endocytic trafficking

胞吞运输
  • 文章类型: Journal Article
    成纤维细胞生长因子(FGF)途径是胚胎发育所需的保守信号传导途径。激活的FGF受体1(FGFR1)驱动多个细胞内信号级联通路,包括ERK/MAPK和PI3K/AKT,统称为规范信令。然而,与Fgfr1空胚胎不同,Fgfr1中含有低态突变的胚胎缺乏激活经典下游信号的能力仍然能够发育到出生,但在所有中胚层来源的组织中表现出严重的缺陷。额外的信号突变的引入进一步降低了Fgfr1的活性,导致更早的致死性,减少的躯体发生,以及更严重的转录输出变化。涉及迁移的基因,ECM相互作用,和磷酸肌醇信号显著下调,蛋白质组学分析确定了与内吞途径成分相互作用的变化,和表达突变受体的细胞显示内吞运输的变化。一起,我们确定了通过涉及规范和非规范Fgfr1途径的机制调节早期中胚层发育的过程,包括与细胞粘附成分的直接相互作用和内吞调节。
    The fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1-null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identified processes regulating early mesoderm development by mechanisms involving both canonical and noncanonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    成纤维细胞生长因子(FGF)途径是胚胎发育所需的保守信号传导途径。激活的FGF受体1(FGFR1)驱动多个细胞内信号级联通路,包括ERK/MAPK和PI3K/AKT,统称为规范信令。然而,与Fgfr1空胚胎不同,Fgfr1中含有低态突变的胚胎缺乏激活经典下游信号的能力仍然能够发育到出生,但在所有中胚层来源的组织中表现出严重的缺陷。额外的信号突变的引入进一步降低了Fgfr1的活性,导致更早的致死性,减少的躯体发生,以及更严重的转录输出变化。涉及迁移的基因,ECM相互作用,和磷酸肌醇信号显著下调,蛋白质组学分析确定了与内吞途径成分相互作用的变化,和表达突变受体的细胞显示内吞运输的变化。一起,我们确定了通过涉及规范和非规范Fgfr1途径的机制调节早期中胚层发育的过程,包括与细胞粘附成分的直接相互作用和内吞调节。
    The Fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1 null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth, but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM-interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identify processes regulating early mesoderm development by mechanisms involving both canonical and non-canonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.
    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
    设计用于选择性靶向神经元的小分子探针的开发将增强对复杂神经元结构和功能的探索。在这些探测器中,NeuO作为先驱脱颖而出,并在研究领域获得了巨大的吸引力。然而,尚未确定神经元选择性背后的机制或细胞定位。这里,我们介绍NeuM,NeuO的衍生物,旨在靶向神经元细胞膜。此外,我们阐明了区分神经元的选择性神经元膜运输背后的机制。在水性缓冲液中,NeuM自主组装成胶束结构,导致其荧光猝灭(Φ=0.001)。暴露于神经元后,NeuM胶束通过网格蛋白介导的内吞作用选择性内化到神经元内体中。通过胞吞再循环途径,NeuM胶束整合到神经元膜中,将荧光NeuM分子分散在膜中(Φ=0.61)。分子动力学模拟表明,NeuM,与NeuO相比,具有最佳的亲脂性和分子长度,促进其稳定掺入磷脂层。NeuM在神经元膜内的稳定整合允许神经元的长期监测,以及复杂的神经元结构的可视化。
    The development of a small-molecule probe designed to selectively target neurons would enhance the exploration of intricate neuronal structures and functions. Among such probes, NeuO stands out as the pioneer and has gained significant traction in the field of research. Nevertheless, neither the mechanism behind neuron-selectivity nor the cellular localization has been determined. Here, we introduce NeuM, a derivative of NeuO, designed to target neuronal cell membranes. Furthermore, we elucidate the mechanism behind the selective neuronal membrane trafficking that distinguishes neurons. In an aqueous buffer, NeuM autonomously assembles into micellar structures, leading to the quenching of its fluorescence (Φ=0.001). Upon exposure to neurons, NeuM micelles were selectively internalized into neuronal endosomes via clathrin-mediated endocytosis. Through the endocytic recycling pathway, NeuM micelles integrate into neuronal membrane, dispersing fluorescent NeuM molecules in the membrane (Φ=0.61). Molecular dynamics simulations demonstrated that NeuM, in comparison to NeuO, possesses optimal lipophilicity and molecular length, facilitating its stable incorporation into phospholipid layers. The stable integration of NeuM within neuronal membrane allows the prolonged monitoring of neurons, as well as the visualization of intricate neuronal structures.
    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
    在顶端丛,内吞货物(例如,血红蛋白)被贩运到专门的细胞器进行消化。这遵循这些寄生虫中的微孔/细胞基质的独特内吞过程。然而,内吞贩运的潜在机制仍然难以捉摸,由于经典的内吞蛋白用于根尖细胞器的生物发生。要解决此问题,我们已经利用了弓形虫尖丛模型的遗传易懂,摄取宿主细胞溶质物质(例如,绿色荧光蛋白[GFP])。我们确定了蛋白质戊烯化和内吞运输之间的关联,使用炔烃标记的点击化学方法,异戊二烯化蛋白质组的特征。基因组编辑,使用成簇的规则间隔短回文repaet/CRISPR相关核酸酶9(CRISPR/Cas9),被有效地用于产生用于23个异戊烯化候选物的功能筛选的基因修饰系。这鉴定了调节内吞GFP囊泡运输的这些蛋白质中的四种。在这些蛋白质中,Rab1B和YKT6.1是高度保守的,但在真核生物中是非经典的内吞蛋白。共聚焦成像分析表明,Rab1B和Ras基本上位于寄生虫的跨高尔基体网络和内体样区室中。Rab1B的条件性敲除导致了向跳楼球的分泌贩运的快速缺陷,建议关键监管机构Rab1B的贩运交叉作用。进一步的实验证实了蛋白质异戊二烯化在调节这些蛋白质的稳定性/活性方面的关键作用(即,Rab1B和YKT6.1)在寄生虫中。我们的发现定义了内吞运输的分子基础,并揭示了Rab1B对弓形虫膜运输的潜在交叉功能。这可能会延伸到其他相关的原生生物,包括疟疾寄生虫.重要性原生动物弓形虫建立了一个允许的生态位,在宿主细胞中,这使得寄生虫能够获得蛋白质等大分子。大量研究表明,寄生虫将经典的内吞成分重新用于分泌分选到顶端细胞器,不清楚内吞转运到溶酶体样隔室的问题。最近的研究表明,内吞运输可能与疟疾寄生虫中的蛋白质异戊二烯化有关。这些信息促使我们在刚地尖丛模型中检查这种关联,并确定所涉及的异戊二烯化蛋白质组的关键成分。通过利用弓形虫的遗传可操作性和宿主GFP采集试验,我们揭示了四种调节内吞货物运输的非经典内吞蛋白(例如,GFP)在弓形虫中。因此,我们扩展了蛋白质戊烯化调节胞吞运输的原理,并阐明了弓形虫和其他潜在相关原生生物的非经典胞吞过程。
    In the apicomplexans, endocytosed cargos (e.g., hemoglobin) are trafficked to a specialized organelle for digestion. This follows a unique endocytotic process at the micropore/cytostome in these parasites. However, the mechanism underlying endocytic trafficking remains elusive, due to the repurposing of classical endocytic proteins for the biogenesis of apical organelles. To resolve this issue, we have exploited the genetic tractability of the model apicomplexan Toxoplasma gondii, which ingests host cytosolic materials (e.g., green fluorescent protein[GFP]). We determined an association between protein prenylation and endocytic trafficking, and using an alkyne-labeled click chemistry approach, the prenylated proteome was characterized. Genome editing, using clustered regularly interspaced short palindromic repaet/CRISPR-associated nuclease 9 (CRISPR/Cas9), was efficiently utilized to generate genetically modified lines for the functional screening of 23 prenylated candidates. This identified four of these proteins that regulate the trafficking of endocytosed GFP vesicles. Among these proteins, Rab1B and YKT6.1 are highly conserved but are non-classical endocytic proteins in eukaryotes. Confocal imaging analysis showed that Rab1B and Ras are substantially localized to both the trans-Golgi network and the endosome-like compartments in the parasite. Conditional knockdown of Rab1B caused a rapid defect in secretory trafficking to the rhoptry bulb, suggesting a trafficking intersection role for the key regulator Rab1B. Further experiments confirmed a critical role for protein prenylation in regulating the stability/activity of these proteins (i.e., Rab1B and YKT6.1) in the parasite. Our findings define the molecular basis of endocytic trafficking and reveal a potential intersection function of Rab1B on membrane trafficking in T. gondii. This might extend to other related protists, including the malarial parasites. IMPORTANCE The protozoan Toxoplasma gondii establishes a permissive niche, in host cells, that allows parasites to acquire large molecules such as proteins. Numerous studies have demonstrated that the parasite repurposes the classical endocytic components for secretory sorting to the apical organelles, leaving the question of endocytic transport to the lysosome-like compartment unclear. Recent studies indicated that endocytic trafficking is likely to associate with protein prenylation in malarial parasites. This information promoted us to examine this association in the model apicomplexan T. gondii and to identify the key components of the prenylated proteome that are involved. By exploiting the genetic tractability of T. gondii and a host GFP acquisition assay, we reveal four non-classical endocytic proteins that regulate the transport of endocytosed cargos (e.g., GFP) in T. gondii. Thus, we extend the principle that protein prenylation regulates endocytic trafficking and elucidate the process of non-classical endocytosis in T. gondii and potentially in other related protists.
    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
    包膜病毒进入宿主细胞需要病毒和宿主细胞膜的融合,从病毒包膜突出的病毒融合蛋白促进的过程。这些病毒融合蛋白需要由宿主因子触发,对于一些病毒,此事件发生在内体和/或溶酶体内。因此,这些“晚期穿透病毒”必须被内化并递送至有助于进入的细胞内囊泡。因为胞吞作用和囊泡运输是紧密调节的细胞过程,晚期穿透病毒还依赖于特定的宿主蛋白来有效递送到融合位点,这表明这些可以作为抗病毒治疗的目标。在这项研究中,我们研究了鞘氨醇激酶(SKs)在病毒进入中的作用,发现鞘氨醇激酶1(SK1)和/或SK2的化学抑制和SK1/2的敲减抑制了埃博拉病毒(EBOV)进入宿主细胞.机械上,抑制SK1/2阻止EBOV到达含有EBOV受体的晚期内体和溶酶体,尼曼选择C1(NPC1)。此外,我们提供的证据表明,由SK1/2抑制引起的运输缺陷独立于通过细胞表面S1P受体的鞘氨醇-1-磷酸(S1P)信号传导而发生.最后,我们发现SK1/2的化学抑制可以防止其他晚期穿透病毒的进入,包括沙粒病毒和冠状病毒,并抑制Huh7.5细胞中具有复制能力的EBOV和SARS-CoV-2的感染。总之,我们的结果强调了SK1/2在内吞运输中的重要作用,它可以靶向抑制晚期穿透病毒的进入,并可以作为开发广谱抗病毒疗法的起点。
    Entry of enveloped viruses in host cells requires the fusion of viral and host cell membranes, a process that is facilitated by viral fusion proteins protruding from the viral envelope. These viral fusion proteins need to be triggered by host factors, and for some viruses, this event occurs inside endosomes and/or lysosomes. Consequently, these \'late-penetrating viruses\' must be internalized and delivered to entry-conducive intracellular vesicles. Because endocytosis and vesicular trafficking are tightly regulated cellular processes, late-penetrating viruses also depend on specific host proteins for efficient delivery to the site of fusion, suggesting that these could be targeted for antiviral therapy. In this study, we investigated a role for sphingosine kinases (SKs) in viral entry and found that chemical inhibition of sphingosine kinase 1 (SK1) and/or SK2 and knockdown of SK1/2 inhibited entry of Ebola virus (EBOV) into host cells. Mechanistically, inhibition of SK1/2 prevented EBOV from reaching late-endosomes and lysosomes that contain the EBOV receptor, Niemann Pick C1 (NPC1). Furthermore, we present evidence that suggests that the trafficking defect caused by SK1/2 inhibition occurs independently of sphingosine-1-phosphate (S1P) signaling through cell-surface S1P receptors. Lastly, we found that chemical inhibition of SK1/2 prevents entry of other late-penetrating viruses, including arenaviruses and coronaviruses, and inhibits infection by replication-competent EBOV and SARS-CoV-2 in Huh7.5 cells. In sum, our results highlight an important role played by SK1/2 in endocytic trafficking, which can be targeted to inhibit entry of late-penetrating viruses and could serve as a starting point for the development of broad-spectrum antiviral therapeutics.
    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
    小GTP酶RhoB与其他Rho蛋白的区别在于其在内体中的独特亚细胞定位,多囊尸体,和核。尽管与RhoA和RhoC具有很高的序列同源性,RhoB主要与肿瘤抑制功能有关,而RhoA和RhoC支持大多数恶性肿瘤的致癌转化。RhoB调节信号分子的内吞运输和细胞骨架重塑,从而控制增长,凋亡,应激反应,免疫功能,和细胞运动在各种情况下。这些功能中的一些可能归因于RhoB独特的内吞区室亚细胞定位。在这里,我们描述了RhoB在亚细胞定位的背景下在癌症抑制中的多效性作用,我们讨论了可能的治疗途径,以追求和强调未来研究的重点。
    The small GTPase RhoB is distinguished from other Rho proteins by its unique subcellular localization in endosomes, multivesicular bodies, and nucleus. Despite high sequence homology with RhoA and RhoC, RhoB is mainly associated with tumor suppressive function, while RhoA and RhoC support oncogenic transformation in most malignancies. RhoB regulates the endocytic trafficking of signaling molecules and cytoskeleton remodeling, thereby controlling growth, apoptosis, stress response, immune function, and cell motility in various contexts. Some of these functions may be ascribed to RhoB\'s unique subcellular localization to endocytic compartments. Here we describe the pleiotropic roles of RhoB in cancer suppression in the context of its subcellular localization, and we discuss possible therapeutic avenues to pursue and highlight priorities for future research.
    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
    果蝇的许多器官表现出刻板的左右(LR)不对称;然而,潜在的机制仍然难以捉摸。这里,我们确定了一种进化保守的泛素结合蛋白,AWP1/医生编号(Drn),作为胚胎前肠LR不对称所需的新因素。我们发现drn在中肠的环状内脏肌细胞中对于JAK/STAT信号传导是必需的,这有助于通过LR不对称核重排导致前肠侧化的第一个已知线索。drn纯合且缺乏母体贡献的胚胎显示出与JAK/STAT信号耗尽的表型相似,这表明Drn是JAK/STAT信号的一般组成部分。没有Drn导致了Domeless(Dome)的特定积累,JAK/STAT信号的受体,在细胞内区室,包括泛素化的货物。圆顶在野生型果蝇中与Drn共定位。这些结果表明,Drn是圆顶的内吞运输所必需的,这是激活JAK/STAT信号和随后的圆顶降解的关键步骤。AWP1/Drn在激活JAK/STAT信号传导和LR不对称发育中的作用在各种生物体中可能是保守的。
    Many organs of Drosophila show stereotypical left-right (LR) asymmetry; however, the underlying mechanisms remain elusive. Here, we have identified an evolutionarily conserved ubiquitin-binding protein, AWP1/Doctor No (Drn), as a factor required for LR asymmetry in the embryonic anterior gut. We found that drn is essential in the circular visceral muscle cells of the midgut for JAK/STAT signaling, which contributes to the first known cue for anterior gut lateralization via LR asymmetric nuclear rearrangement. Embryos homozygous for drn and lacking its maternal contribution showed phenotypes similar to those with depleted JAK/STAT signaling, suggesting that Drn is a general component of JAK/STAT signaling. Absence of Drn resulted in specific accumulation of Domeless (Dome), the receptor for ligands in the JAK/STAT signaling pathway, in intracellular compartments, including ubiquitylated cargos. Dome colocalized with Drn in wild-type Drosophila. These results suggest that Drn is required for the endocytic trafficking of Dome, which is a crucial step for activation of JAK/STAT signaling and the subsequent degradation of Dome. The roles of AWP1/Drn in activating JAK/STAT signaling and in LR asymmetric development may be conserved in various organisms.
    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
    植物可以改变它们的身体结构,比如他们的根架构,胚胎后。例如,拟南芥可以发育侧根作为内源程序的一部分或响应生物和非生物刺激。根周轮细胞被指定为侧根创始人细胞,启动侧根器官发生。我们使用内吞运输诱导剂Sortin2来检查内膜运输在侧根建立细胞规格中的作用。我们的结果表明,Sortin2刺激开启了侧根原基形成的从头程序,该程序不同于生长素驱动的内源性程序。在这种独特的机制中,AUX/IAA28领导的生长素模块上游的侧根建立细胞规范需要细胞外钙摄取和向液泡的内吞运输。生长素依赖性TIR1/AFBF盒和生长素极性运输对于依赖于内吞运输的侧根建立细胞规范是不必要的;但是,需要一组不同的F-box蛋白和功能性SCF复合物。胞吞运输可以构成响应环境条件的器官发生的方便策略。
    Plants can modify their body structure, such as their root architecture, post-embryonically. For example, Arabidopsis thaliana can develop lateral roots as part of an endogenous program or in response to biotic and abiotic stimuli. Root pericycle cells are specified to become lateral root founder cells, initiating lateral root organogenesis. We used the endocytic trafficking inducer Sortin2 to examine the role of endomembrane trafficking in lateral root founder cell specification. Our results indicate that Sortin2 stimulation turns on a de novo program of lateral root primordium formation that is distinct from the endogenous program driven by auxin. In this distinctive mechanism, extracellular calcium uptake and endocytic trafficking toward the vacuole are required for lateral root founder cell specification upstream of the auxin module led by AUX/IAA28. The auxin-dependent TIR1/AFB F-boxes and auxin polar transport are dispensable for the endocytic trafficking-dependent lateral root founder cell specification; however, a different set of F-box proteins and a functional SCF complex are required. The endocytic trafficking could constitute a convenient strategy for organogenesis in response to environmental 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
    严重急性呼吸综合征相关冠状病毒2(SARS-CoV-2)和SARS-CoV-1辅助蛋白Orf3a与质膜标记物共同定位,胞吞途径,和高尔基体。一些报道已经导致将两种Orf3a蛋白注释为病毒体。在这里,我们表明SARS-CoV-2和SARS-CoV-1Orf3a均未形成功能性离子导电孔,并且测得的电导率是过表达中的常见污染物,并且在重建研究中具有高水平的蛋白质。SARS-CoV-2和SARS-CoV-1Orf3a的低温EM结构都显示出狭窄的收缩和带正电荷的水性前庭的存在,这不利于阳离子渗透。我们观察到SARS-CoV-2Orf3a过表达后晚期内体标记Rab7的富集,并与VPS39共免疫沉淀。有趣的是,SARS-CoV-1Orf3a不引起与SARS-CoV-2Orf3a相同的细胞表型,并且不与VPS39相互作用。为了解释这种差异,我们发现一个分歧,SARS-CoV-2Orf3a的非结构化环促进其与VPS39的结合,VPS39是一种HOPS复合物连接蛋白,参与晚期内体和自噬体与溶酶体的融合。我们建议添加的环增强SARS-CoV-2Orf3a的能力,以共同选择宿主细胞运输机制以进行病毒退出或宿主免疫逃避。
    The severe acute respiratory syndrome associated coronavirus 2 (SARS-CoV-2) and SARS-CoV-1 accessory protein Orf3a colocalizes with markers of the plasma membrane, endocytic pathway, and Golgi apparatus. Some reports have led to annotation of both Orf3a proteins as viroporins. Here, we show that neither SARS-CoV-2 nor SARS-CoV-1 Orf3a form functional ion conducting pores and that the conductances measured are common contaminants in overexpression and with high levels of protein in reconstitution studies. Cryo-EM structures of both SARS-CoV-2 and SARS-CoV-1 Orf3a display a narrow constriction and the presence of a positively charged aqueous vestibule, which would not favor cation permeation. We observe enrichment of the late endosomal marker Rab7 upon SARS-CoV-2 Orf3a overexpression, and co-immunoprecipitation with VPS39. Interestingly, SARS-CoV-1 Orf3a does not cause the same cellular phenotype as SARS-CoV-2 Orf3a and does not interact with VPS39. To explain this difference, we find that a divergent, unstructured loop of SARS-CoV-2 Orf3a facilitates its binding with VPS39, a HOPS complex tethering protein involved in late endosome and autophagosome fusion with lysosomes. We suggest that the added loop enhances SARS-CoV-2 Orf3a\'s ability to co-opt host cellular trafficking mechanisms for viral exit or host immune evasion.
    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
    李斯特菌溶血素O(LLO)介导的快速吞噬体逃逸是单核细胞增生李斯特菌细胞内复制和发病的先决条件。在从对早期内体Rab5GTP酶呈阴性且对晚期内体Rab7呈阳性的空泡内化后几分钟内发生逃逸。使用突变分析,我们发现单核细胞增生李斯特菌的最佳细胞内增殖需要李斯特菌侵入素InlB。从这个观察开始,我们在HeLa细胞中确定InlB促进含李斯特菌液泡(LCV)的早期吞噬体逃逸和有效的Rab7获取。向LCV募集III类磷酸肌醇3激酶(PI3K)Vps34并积累其脂质产物,磷脂酰肌醇3-磷酸酯(PI3P),两个关键的内体成熟介质,也依赖于InlB。小干扰RNA(siRNA)敲低实验表明,Rab7募集和早期(LLO介导的)逃逸需要Vps34,并支持InlB依赖性细胞内增殖。一起,我们的数据表明,InlB通过破坏III类PI3K/Vps34信号传导加速LCV转化为逃逸有利的Rab7晚期吞噬体。我们的发现揭示了InlBinvasin在李斯特菌发病机制中作为细胞内促进增殖的毒力因子的新功能。重要性通过操纵内体区室避免溶酶体杀死是一种被认为主要限于针内细胞内病原体的毒力机制。我们的发现很重要,因为它们表明细胞溶质病原体,如单核细胞增生李斯特菌,内化后迅速逃离吞噬体,作为生存策略的一部分,也可以广泛颠覆内吞贩运。他们还澄清说,而不是延迟吞噬体成熟(为LLO依赖性破坏留出时间,正如目前所认为的),通过InlBL.单核细胞增多性细胞似乎有助于吞噬液泡快速转化为有利于逃逸的晚期吞噬体。我们的数据强调了细菌毒力因子的多功能性。在细胞表面,InlBinvasin通过I类PI3K激活诱导受体介导的吞噬作用,而在内化后,它利用III类PI3K(Vsp34)来促进细胞内存活。系统地阐明李斯特菌在整个胞吞途径中干扰PI3K信号传导的机制可能会导致新的抗感染疗法。
    Rapid phagosomal escape mediated by listeriolysin O (LLO) is a prerequisite for Listeria monocytogenes intracellular replication and pathogenesis. Escape takes place within minutes after internalization from vacuoles that are negative to the early endosomal Rab5 GTPase and positive to the late endosomal Rab7. Using mutant analysis, we found that the listerial invasin InlB was required for optimal intracellular proliferation of L. monocytogenes. Starting from this observation, we determined in HeLa cells that InlB promotes early phagosomal escape and efficient Rab7 acquisition by the Listeria-containing vacuole (LCV). Recruitment of the class III phosphoinositide 3-kinase (PI3K) Vps34 to the LCV and accumulation of its lipid product, phosphatidylinositol 3-phosphate (PI3P), two key endosomal maturation mediators, were also dependent on InlB. Small interfering RNA (siRNA) knockdown experiments showed that Vps34 was required for Rab7 recruitment and early (LLO-mediated) escape and supported InlB-dependent intracellular proliferation. Together, our data indicate that InlB accelerates LCV conversion into an escape-favorable Rab7 late phagosome via subversion of class III PI3K/Vps34 signaling. Our findings uncover a new function for the InlB invasin in Listeria pathogenesis as an intracellular proliferation-promoting virulence factor. IMPORTANCE Avoidance of lysosomal killing by manipulation of the endosomal compartment is a virulence mechanism assumed to be largely restricted to intravacuolar intracellular pathogens. Our findings are important because they show that cytosolic pathogens like L. monocytogenes, which rapidly escape the phagosome after internalization, can also extensively subvert endocytic trafficking as part of their survival strategy. They also clarify that, instead of delaying phagosome maturation (to allow time for LLO-dependent disruption, as currently thought), via InlB L. monocytogenes appears to facilitate the rapid conversion of the phagocytic vacuole into an escape-conducive late phagosome. Our data highlight the multifunctionality of bacterial virulence factors. At the cell surface, the InlB invasin induces receptor-mediated phagocytosis via class I PI3K activation, whereas after internalization it exploits class III PI3K (Vsp34) to promote intracellular survival. Systematically elucidating the mechanisms by which Listeria interferes with PI3K signaling all along the endocytic pathway may lead to novel anti-infective therapies.
    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