Phagosome maturation

吞噬体成熟
  • 文章类型: Journal Article
    对吞噬细胞对摄入细菌的灭活过程的理解是宿主-病原体相互作用领域的关键焦点。Dictyostelium是一种模式生物,一直处于揭示这种相互作用机制的最前沿。在这项研究中,我们描述了一种旨在测量盘基网虱吞噬体中产气克雷伯菌失活的方法。
    The understanding of the inactivation process of ingested bacteria by phagocytes is a key focus in the field of host-pathogen interactions. Dictyostelium is a model organism that has been at the forefront of uncovering the mechanisms underlying this type of interaction. In this study, we describe an assay designed to measure the inactivation of Klebsiella aerogenes in the phagosomes of Dictyostelium discoideum.
    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
    小胶质细胞是专门的巨噬细胞,负责通过吞噬和降解清除死亡的神经元和病原体。降解需要由囊泡型或液泡型H转运腺苷三磷酸酶(V-ATPase)提供的吞噬体成熟和酸化,其由细胞质V1结构域和膜嵌入的Vo结构域组成。V-ATPasea亚基,Vo域的一个组成部分,哺乳动物有四种亚型。不同同工型对不同细胞/物种中吞噬体成熟的功能仍存在争议。在这里,我们表明V-ATPaseAtp6v0a1和Tcirg1b/Atp6v0a3亚基的突变导致斑马鱼小胶质细胞中吞噬体的积累。然而,他们的机制是不同的。V-ATPaseAtp6v0a1亚基主要分布在早期和晚期吞噬体中。该亚基的缺陷导致从早期吞噬小体到晚期吞噬小体的缺陷转变。相比之下,V-ATP酶Tcirg1b/Atp6v0a3亚基主要位于溶酶体上并调节晚期吞噬体-溶酶体融合。有缺陷的Tcirg1b/Atp6v0a3,而不是Atp6v0a1亚基导致小胶质细胞中酸化减少和巨自噬/自噬受损。我们进一步表明,小鼠巨噬细胞中的ATP6V0A1/a1和TCIRG1/a3亚基优先位于内体和溶酶体中,分别。阻断这些亚基破坏了早期到晚期的内体转变和内体到溶酶体的融合,分别。一起来看,我们的结果强调了不同的V-ATPase亚基在不同物种的吞噬作用和内吞作用的多个步骤中发挥的重要和保守作用.
    Microglia are specialized macrophages responsible for the clearance of dead neurons and pathogens by phagocytosis and degradation. The degradation requires phagosome maturation and acidification provided by the vesicular- or vacuolar-type H+-translocating adenosine triphosphatase (V-ATPase), which is composed of the cytoplasmic V1 domain and the membrane-embedded Vo domain. The V-ATPase a subunit, an integral part of the Vo domain, has four isoforms in mammals. The functions of different isoforms on phagosome maturation in different cells/species remain controversial. Here we show that mutations of both the V-ATPase Atp6v0a1 and Tcirg1b/Atp6v0a3 subunits lead to the accumulation of phagosomes in zebrafish microglia. However, their mechanisms are different. The V-ATPase Atp6v0a1 subunit is mainly distributed in early and late phagosomes. Defects of this subunit lead to a defective transition from early phagosomes to late phagosomes. In contrast, The V-ATPase Tcirg1b/Atp6v0a3 subunit is primarily located on lysosomes and regulates late phagosome-lysosomal fusion. Defective Tcirg1b/Atp6v0a3, but not Atp6v0a1 subunit leads to reduced acidification and impaired macroautophagy/autophagy in microglia. We further showed that ATP6V0A1/a1 and TCIRG1/a3 subunits in mouse macrophages preferentially located in endosomes and lysosomes, respectively. Blocking these subunits disrupted early-to-late endosome transition and endosome-to-lysosome fusion, respectively. Taken together, our results highlight the essential and conserved roles played by different V-ATPase subunits in multiple steps of phagocytosis and endocytosis across various species.Abbrevations: Apoe: apolipoprotein E; ANXA5/annexin V: annexin A5; ATP6V0A1/a1: ATPase H+-transporting V0 subunit a1; ATP6V0A2/a2: ATPase H+-transporting V0 subunit a2; ATP6V0A4/a4: ATPase H+-transporting V0 subunit a4; dpf: days post-fertilization; EEA1: early endosome antigen 1; HOPS: homotypic fusion and protein sorting; LAMP1: lysosomal associated membrane protein 1; Lcp1: lymphocyte cytosolic protein 1 (L-plastin); Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; NR: neutral red; PBS: phosphate-buffered saline; PtdIns: phosphatidylinositol; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns(3,5)P2: phosphatidylinositol (3,5)-bisphosphate; RAB4: RAB4, member RAS oncogene family; RAB5: RAB5, member RAS oncogene family; RAB7: RAB7, member RAS oncogene family; TCIRG1/Atp6v0a3/a3: T cell immune regulator 1, ATPase H+-transporting V0 subunit a3; V-ATPase: vacuolar-type H+-translocating adenosine triphosphatase; Xla.Tubb2b/NBT: tubulin beta 2B class IIb.
    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
    人类鼻病毒是慢性呼吸道疾病严重加重期间最常见的病毒,比如慢性阻塞性肺病.在这种疾病中,肺泡巨噬细胞显示显著降低的吞噬功能,这可能与细菌超感染有关。然而,人类鼻病毒如何影响巨噬细胞的功能在很大程度上是未知的。用HRV16处理的巨噬细胞表现出缺乏的细菌杀伤活性,吞噬溶酶体生物发生受损,并改变了细胞内区室。使用RNA测序,我们确定了在原代人巨噬细胞中由病毒上调的小GTP酶ARL5b.重要的是,ARL5b的消耗挽救了HRV16暴露后巨噬细胞中细菌清除和内体标志物的定位。在许可细胞中,ARL5b的消耗增加了HRV16病毒粒子的分泌。因此,我们确定ARL5b是巨噬细胞细胞内运输动力学和吞噬溶酶体生物发生的新型调节因子,也是允许细胞中HRV16的限制因子.
    Human rhinovirus is the most frequently isolated virus during severe exacerbations of chronic respiratory diseases, like chronic obstructive pulmonary disease. In this disease, alveolar macrophages display significantly diminished phagocytic functions that could be associated with bacterial superinfections. However, how human rhinovirus affects the functions of macrophages is largely unknown. Macrophages treated with HRV16 demonstrate deficient bacteria-killing activity, impaired phagolysosome biogenesis, and altered intracellular compartments. Using RNA sequencing, we identify the small GTPase ARL5b to be upregulated by the virus in primary human macrophages. Importantly, depletion of ARL5b rescues bacterial clearance and localization of endosomal markers in macrophages upon HRV16 exposure. In permissive cells, depletion of ARL5b increases the secretion of HRV16 virions. Thus, we identify ARL5b as a novel regulator of intracellular trafficking dynamics and phagolysosomal biogenesis in macrophages and as a restriction factor of HRV16 in permissive cells.
    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
    吞噬作用是感染控制和组织稳态的关键免疫功能。在吞噬作用期间,病原体在吞噬溶酶体中内化和降解。对于逃避免疫降解的病原体,普遍的观点是,需要毒力因子来破坏吞噬溶酶体的生物发生。相比之下,我们在这里提出,在细胞进入过程中,来自活动病原体的物理力将它们从典型的降解途径转移开。这种改变的命运始于力量诱导的吞噬细胞突触形成的重塑。我们使用寄生虫弓形虫作为模型,因为活的弓形虫利用滑翔运动积极侵入宿主细胞。为了区分物理力和毒力因子在吞噬作用中的作用,我们利用磁力诱导灭活弓形虫推进进入巨噬细胞。实验和计算机模拟表明,巨大的推进力通过防止受体在吞噬突触处与磷酸酶的空间分离来阻碍受体的生产性激活。因此,灭活的寄生虫被吞没在空泡中,这些空泡无法成熟为降解单位,类似于活活寄生虫的细胞内途径。使用酵母细胞和调理珠,我们确认这个机制是通用的,不是特定于使用的寄生虫。这些结果揭示了免疫逃避的新方面,展示了活动细胞进入过程中的物理力,独立于毒力因子,使病原体能够规避吞噬溶酶体降解。
    Phagocytosis is a critical immune function for infection control and tissue homeostasis. During phagocytosis, pathogens are internalized and degraded in phagolysosomes. For pathogens that evade immune degradation, the prevailing view is that virulence factors are required to disrupt the biogenesis of phagolysosomes. In contrast, we present here that physical forces from motile pathogens during cell entry divert them away from the canonical degradative pathway. This altered fate begins with the force-induced remodeling of the phagocytic synapse formation. We used the parasite Toxoplasma gondii as a model because live Toxoplasma actively invades host cells using gliding motility. To differentiate the effects of physical forces from virulence factors in phagocytosis, we employed magnetic forces to induce propulsive entry of inactivated Toxoplasma into macrophages. Experiments and computer simulations show that large propulsive forces hinder productive activation of receptors by preventing their spatial segregation from phosphatases at the phagocytic synapse. Consequently, the inactivated parasites are engulfed into vacuoles that fail to mature into degradative units, similar to the live motile parasite\'s intracellular pathway. Using yeast cells and opsonized beads, we confirmed that this mechanism is general, not specific to the parasite used. These results reveal new aspects of immune evasion by demonstrating how physical forces during active cell entry, independent of virulence factors, enable pathogens to circumvent phagolysosomal degradation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    [这修正了文章DOI:10.3389/fcimb.203.1220089。].
    [This corrects the article DOI: 10.3389/fcimb.2023.1220089.].
    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
    细胞致死膨胀毒素(Cdt)是由几种人类病原体产生的毒素家族,它们感染皮肤粘膜组织并诱发炎性疾病。暴露于Aggregatibacter放线菌(Aa)Cdt的人巨噬细胞通过规范和非规范的炎性体激活来刺激细胞因子释放。炎症反应依赖于通过毒素磷脂酰肌醇-3,4,5-三磷酸(PIP3)磷酸酶活性的PI3K信号传导阻断;将PIP3转化为磷脂酰肌醇-3,4-二磷酸(PI3,4P2),从而耗尽PIP3库。磷酸肌醇,在吞噬体贩运中也起着关键作用,在吞噬体成熟和随后与溶酶体融合期间充当效应蛋白的结合域。我们现在证明AaCdt操纵吞噬体膜的磷酸肌醇(PI)池并改变Rab5缔合。巨噬细胞暴露于AaCdt减慢了吞噬体的成熟并减少了phago-溶酶体的形成,从而损害巨噬细胞的吞噬功能。此外,暴露于Cdt的巨噬细胞显示出降低的杀菌能力,导致Aggregatibacter放线菌的存活率增加。因此,Cdt可能有助于增加对细菌感染的易感性。这些研究揭示了Cdt功能的未充分开发方面,并为Cdt介导由Cdt产生的生物(如Aa)引起的疾病的发病机理提供了新的认识。
    Cytolethal distending toxins (Cdt) are a family of toxins produced by several human pathogens which infect mucocutaneous tissue and induce inflammatory disease. Human macrophages exposed to Aggregatibacter actinomycetemcomitans (Aa) Cdt respond through canonical and non-canonical inflammasome activation to stimulate cytokine release. The inflammatory response is dependent on PI3K signaling blockade via the toxin\'s phosphatidylinositol-3,4,5-triphosphate (PIP3) phosphatase activity; converting PIP3 to phosphatidylinsoitol-3,4-diphosphate (PI3,4P2) thereby depleting PIP3 pools. Phosphoinositides, also play a critical role in phagosome trafficking, serving as binding domains for effector proteins during phagosome maturation and subsequent fusion with lysosomes. We now demonstrate that AaCdt manipulates the phosphoinositide (PI) pools of phagosome membranes and alters Rab5 association. Exposure of macrophages to AaCdt slowed phagosome maturation and decreased phago-lysosome formation, thereby compromising macrophage phagocytic function. Moreover, macrophages exposed to Cdt showed decreased bactericidal capacity leading to increase in Aggregatibacter actinomycetemcomitans survival. Thus, Cdt may contribute to increased susceptibility to bacterial infection. These studies uncover an underexplored aspect of Cdt function and provide new insight into the virulence potential of Cdt in mediating the pathogenesis of disease caused by Cdt-producing organisms such as Aa.
    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
    线虫秀丽隐杆线虫为研究保守的吞噬和吞噬清除机制提供了许多实验优势。这些包括用于延时成像的体内吞噬事件的定型时间,参与不同吞噬作用步骤的转基因报告基因标记分子的可用性,和动物的透明度用于荧光成像。Further,秀丽隐杆线虫的正向和反向遗传学的简易性使得许多与吞噬清除有关的蛋白质的初步发现成为可能。在这一章中,我们专注于线虫胚胎的大的未分化卵裂球的吞噬作用,吞噬并消除了从第二极体尸体到细胞动力学中体残留物的多种吞噬货物。我们描述了使用荧光延时成像来观察吞噬清除的不同步骤,以及将该过程标准化以区分突变菌株缺陷的方法。这些方法使我们能够从最初的信号传导中揭示新的见解,以诱导吞噬直至吞噬溶酶体中吞噬货物的最终解决。
    The nematode Caenorhabditis elegans offers many experimental advantages to study conserved mechanisms of phagocytosis and phagocytic clearance. These include the stereotyped timing of phagocytic events in vivo for time-lapse imaging, the availability of transgenic reporters labeling molecules involved in different steps of phagocytosis, and the transparency of the animal for fluorescence imaging. Further, the ease of forward and reverse genetics in C. elegans has enabled many of the initial discoveries of proteins involved in phagocytic clearance. In this chapter, we focus on phagocytosis by the large undifferentiated blastomeres of C. elegans embryos, which engulf and eliminate diverse phagocytic cargo from the corpse of the second polar body to cytokinetic midbody remnants. We describe the use of fluorescent time-lapse imaging to observe the distinct steps of phagocytic clearance and methods to normalize this process to distinguish defects in mutant strains. These approaches have enabled us to reveal new insights from the initial signaling to induce phagocytosis up until the final resolution of phagocytic cargo in phagolysosomes.
    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
    巨噬细胞和嗜中性粒细胞等细胞可以内化一组不同的颗粒物,说明细菌和凋亡小体经由过程吞噬。这些颗粒被隔离成吞噬体,然后与早期和晚期内体融合,最终与溶酶体融合成熟成吞噬溶酶体,通过一个被称为吞噬体成熟的过程。最终,粒子降解后,然后,吞噬体片段以通过吞噬体分解来改革溶酶体。随着吞噬体的变化,它们获取和剥离与吞噬体成熟和分解的各个阶段相关的蛋白质。可以通过使用免疫荧光方法在单吞噬体水平上评估这些变化。通常,我们使用间接免疫荧光方法,该方法依赖于针对追踪吞噬体成熟的特定分子标记的一级抗体。通常,吞噬小体进入吞噬溶酶体的进展可以通过对细胞进行溶酶体相关膜蛋白I(LAMP1)染色并通过显微镜或流式细胞术测量每个吞噬小体周围LAMP1的荧光强度来确定。然而,该方法可用于检测任何具有免疫荧光相容抗体的分子标记。
    Cells such as macrophages and neutrophils can internalize a diverse set of particulate matter, illustrated by bacteria and apoptotic bodies through the process of phagocytosis. These particles are sequestered into phagosomes, which then fuse with early and late endosomes and ultimately with lysosomes to mature into phagolysosomes, through a process known as phagosome maturation. Ultimately, after particle degradation, phagosomes then fragment to reform lysosomes through phagosome resolution. As phagosomes change, they acquire and divest proteins that are associated with the various stages of phagosome maturation and resolution. These changes can be assessed at the single-phagosome level by using immunofluorescence methods. Typically, we use indirect immunofluorescence methods that rely on primary antibodies against specific molecular markers that track phagosome maturation. Commonly, progression of phagosomes into phagolysosomes can be determined by staining cells for Lysosomal-Associated Membrane Protein I (LAMP1) and measuring the fluorescence intensity of LAMP1 around each phagosome by microscopy or flow cytometry. However, this method can be used to detect any molecular marker for which there are compatible antibodies for immunofluorescence.
    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
    丝状靶标通过吞噬杯内化,吞噬杯持续几分钟,然后关闭形成吞噬体。这种特性提供了研究吞噬作用中的关键事件的可能性,其空间和时间分辨率比使用球形粒子可能实现的分辨率要高。从吞噬杯到封闭的吞噬体的转变发生在粒子附着后的几秒钟内。在这一章中,我们提供了制备丝状细菌的方法,并描述了如何将它们用作研究吞噬作用的不同方面的靶标。
    Filamentous targets are internalized via phagocytic cups that last for several minutes before closing to form a phagosome. This characteristic offers the possibility to study key events in phagocytosis with greater spatial and temporal resolution than is possible to achieve using spherical particles, for which the transition from a phagocytic cup to an enclosed phagosome occurs within a few seconds after particle attachment. In this chapter, we provide methodologies to prepare filamentous bacteria and describe how they can be used as targets to study different aspects of phagocytosis.
    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
    The decision whether endosomes enter the degradative or recycling pathway in mammalian cells is of fundamental importance for pathogen killing, and its malfunctioning has pathological consequences. We discovered that human p11 is a critical factor for this decision. The HscA protein present on the conidial surface of the human-pathogenic fungus Aspergillus fumigatus anchors p11 on conidia-containing phagosomes (PSs), excludes the PS maturation mediator Rab7, and triggers binding of exocytosis mediators Rab11 and Sec15. This reprogramming redirects PSs to the non-degradative pathway, allowing A. fumigatus to escape cells by outgrowth and expulsion as well as transfer of conidia between cells. The clinical relevance is supported by the identification of a single nucleotide polymorphism in the non-coding region of the S100A10 (p11) gene that affects mRNA and protein expression in response to A. fumigatus and is associated with protection against invasive pulmonary aspergillosis. These findings reveal the role of p11 in mediating fungal PS evasion.
    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