Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Crit Rev Food Sci Nutr ; : 1-13, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37862145

ABSTRACT

Ribosomes that synthesize proteins are among the most central and evolutionarily conserved organelles. Given the key role of proteins in cellular functions, prokaryotic and eukaryotic pathogens have evolved potent toxins to inhibit ribosomal functions and weaken their host. Many of these ribotoxin-producing pathogens are associated with food. For example, food can be contaminated with bacterial pathogens that produce the ribotoxin Shiga toxin, but also with the fungal ribotoxin deoxynivalenol. Shiga toxin cleaves ribosomal RNA, while deoxynivalenol binds to and inhibits the peptidyl transferase center. Despite their distinct modes of action, both groups of ribotoxins hinder protein translation, but also trigger other comparable toxic effects, which depend or not on the activation of the ribotoxic stress response. Ribotoxic stress response-dependent effects include inflammation and apoptosis, whereas ribotoxic stress response-independent effects include endoplasmic reticulum stress, oxidative stress, and autophagy. For other effects, such as cell cycle arrest and cytoskeleton modulation, the involvement of the ribotoxic stress response is still controversial. Ribotoxins affect one organelle yet induce multiple toxic effects with multiple consequences for the cell. The ribosome can therefore be considered as the cellular "Achilles heel" targeted by food borne ribotoxins. Considering the high toxicity of ribotoxins, they pose a substantial health risk, as humans are highly susceptible to widespread exposure to these toxins through contaminated food sources.

2.
Autophagy ; 18(12): 2913-2925, 2022 12.
Article in English | MEDLINE | ID: mdl-35311462

ABSTRACT

Escherichia coli strains are responsible for a majority of human extra-intestinal infections, resulting in huge direct medical and social costs. We had previously shown that HlyF encoded by a large virulence plasmid harbored by pathogenic E. coli is not a hemolysin but a cytoplasmic enzyme leading to the overproduction of outer membrane vesicles (OMVs). Here, we showed that these specific OMVs inhibit the macroautophagic/autophagic flux by impairing the autophagosome-lysosome fusion, thus preventing the formation of acidic autolysosomes and autophagosome clearance. Furthermore, HlyF-associated OMVs were more prone to activate the non-canonical inflammasome pathway. Because autophagy and inflammation are crucial in the host's response to infection especially during sepsis, our findings revealed an unsuspected role of OMVs in the crosstalk between bacteria and their host, highlighting the fact that these extracellular vesicles have exacerbated pathogenic properties.Abbreviations: AIEC: adherent-invasive E. coliBDI: bright detail intensityBMDM: bone marrow-derived macrophagesCASP: caspaseE. coli: Escherichia coliEHEC: enterohemorrhagic E. coliExPEC: extra-intestinal pathogenic E. coliGSDMD: gasdermin DGFP: green fluorescent proteinHBSS: Hanks' balanced salt solutionHlyF: hemolysin FIL1B/IL-1B: interleukin 1 betaISX: ImageStreamX systemLPS: lipopolysaccharideMut: mutatedOMV: outer membrane vesicleRFP: red fluorescent proteinTEM: transmission electron microscopyWT: wild-type.


Subject(s)
Escherichia coli Infections , Escherichia coli , Humans , Escherichia coli/metabolism , Inflammasomes/metabolism , Hemolysin Proteins , Autophagy , Escherichia coli Infections/metabolism
3.
mSphere ; 6(4): e0062421, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34378987

ABSTRACT

The probiotic Escherichia coli strain Nissle 1917 (DSM 6601, Mutaflor), generally considered beneficial and safe, has been used for a century to treat various intestinal diseases. However, Nissle 1917 hosts in its genome the pks pathogenicity island that codes for the biosynthesis of the genotoxin colibactin. Colibactin is a potent DNA alkylator, suspected to play a role in colorectal cancer development. We show in this study that Nissle 1917 is functionally capable of producing colibactin and inducing interstrand cross-links in the genomic DNA of epithelial cells exposed to the probiotic. This toxicity was even exacerbated with lower doses of the probiotic, when the exposed cells started to divide again but exhibited aberrant anaphases and increased gene mutation frequency. DNA damage was confirmed in vivo in mouse models of intestinal colonization, demonstrating that Nissle 1917 produces the genotoxin in the gut lumen. Although it is possible that daily treatment of adult humans with their microbiota does not produce the same effects, administration of Nissle 1917 as a probiotic or as a chassis to deliver therapeutics might exert long-term adverse effects and thus should be considered in a risk-versus-benefit evaluation. IMPORTANCE Nissle 1917 is sold as a probiotic and considered safe even though it has been known since 2006 that it harbors the genes for colibactin synthesis. Colibactin is a potent genotoxin that is now linked to causative mutations found in human colorectal cancer. Many papers concerning the use of this strain in clinical applications ignore or elude this fact or misleadingly suggest that Nissle 1917 does not induce DNA damage. Here, we demonstrate that Nissle 1917 produces colibactin in vitro and in vivo and induces mutagenic DNA damage. This is a serious safety concern that must not be ignored in the interests of patients, the general public, health care professionals, and ethical probiotic manufacturers.


Subject(s)
DNA Damage , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Genome, Bacterial , Mutagenesis , Probiotics , Animals , CHO Cells , Cricetulus , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Female , Genomic Islands , HeLa Cells , Humans , Mice , Mutation
4.
Cell Mol Life Sci ; 78(17-18): 6319-6335, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34308492

ABSTRACT

The Cytolethal Distending Toxin (CDT) is a bacterial genotoxin produced by pathogenic bacteria causing major foodborne diseases worldwide. CDT activates the DNA Damage Response and modulates the host immune response, but the precise relationship between these outcomes has not been addressed so far. Here, we show that chronic exposure to CDT in HeLa cells or mouse embryonic fibroblasts promotes a strong type I interferon (IFN) response that depends on the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) through the recognition of micronuclei. Indeed, despite active cell cycle checkpoints and in contrast to other DNA damaging agents, cells exposed to CDT reach mitosis where they accumulate massive DNA damage, resulting in chromosome fragmentation and micronucleus formation in daughter cells. These mitotic phenotypes are observed with CDT from various origins and in cancer or normal cell lines. Finally, we show that CDT exposure in immortalized normal colonic epithelial cells is associated to cGAS protein loss and low type I IFN response, implying that CDT immunomodulatory function may vary depending on tissue and cell type. Thus, our results establish a direct link between CDT-induced DNA damage, genetic instability and the cellular immune response that may be relevant in the context of natural infection associated to chronic inflammation or carcinogenesis.


Subject(s)
Bacterial Toxins/pharmacology , Interferon Type I/metabolism , Nucleotidyltransferases/metabolism , Up-Regulation/drug effects , Animals , Cell Cycle Checkpoints/drug effects , DNA Breaks, Double-Stranded/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Humans , Interferon Type I/genetics , Mice , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/genetics
5.
Front Cell Infect Microbiol ; 10: 586934, 2020.
Article in English | MEDLINE | ID: mdl-33330131

ABSTRACT

Salmonella Typhimurium expresses on its outer membrane the protein Rck which interacts with the epidermal growth factor receptor (EGFR) of the plasma membrane of the targeted host cells. This interaction activates signaling pathways, leading to the internalization of Salmonella. Since EGFR plays a key role in cell proliferation, we sought to determine the influence of Rck mediated infection on the host cell cycle. By analyzing the DNA content of uninfected and infected cells using flow cytometry, we showed that the Rck-mediated infection induced a delay in the S-phase (DNA replication phase) of the host cell cycle, independently of bacterial internalization. We also established that this Rck-dependent delay in cell cycle progression was accompanied by an increased level of host DNA double strand breaks and activation of the DNA damage response. Finally, we demonstrated that the S-phase environment facilitated Rck-mediated bacterial internalization. Consequently, our results suggest that Rck can be considered as a cyclomodulin with a genotoxic activity.


Subject(s)
Bacterial Outer Membrane Proteins , Salmonella typhimurium , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins , Cell Division , Cell Membrane , Salmonella typhimurium/genetics , Signal Transduction
6.
Sci Rep ; 9(1): 7694, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118484

ABSTRACT

Staphylococcus aureus causes serious medical problems in human and animals. Here we show that S. aureus can compromise host genomic integrity as indicated by bacteria-induced histone H2AX phosphorylation, a marker of DNA double strand breaks (DSBs), in human cervix cancer HeLa and osteoblast-like MG-63 cells. This DNA damage is mediated by alpha phenol-soluble modulins (PSMα1-4), while a specific class of lipoproteins (Lpls), encoded on a pathogenicity island in S. aureus, dampens the H2AX phosphorylation thus counteracting the DNA damage. This DNA damage is mediated by reactive oxygen species (ROS), which promotes oxidation of guanine forming 7,8-dihydro-8-oxoguanine (8-oxoG). DNA damage is followed by the induction of DNA repair that involves the ATM kinase-signaling pathway. An examination of S. aureus strains, isolated from the same patient during acute initial and recurrent bone and joint infections (BJI), showed that recurrent strains produce lower amounts of Lpls, induce stronger DNA-damage and prompt the G2/M transition delay to a greater extent that suggest an involvement of these mechanisms in adaptive processes of bacteria during chronicization. Our findings redefine our understanding of mechanisms of S. aureus-host interaction and suggest that the balance between the levels of PSMα and Lpls expression impacts the persistence of the infection.


Subject(s)
DNA Damage , Staphylococcus aureus/pathogenicity , Acetylcysteine/pharmacology , Arthritis, Infectious/microbiology , Bacterial Toxins/pharmacology , Cell Line, Tumor , DNA Repair , Etoposide/pharmacology , G2 Phase Cell Cycle Checkpoints , Genomic Islands , Guanine/analogs & derivatives , Guanine/metabolism , HeLa Cells/microbiology , Histones/analysis , Host-Pathogen Interactions , Humans , Lipoproteins/pharmacology , Osteitis/microbiology , Osteoblasts/microbiology , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational , Reactive Oxygen Species , Staphylococcal Infections/microbiology
7.
mBio ; 9(2)2018 03 20.
Article in English | MEDLINE | ID: mdl-29559578

ABSTRACT

Colibactins are hybrid polyketide-nonribosomal peptides produced by Escherichia coli, Klebsiella pneumoniae, and other Enterobacteriaceae harboring the pks genomic island. These genotoxic metabolites are produced by pks-encoded peptide-polyketide synthases as inactive prodrugs called precolibactins, which are then converted to colibactins by deacylation for DNA-damaging effects. Colibactins are bona fide virulence factors and are suspected of promoting colorectal carcinogenesis when produced by intestinal E. coli Natural active colibactins have not been isolated, and how they induce DNA damage in the eukaryotic host cell is poorly characterized. Here, we show that DNA strands are cross-linked covalently when exposed to enterobacteria producing colibactins. DNA cross-linking is abrogated in a clbP mutant unable to deacetylate precolibactins or by adding the colibactin self-resistance protein ClbS, confirming the involvement of the mature forms of colibactins. A similar DNA-damaging mechanism is observed in cellulo, where interstrand cross-links are detected in the genomic DNA of cultured human cells exposed to colibactin-producing bacteria. The intoxicated cells exhibit replication stress, activation of ataxia-telangiectasia and Rad3-related kinase (ATR), and recruitment of the DNA cross-link repair Fanconi anemia protein D2 (FANCD2) protein. In contrast, inhibition of ATR or knockdown of FANCD2 reduces the survival of cells exposed to colibactin-producing bacteria. These findings demonstrate that DNA interstrand cross-linking is the critical mechanism of colibactin-induced DNA damage in infected cells.IMPORTANCE Colorectal cancer is the third-most-common cause of cancer death. In addition to known risk factors such as high-fat diets and alcohol consumption, genotoxic intestinal Escherichia coli bacteria producing colibactin are proposed to play a role in colon cancer development. Here, by using transient infections with genotoxic E. coli, we showed that colibactins directly generate DNA cross-links in cellulo Such lesions are converted into double-strand breaks during the repair response. DNA cross-links, akin to those induced by metabolites of alcohol and high-fat diets and by widely used anticancer drugs, are both severely mutagenic and profoundly cytotoxic lesions. This finding of a direct induction of DNA cross-links by a bacterium should facilitate delineating the role of E. coli in colon cancer and engineering new anticancer agents.


Subject(s)
Escherichia coli/metabolism , Peptides/metabolism , Polyketides/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism
9.
Article in English | MEDLINE | ID: mdl-28589102

ABSTRACT

Some bacterial pathogens modulate signaling pathways of eukaryotic cells in order to subvert the host response for their own benefit, leading to successful colonization and invasion. Pathogenic bacteria produce multiple compounds that generate favorable conditions to their survival and growth during infection in eukaryotic hosts. Many bacterial toxins can alter the cell cycle progression of host cells, impairing essential cellular functions and impeding host cell division. This review summarizes current knowledge regarding cyclomodulins, a heterogeneous family of bacterial effectors that induce eukaryotic cell cycle alterations. We discuss the mechanisms of actions of cyclomodulins according to their biochemical properties, providing examples of various cyclomodulins such as cycle inhibiting factor, γ-glutamyltranspeptidase, cytolethal distending toxins, shiga toxin, subtilase toxin, anthrax toxin, cholera toxin, adenylate cyclase toxins, vacuolating cytotoxin, cytotoxic necrotizing factor, Panton-Valentine leukocidin, phenol soluble modulins, and mycolactone. Special attention is paid to the benefit provided by cyclomodulins to bacteria during colonization of the host.


Subject(s)
Bacteria/pathogenicity , Bacterial Physiological Phenomena , Bacterial Toxins/metabolism , Cell Cycle/drug effects , Eukaryotic Cells/microbiology , Adenylate Cyclase Toxin/toxicity , Animals , Antigens, Bacterial/toxicity , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Cholera Toxin/toxicity , Eukaryotic Cells/drug effects , Exotoxins/toxicity , Host-Parasite Interactions , Humans , Leukocidins/toxicity , Macrolides/toxicity , Shiga Toxin/toxicity , Signal Transduction , Virulence Factors/toxicity
10.
EcoSal Plus ; 7(1)2016 07.
Article in English | MEDLINE | ID: mdl-27419387

ABSTRACT

While the DNA damage induced by ionizing radiation and by many chemical compounds and drugs is well characterized, the genotoxic insults inflicted by bacteria are only scarcely documented. However, accumulating evidence indicates that we are exposed to bacterial genotoxins. The prototypes of such bacterial genotoxins are the Cytolethal Distending Toxins (CDTs) produced by Escherichia coli and Salmonella enterica serovar Typhi. CDTs display the DNase structure fold and activity, and induce DNA strand breaks in the intoxicated host cell nuclei. E. coli and certain other Enterobacteriaceae species synthesize another genotoxin, colibactin. Colibactin is a secondary metabolite, a hybrid polyketide/nonribosomal peptide compound synthesized by a complex biosynthetic machinery. In this review, we summarize the current knowledge on CDT and colibactin produced by E. coli and/or Salmonella Typhi. We describe their prevalence, genetic determinants, modes of action, and impact in infectious diseases or gut colonization, and discuss the possible involvement of these genotoxigenic bacteria in cancer.


Subject(s)
Bacterial Toxins , Escherichia coli/pathogenicity , Mutagens , Peptides , Polyketides , Salmonella typhi/pathogenicity , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Cycle , DNA Damage , Escherichia coli/metabolism , Gastrointestinal Tract/microbiology , Humans , Mice , Neoplasms/microbiology , Peptides/genetics , Peptides/metabolism , Polyketides/metabolism , Salmonella typhi/metabolism
11.
Acta Vet Hung ; 63(1): 1-10, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25655410

ABSTRACT

Cytolethal distending toxins (CDT) are considered the prototype of inhibitory cyclomodulins, and are produced by a wide range of Gram-negative pathogenic bacteria, including Escherichia coli strains of various sero- and pathotypes. CDT is a heterotripartite toxin consisting of three protein subunits, CdtA, CdtB and CdtC. The active subunit, CdtB has DNase activity and causes DNA damage and cell cycle arrest in the target cell. However, several studies have highlighted different roles for CdtA and CdtC subunits. In order to reveal the necessity of CdtA and CdtC subunit proteins in the CDT-specific phenotype, expression clones containing the cdt-V subunit genes were constructed. Using cell culture assays, we demonstrated that clones expressing only the CdtB subunit or in combination with only CdtA or CdtC were unable to trigger the specific cell cycle arrest and changes in cell morphology in HeLa cells. At the same time, the recombinant clone harbouring the whole cdt-V operon caused all the CDT-associated characteristic phenotypes. All these results verify that all the three CDT subunit proteins are necessary for the genotoxic effect caused by CDT-V.

12.
PLoS One ; 8(5): e63279, 2013.
Article in English | MEDLINE | ID: mdl-23717407

ABSTRACT

Staphylococcus aureus is a highly versatile, opportunistic pathogen and the etiological agent of a wide range of infections in humans and warm-blooded animals. The epithelial surface is its principal site of colonization and infection. In this work, we investigated the cytopathic effect of S. aureus strains from human and animal origins and their ability to affect the host cell cycle in human HeLa and bovine MAC-T epithelial cell lines. S. aureus invasion slowed down cell proliferation and induced a cytopathic effect, resulting in the enlargement of host cells. A dramatic decrease in the number of mitotic cells was observed in the infected cultures. Flow cytometry analysis revealed an S. aureus-induced delay in the G2/M phase transition in synchronous HeLa cells. This delay required the presence of live S. aureus since the addition of the heat-killed bacteria did not alter the cell cycle. The results of Western blot experiments showed that the G2/M transition delay was associated with the accumulation of inactive cyclin-dependent kinase Cdk1, a key inducer of mitosis entry, and with the accumulation of unphosphorylated histone H3, which was correlated with a reduction of the mitotic cell number. Analysis of S. aureus proliferation in asynchronous, G1- and G2-phase-enriched HeLa cells showed that the G2 phase was preferential for bacterial infective efficiency, suggesting that the G2 phase delay may be used by S. aureus for propagation within the host. Taken together, our results divulge the potential of S. aureus in the subversion of key cellular processes such as cell cycle progression, and shed light on the biological significance of S. aureus-induced host cell cycle alteration.


Subject(s)
Epithelial Cells/microbiology , Staphylococcus aureus/physiology , Animals , CDC2 Protein Kinase/metabolism , Cattle , Cell Proliferation , Cell Size , Epithelial Cells/enzymology , Epithelial Cells/physiology , G2 Phase Cell Cycle Checkpoints , HeLa Cells , Histones/metabolism , Host-Pathogen Interactions , Humans , Mitosis , Mitotic Index , Phosphorylation , Protein Processing, Post-Translational , Staphylococcus aureus/pathogenicity
13.
Proc Natl Acad Sci U S A ; 109(27): E1830-8, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22691497

ABSTRACT

The cycle inhibiting factors (Cifs) are a family of translocated effector proteins, found in diverse pathogenic bacteria, that interfere with the host cell cycle by catalyzing the deamidation of a specific glutamine residue (Gln40) in NEDD8 and the related protein ubiquitin. This modification prevents recycling of neddylated cullin-RING ligases, leading to stabilization of various cullin-RING ligase targets, and also prevents polyubiquitin chain formation. Here, we report the crystal structures of two Cif/NEDD8 complexes, revealing a conserved molecular interface that defines enzyme/substrate recognition. Mutation of residues forming the interface suggests that shape complementarity, rather than specific individual interactions, is a critical feature for complex formation. We show that Cifs from diverse bacteria bind NEDD8 in vitro and conclude that they will all interact with their substrates in the same way. The "occluding loop" in Cif gates access to Gln40 by forcing a conformational change in the C terminus of NEDD8. We used native PAGE to follow the activity of Cif from the human pathogen Yersinia pseudotuberculosis and selected variants, and the position of Gln40 in the active site has allowed us to propose a catalytic mechanism for these enzymes.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Photorhabdus/enzymology , Ubiquitins/chemistry , Ubiquitins/metabolism , Yersinia pseudotuberculosis/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Crystallization , Glutamine/genetics , HeLa Cells , Host-Parasite Interactions/physiology , Humans , Molecular Sequence Data , Mutagenesis/physiology , NEDD8 Protein , Oncogene Protein p21(ras)/metabolism , Photorhabdus/genetics , Polyubiquitin/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Ubiquitins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology
14.
Toxins (Basel) ; 3(4): 356-68, 2011 04.
Article in English | MEDLINE | ID: mdl-22069713

ABSTRACT

Cycle inhibiting factors (Cifs) are type III secreted effectors produced by diverse pathogenic bacteria. Cifs are "cyclomodulins" that inhibit the eukaryotic host cell cycle and also hijack other key cellular processes such as those controlling the actin network and apoptosis. This review summarizes current knowledge on Cif since its first characterization in enteropathogenic Escherichia coli, the identification of several xenologues in distant pathogenic bacteria, to its structure elucidation and the recent deciphering of its mode of action. Cif impairs the host ubiquitin proteasome system through deamidation of ubiquitin or the ubiquitin-like protein NEDD8 that regulates Cullin-Ring-ubiquitin Ligase (CRL) complexes. The hijacking of the ubiquitin-dependent degradation pathway of host cells results in the modulation of various cellular functions such as epithelium renewal, apoptosis and immune response. Cif is therefore a powerful weapon in the continuous arm race that characterizes host-bacteria interactions.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Ubiquitins/genetics , Apoptosis , Bacterial Secretion Systems , Cell Cycle , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Escherichia coli Proteins/genetics , HeLa Cells , Host-Pathogen Interactions , Humans , NEDD8 Protein , Signal Transduction , Ubiquitins/metabolism
15.
PLoS Pathog ; 6(9): e1001128, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20941356

ABSTRACT

The cycle inhibiting factors (Cif), produced by pathogenic bacteria isolated from vertebrates and invertebrates, belong to a family of molecules called cyclomodulins that interfere with the eukaryotic cell cycle. Cif blocks the cell cycle at both the G1/S and G2/M transitions by inducing the stabilization of cyclin-dependent kinase inhibitors p21(waf1) and p27(kip1). Using yeast two-hybrid screens, we identified the ubiquitin-like protein NEDD8 as a target of Cif. Cif co-compartmentalized with NEDD8 in the host cell nucleus and induced accumulation of NEDD8-conjugated cullins. This accumulation occurred early after cell infection and correlated with that of p21 and p27. Co-immunoprecipitation revealed that Cif interacted with cullin-RING ubiquitin ligase complexes (CRLs) through binding with the neddylated forms of cullins 1, 2, 3, 4A and 4B subunits of CRL. Using an in vitro ubiquitylation assay, we demonstrate that Cif directly inhibits the neddylated CUL1-associated ubiquitin ligase activity. Consistent with this inhibition and the interaction of Cif with several neddylated cullins, we further observed that Cif modulates the cellular half-lives of various CRL targets, which might contribute to the pathogenic potential of diverse bacteria.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/pathogenicity , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Ubiquitins/metabolism , Actins/metabolism , Animals , Blotting, Western , Cell Cycle , Cell Nucleus/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , NEDD8 Protein , Protein Transport , Rats , Two-Hybrid System Techniques , Ubiquitination , Ubiquitins/genetics
16.
Microbes Infect ; 12(14-15): 1208-18, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20870031

ABSTRACT

Cycle inhibiting factors (Cif) constitute a broad family of cyclomodulins present in bacterial pathogens of invertebrates and mammals. Cif proteins are thought to be type III effectors capable of arresting the cell cycle at G(2)/M phase transition in human cell lines. We report here the first direct functional analysis of Cif(Pl), from the entomopathogenic bacterium Photorhabdus luminescens, in its insect host. The cif(Pl) gene was expressed in P. luminescens cultures in vitro. The resulting protein was released into the culture medium, unlike the well characterized type III effector LopT. During locust infection, cif(Pl) was expressed in both the hemolymph and the hematopoietic organ, but was not essential for P. luminescens virulence. Cif(Pl) inhibited proliferation of the insect cell line Sf9, by blocking the cell cycle at the G(2)/M phase transition. It also triggered host cell death by apoptosis. The integrity of the Cif(Pl) catalytic triad is essential for the cell cycle arrest and pro-apoptotic activities of this protein. These results highlight, for the first time, the dual role of Cif in the control of host cell proliferation and apoptotic death in a non-mammalian cell line.


Subject(s)
Apoptosis , Bacterial Proteins/metabolism , Cell Cycle , Grasshoppers/microbiology , Photorhabdus/pathogenicity , Virulence Factors/metabolism , Animal Structures/microbiology , Animals , Cell Line , Hemolymph/microbiology
17.
Infect Immun ; 77(12): 5471-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19786559

ABSTRACT

The cycle inhibiting factor (Cif) belongs to a family of bacterial toxins, the cyclomodulins, which modulate the host cell cycle. Upon injection into the host cell by the type III secretion system of enteropathogenic Escherichia coli (EPEC), Cif induces both G(2) and G(1) cell cycle arrests. The cell cycle arrests correlate with the accumulation of p21(waf1) and p27(kip1) proteins that inhibit CDK-cyclin complexes, whose activation is required for G(1)/S and G(2)/M transitions. Increases of p21 and p27 levels are independent of p53 transcriptional induction and result from protein stabilization through inhibition of the ubiquitin/proteasome degradation pathway. In this study, we show that Cif not only induces cell cycle arrest but also eventually provokes a delayed cell death. Indeed, 48 h after infection with EPEC expressing Cif, cultured IEC-6 intestinal cells were positive for extracellular binding of annexin V and exhibited high levels of cleaved caspase-3 and lactate dehydrogenase release, indicating evidence of apoptosis. Cif was necessary and sufficient for inducing this late apoptosis, and the cysteine residue of the catalytic site was required for Cif activity. These results highlight a more complex role of Cif than previously thought, as a cyclomodulin but also as an apoptosis inducer.


Subject(s)
Apoptosis , Enteropathogenic Escherichia coli/pathogenicity , Epithelial Cells/microbiology , Escherichia coli Proteins/physiology , Virulence Factors/physiology , Animals , Annexin A5/metabolism , Caspase 3/metabolism , Cell Line , L-Lactate Dehydrogenase/metabolism , Mutagenesis, Site-Directed , Mutant Proteins/metabolism , Rats
18.
Future Microbiol ; 4(7): 867-77, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19722840

ABSTRACT

During coevolution with their hosts, bacteria have developed functions that allow them to interfere with the mechanisms controlling the proliferation of eukaryotic cells. Cycle inhibiting factor (Cif) is one of these cyclomodulins, the family of bacterial effectors that interfere with the host cell cycle. Acquired early during evolution by bacteria isolated from vertebrates and invertebrates, Cif is an effector protein of type III secretion machineries. Cif blocks the host cell cycle in G1 and G2 by inducing the accumulation of the cyclin-dependent kinase inhibitors p21(waf1/cip1) and p27(kip1). The x-ray crystal structure of Cif reveals it to be a divergent member of a superfamily of enzymes including cysteine proteases and acetyltransferases. This review summarizes and discusses what we know about Cif, from the bacterial gene to the host target.


Subject(s)
Bacteria/pathogenicity , Bacterial Proteins/metabolism , Cell Cycle , Eukaryotic Cells/physiology , Virulence Factors/metabolism , Animals , Bacterial Proteins/chemistry , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Protein Structure, Tertiary , Virulence Factors/chemistry
19.
Microbiology (Reading) ; 155(Pt 10): 3214-3225, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628559

ABSTRACT

Enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC) inject a repertoire of effector proteins into host cells via a type III secretion system (T3SS) encoded by the locus of enterocyte effacement (LEE). OspG is an effector protein initially identified in Shigella that was shown to inhibit the host innate immune response. In this study, we found ospG homologues in EHEC (mainly of serogroup O111) and in Yersinia enterocolitica. The T3SS encoded by the LEE was able to inject these different OspG homologues into host cells. Infection of HeLa cells with EHEC O111 inhibited the NF-kappaB-dependent innate immune response via a T3SS-dependent mechanism. However, an EHEC O111 ospG mutant was still able to inhibit NF-kappaB p65 transfer to the nucleus in infected cells stimulated by tumour necrosis factor alpha (TNF-alpha). In addition, no difference in the inflammatory response was observed between wild-type EHEC O111 and the isogenic ospG mutant in the rabbit ligated intestinal loop model. These results suggest that OspG is not the sole effector protein involved in the inactivation of the host innate immune system during EHEC O111 infection.


Subject(s)
Enterohemorrhagic Escherichia coli/immunology , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/physiology , Immunity, Innate , NF-kappa B/antagonists & inhibitors , Virulence Factors/physiology , Amino Acid Sequence , Animals , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/pathology , Gene Order , HeLa Cells , Humans , Intestines/microbiology , Intestines/pathology , Molecular Sequence Data , Rabbits , Sequence Alignment , Serotyping
20.
PLoS One ; 4(3): e4855, 2009.
Article in English | MEDLINE | ID: mdl-19308257

ABSTRACT

The cycle inhibiting factor (Cif) produced by enteropathogenic and enterohemorrhagic Escherichia coli was the first cyclomodulin to be identified that is injected into host cells via the type III secretion machinery. Cif provokes cytopathic effects characterized by G(1) and G(2) cell cycle arrests, accumulation of the cyclin-dependent kinase inhibitors (CKIs) p21(waf1/cip1) and p27(kip1) and formation of actin stress fibres. The X-ray crystal structure of Cif revealed it to be a divergent member of a superfamily of enzymes including cysteine proteases and acetyltransferases that share a conserved catalytic triad. Here we report the discovery and characterization of four Cif homologs encoded by different pathogenic or symbiotic bacteria isolated from vertebrates or invertebrates. Cif homologs from the enterobacteria Yersinia pseudotuberculosis, Photorhabdus luminescens, Photorhabdus asymbiotica and the beta-proteobacterium Burkholderia pseudomallei all induce cytopathic effects identical to those observed with Cif from pathogenic E. coli. Although these Cif homologs are remarkably divergent in primary sequence, the catalytic triad is strictly conserved and was shown to be crucial for cell cycle arrest, cytoskeleton reorganization and CKIs accumulation. These results reveal that Cif proteins form a growing family of cyclomodulins in bacteria that interact with very distinct hosts including insects, nematodes and humans.


Subject(s)
Bacteria/pathogenicity , Bacterial Proteins/physiology , Cell Cycle , Enterobacter/chemistry , Animals , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cytoskeleton/metabolism , Enterobacter/pathogenicity , Escherichia coli Proteins , Interphase , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...