Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
1.
J Physiol Pharmacol ; 60(3): 23-30, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19826178

ABSTRACT

VacA toxin is one of the most important virulence factors produced by H. pylori even though neither its role nor its action mechanisms are completely understood. First considered as a toxin inducing only cell vacuolation, VacA causes apoptosis of gastric epithelial cells by targeting mitochondria. A hotly debated question about VacA action is its relationship with ammonia, which is produced in vivo by H. pylori urease. While ammonia is strictly required for VacA-dependent vacuolation, its role in VacA-induced apoptosis is much less defined. This study was thus aimed to investigate the relationship between VacA toxin and ammonia in H. pylori-induced mitochondrial damage and apoptosis of human gastric epithelial cells in culture by means of flow cytometry. Our results show that, unlike cell vacuolation, in MKN 28 cells neither apoptosis nor dissipation of mitochondrial transmembrane potential induced by VacA require ammonia. Nevertheless, ammonia significantly potentiates both these VacA-induced effects, but independently of the swelling of VacA-containing endosomes (i.e., vacuolation). Our findings make unlikely the hypothesis that ammonia-dependent swelling and rupture of endosomal vesicles in which VacA is sequestered after cell internalization may allow the toxin to reach mitochondria and trigger apoptosis.


Subject(s)
Ammonia/metabolism , Apoptosis , Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Helicobacter pylori/pathogenicity , Stomach/microbiology , Cell Culture Techniques , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Flow Cytometry , Gastric Mucosa/metabolism , Helicobacter pylori/metabolism , Humans , Membrane Potential, Mitochondrial , Microscopy, Phase-Contrast , Mitochondria/metabolism , Mitochondria/microbiology , Mitochondria/pathology , Stomach/pathology , Vacuoles/metabolism , Vacuoles/microbiology , Vacuoles/pathology
2.
J Neurochem ; 90(1): 9-18, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15198662

ABSTRACT

Formation of neurites and their differentiation into axons and dendrites requires precisely controlled changes in the cytoskeleton. While small GTPases of the Rho family appear to be involved in this regulation, it is still unclear how Rho function affects axonal and dendritic growth during development. Using hippocampal neurones at defined states of differentiation, we have dissected the function of RhoA in axonal and dendritic growth. Expression of a dominant negative RhoA variant inhibited axonal growth, whereas dendritic growth was promoted. The opposite phenotype was observed when a constitutively active RhoA variant was expressed. Inactivation of Rho by C3-catalysed ADP-ribosylation using C3 isoforms (Clostridium limosum, C3(lim) or Staphylococcus aureus, C3(stau2)), diminished axonal branching. By contrast, extracellularly applied nanomolar concentrations of C3 from C. botulinum (C3(bot)) or enzymatically dead C3(bot) significantly increased axon growth and axon branching. Taken together, axonal development requires activation of RhoA, whereas dendritic development benefits from its inactivation. However, extracellular application of enzymatically active or dead C3(bot) exclusively promotes axonal growth and branching suggesting a novel neurotrophic function of C3 that is independent from its enzymatic activity.


Subject(s)
Axons/physiology , Dendrites/physiology , Hippocampus/physiology , Neurons/physiology , rho GTP-Binding Proteins/metabolism , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/pharmacology , Adenosine Diphosphate Ribose/metabolism , Animals , Axons/drug effects , Axons/enzymology , Botulinum Toxins/genetics , Botulinum Toxins/metabolism , Botulinum Toxins/pharmacology , Cells, Cultured , Dendrites/drug effects , Dendrites/enzymology , Gene Transfer Techniques , Genes, Dominant , Hippocampus/enzymology , Hippocampus/ultrastructure , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/pharmacology , Mice , Mice, Inbred Strains , Neurons/enzymology , Neurons/ultrastructure , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics
3.
Biochem Biophys Res Commun ; 289(2): 623-9, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716521

ABSTRACT

Tetanus neurotoxin (TeNT) produced by Clostridium tetani specifically cleaves VAMP/synaptobrevin (VAMP) in central neurons, thereby causing inhibition of neurotransmitter release and ensuing spastic paralysis. Although polysialogangliosides act as components of the neurotoxin binding sites on neurons, evidence has accumulated indicating that a protein moiety is implicated as a receptor of TeNT. We have observed that treatment of cultured mouse neuronal cells with the phosphatidylinositol-specific phospholipase C (PIPLC) inhibited TeNT-induced cleavage of VAMP. Also, we have shown that the blocking effects of TeNT on neuroexocytosis can be prevented by incubation of Purkinje cell preparation with PIPLC. In addition, treatment of cultured mouse neuronal cells with cholesterol sequestrating agents such as nystatin and filipin, which disrupt clustering of GPI-anchored proteins in lipid rafts, prevented intraneuronal VAMP cleavage by TeNT. Our results demonstrate that high sensitivity of neurons to TeNT requires rafts and one or more GPI-anchored protein(s) which act(s) as a pivotal receptor for the neurotoxin.


Subject(s)
Neurons/metabolism , Tetanus Toxin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Binding Sites , Cells, Cultured , Cerebellum/metabolism , Cytosol/chemistry , Dose-Response Relationship, Drug , Electrophysiology , Endocytosis , Filipin/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/physiology , Nystatin/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Protein Binding , Protein Structure, Tertiary , Purkinje Cells/metabolism , Spinal Cord/embryology , Time Factors , Type C Phospholipases/metabolism
4.
Mol Microbiol ; 41(6): 1237-47, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11580831

ABSTRACT

The Rho-GTPases-activating toxin CNF1 (cytotoxic necrotizing factor 1) delivers its catalytic activity into the cytosol of eukaryotic cells by a low pH membrane translocation mechanism reminiscent of that used by diphtheria toxin (DT). As DT, CNF1 exhibits a translocation domain (T) containing two predicted hydrophobic helices (H1-2) (aa 350-412) separated by a short peptidic loop (CNF1-TL) (aa 373-386) with acidic residues. In the DT loop, the loss of charge of acidic amino acids, as a result of protonation at low pH, is a critical step in the transfer of the DT catalytic activity into the cytosol. To determine whether the CNF1 T domain operates similarly to the DT T domain, we mutated several ionizable amino acids of CNF1-TL to lysine. Single substitutions such as D373K or D379K strongly decreased the cytotoxic effect of CNF1 on HEp-2 cells, whereas the double substitution D373K/D379K induced a nearly complete loss of cytotoxic activity. These single or double substitutions did not modify the cell-binding, enzymatic or endocytic activities of the mutant toxins. Unlike the wild-type toxin, single- or double-substituted CNF1 molecules bound to the HEp-2 plasma membrane could not translocate their enzymatic activity directly into the cytosol following a low pH pulse.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cytotoxins/chemistry , Cytotoxins/genetics , Escherichia coli Proteins , Amino Acid Substitution , Bacterial Toxins/metabolism , Base Sequence , Biological Transport, Active , Cell Line , Cell Membrane/metabolism , Cytotoxins/metabolism , DNA Primers/genetics , Endocytosis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Humans , Hydrogen-Ion Concentration , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
J Biol Chem ; 276(50): 46849-55, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11591701

ABSTRACT

Cyclooxygenase (COX)-2 and COX-1 play an important role in prostacyclin production in vessels and participate in maintaining vascular homeostasis. Statins are inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase, which is crucial in cholesterol biosynthesis. Recently, cholesterol-independent effects of statins have been described. In this study, we evaluated the effect of two inhibitors of HMG CoA reductase, mevastatin and lovastatin, on the production of prostacyclin and the expression of COX in human aortic smooth muscle cells. Treatment of cells with 25 microm mevastatin or lovastatin resulted in the induction of COX-2 and increase in prostacyclin production. Mevalonate, the direct metabolite of HMG CoA reductase, and geranylgeranyl-pyrophosphate reversed this effect. GGTI-286, a selective inhibitor of geranylgeranyltransferases, increased COX-2 expression and prostacyclin formation, thus indicating the involvement of geranylgeranylated proteins in the down-regulation of COX-2. Furthermore, Clostridium difficile toxin B, an inhibitor of the Rho GTP-binding protein family, the Rho selective inhibitor C3 transferase, and Y-27632, a selective inhibitor of the Rho-associated kinases, targets of Rho A, increased COX-2 expression whereas the activator of the Rho GTPase, the cytotoxic necrotizing factor 1, blocked interlukin-1alpha-dependent COX-2 induction. These results demonstrate that statins up-regulate COX-2 expression and subsequent prostacyclin formation in human aortic smooth muscle cells in part through inhibition of Rho.


Subject(s)
Aorta/enzymology , Bacterial Proteins , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Isoenzymes/biosynthesis , Leucine/analogs & derivatives , Lovastatin/analogs & derivatives , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Amides/pharmacology , Aorta/metabolism , Apoptosis , Bacterial Toxins/pharmacology , Blotting, Northern , Blotting, Western , Cells, Cultured , Cyclooxygenase 2 , DNA, Complementary/metabolism , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epoprostenol/biosynthesis , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Inhibitory Concentration 50 , Interleukin-1/metabolism , Leucine/pharmacology , Lovastatin/pharmacology , Membrane Proteins , Mevalonic Acid/chemistry , Mevalonic Acid/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Prenylation , Pyridines/pharmacology , Time Factors , rho GTP-Binding Proteins/metabolism
6.
Toxicon ; 39(11): 1673-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11595630

ABSTRACT

The cytotoxic necrotizing factor 1, from uropathogenic Escherichia coli, is the paradigm of Rho-GTPases-activating bacterial toxins. CNF1 is a MW 108kDa A-B protein toxin divided into three domains which are implicated in the three steps of the intoxication process. The N-terminal domain contains the cell receptor function and binds with high affinity to a cell receptor not yet identified. Binding of the toxin is followed by its internalization by endocytosis and its transport into late endosomes. The middle toxin domain contains two hydophobic helices which allow translocation of the toxin across the membrane upon acidification in late endosomes. Finally the carboxy-terminal domain of CNF1 is an enzyme which deamidates Rho-GTP-binding proteins (Rho, Rac and Cdc42) glutamine 63 (for Rho) or glutamine 61 (for Rac and Cdc42). Deamidation of glutamine 63/61 blocks the intrinsic or the GTPase activating protein (GAP)-induced hydrolysis of GTP leading to the permanent activation of the GTPase. Activation of Rho GTPases by CNF1 induces a profound reorganization of the cell actin cytoskeleton. By its properties on Rho GTPases CNF1 is to date an invaluable tool for cell biology studies.


Subject(s)
Bacterial Toxins/toxicity , Cytotoxins/toxicity , Escherichia coli Proteins , Escherichia coli/metabolism , Animals , Bacterial Toxins/genetics , Cells/drug effects , Cells/metabolism , Cytotoxins/genetics , Escherichia coli/genetics , Humans , Structure-Activity Relationship , rho GTP-Binding Proteins/metabolism
7.
Nat Struct Biol ; 8(7): 584-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11427886

ABSTRACT

Certain uropathogenic and neonatal meningitis-causing strains of Escherichia coli express a 114 kDa protein toxin called cytotoxic necrotizing factor 1 (CNF1). The toxin causes alteration of the host cell actin cytoskeleton and promotes bacterial invasion of blood-brain barrier endothelial cells. CNF1 belongs to a unique group of large cytotoxins that cause constitutive activation of Rho guanosine triphosphatases (GTPases), which are key regulators of the actin cytoskeleton. This group also includes E. coli cytotoxic necrotizing factor 2 (CNF2, 114 kDa) and dermonecrotic toxins (DNT, 159 kDa) of Bordetella spp. with related sequences occurring in Yersinia spp. Here we show that the catalytic region of CNF1 exhibits a novel protein fold as determined by its 1.83 A resolution crystal structure. The structure reveals that CNF1 has a Cys-His-main chain oxygen catalytic triad reminiscent of enzymes belonging to the catalytic triad superfamily. The position of the catalytic Cys residue at the base of a deep pocket restricts access to potential substrates and helps explain the high specificity of this and related toxins.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cytotoxins/chemistry , Cytotoxins/metabolism , Escherichia coli Proteins , Escherichia coli/chemistry , rho GTP-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Bacterial Toxins/genetics , Binding Sites , Catalytic Domain , Conserved Sequence/genetics , Crystallography, X-Ray , Cysteine/genetics , Cysteine/metabolism , Cytotoxins/genetics , Enzyme Activation , Escherichia coli/genetics , Histidine/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity
8.
Lab Invest ; 81(3): 375-84, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11310830

ABSTRACT

A prominent histologic feature of Helicobacter pylori infection is a dense infiltration of polymorphonuclear leukocytes (PMNL) in gastric mucosa. H. pylori lipopolysaccharide (LPS) has been recognized as a primary virulence factor evoking acute mucosal inflammatory reaction. Previous works have shown that H. pylori LPS immunologic activities are lower than those of enterobacterial LPS. However, the effect of H. pylori LPS on spontaneous PMNL apoptosis, and mechanisms by which this H. pylori LPS may promote PMNL survival remain to be established. In this study, we investigated, by both morphologic and biochemical approaches, the action of H. pylori LPS on PMNL apoptosis in vitro, using broth culture filtrates (BCF) of H. pylori strains with different genotypes. We found that BCF from H. pylori caused a significant delay in spontaneous PMNL apoptosis and this delay was independent of the VacA, cag pathogenicity island and urease status. We demonstrated that LPS in BCF is responsible for this effect because it was abrogated by the LPS antagonist B287 (a synthetic analog of Rhodobactersphaeroides lipid A). Moreover, BCF from H. pylori induced P42/44MAP kinase activation in PMNL. Similar results were obtained with BCF of an Escherichia coli strain. Taken together these data suggest that longer survival of PMNL induced by H. pylori LPS may increase gastric epithelium injury in H. pylori-associated diseases.


Subject(s)
Apoptosis/immunology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Lipopolysaccharides/immunology , Neutrophils/cytology , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA, Bacterial/analysis , Enzyme Activation/immunology , Enzyme Precursors/metabolism , Helicobacter Infections/immunology , Helicobacter pylori/genetics , Humans , In Situ Nick-End Labeling , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/enzymology , Neutrophils/immunology , Virulence
9.
J Bacteriol ; 183(4): 1147-58, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157926

ABSTRACT

In Escherichia coli, the DsbA'-PhoA hybrid proteins carrying an unfoldable DsbA' fragment can be targeted to the envelope, where they exert their toxicity. Hybrid proteins stick to the periplasmic face of the inner membrane and paralyze the export mechanism, becoming lethal if sufficiently overproduced and if not degraded by the DegP protease (A. Guigueno, P. Belin, and P. L. Boquet, J. Bacteriol. 179:3260-3269, 1997). We isolated a multicopy suppressor that restores viability to a degP strain without modifying the expression level of the toxic fusion. Suppression does not involve activation of the known envelope stress-combative pathways, the Cpx pathway and the sigma(E) regulon. Subclone analysis of the suppressor revealed a 195-bp DNA fragment that is responsible for toxicity suppression. The cloned gene, called uptR, is approximately 130 bp long (including the promoter and a transcription termination signal) and is transcribed into a small RNA (92 nucleotides). Using site-directed mutagenesis, we found that UptR RNA does not require translation for toxicity suppression. UptR-mediated action reduces the amount of membrane-bound toxic hybrid protein. UptR RNA is the first example of a small RNA implicated in extracytoplasmic toxicity suppression. It appears to offer a new way of suppressing toxicity, and its possible modes of action are discussed.


Subject(s)
Bacterial Proteins/toxicity , Escherichia coli/genetics , Periplasm/metabolism , Protein Folding , Protein Transport/genetics , RNA, Bacterial/biosynthesis , Suppression, Genetic , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/toxicity , Bacterial Proteins/metabolism , Base Sequence , Cloning, Molecular , DNA, Ribosomal , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Complementation Test , Membrane Proteins/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/toxicity , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/toxicity , Sequence Analysis, DNA , Sigma Factor/metabolism , Transcription Factors/metabolism
10.
EMBO J ; 19(23): 6361-70, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11101509

ABSTRACT

The pathogenic bacterium Helicobacter pylori produces the cytotoxin VacA, which is implicated in the genesis of gastric epithelial lesions. By transfect ing HEp-2 cells with DNAs encoding either the N-terminal (p34) or the C-terminal (p58) fragment of VacA, p34 was found localized specifically to mitochondria, whereas p58 was cytosolic. Incubated in vitro with purified mitochondria, VacA and p34 but not p58 translocated into the mitochondria. Microinjection of DNAs encoding VacA-GFP and p34-GFP, but not GFP-VacA or GFP-p34, induced cell death by apoptosis. Transient transfection of HeLa cells with p34-GFP or VacA-GFP induced the release of cytochrome c from mitochondria and activated the executioner caspase 3, as determined by the cleavage of poly(ADP-ribose) polymerase (PARP). PARP cleavage was antagonized specifically by co-transfection of DNA encoding Bcl-2, known to block mitochondria-dependent apoptotic signals. The relevance of these observations to the in vivo mechanism of VacA action was supported by the fact that purified activated VacA applied externally to cells induced cytochrome c release into the cytosol.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome c Group/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Caspase 3 , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , Digitonin/pharmacology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Green Fluorescent Proteins , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Microscopy, Electron , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , Reticulocytes/metabolism , Stomach Diseases/metabolism , Transfection
13.
Mol Biol Cell ; 11(11): 3897-909, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11071915

ABSTRACT

Helicobacter pylori vacuolating toxin (VacA) causes vacuolation in a variety of cultured cell lines, sensitivity to VacA differing greatly, however, among the different cell types. We found that the high sensitivity of HEp-2 cells to VacA was impaired by treating the cells with phosphatidylinositol-specific phospholipase C (PI-PLC) which removes glycosylphosphatidylinositol (GPI)-anchored proteins from the cell surface. Incubation of cells with a cholesterol-sequestering agent, that impairs both structure and function of sphingolipid-cholesterol-rich membrane microdomains ("lipid rafts"), also impaired VacA-induced cell vacuolation. Overexpression into HEp-2 cells of proteins inhibiting clathrin-dependent endocytosis (i.e., a dominant-negative mutant of Eps15, the five tandem Src-homology-3 domains of intersectin, and the K44A dominant-negative mutant of dynamin II) did not affect vacuolation induced by VacA. Nevertheless, F-actin depolymerization, known to block the different types of endocytic mechanisms, strongly impaired VacA vacuolating activity. Taken together, our data suggest that the high cell sensitivity to VacA depends on the presence of one or several GPI-anchored protein(s), intact membrane lipid rafts, and an uptake mechanism via a clathrin-independent endocytic pathway.


Subject(s)
Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Clathrin/metabolism , Endocytosis/drug effects , Phosphatidylinositols/metabolism , Actin Cytoskeleton/drug effects , Actins/drug effects , Animals , Bacterial Proteins/metabolism , CHO Cells/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line/drug effects , Cricetinae , Cytochalasin D/pharmacology , Dogs , Dose-Response Relationship, Drug , Endocytosis/physiology , Humans , Iodine Radioisotopes , Nystatin/pharmacology , Proteins/drug effects , Proteins/metabolism , Type C Phospholipases/pharmacology , Vacuoles/drug effects
14.
J Leukoc Biol ; 68(4): 522-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11037974

ABSTRACT

Recruitment of polymorphonuclear leukocytes (PMNL) is a hallmark of both urinary and digestive infections caused by Escherichia coli. Cytotoxic necrotizing factor 1 (CNF-1) is a toxin produced by uropathogenic E. coli strains that mediates its effects via the activation of small GTP-binding proteins. However, the role and the consequences of CNF-1 on PMNL physiology remain largely unknown. In this study, we provide evidence that CNF-1 dramatically affects the PMNL cytoskeleton architecture by inducing an increased content of F-actin. Furthermore, we demonstrate that CNF-1 increases functional features of PMNL, such as superoxide generation and adherence on epithelial T84 monolayers, but significantly decreases their phagocytic function. Our results suggest that CNF-1 may behave as a virulence factor in urinary or digestive infection by stimulating PMNL cytotoxicity as a result of its enhancing effect on their adherence to epithelial cells as well as the production of radical oxygen products. Moreover, the decreased phagocytosis of PMNL induced by CNF-1 likely facilitates growth of bacteria. In these conditions, CNF-1 would intervene in the initiation and in the perpetuation of the inflammatory process.


Subject(s)
Bacterial Toxins/pharmacology , Cytotoxins/pharmacology , Escherichia coli Proteins , Escherichia coli/pathogenicity , Neutrophils/drug effects , Phagocytosis/drug effects , Respiratory Burst/drug effects , Actins/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Epithelial Cells/cytology , Humans , Inflammation/chemically induced , Intestinal Mucosa/cytology , Macrophage-1 Antigen/metabolism , Neutrophils/microbiology , Neutrophils/physiology , Neutrophils/ultrastructure , Opsonin Proteins/pharmacology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Virulence , Zymosan/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/physiology
16.
Microbes Infect ; 2(7): 837-43, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10955965

ABSTRACT

Many bacterial toxins and bacterial enzymes modify small GTPases. Toxins exhibit different enzymatic activities on either the switch 1 or switch 2 domains of these small GTPases leading to inactivation or activation of such intracellular timer molecules. In addition, some virulence factors of certain invasive bacteria such as Salmonella also modulate small GTP binding proteins either by mimicking GTPase exchange factors or GTPase activating proteins.


Subject(s)
Bacteria/pathogenicity , Bacterial Toxins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Animals , Humans , Virulence , rho GTP-Binding Proteins/metabolism
17.
Infect Immun ; 68(9): 5225-33, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10948148

ABSTRACT

Helicobacter pylori infection can induce polymorphonuclear leukocyte (PMNL) infiltration of the gastric mucosa, which characterizes acute chronic gastritis. The mechanisms underlying this process are poorly documented. The lack of an in vitro model has considerably impaired the study of transepithelial migration of PMNL induced by H. pylori. In the present work, we used confluent polarized monolayers of the human intestinal cell line T84 grown on permeable filters to analyze the epithelial PMNL response induced by broth culture filtrates (BCFs) and bacterial suspensions from different strains of H. pylori. We have evaluated the role of the vacuolating cytotoxin VacA and of the cag pathogenicity island (PAI) of H. pylori in PMNL migration via their effects on T84 epithelial cells. We noted no difference in the rates of PMNL transepithelial migration after epithelial preincubation with bacterial suspensions or with BCFs of VacA-negative or VacA-positive H. pylori strains. In contrast, PMNL transepithelial migration was induced after incubation of the T84 cells with cag PAI-positive and cagE-positive H. pylori strains. Finally, PMNL migration was correlated with a basolateral secretion of interleukin-8 by T84 cells, thus creating a subepithelial chemotactic gradient for PMNL. These data provide evidence that the vacuolating cytotoxin VacA is not involved in PMNL transepithelial migration and that the cag PAI, with a pivotal role for the cagE gene, provokes a transcellular signal across T84 monolayers, inducing a subepithelial PMNL response.


Subject(s)
Antigens, Bacterial , Bacterial Proteins/physiology , Helicobacter pylori/pathogenicity , Neutrophils/physiology , Bacterial Adhesion , Cell Movement , Cell Polarity , Humans , Interleukin-8/physiology , Tumor Cells, Cultured
18.
Mol Microbiol ; 37(2): 433-42, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10931337

ABSTRACT

Helicobacter pylori VacA is a secreted toxin that induces multiple structural and functional alterations in eukaryotic cells. Exposure of VacA to either acidic or alkaline pH ('activation') results in structural changes in the protein and a marked enhancement of its cell-vacuolating activity. However, the mechanism by which activation leads to increased cytotoxicity is not well understood. In this study, we analysed the binding and internalization of [125I]-VacA by HeLa cells. We detected no difference in the binding of untreated and activated [125I]-VacA to cells. Binding of acid-activated [125I]-VacA to cells at 4 degrees C was not saturable, and was only partially inhibited by excess unlabelled toxin. These results suggest that VacA binds either non-specifically or to an abundant, low-affinity receptor on HeLa cells. To study internalization of VacA, we used a protease protection assay. Analysis by SDS-PAGE and autoradiography indicated that the intact 87 kDa toxin was internalized in a time-dependent process at 37 degrees C but not at 4 degrees C. Furthermore, internalization of the intact toxin was detected only if VacA was acid or alkaline activated before being added to cells. The internalization of activated [125I]-VacA was not substantially inhibited by the presence of excess unlabelled toxin, but was blocked if cells were depleted of cellular ATP by the addition of sodium azide and 2-deoxy-D-glucose. These results indicate that acid or alkaline pH-induced structural changes in VacA are required for VacA entry into cells, and that internalization of the intact 87 kDa toxin is required for VacA cytotoxicity.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cytotoxins/metabolism , Helicobacter pylori/metabolism , Transcriptional Activation , Vacuoles/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration
19.
Mol Biol Cell ; 11(5): 1775-87, 2000 May.
Article in English | MEDLINE | ID: mdl-10793151

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1), a protein produced by pathogenic strains of Escherichia coli, activates the p21 Rho-GTP-binding protein, inducing a profound reorganization of the actin cytoskeleton. CNF1 binds to its cell surface receptor on HEp-2 cells with high affinity (K(d) = 20 pM). In HEp-2 cells the action of CNF1 is not blocked in the presence of filipin, a drug described to reduce cholera toxin internalization by the caveolae-like mechanism. Moreover, HEp-2 cells, which express a dominant negative form of proteins that impair the formation of clathrin coated-vesicles and internalization of transferrin (Eps15, dynamin or intersectin-Src homology 3), are still sensitive to CNF1. In this respect, the endocytosis of CNF1 is similar to the plant toxin ricin. However, unlike ricin toxin, CNF1 does not cross the Golgi apparatus and requires an acidic cell compartment to transfer its enzymatic activity into the cytosol in a manner similar to that required by diphtheria toxin. As shown for diphtheria toxin, the pH-dependent membrane translocation step of CNF1 could be mimicked at the level of the plasma membrane by a brief exposure to a pH of

Subject(s)
Bacterial Toxins/metabolism , Caveolins , Clathrin/metabolism , Cytosol/metabolism , Cytotoxins/metabolism , Endocytosis/physiology , Escherichia coli Proteins , Animals , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Binding Sites , Biological Transport , Catalytic Domain , Caveolin 1 , Cell Compartmentation , Cell Membrane/metabolism , Coated Vesicles/metabolism , Cytotoxins/genetics , Cytotoxins/pharmacokinetics , Dogs , Endocytosis/drug effects , Golgi Apparatus/metabolism , Humans , Membrane Proteins/metabolism , Microtubules/metabolism , Receptors, Cell Surface/metabolism , Tumor Cells, Cultured
20.
Lett Appl Microbiol ; 30(3): 213-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10747253

ABSTRACT

The presence of cytotoxic necrotizing factor 1 (CNF1), together with various associated virulence factors (alpha-haemolysin, P-, S- and A-fimbriae), was screened in 175 uropathogenic Escherichia coli strains isolated from hospitalized adult patients. The cnf1 gene was detected in 30% of the selected strains independently of the severity of the clinical urinary infection. A significant association between CNF1, haemolytic activity and the products of the pap/sfa genes was found. However, CNF1 appeared not to play a major role in nosocomial E. coli urinary tract infections.


Subject(s)
Bacterial Toxins/metabolism , Cross Infection/microbiology , Cytotoxins/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins , Escherichia coli/isolation & purification , Repressor Proteins , Transcription Factors , Urinary Tract Infections/microbiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Cross Infection/urine , Cytotoxins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/urine , Female , Genes, Bacterial , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Humans , Male , Middle Aged , Mutation , Polymerase Chain Reaction , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...