首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 359 毫秒
1.
鼠伤寒沙门菌通过效应蛋白注入,利用宿主细胞的肌动蛋白骨架网络入侵非吞噬细胞。已知SipC能使肌动蛋白成核,使纤维型肌动蛋白成束,并转运鼠伤寒沙门菌Ⅲ型分泌系统的效应蛋白。但SipC如何使纤维型肌动蛋白成束的分子机制及发挥此活性功能域的作用仍不清楚。该研究利用一系列SipC删除/插入突  相似文献   

2.
沙门菌是一种重要的人兽共患食源性病原菌。其感染宿主后可以凭借独特的免疫逃逸机制逃避宿主免疫系统的清除,潜伏在宿主体内1年至终身不等,从而建立持续性感染。沙门菌持续性感染与毒力岛密切相关,尤其是沙门菌毒力岛(Salmonella pathogenicity islands,SPIs) SPI-1和SPI-2。SPI-1效应蛋白SipB和SipC等以不同的途径影响细菌入侵,诱导细胞自噬或者凋亡;而SPI-2效应蛋白SseI和SseL等可以通过调控不同的信号通路协助沙门菌的胞内存活,为沙门菌持续性感染的发生和发展提供条件。本文主要阐述SipB和SseI等毒力岛效应蛋白在沙门菌持续性感染过程中的作用,同时总结了SPI-6、SPI-7和SPI-19等毒力岛的作用,以期为研究沙门菌持续性感染提供新思路。  相似文献   

3.
耶尔森氏菌属中有3个种对人和动物具有致病性,包括鼠疫杆菌(Yersinia pestis)、小肠结肠炎耶尔森氏菌(Yersinia enterocolitica)和假结核耶尔森氏菌(Yersinia pseudotuberculosis)。研究发现,假结核耶尔森氏菌能通过小鼠小肠派氏淋巴结的M细胞(Microfold cell)进行跨细胞转运。这种转运方式首先是细菌利用其表面蛋白侵袭素Invasin或黏附素Yad A蛋白识别并结合宿主细胞表面的整合素Integrin受体家族成员的β1链,然后细胞膜上的分泌通道打开,细菌利用Ⅲ型分泌系统把效应蛋白注入宿主细胞内,破坏宿主细胞免疫系统进行感染的过程。过去三十年内关于假结核耶尔森氏菌有大量的文献报道,本文综述该菌与宿主细胞之间相互作用分子机制的研究进展,并讨论目前关于假结核耶尔森氏菌的研究方向及热点。  相似文献   

4.
沙门菌(Salmonella)通过向宿主细胞分泌毒力效应蛋白(effector protein)来调控细胞内一系列的信号传导通路,从而有利于沙门菌的侵染和繁殖。NF-κB信号通路在宿主对病原菌的炎症反应及免疫应答中发挥着重要的作用,也是很多毒力效应蛋白调控的靶点。沙门菌致病岛(Salmonella pathogenicity island,SPI)-1上的毒力效应蛋白Sip A、Sop E、Sop E2和Sop B都能激活宿主细胞的NF-κB信号通路,而毒力效应蛋白Spt P、Avr A、Ssp H1以及SPI-2上的Sse L能有效地抑制NF-κB信号通路。研究这些毒力效应蛋白对NF-κB信号通路的时相调控和协同作用,将进一步揭示沙门菌的致病机制。  相似文献   

5.
流感病毒的蛋白和基因组在宿主细胞内能否正确地转运到相关部位,直接影响到病毒颗粒的形态发生。流感病毒跨膜蛋白(HA、NA和M2)主要通过宿主细胞的运输膜泡实现转运,而宿主细胞的蛋白转运机器参与了这一过程。新合成的流感病毒核糖核蛋白复合物(vRNPs)出核后,通过与活化的Rab11相结合,聚集于邻近微管组织中心(MTOC)的胞内体。然后以运输小膜泡的形式,沿着MTOC的微管网络向细胞膜方向转运。跨膜蛋白和基因组在细胞质内的转运受一些宿主因子的调控,如ARHGAP21和小G蛋白Cdc42能够调节NA蛋白向细胞膜转运,Rab11协助vRNPs从MTOC向细胞膜转运。文中主要讨论新合成的流感病毒跨膜蛋白和遗传物质在宿主细胞质内的顺向转运(Anterograde transport)过程与调控。  相似文献   

6.
铜绿假单胞菌(Pseudomonas aeruginosa)Ⅲ型分泌系统(typeⅢsecretion system,TTSS)是重要的细菌致病因子之一,能够将酶蛋白直接注入到宿主细胞内,导致细胞损害的发生。重点研究了TTSS中的popN基因的功能,通过构建popN-突变子,发现该突变子在非诱导条件下,能够分泌酶蛋白,显示popN基因编码的蛋白对TTSS蛋白的分泌具有负调控作用。进一步研究发现,popN-突变子在不同培养基中TTSS的分泌水平存在着显著差异,影响对popN基因的功能的判断。为了解决这一矛盾,从几个方面分析了造成表型差异的可能因素,确定蛋白酶对TTSS分泌蛋白的降解作用,是表型差异存在的主要原因,从而首次系统地阐明popN-突变子在不同培养基中都具有TTSS组成型表达的表型,对于深入研究TTSS的调控机制具有重要意义。  相似文献   

7.
Robert  K  史红艳 《微生物与感染》2005,28(4):48-48
肠致病性大肠埃希菌(EPEC)在肠道细胞的定植可对小肠黏膜细胞产生典型的附着和缺失性(A/E)损伤,其病理改变是局部刷状缘微绒毛的破坏,细菌的紧密黏附及黏附部位细胞骨架的聚集。EPEC通过Ⅲ型分泌系统(TTSS)将效应蛋白运输到宿主细胞内,导致A/E损伤及腹泻。与A/E损伤相关的基因及LEE致病岛编码的效应蛋白包括Tir、  相似文献   

8.
【目的】研究鼠伤寒沙门菌致病岛1(SPI-1)内部的假定调控蛋白STM14_3514的功能及其作用机制。【方法】以鼠伤寒沙门菌模式菌株ATCC 14028为亲本株,构建了STM14_3514基因的缺失突变体及互补菌株,通过小鼠实验、细胞侵袭实验、Western blot及实时荧光定量PCR(q RT-PCR)等实验技术,深入研究了STM14_3514基因对鼠伤寒沙门菌致病过程的影响。【结果】STM14_3514突变提高了细菌对小鼠的致病能力,突变体在小鼠肠道、肝和脾中的定殖能力均增强;细胞实验揭示,突变体致病力提升主要由于STM14_3514突变能显著增强细菌对上皮细胞的侵袭力(2倍,P0.05)。q RT-PCR及Western blot分析表明,STM14_3514显著抑制SPI-1内部主要调控因子hil A及侵袭相关基因的表达。此外,STM14_3514对hil A的抑制由Hil C介导。【结论】STM14_3514是鼠伤寒沙门菌SPI-1内部的负调控因子,能通过Hil C抑制hil A及SPI-1其他入侵基因的表达,该基因的生物学意义可能与细菌进入细胞后对SPI-1的负调控相关。  相似文献   

9.
无论是免疫细胞对病原体的主动吞噬,还是病原体诱导非吞噬细胞的被动吞噬,均是不同细胞膜受体介导的细胞肌动蛋白骨架重排过程,受到单体G蛋白和肌动蛋白骨架相关蛋白的精密调控。细胞内重要信号蛋白,磷脂酰胆碱专一性磷脂酶D(PLD)的活性变化与细胞肌动蛋白骨架重排密切相关,其参与调节了由抗体受体(FcγR)及补体受体(CR3)介导的免疫细胞的主动吞噬,而细胞肌动蛋白骨架解聚蛋白cofilin被磷酸化后可与PLD结合并激活PLD,进而调节肌动蛋白骨架重排。另一方面,cofilin磷酸化状态严格调控李斯特菌感染细胞过程中的肌动蛋白骨架重排。因此,阐明PLD是否在李斯特菌感染细胞过程中被激活并参与调节肌动蛋白骨架重排,将有助于揭示PLD激活对感染发生的调控作用,对透彻理解细菌感染宿主细胞的分子机制具有重要意义。  相似文献   

10.
用荧光定量PCR法检测鼠RAW264.7巨噬细胞感染与未感染鼠伤寒沙门菌后18种铁穗态相关基因的表达,评估宿主与病原体相互作用中铁稳态效应。研究显示,活的鼠伤寒沙门菌感染巨噬细胞1 h后可以诱导转铁蛋白受体表达,引起细胞内动态铁池相关基因的mRNA水平上长。基因表达分析显示,沙门菌通过诱导铁氧还原酶(Steap3)、铁膜转运蛋白(Dmt1)、铁调节因子Tfr2/Hfe以及铁调节蛋白(Irp1和Irp2)的表达主动吸收铁,而经铁转运蛋白(Fpn1)的铁外流并无明显改变。沙门菌在感染后1h积极地驱动了转铁蛋白介导的铁吸收程序。  相似文献   

11.
The Salmonella pathogenicity island 1 (SPI-1) type three secretion system (TTSS) is essential for Salmonella invasion of host cells through its triggering of actin-dependent membrane ruffles. The SPI-1 effectors SipA, SopE, SopE2 and SopB all have actin regulating activities and contribute to invasion. The precise role of actin regulation by SipA in Salmonella invasion remains controversial since divergent data have been presented regarding the relationship between SipA and membrane ruffling. We hypothesized that the contribution of SipA to membrane ruffling and invasion might vary between Salmonella strains. We compared the effects of SipA deletion on Salmonella enterica serovar Typhimurium ( S.  Typhimurium) strains that possess or lack SopE. Loss of SipA reduced invasion in the early stages of infection by SopE+ and SopE- strains but the number of membrane ruffles elicited was unaffected. Salmonella strains lacking both SipA and SopE induced ruffles with very different morphology from those induced by wild-type strains or ones lacking single effectors, including the presence of highly dynamic finger-like protrusions and numerous filopodia. A similar phenotype was found for sipA - sopE -, sipA - sopE2 - and sipA - sopB - mutants. Thus, SipA plays a more prominent role in induction of invasion-competent membrane ruffles by Salmonella lacking a full complement of SPI-1 effectors.  相似文献   

12.
Salmonella enterica serovar Typhimurium encodes two type III secretion systems (TTSSs) within pathogenicity island 1 (SPI-1) and island 2 (SPI-2). These type III protein secretion and translocation systems transport a panel of bacterial effector proteins across both the bacterial and the host cell membranes to promote bacterial entry and subsequent survival inside host cells. Effector proteins contain secretion and translocation signals that are often located at their N termini. We have developed a ruffling-based translocation reporter system that uses the secretion- and translocation-deficient catalytic domain of SopE, SopE78-240, as a reporter. Using this assay, we determined that the N-terminal 45 amino acid residues of Salmonella SopA are necessary and sufficient for directing its secretion and translocation through the SPI-1 TTSS. SopA1-45, but not SopA1-44, is also able to bind to its chaperone, InvB, indicating that SPI-1 type III secretion and translocation of SopA require its chaperone.  相似文献   

13.
A central feature of Salmonella pathogenicity is the bacterium's ability to enter into non-phagocytic cells. Bacterial internalization is the consequence of cellular responses characterized by Cdc42- and Rac-dependent actin cytoskeleton rearrangements. These responses are triggered by the co-ordinated function of bacterial proteins delivered into the host cell by a specialized protein secretion system termed type III. We report here that SopB, a Salmonella inositol polyphosphatase delivered to the host cell by this secretion system, mediates actin cytoskeleton rearrangements and bacterial entry in a Cdc42-dependent manner. SopB exhibits overlapping functions with two other effectors of bacterial entry, the Rho family GTPase exchange factors SopE and SopE2. Thus, Salmonella strains deficient in any one of these proteins can enter into cells at high efficiency, whereas a strain lacking all three effectors is completely defective for entry. Consistent with an important role for inositol phosphate metabolism in Salmonella-induced cellular responses, a catalytically defective mutant of SopB failed to stimulate actin cytoskeleton rearrangements and bacterial entry. Furthermore, bacterial infection of intestinal cells resulted in a marked increase in Ins(1,4,5,6)P4, a consumption of InsP5 and the activation of phospholipase C. In agreement with the in vivo findings, purified SopB specifically dephosphorylated InsP5 to Ins(1,4,5,6)P4 in vitro. Surprisingly, the inositol phosphate fluxes induced by Salmonella were not caused exclusively by SopB. We show that the SopB-independent inositol phosphate fluxes are the consequence of the SopE-dependent activation of an endogenous inositol phosphatase. The ability of Salmonella to stimulate Rho GTPases signalling and inositol phosphate metabolism through alternative mechanisms is an example of the remarkable ability of this bacterial pathogen to manipulate host cellular functions.  相似文献   

14.
Salmonella enterica serovar Typhimurium (S. typhimurium) is a gram-negative facultative intracellular pathogen that can infect a broad range of mammalian hosts. Following invasion of host cells, the majority of S. typhimurium are known to reside in a membrane-bound compartment known as the Salmonella-containing vacuole (SCV). S. typhimurium actively remodels this compartment using bacterial virulence proteins, called effectors, to establish a protected niche where it can replicate. S. typhimurium delivers more than 30 effectors into the host cell cytosol by bacterial type three secretion systems, encoded by Salmonella pathogenicity island 1 or 2 (SPI-1 or SPI-2). Recent studies have revealed a critical role for the SPI-1 effector SopB in 'directing traffic' at early stages of infection, allowing the bacteria to control SCV maturation by modulating its interaction with the endocytic system. At later stages of infection, the SCV establishes a 'nest' near the Golgi where optimal bacterial growth takes place. In this study, we highlight these recent developments in our understanding of SCV trafficking.  相似文献   

15.
Salmonella enterica subspecies 1 serovar Typhimurium encodes a type III secretion system (TTSS) within Salmonella pathogenicity island 1 (SPI-1). This TTSS injects effector proteins into host cells to trigger invasion and inflammatory responses. Effector proteins are recognized by the TTSS via signals encoded in their N termini. Specific chaperones can be involved in this process. The chaperones InvB, SicA, and SicP are encoded in SPI-1 and are required for transport of SPI-1-encoded effectors. Several key effector proteins, like SopE and SopE2, are located outside of SPI-1 but are secreted in an SPI-1-dependent manner. It has not been clear how these effector proteins are recognized by the SPI-1 TTSS. Using pull-down and coimmunoprecipitation assays, we found that SopE is copurified with InvB, the known chaperone for the SPI-1-encoded effector protein Sip/SspA. We also found that InvB is required for secretion and translocation of SopE and SopE2 and for stabilization of SopE2 in the bacterial cytosol. Our data demonstrate that effector proteins encoded within and outside of SPI-1 use the same chaperone for secretion via the SPI-1 TTSS.  相似文献   

16.
Pathogenicity islands (PAIs) are large DNA segments in the genomes of bacterial pathogens that encode virulence factors. Five PAIs have been identified in the Gram-negative bacterium Salmonella enterica. Two of these PAIs, Salmonella pathogenicity island (SPI)-1 and SPI-2, encode type III secretion systems (TTSS), which are essential virulence determinants. These 'molecular syringes' inject effectors directly into the host cell, whereupon they manipulate host cell functions. These effectors are either encoded with their respective TTSS or scattered elsewhere on the Salmonella chromosome. Importantly, SPI-1 and SPI-2 are expressed under distinct environmental conditions: SPI-1 is induced upon initial contact with the host cell, whereas SPI-2 is induced intracellularly. Here, we demonstrate that a single PAI, in this case SPI-5, can encode effectors that are induced by distinct regulatory cues and targeted to different TTSS. SPI-5 encodes the SPI-1 TTSS translocated effector, SigD/SopB. In contrast, we report that the adjacently encoded effector PipB is part of the SPI-2 regulon. PipB is translocated by the SPI-2 TTSS to the Salmonella-containing vacuole and Salmonella-induced filaments. We also show that regions of SPI-5 are not conserved in all Salmonella spp. Although sigD/sopB is present in all Salmonella spp., pipB is not found in Salmonella bongori, which also lacks a functional SPI-2 TTSS. Thus, we demonstrate a functional and regulatory cross-talk between three chromosomal PAIs, SPI-1, SPI-2 and SPI-5, which has significant implications for the evolution and role of PAIs in bacterial pathogenesis.  相似文献   

17.
The hallmark of Salmonella entry into host cells is extensive rearrangements of the host actin cytoskeleton at the site of Salmonella contact with intestinal epithelial cells. SopE, SopE2 and SopB, three type III effectors of Salmonella pathogenicity island 1 (SPI-1), activate the Cdc42 and Rac1 signal transduction pathways to promote these rearrangements. SipA and SipC, two Salmonella type III-secreted actin-binding proteins, directly modulate host actin dynamics to facilitate bacterial uptake. Salmonella-induced actin cytoskeleton rearrangements are therefore the result of the coordinated action of a group of type III-secreted effector proteins.  相似文献   

18.
Maturation and maintenance of the intracellular vacuole in which Salmonella replicates is controlled by virulence proteins including the type III secretion system encoded by Salmonella pathogenicity island 2 (SPI-2). Here, we show that, several hours after bacterial uptake into different host cell types, Salmonella induces the formation of an F-actin meshwork around bacterial vacuoles. This structure is assembled de novo from the cellular G-actin pool in close proximity to the Salmonella vacuolar membrane. We demonstrate that the phenomenon does not require the Inv/Spa type III secretion system or cognate effector proteins, which induce actin polymerization during bacterial invasion, but does require a functional SPI-2 type III secretion system, which plays an important role in intracellular replication and systemic infection in mice. Treatment with actin-depolymerizing agents significantly inhibited intramacrophage replication of wild-type Salmonella typhimurium . Furthermore, after this treatment, wild-type bacteria were released into the host cell cytoplasm, whereas SPI-2 mutant bacteria remained within vacuoles. We conclude that actin assembly plays an important role in the establishment of an intracellular niche that sustains bacterial growth.  相似文献   

19.
Bacterial pathogens have evolved a specialized type III secretion system (T3SS) to translocate virulence effector proteins directly into eukaryotic target cells. Salmonellae deploy effectors that trigger localized actin reorganization to force their own entry into non-phagocytic host cells. Six effectors (SipC, SipA, SopE/2, SopB, SptP) can individually manipulate actin dynamics at the plasma membrane, which acts as a 'signaling hub' during Salmonella invasion. The extent of crosstalk between these spatially coincident effectors remains unknown. Here we describe trans and cisbinary entry effector interplay (BENEFIT) screens that systematically examine functional associations between effectors following their delivery into the host cell. The results reveal extensive ordered synergistic and antagonistic relationships and their relative potency, and illuminate an unexpectedly sophisticated signaling network evolved through longstanding pathogen-host interaction.  相似文献   

20.
Many bacterial pathogens require a type 3 secretion system (T3SS) to establish a niche. Host contact activates bacterial T3SS assembly of a translocon pore in the host plasma membrane. Following pore formation, the T3SS docks onto the translocon pore. Docking establishes a continuous passage that enables the translocation of virulence proteins, effectors, into the host cytosol. Here we investigate the contribution of actin polymerization to T3SS-mediated translocation. Using the T3SS model organism Shigella flexneri, we show that actin polymerization is required for assembling the translocon pore in an open conformation, thereby enabling effector translocation. Opening of the pore channel is associated with a conformational change to the pore, which is dependent upon actin polymerization and a coiled-coil domain in the pore protein IpaC. Analysis of an IpaC mutant that is defective in ruffle formation shows that actin polymerization-dependent pore opening is distinct from the previously described actin polymerization-dependent ruffles that are required for bacterial internalization. Moreover, actin polymerization is not required for other pore functions, including docking or pore protein insertion into the plasma membrane. Thus, activation of the T3SS is a multilayered process in which host signals are sensed by the translocon pore leading to the activation of effector translocation.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号