capsid

衣壳
  • 文章类型: Journal Article
    腺相关病毒(AAV)是基础科学和临床治疗的基础基因传递工具。然而,缺乏机械洞察力,特别是对于由定向进化产生的工程载体,会妨碍其应用。这里,我们采用无偏的人体细胞微阵列平台来确定天然和工程化AAV的细胞外和细胞表面相互作用。我们确定了AAV9衣壳和人白细胞介素3(IL3)之间的自然进化和血清型特异性相互作用,在宿主免疫调节中可能发挥作用,以及实验室进化的低密度脂蛋白受体相关蛋白6(LRP6)相互作用,该相互作用对工程化衣壳具有增强的静脉内给药后非人灵长类动物的血脑屏障穿越。无偏倚的细胞微阵列筛选方法还使我们能够鉴定工程化的富含脑的AAV衣壳的脱靶组织结合相互作用,这些相互作用可以告知载体周围器官嗜性和副作用。我们的衣壳相互作用复合物的低温电子层析成像和AlphaFold建模揭示了LRP6和IL3结合位点。这些结果允许工程AAV在不同生物体中的自信应用,并解锁用于非侵入性治疗递送到大脑的改进的病毒和非病毒载体的未来靶标告知工程。
    Adeno-associated viruses (AAVs) are foundational gene delivery tools for basic science and clinical therapeutics. However, lack of mechanistic insight, especially for engineered vectors created by directed evolution, can hamper their application. Here, we adapt an unbiased human cell microarray platform to determine the extracellular and cell surface interactomes of natural and engineered AAVs. We identify a naturally-evolved and serotype-specific interaction between the AAV9 capsid and human interleukin 3 (IL3), with possible roles in host immune modulation, as well as lab-evolved low-density lipoprotein receptor-related protein 6 (LRP6) interactions specific to engineered capsids with enhanced blood-brain barrier crossing in non-human primates after intravenous administration. The unbiased cell microarray screening approach also allows us to identify off-target tissue binding interactions of engineered brain-enriched AAV capsids that may inform vectors\' peripheral organ tropism and side effects. Our cryo-electron tomography and AlphaFold modeling of capsid-interactor complexes reveal LRP6 and IL3 binding sites. These results allow confident application of engineered AAVs in diverse organisms and unlock future target-informed engineering of improved viral and non-viral vectors for non-invasive therapeutic delivery to the brain.
    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
    实验室环境中的腺相关病毒(AAV)载体的产生通常涉及在贴壁细胞中表达,然后通过密度梯度中的超速离心进行纯化。这种生产方法是,然而,不容易扩展,呈现与病毒共纯化的高水平细胞杂质,并导致空衣壳和满衣壳的混合物。在这里,我们描述了详细的AAV生产方案,通过AAV在悬浮细胞中表达,然后通过AAV亲和纯化和AAV抛光以分离空衣壳和完整衣壳来克服这些限制,导致在全衣壳中高度富集的超纯AAV的高产率。
    The production of Adeno-associated virus (AAV) vectors in the lab setting has typically involved expression in adherent cells followed by purification through ultracentrifugation in density gradients. This production method is, however, not easily scalable, presents high levels of cellular impurities that co-purify with the virus, and results in a mixture of empty and full capsids. Here we describe a detailed AAV production protocol that overcomes these limitations through AAV expression in suspension cells followed by AAV affinity purification and AAV polishing to separate empty and full capsids, resulting in high yields of ultra-pure AAV that is highly enriched in full capsids.
    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
    重组腺相关病毒(rAAV)载体是基因治疗中最有前途的病毒载体之一。然而,人胚肾(HEK)细胞的制造能力有限是rAAV商业化的障碍。我们研究了内质网(ER)蛋白加工和凋亡基因对HEK293细胞中瞬时rAAV产生的影响。我们根据先前的转录组学研究选择了四个候选基因:XBP1,GADD34/PPP1R15A,HSPA6和BCL2。这些基因稳定整合到HEK293宿主细胞中。传统的三质粒瞬时转染用于评估载体生产能力以及过表达的稳定池和亲本细胞的质量。我们表明,在HEK293T细胞中,XBP1,HSPA6和GADD34的过表达使rAAV生产率增加高达100%,并使rAAV的比生产率增加高达78%。此外,当亲本细胞生产力高时,观察到与ER蛋白加工基因过表达相关的更显著的改善,但是在低产量条件下没有检测到实质性的变化。我们还证实了不同血清型(AAV2和AAV8)和不同细胞系(HEK293T和HEK293)的基因组滴度提高;然而,改善的程度可能有所不同。这项研究揭示了ER蛋白加工途径在病毒颗粒合成中的重要性,衣壳组件,和矢量生产。关键点:•内质网(ER)蛋白加工(XBP1,HSPA6和GADD34)的上调导致rAAV生产力最大100%的增加,并且在HEK293T细胞中特定rAAV生产力最大78%的提高•可以在不同的HEK293细胞系中验证生产力的提高,并可用于生产各种AAV血清型,尽管增强的程度可能略有不同•当亲本细胞生产力高时,观察到与过表达ER蛋白加工基因相关的更明显的改善。在低产量条件下注意到最小的变化。
    The recombinant adeno-associated virus (rAAV) vector is among the most promising viral vectors in gene therapy. However, the limited manufacturing capacity in human embryonic kidney (HEK) cells is a barrier to rAAV commercialization. We investigated the impact of endoplasmic reticulum (ER) protein processing and apoptotic genes on transient rAAV production in HEK293 cells. We selected four candidate genes based on prior transcriptomic studies: XBP1, GADD34 / PPP1R15A, HSPA6, and BCL2. These genes were stably integrated into HEK293 host cells. Traditional triple-plasmid transient transfection was used to assess the vector production capability and the quality of both the overexpressed stable pools and the parental cells. We show that the overexpression of XBP1, HSPA6, and GADD34 increases rAAV productivity by up to 100% and increases specific rAAV productivity by up to 78% in HEK293T cells. Additionally, more prominent improvement associated with ER protein processing gene overexpression was observed when parental cell productivity was high, but no substantial variation was detected under low-producing conditions. We also confirmed genome titer improvement across different serotypes (AAV2 and AAV8) and different cell lines (HEK293T and HEK293); however, the extent of improvement may vary. This study unveiled the importance of ER protein processing pathways in viral particle synthesis, capsid assembly, and vector production. KEY POINTS: • Upregulation of endoplasmic reticulum (ER) protein processing (XBP1, HSPA6, and GADD34) leads to a maximum 100% increase in rAAV productivity and a maximum 78% boost in specific rAAV productivity in HEK293T cells • The enhancement in productivity can be validated across different HEK293 cell lines and can be used for the production of various AAV serotypes, although the extent of the enhancement might vary slightly • The more pronounced improvements linked to overexpressing ER protein processing genes were observed when parental cell productivity was high, with minimal variation noted under low-producing conditions.
    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
    Bufavirus(BuV)是原病毒属的细小病毒科的成员。它们没有被包裹,T=1从显示急性腹泻的患者中分离的二十面体ssDNA病毒。缺乏治疗选择和对其疾病机制的有限理解需要在分子和结构水平上研究这些病毒。在本研究中,我们利用聚糖阵列和细胞结合测定来证明BuV1衣壳结合末端唾液酸(SIA)聚糖。此外,使用低温电子显微镜(cryo-EM),显示SIA结合在衣壳表面的2/5倍壁上。有趣的是,稳定SIA结合的衣壳残基在迄今为止鉴定的所有人BuV中是保守的。此外,生物物理分析说明了BuV1衣壳在内溶酶体(pH7.4-pH4)运输过程中的稳定和衣壳在pH3及以下的不稳定,对应于胃的pH值。因此,我们确定了BuV1衣壳在pH7.4、4.0和2.6至2.8的低温EM结构,3.2bias,和2.7贝达,分别。这些结构揭示了在内溶酶体逃逸过程中的衣壳结构重排,并为该过程提供了潜在的机制。从这项研究中获得的结构见解将增加人类致病性细小病毒的一般知识。此外,BuV中保守的SIA受体结合位点的鉴定为设计小分子提供了可能的可靶向表面可接近的口袋,这些小分子被开发为这些病毒的抗病毒药物。
    Bufaviruses (BuV) are members of the Parvoviridae of the Protoparvovirus genus. They are non-enveloped, T = 1 icosahedral ssDNA viruses isolated from patients exhibiting acute diarrhea. The lack of treatment options and a limited understanding of their disease mechanisms require studying these viruses on a molecular and structural level. In the present study, we utilize glycan arrays and cell binding assays to demonstrate that BuV1 capsid binds terminal sialic acid (SIA) glycans. Furthermore, using cryo-electron microscopy (cryo-EM), SIA is shown to bind on the 2/5-fold wall of the capsid surface. Interestingly, the capsid residues stabilizing SIA binding are conserved in all human BuVs identified to date. Additionally, biophysical assays illustrate BuV1 capsid stabilization during endo-lysosomal (pH 7.4-pH 4) trafficking and capsid destabilization at pH 3 and less, which correspond to the pH of the stomach. Hence, we determined the cryo-EM structures of BuV1 capsids at pH 7.4, 4.0, and 2.6 to 2.8 Å, 3.2 Å, and 2.7 Å, respectively. These structures reveal capsid structural rearrangements during endo-lysosomal escape and provide a potential mechanism for this process. The structural insights gained from this study will add to the general knowledge of human pathogenic parvoviruses. Furthermore, the identification of the conserved SIA receptor binding site among BuVs provides a possible targetable surface-accessible pocket for the design of small molecules to be developed as anti-virals for these viruses.
    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大流行导致全球病毒基因组监测的规模扩大。然而,潮湿实验室的限制(经济,基础设施,和人员)在许多资源有限的环境中,将新病毒变体序列信息翻译为有意义的免疫学和结构见解,这些见解对于开发具有广泛作用的对策(特别是对于新兴和重新出现的病毒)仍然是一个挑战。这里,我们描述了一个结合废水监测的工作流程,高通量测序,系统发育学,免疫信息学,和病毒衣壳结构建模,用于对废水中鉴定的未培养的小核糖核酸病毒序列进行基因型到血清型表征。具体来说,我们分析了犬小核糖核酸病毒(CanPV),它们是引起犬科全身性感染的未培养且尚未分配的Picornaviridae家族成员。我们分析了2019年10月至2020年3月以及2020年10月至2021年3月期间美国西南部约70万人的118个存档(储存在-20°C)废水(WW)样本。按月将样品汇集成12个两升体积,使用450nm膜过滤器分配(过滤捕获的固体[FTSs]和滤液),随后使用10,000DaMW截止离心过滤器浓缩至2mL(1000X)。对24种浓缩物进行RNA提取,CanPV完整衣壳单重叠群RT-PCR,Illumina测序,系统发育学,免疫信息学,和结构预测。我们在58.3%(14/24)的样品中检测到CanPV,产生了13,824,046个修剪的Illumina读数和27个CanPV重叠群。系统发育和配对同一性分析显示,8个CanPV基因型(基因型差异<14%)属于四个集群,内部发散<20%。相似性分析,免疫信息学,病毒原粒和衣壳结构预测表明,这四个簇可能是不同的血清学类型,预测的簇区分性B细胞表位聚集在围绕5倍对称轴的峡谷的北部和南部边缘。我们的方法允许通过耦合系统发育来对未培养的小核糖核酸病毒序列进行基因型到血清型表征,免疫信息学,和病毒衣壳结构预测。因此,这绕过了一个主要的潮湿实验室相关瓶颈,从而使资源有限的环境从废水来源的基因组数据跃升为制定预防和其他缓解措施所需的有价值的免疫学见解。
    The SARS-CoV-2 pandemic resulted in a scale-up of viral genomic surveillance globally. However, the wet lab constraints (economic, infrastructural, and personnel) of translating novel virus variant sequence information to meaningful immunological and structural insights that are valuable for the development of broadly acting countermeasures (especially for emerging and re-emerging viruses) remain a challenge in many resource-limited settings. Here, we describe a workflow that couples wastewater surveillance, high-throughput sequencing, phylogenetics, immuno-informatics, and virus capsid structure modeling for the genotype-to-serotype characterization of uncultivated picornavirus sequences identified in wastewater. Specifically, we analyzed canine picornaviruses (CanPVs), which are uncultivated and yet-to-be-assigned members of the family Picornaviridae that cause systemic infections in canines. We analyzed 118 archived (stored at -20 °C) wastewater (WW) samples representing a population of ~700,000 persons in southwest USA between October 2019 to March 2020 and October 2020 to March 2021. Samples were pooled into 12 two-liter volumes by month, partitioned (into filter-trapped solids [FTSs] and filtrates) using 450 nm membrane filters, and subsequently concentrated to 2 mL (1000×) using 10,000 Da MW cutoff centrifugal filters. The 24 concentrates were subjected to RNA extraction, CanPV complete capsid single-contig RT-PCR, Illumina sequencing, phylogenetics, immuno-informatics, and structure prediction. We detected CanPVs in 58.3% (14/24) of the samples generated 13,824,046 trimmed Illumina reads and 27 CanPV contigs. Phylogenetic and pairwise identity analyses showed eight CanPV genotypes (intragenotype divergence <14%) belonging to four clusters, with intracluster divergence of <20%. Similarity analysis, immuno-informatics, and virus protomer and capsid structure prediction suggested that the four clusters were likely distinct serological types, with predicted cluster-distinguishing B-cell epitopes clustered in the northern and southern rims of the canyon surrounding the 5-fold axis of symmetry. Our approach allows forgenotype-to-serotype characterization of uncultivated picornavirus sequences by coupling phylogenetics, immuno-informatics, and virus capsid structure prediction. This consequently bypasses a major wet lab-associated bottleneck, thereby allowing resource-limited settings to leapfrog from wastewater-sourced genomic data to valuable immunological insights necessary for the development of prophylaxis and other mitigation measures.
    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
    活动调节的细胞骨架相关蛋白(Arc/Arg3.1)是一种即时早期基因,在学习和记忆中起着至关重要的作用。Arc蛋白具有与病毒组特异性抗原(Gag)蛋白相似的结构和功能特性,并通过病毒样衣壳介导细胞间RNA转移。然而,电弧衣壳操纵货物的调节因子和分泌途径尚不清楚。这里,我们发现磷脂酰肌醇-3-磷酸(PI3P)通过内体-多囊泡体(MVB)通路介导电弧衣壳的组装和分泌.的确,重建的Arc蛋白优选结合PI3P。在HEK293T细胞中,电弧形成与FYVE共同定位的标点符号,内体PI3P标记,以及Rab5和CD63,早期内体和MVB标记,分别。超分辨率成像解决了电弧在MVB的腔内囊泡内的积累。RalA和RalB的CRISPR双敲除,MVB生物发生和胞吐的关键GTP酶,严重降低了Arc介导的RNA转移效率。培养的大鼠皮质神经元中的RalA/B双敲除增加了成熟树突棘的百分比。摄取从表达Arc的野生型纯化的细胞外囊泡,但不是RalA/B双击倒,小鼠皮质神经元中的细胞降低其表面GlutA1水平。这些结果表明,与HIVGag不同,其膜靶向需要与质膜特异性磷脂酰肌醇(4,5)二磷酸(PI(4,5)P2)相互作用,电弧衣壳的组装是由PI3P在胞吞膜上介导的。了解Arc的分泌途径有助于深入了解其在细胞间货物转移中的作用,并强调结构相似的衣壳蛋白之间的运输机制的共性和区别。
    Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene that plays a vital role in learning and memory. Arc protein has structural and functional properties similar to viral Group-specific antigen (Gag) protein and mediates the intercellular RNA transfer through virus-like capsids. However, the regulators and secretion pathway through which Arc capsids maneuver cargos are unclear. Here, we identified that phosphatidylinositol-3-phosphate (PI3P) mediates Arc capsid assembly and secretion through the endosomal-multivesicular body (MVB) pathway. Indeed, reconstituted Arc protein preferably binds to PI3P. In HEK293T cells, Arc forms puncta that colocalize with FYVE, an endosomal PI3P marker, as well as Rab5 and CD63, early endosomal and MVB markers, respectively. Superresolution imaging resolves Arc accumulates within the intraluminal vesicles of MVB. CRISPR double knockout of RalA and RalB, crucial GTPases for MVB biogenesis and exocytosis, severely reduces the Arc-mediated RNA transfer efficiency. RalA/B double knockdown in cultured rat cortical neurons increases the percentage of mature dendritic spines. Intake of extracellular vesicles purified from Arc-expressing wild-type, but not RalA/B double knockdown, cells in mouse cortical neurons reduces their surface GlutA1 levels. These results suggest that unlike the HIV Gag, whose membrane targeting requires interaction with plasma-membrane-specific phosphatidyl inositol (4,5) bisphosphate (PI(4,5)P2), the assembly of Arc capsids is mediated by PI3P at endocytic membranes. Understanding Arc\'s secretion pathway helps gain insights into its role in intercellular cargo transfer and highlights the commonality and distinction of trafficking mechanisms between structurally resembled capsid proteins.
    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
    对于许多病毒来说,他们的衣壳组装由两个步骤组成。第一步是将病毒结构蛋白单体聚合成结构单元。然后,这些构建块是累积的,并有效地组装成病毒衣壳。第一步中的这些结构单元聚合反应是病毒组装的基础,在这一步中发现了一些药物靶标。在这项工作中,我们专注于第一步。通常,病毒结构单元由少于六个单体组成。也就是说,二聚体,三聚体,四聚体,五聚体,和hexamer。我们提出了这五个结构单元的聚合化学反应的数学模型,分别。然后,我们逐一证明了这些数学模型正平衡解的存在性和唯一性。随后,我们还分析了平衡态的稳定性,分别。这些结果可以提供对体内病毒结构单元聚合化学反应的性质的进一步了解。
    For numerous viruses, their capsid assembly is composed of two steps. The first step is that virus structural protein monomers are polymerized to building blocks. Then, these building blocks are cumulative and efficiently assembled to virus capsid shell. These building block polymerization reactions in the first step are fundamental for virus assembly, and some drug targets were found in this step. In this work, we focused on the first step. Often, virus building blocks consisted of less than six monomers. That is, dimer, trimer, tetramer, pentamer, and hexamer. We presented mathematical models for polymerization chemical reactions of these five building blocks, respectively. Then, we proved the existence and uniqueness of the positive equilibrium solution for these mathematical models one by one. Subsequently, we also analyzed the stability of the equilibrium states, respectively. These results may provide further insight into property of virus building block polymerization chemical reactions in vivo.
    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)感染仍然是一个全球性的公共卫生问题。在2016年2月宣布“国际关注的突发公共卫生事件”之后,该病原体在许多地区的新感染发病率一直在下降。然而,ZIKV仍有可能传播到更多国家的风险。迄今为止,目前尚无疫苗或抗病毒药物可用于预防或治疗寨卡病毒感染。在寨卡疫苗开发中,那些基于蛋白质亚基的细胞作为不可复制的平台是有吸引力的,因为它们具有潜在的增强的安全性,可以在所有人群中使用.然而,这些疫苗通常需要多剂量和佐剂来实现保护性免疫。在这项研究中,我们展示了重组蛋白ZEC的新制剂的免疫学评估,它结合了包膜的结构域III的区域和来自ZIKV的衣壳。使用两种基于核苷酸的佐剂来增强由疫苗候选物ZEC引起的免疫。将ODN39M或c-di-AMP作为免疫调节剂掺入与氢氧化铝组合的制剂中。在免疫活性BALB/c小鼠中进行免疫接种后,该制剂刺激高IgG抗体。尽管IgG亚型表明包含ODN39M的制剂主要有Th1偏向性免疫应答,通过两种免疫调节剂诱导体外刺激后从脾细胞分泌IFNγ测量的细胞免疫应答。这些结果证明了两种免疫调节剂增强重组亚基ZEC作为针对ZIKV的疫苗候选物的免疫原性的能力。
    Zika virus (ZIKV) infection remains a global public health problem. After the \"Public Health Emergencies of International Concern\" declared in February 2016, the incidence of new infections by this pathogen has been decreasing in many areas. However, there is still a likely risk that ZIKV will spread to more countries. To date, there is no vaccine or antiviral drug available to prevent or treat Zika virus infection. In the Zika vaccine development, those based on protein subunits are attractive as a non-replicable platform due to their potentially enhanced safety profile to be used in all populations. However, these vaccines frequently require multiple doses and adjuvants to achieve protective immunity. In this study we show the immunological evaluation of new formulations of the recombinant protein ZEC, which combines regions of domain III of the envelope and the capsid from ZIKV. Two nucleotide-based adjuvants were used to enhance the immunity elicited by the vaccine candidate ZEC. ODN 39M or c-di-AMP was incorporated as immunomodulator into the formulations combined with aluminum hydroxide. Following immunizations in immunocompetent BALB/c mice, the formulations stimulated high IgG antibodies. Although the IgG subtypes suggested a predominantly Th1-biased immune response by the formulation including the ODN 39M, cellular immune responses measured by IFNγ secretion from spleen cells after in vitro stimulations were induced by both immunomodulators. These results demonstrate the capacity of both immunomodulators to enhance the immunogenicity of the recombinant subunit ZEC as a vaccine candidate against ZIKV.
    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:局部基因治疗,包括体内基因组编辑,对皮肤遗传疾病的治疗寄予厚望,尤其是表皮.虽然腺相关病毒(AAV)是体内基因传递的有效载体,缺乏有效的基因传递方法限制了其临床应用。
    目的:优化AAV基因传递系统,使其具有更高的基因传递效率和对表皮和角质形成细胞(KCs)的特异性,使用AAV衣壳和启动子工程技术。
    方法:通过AAVDJ的定点诱变产生在残基中具有突变的AAV变体,据报道所述残基对于确定AAV2对KC的向性是关键的。将这些变体的KC的感染效率和特异性与先前报道的被认为适合于在体外和体内将基因递送至KC的AAV的那些进行比较。此外,我们使用最新的短核心启动子产生了表皮特异性启动子,并将其特异性与现有启动子进行了比较.
    结果:一种称为AAVDJK2的新型AAVDJ变体衣壳在体外和体内对表皮和KCs的基因转导效率和特异性方面优于现有的AAV。一种新的组织特异性启动子,称为K14SCP3启动子,在基因转导效率和对KCs的特异性方面优于现有启动子。
    结论:AAVDJK2衣壳和K14SCP3启动子的组合改善了体内向表皮和体外向KCs的基因递送。新的AAV系统可能有利于新的表皮靶向基因治疗的实验研究和开发。
    BACKGROUND: Local gene therapies, including in vivo genome editing, are highly anticipated for the treatment of genetic diseases in skin, especially the epidermis. While the adeno-associated virus (AAV) is a potent vector for in vivo gene delivery, the lack of efficient gene delivery methods has limited its clinical applications.
    OBJECTIVE: To optimize the AAV gene delivery system with higher gene delivery efficiency and specificity for epidermis and keratinocytes (KCs), using AAV capsid and promoter engineering technologies.
    METHODS: AAV variants with mutations in residues reported to be critical to determine the tropism of AAV2 for KCs were generated by site-directed mutagenesis of AAVDJ. The infection efficiency and specificity for KCs of these variants were compared with those of previously reported AAVs considered to be suitable for gene delivery to KCs in vitro and in vivo. Additionally, we generated an epidermis-specific promoter using the most recent short-core promoter and compared its specificity with existing promoters.
    RESULTS: A novel AAVDJ variant capsid termed AAVDJK2 was superior to the existing AAVs in terms of gene transduction efficiency and specificity for epidermis and KCs in vitro and in vivo. A novel tissue-specific promoter, termed the K14 SCP3 promoter, was superior to the existing promoters in terms of gene transduction efficiency and specificity for KCs.
    CONCLUSIONS: The combination of the AAVDJK2 capsid and K14 SCP3 promoter improves gene delivery to epidermis in vivo and KCs in vitro. The novel AAV system may benefit experimental research and development of new epidermis-targeted gene therapies.
    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
    丝氨酸蛋白酶是肠道病毒生物防治的重要环境贡献者。然而,解释肠道病毒对蛋白酶敏感性的分子相互作用的结构特征仍然无法解释。这里,我们描述了丝氨酸蛋白酶募集到病毒衣壳的分子机制。在使用的病毒类型中,柯萨奇病毒A9(CVA9),但不是CVB5和回声病毒11(E11),被枯草杆菌蛋白酶A以不依赖宿主的方式灭活,而牛胰胰蛋白酶(BPT)仅以宿主依赖性方式降低CVA9感染性。每种蛋白酶与衣壳前体的预测性相互作用模型表明主要靶标为暴露在5倍顶点(DE环VP1)或5/2倍交叉点(C末端VP1)上的内部无序蛋白(IDP)片段病毒衣壳。我们进一步表明,功能性结合蛋白酶/衣壳取决于蛋白酶-VP1复合物的强度和随时间的演变,最后是蛋白酶对周围病毒区域的局部适应。最后,我们预测CVA9衣壳上的三个残基会触发枯草杆菌蛋白酶A的裂解,其中之一可以作为传感器残基,有助于DE环路上的酶识别。总的来说,本研究描述了肠道病毒生物防治的重要生物学机制。
    Serine proteases are important environmental contributors of enterovirus biocontrol. However, the structural features of molecular interaction accounting for the susceptibility of enteroviruses to proteases remains unexplained. Here, we describe the molecular mechanisms involved in the recruitment of serine proteases to viral capsids. Among the virus types used, coxsackievirus A9 (CVA9), but not CVB5 and echovirus 11 (E11), was inactivated by Subtilisin A in a host-independent manner, while Bovine Pancreatic Trypsin (BPT) only reduced CVA9 infectivity in a host-dependent manner. Predictive interaction models of each protease with capsid protomers indicate the main targets as internal disordered protein (IDP) segments exposed either on the 5-fold vertex (DE loop VP1) or at the 5/2-fold intersection (C-terminal end VP1) of viral capsids. We further show that a functional binding protease/capsid depends on both the strength and the evolution over time of protease-VP1 complexes, and lastly on the local adaptation of proteases on surrounding viral regions. Finally, we predicted three residues on CVA9 capsid that trigger cleavage by Subtilisin A, one of which may act as a sensor residue contributing to enzyme recognition on the DE loop. Overall, this study describes an important biological mechanism involved in enteroviruses biocontrol.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号