Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Curr Opin Cell Biol ; 79: 102131, 2022 12.
Article in English | MEDLINE | ID: mdl-36215855

ABSTRACT

Subversion of the host cell cytoskeleton is a virulence attribute common to many bacterial pathogens. On mucosal surfaces, bacteria have evolved distinct ways of interacting with the polarised epithelium and manipulating host cell structure to propagate infection. For example, Shigella and Listeria induce cytoskeletal changes to induce their own uptake into enterocytes in order to replicate within an intracellular environment and then spread from cell-to-cell by harnessing the host actin cytoskeleton. In this review, we highlight some recent studies that advance our understanding of the role of the host cell cytoskeleton in the mechanical and molecular processes of pathogen invasion, cell-to-cell spread and the impact of infection on epithelial intercellular tension and innate mucosal defence.


Subject(s)
Listeria , Shigella , Cytoskeleton/metabolism , Epithelial Cells , Bacteria , Host-Pathogen Interactions
2.
Proc Natl Acad Sci U S A ; 119(34): e2204332119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35976880

ABSTRACT

Attaching and effacing (AE) lesion formation on enterocytes by enteropathogenic Escherichia coli (EPEC) requires the EPEC type III secretion system (T3SS). Two T3SS effectors injected into the host cell during infection are the atypical kinases, NleH1 and NleH2. However, the host targets of NleH1 and NleH2 kinase activity during infection have not been reported. Here phosphoproteomics identified Ser775 in the microvillus protein Eps8 as a bona fide target of NleH1 and NleH2 phosphorylation. Both kinases interacted with Eps8 through previously unrecognized, noncanonical "proline-rich" motifs, PxxDY, that bound the Src Homology 3 (SH3) domain of Eps8. Structural analysis of the Eps8 SH3 domain bound to a peptide containing one of the proline-rich motifs from NleH showed that the N-terminal part of the peptide adopts a type II polyproline helix, and its C-terminal "DY" segment makes multiple contacts with the SH3 domain. Ser775 phosphorylation by NleH1 or NleH2 hindered Eps8 bundling activity and drove dispersal of Eps8 from the AE lesion during EPEC infection. This finding suggested that NleH1 and NleH2 altered the cellular localization of Eps8 and the cytoskeletal composition of AE lesions during EPEC infection.


Subject(s)
Adaptor Proteins, Signal Transducing , Enteropathogenic Escherichia coli , Escherichia coli Infections , Escherichia coli Proteins , Phosphotransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Humans , Microvilli/metabolism , Phosphorylation , Phosphotransferases/metabolism
3.
Cell Host Microbe ; 29(10): 1473-1476, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34648738

ABSTRACT

Shigella is a highly infectious human pathogen, yet mice are naturally resistant to infection. In this issue of Cell Host & Microbe, Luchetti et al. (2021) discuss this species specificity, demonstrating that Shigella directly targets the pore-forming protein Gasdermin D for degradation, thus preventing pyroptosis to enable infection of human cells.


Subject(s)
Intracellular Signaling Peptides and Proteins , Shigella , Animals , Mice , Phosphate-Binding Proteins , Pyroptosis
4.
PLoS Pathog ; 17(6): e1009658, 2021 06.
Article in English | MEDLINE | ID: mdl-34133469

ABSTRACT

During infection, enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC) directly manipulate various aspects of host cell function through the translocation of type III secretion system (T3SS) effector proteins directly into the host cell. Many T3SS effector proteins are enzymes that mediate post-translational modifications of host proteins, such as the glycosyltransferase NleB1, which transfers a single N-acetylglucosamine (GlcNAc) to arginine residues, creating an Arg-GlcNAc linkage. NleB1 glycosylates death-domain containing proteins including FADD, TRADD and RIPK1 to block host cell death. The NleB1 paralogue, NleB2, is found in many EPEC and EHEC strains but to date its enzymatic activity has not been described. Using in vitro glycosylation assays combined with mass spectrometry, we found that NleB2 can utilize multiple sugar donors including UDP-glucose, UDP-GlcNAc and UDP-galactose during glycosylation of the death domain protein, RIPK1. Sugar donor competition assays demonstrated that UDP-glucose was the preferred substrate of NleB2 and peptide sequencing identified the glycosylation site within RIPK1 as Arg603, indicating that NleB2 catalyses arginine glucosylation. We also confirmed that NleB2 catalysed arginine-hexose modification of Flag-RIPK1 during infection of HEK293T cells with EPEC E2348/69. Using site-directed mutagenesis and in vitro glycosylation assays, we identified that residue Ser252 in NleB2 contributes to the specificity of this distinct catalytic activity. Substitution of Ser252 in NleB2 to Gly, or substitution of the corresponding Gly255 in NleB1 to Ser switches sugar donor preference between UDP-GlcNAc and UDP-glucose. However, this switch did not affect the ability of the NleB variants to inhibit inflammatory or cell death signalling during HeLa cell transfection or EPEC infection. NleB2 is thus the first identified bacterial Arg-glucose transferase that, similar to the NleB1 Arg-GlcNAc transferase, inhibits host protein function by arginine glycosylation.


Subject(s)
Arginine/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Glucose/metabolism , Glycosyltransferases/metabolism , Virulence Factors/metabolism , Cell Line , Humans
5.
Methods Mol Biol ; 2291: 317-332, 2021.
Article in English | MEDLINE | ID: mdl-33704761

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) and the related pathogen enteropathogenic Escherichia coli (EPEC) use a type III secretion system to translocate effector proteins into host cells to modulate inflammatory signaling pathways during infection. Here we describe the procedures to investigate effector-driven modulation of host inflammatory signaling pathways in mammalian cells where bacterial effectors are ectopically expressed or in cell lines infected with STEC or EPEC. We focus on the TNF-induced NF-κB response by examining IκBα degradation by immunoblot and p65 nuclear localization in addition to using an NF-κB-dependent luciferase reporter and cytokine secretion assays. These methods can be adapted for examining effector-mediated modulation of other inflammatory stimuli and host signaling pathways.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Animals , Cell Line , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/pathology , Humans , Inflammation/metabolism , Inflammation/microbiology , NF-KappaB Inhibitor alpha/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity
6.
Curr Opin Microbiol ; 59: 58-64, 2021 02.
Article in English | MEDLINE | ID: mdl-32862049

ABSTRACT

Many Gram-negative enteric pathogens, including enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC), Salmonella, Shigella, and Yersinia species have evolved strategies to combat host defence mechanisms. Critical bacterial virulence factors, which often include but are not limited to type III secreted effector proteins, are deployed to cooperatively interfere with key host defence pathways. Recent studies in this area have not only contributed to our knowledge of bacterial pathogenesis, but have also shed light on the host pathways that are critical for controlling bacterial infection. In this review, we summarise recent breakthroughs in our understanding of the mechanisms utilised by enteric bacterial pathogens to rewire critical host innate immune responses, including cell death and inflammatory signaling and cell-intrinsic anti-microbial responses such as xenophagy.


Subject(s)
Gastrointestinal Microbiome , Host-Pathogen Interactions , Immunity, Innate , Animals , Cell Death , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , Signal Transduction , Virulence Factors/genetics , Virulence Factors/metabolism
7.
Article in English | MEDLINE | ID: mdl-32974215

ABSTRACT

During infection, Salmonella species inject multiple type III secretion system (T3SS) effector proteins into host cells that mediate invasion and subsequent intracellular replication. At early stages of infection, Salmonella exploits key regulators of host intracellular vesicle transport, including the small GTPases Rab5 and Rab7, to subvert host endocytic vesicle trafficking and establish the Salmonella-containing vacuole (SCV). At later stages of intracellular replication, interactions of the SCV with Rab GTPases are less well defined. Here we report that Rab1, Rab5, and Rab11 are modified at later stages of Salmonella infection by SseK3, an arginine N-acetylglucosamine (GlcNAc) transferase effector translocated via the Salmonella pathogenicity island 2 (SPI-2) type III secretion system. SseK3 modified arginines at positions 74, 82, and 111 within Rab1 and this modification occurred independently of Rab1 nucleotide binding. SseK3 exhibited Golgi localization that was independent of its glycosyltransferase activity but Arg-GlcNAc transferase activity was required for inhibition of alkaline phosphatase secretion in transfected cells. While SseK3 had a modest effect on SEAP secretion during infection of HeLa229 cells, inhibition of IL-1 and GM-CSF cytokine secretion was only observed upon over-expression of SseK3 during infection of RAW264.7 cells. Our results suggest that, in addition to targeting death receptor signaling, SseK3 may contribute to Salmonella infection by interfering with the activity of key Rab GTPases.


Subject(s)
Salmonella Infections , rab GTP-Binding Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , HeLa Cells , Humans , Salmonella/metabolism
8.
Immunity ; 50(3): 539-541, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893581

ABSTRACT

Immunometabolism is emerging as an important modulator of immune responses. In this issue of Immunity, Li et al. (2019) examine the link between lipopolysaccharide (LPS)-induced glucose metabolism and innate immune signaling and identify how ß-N-acetylglucosamine (O-GlcNAc) modification of the RIPK3 RHIM domain limits inflammation and necroptosis.


Subject(s)
Inflammation , Sugars , Humans , N-Acetylglucosaminyltransferases , Receptor-Interacting Protein Serine-Threonine Kinases , Serine , Threonine
9.
Mol Cell Proteomics ; 18(6): 1138-1156, 2019 06.
Article in English | MEDLINE | ID: mdl-30902834

ABSTRACT

Strains of Salmonella utilize two distinct type three secretion systems to deliver effector proteins directly into host cells. The Salmonella effectors SseK1 and SseK3 are arginine glycosyltransferases that modify mammalian death domain containing proteins with N-acetyl glucosamine (GlcNAc) when overexpressed ectopically or as recombinant protein fusions. Here, we combined Arg-GlcNAc glycopeptide immunoprecipitation and mass spectrometry to identify host proteins GlcNAcylated by endogenous levels of SseK1 and SseK3 during Salmonella infection. We observed that SseK1 modified the mammalian signaling protein TRADD, but not FADD as previously reported. Overexpression of SseK1 greatly broadened substrate specificity, whereas ectopic co-expression of SseK1 and TRADD increased the range of modified arginine residues within the death domain of TRADD. In contrast, endogenous levels of SseK3 resulted in modification of the death domains of receptors of the mammalian TNF superfamily, TNFR1 and TRAILR, at residues Arg376 and Arg293 respectively. Structural studies on SseK3 showed that the enzyme displays a classic GT-A glycosyltransferase fold and binds UDP-GlcNAc in a narrow and deep cleft with the GlcNAc facing the surface. Together our data suggest that salmonellae carrying sseK1 and sseK3 employ the glycosyltransferase effectors to antagonise different components of death receptor signaling.


Subject(s)
Bacterial Proteins/metabolism , Salmonella/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acetylglucosamine/metabolism , Animals , Bacterial Proteins/chemistry , Conserved Sequence , Glutamic Acid/metabolism , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mutagenesis , Mutation/genetics , Protein Domains , RAW 264.7 Cells , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Substrate Specificity , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/metabolism
10.
J Biol Chem ; 292(42): 17337-17350, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28860194

ABSTRACT

The inhibition of host innate immunity pathways is essential for the persistence of attaching and effacing pathogens such as enteropathogenic Escherichia coli (EPEC) and Citrobacter rodentium during mammalian infections. To subvert these pathways and suppress the antimicrobial response, attaching and effacing pathogens use type III secretion systems to introduce effectors targeting key signaling pathways in host cells. One such effector is the arginine glycosyltransferase NleB1 (NleBCR in C. rodentium) that modifies conserved arginine residues in death domain-containing host proteins with N-acetylglucosamine (GlcNAc), thereby blocking extrinsic apoptosis signaling. Ectopically expressed NleB1 modifies the host proteins Fas-associated via death domain (FADD), TNFRSF1A-associated via death domain (TRADD), and receptor-interacting serine/threonine protein kinase 1 (RIPK1). However, the full repertoire of arginine GlcNAcylation induced by pathogen-delivered NleB1 is unknown. Using an affinity proteomic approach for measuring arginine-GlcNAcylated glycopeptides, we assessed the global profile of arginine GlcNAcylation during ectopic expression of NleB1, EPEC infection in vitro, or C. rodentium infection in vivo NleB overexpression resulted in arginine GlcNAcylation of multiple host proteins. However, NleB delivery during EPEC and C. rodentium infection caused rapid and preferential modification of Arg117 in FADD. This FADD modification was extremely stable and insensitive to physiological temperatures, glycosidases, or host cell degradation. Despite its stability and effect on the inhibition of apoptosis, arginine GlcNAcylation did not elicit any proteomic changes, even in response to prolonged NleB1 expression. We conclude that, at normal levels of expression during bacterial infection, NleB1/NleBCR antagonizes death receptor-induced apoptosis of infected cells by modifying FADD in an irreversible manner.


Subject(s)
Apoptosis , Citrobacter rodentium/enzymology , Enteropathogenic Escherichia coli/enzymology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Fas-Associated Death Domain Protein/metabolism , Glycosyltransferases/metabolism , Protein Processing, Post-Translational , Virulence Factors/metabolism , Citrobacter rodentium/pathogenicity , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/pathology , Escherichia coli Proteins/genetics , Fas-Associated Death Domain Protein/genetics , Glycosyltransferases/genetics , HeLa Cells , Humans , Protein Stability , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/genetics , TNF Receptor-Associated Death Domain Protein/metabolism , Virulence Factors/genetics
12.
Infect Immun ; 85(4)2017 04.
Article in English | MEDLINE | ID: mdl-28138023

ABSTRACT

During infection, enteropathogenic Escherichia coli (EPEC) translocates effector proteins directly into the cytosol of infected enterocytes using a type III secretion system (T3SS). Once inside the host cell, these effector proteins subvert various immune signaling pathways, including death receptor-induced apoptosis. One such effector protein is the non-locus of enterocyte effacement (LEE)-encoded effector NleB1, which inhibits extrinsic apoptotic signaling via the FAS death receptor. NleB1 transfers a single N-acetylglucosamine (GlcNAc) residue to Arg117 in the death domain of Fas-associated protein with death domain (FADD) and inhibits FAS ligand (FasL)-stimulated caspase-8 cleavage. Another effector secreted by the T3SS is NleF. Previous studies have shown that NleF binds to and inhibits the activity of caspase-4, -8, and -9 in vitro Here, we investigated a role for NleF in the inhibition of FAS signaling and apoptosis during EPEC infection. We show that NleF prevents the cleavage of caspase-8, caspase-3, and receptor-interacting serine/threonine protein kinase 1 (RIPK1) in response to FasL stimulation. When translocated into host cells by the T3SS or expressed ectopically, NleF also blocked FasL-induced cell death. Using the EPEC-like mouse pathogen Citrobacter rodentium, we found that NleB but not NleF contributed to colonization of mice in the intestine. Hence, despite their shared ability to block FasL/FAS signaling, NleB and NleF have distinct roles during infection.


Subject(s)
Apoptosis , Enteropathogenic Escherichia coli/physiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Virulence Factors/metabolism , Caspases/metabolism , Cell Line , Ectopic Gene Expression , Escherichia coli Proteins/genetics , Fas Ligand Protein/metabolism , Genetic Complementation Test , HEK293 Cells , HeLa Cells , Humans , Mutation , Signal Transduction , Virulence Factors/genetics , fas Receptor/metabolism
13.
Nat Microbiol ; 2: 16258, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28085133

ABSTRACT

Cell death signalling pathways contribute to tissue homeostasis and provide innate protection from infection. Adaptor proteins such as receptor-interacting serine/threonine-protein kinase 1 (RIPK1), receptor-interacting serine/threonine-protein kinase 3 (RIPK3), TIR-domain-containing adapter-inducing interferon-ß (TRIF) and Z-DNA-binding protein 1 (ZBP1)/DNA-dependent activator of IFN-regulatory factors (DAI) that contain receptor-interacting protein (RIP) homotypic interaction motifs (RHIM) play a key role in cell death and inflammatory signalling1-3. RHIM-dependent interactions help drive a caspase-independent form of cell death termed necroptosis4,5. Here, we report that the bacterial pathogen enteropathogenic Escherichia coli (EPEC) uses the type III secretion system (T3SS) effector EspL to degrade the RHIM-containing proteins RIPK1, RIPK3, TRIF and ZBP1/DAI during infection. This requires a previously unrecognized tripartite cysteine protease motif in EspL (Cys47, His131, Asp153) that cleaves within the RHIM of these proteins. Bacterial infection and/or ectopic expression of EspL leads to rapid inactivation of RIPK1, RIPK3, TRIF and ZBP1/DAI and inhibition of tumour necrosis factor (TNF), lipopolysaccharide or polyinosinic:polycytidylic acid (poly(I:C))-induced necroptosis and inflammatory signalling. Furthermore, EPEC infection inhibits TNF-induced phosphorylation and plasma membrane localization of mixed lineage kinase domain-like pseudokinase (MLKL). In vivo, EspL cysteine protease activity contributes to persistent colonization of mice by the EPEC-like mouse pathogen Citrobacter rodentium. The activity of EspL defines a family of T3SS cysteine protease effectors found in a range of bacteria and reveals a mechanism by which gastrointestinal pathogens directly target RHIM-dependent inflammatory and necroptotic signalling pathways.


Subject(s)
Apoptosis , Escherichia coli Proteins/metabolism , Inflammation , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death , Citrobacter rodentium/pathogenicity , Cysteine Proteases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enteropathogenic Escherichia coli/enzymology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/genetics , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Mice , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Type III Secretion Systems
14.
Infect Immun ; 85(2)2017 02.
Article in English | MEDLINE | ID: mdl-27872241

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) is a gastrointestinal pathogen that utilizes a type III secretion system (T3SS) to inject an array of virulence effector proteins into host enterocytes to subvert numerous cellular processes for successful colonization and dissemination. The T3SS effector NleD is a 26-kDa zinc metalloprotease that is translocated into host enterocytes, where it directly cleaves and inactivates the mitogen-activated protein kinase signaling proteins JNK and p38. Here a library of 91 random transposon-based, in-frame, linker insertion mutants of NleD were tested for their ability to cleave JNK and p38 during transient transfection of cultured epithelial cells. Immunoblot analysis of p38 and JNK cleavage showed that 7 mutant derivatives of NleD no longer cleaved p38 but maintained the ability to cleave JNK. Site-directed mutation of specific regions surrounding the insertion sites within NleD revealed that a single amino acid, R203, was essential for cleavage of p38 but not JNK in a direct in vitro cleavage assay, in transiently transfected cells, or in EPEC-infected cells. Mass spectrometry analysis narrowed the cleavage region to within residues 187 and 213 of p38. Mutation of residue R203 within NleD to a glutamate residue abolished the cleavage of p38 and impaired the ability of NleD to inhibit AP-1-dependent gene transcription of a luciferase reporter. Furthermore, the R203 mutation abrogated the ability of NleD to dampen interleukin-6 production in EPEC-infected cells. Overall, this work provides greater insight into substrate recognition and specificity by the type III effector NleD.


Subject(s)
Enteropathogenic Escherichia coli/physiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Host-Pathogen Interactions , JNK Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Sequence , Arginine/metabolism , Cell Line , Cytokines/biosynthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Humans , Mutagenesis, Insertional , Proteolysis , Signal Transduction
15.
Annu Rev Genet ; 50: 493-513, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27893961

ABSTRACT

In many parts of the world, enteropathogenic Escherichia coli (EPEC) are a leading cause of death in children with diarrhea. Much of what we know about the pathogenesis of EPEC infections is based on the study of one or two prototypic strains that have provided deep insight into the precise mechanisms by which EPEC colonizes the intestine, evades host immunity, and spreads from person to person. In some cases, defining the biochemical activity of the host-interacting effector proteins from these prototypic strains has led to the discovery of novel post-translational protein modifications and new understandings of biology and host-pathogen interactions. However, genomic analysis of recent EPEC isolates has revealed that the EPEC pathotype is more diverse than previously appreciated. Although by definition all strains carry the locus of enterocyte effacement, the effector repertoires of different clonal groups are quite divergent, suggesting that there is still a great deal to learn about the genetic basis of EPEC virulence.


Subject(s)
Diarrhea/microbiology , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Apoptosis , Enteropathogenic Escherichia coli/immunology , Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Humans , Immune Evasion , Inflammasomes , Phagocytosis , Virulence/genetics
16.
Infect Immun ; 84(5): 1346-1360, 2016 05.
Article in English | MEDLINE | ID: mdl-26883593

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) interferes with host cell signaling by injecting virulence effector proteins into enterocytes via a type III secretion system (T3SS). NleB1 is a novel T3SS glycosyltransferase effector from EPEC that transfers a single N-acetylglucosamine (GlcNAc) moiety in an N-glycosidic linkage to Arg(117) of the Fas-associated death domain protein (FADD). GlcNAcylation of FADD prevents the assembly of the canonical death-inducing signaling complex and inhibits Fas ligand (FasL)-induced cell death. Apart from the DXD catalytic motif of NleB1, little is known about other functional sites in the enzyme. In the present study, members of a library of 22 random transposon-based, in-frame, linker insertion mutants of NleB1 were tested for their ability to block caspase-8 activation in response to FasL during EPEC infection. Immunoblot analysis of caspase-8 cleavage showed that 17 mutant derivatives of NleB1, including the catalytic DXD mutant, did not inhibit caspase-8 activation. Regions of interest around the insertion sites with multiple or single amino acid substitutions were examined further. Coimmunoprecipitation studies of 34 site-directed mutants showed that the NleB1 derivatives with the E253A, Y219A, and PILN(63-66)AAAA (in which the PILN motif from residues 63 to 66 was changed to AAAA) mutations bound to but did not GlcNAcylate FADD. A further mutant derivative, the PDG(236-238)AAA mutant, did not bind to or GlcNAcylate FADD. Infection of mice with the EPEC-like mouse pathogen Citrobacter rodentium expressing NleBE253A and NleBY219A showed that these strains were attenuated, indicating the importance of residues E253 and Y219 in NleB1 virulence in vivo In summary, we identified new amino acid residues critical for NleB1 activity and confirmed that these are required for the virulence function of NleB1.


Subject(s)
DNA Mutational Analysis , Enteropathogenic Escherichia coli/enzymology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Apoptosis , Arginine/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , DNA Transposable Elements , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Female , Humans , Mice, Inbred C57BL , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Protein Processing, Post-Translational , Virulence
17.
PLoS One ; 10(9): e0138529, 2015.
Article in English | MEDLINE | ID: mdl-26394407

ABSTRACT

Salmonella Typhimurium employs an array of type III secretion system effectors that facilitate intracellular survival and replication during infection. The Salmonella effector SseK3 was originally identified due to amino acid sequence similarity with NleB; an effector secreted by EPEC/EHEC that possesses N-acetylglucoasmine (GlcNAc) transferase activity and modifies death domain containing proteins to block extrinsic apoptosis. In this study, immunoprecipitation of SseK3 defined a novel molecular interaction between SseK3 and the host protein, TRIM32, an E3 ubiquitin ligase. The conserved DxD motif within SseK3, which is essential for the GlcNAc transferase activity of NleB, was required for TRIM32 binding and for the capacity of SseK3 to suppress TNF-stimulated activation of NF-κB pathway. However, we did not detect GlcNAc modification of TRIM32 by SseK3, nor did the SseK3-TRIM32 interaction impact on TRIM32 ubiquitination that is associated with its activation. In addition, lack of sseK3 in Salmonella had no effect on production of the NF-κB dependent cytokine, IL-8, in HeLa cells even though TRIM32 knockdown suppressed TNF-induced NF-κB activity. Ectopically expressed SseK3 partially co-localises with TRIM32 at the trans-Golgi network, but SseK3 is not recruited to Salmonella induced vacuoles or Salmonella induced filaments during Salmonella infection. Our study has identified a novel effector-host protein interaction and suggests that SseK3 may influence NF-κB activity. However, the lack of GlcNAc modification of TRIM32 suggests that SseK3 has further, as yet unidentified, host targets.


Subject(s)
Bacterial Proteins/metabolism , NF-kappa B/metabolism , Salmonella typhimurium/metabolism , Signal Transduction , Transcription Factors/metabolism , Bacterial Proteins/genetics , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunoblotting , Interleukin-8/metabolism , Microscopy, Confocal , Mutation , NF-kappa B/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/microbiology , Protein Binding , RNA Interference , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Transcription Factors/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , trans-Golgi Network/metabolism
18.
Cell Microbiol ; 17(12): 1766-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26096513

ABSTRACT

Upon infection of epithelial cells, enteropathogenic Escherichia coli suppresses host cell inflammatory signalling in a type III secretion system (T3SS) dependent manner. Two key T3SS effector proteins involved in this response are NleE and NleC. NleC is a zinc metalloprotease effector that degrades the p65 subunit of NF-κB. Although the site of p65 cleavage by NleC is now well described, other areas of interaction have not been precisely defined. Here we constructed overlapping truncations of p65 to identify regions required for NleC cleavage. We determined that NleC cleaved both p65 and p50 within the Rel homology domain (RHD) and that two motifs, E22IIE25 and P177VLS180 , within the RHD of p65 were important for recognition and binding by NleC. Alanine substitution of one or both of these motifs protected p65 from binding and degradation by NleC. The E22IIE25 and P177VLS180 motifs were located within the structurally distinct N-terminal subdomain of the RHD involved in DNA binding by p65 on adjacent, parallel strands. Although these motifs have not been recognized previously, both were needed for the correct localization and function of p65. In summary, this work has identified two regions of p65 within the RHD needed for binding and cleavage by NleC and provides further insight into the molecular basis of substrate recognition by a T3SS effector.


Subject(s)
Enteropathogenic Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Metalloproteases/metabolism , Transcription Factor RelA/metabolism , Amino Acid Motifs , DNA Mutational Analysis , Protein Binding , Protein Structure, Tertiary , Proteolysis , Transcription Factor RelA/genetics
19.
Cytokine Growth Factor Rev ; 25(2): 235-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440054

ABSTRACT

Gastrointestinal bacterial pathogens such as enteropathogenic Escherichia coli, Salmonella and Shigella control inflammatory and apoptotic signaling in human intestinal cells to establish infection, replicate and disseminate to other hosts. These pathogens manipulate host cell signaling through the translocation of virulence effector proteins directly into the host cell cytoplasm, which then target various signaling pathways. Death receptors such as TNFR1, FAS and TRAIL-R induce signaling cascades that are crucial to the clearance of pathogens, and as such are major targets for inhibition by pathogens. This review focuses on what is known about how bacterial gut pathogens inhibit death receptor signaling to suppress inflammation and prevent apoptosis.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Receptors, Death Domain/antagonists & inhibitors , Receptors, Death Domain/immunology , Apoptosis/immunology , Bacterial Secretion Systems/immunology , Escherichia coli/immunology , Humans , Inflammation/immunology , Microbiota/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Salmonella/immunology , Shigella/immunology , Signal Transduction/immunology
20.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24025841

ABSTRACT

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/microbiology , Signal Transduction , Virulence Factors/metabolism , Animals , Caspase 8/metabolism , Cell Death , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/physiology , Enteropathogenic Escherichia coli/pathogenicity , Enzyme Activation , Escherichia coli Infections/pathology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , N-Acetylglucosaminyltransferases/metabolism , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/metabolism , fas Receptor/deficiency , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...