LLO

军团菌病
  • 文章类型: Journal Article
    病原菌依靠分泌的毒力因子在易感宿主中引起疾病。然而,在革兰氏阳性细菌中,分泌蛋白激活和调节膜后易位的潜在机制仍然未知.使用蛋白质组学,我们鉴定了几种依赖于分泌伴侣PrsA2的蛋白质。我们追踪了表型,生物化学,以及生物物理分析和计算分析,以检查检测到的关键分泌毒力因子的调节,李斯特菌溶血素O(LLO),及其与细菌病原体单核细胞增生李斯特菌(Lm)的PrsA2的相互作用。Lm毒力的关键是宿主细胞的内化和胆固醇依赖性成孔毒素的后续作用,LLO,这使得细菌能够从宿主细胞吞噬体逃脱。由于Lm是革兰氏阳性生物,细胞膜和细胞壁之间的空间是溶剂暴露。因此,我们假设,随着病原体内化到吞噬体,从中性到酸性pH的下降对于调节PrsA2与LLO的相互作用至关重要。这里,我们证明了PrsA2以pH依赖性方式直接与LLO相互作用。我们表明,PrsA2在中性pH条件下保护和螯合LLO,可以观察到LLO聚集。此外,我们确定了相互作用以及最终LLO的折叠和活性所需的PrsA2的分子特征。此外,蛋白质复合物建模表明PrsA2通过其胆固醇结合域与LLO相互作用。这些发现强调了革兰氏阳性分泌伴侣调节分泌的机制,稳定性,和在与宿主细胞感染相关的条件下折叠成孔毒素。
    目的:Lm是一种普遍存在的食源性病原体,可对脆弱人群造成严重疾病。在感染期间,Lm依赖于广泛的分泌毒力因子库,包括使细菌能够侵入宿主并在细胞之间传播的LLO。膜易位后,分泌因子必须在具有挑战性的细菌细胞膜-壁界面中变得活跃。然而,分泌蛋白折叠和功能所需的机制在很大程度上是未知的。Lm编码一个伴侣,PrsA2,这对分泌因子的活性至关重要。这里,我们表明,PrsA2直接关联并保护主要的Lm毒力因子,LLO,在与宿主胞质溶胶相对应的条件下,其中LLO经历不可逆变性。此外,我们确定了PrsA2的分子特征,使其能够与LLO相互作用。一起,我们的结果表明,Lm和其他革兰氏阳性细菌可能利用分泌的伴侣来调节感染过程中孔形成毒素的活性.
    Pathogenic bacteria rely on secreted virulence factors to cause disease in susceptible hosts. However, in Gram-positive bacteria, the mechanisms underlying secreted protein activation and regulation post-membrane translocation remain largely unknown. Using proteomics, we identified several proteins that are dependent on the secreted chaperone PrsA2. We followed with phenotypic, biochemical, and biophysical assays and computational analyses to examine the regulation of a detected key secreted virulence factor, listeriolysin O (LLO), and its interaction with PrsA2 from the bacterial pathogen Listeria monocytogenes (Lm). Critical to Lm virulence is internalization by host cells and the subsequent action of the cholesterol-dependent pore-forming toxin, LLO, which enables bacterial escape from the host cell phagosome. Since Lm is a Gram-positive organism, the space between the cell membrane and wall is solvent exposed. Therefore, we hypothesized that the drop from neutral to acidic pH as the pathogen is internalized into a phagosome is critical to regulating the interaction of PrsA2 with LLO. Here, we demonstrate that PrsA2 directly interacts with LLO in a pH-dependent manner. We show that PrsA2 protects and sequesters LLO under neutral pH conditions where LLO can be observed to aggregate. In addition, we identify molecular features of PrsA2 that are required for interaction and ultimately the folding and activity of LLO. Moreover, protein-complex modeling suggests that PrsA2 interacts with LLO via its cholesterol-binding domain. These findings highlight a mechanism by which a Gram-positive secretion chaperone regulates the secretion, stability, and folding of a pore-forming toxin under conditions relevant to host cell infection.
    OBJECTIVE: Lm is a ubiquitous food-borne pathogen that can cause severe disease to vulnerable populations. During infection, Lm relies on a wide repertoire of secreted virulence factors including the LLO that enables the bacterium to invade the host and spread from cell to cell. After membrane translocation, secreted factors must become active in the challenging bacterial cell membrane-wall interface. However, the mechanisms required for secreted protein folding and function are largely unknown. Lm encodes a chaperone, PrsA2, that is critical for the activity of secreted factors. Here, we show that PrsA2 directly associates and protects the major Lm virulence factor, LLO, under conditions corresponding to the host cytosol, where LLO undergoes irreversible denaturation. Additionally, we identify molecular features of PrsA2 that enable its interaction with LLO. Together, our results suggest that Lm and perhaps other Gram-positive bacteria utilize secreted chaperones to regulate the activity of pore-forming toxins during infection.
    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
    单核细胞增生李斯特菌(Lm),一种食源性细菌,可以感染人,在免疫功能低下的个体中死亡率很高。李斯特溶素O(LLO),Lm的主要毒力因子,对调节Lm的致病性至关重要。这篇综述得出结论,LLO可能直接或间接激活许多宿主细胞病毒病理生理学过程,如细胞凋亡,焦亡,自噬,坏死和坏死。我们描述了Lm对宿主细胞的侵袭以及随后从主要组织相容性复合物I类(MHC-I)和主要组织相容性复合物II类(MHC-II)中接受LLO表位后CD8T细胞和CD4T细胞对Lm的去除。然后描述了利用LLO的孔形成能力和宿主细胞的免疫应答的几种基于LLO的疫苗的开发。最后,我们的结论是概述了几种天然物质,这些物质已被证明通过与LLO的特定氨基酸残基结合来改变LLO的三维构象,这降低了LLO的致病性,可能是Lm的一种可能的药物治疗方法。
    Listeria monocytogenes (Lm), a foodborne bacterium, can infect people and has a high fatality rate in immunocompromised individuals. Listeriolysin O (LLO), the primary virulence factor of Lm, is critical in regulating the pathogenicity of Lm. This review concludes that LLO may either directly or indirectly activate a number of host cell viral pathophysiology processes, such as apoptosis, pyroptosis, autophagy, necrosis and necroptosis. We describe the invasion of host cells by Lm and the subsequent removal of Lm by CD8 T cells and CD4 T cells upon receipt of the LLO epitopes from major histocompatibility complex class I (MHC-I) and major histocompatibility complex class II (MHC-II). The development of several LLO-based vaccines that make use of the pore-forming capabilities of LLO and the immune response of the host cells is then described. Finally, we conclude by outlining the several natural substances that have been shown to alter the three-dimensional conformation of LLO by binding to particular amino acid residues of LLO, which reduces LLO pathogenicity and may be a possible pharmacological treatment for Lm.
    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
    单核细胞增生李斯特菌,一种食源性病原体,导致高致死率的李斯特菌病,表现出已知正在逐渐增加的多药耐药性(MDR)。替代的抗菌策略对于治疗这种众所周知的病原体有很高的需求。作为治疗细菌感染的新方法,正在探索抗生物膜和抗毒力策略。在这项研究中,一种罕见的抗菌药,名叫setomimycin,对单核细胞增生李斯特菌具有有效的抗菌活性。接下来,在亚最低抑制浓度(亚MIC)下,研究了单独或与卡那霉素和阿米卡星联合使用的结单霉素对单核细胞增生李斯特菌生物膜形成和李斯特菌溶血素O(LLO)产生的抑制作用。结晶紫染色证实,与卡那霉素或阿米卡星联合使用的塞托霉素可以显着减少亚MIC的单核细胞增生李斯特菌生物膜的形成,通过扫描电子显微镜(SEM)和共聚焦激光扫描显微镜(CLSM)进一步评估。同时,塞托霉素的亚MIC可以显着抑制LLO的分泌。此外,与生物膜和主要毒力因子相关的基因转录,比如LLO,鞭毛,和金属蛋白酶,在亚MIC时被塞托霉素抑制。因此,这项研究提供了一个深入的见解,以赛托霉素作为一种替代的抗菌剂对单核细胞增生李斯特菌。
    Listeria monocytogenes, a foodborne pathogen that causes listeriosis with high fatality rate, exhibits multidrug resistance (MDR) known to be progressively increasing. Alternative antibacterial strategies are in high demand for treating this well-known pathogen. Anti-biofilm and anti-virulence strategies are being explored as novel approaches to treat bacterial infections. In this study, one rare antibacterial named setomimycin was isolated from Streptomyces cyaneochromogenes, which showed potent antibacterial activity against L. monocytogenes. Next, the inhibition of biofilm formation and listeriolysin O (LLO) production against L. monocytogenes were investigated at sub-minimal inhibitory concentrations (sub-MICs) of setomimycin alone or combined with kanamycin and amikacin. Crystal violet staining confirmed that setomimycin combining with kanamycin or amikacin could dramatically reduce biofilm formation against L. monocytogenes at sub-MICs, which was further evaluated by scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). In the meantime, sub-MICs of setomimycin could significantly suppress the secretion of LLO. Furthermore, the transcription of genes associated with biofilms and main virulence factors, such as LLO, flagellum, and metalloprotease, were suppressed by setomimycin at sub-MICs. Hence, the study provided a deep insight into setomimycin as an alternative antibacterial agent against L. monocytogenes.
    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
    单核细胞增生李斯特菌(L.M)由先天免疫的几个细胞有效控制,包括肥大细胞(MC)。MC通过L.m诱导其脱颗粒而被激活,细胞因子的产生和杀微生物机制。TLR2是免疫系统不同细胞对L.m感染的最佳控制所必需的。然而,对识别这种细菌的MC受体以及这些相互作用是否介导MC激活知之甚少。在这项研究中,我们分析了在L.m感染期间TLR2是否参与介导不同的MC激活反应.我们发现尽管MC感染了L.M,他们能够清除细菌负荷。此外,MC脱粒并产生ROS,TNF-α,IL-1β,IL-6、IL-13和MCP-1响应细菌感染。有趣的是,L.m诱导信号蛋白的激活:ERK,p38和NF-κB。当TLR2被阻断时,L.m内吞作用,杀菌活性,ROS产生和肥大细胞脱颗粒不受影响。有趣的是,当TLR2对L.m感染的反应被抑制时,只有IL-6和IL-13的产生受到影响。此外,p38激活依赖于TLR2,但不依赖于ERK或NF-κB激活。这些结果表明,TLR2在L.m感染期间仅介导一些MC激活途径,主要与IL-6和IL-13产生有关。
    Listeria monocytogenes (L.m) is efficiently controlled by several cells of the innate immunity, including the Mast Cell (MC). MC is activated by L.m inducing its degranulation, cytokine production and microbicidal mechanisms. TLR2 is required for the optimal control of L.m infection by different cells of the immune system. However, little is known about the MC receptors involved in recognizing this bacterium and whether these interactions mediate MC activation. In this study, we analyzed whether TLR2 is involved in mediating different MC activation responses during L.m infection. We found that despite MC were infected with L.m, they were able to clear the bacterial load. In addition, MC degranulated and produced ROS, TNF-α, IL-1β, IL-6, IL-13 and MCP-1 in response to bacterial infection. Interestingly, L.m induced the activation of signaling proteins: ERK, p38 and NF-κB. When TLR2 was blocked, L.m endocytosis, bactericidal activity, ROS production and mast cell degranulation were not affected. Interestingly, only IL-6 and IL-13 production were affected when TLR2 was inhibited in response to L.m infection. Furthermore, p38 activation depended on TLR2, but not ERK or NF-κB activation. These results indicate that TLR2 mediates only some MC activation pathways during L.m infection, mainly those related to IL-6 and IL-13 production.
    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
    成孔蛋白(PFP)包括由许多病原细菌产生的毒力因子。然而,PFP还包含非毒力因子,如凋亡的Bcl2样蛋白,也发生在非致病性细菌中,甚至发生在所有生命王国中。成孔蛋白是一类古老的蛋白质,在破坏细胞膜方面非常强大。总的来说,与脂质膜结合后,它们从可溶性单体形式转化为寡聚态,然后经历巨大的构象变化形成跨膜孔。因此,PFP使其靶细胞的质膜对溶质具有渗透性,可能导致细胞死亡,或者对细胞功能进行更微妙的操作。最近的冷冻EM和X射线晶体学研究揭示了几种PFP在其孔前和孔状态下的高分辨率结构,然而,关于诱导孔隙形成的线索的许多方面,孔前到孔的构象转变,膜渗透的机制和相关的动力学仍然知之甚少,和这些转变的动态的直接可视化缺失。使用高速原子力显微镜(HS-AFM),各种PFP的低聚动力学和孔前到孔的转变动力学,如李斯特溶素O(LLO),lysenin,和穿孔素-2(PFN2),可以研究。这些研究表明,LLO不会形成规则形状或大小的孔,而是形成膜插入弧,这些弧作为线性体传播和破坏脂质膜。相比之下,lysenin形成稳定的前孔和孔非酰胺环,HS-AFM可以研究它们在膜上的扩散和pH依赖性插入。同样,PFN2在酸化时经历了孔前到孔的转变。HS-AFM系统的开放性允许对过渡进行实时成像,并揭示了所有观察到的分子在3s内过渡到孔状态。在这一章中,我们详细说明了制备脂质的方案,形成支持的脂质双层,并提供实时指南,实际空间HS-AFM观测PFP的作用。
    Pore-forming proteins (PFPs) include virulence factors that are produced by many pathogenic bacteria. However, PFPs also comprise non-virulence factors, such as apoptotic Bcl2-like proteins, and also occur in non-pathogenic bacteria and indeed in all kingdoms of life. Pore-forming proteins are an ancient class of proteins, which are tremendously powerful in damaging cell membranes. In general, upon binding to lipid membranes, they convert from the soluble monomeric form into an oligomeric state, and then undergo a dramatic conformational change to form transmembrane pores. Thus, PFPs render the plasma membrane of their target cells permeable to solutes, potentially leading to cell death, or to more subtle manipulations of cellular functions. Recent cryo-EM and X-ray crystallography studies revealed high-resolution structures of several PFPs in their pre-pore and pore states, however many aspects regarding the cues that induce pore formation, the pre-pore to pore conformational transition, the mechanism of membrane permeation and associated dynamics are still less well understood, and direct visualization of the dynamics of these transitions are missing. Using high-speed atomic force microscopy (HS-AFM), the kinetics of oligomerization and the pre-pore to pore transition dynamics of various PFPs, such as Listeriolysin O (LLO), lysenin, and Perforin-2 (PFN2), could be studied. These studies revealed that LLO does not form pores of regular shape or size, but rather forms membrane inserted arcs that propagate and damage lipid membranes as lineactants. In contrast, lysenin forms stable pre-pore and pore nonameric rings and HS-AFM allowed to study their diffusion on and the pH-dependent insertion into the membrane. Similarly, PFN2 underwent pre-pore to pore transition upon acidification. The openness of the HS-AFM system allowed the transition to be imaged in real time and revealed that all observed molecules transitioned into the pore state within 3s. In this chapter, we detail protocols to prepare lipids, form supported lipid bilayers, and provide guidelines for real-time, real-space HS-AFM observations of PFPs in action.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    A Li-rich layered oxide (LLO) cathode with morphology-dependent electrochemical performance with the composition Li1.23Mn0.538Ni0.117Co0.114O2 in three different microstructural forms, namely, randomly shaped particles, platelets, and nanofibers, is synthesized through the solid-state reaction (SSR-LLO), hydrothermal method (HT-LLO), and electrospinning process (ES-LLO), respectively. Even though the cathodes possess different morphologies, structurally they are identical. The elemental dispersion studies using energy-dispersive X-ray spectroscopy mapping in scanning transmission electron microscopy show uniform distribution of elements. However, SSR-LLO and ES-LLO nanofibers show slight Co-rich regions. The electrochemical studies of LLO cathodes are evaluated in terms of charging/discharging, C-rate capability, and cyclic stability performances. A high reversible capacity of 275 mA h g-1 is achieved in the fibrous LLO cathode which also demonstrates good high-rate capability (80 mA h g-1 at 10 C-rate). These capacities and rate capabilities are superior to those of SSR-LLO [210.5 mA h g-1 (0.1 C-rate) and 4 mA h g-1 (3 C-rate)] and HT-LLO [242 mA h g-1 (0.1 C-rate) and 22 mA h g-1 (10 C-rate)] cathodes. The ES-LLO cathode exhibits 88% capacity retention after 100 cycles at 1 C-rate. A decrease in voltage on cycling is found to be common in all three cathodes; however, minimal voltage decay and capacity loss are observed in ES-LLO upon cycling. Well-connected small LLO particles constituting fibrous microstructural forms in ES-LLO provide an enhanced electrolyte/cathode interfacial area and reduced diffusion path length for Li+. This, in turn, facilitates superior electrochemical performance of the electrospun Co-low LLO cathode suitable for quick charge battery applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Listeria monocytogenes is a foodborne pathogen capable of secreting listeriolysin O (LLO), a pore-forming toxin encoded by the hly gene. While the functions of LLO have been studied extensively, how the production of LLO is modulated by the intestinal environment, devoid of oxygen and enriched in short chain fatty acids (SCFAs), is not completely understood. Using L. monocytogenes strain 10403s, we found that hly transcription was moderately decreased by aerobic SCFA exposures but significantly increased by anaerobic SCFA exposures. Moreover, aerobic, but not anaerobic, exposure to low levels of SCFAs resulted in a significantly higher LLO activity. These results demonstrated that transcriptional and post-transcriptional regulations of LLO production were separately modulated by SCFAs and were responsive to oxygen levels. Examining isogenic mutants revealed that PrfA and SigB play a role in regulating LLO production in response to SCFAs. Effects of SCFAs were also present in the cardiotropic strain 07PF0776 but distinctly different from those in strain 10403s. For both strains, prior exposures to SCFAs altered intracellular infections in Caco-2 and RAW264.7 cells and the plaque sizes in L fibroblasts, a result confirming the ability of L. monocytogenes to adapt to SCFAs in ways that impact its subsequent infection outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Listeria monocytogenes is a Gram-positive pathogen able to cause severe human infections. Its major virulence regulator is the transcriptional activator PrfA, a member of the Crp/Fnr family of transcriptional regulators. To establish a successful L. monocytogenes infection, the PrfA protein needs to be in an active conformation, either by binding the cognate inducer glutathione (GSH) or by possessing amino acid substitutions rendering the protein constitutively active (PrfA*). By a yet unknown mechanism, phosphotransferase system (PTS) sugars repress the activity of PrfA. We therefore took a transposon-based approach to identify the mechanism by which PTS sugars repress PrfA activity. For this, we screened a transposon mutant bank to identify clones able to grow in the presence of glucose-6-phosphate as the sole carbon source. Surprisingly, most of the isolated transposon mutants also carried amino acid substitutions in PrfA. In transposon-free strains, the PrfA amino acid substitution mutants displayed growth, virulence factor expression, infectivity, and DNA binding, agreeing with previously identified PrfA* mutants. Hence, the initial growth phenotype observed in the isolated clone was due to the amino acid substitution in PrfA and unrelated to the loci inactivated by the transposon mutant. Finally, we provide structural evidence for the existence of an intermediately activated PrfA state, which gives new insights into PrfA protein activation.IMPORTANCE The Gram-positive bacterium Listeria monocytogenes is a human pathogen affecting mainly the elderly, immunocompromised people, and pregnant women. It can lead to meningoencephalitis, septicemia, and abortion. The major virulence regulator in L. monocytogenes is the PrfA protein, a transcriptional activator. Using a growth-based selection strategy, we identified mutations in the PrfA protein leading to constitutively active virulence factor expression. We provide structural evidence for the existence of an intermediately activated PrfA state, which gives new insights into PrfA protein activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Listeria monocytogenes (Lm) is a widespread foodborne intracellular pathogen that invades a variety of cells, causing abortions and severe human diseases. After internalization into host cells, pore-forming cytolysin listeriolysin O (LLO) disrupts the phagosome, which allows the bacterium to survive and colonize the cytoplasm, providing the bacterium the chance to infect neighboring cells. Betulin is an extracted natural compound from birch bark with diverse pharmacological activities. Here, we showed that LLO-induced rabbit red blood cell lysis in vitro was inhibited by preincubation with betulin, which suppressed the oligomerization process. Infectious assays performed with human monocyte macrophages indicated that betulin significantly protected cells against Lm-induced cell injury. In addition, Balb/c mice were used to perform a general infection, and betulin administration obviously inhibited organ damage and bacterial burden in livers and spleens of infected mice. In conclusion, betulin obviously inhibited Lm-induced cell injury in vitro and protected against infection in vivo through an antivirulence effect. Our results showed betulin as a new candidate against listeriosis by targeting LLO and highlight the potential of natural product-based medicine to be applied in the treatment of pathogenic infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Case Reports
    ALG11-Congenital Disorder of Glycosylation (ALG11-CDG, also known as congenital disorder of glycosylation type Ip) is an inherited inborn error of metabolism due to abnormal protein and lipid glycosylation. We describe two unrelated patients with ALG11-CDG due to novel mutations, review the literature of previously described affected individuals, and further expand the clinical phenotype. Both affected individuals reported here had severe psychomotor disabilities and epilepsy. Their fibroblasts synthesized truncated precursor glycan structures, consistent with ALG11-CDG, while also showing hypoglycosylation of a novel biomarker, GP130. Surprisingly, one patient presented with normal transferrin glycosylation profile, a feature that has not been reported previously in patients with ALG11-CDG. Together, our data expand the clinical and mutational spectrum of ALG11-CDG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号