Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nature ; 580(7805): E20, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32350466

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
mBio ; 10(6)2019 12 17.
Article in English | MEDLINE | ID: mdl-31848284

ABSTRACT

Listeria monocytogenes is a pathogenic bacterium causing potentially fatal foodborne infections in humans and animals. While the mechanisms used by Listeria to manipulate its host have been thoroughly characterized, how the host controls bacterial virulence factors remains to be extensively deciphered. Here, we found that the secreted Listeria virulence protein InlC is monoubiquitinated by the host cell machinery on K224, restricting infection. We show that the ubiquitinated form of InlC interacts with the intracellular alarmin S100A9, resulting in its stabilization and in increased reactive oxygen species production by neutrophils in infected mice. Collectively, our results suggest that posttranslational modification of InlC exacerbates the host response upon Listeria infection.IMPORTANCE The pathogenic potential of Listeria monocytogenes relies on the production of an arsenal of virulence determinants that have been extensively characterized, including surface and secreted proteins of the internalin family. We have previously shown that the Listeria secreted internalin InlC interacts with IκB kinase α to interfere with the host immune response (E. Gouin, M. Adib-Conquy, D. Balestrino, M.-A. Nahori, et al., Proc Natl Acad Sci USA, 107:17333-17338, 2010, https://doi.org/10.1073/pnas.1007765107). In the present work, we report that InlC is monoubiquitinated on K224 upon infection of cells and provide evidence that ubiquitinated InlC interacts with and stabilizes the alarmin S100A9, which is a critical regulator of the immune response and inflammatory processes. Additionally, we show that ubiquitination of InlC causes an increase in reactive oxygen species production by neutrophils in mice and restricts Listeria infection. These findings are the first to identify a posttranscriptional modification of an internalin contributing to host defense.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Listeria/physiology , Listeriosis/metabolism , Listeriosis/microbiology , Calgranulin B/metabolism , Disease Susceptibility , Epithelial Cells , Humans , Ubiquitination
3.
mBio ; 8(5)2017 10 31.
Article in English | MEDLINE | ID: mdl-29089430

ABSTRACT

Listeria monocytogenes is a bacterial pathogen causing severe foodborne infections in humans and animals. Listeria can enter into host cells and survive and multiply therein, due to an arsenal of virulence determinants encoded in different loci on the chromosome. Several key Listeria virulence genes are clustered in Listeria pathogenicity island 1. This important locus also contains orfX (lmo0206), a gene of unknown function. Here, we found that OrfX is a small, secreted protein whose expression is positively regulated by PrfA, the major transcriptional activator of Listeria virulence genes. We provide evidence that OrfX is a virulence factor that dampens the oxidative response of infected macrophages, which contributes to intracellular survival of bacteria. OrfX is targeted to the nucleus and interacts with the regulatory protein RybP. We show that in macrophages, the expression of OrfX decreases the level of RybP, which controls cellular infection. Collectively, these data reveal that Listeria targets RybP and evades macrophage oxidative stress for efficient infection. Altogether, OrfX is after LntA, the second virulence factor acting directly in the nucleus.IMPORTANCEListeria monocytogenes is a model bacterium that has been successfully used over the last 30 years to refine our understanding of the molecular, cellular, and tissular mechanisms of microbial pathogenesis. The major virulence factors of pathogenic Listeria species are located on a single chromosomal locus. Here, we report that the last gene of this locus encodes a small secreted nucleomodulin, OrfX, that is required for bacterial survival within macrophages and in the infected host. This work demonstrates that the production of OrfX contributes to limiting the host innate immune response by dampening the oxidative response of macrophages. We also identify a target of OrfX, RybP, which is an essential pleiotropic regulatory protein of the cell, and uncover its role in host defense. Our data reinforce the view that the secretion of nucleomodulins is an important strategy used by microbial pathogens to promote infection.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Virulence Factors/genetics , A549 Cells , Animals , Bacterial Load , Bacterial Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Listeriosis/microbiology , Liver/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , RNA-Binding Proteins/genetics , Repressor Proteins , Spleen/microbiology , Virulence , Virulence Factors/metabolism
4.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28827366

ABSTRACT

The pathogenesis of Listeria monocytogenes depends on the ability of this bacterium to escape from the phagosome of the host cells via the action of the pore-forming toxin listeriolysin O (LLO). Expression of the LLO-encoding gene (hly) requires the transcriptional activator PrfA, and both hly and prfA genes are essential for L. monocytogenes virulence. Here, we used the hemolytic activity of LLO as a phenotypic marker to screen for spontaneous virulence-attenuating mutations in L. monocytogenes Sixty nonhemolytic isolates were identified among a collection of 57,820 confirmed L. monocytogenes strains isolated from a variety of sources (0.1%). In most cases (56/60; 93.3%), the nonhemolytic phenotype resulted from nonsense, missense, or frameshift mutations in prfA Five strains carried hly mutations leading to a single amino acid substitution (G299V) or a premature stop codon causing strong virulence attenuation in mice. In one strain, both hly and gshF (encoding a glutathione synthase required for full PrfA activity) were missing due to genomic rearrangements likely caused by a transposable element. The PrfA/LLO loss-of-function (PrfA-/LLO-) mutants belonged to phylogenetically diverse clades of L. monocytogenes, and most were identified among nonclinical strains (57/60). Consistent with the rare occurrence of loss-of-virulence mutations, we show that prfA and hly are under purifying selection. Although occurring at a low frequency, PrfA-/LLO- mutational events in L. monocytogenes lead to niche restriction and open an evolutionary path for obligate saprophytism in this facultative intracellular pathogen.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins/genetics , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Mutation , Peptide Termination Factors/genetics , Amino Acid Substitution , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Biological Evolution , Cloning, Molecular , Erythrocytes/microbiology , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Hemolysis , Humans , Listeria monocytogenes/classification , Listeria monocytogenes/growth & development , Listeriosis/microbiology , Listeriosis/pathology , Mice , Mice, Inbred BALB C , Peptide Termination Factors/metabolism , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Selection, Genetic , Severity of Illness Index , Virulence
5.
Cell ; 161(2): 199-200, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25860603

ABSTRACT

Benanti et al. report that Burkholderia pseudomallei and Burkholderia mallei bacteria express proteins that mimic Ena/Vasp family proteins to polymerize actin, thereby inducing actin-based motility. Thus, bacteria can use the various cellular actin polymerization mechanisms for intra- and inter-cellular dissemination.


Subject(s)
Actins/metabolism , Burkholderia Infections/microbiology , Burkholderia/physiology , Burkholderia/pathogenicity , Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Animals , Humans
6.
J Infect Dis ; 212(2): 223-33, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25589336

ABSTRACT

BACKGROUND: Although the T-cell subset differentiation pathway has been characterized extensively from the view of host gene regulation, the effects of genes of the pathogen on T-cell subset differentiation during infection have yet to be elucidated. Especially, the bacterial genes that are responsible for this shift have not yet been determined. METHODS: Utilizing a single-gene-mutation Listeria panel, we investigated genes involved in the host-pathogen interaction that are required for the initiation of T-cell subset differentiation in the early phase of pathogen infection. RESULTS: We demonstrate that the induction of T helper types 1 and 2 (Th1 and Th2) subsets are separate phenomena and are mediated by distinct Listeria genes. We identified several candidate Listeria genes that appear to be involved in the host-Listeria interaction. Among them, arpJ is the strongest candidate gene for inhibiting Th2 subset induction. Furthermore, the analysis utilizing arpJ-deficient Listeria monocytogenes (Lm) revealed that the tumor necrosis factor (TNF) superfamily (Tnfsf) 9-TNF receptor superfamily (Tnfrsf) 9 interaction inhibits the Th2 response during Lm infection. CONCLUSIONS: arpJ is the candidate gene for inhibiting Th2 T-cell subset induction. The arpJ gene product influences the expression of Tnfsf/Tnfrsf on antigen-presenting cells and inhibits the Th2 T-cell subset differentiation during Listeria infection.


Subject(s)
Cell Differentiation/immunology , Listeria monocytogenes/genetics , Listeriosis/immunology , T-Lymphocytes, Helper-Inducer/physiology , Animals , Cells, Cultured , Genes, Bacterial , Host-Pathogen Interactions , Listeria monocytogenes/immunology , Listeriosis/microbiology , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/virology
7.
FEBS J ; 281(13): 2977-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24814574

ABSTRACT

Ral proteins are small GTPases that play critical roles in normal physiology and in oncogenesis. There is little information on the GTPase-activating proteins (GAPs) that downregulate their activity. Here, we provide evidence that the noncatalytic ß subunit of RalGAPα1/2 ß complexes is involved in mitotic control. RalGAPß localizes to the Golgi and nucleus during interphase, and relocalizes to the mitotic spindle and cytokinetic intercellular bridge during mitosis. Depletion of RalGAPß causes chromosome misalignment and decreases the amount of mitotic cyclin B1, disturbing the metaphase-to-anaphase transition. Overexpression of RalGAPß interferes with cell division, leading to binucleation and multinucleation, and cell death. We propose that RalGAPß plays an essential role in the sequential progression of mitosis by controlling the spatial and temporal activation of Ral GTPases in the spindle assembly checkpoint (SAC) and cytokinesis. Deregulation of RalGAPß might cause genomic instability, leading to human carcinogenesis.


Subject(s)
Anaphase , GTPase-Activating Proteins/physiology , Metaphase , Cell Death , Cell Line, Tumor , Chromatography, Affinity , Chromosome Segregation , GTPase-Activating Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mitosis , Nerve Tissue Proteins/metabolism , Protein Transport , Spindle Apparatus/metabolism , Tubulin/metabolism , ral GTP-Binding Proteins/metabolism
8.
mBio ; 5(2): e00969-14, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24667708

ABSTRACT

For nearly 3 decades, listeriologists and immunologists have used mainly three strains of the same serovar (1/2a) to analyze the virulence of the bacterial pathogen Listeria monocytogenes. The genomes of two of these strains, EGD-e and 10403S, were released in 2001 and 2008, respectively. Here we report the genome sequence of the third reference strain, EGD, and extensive genomic and phenotypic comparisons of the three strains. Strikingly, EGD-e is genetically highly distinct from EGD (29,016 single nucleotide polymorphisms [SNPs]) and 10403S (30,296 SNPs), and is more related to serovar 1/2c than 1/2a strains. We also found that while EGD and 10403S strains are genetically very close (317 SNPs), EGD has a point mutation in the transcriptional regulator PrfA (PrfA*), leading to constitutive expression of several major virulence genes. We generated an EGD-e PrfA* mutant and showed that EGD behaves like this strain in vitro, with slower growth in broth and higher invasiveness in human cells than those of EGD-e and 10403S. In contrast, bacterial counts in blood, liver, and spleen during infection in mice revealed that EGD and 10403S are less virulent than EGD-e, which is itself less virulent than EGD-e PrfA*. Thus, constitutive expression of PrfA-regulated virulence genes does not appear to provide a significant advantage to the EGD strain during infection in vivo, highlighting the fact that in vitro invasion assays are not sufficient for evaluating the pathogenic potential of L. monocytogenes strains. Together, our results pave the way for deciphering unexplained differences or discrepancies in experiments using different L. monocytogenes strains. IMPORTANCE Over the past 3 decades, Listeria has become a model organism for host-pathogen interactions, leading to critical discoveries in a broad range of fields, including bacterial gene regulation, cell biology, and bacterial pathophysiology. Scientists studying Listeria use primarily three pathogenic strains: EGD, EGD-e, and 10403S. Despite many studies on EGD, it is the only one of the three strains whose genome has not been sequenced. Here we report the sequence of its genome and a series of important genomic and phenotypic differences between the three strains, in particular, a critical mutation in EGD's PrfA, the main regulator of Listeria virulence. Our results show that the three strains display differences which may play an important role in the virulence differences observed between the strains. Our findings will be of critical relevance to listeriologists and immunologists who have used or may use Listeria as a tool to study the pathophysiology of listeriosis and immune responses.


Subject(s)
DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Genetic Variation , Genome, Bacterial , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Animals , Blood/microbiology , Disease Models, Animal , Humans , Listeriosis/microbiology , Listeriosis/pathology , Liver/microbiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Sequence Analysis, DNA , Spleen/microbiology , Virulence
9.
Proc Natl Acad Sci U S A ; 110(51): 20521-6, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24306931

ABSTRACT

The intracellular bacterial pathogen Listeria monocytogenes is capable of remodelling the actin cytoskeleton of its host cells such that "comet tails" are assembled powering its movement within cells and enabling cell-to-cell spread. We used cryo-electron tomography to visualize the 3D structure of the comet tails in situ at the level of individual filaments. We have performed a quantitative analysis of their supramolecular architecture revealing the existence of bundles of nearly parallel hexagonally packed filaments with spacings of 12-13 nm. Similar configurations were observed in stress fibers and filopodia, suggesting that nanoscopic bundles are a generic feature of actin filament assemblies involved in motility; presumably, they provide the necessary stiffness. We propose a mechanism for the initiation of comet tail assembly and two scenarios that occur either independently or in concert for the ensuing actin-based motility, both emphasizing the role of filament bundling.


Subject(s)
Listeria monocytogenes/ultrastructure , Listeriosis , Models, Molecular , Stress Fibers/ultrastructure , Cell Line , Cryoelectron Microscopy/methods , Humans , Listeria monocytogenes/metabolism , Stress Fibers/metabolism
10.
J Vis Exp ; (79): e51043, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24084755

ABSTRACT

Bacterial intracellular pathogens can be conceived as molecular tools to dissect cellular signaling cascades due to their capacity to exquisitely manipulate and subvert cell functions which are required for the infection of host target tissues. Among these bacterial pathogens, Listeria monocytogenes is a Gram positive microorganism that has been used as a paradigm for intracellular parasitism in the characterization of cellular immune responses, and which has played instrumental roles in the discovery of molecular pathways controlling cytoskeletal and membrane trafficking dynamics. In this article, we describe a robust microscopical assay for the detection of late cellular infection stages of L. monocytogenes based on the fluorescent labeling of InlC, a secreted bacterial protein which accumulates in the cytoplasm of infected cells; this assay can be coupled to automated high-throughput small interfering RNA screens in order to characterize cellular signaling pathways involved in the up- or down-regulation of infection.


Subject(s)
Bacterial Proteins/analysis , Bacterial Proteins/metabolism , Listeria monocytogenes/pathogenicity , Bacterial Proteins/chemistry , HeLa Cells , Humans , Listeria monocytogenes/chemistry , Listeria monocytogenes/genetics , Listeriosis/microbiology , Microscopy, Fluorescence/methods , RNA, Small Interfering/genetics , Transfection
11.
Proc Natl Acad Sci U S A ; 110(32): 13132-7, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23878253

ABSTRACT

Riboswitches are ligand-binding elements located in 5' untranslated regions of messenger RNAs, which regulate expression of downstream genes. In Listeria monocytogenes, a vitamin B12-binding (B12) riboswitch was identified, not upstream of a gene but downstream, and antisense to the adjacent gene, pocR, suggesting it might regulate pocR in a nonclassical manner. In Salmonella enterica, PocR is a transcription factor that is activated by 1,2-propanediol, and subsequently activates expression of the pdu genes. The pdu genes mediate propanediol catabolism and are implicated in pathogenesis. As enzymes involved in propanediol catabolism require B12 as a cofactor, we hypothesized that the Listeria B12 riboswitch might be involved in pocR regulation. Here we demonstrate that the B12 riboswitch is transcribed as part of a noncoding antisense RNA, herein named AspocR. In the presence of B12, the riboswitch induces transcriptional termination, causing aspocR to be transcribed as a short transcript. In contrast, in the absence of B12, aspocR is transcribed as a long antisense RNA, which inhibits pocR expression. Regulation by AspocR ensures that pocR, and consequently the pdu genes, are maximally expressed only when both propanediol and B12 are present. Strikingly, AspocR can inhibit pocR expression in trans, suggesting it acts through a direct interaction with pocR mRNA. Together, this study demonstrates how pocR and the pdu genes can be regulated by B12 in bacteria and extends the classical definition of riboswitches from elements governing solely the expression of mRNAs to a wider role in controlling transcription of noncoding RNAs.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/genetics , RNA, Antisense/genetics , Riboswitch/genetics , Bacterial Proteins/metabolism , Base Sequence , Blotting, Northern , Blotting, Western , Gene Expression Regulation, Bacterial , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Listeria monocytogenes/metabolism , Mutation , Propylene Glycol/metabolism , Protein Binding , RNA Stability/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Vitamin B 12/metabolism
12.
PLoS Pathog ; 9(1): e1003131, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23382675

ABSTRACT

Listeria monocytogenes (Lm) is a ubiquitous bacterium able to survive and thrive within the environment and readily colonizes a wide range of substrates, often as a biofilm. It is also a facultative intracellular pathogen, which actively invades diverse hosts and induces listeriosis. So far, these two complementary facets of Lm biology have been studied independently. Here we demonstrate that the major Lm virulence determinant ActA, a PrfA-regulated gene product enabling actin polymerization and thereby promoting its intracellular motility and cell-to-cell spread, is critical for bacterial aggregation and biofilm formation. We show that ActA mediates Lm aggregation via direct ActA-ActA interactions and that the ActA C-terminal region, which is not involved in actin polymerization, is essential for aggregation in vitro. In mice permissive to orally-acquired listeriosis, ActA-mediated Lm aggregation is not observed in infected tissues but occurs in the gut lumen. Strikingly, ActA-dependent aggregating bacteria exhibit an increased ability to persist within the cecum and colon lumen of mice, and are shed in the feces three order of magnitude more efficiently and for twice as long than bacteria unable to aggregate. In conclusion, this study identifies a novel function for ActA and illustrates that in addition to contributing to its dissemination within the host, ActA plays a key role in Lm persistence within the host and in transmission from the host back to the environment.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Cecum/metabolism , Colon/metabolism , Intestinal Mucosa/metabolism , Listeria monocytogenes/pathogenicity , Membrane Proteins/metabolism , Animals , Cecum/microbiology , Cell Line , Colon/microbiology , Disease Models, Animal , Feces/microbiology , Host-Pathogen Interactions , Humans , Intestinal Mucosa/microbiology , Listeria monocytogenes/growth & development , Listeria monocytogenes/metabolism , Listeriosis/metabolism , Listeriosis/microbiology , Mice , Virulence Factors/metabolism
13.
J Biol Chem ; 286(40): 35096-103, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21841197

ABSTRACT

Pathogenic Rickettsia species cause high morbidity and mortality, especially R. prowazekii, the causative agent of typhus. Like many intracellular pathogens, Rickettsia exploit the cytoskeleton to enter and spread within the host cell. Here we report that the cell surface antigen sca4 of Rickettsia co-localizes with vinculin in cells at sites of focal adhesions in sca4-transfected cells and that sca4 binds to and activates vinculin through two vinculin binding sites (VBSs) that are conserved across all Rickettsia. Remarkably, this occurs through molecular mimicry of the vinculin-talin interaction that is also seen with the IpaA invasin of the intracellular pathogen Shigella, where binding of these VBSs to the vinculin seven-helix bundle head domain (Vh1) displaces intramolecular interactions with the vinculin tail domain that normally clamp vinculin in an inactive state. Finally, the vinculin·sca4-VBS crystal structures reveal that vinculin adopts a new conformation when bound to the C-terminal VBS of sca4. Collectively, our data define the mechanism by which sca4 activates vinculin and interacts with the actin cytoskeleton, and they suggest important roles for vinculin in Rickettsia pathogenesis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Vinculin/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray/methods , Cytoskeleton/metabolism , Focal Adhesions/metabolism , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Binding , Protein Interaction Mapping/methods , Rickettsia , Sequence Homology, Amino Acid
14.
PLoS Pathog ; 7(8): e1002168, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21829365

ABSTRACT

L. monocytogenes is a facultative intracellular bacterium responsible for listeriosis. It is able to invade, survive and replicate in phagocytic and non-phagocytic cells. The infectious process at the cellular level has been extensively studied and many virulence factors have been identified. Yet, the role of InlK, a member of the internalin family specific to L. monocytogenes, remains unknown. Here, we first show using deletion analysis and in vivo infection, that InlK is a bona fide virulence factor, poorly expressed in vitro and well expressed in vivo, and that it is anchored to the bacterial surface by sortase A. We then demonstrate by a yeast two hybrid screen using InlK as a bait, validated by pulldown experiments and immunofluorescence analysis that intracytosolic bacteria via an interaction with the protein InlK interact with the Major Vault Protein (MVP), the main component of cytoplasmic ribonucleoproteic particules named vaults. Although vaults have been implicated in several cellular processes, their role has remained elusive. Our analysis demonstrates that MVP recruitment disguises intracytosolic bacteria from autophagic recognition, leading to an increased survival rate of InlK over-expressing bacteria compared to InlK(-) bacteria. Together these results reveal that MVP is hijacked by L. monocytogenes in order to counteract the autophagy process, a finding that could have major implications in deciphering the cellular role of vault particles.


Subject(s)
Autophagy , Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Listeriosis/metabolism , Vault Ribonucleoprotein Particles/metabolism , Virulence Factors/metabolism , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Bacterial Proteins/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Listeria monocytogenes/genetics , Listeriosis/genetics , Mice , Mice, Inbred BALB C , Protein Binding , Two-Hybrid System Techniques , Vault Ribonucleoprotein Particles/genetics , Virulence Factors/genetics
15.
Cell Host Microbe ; 8(5): 433-44, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21075354

ABSTRACT

Actin-based motility is used by various pathogens for dissemination within and between cells. Yet host factors restricting this process have not been identified. Septins are GTP-binding proteins that assemble as filaments and are essential for cell division. However, their role during interphase has remained elusive. Here, we report that septin assemblies are recruited to different bacteria that polymerize actin. We observed that intracytosolic Shigella either become compartmentalized in septin cage-like structures or form actin tails. Inactivation of septin caging increases the number of Shigella with actin tails and enhances cell-to-cell spread. TNF-α, a host cytokine produced upon Shigella infection, stimulates septin caging and restricts actin tail formation and cell-to-cell spread. Finally, we show that septin cages entrap bacteria targeted to autophagy. Together, these results reveal an unsuspected mechanism of host defense that restricts dissemination of invasive pathogens.


Subject(s)
Cervix Uteri/microbiology , Colon/microbiology , Cytosol/microbiology , Host-Pathogen Interactions , Septins/metabolism , Shigella flexneri/pathogenicity , Actins/metabolism , Caco-2 Cells/immunology , Caco-2 Cells/microbiology , Caco-2 Cells/ultrastructure , Cervix Uteri/cytology , Colon/cytology , Female , HeLa Cells/immunology , HeLa Cells/microbiology , HeLa Cells/ultrastructure , Humans , Shigella flexneri/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Proc Natl Acad Sci U S A ; 107(40): 17333-8, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855622

ABSTRACT

Listeria monocytogenes is an intracellular pathogen responsible for severe foodborne infections. It can replicate in both phagocytic and nonphagocytic mammalian cells. The infectious process at the cellular level has been studied extensively, but how the bacterium overcomes early host innate immune responses remains largely unknown. Here we show that InlC, a member of the internalin family, is secreted intracellularly and directly interacts with IKKα, a subunit of the IκB kinase complex critical for the phosphorylation of IκB and activation of NF-κB, the major regulator of innate immune responses. Infection experiments with WT Listeria or the inlC-deletion mutant and transfection of cells with InlC reveal that InlC expression impairs phosphorylation and consequently delays IκB degradation normally induced by TNF-α, a classical NF-κB stimulator. Moreover, infection of RAW 264.7 macrophages by the inlC mutant leads to increased production of proinflammatory cytokines compared with that obtained with the WT. Finally, in a peritonitis mouse model, we show that infection with the inlC mutant induces increased production of chemokines and increased recruitment of neutrophils in the peritoneal cavity compared with infection with WT. Together, these results demonstrate that InlC, by interacting with IKKα, dampens the host innate response induced by Listeria during the infection process.


Subject(s)
Bacterial Proteins/immunology , I-kappa B Kinase/metabolism , Immunity, Innate , Protein Subunits/metabolism , Animals , Cell Line , Humans , I-kappa B Kinase/genetics , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Mice , Promoter Regions, Genetic , Protein Subunits/genetics , Tumor Necrosis Factor-alpha/metabolism , Two-Hybrid System Techniques
17.
J Infect Dis ; 202(4): 551-62, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20617901

ABSTRACT

Attachment to mucosal surfaces is the initial event in the pathogenesis of the human foodborne pathogen Listeria monocytogenes. By use of comparative genomics, we identified a L. monocytogenes-specific gene, lapB, that encodes an LPXTG surface protein that is absent from nonpathogenic Listeria species. We showed that lapB expression is positively regulated by PrfA, the major transcriptional activator of the virulence genes of Listeria species, and is up-regulated in mouse spleens during infection. We demonstrated that LapB is an SrtA-anchored surface protein required for adhesion to and entry into mammalian cells and for virulence following intravenous or oral inoculation in mice. Our results highlight LapB as a new L. monocytogenes virulence adhesin with a function that is supported by its unique N-terminal domain through the probable interaction with a cellular receptor.


Subject(s)
Adhesins, Bacterial/physiology , Eukaryotic Cells/microbiology , Listeria monocytogenes/pathogenicity , Virulence Factors/physiology , Adhesins, Bacterial/biosynthesis , Amino Acid Sequence , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Cell Line , Colony Count, Microbial , Computational Biology , Gene Expression Regulation, Bacterial , Humans , Listeriosis/microbiology , Listeriosis/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Termination Factors/metabolism , Up-Regulation , Virulence , Virulence Factors/biosynthesis
18.
Nature ; 464(7292): 1192-5, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20414307

ABSTRACT

During infection, pathogenic bacteria manipulate the host cell in various ways to allow their own replication, propagation and escape from host immune responses. Post-translational modifications are unique mechanisms that allow cells to rapidly, locally and specifically modify activity or interactions of key proteins. Some of these modifications, including phosphorylation and ubiquitylation, can be induced by pathogens. However, the effects of pathogenic bacteria on SUMOylation, an essential post-translational modification in eukaryotic cells, remain largely unknown. Here we show that infection with Listeria monocytogenes leads to a decrease in the levels of cellular SUMO-conjugated proteins. This event is triggered by the bacterial virulence factor listeriolysin O (LLO), which induces a proteasome-independent degradation of Ubc9, an essential enzyme of the SUMOylation machinery, and a proteasome-dependent degradation of some SUMOylated proteins. The effect of LLO on Ubc9 is dependent on the pore-forming capacity of the toxin and is shared by other bacterial pore-forming toxins like perfringolysin O (PFO) and pneumolysin (PLY). Ubc9 degradation was also observed in vivo in infected mice. Furthermore, we show that SUMO overexpression impairs bacterial infection. Together, our results reveal that Listeria, and probably other pathogens, dampen the host response by decreasing the SUMOylation level of proteins critical for infection.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Listeriosis/microbiology , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Bacterial Toxins/metabolism , Cell Line , HeLa Cells , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Mice , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Virulence Factors/metabolism
19.
Infect Immun ; 78(1): 204-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901060

ABSTRACT

Listeria monocytogenes is an intracellular bacterial pathogen that invades epithelial cells by subverting two cellular receptors, E-cadherin and Met. We recently identified type II phosphatidylinositol 4-kinases alpha and beta (PI4KIIalpha and PI4KIIbeta) as being required for bacterial entry downstream of Met. In this work, we investigated whether tetraspanins CD9, CD63, and CD81, which figure among the few described molecular partners of PI4KIIalpha, function as molecular adaptors recruiting PI4KIIalpha to the bacterial entry site. We observed by fluorescence microscopy that CD9, CD63, and CD81 are expressed and detected at the cellular surface and also within intracellular compartments, particularly in the case of CD63. In resting cells, colocalization of tetraspanins and PI4KIIalpha is detectable only in restricted areas of the perinuclear region. Upon infection with Listeria, endogenous CD9, CD63, and CD81 were recruited to the bacterial entry site but did not colocalize strictly with endogenous PI4KIIalpha. Live-cell imaging confirmed that tetraspanins and PI4KIIalpha do not follow the same recruitment dynamics to the Listeria entry site. Depletion of CD9, CD63, and CD81 levels by small interfering RNA demonstrated that CD81 is required for bacterial internalization, identifying for the first time a role for a member of the tetraspanin family in the entry of Listeria into target cells. Moreover, depletion of CD81 inhibits the recruitment of PI4KIIalpha but not that of the Met receptor to the bacterial entry site, suggesting that CD81 may act as a membrane organizer required for the integrity of signaling events occurring at Listeria entry sites.


Subject(s)
Antigens, CD/metabolism , Listeria monocytogenes/physiology , Antigens, CD/genetics , Epithelial Cells , Gene Expression Regulation/immunology , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Minor Histocompatibility Antigens , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Protein Transport , Tetraspanin 28 , Tetraspanin 29 , Tetraspanin 30
SELECTION OF CITATIONS
SEARCH DETAIL
...