Entry inhibitor

进入抑制剂
  • 文章类型: Journal Article
    甲型流感病毒(IAV)是一种广泛存在的病原体,对人类健康构成重大威胁。引起高死亡率和致病性的大流行。鉴于越来越多的IAV耐药菌株的出现,据报道,目前可用的抗病毒药物不足以满足临床需求。因此,不断探索安全,迫切需要有效和广谱的抗病毒药物。这里,我们发现小分子化合物J1在体外和体内都表现出低毒性。此外,J1对包膜病毒具有广谱抗病毒活性,包括IAV,呼吸道合胞病毒(RSV),严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),人类冠状病毒OC43(HCoV-OC43),单纯疱疹病毒1型(HSV-1)和HSV-2。在这项研究中,我们探讨了J1在体内和体外对IAV的抑制作用和作用机制。结果表明,J1抑制了IAV菌株的感染,包括H1N1,H7N9,H5N1和H3N2,以及奥司他韦耐药菌株。机制研究表明,J1主要通过与流感病毒血凝素HA2亚基的特异性相互作用阻断IAV感染,从而阻断膜融合。采用BALB/c小鼠建立IAV诱导的急性肺损伤(ALI)模型。用J1治疗增加存活率和降低病毒滴度,病毒感染小鼠的肺指数和肺部炎症损伤。总之,J1在体外和体内具有显著的抗IAV作用,提供对未来大流行的广谱抗病毒药物发展的见解。
    Influenza A virus (IAV) is a widespread pathogen that poses a significant threat to human health, causing pandemics with high mortality and pathogenicity. Given the emergence of increasingly drug-resistant strains of IAV, currently available antiviral drugs have been reported to be inadequate to meet clinical demands. Therefore, continuous exploration of safe, effective and broad-spectrum antiviral medications is urgently required. Here, we found that the small molecule compound J1 exhibited low toxicity both in vitro and in vivo. Moreover, J1 exhibits broad-spectrum antiviral activity against enveloped viruses, including IAV, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human coronavirus OC43 (HCoV-OC43), herpes simplex virus type 1 (HSV-1) and HSV-2. In this study, we explored the inhibitory effects and mechanism of action of J1 on IAV in vivo and in vitro. The results showed that J1 inhibited infection by IAV strains, including H1N1, H7N9, H5N1 and H3N2, as well as by oseltamivir-resistant strains. Mechanistic studies have shown that J1 blocks IAV infection mainly through specific interactions with the influenza virus hemagglutinin HA2 subunit, thereby blocking membrane fusion. BALB/c mice were used to establish a model of acute lung injury (ALI) induced by IAV. Treatment with J1 increased survival rates and reduced viral titers, lung index and lung inflammatory damage in virus-infected mice. In conclusion, J1 possesses significant anti-IAV effects in vitro and in vivo, providing insights into the development of broad-spectrum antivirals against future pandemics.
    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
    先前的数据表明gp120定向的附着抑制剂temsavir(fosemsavir的活性部分)和CD4定向的附着后抑制剂ibalizumab之间缺乏交叉抗性。最近,对两种抑制剂敏感性降低的HIV-1包膜进行分析,以确定它们是否具有耐药的基因型相关性.将对两种药物的敏感性降低的2个包膜的序列映射到结合temsavir的gp120结构上。使用反向遗传学评估了temsavir结合位点5.0µ内的残基。使用3期BRIGHTE研究参与者的信封进一步评估了关键替代的更广泛适用性和上下文决定因素。通过半最大抑制浓度(IC50)测量坦沙韦敏感性,通过IC50和最大抑制百分比(MPI)测量ibalizumab敏感性。一个包膜需要E113D和T434M的替换以完全恢复坦沙韦敏感性。替代或其组合均不影响ibalizumab敏感性。然而,在第二个信封里,E202取代(HXB2,T202)足以观察到对两种抑制剂的敏感性丧失。一名没有ibalizumab暴露的BRIGHTE参与者在方案定义的病毒学失败时出现了K202E替换,对两种抑制剂的敏感性降低。将T202E引入先前易感的临床分离株中,将temsavir效价降低≥40倍,将ibalizumabMPI从>99%降低至~80%。有趣的是,从高度易感的ibalizumab包膜引入gp120V5区域减轻了E202对ibalizumab的影响,但不能减轻对temsavir的影响.一种罕见的HIV-1gp120E202突变降低了坦沙韦易感性,根据序列上下文,可能导致对ibalizumab的敏感性降低。
    Previous data suggest a lack of cross-resistance between the gp120-directed attachment inhibitor temsavir (active moiety of fostemsavir) and the CD4-directed post-attachment inhibitor ibalizumab. Recently, analysis of HIV-1 envelopes with reduced sensitivity to both inhibitors was undertaken to determine whether they shared genotypic correlates of resistance. Sequences from 2 envelopes with reduced susceptibility to both agents were mapped onto a temsavir-bound gp120 structure. Residues within 5.0 Å of the temsavir binding site were evaluated using reverse genetics. Broader applicability and contextual determinants of key substitutions were further assessed using envelopes from participants in the phase 3 BRIGHTE study. Temsavir sensitivity was measured by half-maximal inhibitory concentration (IC50) and ibalizumab sensitivity by IC50 and maximum percent inhibition (MPI). One envelope required substitutions of E113D and T434M for full restoration of temsavir susceptibility. Neither substitution nor their combination affected ibalizumab sensitivity. However, in the second envelope, an E202 substitution (HXB2, T202) was sufficient for observed loss of susceptibility to both inhibitors. One BRIGHTE participant with no ibalizumab exposure had an emergent K202E substitution at protocol-defined virologic failure, with reduced sensitivity to both inhibitors. Introducing T202E into previously susceptible clinical isolates reduced temsavir potency by ≥ 40-fold and ibalizumab MPI from >99% to ∼80%. Interestingly, introduction of the gp120 V5 region from a highly ibalizumab-susceptible envelope mitigated the E202 effect on ibalizumab but not temsavir. A rare HIV-1 gp120 E202 mutation reduced temsavir susceptibility, and depending on sequence context, could result in reduced susceptibility to ibalizumab.
    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
    由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)引起的2019年冠状病毒病(COVID-19)构成了巨大的社会经济负担,对抗COVID-19势在必行。阻断SARS-CoV-2RBD-ACE2的相互作用是治疗病毒感染的一种有希望的方法,SARS-CoV-2通过刺突蛋白的RBD与宿主细胞的ACE2受体结合以渗入这些细胞。我们使用计算机辅助药物设计技术和细胞实验,通过对SARS-CoV-2和ACE2相互作用的结构分析,筛选对人ACE2受体具有高亲和力和特异性的肽S4。细胞实验表明,肽S4有效抑制SARS-CoV-2和HCoV-NL63病毒感染宿主细胞,并且在有效浓度下对细胞是安全的。基于这些发现,肽S4可能是治疗正在进行的SARS-CoV-2大流行的临床应用的潜在药物。
    Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant socioeconomic burden, and combating COVID-19 is imperative. Blocking the SARS-CoV-2 RBD-ACE2 interaction is a promising therapeutic approach for viral infections, as SARS-CoV-2 binds to the ACE2 receptors of host cells via the RBD of spike proteins to infiltrate these cells. We used computer-aided drug design technology and cellular experiments to screen for peptide S4 with high affinity and specificity for the human ACE2 receptor through structural analysis of SARS-CoV-2 and ACE2 interactions. Cellular experiments revealed that peptide S4 effectively inhibited SARS-CoV-2 and HCoV-NL63 viruses from infecting host cells and was safe for cells at effective concentrations. Based on these findings, peptide S4 may be a potential pharmaceutical agent for clinical application in the treatment of the ongoing SARS-CoV-2 pandemic.
    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
    寨卡病毒(ZIKV)是蚊媒黄病毒之一,对神经系统表现出独特的嗜性,并与格林-巴利综合征和先天性寨卡综合征(CZS)有关。登革热病毒(DENV)和黄热病病毒(YFV),另外两种蚊媒黄病毒,也已经传播了很长时间并导致严重的疾病,如登革热出血热和黄热病,分别。然而,目前还没有安全有效的抗病毒药物被批准用于治疗这些黄病毒的感染或合并感染。这里,我们发现扎鲁司特,一种怀孕安全的白三烯受体拮抗剂,在不同细胞系中对来自不同谱系的ZIKV菌株的感染表现出有效的抗病毒活性,以及针对DENV-2和YFV17D的感染。机制研究表明,扎鲁司特通过破坏病毒体的完整性,直接和不可逆地灭活这些黄病毒,导致病毒感染性的丧失,从而抑制病毒感染的进入步骤。考虑到它对黄病毒的功效,它对孕妇的安全性,以及它的神经保护作用,扎鲁司特是预防和治疗ZIKV感染或合并感染的有前途的候选药物,DENV,YFV,即使是孕妇。
    Zika virus (ZIKV) is one of the mosquito-borne flaviviruses that exhibits a unique tropism to nervous systems and is associated with Guillain-Barre syndrome and congenital Zika syndrome (CZS). Dengue virus (DENV) and yellow fever virus (YFV), the other two mosquito-borne flaviviruses, have also been circulating for a long time and cause severe diseases, such as dengue hemorrhagic fever and yellow fever, respectively. However, there are no safe and effective antiviral drugs approved for the treatment of infections or coinfections of these flaviviruses. Here, we found that zafirlukast, a pregnancy-safe leukotriene receptor antagonist, exhibited potent antiviral activity against infections of ZIKV strains from different lineages in different cell lines, as well as against infections of DENV-2 and YFV 17D. Mechanistic studies demonstrated that zafirlukast directly and irreversibly inactivated these flaviviruses by disrupting the integrity of the virions, leading to the loss of viral infectivity, hence inhibiting the entry step of virus infection. Considering its efficacy against flaviviruses, its safety for pregnant women, and its neuroprotective effect, zafirlukast is a promising candidate for prophylaxis and treatment of infections or coinfections of ZIKV, DENV, and YFV, even in pregnant women.
    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
    拉沙病毒(LASV)是人类拉沙热的病原体,其在严重的情况下表现为每年导致数千人死亡的出血热。然而,目前尚无批准的疫苗或抗病毒药物。最近,我们使用表达LASV糖蛋白GP(VSV-LASVGP)的重组水泡性口炎病毒(VSV)筛选了约2,500种化合物,并鉴定了P-糖蛋白抑制剂为潜在的LASV进入抑制剂.这里,我们显示另一个确定的候选人,hexestrol(HES),雌激素受体激动剂,也是LASV进入抑制剂。HES以0.63μM的50%抑制浓度(IC50)抑制VSV-LASVGP复制。重要的是,HES还抑制真正的LASV复制,IC50值为0.31µM-0.61µM。添加时间和基于细胞的膜融合测定表明HES在病毒进入期间抑制膜融合步骤。替代雌激素受体激动剂不抑制VSV-LASVGP复制,这表明雌激素受体本身不太可能参与HES的抗病毒活性。HES抗性突变体的产生揭示了LASVGP氨基酸位置446(F446)的苯丙氨酸,它位于跨膜区域,赋予了对HES的抵抗力。尽管F446的突变增强了LASVGP的膜融合活性,它表现出减少的VSV-LASVGP复制,很可能是由于LASVGP融合前状态的不稳定性。总的来说,我们的结果表明,HES是一种有前景的抗LASV药物,其通过抑制LASV进入的膜融合步骤起作用.这项研究还强调了LASVGP跨膜区作为抗LASV药物靶标的重要性。IMPORTANCELassa病毒(LASV),拉沙热的病原体,就其对西非公共卫生的影响而言,是最具破坏性的哺乳动物病毒。然而,目前尚无批准的抗病毒药物或疫苗。这里,我们确定了hexestrol(HES),雌激素受体激动剂,作为潜在的抗病毒候选药物。我们表明,雌激素受体本身不参与抗病毒活性。HES直接与LASVGP结合并阻断膜融合,从而抑制LASV感染。通过产生抗HES病毒,我们发现LASVGP跨膜区446位(F446)的苯丙氨酸在HES的抗病毒活性中起着至关重要的作用。F446突变导致病毒复制减少,可能是由于LASVGP融合前状态的不稳定性。这些发现突出了HES作为开发靶向LASV的抗病毒化合物的有希望的候选物的潜力。
    Lassa virus (LASV) is the causative agent of human Lassa fever which in severe cases manifests as hemorrhagic fever leading to thousands of deaths annually. However, no approved vaccines or antiviral drugs are currently available. Recently, we screened approximately 2,500 compounds using a recombinant vesicular stomatitis virus (VSV) expressing LASV glycoprotein GP (VSV-LASVGP) and identified a P-glycoprotein inhibitor as a potential LASV entry inhibitor. Here, we show that another identified candidate, hexestrol (HES), an estrogen receptor agonist, is also a LASV entry inhibitor. HES inhibited VSV-LASVGP replication with a 50% inhibitory concentration (IC50) of 0.63 µM. Importantly, HES also inhibited authentic LASV replication with IC50 values of 0.31 µM-0.61 µM. Time-of-addition and cell-based membrane fusion assays suggested that HES inhibits the membrane fusion step during virus entry. Alternative estrogen receptor agonists did not inhibit VSV-LASVGP replication, suggesting that the estrogen receptor itself is unlikely to be involved in the antiviral activity of HES. Generation of a HES-resistant mutant revealed that the phenylalanine at amino acid position 446 (F446) of LASVGP, which is located in the transmembrane region, conferred resistance to HES. Although mutation of F446 enhanced the membrane fusion activity of LASVGP, it exhibited reduced VSV-LASVGP replication, most likely due to the instability of the pre-fusion state of LASVGP. Collectively, our results demonstrated that HES is a promising anti-LASV drug that acts by inhibiting the membrane fusion step of LASV entry. This study also highlights the importance of the LASVGP transmembrane region as a target for anti-LASV drugs.IMPORTANCELassa virus (LASV), the causative agent of Lassa fever, is the most devastating mammarenavirus with respect to its impact on public health in West Africa. However, no approved antiviral drugs or vaccines are currently available. Here, we identified hexestrol (HES), an estrogen receptor agonist, as the potential antiviral candidate drug. We showed that the estrogen receptor itself is not involved in the antiviral activity. HES directly bound to LASVGP and blocked membrane fusion, thereby inhibiting LASV infection. Through the generation of a HES-resistant virus, we found that phenylalanine at position 446 (F446) within the LASVGP transmembrane region plays a crucial role in the antiviral activity of HES. The mutation at F446 caused reduced virus replication, likely due to the instability of the pre-fusion state of LASVGP. These findings highlight the potential of HES as a promising candidate for the development of antiviral compounds targeting LASV.
    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
    自从COVID-19大流行开始以来,广泛的药物再利用努力寻求鉴定具有各种作用机制的小分子抗病毒药物.这里,我们旨在综述与SARS-CoV-2Spike蛋白直接结合的小分子病毒进入和融合抑制剂的研究进展。在大流行的早期,在药物再利用筛选中鉴定出许多小分子,据报道在体外SARS-CoV-2病毒进入或融合抑制剂中有效。然而,给出了关于Spike上小分子结合位点确切位置的最少实验信息,目前尚不清楚某些候选抑制剂的具体作用机制或确切结合位点在Spike上的位置.无数研究人员的工作取得了巨大进步,鉴定了许多靶向S1受体结合域(RBD)或N末端域(NTD)上的元件并破坏S1受体结合功能的病毒进入抑制剂。在这次审查中,我们还将重点强调靶向抑制S2融合功能的融合抑制剂,通过破坏融合后S2构象的形成,或者通过稳定融合前S2构象的结构元件以防止与S2功能相关的构象变化。我们重点介绍了S1/S2界面和S2亚基上经过实验验证的结合位点。尽管迄今为止关注变体(VOCs)中对Spike蛋白的大多数替换都位于S1亚基,S2亚基序列更保守,在S2结合位点附近仅观察到少数取代。已显示几种最近靶向S2的小分子对最近的VOC突变株具有强大的活性和/或对其他更远距离相关的冠状病毒具有更大的广谱抗病毒活性。
    Since the beginning of the COVID-19 pandemic, extensive drug repurposing efforts have sought to identify small-molecule antivirals with various mechanisms of action. Here, we aim to review research progress on small-molecule viral entry and fusion inhibitors that directly bind to the SARS-CoV-2 Spike protein. Early in the pandemic, numerous small molecules were identified in drug repurposing screens and reported to be effective in in vitro SARS-CoV-2 viral entry or fusion inhibitors. However, given minimal experimental information regarding the exact location of small-molecule binding sites on Spike, it was unclear what the specific mechanism of action was or where the exact binding sites were on Spike for some inhibitor candidates. The work of countless researchers has yielded great progress, with the identification of many viral entry inhibitors that target elements on the S1 receptor-binding domain (RBD) or N-terminal domain (NTD) and disrupt the S1 receptor-binding function. In this review, we will also focus on highlighting fusion inhibitors that target inhibition of the S2 fusion function, either by disrupting the formation of the postfusion S2 conformation or alternatively by stabilizing structural elements of the prefusion S2 conformation to prevent conformational changes associated with S2 function. We highlight experimentally validated binding sites on the S1/S2 interface and on the S2 subunit. While most substitutions to the Spike protein to date in variants of concern (VOCs) have been localized to the S1 subunit, the S2 subunit sequence is more conserved, with only a few observed substitutions in proximity to S2 binding sites. Several recent small molecules targeting S2 have been shown to have robust activity over recent VOC mutant strains and/or greater broad-spectrum antiviral activity for other more distantly related coronaviruses.
    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
    尽管在开发疫苗和抗病毒药物以对抗COVID-19方面取得了相当大的进展,但SARS-CoV-2基因组的快速突变限制了当前疫苗和治疗干预措施的持久性和有效性。因此,它需要开发新的治疗方法或重新利用现有的靶向病毒生命周期的药物,宿主因素,或者两者兼而有之。这里,我们报道了SRX3177,一种有效的三重活性CDK4/6-PI3K-BET抑制剂,在亚微摩尔浓度下以IC50值阻断SARS-CoV-2Omicron变体的复制,而在其IC50浓度及以下对Calu-3细胞的细胞增殖没有任何影响。当SRX3177与EIDD-1931(小分子前药Molnupiravir的活性部分)或MU-UNMC-2(SARS-CoV-2进入抑制剂)以固定剂量矩阵组合时,观察到协同效应,导致单个化合物的剂量显着降低,以实现对SARS-CoV-2复制的类似抑制。研究结果表明,SRX3177/MPV或SRX3177/UM-UNMC-2的组合具有进一步开发作为SARS-CoV-2和任何未来β冠状病毒爆发的组合疗法的潜力。
    Despite considerable progress in developing vaccines and antivirals to combat COVID-19, the rapid mutations of the SARS-CoV-2 genome have limited the durability and efficacy of the current vaccines and therapeutic interventions. Hence, it necessitates the development of novel therapeutic approaches or repurposing existing drugs that target either viral life cycle, host factors, or both. Here, we report that SRX3177, a potent triple-activity CDK4/6-PI3K-BET inhibitor, blocks replication of the SARS-CoV-2 Omicron variant with IC50 values at sub-micromolar concentrations without any impact on the cell proliferation of Calu-3 cells at and below its IC50 concentration. When SRX3177 is combined with EIDD-1931 (active moiety of a small-molecule prodrug Molnupiravir) or MU-UNMC-2 (a SARS-CoV-2 entry inhibitor) at a fixed doses matrix, a synergistic effect was observed, leading to the significant reduction in the dose of the individual compounds to achieve similar inhibition of SARS-CoV-2 replication. Herein, we report that the combination of SRX3177/MPV or SRX3177/UM-UNMC-2 has the potential for further development as a combinational therapy against SARS-CoV-2 and in any future outbreak of beta coronavirus.
    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
    补体系统是针对微生物感染的先天免疫机制。它涉及一系列通过经典激活的效应分子,凝集素和替代途径。因此,许多病原体结合或在其结构中包含补体途径的负调节因子作为逃避机制。因子H(FH)是补体替代途径的负调节因子,可保护宿主的“自身”细胞免受非特异性补体攻击。已显示FH结合包括人甲型流感病毒(IAV)的病毒。除了参与调节补体激活,FH也已显示其自身执行一系列功能,包括其与病原体的直接相互作用。这里,我们表明人类FH可以直接与人类和鸟类来源的IAVs结合,并且该相互作用是通过IAV表面糖蛋白血凝素(HA)介导的。HA结合到FH结构上的常见病原体结合足迹,补体控制蛋白模块,CCP5-7和CCP15-20。FH与H1和H3的结合表明,相互作用与两个HAs的受体结合位点重叠,但是H3HA的足迹比H1HA更广泛。HA-FH相互作用阻碍了H1N1和H3N2IAV株的初始进入,但其对人肺细胞中病毒多循环复制的影响是菌株特异性的。与FH预孵育可显著抑制H3N2病毒与细胞的结合,而人H1N1,禽H9N2和H5N3IAV毒株的复制率和子代病毒释放没有变化。我们已经绘制了FH和IAV之间的相互作用,其对病毒或宿主的体内意义尚待阐明。
    The complement system is an innate immune mechanism against microbial infections. It involves a cascade of effector molecules that is activated via classical, lectin and alternative pathways. Consequently, many pathogens bind to or incorporate in their structures host negative regulators of the complement pathways as an evasion mechanism. Factor H (FH) is a negative regulator of the complement alternative pathway that protects \"self\" cells of the host from non-specific complement attack. FH has been shown to bind viruses including human influenza A viruses (IAVs). In addition to its involvement in the regulation of complement activation, FH has also been shown to perform a range of functions on its own including its direct interaction with pathogens. Here, we show that human FH can bind directly to IAVs of both human and avian origin, and the interaction is mediated via the IAV surface glycoprotein haemagglutinin (HA). HA bound to common pathogen binding footprints on the FH structure, complement control protein modules, CCP 5-7 and CCP 15-20. The FH binding to H1 and H3 showed that the interaction overlapped with the receptor binding site of both HAs, but the footprint was more extensive for the H3 HA than the H1 HA. The HA - FH interaction impeded the initial entry of H1N1 and H3N2 IAV strains but its impact on viral multicycle replication in human lung cells was strain-specific. The H3N2 virus binding to cells was significantly inhibited by preincubation with FH, whereas there was no alteration in replicative rate and progeny virus release for human H1N1, or avian H9N2 and H5N3 IAV strains. We have mapped the interaction between FH and IAV, the in vivo significance of which for the virus or host is yet to be elucidated.
    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
    人类呼吸道病毒对国家卫生系统产生巨大影响,社会,由于这种病原体的快速空气传播和流行传播,而有效的特异性抗病毒药物来对抗感染仍然缺乏。这里,我们确定了两种Keggin型多金属氧酸盐(POM),[TiW11CoO40]8-(TiW11Co)和[Ti2PW10O40]7-(Ti2PW10),具有针对有包膜和无包膜人类呼吸道病毒的广谱活性,即,冠状病毒(HCoV-OC43),鼻病毒(HRV-A1),呼吸道合胞病毒(RSV-A2),和腺病毒(AdV-5)。Ti2PW10对所有测试的病毒显示出非常有利的选择性指数(SI>700),并进一步研究了其针对人类冠状病毒和鼻病毒的抗病毒潜力。发现该POM抑制多种HCoV和HRV菌株的复制,在不同的细胞系统中。Ti2PW10不影响病毒结合或细胞内病毒复制,但选择性地抑制了病毒的进入。在POM存在下病毒的连续传代揭示了HRV-A1或HCoV-OC43的Ti2PW10抗性变体的发展的高度障碍。此外,Ti2PW10能够在人类鼻上皮的3D模型中抑制HRV-A1的产生,重要的是,抗病毒治疗未确定细胞毒性或组织损伤.还为Ti2PW10开发了用于鼻递送的粘膜粘附热敏原位水凝胶制剂。总的来说,对细胞系和人鼻上皮具有良好的生物相容性,广谱活动,和缺乏抗病毒抗性的发展揭示了Ti2PW10作为治疗急性呼吸道病毒性疾病发展的抗病毒候选物的潜力,保证进一步的研究,以确定聚阴离子的具体目标,并评估其临床潜力。
    Human respiratory viruses have an enormous impact on national health systems, societies, and economy due to the rapid airborne transmission and epidemic spread of such pathogens, while effective specific antiviral drugs to counteract infections are still lacking. Here, we identified two Keggin-type polyoxometalates (POMs), [TiW11CoO40]8- (TiW11Co) and [Ti2PW10O40]7- (Ti2PW10), endowed with broad-spectrum activity against enveloped and non-enveloped human respiratory viruses, i.e., coronavirus (HCoV-OC43), rhinovirus (HRV-A1), respiratory syncytial virus (RSV-A2), and adenovirus (AdV-5). Ti2PW10 showed highly favorable selectivity indexes against all tested viruses (SIs >700), and its antiviral potential was further investigated against human coronaviruses and rhinoviruses. This POM was found to inhibit replication of multiple HCoV and HRV strains, in different cell systems. Ti2PW10 did not affect virus binding or intracellular viral replication, but selectively inhibited the viral entry. Serial passaging of virus in presence of the POM revealed a high barrier to development of Ti2PW10-resistant variants of HRV-A1 or HCoV-OC43. Moreover, Ti2PW10 was able to inhibit HRV-A1 production in a 3D model of the human nasal epithelium and, importantly, the antiviral treatment did not determine cytotoxicity or tissue damage. A mucoadhesive thermosensitive in situ hydrogel formulation for nasal delivery was also developed for Ti2PW10. Overall, good biocompatibility on cell lines and human nasal epithelia, broad-spectrum activity, and absence of antiviral resistance development reveal the potential of Ti2PW10 as an antiviral candidate for the development of a treatment of acute respiratory viral diseases, warranting further studies to identify the specific target/s of the polyanion and assess its clinical potential.
    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
    乙型肝炎病毒(HBV)引起的慢性肝病是公认的导致肝硬化的主要原因,肝纤维化,还有肝癌.牛磺胆酸钠共转运多肽(NTCP),触发HBV感染的肝细胞的特定膜受体,是一个有希望的目标,对HBV进入。在这项研究中,五环三萜类(PT),包括甘草次酸(GA),齐墩果酸(OA),分别通过与鬼臼毒素的分子杂交对熊果酸(UA)和桦木酸(BA)进行修饰,并产生了32个新的缀合物。在HepG2.2.15细胞中评估缀合物的抗HBV活性。结果表明,66%的结合物对宿主细胞具有较低的毒性,对两种HBV抗原有显著的抑制作用,尤其是HBsAg。值得注意的是,化合物BA-PPT1,BA-PPT3,BA-PPT4和UA-PPT3不仅抑制HBsAg的分泌,而且抑制HBVDNA复制。通过SPR测定观察到与HBVPreS1抗原相比的活性缀合物与NTCP的结合的显著差异。发现作用机制是这些化合物与NTCP157-165表位的竞争性结合,阻断HBV进入宿主细胞。分子对接结果表明,BA-PPT3主要通过π-阳离子与靶蛋白的氨基酸残基相互作用,氢键和疏水相互作用,提示其作为靶向NTCP受体的有前途的HBV进入抑制剂的潜力。
    Chronic liver diseases caused by hepatitis B virus (HBV) are the accepted main cause leading to liver cirrhosis, hepatic fibrosis, and hepatic carcinoma. Sodium taurocholate cotransporting polypeptide (NTCP), a specific membrane receptor of hepatocytes for triggering HBV infection, is a promising target against HBV entry. In this study, pentacyclic triterpenoids (PTs) including glycyrrhetinic acid (GA), oleanolic acid (OA), ursolic acid (UA) and betulinic acid (BA) were modified via molecular hybridization with podophyllotoxin respectively, and resulted in thirty-two novel conjugates. The anti-HBV activities of conjugates were evaluated in HepG2.2.15 cells. The results showed that 66% of the conjugates exhibited lower toxicity to the host cells and had significant inhibitory effects on the two HBV antigens, especially HBsAg. Notably, the compounds BA-PPT1, BA-PPT3, BA-PPT4, and UA-PPT3 not only inhibited the secretion of HBsAg but also suppressed HBV DNA replication. A significant difference in the binding of active conjugates to NTCP compared to the HBV PreS1 antigen was observed by SPR assays. The mechanism of action was found to be the competitive binding of these compounds to the NTCP 157-165 epitopes, blocking HBV entry into host cells. Molecular docking results indicated that BA-PPT3 interacted with the amino acid residues of the target protein mainly through π-cation, hydrogen bond and hydrophobic interaction, suggesting its potential as a promising HBV entry inhibitor targeting the NTCP receptor.
    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