only vs. SARS-CoV-2 variants but also against other coronaviruses. Even though the FDA-approved drug nirmatrelvir, a Mpro inhibitor, has boosted the antiviral therapy for the treatment of COVID-19, the drug shows several drawbacks that hinder its clinical application. Herein, we report the synthesis of new thiazolidine-4-one derivatives endowed with inhibitory potencies in the micromolar range against SARS-CoV-2 Mpro. In silico studies shed light on the key structural requirements responsible for binding to highly conserved enzymatic residues, showing that the thiazolidinone core acts as a mimetic of the Gln amino acid of the natural substrate and the central role of the nitro-substituted aromatic portion in establishing π-π stacking interactions with the catalytic His-41 residue.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Comparative Study
    主要蛋白酶(Mpro)仍然是感染干预后COVID-19的重要治疗靶标,因为它在处理SARS-CoV-2基因组编码的大多数病毒蛋白中发挥了关键作用。病毒进入后,+ssRNA基因组被翻译成两个长的多蛋白(pp1a或移码依赖性pp1ab),包含病毒免疫调节所需的所有非结构蛋白(nsps),复制,最终,病毒体组装.这些nsp中包括半胱氨酸蛋白酶Mpro(nsp5),它从多蛋白中自我切除,二聚化,然后连续切割在多蛋白内每个nsp之间发现的15个切割位点连接中的11个。最近已经详细描述了Mpro的许多结构(通常与各种抑制剂或肽抑制剂结合),包括与每个多蛋白切割序列结合的Mpro结构,表明Mpro可以在其活性位点内适应广泛的靶标。然而,到目前为止,Mpro与其每个天然切割位点序列相互作用的动力学表征仍然不完整。在这里,我们提出了一种稳健且具有成本效益的基于FRET的系统,该系统受益于更一致的底物呈递,与以前报道的使用化学修饰肽的FRET系统相比,该底物在组织中也更接近天然多蛋白环境。使用这个系统,我们能够证明,虽然每个网站都保持着类似的迈克尔斯常数,Mpro的催化效率在切割位点序列之间变化很大,建议对nsp处理顺序有明确的偏好。
    The main protease (Mpro) remains an essential therapeutic target for COVID-19 post infection intervention given its critical role in processing the majority of viral proteins encoded by the genome of severe acute respiratory syndrome related coronavirus 2 (SARS-CoV-2). Upon viral entry, the +ssRNA genome is translated into two long polyproteins (pp1a or the frameshift-dependent pp1ab) containing all the nonstructural proteins (nsps) required by the virus for immune modulation, replication, and ultimately, virion assembly. Included among these nsps is the cysteine protease Mpro (nsp5) which self-excises from the polyprotein, dimerizes, then sequentially cleaves 11 of the 15 cut-site junctions found between each nsp within the polyprotein. Many structures of Mpro (often bound to various small molecule inhibitors or peptides) have been detailed recently, including structures of Mpro bound to each of the polyprotein cleavage sequences, showing that Mpro can accommodate a wide range of targets within its active site. However, to date, kinetic characterization of the interaction of Mpro with each of its native cleavage sequences remains incomplete. Here, we present a robust and cost-effective FRET based system that benefits from a more consistent presentation of the substrate that is also closer in organization to the native polyprotein environment compared to previously reported FRET systems that use chemically modified peptides. Using this system, we were able to show that while each site maintains a similar Michaelis constant, the catalytic efficiency of Mpro varies greatly between cut-site sequences, suggesting a clear preference for the order of nsp processing.
    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
    导语:全球病毒大流行迅速蔓延,导致许多人经历感染。冠状病毒(CoV)是感染不同类型哺乳动物的许多病毒家族之一。它们可以传播给人类并引起胃肠道,神经学,和呼吸问题。本研究发现类黄酮化合物是有前途的分子剂,具有潜在的抗病毒活性抗病毒蛋白,特别是主要蛋白酶(Mpro)。方法:本研究对来自药用植物的天然化合物进行了全面的计算机筛选。它包括参数评估,如药物相似度,药代动力学,分子对接,毒性评价,生物利用度评估,和分子靶标探索。在这种系统的方法中,主要目的是鉴定潜在的先导化合物.将这些植物化学物质作为候选药物进行研究,以详细了解其分子特性。结果:异槲皮素的Mpro结合能值为-10.637,-12.752,-7.813,-15.732,-6.449,-5.578,-8.037和-8.52kcal/mol,那利鲁丁,杨梅素,橙皮苷,水飞蓟宾,黄芩素,taxifolin,还有petunidin.对两种类黄酮化合物-橙皮苷和narirutin-在冠状病毒蛋白中稳定超过100纳秒进行了分子模拟。结论:我们进行的计算研究是有希望的,但是为了验证这些化合物的作用,需要进一步的实验研究,研究的关键组成部分是进行体外和体内实验。
    Introduction: The global viral pandemic has rapidly spread, leading to many individuals experiencing the infection. Coronaviruses (CoVs) are among many viral families that infect different types of mammals. They can spread to humans and cause gastrointestinal, neurological, and respiratory problems. The present investigation has discovered flavonoid compounds as promising molecular agents with potential antiviral activity against virus proteins, specifically main protease (Mpro). Methodology: A comprehensive in silico screening of natural compounds derived from medicinal plants was performed in the present study. It included parameter assessments such as drug-likeness, pharmacokinetics, molecular docking, toxicity evaluations, bioavailability assessments, and molecular target exploration. In this systematic approach, the primary objective was to identify potential lead compounds. These phytochemicals were investigated as drug candidates to provide a detailed understanding of their molecular properties. Results: The Mpro binding energy values were -10.637, -12.752, -7.813, -15.732, -6.449, -5.578, -8.037, and -8.52 kcal/mol for isoquercetin, narirutin, myricetin, hesperidin, silibinin, baicalein, taxifolin, and petunidin. Molecular simulations were conducted on two flavonoid compounds - hesperidin and narirutin - stable over 100 nanoseconds in the Coronavirus protein. Conclusions: The computational study we conducted is promising, but to validate the action of these compounds, further experimental studies are needed, with a critical component of the research being the conduct of in vitro and in vivo experiments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号