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 31 毫秒
1.
Yersinia type III machines secrete protein substrates across the bacterial envelope and, following assembly of their secretion needles, transport effector Yops into host cells. According to their destination during type III secretion, early, middle, and late secretion substrates can be distinguished; however, the signals and mechanisms whereby these proteins are recognized and transported by the secretion machine are not understood. Here, we examine several hybrids between secretion substrates and the impassable reporter protein glutathione S-transferase (GST). YscP-GST and YopR-GST blocked type III secretion; however, YscF-, YopD-, YopN-, and LcrV-GST did not. Unlike YopR-GST, which can block type III machines only during their assembly, expression of YscP-GST led to an immediate and complete block of all secretion. The secretion signal of YscP was mapped to its first 10 codons or amino acids; however, YscPΔ2-15-GST, lacking this secretion signal, imposed a partial blockade. YscP-GST copurified with the type III ATPase complex (YscN, YscL, and YscQ) and with YscO, suggesting that the association of specific machine components with the impassable substrate may cause the block in type III secretion.  相似文献   

2.
The Yersinia pestis low-Ca2+ response stimulon is responsible for the environmentally regulated expression and secretion of antihost proteins (V antigen and Yops). We have previously shown that yscO encodes a secreted core component of the Yop secretion (Ysc) mechanism. In this study, we constructed and characterized in-frame deletions in the adjacent gene, yscP, in the yscN-yscU operon. The DeltaP1 mutation, which removed amino acids 246 to 333 of YscP, had no effect on Yop expression or secretion, and the mutant protein, YscP1, was secreted, as was YscP in the parent. In contrast, the DeltaP2 strain expressed and secreted less of each Yop than did the parent under the inductive conditions of 37 degrees C and the absence of Ca2+, with an exception being YopE, which was only minimally affected by the mutation. The YscP2 protein, missing amino acids 57 to 324 of YscP, was expressed but not secreted by the DeltaP2 mutant. The effect of the DeltaP2 mutation was at the level of Yop secretion because YopM and V antigen still showed limited secretion when overproduced in trans. Excess YscP also affected secretion: overexpression of YscP in the parent, in either yscP mutant, or in an lcrG mutant effectively shut off secretion. However, co-overexpression of YscO and YscP had no effect on secretion, and YscP overexpression in an lcrE mutant had little effect on Yop secretion, suggesting that YscP acts, in conjunction with YscO, at the level of secretion control of LcrE at the bacterial surface. These findings place YscP among the growing family of mobile Ysc components that both affect secretion and themselves are secreted by the Ysc.  相似文献   

3.
YopD-like translocator proteins encoded by several Gram-negative bacteria are important for type III secretion-dependent delivery of anti-host effectors into eukaryotic cells. This probably depends on their ability to form pores in the infected cell plasma membrane, through which effectors may gain access to the cell interior. In addition, Yersinia YopD is a negative regulator essential for the control of effector synthesis and secretion. As a prerequisite for this functional duality, YopD may need to establish molecular interactions with other key T3S components. A putative coiled-coil domain and an α-helical amphipathic domain, both situated in the YopD C terminus, may represent key protein-protein interaction domains. Therefore, residues within the YopD C terminus were systematically mutagenized. All 68 mutant bacteria were first screened in a variety of assays designed to identify individual residues essential for YopD function, possibly by providing the interaction interface for the docking of other T3S proteins. Mirroring the effect of a full-length yopD gene deletion, five mutant bacteria were defective for both yop regulatory control and effector delivery. Interestingly, all mutations clustered to hydrophobic amino acids of the amphipathic domain. Also situated within this domain, two additional mutants rendered YopD primarily defective in the control of Yop synthesis and secretion. Significantly, protein-protein interaction studies revealed that functionally compromised YopD variants were also defective in self-oligomerization and in the ability to engage another translocator protein, LcrV. Thus, the YopD amphipathic domain facilitates the formation of YopD/YopD and YopD/LcrV interactions, two critical events in the type III secretion process.  相似文献   

4.
Enteropathogenic Escherichia coli employs a type III secretion system (T3SS) to translocate virulence effector proteins directly into enterocyte host cells, leading to diarrheal disease. The T3SS is encoded within the chromosomal locus of enterocyte effacement (LEE). The function of some of the LEE-encoded proteins remains unknown. Here we investigated the role of the Orf16 protein in T3SS biogenesis and function. An orf16 deletion mutant showed translocator and effector protein secretion profiles different from those of wild-type cells. The orf16 null strain produced T3S structures with abnormally long needles and filaments that caused weak hemolysis of red blood cells. Furthermore, the number of fully assembled T3SSs was also reduced in the orf16 mutant, indicating that Orf16, though not essential, is required for efficient T3SS assembly. Analysis of protein secretion revealed that Orf16 is a T3SS-secreted substrate and regulates the secretion of the inner rod component EscI. Both pulldown and yeast two-hybrid assays showed that Orf16 interacts with the C-terminal domain of an inner membrane component of the secretion apparatus, EscU; the inner rod protein EscI; the needle protein EscF; and the multieffector chaperone CesT. These results suggest that Orf16 regulates needle length and, along with EscU, participates in a substrate specificity switch from early substrates to translocators. Taken together, our results suggest that Orf16 acts as a molecular measuring device in a way similar to that of members of the Yersinia YscP and flagellar FliK protein family. Therefore, we propose that this protein be renamed EscP.  相似文献   

5.
The assembly of the type III secretion injectisome culminates in the formation of the needle. In Yersinia, this step requires not only the needle subunit (YscF), but also the small components YscI, YscO, YscX and YscY. We found that these elements act after the completion of the transmembrane export apparatus. YscX and YscY co-purified with the export apparatus protein YscV, even in the absence of any other protein. YscY-EGFP formed fluorescent spots, suggesting its presence in multiple copies. YscO and YscX were required for export of the early substrates YscF, YscI and YscP, but were only exported themselves after the substrate specificity switch had occurred. Unlike its flagellar homologue FliJ, YscO was not required for the assembly of the ATPase YscN. Finally, we investigated the role of the small proteins in export across the inner membrane. No export of the reporter substrate YscP(1-137) -PhoA into the periplasm was observed in absence of YscI, YscO or YscX, confirming that these proteins are required for export of the first substrates. In contrast, YscP(1-137) -PhoA accumulated in the periplasm in the absence of YscF, suggesting that YscF is not required for the function of the export apparatus, but that its polymerization opens the secretin YscC.  相似文献   

6.
The length of the needle ending the Yersinia Ysc injectisome is determined by YscP, a protein acting as a molecular ruler. In addition, YscP is required for Yop secretion. In the present paper, by a systematic deletion analysis, we localized accurately the region required for Yop secretion between residues 405 and 500. As this C-terminal region of YscP has also been shown to control needle length it probably represents the substrate specificity switch of the machinery. By a bioinformatics analysis, we show that this region has a globular structure, an original alpha/beta fold, a P-x-L-G signature and presumably no catalytic activity. In spite of very limited sequence similarities, this structure is conserved among the proteins that are presumed to control the needle length in many different injectisomes and also among members of the FliK family, which control the flagellar hook length. This region thus represents a new protein domain that we called T3S4 for Type III secretion substrate specificity switch. The T3S4 domain of YscP can be replaced by the T3S4 domain of AscP (Aeromonas salmonicida) or PscP (Pseudomonas aeruginosa) but not by the one from FliK, indicating that in spite of a common global structure, these domains need to fit their partner proteins in the secretion apparatus.  相似文献   

7.
Type III secretion systems (T3SSs) are multiprotein molecular devices used by many Gram-negative bacterial pathogens to translocate effector proteins into eukaryotic cells. A T3SS is also used for protein export in flagellar assembly, which promotes bacterial motility. The two systems are evolutionarily related, possessing highly conserved components in their export apparatuses. Enteropathogenic Escherichia coli (EPEC) employs a T3SS, encoded by genes in the locus of enterocyte effacement (LEE) pathogenicity island, to colonize the human intestine and cause diarrheal disease. In the present work, we investigated the role of the LEE-encoded EscO protein (previously Orf15 or EscA) in T3SS biogenesis. We show that EscO shares similar properties with the flagellar FliJ and the Yersinia YscO protein families. Our findings demonstrate that EscO is essential for secretion of all categories of T3SS substrates. Consistent with its central role in protein secretion, it was found to interact with the ATPase EscN and its negative regulator, EscL, of the export apparatus. Moreover, we show that EscO stimulates EscN enzymatic activity; however, it is unable to upregulate ATP hydrolysis in the presence of EscL. Remarkably, EscO partially restored the swimming defect of a Salmonella flagellar fliJ mutant and was able to stimulate the ATPase activity of FliI. Overall, our data indicate that EscO is the virulence counterpart of the flagellar FliJ protein.  相似文献   

8.
The virulence of a large number of Gram-negative bacterial pathogens depends on the type III secretion (T3S) system, which transports select bacterial proteins into host cells. An essential component of the Yersinia T3S system is YscD, a single-pass inner membrane protein. We report here the 2.52-Å resolution structure of the cytoplasmic domain of YscD, called YscDc. The structure confirms that YscDc consists of a forkhead-associated (FHA) fold, which in many but not all cases specifies binding to phosphothreonine. YscDc, however, lacks the structural properties associated with phosphothreonine binding and thus most likely interacts with partners in a phosphorylation-independent manner. Structural comparison highlighted two loop regions, L3 and L4, as potential sites of interactions. Alanine substitutions at L3 and L4 had no deleterious effects on protein structure or stability but abrogated T3S in a dominant negative manner. To gain insight into the function of L3 and L4, we identified proteins associated with YscD by affinity purification coupled to mass spectrometry. The lipoprotein YscJ was found associated with wild-type YscD, as was the effector YopH. Notably, the L3 and L4 substitution mutants interacted with more YopH than did wild-type YscD. These substitution mutants also interacted with SycH (the specific chaperone for YopH), the putative C-ring component YscQ, and the ruler component YscP, whereas wild-type YscD did not. These results suggest that substitutions in the L3 and L4 loops of YscD disrupted the dissociation of SycH from YopH, leading to the accumulation of a large protein complex that stalled the T3S apparatus.  相似文献   

9.
Comparative genomic studies have identified many proteins that are found only in various Chlamydiae species and exhibit no significant sequence similarity to any protein in organisms that do not belong to this group. The CT670 protein of Chlamydia trachomatis is one of the proteins whose genes are in one of the type III secretion gene clusters but whose cellular functions are not known. CT670 shares several characteristics with the YscO protein of Yersinia pestis, including the neighboring genes, size, charge, and secondary structure, but the structures and/or functions of these proteins remain to be determined. Although a BLAST search with CT670 did not identify YscO as a related protein, our analysis indicated that these two proteins exhibit significant sequence similarity. In this paper, we report that the CT670 crystal, solved at a resolution of 2 Å, consists of a single coiled coil containing just two long helices. Gel filtration and analytical ultracentrifugation studies showed that in solution CT670 exists in both monomeric and dimeric forms and that the monomer predominates at lower protein concentrations. We examined the interaction of CT670 with many type III secretion system-related proteins (viz., CT091, CT665, CT666, CT667, CT668, CT669, CT671, CT672, and CT673) by performing bacterial two-hybrid assays. In these experiments, CT670 was found to interact only with the CT671 protein (YscP homolog), whose gene is immediately downstream of ct670. A specific interaction between CT670 and CT671 was also observed when affinity chromatography pull-down experiments were performed. These results suggest that CT670 and CT671 are putative homologs of the YcoO and YscP proteins, respectively, and that they likely form a chaperone-effector pair.Chlamydiae are obligate intracellular bacteria that infect a variety of eukaryotes, including humans, animals, insects, and free-living amoebae (8, 31, 51). They are highly pathogenic and cause genital tract, ocular, and respiratory infections in humans (9, 55). A key characteristic of Chlamydiae is their biphasic developmental cycle, in which the bacteria alternate between two morphologies: the elementary body and the reticulate body (1, 31). The Chlamydiae species, like many other Gram-negative pathogenic bacteria, contain a type III secretion (T3S) system, which plays a major role in their pathogenicity (4, 7, 44). This key system aids pathogenicity by exporting bacterial proteins, termed effectors, into the host cell via a syringe-like nanomachine called an injectisome (4, 7). Once secreted into the host cell, these effectors may manipulate host cell functions to the advantage of the pathogen (44, 54).Because of the unique chlamydial developmental cycle currently there are no tractable methods for genetic manipulation of these organisms (25, 49). Detailed bioinformatic investigations have identified approximately 200 proteins unique to various taxonomic levels of the Chlamydiae phylum (19, 21). Included in this pool of proteins are proteins whose genes occur in chlamydial T3S system loci (23, 44). Most of the Chlamydiae-specific genes in these loci encode proteins whose functions are unknown or putative and are referred to as Chlamydiae-specific putative T3S-related proteins. These genes include the ct670 and ct671 genes, which are downstream of the gene for the ATPase of the T3S system (ct669) and upstream of yscQ (ct672). CT670 is a Chlamydiales-specific protein with no known homologs (based on BLAST searches) in other species outside this group (21). However, based on similarities in size, charge distribution, and predicted secondary structure, CT670 is thought to be the Chlamydia trachomatis equivalent of YscO, a mobile core component of the T3S system in Yersinia (40). Previous studies of CPn0706, the Chlamydophila pneumoniae homolog of CT670, indicated that this protein was localized in the inclusion in infected cells, but it was not detected in the inclusion membrane or host cytosol, suggesting that it is not a secreted protein (24). Moreover, CT670 is not expected to be a type III secreted effector on the basis of the results obtained by a computational approach that was used to predict type III secreted effectors by comparison of sequences to sequences of known effectors (47). CT671 is a Chlamydiaceae-specific protein with no known homologs in any other species outside this family (21), but it is predicted to be a homolog of YscP, which is the molecular ruler and substrate specificity switch in Yersinia species (2). This protein is secreted by a heterologous T3S system and was predicted to be a T3S effector using the computational approach mentioned above (47, 53). Furthermore, the CCA00037 protein, the CT671 homolog in Chlamydophila caviae, has been visualized in the host cytosol of C. caviae-infected cells using specific antibodies, which provided evidence that it is secreted (53). Although CT670 and CT671 do not appear to be related to YscO and YscP, respectively, on the basis of the results of BLAST searches, it is possible that they have similar roles in the chlamydial T3S system based on their genetic neighborhood and other characteristics noted above. Further, because of their Chlamydiae specificity, they may provide a unique characteristic of the chlamydial T3S system. To examine this possibility, detailed investigations of the three-dimensional (3D) structure of CT670, its self-association properties, and its binding partners were carried out in the present study. Here we report elucidation of the CT670 crystal structure at a resolution of 2.0 Å. CT670 crystallized as a monomer with an elongated two-helix coiled coil. Using analytical ultracentrifugation, CT670 was found to be mostly monomeric in solution, but a dimeric form was also detected. Furthermore, by performing protein-protein interaction studies involving bacterial two-hybrid assays, as well as biochemical experiments, we obtained evidence showing that CT670 interacts specifically with CT671, which would be expected if these proteins have functions similar to those of YscO and YscP, respectively.  相似文献   

10.
Wood SE  Jin J  Lloyd SA 《Journal of bacteriology》2008,190(12):4252-4262
Pathogenic yersiniae utilize a type III secretion system to inject antihost factors, called Yops, directly into the cytosol of eukaryotic cells. The Yops are injected via a needle-like structure, comprising the YscF protein, on the bacterial surface. While the needle is being assembled, Yops cannot be secreted. YscP and YscU switch the substrate specificity of the secretion system to enable Yop export once the needle attains its proper length. Here, we demonstrate that the inner rod protein YscI plays a critical role in substrate specificity switching. We show that YscI is secreted by the type III secretion system and that YscI secretion by a yscP mutant is abnormally elevated. Furthermore, we show that mutations in the cytoplasmic domain of YscU reduce YscI secretion by the yscP null strain. We also demonstrate that mutants expressing one of three forms of YscI (those with mutations Q84A, L87A, and L96A) secrete substantial amounts of Yops yet exhibit severe defects in needle formation. In the absence of YscP, mutants with the same changes in YscI assemble needles but are unable to secrete Yops. Together, these results suggest that the formation of the inner rod, not the needle, is critical for substrate specificity switching and that YscP and YscU exert their effects on substrate export by controlling the secretion of YscI.  相似文献   

11.
Pseudomonas syringae delivers virulence effector proteins into plant cells via an Hrp1 type III secretion system (T3SS). P. syringae pv. tomato DC3000 HrpP has a C-terminal, putative T3SS substrate specificity switch domain, like Yersinia YscP. A ΔhrpP DC3000 mutant could not cause disease in tomato or elicit a hypersensitive response (HR) in tobacco, but the HR could be restored by expression of HrpP in trans. Though HrpP is a relatively divergent protein in the T3SS of different P. syringae pathovars, hrpP from P. syringae pv. syringae 61 and P. syringae pv. phaseolicola 1448A restored HR elicitation and pathogenicity to DC3000 ΔhrpP. HrpP was translocated into Nicotiana benthamiana cells via the DC3000 T3SS when expressed from its native promoter, but it was not secreted in culture. N- and C-terminal truncations of HrpP were tested for their ability to be translocated and to restore HR elicitation activity to the ΔhrpP mutant. No N-terminal truncation completely abolished translocation, implying that HrpP has an atypical T3SS translocation signal. Deleting more than 20 amino acids from the C terminus abolished the ability to restore HR elicitation. HrpP fused to green fluorescent protein was no longer translocated but could restore HR elicitation activity to the ΔhrpP mutant, suggesting that translocation is not essential for the function of HrpP. No T3SS substrates were detectably secreted by DC3000 ΔhrpP except the pilin subunit HrpA, which unexpectedly was secreted poorly. HrpP may function somewhat differently than YscP because the P. syringae T3SS pilus likely varies in length due to differing plant cell walls.Many proteobacterial pathogens use a type III secretion system (T3SS) as their primary mechanism to overcome and infect eukaryotic hosts. T3SSs are complex macromolecular machines that span both the bacterial cell envelope and host cell barriers to deliver proteins, commonly termed effectors, from the bacterial cytoplasm into the host cytoplasm (13, 19). After delivery into the host, effector proteins manipulate host cell function and suppress host defenses, allowing bacterial proliferation and disease development (6, 20). Bacteria that rely on T3SS to cause disease include plant pathogens such as Pseudomonas syringae, Ralstonia solanacearum, Erwinia and Xanthomonas species and animal pathogens in the genera Yersinia, Salmonella, Shigella, Escherichia, and Pseudomonas. While the repertoire of effectors delivered by a given T3SS is unique, the T3SS machinery is more universal (13). T3SS includes a core set of eight conserved proteins. These proteins, which are also conserved in bacterial flagellar biogenesis machines, make up the multiringed base structure, or basal body, that spans the bacterial membranes and cell wall. T3SS machines are also comprised of less-conserved and unique proteins that vary between systems. These include regulatory proteins that orchestrate construction of the machine and the extracellular components that function to translocate effectors across host barriers.The extracellular portion of the T3SS is comprised of the pilus or needle appendage (in plant or animal pathogens, respectively), which acts as a conduit for effector delivery, and the translocon complex, which creates the pore in the host cell membrane. These substructures vary between different T3SSs; presumably these external structures have adapted to allow different bacteria to infect different types of host cells. For Yersinia enterocolitica to infect macrophage cells, the T3SS needle must be a particular length (∼58 nm) to bridge the lipopolysaccharides extending from the bacterial outer membrane and reach the host cell membrane (35). Several other animal pathogens have T3SS needles of a defined length (48). Enteropathogenic Escherichia coli also has an additional extension beyond the needle called the EspA filament that functions to span the mucous layer found outside enterocyte cells (13). In plant pathogens, however, the extracellular gap between a bacterium and a plant cell includes a thick plant cell wall that is variable in width between plant species. Consequently, plant pathogenic Pseudomonas syringae has a pilus that can measure over 1 μm in vitro (25).Another major difference between the T3SS machineries of animal and plant pathogens is their translocon complexes. In animal pathogens, these are typically comprised of three essential proteins, but there is growing evidence that plant pathogen translocons employ diverse, functionally redundant components (28). There is growing interest in understanding the regulatory players that orchestrate the construction of diverse machinery. It is hypothesized that the assembly of the T3SS must involve several tightly regulated steps that allow secretion of the required components, followed by that of effectors upon completion. Of particular interest here is the control of pilus/needle subunit secretion, which is necessary when the pilus/needle is being constructed but would presumably compete with translocon and effector secretion after the T3SS is complete.We study the model plant pathogen P. syringae pv. tomato (Pto) DC3000, the causal agent of bacterial speck of tomato and Arabidopsis thaliana (8). DC3000 has a T3SS that delivers ca. 28 effectors and is essential for pathogenesis (11, 12, 30, 43). The P. syringae T3SS is encoded by hrp and hrc genes (hypersensitive response and pathogenicity/conserved), which are located in a pathogenicity island on the chromosome (4). hrc genes encode the conserved core components present in every T3SS. hrp genes encode T3SS components that are divergent or unique to P. syringae and enterobacterial plant pathogens, which also possess Hrp1 class T3SS (13). In contrast, plant pathogenic Ralstonia and Xanthomonas spp. have Hrp2 class T3SS, as indicated by several different Hrp proteins and distinct regulatory systems.To better understand the T3SS machinery, we previously conducted a survey of the hrp genes of P. syringae pv. syringae (Psy) 61 to complete the inventory of all those encoding proteins capable of traveling the T3SS into plant cells when expressed from a constitutive promoter (39). We hypothesized that these proteins might aid in pilus or translocon construction or regulate the construction process. HrpP was one protein found to be a T3SS substrate and important for secretion and translocation of the model effector AvrPto. Importantly, HrpP is related to a well-studied protein from Yersinia enterocolitica, YscP, which is a T3SS-secreted protein and a regulator responsible for switching the T3SS from secreting needle subunits to secreting effector proteins (15, 38, 47). It has also been shown that secretion of YscP into the culture medium is not essential for the switch function and that there may be two type III secretion signals embedded in YscP (2).The phenotype of a yscP mutant is unregulated secretion of the needle subunit, no secretion of effectors, and production of needles of indeterminate length. The switching phenotype requires a domain at the C terminus of YscP called the type III secretion substrate specificity switch (T3S4) domain, which is a conserved feature unifying its homologs (1). YscP has been proposed to act as a molecular ruler because the length of the YscP protein is directly correlated with the length of the Ysc needle (26). According to this model, when the needle has reached its proper length, YscP signals to the T3SS machinery to stop secreting needle subunits and begin secreting effector proteins. However, other functional models have been hypothesized for homologs of YscP. A recent study of the Salmonella enterica serovar Typhimurium YscP homolog InvJ showed that an invJ mutant lacked an inner rod. When the inner rod protein PrgJ was overexpressed, the length of the needle decreased relative to that of the wild type, leading the researchers to conclude that InvJ controls the inner rod, which in turn controls needle length (33). Recent evidence in Yersinia has lent more support to this model. YscP was found to negatively control secretion of YscI, the inner rod protein (51). Also, certain YscI mutations affected needle assembly but not effector secretion, implying that YscI may be a key player in substrate switching. Little is known about HrpB, the inner rod homolog in P. syringae (22), other than that the protein can be translocated into plant cells and is essential for T3SS function (39).Other models for length control/substrate switching have been proposed, such as the “C-ring cup model” in flagella, which was based on the observation that certain mutations in proteins that make up the inner membrane C ring of the basal body lead to shorter hooks (the flagellar equivalent of the needle), thus suggesting that C-ring capacity controls hook length (32). A more recent, flagellar “molecular-clock” model suggests that because overexpression of hook subunits leads to longer hooks and hook polymerization-defective mutants make shorter hooks, hook polymerization initiates a countdown, and the timing, in cooperation with the YscP homolog FliK, determines final hook length (34).HrpP is considered a member of the YscP/FliK family due mostly to the presence of a T3S4 domain at its C terminus. HrpP is also proline rich (10.6%), which is considered a characteristic of the family. The most striking feature of HrpP is its small size; the protein is 189 amino acids, compared with YscP from Y. enterocolitica, which is 453 amino acids and 8.4% proline. We were intrigued by how HrpP functions in P. syringae to regulate a pilus that can measure several hundred nanometers in length. Also, unlike animal pathogen needles and flagellar hooks, the pilus of P. syringae is predicted to be indeterminate in length, based on the fact that plant cell walls vary in width between species (40).We hypothesized that HrpP would be a main player in regulating pilus construction in P. syringae by allowing the system to make the transition between secretion of pilus subunits and secretion of translocon or effector proteins, though perhaps by a novel mechanism. In this study, we more precisely define the role of HrpP in the P. syringae T3SS. We show that HrpP is a T3SS substrate in DC3000, is translocated into plant cells at levels equivalent to those of effectors, and is essential for the function of the T3SS. Though it is highly translocated and variable, we found that HrpP from different P. syringae pathovars could complement the DC3000 hrpP mutant. Analysis of truncations of HrpP and an impassible HrpP-green fluorescent protein (GFP) fusion suggests that it has structural similarities to YscP, but surprisingly, HrpP was found to be required for full secretion of the pilus subunit HrpA as well as for translocation of HrpB.  相似文献   

12.
Many Gram-negative bacteria use a type III secretion (T3S) system to directly inject effector molecules into eucaryotic cells in order to establish a symbiotic or pathogenic relationship with their host. The translocation of many T3S proteins requires specialized chaperones from the bacterial cytosol. SycD belongs to a class of T3S chaperones that assists the secretion of pore-forming translocators and, specifically chaperones the translocators YopB and YopD from enteropathogenic Yersinia enterocolitica. In addition, SycD is involved in the regulation of virulence factor biosynthesis and secretion. In this study, we present two crystal structures of Y. enterocolitica SycD at 1.95 and 2.6 Å resolution, the first experimental structures of a T3S class II chaperone specific for translocators. The fold of SycD is entirely α-helical and reveals three tetratricopeptide repeat-like motifs that had been predicted from amino acid sequence. In both structures, SycD forms dimers utilizing residues from the first tetratricopeptide repeat motif. Using site-directed mutagenesis and size exclusion chromatography, we verified that SycD forms head-to-head homodimers in solution. Although in both structures, dimerization largely depends on the same residues, the two assemblies represent alternative dimers that exhibit different monomer orientations and overall shape. In these two distinct head-to-head dimers, both the concave and the convex surface of each monomer are accessible for interactions with the SycD binding partners YopB and YopD. A SycD variant carrying two point mutations in the dimerization interface is properly folded but defective in dimerization. Expression of this stable SycD monomer in Yersinia does not rescue the phenotype of a sycD null mutant, suggesting a physiological relevance of the dimerization interface.  相似文献   

13.
A common virulence mechanism among bacterial pathogens is the use of specialized secretion systems that deliver virulence proteins through a translocation channel inserted in the host cell membrane. During Yersinia infection, the host recognizes the type III secretion system mounting a pro-inflammatory response. However, soon after they are translocated, the effectors efficiently counteract that response. In this study we sought to identify YopD residues responsible for type III secretion system function. Through random mutagenesis, we identified eight Y. pseudotuberculosis yopD mutants with single amino acid changes affecting various type III secretion functions. Three severely defective mutants had substitutions in residues encompassing a 35 amino acid region (residues 168–203) located between the transmembrane domain and the C-terminal putative coiled-coil region of YopD. These mutations did not affect regulation of the low calcium response or YopB-YopD interaction but markedly inhibited MAPK and NFκB activation. When some of these mutations were introduced into the native yopD gene, defects in effector translocation and pore formation were also observed. We conclude that this newly identified region is important for YopD translocon function. The role of this domain in vivo remains elusive, as amino acid substitutions in that region did not significantly affect virulence of Y. pseudotuberculosis in orogastrically-infected mice.  相似文献   

14.
The switch in export specificity of the type III flagellar protein export apparatus from rod/hook type to filament type is believed to occur upon completion of hook assembly by way of an interaction of the type III secretion substrate specificity switch (T3S4) domain of the hook-length control protein FliK, with the integral membrane export apparatus component FlhB. The T3S4 domain of FliK (FliKT3S4) consisting of amino acid residues 265-405 has an unstable and flexible conformation in its last 35 residues (FliKCT). To investigate the role of FliKT3S4 in substrate specificity switching, we studied the effect of deletions and point mutations within this domain and characterized suppressor mutations. Deletions of ten amino acid residues within the region of residues 301-350 and five amino acids of residues 401-405 abolished switching of export specificity. Site directed mutagenesis showed that highly conserved residues, Val302, Ile304, Leu335, Val401 and Ala405, are essential, and that the five C terminal residues (401-405) are restricted in conformation for the switching process. Suppressor mutant analysis of the fliK(S319Y) mutant, which produces extended hooks with filaments attached due to delayed switching, suggested that FliKT3S4 interacts with the C terminal half of the cytoplasmic domain of FlhB (FlhBC). We propose a two step binding model of FliKT3S4 and FlhBC, in which residues 301-350 of FliK bind to FlhBC upon hook assembly completion at about 55 nm, and then unfolded FliKCT binds to FlhBC to trigger the switch in substrate specificity.  相似文献   

15.
Pathogenic Yersinia species use a type III secretion system to inhibit phagocytosis by eukaryotic cells. At 37 degrees C, the secretion system is assembled, forming a needle-like structure on the bacterial cell surface. Upon eukaryotic cell contact, six effector proteins, called Yops, are translocated into the eukaryotic cell cytosol. Here, we show that a yscP mutant exports an increased amount of the needle component YscF to the bacterial cell surface but is unable to efficiently secrete effector Yops. Mutations in the cytoplasmic domain of the inner membrane protein YscU suppress the yscP phenotype by reducing the level of YscF secretion and increasing the level of Yop secretion. These results suggest that YscP and YscU coordinately regulate the substrate specificity of the Yersinia type III secretion system. Furthermore, we show that YscP and YscU act upstream of the cell contact sensor YopN as well as the inner gatekeeper LcrG in the pathway of substrate export regulation. These results further strengthen the strong evolutionary link between flagellar biosynthesis and type III synthesis.  相似文献   

16.
17.
YscU, a component of the Yersinia type III secretion machine, promotes auto-cleavage at asparagine 263 (N263). Mutants with an alanine substitution at yscU codon 263 displayed secretion defects for some substrates (LcrV, YopB and YopD); however, transport of effector proteins into host cells (YopE, YopH, YopM) continued to occur. Two yscU mutations were isolated that, unlike N263A , completely abolished type III secretion; YscUG127D promoted auto-cleavage at N263, whereas YscUG270N did not. When fused to glutathione S-transferase (Gst), the YscU C-terminal cytoplasmic domain promoted auto-cleavage and Gst-YscUC also exerted a dominant-negative phenotype by blocking type III secretion. Gst–YscUC/N263A caused a similar blockade and Gst–YscUC/G270N reduced secretion. Gst–YscUC and Gst–YscUC/N263A bound YscL, the regulator of the ATPase YscN, whereas Gst–YscUC/G270N did not. When isolated from Yersinia , Gst–YscUC and Gst–YscUC/N263A associated with YscK–YscL–YscQ; however, Gst–YscUC/G270N interacted predominantly with the machine component YscO, but not with YscK–YscL–YscQ. A model is proposed whereby YscU auto-cleavage promotes interaction with YscL and recruitment of ATPase complexes that initiate type III secretion.  相似文献   

18.
The plant-pathogenic bacterium Xanthomonas campestris pv. vesicatoria employs a type III secretion (T3S) system to translocate bacterial effector proteins into eukaryotic host cells. The membrane-spanning secretion apparatus consists of 11 core components and several associated proteins with yet unknown functions. In this study, we analyzed the role of HrpB1, which was previously shown to be essential for T3S and the formation of the extracellular T3S pilus. We provide experimental evidence that HrpB1 localizes to the bacterial periplasm and binds to peptidoglycan, which is in agreement with its predicted structural similarity to the putative peptidoglycan-binding domain of the lytic transglycosylase Slt70 from Escherichia coli. Interaction studies revealed that HrpB1 forms protein complexes and binds to T3S system components, including the inner membrane protein HrcD, the secretin HrcC, the pilus protein HrpE, and the putative inner rod protein HrpB2. The analysis of deletion and point mutant derivatives of HrpB1 led to the identification of amino acid residues that contribute to the interaction of HrpB1 with itself and HrcD and/or to protein function. The finding that HrpB1 and HrpB2 colocalize to the periplasm and both interact with HrcD suggests that they are part of a periplasmic substructure of the T3S system.  相似文献   

19.
Human pathogenic Yersinia species share a virulence plasmid encoding the Ysc‐Yop type III secretion system (T3SS). A plasmid‐encoded anti‐activator, LcrQ, negatively regulates the expression of this secretion system. Under inducible conditions, LcrQ is secreted outside of bacterial cells and this activates the T3SS, but the mechanism of targeting LcrQ for type III secretion remains largely unknown. In this study, we characterized the regulatory role of the export apparatus component YscV. Depletion or overexpression of YscV compromised Yop synthesis and this primarily prevented secretion of LcrQ. It followed that a lcrQ deletion reversed the repressive effects of excessive YscV. Further characterization demonstrated that the YscV residues 493–511 located within the C‐terminal soluble cytoplasmic domain directly bound with LcrQ. Critically, YscV‐LcrQ complex formation was a requirement for LcrQ secretion, since YscVΔ493–511 failed to secrete LcrQ. This forced a cytoplasmic accumulation of LcrQ, which predictably caused the feedback inhibition of Yops synthesis. Based on these observations, we proposed a model for the YscV‐dependent secretion of LcrQ and its role in regulating Yop synthesis in Yersinia.  相似文献   

20.
The innate immune system is the first line of defense against invading pathogens. Innate immune cells recognize molecular patterns from the pathogen and mount a response to resolve the infection. The production of proinflammatory cytokines and reactive oxygen species, phagocytosis, and induced programmed cell death are processes initiated by innate immune cells in order to combat invading pathogens. However, pathogens have evolved various virulence mechanisms to subvert these responses. One strategy utilized by Gram-negative bacterial pathogens is the deployment of a complex machine termed the type III secretion system (T3SS). The T3SS is composed of a syringe-like needle structure and the effector proteins that are injected directly into a target host cell to disrupt a cellular response. The three human pathogenic Yersinia spp. (Y. pestis, Y. enterocolitica, and Y. pseudotuberculosis) are Gram-negative bacteria that share in common a 70 kb virulence plasmid which encodes the T3SS. Translocation of the Yersinia effector proteins (YopE, YopH, YopT, YopM, YpkA/YopO, and YopP/J) into the target host cell results in disruption of the actin cytoskeleton to inhibit phagocytosis, downregulation of proinflammatory cytokine/chemokine production, and induction of cellular apoptosis of the target cell. Over the past 25 years, studies on the Yersinia effector proteins have unveiled tremendous knowledge of how the effectors enhance Yersinia virulence. Recently, the long awaited crystal structure of YpkA has been solved providing further insights into the activation of the YpkA kinase domain. Multisite autophosphorylation by YpkA to activate its kinase domain was also shown and postulated to serve as a mechanism to bypass regulation by host phosphatases. In addition, novel Yersinia effector protein targets, such as caspase-1, and signaling pathways including activation of the inflammasome were identified. In this review, we summarize the recent discoveries made on Yersinia effector proteins and their contribution to Yersinia pathogenesis.  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