viral entry

病毒进入
  • 文章类型: Journal Article
    狂犬病病毒在全球范围内每年估计造成59,000人死亡,有希望的治疗方法是必要的。在这项研究中,亲和标签纯化质谱用于描述RABV糖蛋白和宿主蛋白的相互作用,PDIA3/ERP57被确定为RABV感染的潜在抑制剂。PDIA3限制性RABV感染的机制如下:PDIA3通过选择性巨自噬/自噬途径靶向赖氨酸332介导RABVG蛋白降解;AP3B1(衔接子相关蛋白复合物3亚基β1)在PDIA3触发的G蛋白选择性降解中是不可缺少的;PDIA3与NCAM1/NCAM(神经细胞粘附分子1)竞争性结合以阻断RABVG,阻碍病毒进入宿主细胞。PDIA3与RABVG蛋白结合的190-199aa残基对于防御RABV是必需的且足以。这些结果证明了靶向PDIA3或利用PDIA3190-199aa肽治疗临床狂犬病的生物制剂的治疗潜力。
    Rabies virus causes an estimated 59,000 annual fatalities worldwide and promising therapeutic treatments are necessary to develop. In this study, affinity tag-purification mass spectrometry was employed to delineate RABV glycoprotein and host protein interactions, and PDIA3/ERP57 was identified as a potential inhibitor of RABV infection. PDIA3 restricted RABV infection with follow mechanisms: PDIA3 mediated the degradation of RABV G protein by targeting lysine 332 via the selective macroautophagy/autophagy pathway; The PDIA3 interactor, AP3B1 (adaptor related protein complex 3 subunit beta 1) was indispensable in PDIA3-triggered selective degradation of the G protein; Furthermore, PDIA3 competitively bound with NCAM1/NCAM (neural cell adhesion molecule 1) to block RABV G, hindering viral entry into host cells. PDIA3 190-199 aa residues bound to the RABV G protein were necessary and sufficient to defend against RABV. These results demonstrated the therapeutic potential of biologics that target PDIA3 or utilize PDIA3 190-199 aa peptide to treat clinical rabies.Abbreviation: aa: amino acids; ANXA2: annexin A2; AP-MS: affinity tag purification-mass spectrometry; AP3B1: adaptor related protein complex 3 subunit beta 1; ATP6V1A: ATPase H+ transporting V1 subunit A; ATP6V1H: ATPase H+ transporting V1 subunit H; BafA1: bafilomycin A1; CHX: cycloheximide; co-IP: co-immunoprecipitation; DDX17: DEAD-box helicase 17; DmERp60: drosophila melanogaster endoplasmic reticulum p60; EBOV: Zaire ebolavirus virus; EV: empty vector; GANAB: glucosidase II alpha subunit; G protein: glycoprotein; GRM2/mGluR2: glutamate metabotropic receptor 2; HsPDIA3: homo sapiens protein disulfide isomerase family A member 3; IAV: influenza virus; ILF2: interleukin enhancer binding factor 2; KO: knockout; MAGT1: magnesium transporter 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MmPDIA3: mus musculus protein disulfide isomerase associated 3; NCAM1/NCAM: neural cell adhesion molecule 1; NGFR/p75NTR: nerve growth factor receptor; NGLY1: N-glycanase 1; OTUD4: OTU deubiquitinase 4; PDI: protein disulfide isomerase; PPIs: protein-protein interactions; RABV: rabies virus; RUVBL2: RuvB like AAA ATPase 2; SCAMP3: secretory carrier membrane protein 3; ScPdi1: Saccharomyces cerevisiae s288c protein disulfide isomerase 1; SLC25A6: solute carrier family 25 member 6; SQSTM1/p62: sequestosome 1; VSV: vesicular stomatitis virus.
    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
    orf病毒(ORFV)对家庭小反刍动物的健康构成严重威胁(即,绵羊和山羊)和全球范围内的人类,每年给畜牧业造成约1.5亿美元的损失。然而,涉及ORFV感染和复制的宿主因素仍然难以捉摸。在这项研究中,我们比较了ORFV感染或未感染的绵羊睾丸间质细胞(STIC)的RNA-seq谱,并鉴定了一个新的宿主基因,钾电压门控通道亚家族E成员4(KCNE4),作为参与ORFV感染的关键宿主因子。RNA-seq数据和RT-qPCR测定均显示,在感染后(hpi)9至48小时,感染的STIC中KCNE4的表达显著增加。另一方面,RT-qPCR检测到ORFV感染后KCNE4siRNA和KCNE4敲除(KO)HeLa细胞转染的STIC中ORFV拷贝数减少,在24hpi时,KOHeLa细胞中ORFV-GFP的荧光比率降低,表明KCNE4对ORFV感染至关重要。此外,附着和内化分析显示ORFV附着减少,内化,复制,并由KOHeLa细胞释放,证明了KCNE4对ORFV进入细胞的潜在抑制作用。用KCNE4抑制剂如奎尼丁和氟西汀预处理显著抑制ORFV感染。我们所有的发现都揭示了KCNE4作为ORFV进入和复制的新型宿主调节因子,为ORFV感染的相互作用机制提供了新的见解。该研究还强调了K+通道可能是阻止病毒感染和疾病的药物靶标。
    The orf virus (ORFV) poses a serious threat to the health of domestic small ruminants (i.e., sheep and goats) and humans on a global scale, causing around $150 million in annual losses to livestock industry. However, the host factors involved in ORFV infection and replication are still elusive. In this study, we compared the RNA-seq profiles of ORFV-infected or non-infected sheep testicular interstitial cells (STICs) and identified a novel host gene, potassium voltage-gated channel subfamily E member 4 (KCNE4), as a key host factor involved in the ORFV infection. Both RNA-seq data and RT-qPCR assay revealed a significant increase in the expression of KCNE4 in the infected STICs from 9 to 48 h post infection (hpi). On the other hand, the RT-qPCR assay detected a decrease in ORFV copy number in both the STICs transfected by KCNE4 siRNA and the KCNE4 knockout (KO) HeLa cells after the ORFV infection, together with a reduced fluorescence ratio of ORFV-GFP in the KO HeLa cells at 24 hpi, indicating KCNE4 to be critical for the ORFV infection. Furthermore, the attachment and internalization assays showed decreased ORFV attachment, internalization, replication, and release by the KO HeLa cells, demonstrating a potential inhibition of ORFV entry into the cells by KCNE4. Pretreatment with the KCNE4 inhibitors such as quinidine and fluoxetine significantly repressed the ORFV infection. All our findings reveal KCNE4 as a novel host regulator of the ORFV entry and replication, shedding new insight into the interactive mechanism of ORFV infection. The study also highlights the K+ channels as possible druggable targets to impede viral infection and disease.
    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冠状病毒进入宿主细胞的机制之一涉及内体酸化。有人提出,在酸性条件下,SARS-CoV-2尖峰糖蛋白的融合肽近端区域(FPPR)充当pH依赖性开关,通过影响受体结合域(RBD)的定位来调节免疫应答可及性。这将提供RBD开口与环境pH的间接偶联。这里,我们探索了SARS-CoV-2刺突糖蛋白中FPPR的这种可能的pH依赖性构象平衡。我们分析了来自蛋白质数据库的数百个实验确定的尖峰结构,并进行了pH复制交换分子动力学,以探索FPPR构象取决于pH和RBD定位的程度。对实验结构的荟萃分析确定了FPPR在其中解决了该柔性区域的结构中的交替构象。然而,结果不支持FPPR构象与RBD位置或低温EM实验报告的pH之间的相关性.我们使用PDB结构计算了FPPR区可滴定侧链的pKa值,但是这些pKa值在其他采用相同FPPR构象类型的替代PDB结构之间显示出很大的差异。这妨碍了不同FPPR构象中pKa值的比较,以合理化pH依赖性构象变化。我们通过全原子模拟补充了这些基于PDB的分析,并使用恒定pH复制交换分子动力学来估计柔性和显式水的pKa值。所得的滴定曲线在模拟之间显示出良好的再现性,但他们也表明,不同FPPR构象的滴定曲线在误差棒内是相同的。总之,我们无法找到支持先前发表的FPPRpH依赖性平衡假说的证据:无论是现有的实验数据还是恒定pHMD模拟.该研究强调了尖峰系统的复杂性,并为进一步探索pH和SARS-CoV-2病毒进入机制之间的相互作用开辟了途径。
    One of the entry mechanisms of the SARS-CoV-2 coronavirus into host cells involves endosomal acidification. It has been proposed that under acidic conditions, the fusion peptide proximal region (FPPR) of the SARS-CoV-2 spike glycoprotein acts as a pH-dependent switch, modulating immune response accessibility by influencing the positioning of the receptor binding domain (RBD). This would provide indirect coupling of RBD opening to the environmental pH. Here, we explored this possible pH-dependent conformational equilibrium of the FPPR within the SARS-CoV-2 spike glycoprotein. We analyzed hundreds of experimentally determined spike structures from the Protein Data Bank and carried out pH-replica exchange molecular dynamics to explore the extent to which the FPPR conformation depends on pH and the positioning of the RBD. A meta-analysis of experimental structures identified alternate conformations of the FPPR among structures in which this flexible regions was resolved. However, the results did not support a correlation between the FPPR conformation and either RBD position or the reported pH of the cryo-EM experiment. We calculated pKa values for titratable side chains in the FPPR region using PDB structures, but these pKa values showed large differences between alternate PDB structures that otherwise adopt the same FPPR conformation type. This hampers the comparison of pKa values in different FPPR conformations to rationalize a pH-dependent conformational change. We supplemented these PDB-based analyses with all-atom simulations and used constant-pH replica exchange molecular dynamics to estimate pKa values in the context of flexibility and explicit water. The resulting titration curves show good reproducibility between simulations, but they also suggest that the titration curves of the different FPPR conformations are the same within the error bars. In summary, we were unable to find evidence supporting the previously published hypothesis of an FPPR pH-dependent equilibrium: neither from existing experimental data nor from constant-pH MD simulations. The study underscores the complexity of the spike system and opens avenues for further exploration into the interplay between pH and SARS-CoV-2 viral entry mechanisms.
    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:在寻找抗COVID-19疗法时,1,2,3,4,6-五-O-没食子酰-βD-吡喃葡萄糖苷,一种从许多传统草药中分离出来的天然多酚化合物,已被报道为RBD-ACE2结合抑制剂和靶向SARSCoV-2的主要蛋白酶和RNA依赖性RNA聚合酶的广谱抗冠状病毒抑制剂。为了促进1,2,3,4,6-五-O-没食子酰-β-D-吡喃葡萄糖苷的构效关系研究,我们描述了它的化学合成和表征,以及它对SARS-CoV-2与宿主ACE2受体相互作用的活性。
    方法:由3,4,5-三苄氧苯甲酸和β-D-吡喃葡萄糖苷通过两个定量步骤合成了1,2,3,4,6-五烷基-O-没食子酰基-β-D-吡喃葡萄糖苷:DCC介导的酯化和钯催化的去苄基化。使用SARS-CoV-2尖峰三聚体(S1S2)ACE2抑制剂筛选比色测定试剂盒评估合成的分子,SARS-CoV2尖峰S1RBDACE2抑制剂筛选比色测定试剂盒,和使用Spike(SARS-CoV-2)假型慢病毒的细胞中和测定法,ACE2-HEK293重组细胞系。
    结果:化学合成的产物阻断了SARSCoV-2的尖峰三聚体与人ACE2受体的结合,IC50=22±2µM。它还以IC50=27±3µM阻断ACE2:尖峰RBD结合。重要的是,它抑制了SARS2-CoV2-Spike假型化慢病毒对ACE2HEK293细胞系的感染性,IC50=20±2µM。
    结论:总体而言,化学合成的1,2,3,4,6-五-O-没食子酰-β-D-吡喃葡萄糖苷是开发抗SARS-CoV-2疗法的先导分子,该疗法通过阻断病毒进入宿主细胞来阻断病毒感染的初始阶段。
    BACKGROUND: In the search for anti-COVID-19 therapy, 1,2,3,4,6-pentakis-O-galloyl-βD-glucopyranoside, a natural polyphenolic compound isolated from many traditional medicinal herbs, has been reported as an RBD-ACE2 binding inhibitor and as a broad-spectrum anticoronaviral inhibitor targeting the main protease and RNA-dependent RNA polymerase of SARSCoV-2. To facilitate the structure-activity relationship studies of 1,2,3,4,6-pentakis-O-galloyl-β-Dglucopyranoside, we describe its chemical synthesis and characterization, as well as its activity towards the SARS-CoV-2 spike interaction with host ACE2 receptor.
    METHODS: 1,2,3,4,6-Pentakis-O-galloyl-β-D-glucopyranoside was synthesized in two quantitative steps from 3,4,5-tribenzyloxybenzoic acid and β-D-glucopyranoside: DCC-mediated esterification and palladium-catalyzed per-debenzylation. The synthesized molecule was evaluated using a SARS-CoV-2 spike trimer (S1 + S2) ACE2 inhibitor screening colorimetric assay kit, SARS-CoV2 spike S1 RBD ACE2 inhibitor screening colorimetric assay kit, and a cellular neutralization assay using the Spike (SARS-CoV-2) Pseudotyped Lentivirus, ACE2-HEK293 recombinant cell line.
    RESULTS: The chemically synthesized product blocked the binding of the spike trimer of SARSCoV-2 to the human ACE2 receptor with IC50=22±2 µM. It also blocked ACE2:spike RBD binding with IC50=27±3 µM. Importantly, it inhibited the infectivity of SARS2-CoV2-Spike pseudotyped lentivirus on the ACE2 HEK293 cell line with IC50=20±2 µM.
    CONCLUSIONS: Overall, the chemically synthesized 1,2,3,4,6-pentakis-O-galloyl-β-D-glucopyranoside represents a lead molecule to develop anti-SARS-CoV-2 therapies that block the initial stage of the viral infection by blocking the virus entry to the host cell.
    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
    已显示许多共受体促进SARS-CoV-2的hACE2依赖性或hACE2非依赖性感染。Yu等人最近发表在mBio上的一项研究。表明组胺受体H1(HRH1)通过直接结合病毒刺突蛋白(F.Yu,X.Liu,H.Ou,X.李,etal.,mBioe01088-24,2024,https://doi.org/10.1128/mbio.01088-24)。此外,他们提供了令人信服的证据,证明针对HRH1的抗组胺药物能有效抑制SARS-CoV-2进入.这项研究强调了可再利用的抗组胺药对COVID-19的治疗潜力。
    Numerous coreceptors have been shown to facilitate hACE2-dependent or hACE2-independent infection by SARS-CoV-2. A recent study published in mBio by Yu et al. showed that the histamine receptor H1 (HRH1) functions as an alternative receptor for SARS-CoV-2 via direct binding to viral spike proteins (F. Yu, X. Liu, H. Ou, X. Li, et al., mBio e01088-24, 2024, https://doi.org/10.1128/mbio.01088-24). Furthermore, they present compelling evidence that antihistamine drugs targeting HRH1 potently inhibit SARS-CoV-2 entry. This study highlights the therapeutic potential of repurposable antihistamines against COVID-19.
    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
    沙蝇传播的托斯卡纳病毒(TOSV)是一种有包膜的三节段负单链RNAPhlebovirus。它是一种新兴病毒,主要在欧洲西南部和北非流行。尽管TOSV感染通常无症状或导致轻度发热疾病,它具有神经毒性,在某些地区是夏季脑膜炎的三个最常见原因之一。尽管有这种临床意义,我们对调节静脉病毒感染的分子方面和宿主因素的理解是有限的。这项研究表征了TOSV感染的早期步骤。我们的发现揭示了Ser/Thr激酶的Numb相关激酶家族的两个成员,即衔接子相关激酶1(AAK1)和细胞周期蛋白G相关激酶(GAK),在调节TOSV进入的早期阶段发挥作用。FDA批准的靶向这些激酶的抑制剂表现出对TOSV感染的显著抑制。这项研究表明,AAK1和GAK代表了抑制TOSV感染的药物靶标,潜在的,相关的滴虫病毒。
    Sandfly-borne Toscana virus (TOSV) is an enveloped tri-segmented negative single-strand RNA Phlebovirus. It is an emerging virus predominantly endemic in southwestern Europe and Northern Africa. Although TOSV infection is typically asymptomatic or results in mild febrile disease, it is neurovirulent and ranks among the three most common causes of summer meningitis in certain regions. Despite this clinical significance, our understanding of the molecular aspects and host factors regulating phlebovirus infection is limited. This study characterized the early steps of TOSV infection. Our findings reveal that two members of the Numb-associated kinases family of Ser/Thr kinases, namely adaptor-associated kinase 1 (AAK1) and cyclin G-associated kinase (GAK), play a role in regulating the early stages of TOSV entry. FDA-approved inhibitors targeting these kinases demonstrated significant inhibition of TOSV infection. This study suggests that AAK1 and GAK represent druggable targets for inhibiting TOSV infection and, potentially, related Phleboviruses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由于人血管紧张素转换酶2(ACE2)是SARS-CoV-2的主要受体,因此表征允许SARS-CoV-2进入人细胞的ACE2区域对于设计基于肽的抗病毒阻断剂和阐明病毒的发病机理至关重要。我们鉴定并合成了涉及S1受体结合域(RBD)-ACE2界面的25聚体模拟肽(包含ACE2α1的22-46位)。模拟物(野生型,WT)ACE2肽在体外显着抑制SARS-CoV-2感染人肺Calu-3细胞。在图形蛋白建模中预测ACE2α-螺旋α1的残基F28,K31,F32,F40和Y41对原始蛋白至关重要,Delta,和SARS-CoV-2的Omicron菌株建立了SpikeRBD-ACE2界面。用丙氨酸(A)或天冬氨酸(D)取代这些残基消除了肽的抗病毒保护作用,表明这些位置对于病毒进入肺细胞至关重要。WTACE2肽,但不是A或D突变的肽,与SARS-CoV-2S1RBD表现出显著的相互作用,如分子动力学模拟所示。通过确定ACE2α-螺旋α1的关键氨基酸残基,这是SpikeRBD-ACE2界面所必需的,并且在细胞的体外病毒感染过程中动员,我们证明WTACE2肽可保护易感K18-hACE2小鼠免受体内SARS-CoV-2感染,并可有效治疗COVID-19。
    Since human angiotensin-converting enzyme 2 (ACE2) serves as a primary receptor for SARS-CoV-2, characterizing ACE2 regions that allow SARS-CoV-2 to enter human cells is essential for designing peptide-based antiviral blockers and elucidating the pathogenesis of the virus. We identified and synthesized a 25-mer mimetic peptide (encompassing positions 22-46 of the ACE2 alpha-helix α1) implicated in the S1 receptor-binding domain (RBD)-ACE2 interface. The mimetic (wild-type, WT) ACE2 peptide significantly inhibited SARS-CoV-2 infection of human pulmonary Calu-3 cells in vitro. In silico protein modeling predicted that residues F28, K31, F32, F40, and Y41 of the ACE2 alpha-helix α1 are critical for the original, Delta, and Omicron strains of SARS-CoV-2 to establish the Spike RBD-ACE2 interface. Substituting these residues with alanine (A) or aspartic acid (D) abrogated the antiviral protective effect of the peptides, indicating that these positions are critical for viral entry into pulmonary cells. WT ACE2 peptide, but not the A or D mutated peptides, exhibited significant interaction with the SARS-CoV-2 S1 RBD, as shown through molecular dynamics simulations. Through identifying the critical amino acid residues of the ACE2 alpha-helix α1, which is necessary for the Spike RBD-ACE2 interface and mobilized during the in vitro viral infection of cells, we demonstrated that the WT ACE2 peptide protects susceptible K18-hACE2 mice against in vivo SARS-CoV-2 infection and is effective for the treatment of 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
    冠状病毒的进入是由宿主细胞受体的尖峰识别启动的,涉及蛋白质和/或聚糖受体。最近,TMPRSS2被鉴定为HCoV-HKU1的蛋白质受体,与唾液酸聚糖作为聚糖受体一起。然而,病毒进入的潜在机制仍然未知.这里,我们调查了不活跃的HCoV-HKU1C峰值,聚糖激活,和功能锚定状态,揭示唾液酸聚糖结合诱导NTD的构象变化,并促进尖峰的相邻RBD打开以识别TMPRSS2,表现出HCoV-HKU1进入的协同机制。HCoV-HKU1的RBD具有通过三个先前未发现的接口识别TMPRSS2的插入子域。此外,HCoV-HKU1A的结构研究与诱变和结合测定相结合,证实了HCoV-HKU1采用的保守受体识别模式。这些研究促进了我们对进入过程中复杂的病毒与宿主相互作用的理解,为开发针对冠状病毒相关疾病的新疗法奠定基础。
    The entry of coronaviruses is initiated by spike recognition of host cellular receptors, involving proteinaceous and/or glycan receptors. Recently, TMPRSS2 was identified as the proteinaceous receptor for HCoV-HKU1 alongside sialoglycan as a glycan receptor. However, the underlying mechanisms for viral entry remain unknown. Here, we investigated the HCoV-HKU1C spike in the inactive, glycan-activated, and functionally anchored states, revealing that sialoglycan binding induces a conformational change of the NTD and promotes the neighboring RBD of the spike to open for TMPRSS2 recognition, exhibiting a synergistic mechanism for the entry of HCoV-HKU1. The RBD of HCoV-HKU1 features an insertion subdomain that recognizes TMPRSS2 through three previously undiscovered interfaces. Furthermore, structural investigation of HCoV-HKU1A in combination with mutagenesis and binding assays confirms a conserved receptor recognition pattern adopted by HCoV-HKU1. These studies advance our understanding of the complex viral-host interactions during entry, laying the groundwork for developing new therapeutics against coronavirus-associated diseases.
    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
    目的:牛磺胆酸钠共转运多肽(NTCP)已被确定为乙型肝炎病毒(HBV)的细胞受体。然而,表达NTCP的肝细胞对HBV感染表现出不同的敏感性。本研究旨在探讨其他宿主因素是否调节HBV感染的过程。
    方法:从乙型肝炎儿童获得的肝活检样本用于单细胞测序和易感性分析。原代人肝细胞,HepG2-NTCP细胞,和人肝嵌合小鼠用于分析候选宿主因素对HBV感染的影响。
    结果:单细胞测序和易感性分析显示神经纤毛蛋白-1(NRP1)表达与HBV感染之间呈正相关。在HBV感染的细胞模型中,NRP1过表达前HBV接种显着增强病毒的附着和内化,并在NTCP存在下促进病毒感染。机理研究表明NRP1与LHBs和NTCP形成复合物。NRP1b结构域介导其与HBV包膜蛋白LHBs的preS1结构域中位置88和92处的保守精氨酸残基的相互作用。这种NRP1-preS1相互作用随后促进了preS1与NTCP的结合,促进病毒感染。此外,NRP1拮抗剂EG00229破坏NRP1-preS1相互作用显着减弱NTCP和preS1之间的结合亲和力,从而在体外和体内抑制HBV感染。
    结论:我们的研究结果表明,NRP1是HBV感染的新型宿主因子,与preS1和NTCP相互作用以调节HBV进入肝细胞。
    HBV感染是一个全球性的公共卫生问题,但对HBV早期感染过程的认识仍然有限。通过单细胞测序,我们发现了一个新的宿主因子,NRP1,通过与HBVpreS1和NTCP相互作用来调节HBV进入。此外,针对NRP1的拮抗剂可以在体外和体内抑制HBV感染。本研究可进一步促进我们对HBV早期感染过程的理解。
    OBJECTIVE: Sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the cellular receptor for hepatitis B virus (HBV). However, hepatocytes expressing NTCP exhibit varying susceptibilities to HBV infection. This study aimed to investigate whether other host factors modulate the process of HBV infection.
    METHODS: Liver biopsy samples obtained from children with hepatitis B were used for single-cell sequencing and susceptibility analysis. Primary human hepatocytes, HepG2-NTCP cells, and human liver chimeric mice were used to analyze the effect of candidate host factors on HBV infection.
    RESULTS: Single-cell sequencing and susceptibility analysis revealed a positive correlation between neuropilin-1 (NRP1) expression and HBV infection. In the HBV-infected cell model, NRP1 overexpression before HBV inoculation significantly enhanced viral attachment and internalization, and promoted viral infection in the presence of NTCP. Mechanistic studies indicated that NRP1 formed a complex with LHBs and NTCP. The NRP1 b domain mediated its interaction with conserved arginine residues at positions 88 and 92 in the preS1 domain of the HBV envelope protein LHBs. This NRP1-preS1 interaction subsequently promoted the binding of preS1 to NTCP, facilitating viral infection. Moreover, disruption of the NRP1-preS1 interaction by the NRP1 antagonist EG00229 significantly attenuated the binding affinity between NTCP and preS1, thereby inhibiting HBV infection both in vitro and in vivo.
    CONCLUSIONS: Our findings indicate that NRP1 is a novel host factor for HBV infection, which interacts with preS1 and NTCP to modulate HBV entry into hepatocytes.
    UNASSIGNED: HBV infection is a global public health problem, but the understanding of the early infection process of HBV remains limited. Through single-cell sequencing, we identified a novel host factor, NRP1, which modulates HBV entry by interacting with HBV preS1 and NTCP. Moreover, antagonists targeting NRP1 can inhibit HBV infection both in vitro and in vivo. This study could further advance our comprehension of the early infection process of HBV.
    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
    许多宿主因素,除了人血管紧张素转换酶2(hACE2),已被确定为严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的共受体,表现出广泛的病毒嗜性和多样化的可药用潜力。我们和其他人发现抗组胺药,特别是组胺受体H1(HRH1)拮抗剂,有效抑制SARS-CoV-2感染。在这项研究中,我们提供了令人信服的证据,证明HRH1通过直接与病毒刺突蛋白结合而成为SARS-CoV-2的替代受体.HRH1还通过与hACE2相互作用协同增强hACE2依赖性病毒进入。抗组胺药物通过竞争性结合HRH1,从而破坏刺突蛋白与其受体之间的相互作用,从而有效地预防病毒感染。多种抑制试验表明,抗组胺药广泛抑制各种SARS-CoV-2突变体的感染,平均IC50为2.4µM。通过真正的SARS-CoV-2感染测定和人源化小鼠攻击实验进一步证实了这些药物的预防功能,证明抗组胺药对抗冠状病毒病的治疗潜力19.重要意义除了人血管紧张素转换酶2外,严重急性呼吸综合征冠状病毒2(SARS-CoV-2)还可以利用替代辅因子来促进病毒进入。在这项研究中,我们发现组胺受体H1(HRH1)不仅是SARS-CoV-2的独立受体,而且通过与ACE2直接相互作用,协同增强ACE2依赖性病毒的进入.进一步的研究表明,HRH1通过直接结合刺突蛋白的N端结构域来促进SARS-CoV-2的进入。相反,抗组胺药,主要是HRH1拮抗剂,可以竞争性地结合HRH1,从而防止病毒进入。这些发现表明,可再利用的抗组胺药物的给药可能是对抗冠状病毒疾病19的治疗性干预措施。
    Numerous host factors, in addition to human angiotensin-converting enzyme 2 (hACE2), have been identified as coreceptors of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), demonstrating broad viral tropism and diversified druggable potential. We and others have found that antihistamine drugs, particularly histamine receptor H1 (HRH1) antagonists, potently inhibit SARS-CoV-2 infection. In this study, we provided compelling evidence that HRH1 acts as an alternative receptor for SARS-CoV-2 by directly binding to the viral spike protein. HRH1 also synergistically enhanced hACE2-dependent viral entry by interacting with hACE2. Antihistamine drugs effectively prevent viral infection by competitively binding to HRH1, thereby disrupting the interaction between the spike protein and its receptor. Multiple inhibition assays revealed that antihistamine drugs broadly inhibited the infection of various SARS-CoV-2 mutants with an average IC50 of 2.4 µM. The prophylactic function of these drugs was further confirmed by authentic SARS-CoV-2 infection assays and humanized mouse challenge experiments, demonstrating the therapeutic potential of antihistamine drugs for combating coronavirus disease 19.IMPORTANCEIn addition to human angiotensin-converting enzyme 2, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can utilize alternative cofactors to facilitate viral entry. In this study, we discovered that histamine receptor H1 (HRH1) not only functions as an independent receptor for SARS-CoV-2 but also synergistically enhances ACE2-dependent viral entry by directly interacting with ACE2. Further studies have demonstrated that HRH1 facilitates the entry of SARS-CoV-2 by directly binding to the N-terminal domain of the spike protein. Conversely, antihistamine drugs, primarily HRH1 antagonists, can competitively bind to HRH1 and thereby prevent viral entry. These findings revealed that the administration of repurposable antihistamine drugs could be a therapeutic intervention to combat coronavirus disease 19.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号