Mpro

Mpro
  • 文章类型: Journal Article
    蛋白质是动态大分子。了解蛋白质的热可接近构象对于确定重要的转变和设计疗法至关重要。可接近的构象受到蛋白质结构的高度限制,因此由于外部扰动而引起的协同结构变化可能会跟踪固有的构象转变。这些过渡可以被认为是通过构象景观的路径。晶体学药物片段筛选是高通量扰动实验,其中将数千个药物靶标的晶体浸入小分子药物前体(片段)并检查片段结合,在靶蛋白上绘制潜在的药物结合位点。这里,我们描述了一个开源的Python包,COLAV(形式化LAndscape可视化),从这种大规模的晶体学扰动研究中推断构象景观。我们将COLAV应用于两个医学上重要系统的药物片段筛选:蛋白酪氨酸磷酸酶1B(PTP-1B),调节胰岛素信号,和SARSCoV-2主蛋白酶(MPro)。有足够的片段结合结构,我们发现,这样的药物筛选也能够详细绘制蛋白质的构象景观。
    Proteins are dynamic macromolecules. Knowledge of a protein\'s thermally accessible conformations is critical to determining important transitions and designing therapeutics. Accessible conformations are highly constrained by a protein\'s structure such that concerted structural changes due to external perturbations likely track intrinsic conformational transitions. These transitions can be thought of as paths through a conformational landscape. Crystallographic drug fragment screens are high-throughput perturbation experiments, in which thousands of crystals of a drug target are soaked with small-molecule drug precursors (fragments) and examined for fragment binding, mapping potential drug binding sites on the target protein. Here, we describe an open-source Python package, COLAV (COnformational LAndscape Visualization), to infer conformational landscapes from such large-scale crystallographic perturbation studies. We apply COLAV to drug fragment screens of two medically important systems: protein tyrosine phosphatase 1B (PTP-1B), which regulates insulin signaling, and the SARS CoV-2 Main Protease (MPro). With enough fragment-bound structures, we find that such drug screens also enable detailed mapping of proteins\' conformational landscapes.
    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的变体由于其增加的传播性和逃避自然免疫的能力而对公共卫生构成重大挑战,疫苗保护,和单克隆抗体疗法。高度传播的Omicron变体和随后的亚变体的出现,特征在于刺突蛋白中超过32个突变的广泛阵列,加剧了对逃避疫苗的担忧。作为回应,多种抗病毒疗法已获得FDA紧急使用批准,靶向SARS-CoV-2RNA依赖性RNA聚合酶(RdRp)和主要蛋白酶(Mpro)区域,已知在新变体中突变相对较少。在这项研究中,我们评估了尼马特雷韦(PF-07321332)和其他具有临床意义的SARS-CoV-2抗病毒药物对多种SARS-CoV-2变体的疗效,包括新鉴定的Omicron亚变体XBB1.5和JN.1,使用活病毒抗病毒测定。我们的发现表明,虽然最后的Omicron亚变体在我们的动物模型中表现出更高的致病性,nirmatrelvir和其他临床相关的抗病毒药物始终保持其对所有测试变体的疗效,包括XBB1.5亚变体。
    Variants of SARS-CoV-2 pose significant challenges in public health due to their increased transmissibility and ability to evade natural immunity, vaccine protection, and monoclonal antibody therapeutics. The emergence of the highly transmissible Omicron variant and subsequent subvariants, characterized by an extensive array of over 32 mutations within the spike protein, intensifies concerns regarding vaccine evasion. In response, multiple antiviral therapeutics have received FDA emergency use approval, targeting the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and main protease (Mpro) regions, known to have relatively fewer mutations across novel variants. In this study, we evaluated the efficacy of nirmatrelvir (PF-07321332) and other clinically significant SARS-CoV-2 antivirals against a diverse panel of SARS-CoV-2 variants, encompassing the newly identified Omicron subvariants XBB1.5 and JN.1, using live-virus antiviral assays. Our findings demonstrate that while the last Omicron subvariants exhibited heightened pathogenicity in our animal model, nirmatrelvir and other clinically relevant antivirals consistently maintained their efficacy against all tested variants, including the XBB1.5 subvariant.
    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
    在严重的COVID-19病例中,炎症反应加剧会引发细胞因子风暴,从而使预后恶化。具有抗病毒和抗炎活性的化合物有望成为COVID-19治疗的候选药物,因为无论疾病阶段如何,它们都可能对SARS-CoV-2感染起作用。冠状病毒中最有吸引力的药物靶标之一是主要蛋白酶(MPro)。这种酶对于将多蛋白切割成病毒复制所需的非结构蛋白至关重要。这篇综述的目的是鉴定具有抗病毒和抗炎特性的SARS-CoV-2MPro抑制剂。当无法获得来自晶体结构的数据时,通过分子对接分析了SARS-CoV-2MPro结合位点内化合物的相互作用。选择18个化合物并将其分为5个不同的超类。其中五个对MPro具有很高的效力:GC-376,黄芩素,柚皮苷,肝素,和卡莫弗,IC50值低于0.2μM。所选择的MPro抑制剂具有减轻肺水肿和减少细胞因子释放的潜力。这些分子主要针对三个关键的炎症途径:NF-κB,JAK/STAT,和MAPK,以前都与COVID-19发病机制有关。化合物的结构占据了MPro的S1/S2底物结合亚位点。它们与催化二元体的残基(His41和Cys145)和/或氧阴离子孔(Gly143,Ser144和Cys145)相互作用,对于底物识别至关重要。此处存在的具有潜在抗炎活性的MProSARS-CoV-2抑制剂可以进行优化,以获得最大的疗效和安全性,并可作为轻度和重度COVID-19的潜在治疗方法进行探索。
    In severe COVID-19 cases, an exacerbated inflammatory response triggers a cytokine storm that can worsen the prognosis. Compounds with both antiviral and anti-inflammatory activities show promise as candidates for COVID-19 therapy, as they potentially act against the SARS-CoV-2 infection regardless of the disease stage. One of the most attractive drug targets among coronaviruses is the main protease (MPro). This enzyme is crucial for cleaving polyproteins into non-structural proteins required for viral replication. The aim of this review was to identify SARS-CoV-2 MPro inhibitors with both antiviral and anti-inflammatory properties. The interactions of the compounds within the SARS-CoV-2 MPro binding site were analyzed through molecular docking when data from crystallographic structures were unavailable. 18 compounds were selected and classified into five different superclasses. Five of them exhibit high potency against MPro: GC-376, baicalein, naringenin, heparin, and carmofur, with IC50 values below 0.2 μM. The MPro inhibitors selected have the potential to alleviate lung edema and decrease cytokine release. These molecules mainly target three critical inflammatory pathways: NF-κB, JAK/STAT, and MAPK, all previously associated with COVID-19 pathogenesis. The structures of the compounds occupy the S1/S2 substrate binding subsite of the MPro. They interact with residues from the catalytic dyad (His41 and Cys145) and/or with the oxyanion hole (Gly143, Ser144, and Cys145), which are pivotal for substrate recognition. The MPro SARS-CoV-2 inhibitors with potential anti-inflammatory activities present here could be optimized for maximum efficacy and safety and be explored as potential treatment of both mild and severe COVID-19.
    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
    背景:猪流行性腹泻病毒(PEDV)主要引起急性和严重的猪流行性腹泻(PED),并且在新生仔猪中高度致命。没有可靠的治疗方法,这对仔猪来说是一个重大的全球健康问题。木犀草素是对几种病毒具有抗病毒活性的类黄酮。
    结果:我们评估了木犀草素在PEDV感染的Vero和IPEC-J2细胞中的抗病毒作用,并确定IC50值为23.87µM和68.5µM,分别。发现PEDV内化,木犀草素处理后,复制和释放显着减少。由于木犀草素可以与人ACE2和SARS-CoV-2主要蛋白酶(Mpro)结合以促进病毒进入,我们首先通过分子对接确定木犀草素与PEDV-S在pACE2上共享相同的核心结合位点,并通过表面等离子体共振(SPR)分析在剂量依赖性增加时显示pACE2阳性结合,亲和常数为71.6µM.然而,pACE2不能与PEDV-S1结合。因此,木犀草素抑制PEDV内化,不依赖于PEDV-S与pACE2的结合。此外,在三维对接模型中,木犀草素被牢固地嵌入Mpro活动口袋的凹槽中,和荧光共振能量转移(FRET)测定证实木犀草素抑制PEDVMpro活性。此外,我们还观察到PEDV诱导的促炎细胞因子抑制和Nrf2诱导的HO-1表达。最后,在10次细胞培养传代后分离出一个耐药突变体,伴随着木犀草素浓度的增加,在第10代鉴定的PEDV对木犀草素的敏感性降低。
    结论:我们的结果推动了木犀草素的抗PEDV机制和抗PEDV特性,可以用来对抗PED.
    BACKGROUND: Porcine epidemic diarrhea virus (PEDV) mainly causes acute and severe porcine epidemic diarrhea (PED), and is highly fatal in neonatal piglets. No reliable therapeutics against the infection exist, which poses a major global health issue for piglets. Luteolin is a flavonoid with anti-viral activity toward several viruses.
    RESULTS: We evaluated anti-viral effects of luteolin in PEDV-infected Vero and IPEC-J2 cells, and identified IC50 values of 23.87 µM and 68.5 µM, respectively. And found PEDV internalization, replication and release were significantly reduced upon luteolin treatment. As luteolin could bind to human ACE2 and SARS-CoV-2 main protease (Mpro) to contribute viral entry, we first identified that luteolin shares the same core binding site on pACE2 with PEDV-S by molecular docking and exhibited positive pACE2 binding with an affinity constant of 71.6 µM at dose-dependent increases by surface plasmon resonance (SPR) assay. However, pACE2 was incapable of binding to PEDV-S1. Therefore, luteolin inhibited PEDV internalization independent of PEDV-S binding to pACE2. Moreover, luteolin was firmly embedded in the groove of active pocket of Mpro in a three-dimensional docking model, and fluorescence resonance energy transfer (FRET) assays confirmed that luteolin inhibited PEDV Mpro activity. In addition, we also observed PEDV-induced pro-inflammatory cytokine inhibition and Nrf2-induced HO-1 expression. Finally, a drug resistant mutant was isolated after 10 cell culture passages concomitant with increasing luteolin concentrations, with reduced PEDV susceptibility to luteolin identified at passage 10.
    CONCLUSIONS: Our results push forward that anti-PEDV mechanisms and resistant-PEDV properties for luteolin, which may be used to combat PED.
    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
    冠状病毒病19大流行,由严重急性呼吸综合征冠状病毒2(SARS-CoV-2)引起,导致全球健康危机,数百万例确诊病例和相关死亡。SARS-CoV-2的主要蛋白酶(Mpro)对于病毒复制至关重要,并为药物开发提供了有吸引力的靶标。尽管某些药物获得批准,继续寻找有效的治疗方法。在这项研究中,我们系统地评估了Mpro的342个完整晶体结构,以确定基于结构的虚拟筛选(SBVS)的最佳构象。我们的分析表明,结构之间的结构灵活性有限。三个对接程序,AutoDockVina,rDock,和Glide被用来评估虚拟筛查的效率,揭示了选定的Mpro结构的不同表现。我们发现结构5RHE,7DDC,和7DPU(PDBIds)一致显示最低EF,AUC,和BEDROCK得分。此外,这些结构在所有对接程序中表现出最差的姿态预测结果。两个结构差异导致对接性能的变化:7DDC和7DPU中缺少S1子位点,并且在7DDC的S2子站点中存在一个子袋,7DPU,5RHE这些发现强调了为SBVS选择合适的Mpro构象的重要性,为推进药物发现工作提供有价值的见解。
    The coronavirus disease 19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global health crisis with millions of confirmed cases and related deaths. The main protease (Mpro) of SARS-CoV-2 is crucial for viral replication and presents an attractive target for drug development. Despite the approval of some drugs, the search for effective treatments continues. In this study, we systematically evaluated 342 holo-crystal structures of Mpro to identify optimal conformations for structure-based virtual screening (SBVS). Our analysis revealed limited structural flexibility among the structures. Three docking programs, AutoDock Vina, rDock, and Glide were employed to assess the efficiency of virtual screening, revealing diverse performances across selected Mpro structures. We found that the structures 5RHE, 7DDC, and 7DPU (PDB Ids) consistently displayed the lowest EF, AUC, and BEDROCK scores. Furthermore, these structures demonstrated the worst pose prediction results in all docking programs. Two structural differences contribute to variations in docking performance: the absence of the S1 subsite in 7DDC and 7DPU, and the presence of a subpocket in the S2 subsite of 7DDC, 7DPU, and 5RHE. These findings underscore the importance of selecting appropriate Mpro conformations for SBVS, providing valuable insights for advancing drug discovery efforts.
    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:最近提出了SARS-CoV-2的病毒主要蛋白酶(Mpro)作为抑制病毒在宿主中复制的关键靶标。因此,可以结合Mpro催化位点的分子可以被认为是治疗SARS-CoV-2感染的潜在药物候选物。在这里,我们提出了一个最先进的分析平台的应用,该平台结合了代谢组学和蛋白质结构分析,以挖掘源自天然基质的潜在活性化合物。即,蓝莓提取物.
    方法:实验的重点是寻找Mpro的MS共价抑制剂,其结构中含有能够与酶催化位点的亲核氨基酸结合的儿茶酚/连苯三酚部分。
    结果:在确定的潜在候选人中,delphinidin-3-葡萄糖苷显示了最有希望的结果。已在体外对感染SARS-CoV-2的VeroE6细胞证实了其抗病毒活性,显示出与已知的Mpro抑制剂黄芩苷几乎相当的剂量依赖性抑制作用。还通过计算研究评估了飞跃素-3-葡萄糖苷与Mpro口袋的相互作用。
    结论:所描述的HRMS分析平台被证明可有效地鉴定共价结合Mpro的化合物,并且在抑制SARS-CoV-2复制方面具有活性,如Delphinidin-3-葡萄糖苷。
    BACKGROUND: The viral main protease (Mpro) of SARS-CoV-2 has been recently proposed as a key target to inhibit virus replication in the host. Therefore, molecules that can bind the catalytic site of Mpro could be considered as potential drug candidates in the treatment of SARS-CoV-2 infections. Here we proposed the application of a state-of-the-art analytical platform which combines metabolomics and protein structure analysis to fish-out potential active compounds deriving from a natural matrix, i.e., a blueberry extract.
    METHODS: The experiments focus on finding MS covalent inhibitors of Mpro that contain in their structure a catechol/pyrogallol moiety capable of binding to the nucleophilic amino acids of the enzyme\'s catalytic site.
    RESULTS: Among the potential candidates identified, the delphinidin-3-glucoside showed the most promising results. Its antiviral activity has been confirmed in vitro on Vero E6 cells infected with SARS-CoV-2, showing a dose-dependent inhibitory effect almost comparable to the known Mpro inhibitor baicalin. The interaction of delphinidin-3-glucoside with the Mpro pocket observed was also evaluated by computational studies.
    CONCLUSIONS: The HRMS analytical platform described proved to be effective in identifying compounds that covalently bind Mpro and are active in the inhibition of SARS-CoV-2 replication, such as delphinidin-3-glucoside.
    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
    虽然研究已经确定了SARS-CoV-2的主要蛋白酶(Mpro)的几种抑制剂,但这些化合物的很大一部分在还原剂存在下表现出降低的活性。引起人们对其体内有效性的担忧。此外,使用病毒颗粒的细胞试验的常规生物安全3级(BSL-3)对基于细胞的试验中Mpro抑制剂功效的广泛评估造成了限制.这里,我们建立了一个与BSL-1相容的细胞试验来评估Mpro抑制剂的体内潜力.该测定利用表达含有N-末端谷胱甘肽S-转移酶(GST)和C-末端血凝素(HA)标签的标记的Mpro构建体的哺乳动物细胞并监测Mpro自身消化。使用此方法,GC376和boceprevir有效抑制Mpro自身消化,表明它们在体内的潜在活性。相反,卡莫富和依布硒在该测定中没有表现出明显的抑制作用。我们使用这种方法进一步研究了硒酮对Mpro的抑制潜力。结合能的计算分析表明,非共价相互作用在促进C145残基的共价修饰中起关键作用。导致Mpro抑制。我们的方法很简单,成本效益高,并容易适用于标准实验室,使具有不同传染病专业知识水平的研究人员可以使用它。
    While research has identified several inhibitors of the main protease (Mpro) of SARS-CoV-2, a significant portion of these compounds exhibit reduced activity in the presence of reducing agents, raising concerns about their effectiveness in vivo. Furthermore, the conventional biosafety level 3 (BSL-3) for cellular assays using viral particles poses a limitation for the widespread evaluation of Mpro inhibitor efficacy in a cell-based assay. Here, we established a BSL-1 compatible cellular assay to evaluate the in vivo potential of Mpro inhibitors. This assay utilizes mammalian cells expressing a tagged Mpro construct containing N-terminal glutathione S-transferase (GST) and C-terminal hemagglutinin (HA) tags and monitors Mpro autodigestion. Using this method, GC376 and boceprevir effectively inhibited Mpro autodigestion, suggesting their potential in vivo activity. Conversely, carmofur and ebselen did not exhibit significant inhibitory effects in this assay. We further investigated the inhibitory potential of selenoneine on Mpro using this approach. Computational analyses of binding energies suggest that noncovalent interactions play a critical role in facilitating the covalent modification of the C145 residue, leading to Mpro inhibition. Our method is straightforward, cost-effective, and readily applicable in standard laboratories, making it accessible to researchers with varying levels of expertise in infectious 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
    COVID-19大流行主要由SARS-CoV-2引起,其主要蛋白酶,Mpro,在病毒复制中起关键作用,并作为抑制不同变体的潜在靶标。在这项研究中,从甘草酸(GL)衍生物与氨基酸甲酯/乙酯中鉴定出有效的Mpro抑制剂。在17种半合成的衍生物中,化合物2,6,9和15,与甲硫氨酸甲酯,D-酪氨酸甲酯,谷氨酸甲酯,和碳水化合物部分中的蛋氨酸,分别,显著抑制野生型SARS-CoV-2Mpro介导的蛋白水解,IC50值范围为0.06μM至0.84μM。他们还证明了抑制突变Mpro变体的反式切割的功效(Mpro_P132H,Mpro_E166V,Mpro_P168A,Mpro_Q189I),IC50值范围为0.05至0.92μM,超过尼马特雷韦(IC50:1.17-152.9μM)。分子模型显示,与Nirmatrelvir相比,Mpro_E166V与化合物的结构复合物中与Valine166的相互作用更强。此外,这些化合物有效地抑制了野生型SARS-CoV-2单轮感染颗粒(SRIPs)的进入后病毒过程,减轻病毒细胞病变效应和减少复制子驱动的GFP报告信号,以及野生型的体外感染性,Mpro_E166V,和Mpro_Q189ISRIP,EC50值范围为0.02至0.53μM。然而,与野生型SRIP(EC50:0.06μM)相比,nirmatrelvir在抑制携带Mpro_E166V(EC50:>20μM)和Mpro_Q189I(EC50:13.2μM)的突变型SARS-CoV-2SRIP的复制方面表现出显着降低。总的来说,这项研究确定了四种GL衍生物是有前途的先导化合物,可用于开发针对各种SARS-CoV-2菌株的治疗方法,包括Omicron,和耐尼马特雷韦的变种。
    COVID-19 pandemic is predominantly caused by SARS-CoV-2, with its main protease, Mpro, playing a pivotal role in viral replication and serving as a potential target for inhibiting different variants. In this study, potent Mpro inhibitors were identified from glycyrrhizic acid (GL) derivatives with amino acid methyl/ethyl esters. Out of the 17 derivatives semisynthesized, Compounds 2, 6, 9, and 15, with methionine methyl esters, D-tyrosine methyl esters, glutamic acid methyl esters, and methionines in the carbohydrate moiety, respectively, significantly inhibited wild-type SARS-CoV-2 Mpro-mediated proteolysis, with IC50 values ranging from 0.06 μM to 0.84 μM. They also demonstrated efficacy in inhibiting trans-cleavage by mutant Mpro variants (Mpro_P132H, Mpro_E166V, Mpro_P168A, Mpro_Q189I), with IC50 values ranging from 0.05 to 0.92 μM, surpassing nirmatrelvir (IC50: 1.17-152.9 μM). Molecular modeling revealed stronger interactions with Valine166 in the structural complex of Mpro_E166V with the compounds compared to nirmatrelvir. Moreover, these compounds efficiently inhibited the post-entry viral processes of wild-type SARS-CoV-2 single-round infectious particles (SRIPs), mitigating viral cytopathic effects and reducing replicon-driven GFP reporter signals, as well as in vitro infectivity of wild-type, Mpro_E166V, and Mpro_Q189I SRIPs, with EC50 values ranging from 0.02 to 0.53 μM. However, nirmatrelvir showed a significant decrease in inhibiting the replication of mutant SARS-CoV-2 SRIPs carrying Mpro_E166V (EC50: >20 μM) and Mpro_Q189I (EC50: 13.2 μM) compared to wild-type SRIPs (EC50: 0.06 μM). Overall, this study identifies four GL derivatives as promising lead compounds for developing treatments against various SARS-CoV-2 strains, including Omicron, and nirmatrelvir-resistant 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
    许多先前的研究已经确定了可以有效对抗严重急性呼吸综合征冠状病毒2(SARS-CoV-2)感染的治疗靶标,包括血管紧张素转换酶2(ACE2)受体,RNA依赖性RNA聚合酶(RdRp),和主要蛋白酶(Mpro)。并行,抗病毒化合物像阿巴卡韦,阿昔洛韦,阿德福韦,金刚烷胺,Amprenaver,darunavir,去羟肌苷,奥司他韦,喷昔洛韦,和替诺福韦正在调查他们的潜在的药物再利用,以解决这种感染。该研究的目的是确定在计算机上修饰上述抗病毒剂的官能团的效果。在Maestro软件(11.1版)上使用遗传优化的配体对接算法,修饰的抗病毒药物停靠在ACE2受体上,RdRp,还有Mpro.使用QuickProp(Maestrov11.1),PASS(物质的活性谱预测),和Swissadme一起,ADMET(吸收,分布,新陈代谢,排泄,和毒性)修饰的抗病毒药物,以及它们的生物利用度和预测的活性谱,决心。使用Discoverystudio软件进行对接后分析。在10种抗病毒药物中,达鲁那韦的N(CH3)2衍生物,Amprenavir的N(CH3)2衍生物和darunavir的NCH3衍生物与ACE2受体表现出最佳的结合亲和力(对接评分:-10.333,-9.527和-9.695kJ/mol,分别)。此外,阿巴卡韦的NCH3衍生物(-6.506kJ/mol),去羟肌苷的NO2衍生物(-6.877kJ/mol),达鲁那韦的NCH3衍生物(-7.618kJ/mol)对Mpro具有良好的亲和力。总之,计算机筛选的结果可以作为未来实验工作的有用信息。
    Numerous prior studies have identified therapeutic targets that could effectively combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, including the angiotensin-converting enzyme 2 (ACE2) receptor, RNA-dependent RNA polymerase (RdRp), and Main protease (Mpro). In parallel, antiviral compounds like abacavir, acyclovir, adefovir, amantadine, amprenavir, darunavir, didanosine, oseltamivir, penciclovir, and tenofovir are under investigation for their potential in drug repurposing to address this infection. The aim of the study was to determine the effect of modifying the functional groups of the aforementioned antivirals in silico. Using the genetic optimization for ligand docking algorithm on software Maestro (version 11.1), the modified antivirals were docked onto ACE2 receptor, RdRp, and Mpro. Using QuickProp (Maestro v11.1), PASS (prediction of activity spectra for the substances), and altogether with SwissADME, the ADMET (absorption, distribution, metabolism, excretion, and toxicity) of the modified antivirals, as well as their bioavailability and the predicted activity spectra, were determined. Discovery studio software was used to undertake post-docking analysis. Among the 10 antivirals, N(CH3)2 derivative of darunavir, N(CH3)2 derivative of amprenavir and NCH3 derivative of darunavir exhibited best binding affinities with ACE2 receptor (docking scores: -10.333, -9.527 and -9.695 kJ/mol, respectively). Moreover, NCH3 derivative of abacavir (-6.506 kJ/mol), NO2 derivative of didanosine (-6.877 kJ/mol), NCH3 derivative of darunavir (-7.618 kJ/mol) exerted promising affinity to Mpro. In conclusion, the results of the in silico screenings can serve as a useful information for future experimental works.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号