effector protein

效应蛋白
  • 文章类型: Journal Article
    细胞内细菌病原体转移多个细胞途径以建立其生态位并在其宿主内持续存在。伯内蒂柯西拉,Q热的病原体,通过其4型分泌系统分泌细菌效应蛋白以产生含柯西氏菌的液泡(CCV)。这些效应子对脂质和蛋白质运输的操纵对于细菌复制和毒力至关重要。这里,我们已经表征了CCV的脂质组成,并发现效应子副与磷酸肌醇和富含磷脂酰丝氨酸和溶异双磷脂酸的膜相互作用。值得注意的是,异位表达Vice的真核细胞在形态和组成上都存在类似于早期CCV的区室。我们发现这些区室的生物发生依赖于副的双重功能。效应蛋白最初定位于真核细胞的质膜,在那里它通过大细胞胞吞作用触发大液泡的内化。然后,通过干扰ESCRT机械来稳定这些隔间。总的来说,我们的研究结果表明,Vice是一种必需的C.burnetii效应蛋白,能够劫持两个主要的细胞途径来塑造细菌复制性生态位。
    Intracellular bacterial pathogens divert multiple cellular pathways to establish their niche and persist inside their host. Coxiella burnetii, the causative agent of Q fever, secretes bacterial effector proteins via its Type 4 secretion system to generate a Coxiella-containing vacuole (CCV). Manipulation of lipid and protein trafficking by these effectors is essential for bacterial replication and virulence. Here, we have characterized the lipid composition of CCVs and found that the effector Vice interacts with phosphoinositides and membranes enriched in phosphatidylserine and lysobisphosphatidic acid. Remarkably, eukaryotic cells ectopically expressing Vice present compartments that resemble early CCVs in both morphology and composition. We found that the biogenesis of these compartments relies on the double function of Vice. The effector protein initially localizes at the plasma membrane of eukaryotic cells where it triggers the internalization of large vacuoles by macropinocytosis. Then, Vice stabilizes these compartments by perturbing the ESCRT machinery. Collectively, our results reveal that Vice is an essential C. burnetii effector protein capable of hijacking two major cellular pathways to shape the bacterial replicative niche.
    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
    许多细菌病原体已经进化出有效的策略来干扰泛素化网络以逃避先天免疫系统的清除。这里,我们报道了OTUB1,哺乳动物细胞中最丰富的去泛素酶(DUB)之一,在嗜肺军团菌感染期间同时进行规范和非规范的泛素化。效应子SidC和SdcA在多个赖氨酸残基上催化OTUB1泛素化,导致其与含军团菌的液泡相关联。通过SidC和SdcA的赖氨酸泛素化促进OTUB1和DEPTOR之间的相互作用,MTORC1通路的抑制剂,从而抑制MTORC1信号。MTORC1的抑制导致宿主蛋白合成的抑制和宿主巨自噬/自噬在肺炎杆菌感染期间的促进。此外,SidE家族效应子(SidEs)的成员在Ser16和Ser18处诱导OTUB1的磷酸核糖(PR)连接的泛素化,并阻断其DUB活性。OTUB1的赖氨酸和丝氨酸泛素化的水平进一步受到具有拮抗SidC活性的效应子的调节,SdcA和侧面,包括Lem27Dupa,Dupb,SidJ和SdjA.我们的研究揭示了调节宿主DUB活性的效应子介导的复杂机制。缩写:BafA1:bafilomycinA1;BMDMs:骨髓衍生的巨噬细胞;DUB:去泛素酶;Dot/Icm:细胞器运输/细胞内繁殖缺陷;DEPTOR:含有MTOR相互作用蛋白的DEP结构域;GAPDH:甘油醛-3-磷酸脱氢酶;嗜肺军团菌:嗜肺军团菌;LCV:含军团菌复合酶1的MTUO泛素醛结合1;PR-Ub:磷酸核糖(PR)连接的泛素;PTM:翻译后修饰;SDS-PAGE:十二烷基硫酸钠-聚丙烯酰胺凝胶电泳;SidEs:SidE家族效应子;Ub:泛素。
    Many bacterial pathogens have evolved effective strategies to interfere with the ubiquitination network to evade clearance by the innate immune system. Here, we report that OTUB1, one of the most abundant deubiquitinases (DUBs) in mammalian cells, is subjected to both canonical and noncanonical ubiquitination during Legionella pneumophila infection. The effectors SidC and SdcA catalyze OTUB1 ubiquitination at multiple lysine residues, resulting in its association with a Legionella-containing vacuole. Lysine ubiquitination by SidC and SdcA promotes interactions between OTUB1 and DEPTOR, an inhibitor of the MTORC1 pathway, thus suppressing MTORC1 signaling. The inhibition of MTORC1 leads to suppression of host protein synthesis and promotion of host macroautophagy/autophagy during L. pneumophila infection. In addition, members of the SidE family effectors (SidEs) induce phosphoribosyl (PR)-linked ubiquitination of OTUB1 at Ser16 and Ser18 and block its DUB activity. The levels of the lysine and serine ubiquitination of OTUB1 are further regulated by effectors that function to antagonize the activities of SidC, SdcA and SidEs, including Lem27, DupA, DupB, SidJ and SdjA. Our study reveals an effectors-mediated complicated mechanism in regulating the activity of a host DUB.Abbreviations: BafA1: bafilomycin A1; BMDMs: bone marrow-derived macrophages; DUB: deubiquitinase; Dot/Icm: defective for organelle trafficking/intracellular multiplication; DEPTOR: DEP domain containing MTOR interacting protein; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; L. pneumophila: Legionella pneumophila; LCV: Legionella-containing vacuole; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; MTORC1: mechanistic target of rapamycin kinase complex 1; OTUB1: OTU deubiquitinase, ubiquitin aldehyde binding 1; PR-Ub: phosphoribosyl (PR)-linked ubiquitin; PTM: posttranslational modification; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SidEs: SidE family effectors; Ub: ubiquitin.
    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
    叶片变黄是一种众所周知的表型,可以吸引以韧皮部为食的昆虫。然而,目前尚不清楚昆虫介导的植物病原体如何诱导宿主叶片变黄以促进昆虫媒介自身传播。这里,我们报道了由水稻橙叶植物质(ROLP)分泌的效应蛋白抑制叶绿素生物合成并诱导叶片黄化以吸引叶斗载体,从而推测促进病原体传播。这个效应器,指定的分泌型ROLP蛋白1(SRP1),首先通过ROLP分泌到水稻韧皮部中,随后通过与叶绿体谷氨酰胺合成酶(GS2)相互作用转移到叶绿体。SRP1和GS2之间的直接相互作用破坏了GS2全酶的十聚体形成,削弱其酶活性,从而抑制叶绿素前体谷氨酸和谷氨酰胺的合成。SRP1在水稻植株中的转基因表达降低了GS2活性和叶绿素前体的积累,最终诱发叶片黄化。此过程与先前的证据相关,即水稻植物中GS2表达的敲除会导致类似的黄萎病表型。始终如一,这些变黄的叶子吸引了更多的叶斗矢量,导致载体更频繁地探测,并可能促进更有效的植物质传播。一起,这些结果揭示了植物为增强对昆虫媒介的吸引力而操纵受感染植物叶色的机制。
    Leaf yellowing is a well-known phenotype that attracts phloem-feeding insects. However, it remains unclear how insect-vectored plant pathogens induce host leaf yellowing to facilitate their own transmission by insect vectors. Here, we report that an effector protein secreted by rice orange leaf phytoplasma (ROLP) inhibits chlorophyll biosynthesis and induces leaf yellowing to attract leafhopper vectors, thereby presumably promoting pathogen transmission. This effector, designated secreted ROLP protein 1 (SRP1), first secreted into rice phloem by ROLP, was subsequently translocated to chloroplasts by interacting with the chloroplastic glutamine synthetase (GS2). The direct interaction between SRP1 and GS2 disrupts the decamer formation of the GS2 holoenzyme, attenuating its enzymatic activity, thereby suppressing the synthesis of chlorophyll precursors glutamate and glutamine. Transgenic expression of SRP1 in rice plants decreased GS2 activity and chlorophyll precursor accumulation, finally inducing leaf yellowing. This process is correlated with the previous evidence that the knockout of GS2 expression in rice plants causes a similar yellow chlorosis phenotype. Consistently, these yellowing leaves attracted higher numbers of leafhopper vectors, caused the vectors to probe more frequently, and presumably facilitate more efficient phytoplasma transmission. Together, these results uncover the mechanism used by phytoplasmas to manipulate the leaf color of infected plants for the purpose of enhancing attractiveness to insect vectors.
    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
    Pyroptosis is a type of programmed cell death mediated by the Gasdermin family. It is triggered in response to pathogen infection or other danger signals. The activation of Gasdermins leads to pyroptosis and the release of large amounts of inflammatory cytokines. Pyroptosis plays a crucial role in combating pathogen infections, as it helps to eliminate infected cells and activate the immune system. However, pathogens have already developed sophisticated strategies to evade or inhibit pyroptosis, allowing them to persist and facilitate infection. This review provides an overview of the discovery of pyroptosis and its importance in anti-infectious immunity. We also discuss several new strategies for inhibiting pyroptosis by pathogens. A thorough learning of the occurrence and regulation of pyroptosis may reveal the pathogenesis of related infectious diseases and contribute to developing effective anti-infective therapeutic strategies.
    细胞焦亡是一种由Gasdermin家族蛋白介导的新型程序性细胞死亡。当宿主细胞感应病原体感染或其他危险信号时,Gasdermin家族蛋白被切割活化并诱导细胞焦亡。细胞焦亡过程往往伴随大量炎性细胞因子释放,这些炎性细胞因子在宿主清除病原体过程中发挥着至关重要作用,而病原体在与宿主长期“博弈”过程中也进化出抑制细胞焦亡的策略以实现免疫逃逸。本文介绍了细胞焦亡的发现历程及其在抗感染免疫中的重要功能,并总结了病原体抑制细胞焦亡的多种新策略及其相关研究进展。深入理解细胞焦亡的发生及调控机制,可揭示相关感染性疾病的发病机制并有助于开发有效的抗感染治疗策略。.
    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
    肠沙门氏菌是人类细菌性食源性疾病的主要原因,每年造成数百万例病例。该病原体生存的关键策略是将称为效应子的细菌毒力因子转移到宿主细胞中。主要通过与宿主蛋白的蛋白-蛋白相互作用起作用。沙门氏菌基因组编码几种旁系效应子,据信这些效应子是在整个进化过程中由复制事件引起的。这些旁系同源物可以共享结构相似性和酶活性,但也显示出宿主细胞靶标或相互作用伙伴的差异以及对沙门氏菌细胞内生命周期的贡献。同源效应子SopD和SopD2共有63%的氨基酸序列相似性和广泛的结构同源性,但在分泌动力学方面表现出差异。细胞内定位,主机目标,以及在感染中的作用。SopD和SopD2目标宿主RabGTPases,它们代表了介导多种细胞功能的细胞内运输的关键调节剂。虽然SopD和SopD2都操纵Rab函数,这些旁系同源物显示出Rab特异性的差异,效应子也进化出了多种操作GTP酶的作用机制。这里,我们在宿主-病原体相互作用的背景下重点介绍了这对有趣的旁系效应子,并讨论了这项研究如何为效应子进化提供有价值的见解。
    Salmonella enterica is a leading cause of bacterial food-borne illness in humans and is responsible for millions of cases annually. A critical strategy for the survival of this pathogen is the translocation of bacterial virulence factors termed effectors into host cells, which primarily function via protein-protein interactions with host proteins. The Salmonella genome encodes several paralogous effectors believed to have arisen from duplication events throughout the course of evolution. These paralogs can share structural similarities and enzymatic activities but have also demonstrated divergence in host cell targets or interaction partners and contributions to the intracellular lifecycle of Salmonella. The paralog effectors SopD and SopD2 share 63% amino acid sequence similarity and extensive structural homology yet have demonstrated divergence in secretion kinetics, intracellular localization, host targets, and roles in infection. SopD and SopD2 target host Rab GTPases, which represent critical regulators of intracellular trafficking that mediate diverse cellular functions. While SopD and SopD2 both manipulate Rab function, these paralogs display differences in Rab specificity, and the effectors have also evolved multiple mechanisms of action for GTPase manipulation. Here, we highlight this intriguing pair of paralog effectors in the context of host-pathogen interactions and discuss how this research has presented valuable insights into effector evolution.
    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
    许多强大的方法已被用来阐明全球转录组,蛋白质组学,或对病原体感染的宿主细胞的代谢反应。然而,宿主糖对细菌感染的反应仍未被探索,因此,我们对细菌病原体操纵宿主糖有利于感染的分子机制的理解仍然不完整。这里,我们通过对细菌病原体布鲁氏菌感染期间的宿主糖进行系统分析来解决这一差距。导致布鲁氏菌病。我们发现,令人惊讶的是,布鲁氏菌效应蛋白(EP)Rhg1通过与控制N连接蛋白糖基化的寡糖转移酶复合物的成分相互作用来诱导宿主细胞N糖的整体重编程,和Rhg1调节布鲁氏菌病小鼠模型中的布鲁氏菌复制和组织定植,证明布鲁氏菌利用EPRhg1重新编程宿主N-糖并促进细菌细胞内寄生,从而为宿主细胞感染的细菌控制提供范例。
    Many powerful methods have been employed to elucidate the global transcriptomic, proteomic, or metabolic responses to pathogen-infected host cells. However, the host glycome responses to bacterial infection remain largely unexplored, and hence, our understanding of the molecular mechanisms by which bacterial pathogens manipulate the host glycome to favor infection remains incomplete. Here, we address this gap by performing a systematic analysis of the host glycome during infection by the bacterial pathogen Brucella spp. that cause brucellosis. We discover, surprisingly, that a Brucella effector protein (EP) Rhg1 induces global reprogramming of the host cell N-glycome by interacting with components of the oligosaccharide transferase complex that controls N-linked protein glycosylation, and Rhg1 regulates Brucella replication and tissue colonization in a mouse model of brucellosis, demonstrating that Brucella exploits the EP Rhg1 to reprogram the host N-glycome and promote bacterial intracellular parasitism, thereby providing a paradigm for bacterial control of host cell 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
    根据幽门螺杆菌菌株是否含有或缺乏称为cag致病性岛(cagPAI)的染色体区域,可以将其大致分为两组。cagPAI阳性菌株在人胃中的定植与胃癌和消化性溃疡疾病的风险增加有关,与CagPAI阴性菌株的定植相比。cagPAI编码分泌的效应蛋白(CagA)和IV型分泌系统(CagT4SS)的组分,其将CagA和非蛋白质底物递送到宿主细胞中。动物模型实验表明CagA和CagT4SS刺激胃粘膜炎症反应并促进胃癌的发展。在这次审查中,我们讨论了最近的研究,定义了CagA和CagT4SS的结构和功能特征,以及含有cagPAI的幽门螺杆菌菌株促进胃癌和消化性溃疡疾病发展的机制。
    Helicobacter pylori strains can be broadly classified into two groups based on whether they contain or lack a chromosomal region known as the cag pathogenicity island (cag PAI). Colonization of the human stomach with cag PAI-positive strains is associated with an increased risk of gastric cancer and peptic ulcer disease, compared to colonization with cag PAI-negative strains. The cag PAI encodes a secreted effector protein (CagA) and components of a type IV secretion system (Cag T4SS) that delivers CagA and non-protein substrates into host cells. Animal model experiments indicate that CagA and the Cag T4SS stimulate a gastric mucosal inflammatory response and contribute to the development of gastric cancer. In this review, we discuss recent studies defining structural and functional features of CagA and the Cag T4SS and mechanisms by which H. pylori strains containing the cag PAI promote the development of gastric cancer and peptic ulcer disease.
    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
    \'念珠菌phytoplasmamelia\'是与中国黄化病有关的病原体,这已成为阿根廷中国林业生产的主要植物检疫问题。尽管有经济影响,这种植物质的基因组信息尚未公布,这阻碍了它在基因组水平上的表征。在这项研究中,我们使用宏基因组学方法分析了Ca的基因组草案。P.Meliae\'菌株ChTYXIII。草案集合由21个重叠群组成,总长度为751.949bp,注释揭示了669个CDS,34个tRNA,和1组rRNA操纵子。代谢途径分析表明,ChTYXIII包含用于糖酵解的完整核心基因和用于蛋白质易位的功能性Sec系统。我们基于133个单拷贝基因和基因组到基因组指标的系统基因组分析支持将其分类为独特的Ca。P.MPV进化枝内的物种。我们还确定了31个推定效应子,包括SAP11的同源物和其他仅在该病原体中描述的同源物。我们的直系同源分析揭示了Ca基因组中的37个PMU核心基因。P.Meliae\'ChTYXIII,导致2个完整的PMU的鉴定。我们的工作为Ca提供了重要的基因组信息。马柳和其他植物为16SrXIII(MPV)组。
    \'Candidatus Phytoplasma meliae\' is a pathogen associated with chinaberry yellowing disease, which has become a major phytosanitary problem for chinaberry forestry production in Argentina. Despite its economic impact, no genome information of this phytoplasma has been published, which has hindered its characterization at the genomic level. In this study, we used a metagenomics approach to analyze the draft genome of the \'Ca. P. meliae\' strain ChTYXIII. The draft assembly consisted of twenty-one contigs with a total length of 751.949 bp, and annotation revealed 669 CDSs, 34 tRNAs, and 1 set of rRNA operons. The metabolic pathways analysis showed that ChTYXIII contains the complete core genes for glycolysis and a functional Sec system for protein translocation. Our phylogenomic analysis based on 133 single-copy genes and genome-to-genome metrics supports the classification as unique \'Ca. P. species\' within the MPV clade. We also identified 31 putative effectors, including a homolog to SAP11 and others that have only been described in this pathogen. Our ortholog analysis revealed 37 PMU core genes in the genome of \'Ca. P. meliae\' ChTYXIII, leading to the identification of 2 intact PMUs. Our work provides important genomic information for \'Ca. P. meliae\' and others phytoplasmas for the 16SrXIII (MPV) group.
    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
    布鲁氏菌属革兰氏阴性细胞内细菌病原体,可引起世界范围内的人畜共患疾病布鲁氏菌病。布鲁氏菌可以感染许多哺乳动物,包括人类、家畜和野生动物。布鲁氏菌操纵各种宿主细胞过程以侵入和繁殖专业和非专业吞噬细胞。然而,宿主靶标及其通过布鲁氏菌促进感染过程的调节仍然不清楚。这里,我们报道了宿主泛素特异性蛋白酶,USP8,通过质膜受体负调节布鲁氏菌对巨噬细胞的侵袭,CXCR4。在沉默或化学抑制USP8后,CXCR4受体的膜定位被富集,这增加了布鲁氏菌对巨噬细胞的入侵。通过化学抑制14-3-3蛋白激活USP8影响布鲁氏菌对巨噬细胞的侵袭。布鲁氏菌在感染巨噬细胞的感染早期抑制了Usp8的表达。此外,我们发现只有活的布鲁氏菌可以负调节Usp8的表达,提示布鲁氏菌分泌效应蛋白在调节基因表达中的作用。随后的研究表明,布鲁氏菌效应蛋白,来自布鲁氏菌的含TIR结构域的蛋白质,TcpB,通过靶向环AMP反应元件结合蛋白途径下调Usp8的表达发挥重要作用。用USP8抑制剂处理小鼠可提高B.melitensis的存活率,而用CXCR4或14-3-3拮抗剂治疗的小鼠显示细菌负荷减少。我们的实验数据证明了Usp8在宿主防御微生物入侵中的新作用。本研究提供了对宿主防御的微生物颠覆的见解,这些信息可能最终有助于开发针对传染病的新型治疗干预措施。
    Brucella species are Gram-negative intracellular bacterial pathogens that cause the worldwide zoonotic disease brucellosis. Brucella can infect many mammals, including humans and domestic and wild animals. Brucella manipulates various host cellular processes to invade and multiply in professional and non-professional phagocytic cells. However, the host targets and their modulation by Brucella to facilitate the infection process remain obscure. Here, we report that the host ubiquitin-specific protease, USP8, negatively regulates the invasion of Brucella into macrophages through the plasma membrane receptor, CXCR4. Upon silencing or chemical inhibition of USP8, the membrane localization of the CXCR4 receptor was enriched, which augmented the invasion of Brucella into macrophages. Activation of USP8 through chemical inhibition of 14-3-3 protein affected the invasion of Brucella into macrophages. Brucella suppressed the expression of Usp8 at its early stage of infection in the infected macrophages. Furthermore, we found that only live Brucella could negatively regulate the expression of Usp8, suggesting the role of secreted effector protein of Brucella in modulating the gene expression. Subsequent studies revealed that the Brucella effector protein, TIR-domain containing protein from Brucella, TcpB, plays a significant role in downregulating the expression of Usp8 by targeting the cyclic-AMP response element-binding protein pathway. Treatment of mice with USP8 inhibitor resulted in enhanced survival of B. melitensis, whereas mice treated with CXCR4 or 14-3-3 antagonists showed a diminished bacterial load. Our experimental data demonstrate a novel role of Usp8 in the host defense against microbial intrusion. The present study provides insights into the microbial subversion of host defenses, and this information may ultimately help to develop novel therapeutic interventions for infectious diseases.
    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
    Ustilagomaydis是一种生物营养真菌,可在玉米的所有地上部分引起肿瘤形成。在渗透和定殖过程中,蛋黄菌分泌效应蛋白,成功克服植物免疫反应,并重新编程宿主生理学以促进感染。在这项研究中,我们在功能上表征了美国蛋黄连效应蛋白无上型(TPL)相互作用蛋白6(Tip6)。我们发现Tip6通过其两个乙烯响应元件结合因子相关的两亲性阻遏(EAR)基序与RELK2的N端相互作用。我们表明,EAR基序对于Tip6的毒力功能至关重要,对于改变RELK2的核分布模式至关重要。我们建议Tip6模拟植物抑制蛋白对RELK2的募集,从而破坏宿主的转录调控。我们表明,在Tip6的存在下,大量的AP2/ERFB1亚家族转录因子被错误调节。我们的研究表明了一种调节机制,其中美国蛋黄连效应子Tip6利用抑制结构域来招募辅抑制因子RELK2以破坏宿主植物的转录网络。
    Ustilago maydis is a biotrophic fungus that causes tumor formation on all aerial parts of maize. U. maydis secretes effector proteins during penetration and colonization to successfully overcome the plant immune response and reprogram host physiology to promote infection. In this study, we functionally characterized the U. maydis effector protein Topless (TPL) interacting protein 6 (Tip6). We found that Tip6 interacts with the N-terminus of RELK2 through its two Ethylene-responsive element binding factor-associated amphiphilic repression (EAR) motifs. We show that the EAR motifs are essential for the virulence function of Tip6 and critical for altering the nuclear distribution pattern of RELK2. We propose that Tip6 mimics the recruitment of RELK2 by plant repressor proteins, thus disrupting host transcriptional regulation. We show that a large group of AP2/ERF B1 subfamily transcription factors are misregulated in the presence of Tip6. Our study suggests a regulatory mechanism where the U. maydis effector Tip6 utilizes repressive domains to recruit the corepressor RELK2 to disrupt the transcriptional networks of the host plant.
    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