Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cell Microbiol ; 15(9): 1560-71, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23480519

ABSTRACT

The first step in attachment of Chlamydia to host cells is thought to involve reversible binding to host heparan sulfate proteoglycans (HSPGs), polymers of variably sulfated repeating disaccharide units coupled to diverse protein backbones. However, the key determinants of HSPG structure that are involved in Chlamydia binding are incompletely defined. A previous genome-wide Drosophila RNAi screen suggested that the level of HSPG 6-O sulfation rather than the identity of the proteoglycan backbone maybe a critical determinant for binding. Here, we tested in mammalian cells whether SULF1 or SULF2, human endosulfatases, which remove 6-O sulfates from HSPGs, modulate Chlamydia infection. Ectopic expression of SULF1 or SULF2 in HeLa cells, which decreases cell surface HSPG sulfation, diminished C. muridarum binding and decreased vacuole formation. ShRNA depletion of endogenous SULF2 in a cell line that primarily expresses SULF2 augmented binding and increased vacuole formation. C. muridarum infection of diverse cell lines resulted indownregulation of SULF2 mRNA. In a murine model of acute pneumonia, mice genetically deficient in both endosulfatases or in SULF2 alone demonstrated increased susceptibility to C. muridarum lung infection. Collectively, these studies demonstrate that the level of HSPG 6-O sulfation is a critical determinant of C. muridarum infection in vivo and that 6-O endosulfatases are previously unappreciated modulators of microbial pathogenesis.


Subject(s)
Bacterial Adhesion , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Heparitin Sulfate/metabolism , Sulfotransferases/immunology , Animals , Chlamydia Infections/microbiology , Chlamydia muridarum/growth & development , Disease Models, Animal , Disease Susceptibility , HeLa Cells , Humans , Mice , Mice, Knockout , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Sulfatases/deficiency , Sulfatases/immunology , Sulfotransferases/deficiency , Sulfotransferases/metabolism
2.
Cell Microbiol ; 7(5): 725-39, 2005 May.
Article in English | MEDLINE | ID: mdl-15839901

ABSTRACT

Chlamydia spp. are major causes of important human diseases, but dissecting the host-pathogen interactions has been hampered by the lack of bacterial genetics and the difficulty in carrying out forward genetic screens in mammalian hosts. RNA interference (RNAi)-based methodologies for gene inactivation can now be easily carried out in genetically tractable model hosts, such as Drosophila melanogaster, and offer a new approach to identifying host genes required for pathogenesis. We tested whether Chlamydia trachomatis infection of D. melanogaster S2 cells recapitulated critical aspects of mammalian cell infections. As in mammalian cells, C. trachomatis entry was greatly reduced by heparin and cytochalasin D. Inclusions were formed in S2 cells, acquired Golgi-derived sphingolipids, and avoided phagolysosomal fusion. Elementary body (EB) to reticulate body (RB) differentiation was observed, however, no RB to EB development or host cell killing was observed. RNAi-mediated inactivation of Rac, a Rho GTPase recently shown to be required for C. trachomatis entry in mammalian cells, inhibits C. trachomatis infection in S2 cells. We conclude that Drosophila S2 cells faithfully mimic early events in Chlamydia host cell interactions and provides a bona fide system to systematically dissect host functions important in the pathogenesis of obligate intracellular pathogens.


Subject(s)
Chlamydia Infections/parasitology , Chlamydia trachomatis/metabolism , Disease Models, Animal , Drosophila melanogaster/cytology , Drosophila melanogaster/parasitology , Actins/physiology , Animals , Cells, Cultured , Chlamydia trachomatis/ultrastructure , Cytochalasin D/pharmacology , Drosophila melanogaster/ultrastructure , Golgi Apparatus/metabolism , Heparin/pharmacology , Humans , Lysosomes/parasitology , Lysosomes/ultrastructure , Microscopy, Electron, Transmission , Sphingomyelins/metabolism
3.
Mol Biol Cell ; 16(5): 2577-85, 2005 May.
Article in English | MEDLINE | ID: mdl-15772151

ABSTRACT

Several Pseudomonas aeruginosa strains are internalized by epithelial cells in vitro and in vivo, but the host pathways usurped by the bacteria to enter nonphagocytic cells are not clearly understood. Here, we report that internalization of strain PAK into epithelial cells triggers and requires activation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B/Akt (Akt). Incubation of Madin-Darby canine kidney (MDCK) or HeLa cells with the PI3K inhibitors LY294002 (LY) or wortmannin abrogated PAK uptake. Addition of the PI3K product phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3] to polarized MDCK cells was sufficient to increase PAK internalization. PtdIns(3,4,5)P3 accumulated at the site of bacterial binding in an LY-dependent manner. Akt phosphorylation correlated with PAK invasion. The specific Akt phosphorylation inhibitor SH-5 inhibited PAK uptake; internalization also was inhibited by small interfering RNA-mediated depletion of Akt phosphorylation. Expression of constitutively active Akt was sufficient to restore invasion when PI3K signaling was inhibited. Together, these results demonstrate that the PI3K signaling pathway is necessary and sufficient for the P. aeruginosa entry and provide the first example of a bacterium that requires Akt for uptake into epithelial cells.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Animals , Cell Line , Dogs , Enzyme Activation , Epithelial Cells/microbiology , HeLa Cells , Humans , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol Phosphates/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/genetics , Signal Transduction
4.
Infect Immun ; 72(12): 7367-73, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557670

ABSTRACT

Using pharmacologic and biochemical criteria, we evaluated whether uptake of four different Chlamydia trachomatis serovars, D, E, K, and L2, was dependent upon lipid rafts. Our data suggest that lipid raft-mediated entry is not required for C. trachomatis infection of cultured epithelial cells.


Subject(s)
Chlamydia trachomatis/pathogenicity , Membrane Microdomains/physiology , Caveolin 1 , Caveolins/analysis , Cholera Toxin/pharmacology , G(M1) Ganglioside/analysis , HeLa Cells , Humans , Porins/analysis , beta-Cyclodextrins/pharmacology
5.
Infect Immun ; 72(1): 546-58, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688136

ABSTRACT

ExoT is a type III secreted effector protein found in almost all strains of Pseudomonas aeruginosa and is required for full virulence in an animal model of acute pneumonia. It is comprised of an N-terminal domain with GTPase activating protein (GAP) activity towards Rho family GTPases and a C-terminal ADP ribosyltransferase (ADPRT) domain with minimal activity towards a synthetic substrate in vitro. Consistent with its activity as a Rho family GTPase, ExoT has been shown to inhibit P. aeruginosa internalization into epithelial cells and macrophages, disrupt the actin cytoskeleton through a Rho-dependent pathway, and inhibit wound repair in a scrape model of injured epithelium. We have previously shown that mutation of the invariant arginine of the GAP domain to lysine (R149K) results in complete loss of GAP activity in vitro but only partially inhibits ExoT anti-internalization and cell rounding activity. We have constructed in-frame deletions and point mutations within the ADPRT domain in order to test whether this domain might account for the residual activity observed in ExoT GAP mutants. Deletion of a majority of the ADPRT domain (residues 234 to 438) or point mutations of the ADPRT catalytic site (residues 383 to 385) led to distinct changes in host cell morphology and substantially reduced the ability of ExoT to inhibit in vitro epithelial wound healing over a 24-h period. In contrast, only subtle effects on the efficiency of ExoT-induced bacterial internalization were observed in the ADPRT mutant forms. Expression of each domain individually in Saccharomyces cerevisiae was toxic, whereas expression of each of the catalytically inactive mutant domains was not. Collectively, these data demonstrate that the ADPRT domain of ExoT is active in vivo and contributes to the pathogenesis of P. aeruginosa infections.


Subject(s)
ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/toxicity , Pseudomonas aeruginosa/pathogenicity , ADP Ribose Transferases/genetics , Actins/metabolism , Animals , Cell Line , Cytoskeleton/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , GTPase-Activating Proteins , Gene Deletion , HeLa Cells , Humans , Mass Spectrometry , Point Mutation , Transfection , Wound Healing
6.
Infect Immun ; 70(4): 2198-205, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11895987

ABSTRACT

The Pseudomonas aeruginosa protein ExoT is a bacterial GTPase-activating protein (GAP) that has in vitro activity toward Rho, Rac, and Cdc42 GTPases. Expression of ExoT both inhibits the internalization of strain PA103 by macrophages and epithelial cells and is associated with morphological changes (cell rounding and detachment) of infected cells. We find that expression of ExoT leads to the loss of GTP-bound RhoA, Rac1, and Cdc42 in transfected HeLa cells, demonstrating that ExoT has GAP activity in vivo toward all three GTPases. GAP activity is absolutely dependent on the presence of arginine at position 149 but is not affected by whether ExoT is expressed in the absence or presence of other P. aeruginosa type III secreted proteins. We also demonstrate that expression of ExoT in epithelial cells is sufficient to cause stress fiber disassembly by means of ExoT's GAP activity toward RhoA.


Subject(s)
Bacterial Toxins/metabolism , GTPase-Activating Proteins/metabolism , Pseudomonas aeruginosa/pathogenicity , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cytoskeleton/metabolism , Cytotoxins , Dogs , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Pseudomonas aeruginosa/metabolism
7.
Annu Rev Microbiol ; 55: 407-35, 2001.
Article in English | MEDLINE | ID: mdl-11544362

ABSTRACT

Many pathogens must surmount an epithelial cell barrier in order to establish an infection. While much has been learned about the interaction of bacterial pathogens with cultured epithelial cells, the influence of cell polarity on these events has only recently been appreciated. This review outlines bacterial-host epithelial cell interactions in the context of the distinct apical and basolateral surfaces of the polarized epithelium that lines the lumens of our organs.


Subject(s)
Bacteria/metabolism , Epithelial Cells/metabolism , Bacteria/pathogenicity , Cell Polarity , Epithelial Cells/microbiology , Virulence
8.
Cell Microbiol ; 3(4): 223-36, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298646

ABSTRACT

The nosocomial pathogen Pseudomonas aeruginosa causes clinical infection in the setting of pre-existing epithelial tissue damage, an association that is mirrored by the increased ability of P. aeruginosa to bind, invade and damage injured epithelial cells in vitro. In this study, we report that P. aeruginosa inhibits the process of epithelial wound repair in vitro through the type III-secreted bacterial protein ExoT, a GTPase-activating protein (GAP) for Rho family GTPases. This inhibition primarily targets cells at the edge of the wound, and causes actin cytoskeleton collapse, cell rounding and cell detachment. ExoT-dependent inhibition of wound repair is mediated through the GAP activity of this bacterial protein, as mutations in ExoT that alter the conserved arginine (R149) within the GAP domain abolish the ability of P. aeruginosa to inhibit wound closure. Because ExoT can also inhibit P. aeruginosa internalization by phagocytes and epithelial cells, this protein may contribute to the in vivo virulence of P. aeruginosa by allowing organisms both to overcome local host defences, such as an intact epithelial barrier, and to evade phagocytosis by immune effector cells.


Subject(s)
Bacterial Toxins/pharmacology , Cytotoxins/pharmacology , GTPase-Activating Proteins/physiology , Pseudomonas aeruginosa/pathogenicity , Wound Healing , Actin Cytoskeleton/microbiology , Actins/metabolism , Arginine/metabolism , Bacterial Toxins/genetics , Cells, Cultured , Epithelial Cells/pathology , Focal Adhesions , GTPase-Activating Proteins/genetics , Humans , Lung/microbiology , Lung/pathology , Phagocytosis , Point Mutation , Pseudomonas aeruginosa/genetics , Virulence
9.
Cell Microbiol ; 3(2): 85-98, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11207623

ABSTRACT

The Gram-negative pathogen Pseudomonas aeruginosa invades epithelial cells in vivo and in vitro. We have examined the pathway(s) by which epithelial cells internalize P. aeruginosa strain PA103 using Madin-Darby canine kidney (MDCK) cells. We have recently demonstrated that P. aeruginosa internalization occurs by an actin-dependent Toxin B-inhibited pathway which becomes downregulated as epithelial cells become polarized, suggesting that one or more of the Rho family GTPases is involved in bacterial internalization. Here, we demonstrate that activation of the Rho family GTPases by cytotoxic necrotizing factor 1 (CNF-1) stimulates P. aeruginosa internalization. Examination of the roles of the individual Rho family GTPases in internalization shows that expression of a constitutively active allele of RhoA (RhoAV14), but not of constitutively active Rac1 (Rac1V12) or Cdc42 (Cdc42V12), is sufficient to increase uptake of PA103pscJ. This relative increase persists when bacterial infection is established at the basolateral surface of polarized cells, suggesting that the effect of RhoAV14 is not simply due to its known ability to disrupt tight junction integrity in polarized cells. RhoAV14-mediated stimulation of bacterial uptake is actin dependent as it is abrogated by exposure to latrunculin A. We also find that endogenous Rho GTP levels in epithelial cells are increased by infection with an internalized strain of P. aeruginosa; conversely, a poorly internalized isogenic strain expressing the bacterial anti-internalization protein ExoT causes decreased Rho GTP levels. Experimental inhibition of Rho, either by expressing dominant negative RhoAN19 or by inhibiting native Rho using a membrane permeable fusion construct of a Rho-specific inhibitor, C3 ADP-ribosyltransferase, does not inhibit PA103pscJ internalization in MDCK or HeLa cells. Models consistent with these data are presented.


Subject(s)
Epithelial Cells/microbiology , Escherichia coli Proteins , Pseudomonas aeruginosa/pathogenicity , rho GTP-Binding Proteins/metabolism , Animals , Bacterial Toxins/pharmacology , Cells, Cultured , Cytotoxins/pharmacology , Dogs , Enzyme Activation , Humans , Models, Biological , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
10.
Infect Immun ; 68(12): 7100-13, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083836

ABSTRACT

Pseudomonas aeruginosa, an important nosocomial pathogen of humans, expresses a type III secretion system that is required for virulence. Previous studies demonstrated that the lung-virulent strain PA103 has the capacity to be either cytotoxic or invasive. Analyses of mutants suggest that PA103 delivers a negative regulator of invasion, or anti-internalization factor, to host cells via a type III secretion system. In this work we show that the type III secreted protein ExoT inhibits the internalization of PA103 by polarized epithelial cells (Madin-Darby canine kidney cells) and J774.1 macrophage-like cells. ExoS, which is closely related to ExoT but has additional ADP-ribosylating activity, can substitute for ExoT as an anti-internalization factor. ExoT contains a signature arginine finger domain found in GTPase-activating proteins. Mutation of the conserved arginine in ExoT diminished its anti-internalization activity and altered its ability to disrupt the actin cytoskeleton. Cell fractionation experiments showed that ExoT is translocated into host cells and that mutation of the arginine finger did not disrupt translocation. In a mouse model of acute pneumonia, PA103DeltaUDeltaT reached the lungs as efficiently as PA103DeltaU but showed reduced colonization of the liver. This finding suggests that the ability to resist internalization may be important for virulence in vivo.


Subject(s)
ADP Ribose Transferases , Actins/metabolism , Bacterial Toxins/toxicity , Cytoskeleton/physiology , Exotoxins/toxicity , Macrophages/microbiology , Pseudomonas aeruginosa/pathogenicity , Virulence Factors , Animals , Arginine , Biological Transport , Cytoplasm/metabolism , Exotoxins/chemistry , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Virulence , Pseudomonas aeruginosa Exotoxin A
11.
Infect Immun ; 68(3): 1080-5, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10678910

ABSTRACT

Using polystyrene microspheres coated with heparin or heparan sulfate, it was shown that coated microspheres specifically bound eukaryotic cells and were endocytosed by nonprofessional phagocytic cells. Coated microspheres displayed properties of binding to eukaryotic cells that were similar to those of chlamydiae, and the microspheres were competitively inhibited by chlamydial organisms. Endocytosis of heparin-coated beads resulted in the tyrosine phosphorylation of a similar set of host proteins as did endocytosis of chlamydiae; however, unlike viable chlamydial organisms, which prevent phagolysosomal fusion, endocytosed beads were trafficked to a lysosomal compartment. These findings suggest that heparin-coated beads and Chlamydia trachomatis enter eukaryotic cells by similar pathways.


Subject(s)
Bacterial Adhesion , Chlamydia trachomatis/physiology , Heparin/metabolism , HeLa Cells , Heparitin Sulfate/metabolism , Heparitin Sulfate/pharmacology , Humans , Microspheres , Phosphorylation , Tyrosine/metabolism
12.
Cell Microbiol ; 2(6): 627-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11207614

ABSTRACT

Chlamydia trachomatis, an important cause of human disease, is an obligate intracellular bacterial pathogen that relies on the eukaryotic host cell for its replication. Recent reports have revealed that the C. trachomatis vacuole receives host-derived sphingolipids by fusing with trans-Golgi network (TGN)derived secretory vesicles. Here, it is shown that these lipids are required for the growth of the bacteria. C. trachomatis was unable to replicate at 39 degrees C in the Chinese hamster ovary (CHO)-derived cell line SPB-1, a cell line incapable of synthesizing sphingolipids at this temperature because of a temperature-sensitive mutation in the serine palmitoyltransferase (SPT) gene. Complementation with the wild-type SPT gene or addition of exogenous cell-permeable sphingolipid precursors to the mutant cells restored their ability to support chlamydial replication. L-cycloserine (L-CS) and fumonisin B1 (FB1), inhibitors of sphingolipid biosynthesis, decreased the proliferation of the bacteria in eukaryotic cells at concentrations that also decreased host cell sphingolipid synthesis. In the case of FB1, the vacuoles appeared aberrant; the addition of sphingolipid precursors was able to reverse the altered morphology of the FB1-treated vacuoles. Collectively, these data strongly suggest that the growth and replication of chlamydiae is dependent on synthesis of sphingolipids by the eukaryotic host cell and may contribute to this organism's obligate intracellular parasitism.


Subject(s)
Cell Line/enzymology , Chlamydia Infections/microbiology , Chlamydia trachomatis/growth & development , Sphingolipids/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Bacterial Adhesion , CHO Cells/enzymology , Chlamydia trachomatis/pathogenicity , Cricetinae , Humans , Serine C-Palmitoyltransferase
13.
Infect Immun ; 67(10): 5530-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10496945

ABSTRACT

Pseudomonas aeruginosa is a gram-negative opportunistic pathogen that is cytotoxic towards a variety of eukaryotic cells. To investigate the effect of this bacterium on macrophages, we infected J774A.1 cells and primary bone-marrow-derived murine macrophages with the P. aeruginosa strain PA103 in vitro. PA103 caused type-III-secretion-dependent killing of macrophages within 2 h of infection. Only a portion of the killing required the putative cytotoxin ExoU. By three criteria, terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling assays, cytoplasmic nucleosome assays, and Hoechst staining, the ExoU-independent but type-III-secretion-dependent killing exhibited features of apoptosis. Extracellular bacteria were capable of inducing apoptosis, and some laboratory and clinical isolates of P. aeruginosa induced significantly higher levels of this form of cell death than others. Interestingly, HeLa cells but not Madin-Darby canine kidney cells were susceptible to type-III-secretion-mediated apoptosis under the conditions of these assays. These findings are consistent with a model in which the P. aeruginosa type III secretion system transports at least two factors that kill macrophages: ExoU, which causes necrosis, and a second, as yet unidentified, effector protein, which induces apoptosis. Such killing may contribute to the ability of this organism to persist and disseminate within infected patients.


Subject(s)
Apoptosis , Macrophages/pathology , Pseudomonas aeruginosa/pathogenicity , Animals , Cell Line , Dogs , Epithelial Cells/pathology , HeLa Cells , Humans , Macrophages/immunology , Mice , Phagocytosis
14.
Infect Immun ; 67(7): 3207-14, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10377092

ABSTRACT

The interaction of Pseudomonas aeruginosa type IV pili and the glycosphingolipid asialo-GM1 (aGM1) can mediate bacterial adherence to epithelial cells, but the steps subsequent to this adherence have not been elucidated. To investigate the result of the interaction of pili and aGM1, we used polarized epithelial monolayers of Madin-Darby canine kidney (MDCK) cells in culture, which contained little detectable aGM1 on their apical surface but were able to incorporate exogenous aGM1. Compared to an untreated monolayer, P. aeruginosa PA103 displayed an eightfold increase in association with and fivefold more cytotoxicity toward MDCK cells pretreated with aGM1. Cytotoxicity of either carrier-treated or aGM1-treated monolayers required the type III secreted protein ExoU. Asialo-GM1 pretreatment of MDCK monolayers likewise augmented bacterial internalization of an isogenic invasive strain approximately fourfold. These increases were not seen in monolayers treated with GM1, the sialyated form of the glycolipid, and were inhibited by treatment with an antibody to aGM1. Also, the aGM1-mediated adhesion, cytotoxicity, and internalization required intact type IV pili since nonpiliated PA103 mutants were unaffected by aGM1 pretreatment of MDCK cells. These results demonstrate that epithelial cell injury and bacterial internalization can proceed from the same adhesin-receptor interaction, and they indicate that P. aeruginosa exoproducts solely determine the steps subsequent to adhesion.


Subject(s)
Bacterial Adhesion/physiology , Fimbriae, Bacterial/physiology , G(M1) Ganglioside/physiology , Pseudomonas aeruginosa/physiology , Animals , Cell Line , Dogs , Epithelial Cells/microbiology
15.
Infect Immun ; 67(7): 3625-30, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10377148

ABSTRACT

Type IV pili of the opportunistic pathogen Pseudomonas aeruginosa mediate twitching motility and act as receptors for bacteriophage infection. They are also important bacterial adhesins, and nonpiliated mutants of P. aeruginosa have been shown to cause less epithelial cell damage in vitro and have decreased virulence in animal models. This finding raises the question as to whether the reduction in cytotoxicity and virulence of nonpiliated P. aeruginosa mutants are primarily due to defects in cell adhesion or loss of twitching motility, or both. This work describes the role of PilT and PilU, putative nucleotide-binding proteins involved in pili function, in mediating epithelial cell injury in vitro and virulence in vivo. Mutants of pilT and pilU retain surface pili but have lost twitching motility. In three different epithelial cell lines, pilT or pilU mutants of the strain PAK caused less cytotoxicity than the wild-type strain but more than isogenic, nonpiliated pilA or rpoN mutants. The pilT and pilU mutants also showed reduced association with these same epithelial cell lines compared both to the wild type, and surprisingly, to a pilA mutant. In a mouse model of acute pneumonia, the pilT and pilU mutants showed decreased colonization of the liver but not of the lung relative to the parental strain, though they exhibited no change in the ability to cause mortality. These results demonstrate that pilus function mediated by PilT and PilU is required for in vitro adherence and cytotoxicity toward epithelial cells and is important in virulence in vivo.


Subject(s)
Adenosine Triphosphatases , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Fimbriae Proteins , Molecular Motor Proteins , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Animals , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genes, Bacterial , Mice , Pseudomonas Infections/pathology , Virulence/genetics
16.
J Bacteriol ; 180(9): 2359-66, 1998 May.
Article in English | MEDLINE | ID: mdl-9573186

ABSTRACT

We have characterized the Chlamydia trachomatis ribosomal promoter, rRNA P1, by measuring the effect of substitutions and deletions on in vitro transcription with partially purified C. trachomatis RNA polymerase. Our analyses indicate that rRNA P1 contains potential -10 and -35 elements, analogous to Escherichia coli promoters recognized by E-sigma70. We identified a novel AT-rich region immediately downstream of the -35 region. The effect of this region was specific for C. trachomatis RNA polymerase and strongly attenuated by single G or C substitutions. Upstream of the -35 region was an AT-rich sequence that enhanced transcription by C. trachomatis and E. coli RNA polymerases. We propose that this region functions as an UP element.


Subject(s)
Chlamydia trachomatis/genetics , Promoter Regions, Genetic , RNA, Bacterial/genetics , RNA, Ribosomal/genetics , Transcription, Genetic , Base Sequence , DNA Mutational Analysis , Escherichia coli/genetics , Molecular Sequence Data
17.
Infect Immun ; 66(4): 1413-20, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9529061

ABSTRACT

Previous characterization of Pseudomonas aeruginosa clinical isolates has demonstrated an inverse correlation between cytotoxicity and internalization by epithelial cells. To further investigate this relationship, we tested PA103, a cytotoxic P. aeruginosa strain, and 33 isogenic noncytotoxic transposon mutants for internalization by Madin-Darby canine kidney cells. The majority of the mutants were not internalized, demonstrating that an inverse correlation between cytotoxicity and bacterial uptake by epithelial cells is not absolute. Six of the noncytotoxic mutants, however, demonstrated measurable levels of internalization by standard aminoglycoside exclusion assays even though internalization of wild-type strain PA103 was not detectable. All six had evidence of protein secretion defects involving two proteins, a 40-kDa protein and a 32-kDa protein. These proteins, designated PepB (for Pseudomonas exoprotein B) and PepD, respectively, each had characteristics of type III transported proteins. In addition, nucleotide sequencing studies demonstrated that PepB and PepD are homologs of YopB and YopD, respectively, type III secreted proteins of Yersinia spp. necessary for the translocation of effector molecules into the cytoplasmic compartment of eukaryotic cells. Thus, while many mutations in PA103 result in loss of cytotoxicity without an appreciable increase in internalization, defects in transport of type III secretion proteins PepB and PepD correlate with both loss of cytotoxicity and gain of internalization. These results are consistent with type III secretion of an inhibitor of internalization that requires PepB and PepD for translocation into the host cell.


Subject(s)
Bacterial Proteins/metabolism , Pseudomonas aeruginosa/physiology , Amino Acid Sequence , Aminopeptidases/metabolism , Animals , Cell Line , Cloning, Molecular , Dogs , Epithelial Cells/microbiology , Glutamyl Aminopeptidase , Molecular Sequence Data , Mutation
18.
Mol Microbiol ; 27(4): 807-18, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9515706

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen and a leading cause of hospital-acquired pneumonia. We identified a 73kDa protein, designated Pseudomonas exoprotein A (PepA), that was secreted by P. aeruginosa strain PA103. PepA was necessary for in vitro killing of epithelial cells as well as virulence in a mouse model of acute pneumonia. Several properties of PepA suggested that it was secreted by a type III system. Secretion occurred without cleavage of a signal peptide and in low-calcium environments in the presence of a divalent cation chelator, as is the case for characterized P. aeruginosa type III secreted proteins. Secretion of PepA was absent from isogenic mutants with defective type III pathways. Finally, amino-terminal peptide sequence analysis indicated that the amino-terminal five residues of PepA were identical to those of ExoS and ExoT, two type III secreted proteins of P. aeruginosa. After secretion, PepA underwent cleavage at two sites, each with the sequence A-X-K-S, suggesting that the cleavage may be caused by a protease. The gene encoding PepA, designated pepA, was cloned and sequenced, and comparisons with the genetic database using BLAST alignments indicated that the nucleotide sequence of pepA and the inferred protein sequence of PepA had no homology to known sequences. A nucleotide sequence identical to the consensus element for binding of ExsA, a transcriptional activator of P. aeruginosa type III secretion genes, was located 84 bp 5' of the translational start codon. Analysis of transposon insertion mutants indicated that the carboxy terminus was required for cytotoxicity. Examination of respiratory clinical isolates demonstrated that pepA was a variable trait and probably acquired by horizontal transmission. Consistent with this hypothesis was the identification of a putative insertion element 94 bp 5' of the PepA translational start site. Analysis of G + C content of the PepA coding sequence and the adjacent insertion element suggested that they were acquired together from a different species. In summary, PepA is a secreted protein of P. aeruginosa that is necessary for epithelial cell cytotoxicity in vitro and virulence in a mouse model of pneumonia.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Disease Models, Animal , Dogs , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pneumonia/microbiology , Pseudomonas aeruginosa/isolation & purification , Sequence Analysis, DNA , Virulence/genetics
19.
Infect Immun ; 65(12): 5301-8, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9393830

ABSTRACT

Infection of epithelial cells by two biovars of Chlamydia trachomatis results in the tyrosine phosphorylation of several host proteins. The most prominent change in host protein tyrosine phosphorylation involves a complex of proteins with molecular masses of 75 to 85 kDa (pp75/85) and 100 kDa (pp100). The C. trachomatis-induced tyrosine phosphorylation of pp75/85 and pp100 is observed in several cell lines, including epithelial cells, fibroblasts, and macrophages. Subcellular fractionation and detergent solubility properties of pp75/85 are consistent with its association with the cytoskeleton. Phosphoamino acid analysis demonstrates that the pp75/85 complex is phosphorylated on both tyrosine and serine residues. Immunofluorescence studies of chlamydia-infected cells by using fluorescein isothiocyanate-phalloidin and antibodies to phosphotyrosine and cortactin demonstrate that tyrosine-phosphorylated proteins, as well as cortactin, are localized to the chlamydial vacuole and that this process is facilitated by actin.


Subject(s)
Chlamydia Infections/metabolism , Chlamydia trachomatis , Microfilament Proteins/metabolism , Animals , Biological Transport , Cortactin , HeLa Cells , Humans , Mice , Phosphorylation , Proteins/metabolism , Tyrosine/metabolism
20.
Mol Microbiol ; 24(6): 1249-62, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9218773

ABSTRACT

We have developed a simple, reproducible and rapid genetic screen for Pseudomonas aeruginosa-induced epithelial cell cytotoxicity in cultures of MDCK cells. This screen was used to isolate isogenic transposon-tagged non-cytotoxic mutants of a cytotoxic and lung-virulent strain of P. aeruginosa (PA103). The transposon-insertion site was determined by using an inverse polymerase chain reaction followed by DNA-sequence analysis. On the basis of phenotype and sequence analysis, these mutants fell into four classes. One class had absent or defective pill, based on their resistance to phage PO4 and/or loss of twitching motility (twt-). A second class exhibited decreased adherence. A third class of mutants exhibited probable defects in the machinery or targets of type III protein secretion. A final class of mutants exhibited decreased but not absent cytotoxicity. This class included members of the first three classes as well as other mutants. These results suggest that localized cytotoxicity is likely to require several steps and several components, including pili and other (unidentified) extracellular proteins. The type III protein-secretion apparatus appears to be involved in this process.


Subject(s)
Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Amino Acid Sequence , Animals , Bacterial Adhesion , Bacterial Proteins/genetics , Cell Line , DNA Transposable Elements , Dogs , Epithelial Cells , Fimbriae, Bacterial , Genes, Bacterial , Lung/cytology , Lung/microbiology , Molecular Sequence Data , Mutagenesis , Polymerase Chain Reaction , Sequence Analysis, DNA , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...